.1! $1.. . II :54! 1 2 .(‘V C; Alt 6 L. . . .'A ..3“:A>Jncn.fi. x £32 2. t: 9 .. é... . u a .x‘nhhwu‘. .._..n...uux..u..w! u... i. 8.. ., :35... F9. 52.. . Lazihx. - w .4 52...... . v 1,. w :33’ :‘ 52...}..‘d5 \az.t.z .5 :0). In .L . .II..... 17.6; 5\ l: ‘91.: r 332:4 )3}: ‘ 4;. $941.51... ‘1‘ :‘1: IY§1IL5151. was... lllllflllllllllll”I”MINIMUM!WW‘IlnlllHllll ‘3 3014112902 , LIBRARY (R 03.5; ,, Mlehlgan State University This is to certify that the dissertation entitled Investigations of Modified Edman Reagents and Modified Anabolic Steroids for the Electrospray Mass Spectrometric Detection prese1_ ted by Kuruppu A. N. Dharmasiri has been accepted towards fulfillment of the requirements for Ph . D . degree in Chemistry swim Major professor Dated) NH ”7% MS U is an Affirmative Action/Equal Opportunity Institution 0-12771 PLACE ll RETURN Boxmmnwofltbdnekommtnyourneow. TO AVOID FINES Mum on or More date duo. DATE DUE DATE DUE DATE DUE MSU leAn Afflnnltlvo mauve“ Oppommuy 1mm WIN-9.1 INVESTIGATIONS OF MODIFIED EDMAN REAGENTS AND MODIFIED ANABOLIC STEROIDS FOR THE ELECTROSPRAY MASS SPECTROMTRIC DETECTION By Kuruppu A. N. Dharmasiri A DESSERTATION Submitted to Michigan State University in partial fulfilment of the requirements for the degree of DOCTOR OF PHILOSOPHY Department of Chemistry 1 994 ABSTRACT INVESTIGATIONS OF MODIFIED EDMAN REAGENTS AND MODIFIED AN ABOLIC STEROIDS FOR THE ELECTROSPRAY MASS SPECTROMETRIC DETECTION By Kuruppu A. N. Dharmasiri Electrospray mass spectrometry (ESI/MS) has become a widely used technique owing to its excellent sensitivity and capacity to mass analyze high-molecular weight compounds due to multiply charging. This technique has gained wide acceptance in biological mass spectrometry due to the ease of coupling with liquid chromatographic systems. In this study, an electrospray source was designed and fabricated for the HP 5985 single quadrupole mass spectrometer. An interface between the mass spectrometer and a personal computer has been developed. The molecules containing pre-formed charged groups or basic functionalities greatly improve the ESI/MS response. The phenylthiohydantoin amino acids in the Edman degradation methodology are identified solely on the retention time on the chromatogram after the separation by high performance liquid chromatography followed by UV detection. New Edman reagents containing a basic pyridyl group were introduced to improve the detectability and the reliability of thiohydantoin derivatives using ESI/MS. The low level detection of anabolic steroids is difficult due to the extensive fragmentations under the conventional gas chromatography-mass spectrometric techniques. Anabolic ketosteroids were modified to form hydrazone derivatives or oxime derivatives with Girard’s T reagent or hydroxylamine hydrochloride, respectively. The modifications improve the detectability into the femtogram level using electrospray mass spectrometry. Tandem mass spectral studies showed that a loss of 59 Daltons was common to all derivatives. A selective method to detect the hydrazone derivatives was developed using the neutral loss scanning mode of a triple quadrupole mass spectrometer. Feasibility studies based on analysis of stanozolol using electrospray mass spectrometry combined with the neutral loss scanning mode or liquid chromatography showed that this technology is suitable for detecting stanozolol in urine samples. to my parents iv ACKNOWLEDGMENTS I would like to thank my research advisor, Dr. J. Throck Watson, for his support and encouragement throughout my graduate school career. I would also like to thank Dr. Doug Gage for continual encouragement he has given, Dr. Z-H. Huang for sharing his expertise on organic chemistry, Dr. Rawl Hollingsworth for his instruments, Dr. Jack Holland for advice on instrumentation, and Drs. John Allison and John McKraken for serving as committee members. I would also like to thank Mike Davenport for helping me build my confidence with electronics. This list is incomplete without Joe Laykem who gave me very helpful suggestions on HPLC and peptide chemistry. I want to acknowledge Mel, Melinda, Bev, and Curt along with the rest of the crew at the MSU-Mass Spectrometry Facility for their assistance and for making a pleasant working environment. I would also thank to Dr. Eugene Zaluzec for his automechanic lessons in addition to useful advice on chemistry. I enjoyed the cooparation of my friends Tim Nieuwenhuis, Kate Noon, and Patrick Lukulay. I thank them for their moral support in getting accustomed to MSU. I enjoyed working with past and present Watson group members. I am greatly indebted to my parents for creating in me a desire for the higher education. Finally, I thank my family for being patient with me and for being understanding. TABLE OF CONTENTS LIST OF FIGURES LIST OF TABLES CHAPTER 1 INTRODUCTION 1. Introduction and Objectives ........................................................................................... 1 I]. History and Development of Electrospray .................................................................... 2 111. Operating Principles and Mechanisms of Electrospray ................................................. 4 A. Production of Charged Droplets at the E81 Capillary tip .................................... 5 1. Efl‘ect of Surface Tension on E81 .................................. . .......................... 7 2. Dependence of Droplet Radius on Liquid Flow Rate ............................... 10 B. Shrinkage of Charged ES Droplets ..................................................................... 10 C. Formation of Gas-phase Ions ............................................................................. 12 1. Charged Residue Model .......................................................................... l3 2. Iribame and Thomson Ion Evaporation Theory ....................................... 13 3. Emission of Gas-Phase Ions from Taylor Cone Tip ................................. 16 D. Dependence of the Ion Intensity on the Concentration ....................................... 16 IV. Characteristics of ESI/MS Data .................................................................................. 20 A. Types of Analytes Required for ESI/MS ............................................................ 20 B. Interpretation of Multiple Charged Envelope ...................................................... 23 C. Why Derivatives ? .............................................................................................. 24 D. Oxidation of Analytes under the High Electric Field (ESI conditions) ................. 24 V. Mass Analyzers Coupled with ESI ................................................................................ 25 A Quadrupole Mass Analyzer ................................................................................ 25 B. Magnetic Sector Instrument ............................................................................... 25 vi C. Quadrupole Ion Trap Mass Analer .................................................................. 26 D. Fourier Transform Mass Analyzer ...................................................................... 27 E. Time of Flight Mass Spectrometer ..................................................................... 27 VI. Sample Introduction Techniques ................................................................................. 28 A.Coupling with Liquid Chromatographic systems ................................................. 28 B. Capillary Electrophoresis (CE) ........................................................................... 29 C. Ion Chromatography (IC) .................................................................................. 30 VII. Applications of ESI-MS ............................................................................................ 30 A. ESI/MS of Small Molecules ............................................................................... 31 B. Multiple Charging and High Molecular Weight Determination ............................ 31 1. Large Polypeptides and Proteins ............................................................. 31 2. Other High Molecular Weight Compounds .............................................. 32 C. Studies of Higher Order Structures .................................................................... 33 VIII. References ............................................................................................................... 3 5 Chapter 2 DEVELOPMENT OF INSTRUMENTATION FOR ESI/MS I. Introduction .................................................................................................................. 42 11. HP 5985 Single Quadrupole Mass Spectrometer .......................................................... 43 A Operating principles of Quadrupole Mass Spectrometer ..................................... 44 B. Vacuum System of HP 5985 MS ........................................................................ 45 111. Design Considerations for the ESI Interface ................................................................ 47 A. Vacuum System and Sensitivity ......................................................................... 47 B. Transport of Gas and Ions into Vacuum ............................................................. 48 VI. Fabrication of Source and Flange ................................................................................ 50 A Instrumentation for Therrnospray MS ............................................................... 52 B. SIMION Ion Simulation .................................................................................... 53 C. Source Modification ......................................................................................... 55 D. Vacuum Flange Modification ............................................................................. 58 VII E. Design and Fabrication of Ion Transport Capillary .............................................. 58 F. Electrospray Needle Assembly ............................................................................ 63 G. Electropolishing of the Capillary Tubes .............................................................. 64 V. Results and Discussion ................................................................................................. 67 A Data Collection .................................................................................................. 69 VI. Design and Construction of Computer Interface .......................................................... 72 A. Introduction ....................................................................................................... 72 B. Computer Interface ............................................................................................ 76 C. Data Acquisition and Processing Program .......................................................... 77 VII. TQMS ....................................................................................................................... 82 A. MS/MS Scan Modes with TQMS ...................................................................... 85 l. Daughter Ion Scan .................................................................................. 85 2. Parent Ion Scan ...................................................................................... 86 3. Functional Relationship Scan .................................................................. 86 4. Selected Reaction Monitoring ................................................................. 87 VIII. References ............................................................................................................... 91 Chapter 3 PEPTIDE SEQUENCING I. Introduction .................................................................................................................. 93 A. Edman Degradation ........................................................................................... 94 II. Thiohydantoin Amino Acid Analysis by Mass Spectrometry ......................................... 98 A Electron Impact (E1) ......................................................................................... 98 B. Chemical Ionization (CI) .................................................................................... 99 C. Two-Step Laser Desorption/Multiphoton Ionization .......................................... 100 D. Thermospray ................................................................................................... 101 E. Atmospheric Pressure Chemical Ionization ........................................................ 102 F. Electron Capture Negative Ionization ................................................................ 102 G. Electrospray ..................................................................................................... 102 viii 111. Other Derivatives ........................................................................................................ 103 A. Fluorescent Derivatives ..................................................................................... 103 B. Radioactive Derivatives .................................................................................... 105 IV. Automated peptide Sequenator ................................................................................... 107 A Liquid Phase ..................................................................................................... 107 B. Solid Phase ....................................................................................................... 108 C. Gas Phase ......................................................................................................... 109 V. Other Mass Spectrometric Techniques for Peptide Sequencing ..................................... 109 A. Fast Atom Bombardment .................................................................................. 110 B. Electrospray ..................................................................................................... 111 C. Matrix Assisted Laser Desorption ..................................................................... 115 VI. References .................................................................................................................. 116 Chapter 4. DEVELOPMENT OF EDMAN REAGENTS FOR ESI/MS DETECTION I. Introduction .................................................................................................................. 122 H. Alternative Edman Reagents ......................................................................................... 123 A. t-BAMPITC ..................................................................................................... 123 1. Introduction ............................................................................................ 123 2. Experimental ........................................................................................... 125 3. Results and Discussion ............................................................................ 126 B. Designing New Reagents .................................................................................. 127 III. Preparation of Isothiocyanate ...................................................................................... 127 A Method I ........................................................................................................... 128 B. Method 11 .......................................................................................................... 128 C. Separation of Mixture ........................................................................................ 132 1. Preparative Thin Layer Chromatography ................................................. 132 2. Column Chromatography ........................................................................ 133 3. Results and Discussion ............................................................................ 134 ix D. Characterization of Isothiocyanates .................................................................... 134 1. Colorimetric Detection ............................................................................ 134 2. Mass Spectrometry ................................................................................. 135 IV. Thiohydantoin amino acids .......................................................................................... 136 A. Synthesis ........................................................................................................... 136 B. Extraction .......................................................................................................... 136 C. Mass Spectral Analysis of Thiohydantoins .......................................................... 137 D. ESI/CID/MS/MS ............................................................................................... 144 V. Studies on Reaction Completeness ............................................................................... 150 A. Experimental ..................................................................................................... 150 B. Results and Discussion ....................................................................................... 150 l. Coupling Reaction with PMITC .............................................................. 150 2. Results and Discussion ............................................................................ 151 D. Cleavage Reaction of PyTC-peptides ................................................................. 151 E. Results and Discussion ....................................................................................... 153 VI. Peptide Sequencing ..................................................................................................... 155 VII. Post-translationally Modified Amino Acids ................................................................ 156 VIII. Conclusion ............................................................................................................... 160 IX. References .................................................................................................................. 161 Chapter V. MODIFICATION OF ANABOLIC STEROIDS FOR ESI/MS I. Introduction .................................................................................................................. 163 11. Modification of Steroids for ESI/MS ............................................................................ 165 III. Hydrazone Derivative Formation ................................................................................. 166 A Mass Spectral Analysis ...................................................................................... 166 B. Detection Limits of Hydrazones ......................................................................... 170 C. ESI/MS/MS Studies of Hydrazones ................................................................... 174 D. Study of Fragmentations with Offset Energy and Pressure ................................. 175 X E. FAB/MS Analysis of Hydrazones ....................................................................... 178 F. High Energy CID on F AB/MS ........................................................................... 180 G. Comparison of ESI/CID/MS/MS with F AB/CID/MS/MS .................................. 180 H. Neutral Loss Scanning Studies of Hydrazone Derivatives ................................... 182 I. LC/MS Analysis .................................................................................................. 182 IV. Feasibility Study of Steroid Detection in Urine ............................................................ 186 A. MS/MS Studies of the Derivatized Urine Extract ............................................... 188 V. Kinetic Studies of the Hydrazone Formation Reaction .................................................. 191 A. Dependence of Reaction Conversion (%) with Temperature ............................... 191 B. Dependence of Reaction Conversion (%) with Acid Concentration .................... 192 VI. Source CID during ESI/MS ........................................................................................ 192 A. MS/MS of the (M-59)+ Ion Derived from Methyltestosterone Hydrazone .......... 193 B. MS/MS of the (M-87)+ Ion Derived from Methyltestosterone Hydrazone .......... 194 VII. Oxime Derivative Formation ...................................................................................... 199 A. Results and Discussion ....................................................................................... 199 B. Tandem Mass Spectrometric Studies of Oximes ................................................. 205 C. Detection Limits of Steroid Oximes ............... ................................................... 205 VIII. Investigation of Stanozolol by ESI/MS ..................................................................... 207 A. Introduction ...................................................................................................... 207 B. Mass Spectral Analysis ...................................................................................... 208 C. Tandem Mass Spectrometry of Stanozolol ......................................................... 211 D. Comparison of ESI/MS/MS with FAB/MS/MS ................................................. 213 B. Effect of Solvent Composition ........................................................................... 215 F. ESI/MS/MS of CH3CN Adduct Ion of ST ......................................................... 215 G. HPLC of Stanozolol .......................................................................................... 218 H. LC/MS of Stanozolol ........................................................................................ 218 I. Urine Extract ...................................................................................................... 220 xi IX. Conclusions ................................................................................................................ 221 X. References ................................................................................................................... 224 Figure. 1-1. Figure 1-2. Figure 1-3. Figure 1-4. Figure 1-5. Figure 1.6. Figure 2-1. Figure 2-2. Figure 2-3. Figure 2-4. Figure 2-5. Figure 2-6. List of Figures Components of the electrospray mass spectrometer used in this study ....... 5 Schematic representation of life time of parent and offspring droplets ....... l4 Schematic representation of Initial state (a) and transition state (b), proposed by Iribarne and Thomson. Evaporating ions leaves as a cluster M+(SL)m, where SL are solvent molecules, r is the distance of ions from surface ..................................................................................................... 18 ESI/MS mass spectrum of 1, 10 phenanthroline (2.7 x 10‘5M) and (CH3)4N1 (6.5 x ro-8M) in methanol/water (SO/50% v/v). Adapted from ref 12 ....................................................................................................... 21 The electrospray mass spectrum of myoglobin (M.W. = 16,951). The data were collected on a TSQ 700 mass spectrometer. 10 pmol were consumed to obtain the spectrum ............................................................. 22 Hypothetical mass spectrum containing multiply charged ions ................... 23 Stability diagram of (a, q) space showing regions (patchwork area) that corresponds to mathematically stable ion trajectories in the quadrupole mass spectrometer .................................................................................... 46 Expansion of gas into vacuum; (a). simplified scheme with shockwaves and silence zone; (b). generation of a beam of gas and ions by sampling from a silent zone, the skimmer penetrates the mach disk; (c). sampling of gas and ions with a skimmer located beyond the mach disk ....................... 51 The Vestec thermospray interface for the HP 5985 mass spectrometer ..... 54 The plots from the SIMION ion simulation. (a) Heated capillary is right angle to the axis of the mass analyzer. (b). Heated capillary is parallel to the mass analyzer. In both cases, source block is at ground potential ...... 56 The ESI source designed for the HP 5985 mass spectrometer ................... 60 The ion transport capillary tube designed for the HP 5985 mass spectrometer ....................... ............ .. .......................................... 62 xiii Figure 2-7. Figure 2-8. Figure 2-9. Figure 2-10 Figure 2-11. Figure 2-12. Figure 2-13. Figure 2-14. Figure 2-15. Figure 2-16. Figure 2-17. Figure 2-18. Figure 2-19. Figure 2-20. Figure 3-1. Figure 3-2. Electrospray Needle. The probe can be used with or without nebulizer gas ........................................................................................................... 65 The complete electrospray interface on the HP 5985 mass spectrometer...66 The circuit diagram for measuring ESI current ......................................... 73 ESI/MS mass spectrum of Lys-Tyr-Lys (M.W. = 43 7) ............................. 74 ESI/MS mass spectrum of Angiotensin III (M. W. = 917). Flow rate = lOuL/min. ESI needle at 3.9kV. Heated capillary at 100°C and 100V and the Skimmer at 15V ................................................................................. 75 The block diagram of the digital interface for the HP 5985 mass spectrometer ............................................................................................ 78 The schematic diagram of the CIO-DIO48 Interface board ....................... 79 The schametic diagram of the custom made adapter to go into the digital board on the HP 5985 mass spectrometer ................................................. 80 The cables from CIO-DIO48 board to the HP 5985 mass spectrometer....8l The control panel of the modified data system for the HP 5985 mass spectrometer ............................................................................................ 83 The ESI/MS mass spectrum of stanozolol acquired using the new data system ...................................................................................................... 84 The reconstructed total ion chromatogram of a LC/ESI/MS analysis of (100pg) testosterone hydrazone. C-18 (15cm x 800 m, 3 urn particles) column. Flow rate 40pL/min of CH3CN/HZO/TF A (40/59.9/0.1 %v/v/v). Scan rate 350-450 in 1 sec ....................................................................... 84 The Finnigan Electrospray source on the TSQ 700 mass spectrometer. It incorporates a tube lens and an octapole rods to improve the ion transmission efficiency .............................................................................. 89 The TSQ 700 triple quadrupole mass spectrometer ................................... 90 Chemical Scheme for Edman Degradation of Peptide ............................... 97 Structures of proposed fluorescent reagents for Edman sequencing .......... 106 xiv Figure 3-3. Figure 3-4. Figure 4-1. Figure 4-2. Figure 4-3. Figure 4-4. Figure 4-5. Figure 4-6. Figure 4-7. Figure 4-8. Figure 4-9. Figure 4-10. Figure 4-11. Figure 4-12. Fragmentation of the peptide backbone and nomenclature proposed by Roepstorff and Fohlmann ......................................................................... 112 Structures of the common ions encountered in FAB-CAD-MS/MS and their nomenclature as revised by Biemann ................................................. 113 The structures of conventional Edman reagent (a), phenylthiohydantoin derivative (b), reagents proposed by Aebersold et al. (c), and Basic et al. ((1) ............................................................................................................ 124 Structures of t-BAMPITC and arninomethylphenylthiohydantoin amino acid .......................................................................................................... 125 The modified Edman reagents .................................................................. 127 The reaction scheme of isothiocyanate formation by Makaiyama Method. 130 Reaction scheme for the synthesis of DPT ................................................ 131 Scheme of isothiocyanate formation using DPT isothiocyanate transfer reagent ..................................................................................................... 131 FAB/CAD/MS/MS spectrum of val-PMTH .............................................. 138 Portion of the ESI/MS mass spectrum of PyTH-val (5pmol). Sample was injected into a stream of CH3OH/HzO/HOAC (50/49.9/0.1 %v/v/v) at a flow rate of IOOuUmin ............................................................................ 140 The mass spectral profile obtained by injecting increasing amounts of purified PyTH-phe derivative into a stream of CH3 OH/HZO/HOAC (50/49.9/0.1%v/v/v) at a flow rate of lOOuUmin ...................................... 141 Portion of the ESI/MS mass spectrum of PMTH-val (5pmol). Sample was injected to a stream of CH3OH/HzO/HOAC (50/49.9/0.1 %v/v/v) at a flow rate of lOOuI/min ......................................................................... 142 The profile obtained for the replicate injections of the PM-thiohydantoin amino acid in selected ion (m/z 298) monitoring mode. The conditions were maintained as in the previous experiment ......................................... 143 Proposed fi'agmentation patterns for the PyTH-lys under ESI/CID/MS/MS ..................................................................................... 144 Figure 4-13. Figure 4-14. Figure 4-15. Figure 4-16 Figure 4-17. Figure 4-18. Figure 4-19. Figure 4-20. Figure 4-21. Figure 4-22. Figure 5-1. Figure 5-2. ESI/MS/MS mass spectrum of PyTH-lys. Collision gas pressure was at 3mtorr and energy was at 30eV. Sample was infused at a flow rate of 3|.tL/min. Mass spectrometer was scanned 100 - 450 Daltons in 0.5 sec....145 ESI/CID/MS/MS spectrum of PMTH-asp. Collision energy at 30eV and pressure at 3 mtorr and the proposed fi'agrnentations ................................ 146 ESI/CID/MS/MS mass spectrum of PyTH-leu and the proposed fragmentation pattern. Collision energy 30eV and pressure 3 mtorr .......... 148 ESI/MS/MS mass spectrum of PyTH-ile amino acid derivative and the proposed fragmentation pattern. Conditions were similar to those of Figure 4-15 .............................................................................................. 149 Kinetic assessment of coupling reaction between pyridyl isothiocyanate and bradykinin (RPPGFSPFR). The peptide was treated with 2.5% PleC in a buffer solution and coupling reaction was assessed by analyzing the reaction mixture by RP-HPLC ............................................. 152 Kinetic assessment of coupling reaction between pyridylrnethyl isothiocyanate and bradykinin (RPPGFSPFR). The peptide was treated with 2.5% PMITC in a buffer solution and coupling reaction was assessed as in Figure 4-17 ........................................................................ 152 F AB/MS mass spectra showing the cleavage of thiocabamyl peptide (VGVAPG) with anhydrous TFA at 50°C. (a) PyTC-peptide afier 10 min., (b). PMTC-peptide afier 2min., and (c) PTC-peptide after 5 min .......................................................................................................... 154 HPLC chromatogram for the PTH amino acid standard containing hydroxy-proline. Hydroxy-proline eluted as two peaks ............................. 157 ESI/MS mass spectrum of hydroxy-proline (5pmol). Sample was injected into a stream of stream of CH3OH/I120/I-IOAC (50/49.9/0.1 %v/v/v) at a flow rate of lOOuL/min .......................................................................... 158 The ESI/MS mass spectrum of PMTH-O-phospho serine (5pmol). Sample was injected to a stream of CH3OH/HzO/HOAC (50/49.9/0.1 %v/v) at a flow rate of 100uL/min ........................................................... 159 Numbering system for the steroid carbon skeleton .................................... 167 The Structures Anabolic Steroids banned By IOC .................................... 168 Figure 5-3. Figure 5-4. Figure. 5-5. Figure. 5-6. Figure 5-7. Figure 5-8. Figure 5-9. Figure 5-10. Figure 5-11. Figure. 5-12. Figure. 5-13. The reaction scheme for the formation of steroid hydrazone. The Girard's T reagent reacts with the keto group of the steroid to form hydrazone derivative ................................................................................................. 171 Portion of the ESI/MS mass spectrum of fluoxymesterone hydrazone chloride (molecular weight of cation is 450u) ........................................... 172 Detection limits for the methyltestosterone hydrazone. The samples were injected into a stream of CH3OH/H20/HOAC (50/49.9/0. 1% v/v) flowing at a rate of 100uI./rr1in in increasing concentration. The injection volume was SuL. The amounts are indicated above the corresponding peak ......................................................................................................... 173 ESI/MS/MS spectrum of the molecular cation of methyltestosterone hydrazone. The spectrum was obtained by infusing the sample at a flow rate of 3|.LL/min. and at a concentration of 100 pg/uL. The mass spectrometer was scanned at a rate of 980u/sec and spectra were averaged for one minute. The collision energy was set at 40eV (lab). Ar was used as the collision gas and the collision cell pressure was 3 mtorr... 17 7 Variation of product ion abundances with collision offset energy at the collision cell gas pressure of 3mtorr .......................................................... 179 The variation of product ion abundances with collision cell gas pressure at the collision offset energy of 40eV ........................................................ 179 The FAB/MS spectrum of methyltestosterone hydrazone (1 ng). Glycerol was used as the matrix and matrix peaks are indicated by an asterisk ........ 181 The FAB/CAD/MS/MS spectrum of methyltestosterone ........................... 180 The neutral loss scan mass spectrum of nandrolone hydrazone .................. 184 Total ion current (RTIC) chromatogram for LC/MS analysis of hydrazones of model compounds (a) and mass chromatograms for (b) fluoxymesterone hydrazone (m/z 450), (c) methyltestosterone hydrazone (m/z 416), (d) testosterone hydrazone (m/z 402) and (e) nandrolone hydrazone (m/z 388) ................................................................................ 185 Total ion current chromatogram (RTIC) for LC/MS analysis of hydrazones of urine extract (a) and mass chromatograms for (b) fluoxymesterone hydrazone (m/z 450), (c) methyltestosterone hydrazone (m/z 416), and (d) nandrolone hydrazone (m/z 388) ................................. 187 xvii Figure. 5-14. Figure 5-15. Figure 5-16. Figure 5-17. Figure 5-18. Figure 5-19. Figure 5-20. Figure 5-21. Figure 5-22. Figure 5-23. Figure 5-24. Figure 5-25. Figure 5-26. Figure 5-27. Portion of the electrospray mass spectrum of extracted urine sample after spiking with methyltestosterone (10ng/mL) followed by derivatization with Girard's T reagent. The sample was infused at a flow rate of 5 uL/min without separation. The spectrum is dominated by signals from the impurities in the urine sample .............................................................. 189 The simplified mass spectrum of the sample used to obtain figure 5-14, by neutral loss scanning (59u) of the quadrupoles (Q1 and Q3). The collision energy was set to 40eV and collision gas pressure at 3mtorr ....... 190 Variation of percent reaction conversion with time and temperature for hydrazone formation ................................................................................ 195 Efi‘ect of HOAC concentration on percent reaction conversion ................. 195 ESI/MS mass spectrum obtained by inducing CID at the octapole. The octapole ofi‘set was set to 40eV ................................................................ 196 The MS/MS spectrum of the ion at m/z (M-59)+ derived fi'om methyltestosterone (m/z 416) by source CID ............................................ 197 The MS/MS spectrum of the ion at m/z (M-87)+ derived fi'om methyltestosterone (m/z 416) by source CID ............................................ 198 Formation of testosterone oxime fiom testosterone and hydroxylamine hydrochloride ........................................................................................... 201 ESI mass spectrum of methyltestosterone oxime in CH3OH/H20/HOAC (50/49.9/0.1 % v/v/v) ............................................................................... 202 ESI mass spectrum of methyltestosterone oxime in ACN/HZO/HOAC (50/49.9/0.l % v/v/v) ............................................................................... 203 ESI/CID/MS/MS spectrum of (M+CH3CN+H)"” for testosterone oxime. The offset energy was set at SeV and the collision energy at 3 mtorr (Ar).204 ESI/CID/MS/MS spectrum of testosterone oxime .................................... 206 Structure of anabolic stanozolol, and preferred metabolic hydroxylation sites indicated by an arrow ....................................................................... 209 ESI/MS mass spectrum of stanozolol in CH3OH and injected to a stream of CH3OH/H20/HOAC (50/49.9/0.1 % v/v/v)- The capillary at 200°C and needle at 4.5kV ................................................................................. 210 xviii Figure 5-28. Figure 5-29. Figure 5-30. Figure 5-31. Figure 5-32. Figure 5-33. Figure 5-34. ESI/MS/MS mass spectrum of stanozolol. Collision energy was 55eV and the gas pressure was 3mtorr ............................................................... 212 The possible fragmentation pathways of stanozolol molecule .................... 213 FAB/CAD/MS/MS mass spectrum of stanozolol ESI/MS mass spectrum of stanozolol in CH3OH as injected into a stream of CH3CN/H20/HOAC (50/49.9/0.1 % v/v/v)- The capillary was at 200°C and needle at 4.5kV ....................................................................... 216 The ESI/MS/MS mass spectrum of the CH3CN adduct ion of ST. The offset energy was at SeV and the collision cell pressure was at 2 mtorr ..... 217 The reconstructed mass chromatograms for (Mi-H)+ (m/z 329) and (M+CH3CN+I-I)+ (m/z 370) for stanozolol (1ng injected) during an LC/MS run ............................................................................................... 219 Selected reaction monitoring profile of stanozolol in urine extract (10uL). The solvent system was CH30H/H20/HOAC (50/49.9/0.1 %v/v) at a rate of 100uL/min ................................................................... 223 xix LIST OF TABLES Table 1-1. Onset voltages Von and surface tension 7 for ES with different solvents... 8 Table 1-2. Experimentally determined coefficients for the equation 1-14. All coefiicients relative to kgs+ = 1. These coefiicients are valid only at concentrations above 10' M .................................................................... 19 Table 2-1: Function of the quadrupoles in various TQMS scan modes ....................... 87 Table 5-2. The steroids used to prepare hydrazone derivatives .................................. 170 Table 5-3. A list of peaks observed in MS/MS studies of hydrazone molecular cation M+. ......................................................................................................... 176 CHAPTER 1 INTRODUCTION AND OBJECTIVES I. INTRODUCTION Since its discovery in 1912, mass spectrometry (MS) has developed into one of the most powerfirl and versatile techniques available for the study of biomolecules. Over the years, MS has been applied not only to problems of structure determination, but to assessment of varied processes ranging from drug metabolism to enzyme kinetics. Rapid advances in the biological sciences, especially in molecular biology and biochemistry, have led to an increase in the demand for chemical and structural information on biologically active molecules. Recent innovations in ionization methods, improvements in ion detection, increases in mass range of instruments, the use of tandem instruments, and improved interfaces for separation techniques are providing mass spectrometrists with the capabilities needed to analyze a wide variety of biologically active materials. Recent advances in mass spectrometric techniques such as electrospray ionization mass spectrometry (ESI/MS) and matrix assisted laser desorption ionization mass spectrometry (MALDI) have the capacity to establish molecular weights of biomolecules, even those larger than 100,000 Daltons. Analytical methods involving MS have been reported with detection limits in the femtogram (10'15g) and attomole (10'1 3 mol) range (1, 2). In trace analysis of biologically important compounds, a single peak representing the analyte molecule is desired for sensitive detection. The soft ionization techniques such as ESI/MS or MALDI have the capabilities to give a single peak representing the 1 2 molecule. ESI/MS has shown distinct advantages in biological mass spectrometry, because of its capabilities to retrofit into almost all the types of mass analyzers and amenable to various kinds, of separation techniques. In this manuscript, ESI/MS in the area of bioanalytical applications will be discussed in greater detail. The intention of this chapter is to introduce (i) the principles of electrospray ionization (ESI), (ii) the operating principles of ESI/MS, (iii) coupling of ESI with various mass analyzers, (iv) coupling of various separation methods with ESI/MS, and (v) a brief summary of applications of ESI/MS. A. Objectives of the Research Project The primary goals of this research investigative project are (1) the modification of an HP 5985 single quadrupole mass spectrometer for electrospray ionization, (2) to improve the reliability of Edman degradation chemistry by using mass spectrometry as a detector, and (3) to develop LC/MS techniques to detect anabolic steroids in biological fluids. II. History and Development of Electrospray Electrospray ionization-mass spectrometry (ESI/MS) has its origins in research that long preceded the current flurry of activity. The study of the electrospray phenomenon extends back perhaps over two and one-half centuries to the work of Bose in 1745 (3) and certainly to that of Zeleny early in this century (1917) (4). The seminal research into the use of electrospray as an ionization method for macromolecules was due to Malcom Dole and co-workers (5, 6), who more than twenty years ago, performed extensive studies into the electrospray process and defined many of the important experimental parameters. The purpose of Dole's studies was to use ESI to produce gas- 3 phase macro-ions. Dole's approach was largely an adoption of the electrospray studies performed in the 1930's by Chapman, in which ion mobility studies were conducted for electrospray ionization of low-molecular weight salt solutions (7). Experimental evidence was presented by Dole for ionization of Zein (M.W.=50,000 Da) (8) and hen egg lysozyrne (M.W.=14306 Da) (9). Interpretation of their results were problematic, however, because a mass spectrometer was not available, and only ion retardation (5, 6, 8) and ion mobility (9) measurements were obtained. ~ In 1984, ESI combined with mass spectrometry was reported, essentially simultaneously, by both Yamashita and Fenn (10) and Aleksandrov et al. (11). Fenn and co-workers also demonstrated ESI/MS in the negative ion mode (12), building upon the original negative ion work by Dole and co-workers (5, 6, 8). Using magnetic sector instruments, the Russian researchers independently demonstrated the on-line combination with liquid chromatography in 1984 (11), and over the next few years applied ESI-MS to oligosaccharides (13), and intact polypeptides up to molecular weight of 1500 Daltons ( l4), and developed chemical digestion-based methods for their sequence determination (15). Of particular importance to ESI analysis of macromolecules is the phenomenon of multiple charging. The multiple charging of lysozyrne by ESI was reported by Dole on the basis of ion mobility measurements (9), but difficulties in the interpretation led them to suggest a charge state of only 3+, substantially lower than that shown by subsequent MS studies. In 1985, both Penn and co-workers (16) and Aleksandrov et al. (13, 14) reported dominant contributions for doubly charged ions for polypeptides such as bradykinin (M.W=1060) and gramicidin S (M.W=1141). Most significant in the work by Penn and co-workers was the observation of multiple charging with as many as 23 charges with polyethylene glycols due to the attaChment of sodium ions (17). Since the higher- molecular-weight polyethylene glycol samples had relatively broad molecular weight 4 distributions, only unresolved ”humps" of ions were observed for these materials. In 1988, F enn and co-workers (18) first reported ESI/MS spectra of intact multiply protonated molecules of proteins up to 40,000 Daltons having as many as 45 positive charges. Several research groups quickly duplicated and extended the applications of ESI/MS (19- 21). Since its introduction in 1988 as a tool for the ionization of large biomolecules, the practice of ESI/MS has grown impressively. Although its inception was nearly simultaneous with MALDI mass spectrometry for similar applications (22), use of ESI/MS has been substantially greater. The most important reasons are the ability to retrofit an ESI source to existing mass analyzers easily and the speed with which commercial instrumentation became available. ESI/MS has clear advantages for analysis of liquid samples and solutions, particularly the efiluents fiom rnicroscale or nanoscale capillary separation methods. In addition, tandem mass spectrometry has been shown to be usefill for dissociation studies of the multiply charged biomolecules produced by ESI, allowing much greater dissociation efliciencies than for singly charged molecular ions (23). ESI combined with high resolution instruments such as magnetic sector (24) and Fourier transform mass spectrometers (25) gives a better accuracy for high molecular weight compounds. By combining the above capabilities with the high sensitivity of ESI/MS serves as a very attractive tool for the mass spectrometry community, especially those who are working with biomolecules. 111. Operating Principles and Mechanisms of the Electrospray Ionization Process Important components of the electrospray mass spectrometric system used in this study are shown in Figure l. The solution is forced through a small capillary tube held at high electric potential. A fine mist or spray is formed at the tip of the ESI needle and is drawn toward a heated capillary tube held at a lower potential. The analyte and solvent 5 molecules emerging from the capillary tube are sampled through a skimmer and ions are focused by a set of lenses into the mass analyzer. The ions are detected by a continuous dynode electron multiplier. Heated C '11 ESI Needle 8‘” my / . l , Mass Analyzer 3-5 kV \ . 100V Skimmer Lenses Figure. 1-1. Components of the electrospray mass spectrometer used in this study There are four major processes in ESI/MS: the production of charged droplets fi'om electrolyte dissolved in a solvent, shrinkage of charged droplets by solvent evaporation and repeated disintegration, the mechanism of gas phase ion formation, and secondary processes, by which gas-phase ions are modified in the atmospheric and ion sampling regions of the mass spectrometer. A. Production of Charged Droplets at the ESI Capillary Tip When a capillary tube is held at high potential and placed near a counter electrode at ground potential, an electric field between the capillary tube and the electrode is created. The magnitude of the electric field at the capillary tip, EC, for a given potential, V0, is given by the equation 1-1. (26, 27) 6 Ec=2Vc/rc ln(4wrc) 1’1 where rc is the radius of the capillary tube and d is the distance between the capillary tube and the counter electrode. Equation l-l provides the field at the capillary tip in the absence of a spraying solution. The field EC is proportional to the applied potential Vc on the capillary. EC is essentially inversely proportional to re; it decreases slowly, with a logarithmic dependence, with the distance d. For example, in a typical system of an operating voltage Vc of 2kV, capillary radius rc of 0.2mm and distance d of 2cm produces an electric field EC of3.3 x 106 V/m. The imposed field E will also partially penetrate the liquid at the capillary tip. When a positive potential is applied to the capillary tip, some positive ions in the liquid will drifl toward the liquid surface and some negative ions will drift away from it until the imposed field inside the liquid is essentially removed by this charge redistribution. However, the accumulated positive charge at the surface leads to destabilization of the surface because the positive ions are drawn downfield but cannot escape fi'om the liquid. The surface is drawn out downfield such that a liquid cone forms. This is called a Taylor cone (28). At a sufficiently high field E, the cone is not stable and a liquid filament with a diameter of a few micrometers, whose surface is enriched with positive charges, is emitted from the Taylor cone tip. At some distance downstream, the liquid filament becomes unstable and forms separate droplets. The droplets' surfaces are enriched with positive ions for which there are no negative counter ions in the droplet. The length of the unbroken filament decreases if the field E is increased. At higher fields, a multispray condition is reached in which the central cone disappears and droplet emission occurs from a crown of four to six short liquid tips formed at the rim of the capillary (29). This ion separation mechanism, which is called the electrophoretic mechanism, is also most 7 plausible on energetic grounds. Pfeifer and Hendricks (27) were probably the first authors who explicitly proposed and discussed the electrophoretic mechanism. More recently, Hayati, Bailey and Tadros (30) also endorsed the electrophoretic charging mechanism in a comprehensive examination of features of the ESI process. The number of excess charges Q on a droplet is large relative to the number of solute molecules in the droplets. For example, flash microphotographs taken under typical operating conditions for large molecules (3 uL/min of lumol analyte in 1:1 methanol/water) show initial droplet diameters of approximately 2.8um (31). Simultaneous measurement of spray current indicates that each of those initial droplets has approximately 44,000 charges, corresponding to a Q/Ni value of approximately 6 (31). 1. Effect of Surface Tension on ESI Smith who also assumed the electrophoretic charging mechanism provided a very useful equation (1-2) for the voltage required for the onset of charged-droplet emission (32). 2Tr cosO _ ”2 V0n= A [—c—eo—QJ ln(4h/rc) 1-2 where 90 is the cone half-angle, so is pennitivity of free space, and A is a constant By combining equation 1-1 with 1-2, one obtains the onset voltage Von Von e 2 x105(7rc)1/21n(4d/rd 1-3 8 where y is the surface tension of the solvent, rc is the capillary radius, and d is the distance between capillary tip and the counter electrode. Experimental verification of equation 1-3 has been provided by Smith (32) and Kebarle et al. (33, 34). For stable ESI operation one needs to apply a few hundred volts higher than Von- Table 1 shows the evaluated Von values for difi‘erent solvents for d = 4cm and rc = 0.1mm (35). The surface of the solvent with the highest surface tension is the most difficult to stretch out into a Taylor cone and a liquid filament; this leads to the requirement for the highest Von- Table 1-1. Onset voltages Von and surface tension 7 for ESI with different solvents CH3 OH CH3CN (CH3)2$O H20 gm (kV) 2.2 2.5 3.0 4.0 y (N/m2) 0.0226 0.030 0.043 0.073 Henion et al. (36) and Chait et al. (37) heated the electrospray needle assembly to promote droplet formation when aqueous solutions are electrosprayed. At high temperatures, the surface tension becomes low and the spray process takes place readily. Buchanan et al. (3 8) studied the effects of surface tension and surface activity on electrospray response. They investigated the ESI/MS response with varying the concentration of cationic, anionic, and non-ionic surfactants. An increase in ESI/MS response was observed with increasing concentration of cationic surfactant and non-ionic surfactants and at a certain limit (0.3mM for Triton X-100), the ESI/MS response drops ofl' sharply. These surfactants decrease the surface tension with increasing concentration and alter a certain limit, surfactants may compete for the excess charges in the droplets. 9 The capillary current I which results fiom charged droplets leaving the capillary, is of interest because it provides a quantitative measure of the excess positive electrolyte ions produced by the spray. A theoretical derivation giving the dependence of I on experimental parameters has been proposed by Hendricks (27). 1=AHVfEeon n=0.2-0.3 14 where A H is a constant that can be evaluated and depends on the dielectric constant and surface tension of the solvent, Vf is the flow rate (volume/time), E is the electric field at the tip, a is the conductivity of the solution, and v, e, n are experimentally determined Mcients. Kebarle et al. compared the capillary current with the mass spectrometrically detected intensity of protonated molecules and they found no close relationship (35). When strong electrolytes are used at concentrations not exceeding 10'2 M, the conductivity follows the relationship 0' = Amo C 1-5 By combining equations 1-4 and 1-5, equation 1-6 can be obtained. 1=AHVfVE3(A,,,0 C)" 1-6 The following coefiicients were experimentally determined; v = 0.5, e as 0.5, n = 0.2-0.3 (27). Because of the small exponents, the changes of I with flow rate, electric field, and concentration of the electrolyte is small. 1 O 2. Dependence of Droplet Radius on Liquid Flow Rate Fernandez de la Mora (39) has proposed a relationship for the radius R of the ESI produced droplets from sprays R a (pl/127913 1-7 where p is the density, If, flow rate, and y is the surface tension of the solvent. This predicts that the radius of the droplets will increase with increasing flow rate. But in ESI/MS, smaller droplets are desirable. Therefore, smaller volume flow rates are suitable for better charging of the droplets. B. Shrinkage of Charged ESI Droplets The work by Gomez and Tang (39) and Davis and co-workers (40) has provided data for charged droplets at a flow rate of SuIJmin with the concentration not exceeding 10'3 M. The droplets are small and have a narrow distribution of sizes, so they can be considered monodispersed. The size distribution peaks at radius R0 as 1.5 um, and the droplets have a charge of Q0 as 10'14C, which corresponds to N = 50,000 singly charged ions (39). The Rayleigh equation which gives the condition in which the charge Q becomes just sufiicient to overcome the surface tension that holds the droplet together is QR2 = 641t280‘Y RR3 1-8 where so is the permittivity of vacuum, 7 is the surface tension, RR is Rayleigh limit radius, and QR is the Rayleigh limit charge. 11 Q? = 1.25 x10"10 RR3 1-9 The equation 1-9 gives the condition for methanol, whose y = 0.0226 N/mz; the numerical factor for Q in coulombs and R in meters. The initial charge Q0 10' 14C observed by Gomez and (Tang corresponds to only 50% of the Rayleigh limit charge QR for R0 = 1.5 pm. Gomez and Tang, who also studied the charge-to-volume ratio of larger ESI-produced droplets, found that Q0 comes closer to the Rayleigh limit for larger droplets. Droplets with the radius of micrometer or larger are known to maintain their charge; they do not emit gas-phase ions (40). The droplets shrink by evaporation of solvent molecules until they come closer to the Rayleigh limit where they become unstable and undergo fission into smaller droplets as shown in Figure 1-2. Recent work has shown that droplets with sizes in the l-llm range fission somewhat before, (at ~ 80% of the calculated value) the Rayleigh limit (39, 40). The droplet will shrink by solvent evaporation until the radius R meets that condition, and then fission will occur. The droplet does not split evenly into two smaller droplets of approximately equal mass and charge (40). Typically, the droplets are observed to vibrate alternatively between oblate and prolate shapes. These elastic vibrations stimulate disruptions in which the ”parent” droplet emits a tail of much smaller offspring droplets. This disruption pattern is similar to the disruption at the tip of the Taylor cone. The emitted stream of offspring droplets carries ofi‘ only about 2% of the mass of the parent droplet but 15% of the parent's charge. The radius of the offspring droplets, which are quite monodispersed, is roughly one-tenth of the radius of the parent (41). A simple calculation (42) shows that a 2% loss of mass leads to ~20 offspring droplets of this radius. The offspring droplets are not only much smaller than the parent but also have a much higher charge-to-mass ratio. 12 The time required for the parent droplet to reach the size R, that leads to first fission can be estimated (42) with use of expressions providing the rate of solvent evaporation fiom small droplets (43). When relatively volatile solvents are involved and the droplets are a few micrometers in diameter or smaller, the evaporation rate follows the surface evaporation limit law (43), which leads to a simple dependence (5, 40) of the droplet radius on time 1, WM ' 4ngT where v is the average molecular velocity of the solvent gas, p0 is the vapor pressure of R=Ro 1 1-10 the solvent at the temperature of the droplet, M molar mass of the solvent molecules, p is the density of the solvent, Rg is the gas constant, and T is the temperature of the droplet. The condensation coefficient, and its value is ~ 0.04 for both water and ethanol (42, 43). The following equation for methanol evaporation at 35°C was obtained assuming a condensation coefiicient, a = 0.04 for methanol and substituting the corresponding values. R=Ro-1.2xlo-3t 1-11 The time dependence process is given in Figure. 1-3. For a methanol droplet having initial diameter of 1.5 um and charge of 8 x 10'15C, at 35°C. The time required for the first fission is ~400 us. The change of R due to fission is very small. Subsequent fissions occur when the parent droplet again reaches 80% of the Rayleigh limit. These fissions require shorter times, ~ 60 us, which decrease fi'om fission to fission. C. Formation of Gas-Phase Ions Despite an explosive growth in their use over the past few years, especially in ESI, there has been very little identification and elucidation of component mechanisms in the 13 process by which solute species in a charged droplet are transformed into free gas-phase ions. Two difi‘erent mechanisms have been proposed to account for the formation of gas- phase ions fiom the small charged droplets: (a). charged residue mechanism, and (b). Iribame and Thomson ion evaporation theory. 1. The Charged Residue Mechanism The charged residue mechanism depends on the formation of extremely small droplets R z lnm, which contain only one ion. Solvent evaporation fiom such a droplet will lead to conversion of the droplet to a gas-phase ion. Such a mechanism was assumed by Dole (5), the first investigator of gas-phase ion production by ESI. A more detailed consideration and support of this mechanism was given by Rollgen (44). The charged-residue model for macromolecular gas-phase ion formation proposed by Dole and co-workers (5) over two decades ago is based on the combination of solvent evaporation and coulombic explosions as shown in Figure 1-2, resulting in a droplet size sufficiently small such that it contains only one molecular ion. 2. Iribame and Thomson Ion Evaporation Theory The Iribame and Thomson theory (45, 46) assumes ion evaporation fiom very small and highly charged droplets. Typically, the droplets from which ion emission becomes competitive with Rayleigh fission have R z 8nm and N as 70 elementary charges (45, 46). Under these conditions, the droplets do not undergo fission but emit gas-phase ions. As N decreases, emission can still be maintained as a result of a decrease of R by solvent evaporation. Thus, the Iribame mechanism does not require the production of very small droplets (R as lnm) that contain only one ion. Iribame emission can occur even when the droplet contains other solutes such as charge-paired electrolytes. Iribame and Thomson based their theory on a derived equation that provides detailed predictions for the rate of ion emission fi'om the charged droplets. In particular, it 14 g: 152250 51250 ’ 0.945 I 0 At = 462 ms ‘ 43560 43560 At = 74 ms 0.343 —> 0 + j 384 O O O 0 37026 37026 0.09 20 droplets 0.844 0.761 ——.> At = 70 ms + 326 00 O 0 37026 0-08 00.756 + 278 0.03 278 O O O O -—-> 0 007 Al ‘3 39 ms 236 0 0.003 + 2 o o o o 0.003 Figure 1-2. Schematic representation of lifetime of parent and offspring droplets Droplet at top left is a typical parent droplet created near the ES capillary tip at low flow rates. Evaporation of solvent at constant charge leads to uneven fission. The numbers beside the droplets give the radius R (um) and number of elementary charges N on the droplet; t corresponds to the time required for evaporative droplet shrinkage to size where fission occurs. Only the first three successive fissions of a parent droplet are shown. 15 predicts the dependence of the rates on the chemical properties of the ions. Observed differences in the gas-phase ion intensities I A and I B of ions A+ and B+ present at equal concentrations in the sprayed solutions were compared with predictions of the theory by Iribame and Thomson and subsequently by other workers (47-50) as well. In general, qualitative agreement between experiment and theory was obtained. The Iribame treatment is based on transition state theory. The rate constant It] for emission of ions from droplets is a: k, = %T exp(-AG IR?) 1-12 where k is the Boltzrnan constant, T is thetemperature of the droplet, and h is the Planck constant. The fi'ee energy of activation, AG: , was evaluated on the basis of the model shown in the Figure 1-3. The barrier in the Iribame transition state is due to the opposing electrostatic forces: the repulsion of the escaping ion by the other charges of the droplet and the attraction between the escaping ion and the droplet because of the polarizability of the solvent medium of the droplet. The attraction is larger at short distances between the ion and the droplet surface, but it falls off faster than the repulsion as the distance is increased. The equation for AG: was found (45, 46) to depend on four parameters. The first two are N, the number of charges on the droplet, and R, the radius of the droplet. The rate constant It] increases with N and decreases with R. The other two parameters express the specific properties of the ions involved; the solvation energy and the distance of the ion charges from the surface of the droplet. The escaping ion in Figure 1-3 is not the naked M+ but an ion-solvent molecule cluster M+(sol)m containing m solvent molecules. AG"F for the naked M+ is not the lowest, but when it leaves with several solvent . . iF . molecules, it has a lower actlvatlon energy. (for example, AG required to transfer Na+g 16 to the gas phase is ~ 98kcal/mol, whereas, Na+(HzO)7 requires only ~ 56kcal/mol) (42, 51). The strongly solvated ions have large transfer energies, and for these ions the Iribame equation predicts large activation barriers and hence low rate constants. 3. Emission of Gas-Phase Ions from the Taylor Cone Tip Very recently Siu and co-workers (52) proposed that the gas-phase ions are emitted from the Taylor cone tip. The authors present evidence by obtaining ESI/MS spectra in two different acids. Also, they compared mass spectra obtained in water and in a nonvolatile solvent, ethylene glycol. D. Dependence of Ion Intensity in ESI/MS on the Concentration of the Analytes in the Electrosprayed Solution When two electrolytes such as A+X' and BTY‘ are present in the solution, both A+ and B+ ions will be present among the excess positive ions that constitute the charges of the droplets. However, because of the very weak dependence of I on the total electrolyte concentration, addition of BY to AX will not materially increase the current, i.e., the total excess charge. On the other hand, B+ will compete with A+ among the excess charges on the droplets. This means that the amount of gas-phase ions A+ produced from the charged droplets will decrease as BY is added to the solution. Kebarle et al. (42, 53) introduced the relationship between I and the concentration of components present in the solution. ‘QMfl = LB u-figmfl+@mql 17 The authors investigated the ion intensity from several alkaloids, quaternary ammonium ions, and alkali metal ions in the presence of N114+ ions. A decrease in analyte (A+) ion current was observed as predicted by the equation, when B+ = NH4+ concentration is increased while the Al” concentration is kept constant. When analyte concentration increases while [A+]=[B+], the predicted pattern was observed above 10—5M. Experimentally determined ratios of coefficients for some compounds are given in Table 1-2. 1. Comparison of Coefiicients with Iribame Theory and Charged Residue Mechanism Comparisons of the experimentally determined coefficients with the Iribame rate constants and expected surface activities should be made only in the high concentration range, where depletion is absent. The range of coefficient values in Table 1-2 is not very large. This is a desired result for ESI/MS standpoint, and we can expect to detect with a fair sensitivity any analyte ion present in the solution (3 5). A general correlation between low solvation energies and high surface activities are expected. Because of the absence of suitable quantitative information of solvation energies and surface activities in the literature, it is difficult to establish the extent to which each factor contributes to the value of the experimentally determined coefficient. The charged residue mechanism, as originally stated (5, 44), did not provide criteria for selectivity on the basis of physicochemical properties of the ions. It is obvious that the surface activity is the factor that should lead to selectivity for the charged residue mechanism. Ions that are enriched on the surface will preferentially end up in a final droplet that contains a single ion. At present, it is not possible to state with certainty which theory, ion evaporation or charged residue mechanism, fits better with the available evidence. Fortunately, from a practical standpoint, qualitative predictions for high 18 (a) Initial state /a.%a Ion solvent cluster (b) Transition State Figure 1-3. Schematic representation of Initial state (a) and transition state (b), proposed by Iribame and Thomson. Evaporating ion leaves as a cluster M+(SL)m, where SL are solvent molecules, and r is the distance of ions from surface. l9 selectivity on the basis of low solvation energy or high surface activity will both be valid, because the two parameters are positively correlated (3 5). Table 1-2. Experimentally determined coefficients for the equation 1-14. All coeficients relative to sz+ = 1. These coeficients are valid only at concentrations above 10'5 M. Ion k Cs” 1 Li+ 1.6 Na+ 1.6 K+ 1.0 NH4+ 1.3 MorphineH+ 3 CodeineH+ 5 HeroinH+ 6 CocaineH+ 10 Ni2+(trypyridyl)2 5 (Butyl)4N+ 2 (Ethyl)4N+ 5 (n-propyl)4N+ 8 (n-pentyl)4N+ 14 n-C7H15NH3+ 10 n-C11H21NH3T 10 20 IV. Characteristics of ESI/MS Data In recent years, ESI mass spectrometry has captured the interest of several researchers owing to its high sensitivity, the simplicity of the interface, the extreme mildness of the process, and the possibility of high molecular weight determination using multiply charged ions. Because of the mildness of the ESI process, fragrnentations were not observed under normal ESI conditions for most of the compounds. In the ESI spectrum of small molecules, only peaks representing the original molecule were observed (1, 2, 10, 12). The molecules that can form ionic adducts or that have pro-formed charges in solution are observed in the ESI mass spectra as singly charged ions or multiply charged ions. Figure 1-4 shows the ESI mass spectra of 1,10 phenanthroline and (CH3)4NI (12). The most important finding in recent years is the multiple charging in the electrospray ionization of proteins and peptides. This gives the ability to analyze biomolecules directly fiom aqueous phase. The ESI mass spectra appear as an envelope of peaks representing the parent molecule with different charge states. The adduct ion is usually H‘l’, Na+ (10, 12), Ca2+, or Mg2+ (54) ions . Figure 1-5 shows the ESI mass spectrum of myoglobin (M.W. = 16,951). The peaks in the ESI mass spectrum corresponds to the myoglobin molecule, but with the addition of difierent number of H+ ions. A. Types of Analytes Required for ESI/MS On the basis of previous work, it has become clear that ESI is a method by which ions present in solution are transferred to the gas phase (55-59). The types of analytes that can be analyzed by ESI/MS are those can form ionic species in solution. ESI transfers ions from solution to the gas phase for a variety of ions: the simple singly charged electrolytes such as Na+ and Cl'; group 11 ions such as Ca2+, Sr2+, and Ba2+, as well as doubly and triply charged transition metal and lanthanide ions and complexes thereof; protonated basic and deprotonated acidic compounds, compounds having pre-formed 21 charges, bioorganic ions such as multiply protonated peptides and proteins; and deprotonated negatively charged nucleic acids (3 5). m1" “' 9% 8‘ '63 5 “waist” ‘65 H o .E Ma'H+ .9. o l I I M W m/z Figure 1.4. ESI/MS mass spectrum of 1, 10 phenanthroline (2.7 x 10'5M) and (CH3)4NI (6.5 x 10"8 M) in methanol/water (SO/50% v/v). Adapted from ref. 12. 22 1131 100- 9r3 999 so~ 893 60- 12,3 4-12 1414 8Q1 1395 ,0- 7721 +-ll 1542 H 4-10 711 1696 20- W 707 ‘1 TA LIMA I ' l I ' l "—I ' 1 600 800 1000 1200 1400 1600 1800 2000 Figure 1-5. The electrospray mass spectrum of myoglobin (M.W. = 16,951). The data were collected on a TSQ 700 mass spectrometer. 10 pmol were consumed to obtain the spectrum. 23 B. Interpretation of Multiple Charged Envelope (n+1) (n+2) n+ Intensity (n+3) m1 m2 m3 m4 Figure 1-6. Hypothetical mass spectrum containing multiply charged ions. where m1, m2, m3, and m4 are m/z values corresponds to peaks containing the number of charges (n+3), (n+2), (n+1), and 11 respectively. Assume the molecular weight of the compound is M, and the number of charges on the other ions as labeled. The adduct ion is x+ and usually it can be 11+, Na+, 1014+, Ca2+ ong2+. m3 = M+(n+1)X+ 144 n+1 + m4 = fl 1-15 11 by solving the above equations, charge state 11 and molecular weight M can be obtained. 24 n = ...—{ELL}— 1'16 m4-m3 M =n (m4-1) 1-17 C. Why Derivatives ? The ESI technique transfers ions fi'om solution to the gas phase. The molecules without pro-formed charges, acidic or basic filnctional groups cannot be easily detected under ESI/MS conditions because they do not exist as ionic species in solution. For a molecule without a ESI active group, one of the above mentioned functional groups must be introduced to obtain a reasonably high response (1, 54). In chapter 5, the enhancement of response of neutral steroid molecules after modification with a quaternary ammonium group will be discussed in detail. D. Oxidation of Analytes under the High Electric Field In the electrospray process, highly charged droplets are formed under a high electric field. Normally, in the positive ion mode, a high voltage of positive value is applied to the ESI needle tip carrying the sample solution. Proof for the occurrence of electrochemical oxidation at the metal capillary was provided by Blades et al. (48). When a Zn capillary tip was used, the release of Zn2+ to the solution could be detected with ESI/MS. Similar results were observed with stainless steel capillaries that releases F «9+ to the solution. The electrospray-induced oxidation due to the high electric field has been observed for peptides containing methionyl, tryptophanyl, or tyrosyl residues (57). The oxidation process is dependent on the field strength and the flow rate of the solution passing through the capillary tube. 25 V. Mass Analyzers for Electrospray Mass Spectrometry The intention of this part is to introduce the various types of mass analyzers that have been coupled with an ESI source. These mass analyzers have their own unique capabilities and advantages over one another. A. Quadrupole Mass Analyzers The most commonly used mass analyzer for ESI/MS is the quadrupole mass filter. The quadrupole mass analyzers can tolerate a little higher pressures than the other types of mass analyzers. They have relatively higher scan rates. The mass range of new quadrupole mass spectrometers have been extended to 3000 Daltons. The acceleration voltage of a quadrupole mass spectrometer is about 5-15V. The resolving power of most commercial quadrupole mass spectrometers is limited to unit mass resolution. The details of quadrupole theory will be discussed in chapter 2. B. Magnetic Sector Instruments There are benefits in interfacing an electrospray ion source to a high-performance magnetic sector mass spectrometer. The major benefits are better mass accuracy, higher mass resolution, higher mass (mass/charge) range, and the possibility of high energy collisional activation of multiply charged ions (58). Better mass accuracy, for example, is useful in determining post-translational modifications as well as point mutations in proteins. Higher resolution aids in mixture analysis and extends the mass limit of biopolymers that can be mass analyzed by providing better separation of the multiple charged ions. A higher mass-range instrument (4000 Daltons) fitted with an electrospray ion source permits observation of high molecular weight materials that do not support sufiicient charges to fall into the mass range of a quadrupole instrument (59). 26 Aleksandrov et al. (10) interfaced electrospray ionization to a magnetic sector mass spectrometer and reported obtaining singly charged ions of small molecules using 10-100 nmol of material. Fragment ions were obtained by increasing the potential difference between the aperture plates separating the atmospheric-pressure region fi'om the vacuum region of the source-housing chamber. Allen and Lewis reported interfacing an electrospray ion source to a single-focusing magnetic-sector mass spectrometer (60). They were able to observe doubly and singly charged ions for grarnicidin (M.W.=1141 Da) and bradykinin (M.W.=1060 Da). Early results with ESI on a magnetic instrument produced spectra of peptides but the sensitivity was poor relative to that reported for quadrupole instruments, and in addition, highly charged ions fi'om proteins were not observed (58). Meng et al. (61) improved the sensitivity and resolution by reducing the pressure in the kilovolt acceleration region. They accomplished this by incorporating two stages of rotary pumvins- ESI on the magnetic sector mass spectrometers has been used to determine the accurate masses of proteins and peptides (62-64). Sequence information has been obtained by in-source fragmentation of electrospray-generated peptide ions. The complete sequence of several peptides has been determined fiom their fragment ions consuming only low picomole amounts of material (64, 65). C. Quadrupole Ion Trap Mass Spectrometry Use of the quadrupole ion trap mass spectrometer has undergone remarkable growth in the past decade due largely to new developments in ion trap technology and the subsequent commercial introduction of analytical instruments based on the three- dirnensional quadrupole (66). The development was catalyzed by the introduction of the mass-selective instability mode of operation (67) and the use of about 1 mtorr of He as a 27 bath gas to improve sensitivity and resolution (68). The development includes, for example, techniques for mass spectrometry/mass spectrometry (MS/MS) (69), multiple stages of mass spectrometry (MSn, n>2) (70), mass extension (71). Dissociation eficiency was reported to be greater than 50% (72). Van Berkel et al. (73) reported the coupling of an ESI source with an ion trap mass spectrometer. The authors described the ESI/MS analysis of various classes of molecules including, direct red 81, bradykinin, melittin, cytochrome c, myoglobin, and bovine albumin. The ability to obtain more information was demonstrated by employing M83 and MS4 dissociation experiments for multiply charged peptide ions. D. Fourier Transform Mass Spectrometry The potential of FT ion cyclotron resonance mass spectrometry for precise mass measurement has long been recognized (74). Fourier transform mass spectrometry (FTMS) has a capability to achieve very high resolving power. The pioneering work by Mclarfi'erty and co-workers (75) led to the development of ESI on FTMS. Recently they reported the resolving power of 5 x 105 for a 29-kDa protein (for the ion having 21+ charges) with less than l-ppm mass measuring error (75). Winger et al. (76) reported the resolving power of 700,000 for bovine insulin (4+ charge state). E. Time of Flight Mass Spectrometry The time of flight mass analyzer, is an inherently pulsed techniqUe requiring spatially and temporally well-defined ion packets with which to conduct mass analyses. Due to the high fi'equency of charged droplet formation and the droplet spatial randomization during evaporation and ion production, the ion current entering the vacuum through the nozzle or capillary from atmospheric pressure is essentially a continuous current with no discernible periodic oscillation (77, 78). Attempts to couple these two 28 techniques necessarily impose a penalty in terms of sensitivity because of reduced duty cycle. Boyle et al. (78) introduced an electrospray ion storage time-of-flight mass spectrometer. Recently, Lubman and co-workers (79) reported on a time of flight mass spectrometer coupled with an ion trap capable of storing and sampling ESI produced ions continuously. This technique is capable of obtaining the typical sensitivity and speed of time of flight mass spectrometry in coupling with a liquid chromatographic system. VI. Sample Introduction Techniques A. Coupling with Liquid Chromatographic Systems The ESI/MS is a liquid ionization technique and it allows to introduce the sample in solution. The most common way of introducing a sample is through a flow injection or by infusion using a infilsion pump. High-resolution separation prior to mass spectral analysis of biological materials is highly desirable since essentially all biochemical systems of interest are comprised of mixtures of varying complexity. Even proteins in a highly ”purified” state are often mixtures due to naturally occurring microheterogeneity, or are converted to mixtures prior to analysis by chemical or enzymatic degradation procedures. Since sample sizes are generally limited, there are strong incentives to conduct biochemical research on the smallest scale possible. It is not surprising therefore that combined separations-ESI-MS analysis is of broad interest and that greater sensitivity is almost always desired. Liquid chromatography is used to separate a wide variety of biologically important compounds, and has been interfaced to the ESI mass spectrometer. For unassisted ESI, flow rates of l-IOuI/min are the best for stable operation, depending somewhat on the solution composition (17, 80). Packed capillary LC has also been coupled to mass spectrometry using both unassisted (81) and assisted ESI (82). Capillary diameters of typically 250nm, and as low as 50 pm with the flow rates of less than a few llL/min have 29 been used. Now it is possible to introduce flow rates on the order of 300 uIJmin that are compatible with 2.1 mm diameter columns into commercially available ESI sources (83). Reversed phase chromatographic columns as well as normal phase columns have been used prior to the ESI/MS analysis. Recently, perfirsion chromatography, a rapid separation technique, has been combined with ESI/MS (84, 85). Separation times on the order of minutes were reported. B. Coupling with Capillary Electrophoresis Capillary Electrophoresis (CE) is attracting attention as a method for rapid high- resolution separation of very small sample volumes ‘ of complex mixtures. In combining with the inherent sensitivity and selectivity of mass spectrometry, CE-MS becomes a potentially powerful bioanalytical technique The combination of CE with a quadrupole mass spectrometer based on ESI has been described. The coupling of CE with an ESI source is not straightforward as that of HPLC. The main limitations are the nanoscale flow rates and the terminating potential. To overcome these limitations, Smith et al. (86) and Henion et al. (87) developed CE-ESI probes. These authors used sheath liquid flow in order to introduce a flow of solvent to flush the end of the CE capillary continuously. This removes the migrating solutes fiom the CE capillary and facilitates the electrospray process. The sheath liquid electrode allows the ESI interface to be operated for almost any high ionic strength buffers that could not otherwise be electrosprayed. Using CE-ESI/MS, various classes of molecules have been analyzed with good sensitivity and high resolution, including quaternary phosphonium salts, amines (86), peptides and proteins (88). Small peptides are easily amenable to CE-ESI/MS analysis with low femtomole or better detection limits (89). ‘ 30 C. Coupling with Ion Chromatography The ion chromatography (IC) system can be interfaced with ESI/MS via a postsuppressor (90). 1C is widely used to separate ionic component in a mixture. Coupling of IC with ESI/MS gives several advantages including high sensitivity. But the ionic strength of bufi‘ers used in IC prohibits the formation of ions in ESI. Therefore, an ion suppresser was introduced alter the column but before the ESI needle to remove excess ions in the bufl‘er. The micromembrane suppresser selectively removes over 99.9% of the ion-pair agents required for ion chromatography from the eluent. The resulting solution consists of analyte, organic modifier, and water, a solution which is compatible with ESI. This work describes the separation and detection of quaternary ammonium drugs and tetraalkylammonium compounds of industrial importance. A limit of detection of 40 pg injected on-column for tetraalkylammonium cations was reported. The separation, detection, and identification of some alkyl sulfates and sulfonates were also shown with operation in the negative ion mode (90). VII. Applications of ESI-MS Since the introduction of ESI/MS in 1988 as a tool for the ionization of large biomolecules without significant decomposition, the practice of ESI/MS has grown impressively. Several review articles appeared recently describing ESI/MS use in molecular weight determination of large biomolecules (89, 91). ESI/MS provides accurate molecular weights of biopolymers that cannot be obtained by conventional methods such as fast atom bombardment (FAB) and thermospray mass spectrometry. Multiple charging enables high molecular weight determinations. Molecular weights greater than 197 kDa have been determined successfully by ESI-MS (92). No attempt will be made here to exhaustively catalog this work. Instead, some major classes of 31 compounds which have been addressed by ESI/MS will be t0uched upon, with the inclusion of some appropriate references. A ESI/MS of Small Molecules Fenn et al. (12, 16) described their early findings of ESI mass spectrometry using small molecules. Fenn and co-workers tried a wide range of solute species including alcohols, acids, esters, amines, catechol amines, cholines, nucleotides, amino acids, and small peptides (91) to evaluate gas phase ion formation fiom liquid phase and detection by ESI/MS. In every case, the spectra were comprised of peaks for the solute species itself when it was an ion or for its adducts with anions or cations when it was not an ion. They demonstrated the ability to obtain the mass spectra from nonvolatile and thermally labile compounds without significant decomposition or fi'agmentation. Bruins et al. (93) analyzed monosulfonated azo dyes, steroid glucuronides, and steroid sulfates in the negative mode. A A detection limit of 10 pg for diethylstilbestrol was achieved with nebulizer assisted ESI/MS. In recent years, ESI/MS has become a very popular technique in small polar molecule analysis and still is growing its popularity. B. Multiple Charging and High Molecular Weight Determination 1. Large Polypeptides and Proteins A feature of ESI mass spectra of most proteins is that the average charge state increases in an approximately linear fashion with molecular weight, extending the mass range of the MS experiment by the addition of compensating charge. The net number of charged sites in solution appears to be the principal factor affecting the maximum extent of charging observed in ESI mass spectra. For most proteins (prepared in aqueous solution, pH< 4) an approximate linear correlation has been observed between the maximum charge state detected and the number of basic anrino acid residues (e.g., arginine, lysine, histidine) 3.2 plus the NH2 terminus (89). Under typical acidic conditions most, if not all, basic residues will be protonated in solution. The acidic residues can be deprotonated in solution at pH greater than ~ 5, and such conditions would be expected to lower the observed positive ion ESI charge state distribution. Smith et al. (89) compiled a list of proteins successfully analyzed by ESI/MS, along with the maximum number of charges observed and the number of positive charges anticipated under acidic conditions. An example of multiple charging resulting fi'om electrospray ionization is shown in Figure 1-5. An envelope of multiple charged ions was observed for the myoglobin. g Negative ion ESI has been demonstrated for a variety of small molecules with acidic functionalities such as carboxylic, phosphoric, and sulfonic acid groups (89). However, very few negative ion ESI mass spectra of polypeptides have been reported (27). One early example was a negative ESI mass spectrum of aqueous solution of bovine A-chain of insulin (M.W. = 2532) (89) in which 4 cysteine residues are oxidized. Multiple charging to the 5- (deprotonated) charge was observed. Most polypeptides in neutral and acidic solutions do not produce multiple charged ions in the negative mode, because acidic residues such as glutarnic acid and aspartic acid typically have pKa's around 5 or higher in proteins. 2. Other High Molecular Weight Compounds Molecules having preformed charges, basic sites or acidic sites have been directly analyzed by ESI mass spectrometry. Other classes of polymers that have no such groups have been analyzed after oxidation, reduction or transformation to metal cation complexes. Fenn et al. (94) reported the formation of ions in an ESI source from solutions of poly(ethylene glycol) (PEG) samples with average molecular weights ranging fiom 200 to 33 17,500. Mass analysis of these ions provided evidence that up to at least 23 sodium cations could be deposited on the larger oligomers. ‘ The feasibility of ESI/MS in polymer analysis has been evaluated by Kallos'et al. (95). using a polyarnidoamine Starburst polymer. The repeat unit of the polymer is a polyamidoamine. The molecular weight information was obtained for a mixture without prior separation. The fullerenes C60 and C70 were investigated by using ESI/MS (96). C50', C5002', C7o', and C7002“ ions were detected by reducing C60 and C70 using NaK amalgam in benzene + dimethoxyethane solvent. Wilson et al. (54) derivatized C50 and C70 with a reagent to obtain a signal on ESI/MS. .C. Studies. on Higher Order Structures of Proteins Information on higher-order structure (secondary, tertiary and quaternary) is of fundamental biological importance. The native state of proteins is typically folded into well-defined three-dimensional structures by relatively weak intramolecular forces (e. g., hydrogen bonding). Protein function in biological systems generally involves interactions of specific structural conformations essential for activity (97). In their native state, globular proteins are tightly folded, compact structures. They can be denatured and caused to unfold by subjecting them to high temperatures, extremes of pH, detergents, and solutions containing high concentrations of compounds such as urea, guanidium chloride, and organic solvents (97). ESI/MS has been used to study the higher order structural changes recently. Chowdhury et al. (98) investigated the structural conformational changes of bovine cytochrome c at various pH values (pH=2.6-5 .2). These authors found three different charge state distribution patterns at difi‘erent pH values. These results agreed with the earlier reported conformations. 34 L00 et al. found that the addition of methanol denaturant in excess of 40% v/v was required to eliminate the lower charge state (globular or native state) of Ubiquitin peptide (M.W.=8,564.8). The native globular form dominates (7+ and 8+) for Ubiquitin in a completely aqueous solution with a small contribution from the denatured helix structure (10+ to 13+), ascribed to mixing of the analyte solution with the acetonitrile sheath (99). . Le Blanc et al. (100) studied the efi‘ects of heat on the electrospray spectra of a number of globular proteins, observing a dramatic increase with increasing temperature of the charge states of ions produced from equine cytochrome c and chicken egg lysozyrne as well as a strong increase of the ion intensity fi'om cytochrome c. Hydrogen exchange with deuterium followed by ESI/MS appears to be a sensitive and useful method for probing secondary structure in peptides and proteins. Katta et al. (101) demonstrated the possibility of using hydrogen exchange ESI/MS for probing the conformational changes of proteins in solution. The ESI/MS measurements provide information about the rate of exchange and the extent of exchange (102, 103). Electrospray mass spectrometry is a very mild technique for producing ions directly from solution at atmospheric pressure. ESI/MS can be used to observe non- covalently bound adducts. Henion et al. (104, 105) described non-covalent receptor- ligand complexes using ion-spray MS. In several reports, non-covalent enzyme-substrate and enzyme-product complexes have been observed. 10. ll. 12. l3. 14. References Aebersold, R.; Bures, E. J .; Goghari, M. H.; Shusan, B.; Covey, T. C. Protien Science, 1992, I, 494 Conboy, J. J .; Henion, J. D.; Martin, M. W.; Zweigenbaum, J. A. Anal. Chem, 1990, 62, 800 Bose, G. M. Researches sur 1e cause et sur la veritable theorie de I 'electricite, Wittenberg, 1745 Zeleny, J. J. Phys. Rev. 1917, 10, 1-6 Dole, M.; Mack, L. L.; Hines, R. L.; Mobley, R. C.; Ferguson, L. D.; Alice, M. B. J. Chem. Phys. 1968, 49, 2240 Mack, L. L.; Kralik, P.; Rheude, A; Dole, M. J. J. Chem. Phys. 1970, 52, 4977 Chapman, S. Phys. Rev. 1937, 52, 184 Clegg, G. A; Dole, M. Biopolmers 1971, 10, 821 Gieniec, J.; Mack, L. L.; Nakarnae, K.; Gupta, C.; Kumar, V.; Dole, M. Biomed. Mass. Spectrom. 1984, 11, 259 Aleksandrov, M. L.; Gall, L. N.; Krasnov, V. N.; Nikolaev, V. I.; Pavlenko, V. A; Shkurov, V. A. Dokl. Akad Nauk. SSSR 1984, 277, 379 Aleksandrov, M. L.; Gall, L. N.; Krasnov, V. N.; Nikolaev, V. 1.; Pavlenko, V. A; Shkurov, V. A; Bararn, G. 1.; Gracher, M. A; Knorre, V. D.; Kusner, Y. S. Bioorg. Kim. 1984, 10, 710 Yarnashita, M.; Fenn, J. B. J. Phys. Chem. 1984, 88, 4671 Aleksandrov, M. L.; Besuklandnikov, P. V.; Grachev, M. A; Elyakova, L. A; Zyyaginsteva, T. N.; Kondratsev, V. M.; Kusner, Yu. 8.; Mirgorodskaya, O. A; Fridlyansky, G. V. Bioorg. Kim. 1986, 12, 1689 Aleksandrov, M. L.; Barama, G. I.; Gall, L. N.; Krasnov, N. V.; Kusner, Yu. S.; Mirgorodskaya, O. A; Nikolaev, V. 1.; Shkurov, V. A; Bioorg. Kim 1985, 11, 700 35 15. 16. 17. 18. 19. 20. 21. 22. 23. 24. 25. 26. 27. 28. 29. 30. 31. 32. 36 Mirgorodskaya, O. A; Nikolaev, V. 1.; Shkurov, V. A; Bioorg. Kim 1985, 11, 705 Whitehouse, C. M.; Dreyer, R. N.; Yamashita, M.; Fenn, J. B. Anal. Chem, 1985, 57, 675 Wong, S. F.; Meng, C. K.; Fenn. J. B. J. Phys. Chem. 1988, 92, 546 Meng. C. K.; Mann. M.; Fenn. J. B. Z. Phys. D. 1988, 10, 361 L00, J. A; Udseth, H. R.; Smith, R D.; Blamed Environ. Mass Spectrom. 1988, I 7, 41 1 L00, J. A; Udseth, H. R.; Smith, R. D.; Rapid Commun. Mass Spectrom 1988, 2, 207 Covey, T. R.; Boner, R. F.; Shusan, B. I.; Henion, J. D. Rapid Commun. Mass Spectrom 1988, 2, 249 Karas, M.; Killenkamp, F. Anal. Chem, 1988, 60, 2299 Smith, R. D.; Loo, J. A; Loo, R. R. 0.; Busman, M; Udseth, H. R Mass Spec. Rev. 1991, 10, 359 Meng, C-K.; McEwen, C. N.; Larsen, B. S. Rapid Commun. Mass Spectrom. 1990, 4, 147 Beu, S. C.; Senko, M. W.; Quinn, J. P.; Wampler III, F. M.; Mclarfi‘erty, F. M. J. Am. Soc. Mass Spetrom. 1993, 4, 557-565 Loeb, L. B.; Kip, A. F.; Hudson, G. G.; Bennet, W. H. Phys. Rev. 1941, 60, 714 Pfeifer, R. J.; Hendricks, C. D. AIAA J. 1968, 6, 496 Taylor, G. 1. Proc. R. Soc. London A 1964, A280, 383 Ikonomou, M. G.; Blades, A. T.; Kebarle, P. Anal. Chem. 1990, 62, 957 Hayati, 1.; Bailey, A. I.; Tadros, T. F. J. Colloid Interface Sci. 1987, 117, 205 and 222 Taflin, D. C.; Ward, T. L.; Davis, E. J. Langmuir 1989, 5, 376 Smith, D. P. H. IEEE Trans. Ind Appl. 1986, 1A-22, 527 33. 34. 35. 36. 37. 38. 39. 40. 41. 42. 43. 45. 46. 47. 48. 49. 37 Ikonomou, M. G.; Blades, A. T.; Kebarle, P. J. Am. Soc. Mass Spectrom. 1991, 2, 497 Wamper, F. W.; Blades, A. T.; Kebarle, P. J. Am. Soc. Mass Spectrom. 1993, 4, 289 ' Kebarle, P.; Tang, L. Anal. Chem. 1993, 65, 972A Lee, E. D.; Henion, J. D. Rapid Commun. Mass Spectrom. 1992, 6, 727 Mirza, U. A; Cohen, S. L.; Chait, B. T. Anal. Chem. 1993, 65, 1-6 Buchanan, M. V. Proceedings 41st ASMS conference on Mass Spectrometry and Allied Topics, San Francisco, CA, May 31-June 4, 1993, 764 Gomez, A; Tang, K. Proc. 5th International Conference on Liquid Atomization and Spray Systems I (CLASS-91); Smerjians, H. G., Ed: NIST: Gaithersburg, MD; Special Publication 813 Davis, E. J. ISA Trans. 1987, 26, 1 Willoughby, R.; Sheehan, E.; Jarrell, A; Masecic, T.; Pedder, R.; Penn, S. proceedings 41 st ASMS conference on Mass Spectrometry and Allied Topics, San Francisco, CA, May 31-June 4, 1993 Tang, L.; Kebarle, P. Anal. Chem. 1993, 65, 3654-3668 Davies, C. N. In Fundamentals of Aerosol Science; Shaw, D. T., Ed; John Wiley and Sons: New York, 1978, 154 Rollgen, F. W.; Bramer-Wegner, E.; Buttering, L. J. Phys. Colloq. 1984, 45, supplement 12, C9-297 Iribame, J. V.; Thomson, B. A. J. Chem. Phys. 1976, 64, 2287 Thomson, B. A; Iribame, J. V. J. Chem. Phys. 1979, 71, 4451 Fenn, J. B.; Mann, M.; Meng, C. K.; Wong, S. F.; Whitehouse, C. M. Science 1989, 246, 64 Ikonomou, M. 6.; Blades, A. T.; Kebarle, P. Anal. Chem. 1991, 63, 1989 Sakari, M.; Yergey, A. L.; Siu, K. W. M., LeBlanc, J. C. Y.; Guevremont, R.; Berrnan, R. 8. Anal. Sci. 1991, 7, 199 50. 51. 52. 53. 54. 55. 56. 57. 58. 59. 60. 61. 62. 63. 65. 67. 38 Rafaelli, A; Bruins, A. P. Rapid Common. Mass Spectrom. 1991, 5, 269 Hiraoka, K. Rapid Common. Mass Spectrom. 1992, 6, 463 Siu, K. W. M.; Guevremont, R.; LeBlanc, J. C. Y.; O'Brien, R. T.; Berrnan, S. S. Org. Mass Spectrom, 1993, 28, 579 Tang, L.; Kebarle, P. Anal. Chem. 1991, 63, 2709 Wilson, S. R.; Wu, Y. J. Am. Soc. Mass Spectrom. 1993, 4, 596-603 Pfeifer, R. J .; Hendricks, C. D. AIAA J. 1968, 6, 496 Blades, A. T.; Ikonomou, M. G.; Kebarle, P. Anal. Chem. 1991, 63, 2109 Morand, K.; Talbo, G.; Mann, M. Rapid Common. Mass Spectrom. 1993, 7, 738- 1411111, M. H.; Lewis, I. A. S. Rapid Common. Mass Spectrom. 1989, 3, 255 Meng, C.-K.; McEwen, C. N.; Larsen, B. S. Rapid Common. Mass Spectrom. 1990, 4, 369-372 . Allen, M. H.; Lewis, I. A. S. Rapid Common. Mass Spectrom. 1989, 3, 255 Meng, C.-K.; McEwen, C. N.; Larsen, B. S. Rapid Common. Mass Spectrom. 1990, 4, 147-150 Wada, Y.; Tarnura, J.; Musselrnan, B. D.; Kassel, D. B.; Sakurai, T.; Matsuo, T. Rapid Common. Mass Spectrom. 1992, 6, 9-13 McEwen, C. N.; Larsen, B. S. Rapid Common. Mass Spectrom. 1992, 6, 173-178 Chapman, J. R.; Gallagher, R. T.; Barton, E. C.; Curtis, J. M.; Derrick, P. J. Org. Mass Spectrom. 1992, 27, 195-203 Meng, C. K.; McEwen, C. N.; Larsen, B. S. Rapid Common. Mass Spectrom. 1990, 4, 15-1-155 Nourse, B. D.; Coks, R. G. Anal. Chim. Acta 1990, 228, 1 Stafi‘ord, G. C.; Kelley, P. E.; Syka, J. E. P.; Reynolds, W. E. .1111. J. Mass Spectrom. Ion Processes, 1984, 60, 85 68. 69. 70. 71. 72. 73. 74. 75. 76. 77. 78. 79. 80. 81. 82. 83. 39 Kelley, P. E.; Stafi‘ord, G. C.; Fies, W. J.; Syka, J. E. P.; McIver,Hunter, R L.; Todd, J. F. J.; Bexon, J. J. ASMS San Antonio, TX, 1984, pp 505 Louris, J. N.; Cooks, R. G.; Syka, J. E. R; Kelly, P. E.; Stafi‘ord Jr, G. C. Todd, J. F. J. Anal Chem, 1987, 59, 1677 Louris, J. N.; Brodbelt-Lustig, J. S.; Cooks, R G.; Glish, G. L.; VanBerkel, G. L.; McLuckey, S. A. Int. J. Mass Spectrom. Ion Proc. 1990, 96, 117 Kraiser, R. E.; Louris, J. N.; Amy, J. W.; Cooks, R. G. Rapid Common. Mass Spectrom. 1989, 3, 225 Cox, K. A; Williams, J. D.; Cooks, R. 6.; Kaiser Jr, R E. Biol. Mass Spectrom. 1992, 21, 226-241 Van Berkel, G. J .; Glish, G. L.; McLuckey, S. A. Anal. Chem, 1990, 62, 1284 Wilkins, C. L.; Gross, M. L. Anal. Chem. 1981, 53, 1661A Ben, S. C.; Senko, M. W.; Quinn, J. P.; Wampler III, F. M.; Mclarfi‘erty, F. M. J. Am. Soc. Mass Spetrom. 1993, 4, 557-565 Winger, B. E.; Hofstadler, S. A; Bruce, J. E.; Udseth, H. R.; Smith, R. D. J. Am. Soc. Mass Spetrom. 1993, 4, 566-577 Charbonnier, F.; Rolando, C.; Saru, F. Rapid Common. Mass Spectrom. 1993, 7, 707-710 Boyle, J. G.; Whitehouse, C. M.; Fenn, J. B. Rapid Common. Mass Spectrom. 1991, 5, 400-405 Chien, B. M.; Micheal, S. M.; Lubman, D. M. Int. J. Mass Spectrom. Ion Processes 1994, 131, 149 Smith, R D.; Loo, J. A; Edmonds, C. G.; Barinaga, C. J.; Udseth, H. R Anal. Chem. 1990, 62, 882 Lane, 8. J.; Blinded, K. A; Taylor, N. L.; Watkins, P. J. F., Harrison, M. E. Rapid Common. Mass Spectrom. 1993, 7, 953 Carr, S. A; Hemling, M. E.; Bean, M. F.; Roberts, G. D. Anal. Chem. 1991, 63, 2802 TSQ 7000 API manual, Finnigan Mat, San Jose, CA, 1993 84. 85. 86. 87. 88. 89. 91. 92. 93. 94. 95. 96. 97. 98. 100. 40 Kessal, D. B.; Shusan, B.; Sakuma, T.; Salzmann, J-P. Anal. Chem. 1994, 66, 236 Lin, H-U.; Voyksner, R. D. Rapid Common. Mass Spectrom. 1994, 8, 333 Smith, R. D.; Barinaga, C. J.; Udseth, H. R Anal. Chem. 1988, 60, 1948 Lee, E. D.; Muck, W.; Henion, J. D.; Covey, T. R. J. Chromtogr. 1988, 458, 313 Smith, R. D.;Udseth, H. R; Barinaga, C. J.; Edmonds, J. Chromatogr. 1991, 559, 197-208 Smith, R. D.; Loo, J. A; Loo, R R O; Busman, M.; Udseth, H. R. Mass Spectrometry Reviews, 1991, 10, 359-451 Conboy, J. J.; Henion, J. D.; Martin, M. W.; Zweigenbaunr, J. A. Anal. Chem. 1990, 62, 800-807 Fenn, J. B. ,Mann, M.; Meng, C. K.; Wong, S. F.; Whitehouse, C. M. Mass Spectrometry Reviews, 1990, 9, 37- 70 Feng, R.; Bouthiller, F.; Konishi, Y.; Cygler, M. Proceedings 39th ASMS Conference of Mass Spectrometry and Allied Topics, Nashville, TN, May 29-June 3, 1991 Bruins, A. P.; Covey, '1". 11.; Henion, J. D. Anal. Chem. 1937, 59, 2642 Wong, S. F.; Meng, C. K.; Fenn, J. B. J. Phys. Chem. 1988, 92, 546-550 Kallos, G. J.; Tomalia, D. A; Hedstrand, D. M.; Lewis, S.; Zhou, J. Rapid Common. Mass Spectrom. 1991, 5, 383-386 Hiraoka, K.; Kudaka, I.; Fujimaki, S.; Shinohara, H. Rapid Common. Mass Spectrom. 1992, 6, 254-256 Creighton, T. E.; Proteins, structures and Molecular Principles, Freeman, W. H. and Co., New York 1984 Katta, V.; Chait, B. T. J. Am. Chem. Soc. 1991, 113, 8534-8535 Loo, J. A; Loo, R. R. O.; Udseth, H. R.; Edmonds, C. G.; Smith, R. D. Rapid Commun. Mass Spectrom. 1991, 5, 101 Le Blane, J. C. Y. ,Beuchemin, D.; Siu, K. W.; Guevremont, R. ,Berrnan, S. S. Org.MassSpectrom. 1991,26, 831 101. 102. 103. 104. 105. 41 Katta, V.; Chait, B. T. Rapid Common. Mass Spectrom. 1991, 5, 214-217 Stevenson, C. L.; Andregg, R. J.; Borchardt, R. T. J. Am. Soc. Mass Spectrom. l993,4,646 Smith, J. B.; Thevenon—Emeric, G.; Smith, D. L.; Green, B. Anal. Biochem, 1993, 193, 118 Ganem, 3.; Li, Y-T.; Henion, J. D. J. Am. Chem. Soc. 1991, 113, 6294-6296 Ganem, 3.; Li, Y-T.; Henion, J. D. J. Am. Chem. Soc. 1991, 113, 7818-7819 Chapter 2 DEVELOPMENT OF INSTRUMENTATION FOR ESI/MS I. Introduction Electrospray mass spectrometry (ESI/MS) has become a very powerful technique in modern mass spectrometry in a short period of time. The analysis of thermally labile and polar molecules using mass spectrometry became routine afier interfacing ESI/MS with chromatographic systems. ESI/MS provides the ability to obtain accurate molecular weight information of polypeptides and other polymer materials which are refractory to conventional ionization techniques by forming a multiple charged envelope of ions with high sensitivity. The sensitivity for polypeptides (1) and small molecules having pre- formed charges (2) has been demonstrated at the femtomole level or better. Fenn et al. (3, 4) described their first electrospray mass spectrometer with impressive results for the analysis of non-volatile molecules in 1984. They attracted the attention of mass spectrometrists for their re-invention of ESI/MS. The essential features of the Yamashita and Penn source are a hypodermic needle, a cylindrical electrode around the needle, a nozzle, and a skimmer. Liquid sample was introduced through a stainless steel hypodermic needle and maintained at 3-10 kV relative to ground. The distance from the end of the needle to an end plate containing the nozzle was in the range of 20-30mm. Surrounding this region was a cylindrical electrode 30 mm in diameter and maintained at 500-600V relative to ground. Dry nitrogen typically at 800 torr was passed through the nozzle region at a velocity of several cm/s in order to sweep out solvent vapor from the evaporating droplets. The region downstream of the nozzle was maintained at a few mtorr or less by an oil difliision pump with a speed of a 1000Us. A portion of the resulting free jet was passed through a skimmer with a 4-mm aperture 42 43 situated after the nozzle into a vacuum chamber containing the quadrupole mass filter and pumped by a pair of bafiled 6-in oil diflirsion pumps with a combined speed of the order of 3000113. Afier the introduction of the original ESI interface, several research groups have duplicated the interface and the results very quickly (5, 6, 7, 8). In all the ESI sources, there are several common features. The ions are formed by forcing liquid through a small stainless steel capillary tube held at high potential. At the tip of the capillary tube, a fine mist of charged droplets is formed. The highly charged droplets are desolvated and ions are transported to the mass analyzer through a set of ion focusing lenses or RF -only quadrupoles. There are several different methods reported for declustering the solvated ions during the ESI process. Mainly three techniques were employed to’ achieve declustering by using a heated counter-current flow of gas through the nozzle-skimmer region (3, 4, 5), a heated metal capillary to control desolvation and transport ions (7), or a heated ion source block (8). The flow of ions and gas going into the mass spectrometer was controlled by using a skimmer and a nozzle (4, 5, 6), or a small diameter capillary and a skimmer (7). These systems use high pumping capacity pumps to achieve reasonable low pressures (10'5 torr or low) inside the mass analyzers. In order to take the advantage of the sensitivity of ESI/MS, an electrospray interface was designed, built in-house, and coupled with a single quadrupole mass spectrometer (HP 5985). An interface between the mass spectrometer and a 486, 33- MHz personal computer has been developed. The interface was assessed for the operational use with model compounds. The intention of this chapter is to introduce the considerations for source design, fabrication, and performance of the newly built source. 44 11. HP 5985 Single Quadrupole Mass Spectrometer This project was started with the HP 5985 single quadrupole mass spectrometer and its components. A thermospray unit, another LC/MS interface fi'om Vestec Corporation (Houston) was used with this mass spectrometer. In this section, the operation principles of a quadrupole mass spectrometer and the vacuum system of the HP 5985 mass spectrometer will be briefly discussed. A Operating Principles of Quadrupole Mass Spectrometer The HP 5985 mass spectrometer is a single quadrupole instrument. The quadrupole mass spectrometer employs a combination of direct current (DC) and radio- fiequency (RF) potentials to separate ions passing through the quadrupole rods according to the m/z values. Ideal performance is obtained with rods having a hyperbolic cross section (9-11), however, cylindrical rods are most often used and provide satisfactory results. Opposite rods are connected together electrically and to RF and DC voltage generators. As the ions travel through the central longitudinal region between the rods, they are influenced by the combined DC and oscillating RF fields. As the ions drift toward the detector along the Z-axis, their motion in the X-Y plane is described by the Mathieu equation (1 1), that defines ax and qx parameters as follows: ax = -ay = 4ZeU/mro2ro2 eqn 2-1 qx = -qy = ZzeV/mcozro2 eqn 2-2 where r0 is half the distance between opposite quadrupole rods, 0) is the alternating frequency (RF), U is the magnitude of the DC field, and V is the amplitude of the RF-field. Figure 2-1 represents the a-q stability diagram; the tip of the diagram represents coordinates of those ions which will have stable trajectories through the quadrupole field. 45 The slope of the m/z scan line is defined by the ratio 2U/V. Altering the slope Of the mass scan line changes the mass-resolution Of the ions. In a typical quadrupole mass spectrometric analysis, voltages are selected tO achieve unit mass resolution. If the DC/RF ratio were adjusted to raise the Operating line even higher, the resolving power Of the instrument would increase, but the sensitivity would decrease, because fewer ions would follow a stable trajectory to reach the detector. In the "RF-only" mode, there is no DC field, U = 0, the Operating line lies horizontally along the abscissa, indicating that ions having Operating point between q = 0 and q = 0.908 could be transmitted through the RF-Only quadrupole filter. However, as suggested by Miller and Denton (12), the transmission efficiency for ions Of all m/z values is not 100% in an RF-Only quadrupole. Not only is the transmission Of ions through a quadrupole influenced by the stability Of the ions as defined by the Mathieu stability diagram, but by (i) the acceptance aperture Of the quadrupole and (ii) the spatial focusing conditions that arise due to the trajectory Of the ions through the quadrupole (12). Therefore mass discrimination efi‘ects are apparent, which implies that RF-0nly quadrupoles are not strictly total ion transmission chambers, that is, ions Of different m/z ratios are transmitted through RF-Only quadrupole rod assemblies with different degrees Of efiiciency. B. Vacuum System Of the HP 5985 Mass Spectrometer The vacuum system of the HP 5985 mass spectrometer uses conventional multistage Oil difl'usion pumps which are pumped by mechanical pumps. The difi‘usion pumps and their associated baffles are water cooled. A diffirsion pump with 600L/sec speed for the EI-CI ion source provides adequate pumping speed to handle the large gas flow rates used in the CI mode. A smaller 150 Usec unit takes care of pumping the mass 0.2 ' 0.1 46 Mass ...—v Scan Line Figure 2-1. Stability diagram Of (a, q) space showing regions (patchwork area) that corresponds to mathematically stable ion trajectories in the quadrupole mass spectrometer. 47 filter and the detector portion where gas flow rates are much smaller. The pressure inside the source housing and the analyzer assembly is monitored using the ion gauges at the base Of the chambers. In electrospray operation, the vacuum system Of the mass spectrometer was used without modifications. III. Design Consideration for the ESI Interface Electrospray ionization is an atmospheric pressure ionization (API) technique. Two problems are intimately related in API mass spectrometry: the transport Of ions from an atmospheric pressure region into the vacuum system Of the mass spectrometer, and the strong cooling Of a mixture Of gas and ions when expanding into a vacuum. The resulting condensation Of polar neutrals (notably water vapor) on analyte ions produces cluster ions having a mass far beyond the range Of most mass analyzers (8). The problem Of declustering was attacked by several methods such as heating the ion source region (8), using heated counter gas flow (3,5,6), or passing through a heated capillary tube (7). In this design, the ions will be transported into the mass spectrometer through a heated capillary tube (100°C) to avoid declustering upon expansion into vacuum. A Vacuum System and Sensitivity The goal Of designing a vacuum system for atmospheric pressure ionization mass spectrometry (API/MS) is to transport ions, produced in atmospheric pressure into the mass analyzer and to introduce as many ions, but as little gas as possible. Ideally, ions should be separated fiom gas. In a simple approximation, the sensitivities Of different API mass spectrometers can be judged fiom the throughput Of gas in the relevant vacuum stage (13). 48 API/MS instruments can be divided into single-stage vacuum system and multiple- stage systems. In a single-stage vacuum system, the flow Of ions into the mass analyzer is proportional to the gas flow through the sampling orifice. The throughput Of gas into the analyzer is given by the pumping speed multiplied by the pressure in the vacuum system (13). The purpose Of the multi-stage vacuum system is to use a larger orifice, which is not easily blocked and can take more gas and ions into the mass spectrometer. In a typical differentially pumped system, the throughput of gas into the ion Optics stage is most important. The bigger the pump and the higher the pressure tolerated by the ion Optics, the higher the throughput Of gas plus ions. In the ion Optics region, ions are focused and transmitted towards the mass analyzer, while gas is pumped away, thereby increasing the ion-tO-gas ratio. The region between the nozzle and the skimmer is called the molecular- bearn stage (Figure 2-2). The molecular-beam stage is supposed to transfer a portion Of gas plus ion beam without changing the ion-tO-gas ratio. Therefore, the molecular-beam stage does not help to increase the sensitivity by an increase Of ion-tO-gas ratio. However, a better shaped beam is presented to the mass analyzer, which will increase the transmission of ions through the analyzer. A significant advantage is that a bigger orifice makes the instrument more rugged. The prediction of sensitivity based on vacuum considerations alone is a first approximation. The effects Of the quality Of the molecular beam components, ion Optics, and mass analyzer cannot be included in a simple generalization. B. Transport of Gas and lens into Vacuum A simple scheme Of the expansion Of a gas into a low-pressure region is given in Figure 2-2. The principles Of the generation Of molecular beams (14,15) are explained in 49 qualitative terms. Inside the nozzle Opening and behind the nozzle, the gas molecules acquire a high velocity, and gas molecules with entrained ions follow straight streamlines originating approximately in the nozzle, the highest intensity of the gas flow being on the axis Of the nozzle. Far away from the nozzle, the gas is pumped away, and gas molecules move at random. In the transition between directed motion and random motion, called shock waves, ions and gas molecules undergo many collisions, with scattering Of the beam of ions and molecules as the result. The region inside the barrel shock wave, and between the nozzle and the mach disk, is called the silent zone, where ions and gas move at equal speed in the same direction and undergo strong and rapid cooling. The location Of the mach disk is important for the design Of an API mass spectrometer. The distance fi'om the nozzle is given by eqn 2-3. 1M = 0.67 D0 (Po/P1)“2 eqn 2-3 where Do is the orifice or nozzle diameter P0 is the upstream pressure (atmospheric pressure) P1 is the downstream pressure in the vacuum chamber xM is firlly determined by the pumping speed available in the volume near the nozzle Opening (16). A small pump or narrow pumping line or other Obstruction will result in a small x”. Often the flow through the nozzle is tOO large for the vacuum pumps Of the mass analyzer. The central portion Of the beam can be sampled into the next vacuum stage by means Of a skimmer (14-17) as in Figure 2-2b and 2-2c. In Figure 2-2b, the core of the beam is sampled from the silent zone, and the ions plus gas continue their movement in straight lines. Collection Of ions fiom the gas flow 50 through the skimmer should be efiicient, because Of the directional effect Of the molecular beam. However, since the gas flowing in the molecular beam has strongly cooled upon expansion, ions will cluster with water molecules, if present. In Figure 2-2c, a sample is taken from behind the mach disk. Ions and gas have undergone extensive scattering, and the extraction and focusing Of ions is more difficult. On the other hand, the gas temperature has risen through collisions in the mach disk, with breaking Of hydrogen bonds in cluster ions. The circumference Of the skimmer Opening should be very sharp and fi'ee fiom burrs. The full angle Of the skimmer cone is usually about 60° in a molecular beam apparatus (14,15); a full angle up to 110° appears to be usable without adverse effects. A larger angle and/or imperfections at the skimmer orifice will result in disturbance Of the molecular beam if sampling is attempted from the silent zone. A lower transmission of ions has been reported in such a case (18). Even with nearly perfect Skimmers, disturbance Of the molecular beam is unavoidable near the edges. As a result, the effective skimmer Opening in molecular beam experiments is always smaller than the physical Opening. IV. Fabrication of Source and Flange The HP 5985 mass spectrometer was modified to accomadate the ESI source. A thermospray interface was earlier fitted in the mass spectrometer. The instrumentation for thermospray mass spectrometry will be discussed before the details Of ESI source fabrications and modifications. 5] Vacuum 1 atm # silent zone Ions, N2 <—:—> (a) <—— mach disk Nozzle barrel shock -3 1 torr 1 X 10 torr 1 atm Ions, N2 /' . . (b) \ Ions, N2 NOZZIC skimmer . -3 1 atm 1 X 10 ton- / J Ions, N2 T» T. Ions, N2 (C) Nozzle skimmer <———> XM Figure 2—2. Expansion Of gas into vacuum; (a). simplified scheme with shockwaves and silence zone; (b). generation Of a beam Of gas and ions by sampling from a silent zone, the skimmer penetrates the mach disk; (c). sampling Of gas and ions with a skimmer located beyond the mach disk. 52 A. Instrumentation for Thermospray Mass Spectrometry Thermospray (TS) ionization is a mild ionization technique that uses heat to vaporize the liquid stream. A partial or complete vaporization Of the liquid stream is carried out passing it through a heated capillary tube. At the tip Of the capillary tube a supersonic jet of vapor is created, normally containing a mist Of fine particles and solution droplets. The droplets are desolvated by the nearby hot surfaces and excess solvent vapor is removed by an additional pumping stage. In the "filament Ofi" mode Of Operation, normally the solutes are mixed with a volatile bufi‘er such as ammonium acetate. It is assumed that a gas phase reaction occurs between analyte and N114+ molecules to transfer protons tO analyte molecules or to form adduct ions with NH4+ (19,20). In the "filament on" mode Of Operation, a discharge electrode is used tO ionize analyte molecules. The ions are sampled through a skimmer and mass analyzed by a mass analyzer. The solvated ions are desolvated while passing through the source block and a heated lens system. The major components Of the thermospray source are: a heated source block, a sampling cone, a vaporizer probe, a discharge electrode, a pumping line to remove excess vapor, and thermocouples to monitor the temperature of the vaporizer probe, the source block, and vapor. Figure 2-3 shows the Vestec thermospray source (for HP 5985 or newer models). The TS probe produces a supersonic jet Of vapor containing a mist of fine droplets or particles. As the droplets travel at high velocity through the heated ion source, they continue to vaporize due to rapid heat input fi'om the surrounding hot vapor. The ion entrance aperture to the mass analyzer is lOcated at the apex Of a cone perpendicular to the vapor jet from the thermospray vaporizer and near the center. In a fi'eely expanding supersonic jet, heavier solutes tend to remain close to the jet axis. The purpose Of the sampling cone is to extract these heavy ions selectively in order to increase sensitivity. The desolvation Of ions takes place at the heated TS source as well as at the heated lens 53 region. The solvent vapor was pumped down by the l/2-in pumping line connected to a cold trap and a 300 um mechanical roughing pump. In this Vestec thermospray interface, the vaporizer probe is placed right angles to the axis Of the mass analyzer. The ions emerging from the skimmer are focused by a set of lenses and analyzed by the quadrupole mass analyzer. The ions are detected by a continuous dynode electron multiplier. HP 5985 single quadrupole mass spectrometer with Vestec thermospray interface having 1/2-in pumping line with the existing vacuum system were not able to maintain the vacuum inside the mass spectrometer when the HPLC effluent was introduced into the mass spectrometer. Therefore, this interface at the MSU Mass Spectrometry Facility was abandoned without use for long time. B. SIMION Ion Simulation The SIMION simulation program (21) was used to study the trajectories Of ions produced by electrospray. The SMON program is an electrostatic lens analysis and design program that allow estimates Of the trajectories of ions in an electric field. When the ESI probe and the ion transport capillary tube is perpendicular to the axis of the skimmer, the ion trajectories did not divert towards the skimmer, when the ESI source was at ground potential and the skimmer at 15V. The potential on the skimmer and the pressure gradient was not enough tO draw ions towards the skimmer. A repeller at high voltage (several kV) has to be introduced in front of the skimmer to divert ions towards the skimmer. When the probe was placed parallel to the axis Of the skimmer, the trajectories Of the ion beam enter the mass analyzer through the skimmer. The simulation studies indicated that the parallel geometry would be the best for the introduction Of ions into the 54 Ions to MS Filament or Discharge electrode Sampling Cone WWW/ J \/ Solvent Vapor tO Trap and Rotary pump Figure 2-3. The Vestec thermospray interface for the HP 5985 mass spectrometer. 55 mass analyzer. Therefore, the parallel geometry was adapted. Figure 2-4 shows the plots fiom the SIMION ion simulation. C. Source Modification As mentioned earlier, the most difiicult problem in API/MS technique is to achieve high degree Of vacuum inside the mass spectrometer. By decreasing the pressure before the skimmer (inside the E81 source), pressure inside the second vacuum stage can be decreased. This can be accomplished by increasing the conductance Of the vacuum line connected to the E81 source chamber. Throughput is the quantity Of gas (the volume Of gas at a known pressure) that passes a plane in a known time ant (PV) = Q (22). The flow Of gas in a channel is dependent on the pressure drop across the tube as well as the geometry Of the channel. For a long round pipe the throughput (in viscous flow) is given by the Hagen- Poiseuille equation, eqn 2-4 (22). Q: m4 (Pl+P2)(Pl-P2) eqn 24 1281]! 2 where dis the diameter Of the tube, t is the length Of the tube, a is the viscosity of the gas. Division Of the throughput by the pressure drop across a channel held at constant temperature yields a property known as the intrinsic conductance C of the channel as given by equation 2-5. Q C = n 2-5 P2-P1 “1 56 \ Electric Field Lines Mass Analyzer (3) Skimmer ESI Needle (3000V) Tr ectories loo 3,] / Ion Tra/nsport Capillary (100W\\\ Skimmer ——> C3 5: (h) f ’ Ion Transport. Capillary (100V) ll we ESI needle (sooev? @ Figure 2-4. The plots fiom the SIMION ion simulation. (a). Heated capillary is right angle to the axis of the mass analyzer. (b). Heated capillary is parallel to the mass analyzer. In both cases, source block is at ground potential. Electric Field Lines 57 where P1 and P2 refer tO the pressures measured in large volumes connected tO each end Of the channel or component. The units Of throughput Q is Pa-m3/s (or Pa-L/s) and that Of conductance or pumping speed C is m3/s (or Us). The conductance Of a long round pipe (for air at 0°C) is given by the equation 2.6. +P C (US) = 1.38 x106 9; (112—2) eqn 2-6 Therefore the conductance Of a pipe is proportional to the (diameter)4 in the viscous flow regime (22). By increasing the diameter by a factor Of 2 causes tO increase the conductance by a factor Of 16. The conductance Of the existing pumping line can be increased by increasing the diameter Of the vacuum line. The maximum diameter Of the vacuum line was determined by the space available on the vacuum flange and that Of the source chamber. A l-in pumping line was attached to the ESI source through the vacuum flange to increase the conductance Of pumping line. The increased conductance of the pumping line can remove excess solvent vapor entering the ESI source. A source was fabricated to accommodate the l-in vacuum line. Figure 2-5 shows the new ESI source. A source block was (Figure 2-5) designed to control the flux Of ions and gas entering into the mass spectrometer. This design facilitates three stages Of differential pumping: molecular beam, ion optics, and mass analyzer. A 0.5 mm-ID skimmer was used to sample a fiaction of the flow Of ions and gas emerging from the ion transport capillary before entering the ion Optics region. The skimmer is electrically isolated from the source block using a rubber O-ring placed in between the skimmer and the source block. The skimmer was connected to a voltage supply (0-50V). The place Of the skimmer was designed so that it can be placed co-axially with the mass analyzer. The ion transport 58 capillary tube can be placed cO-axially with the skimmer and a fiaction of the emerging ion beam can be sampled and focused into the quadrupole mass analyzer. A l-inch pumping line was connected to the source tO remove excess solvent vapor. The pumping line was electrically isolated fiom the source using a Vespel spacer. The ion transport capillary was electrically isolated from the source using a Teflon spacer. The spacers provide vacuum seals as well as electrical insulation for the individual components. The source block was mounted on a platform inside the source housing. D. Vacuum Flange Modification The vacuum flange also had to be modified to accommodate the vacuum line and the ion transport capillary. A l-in Swagelok® welding fitting was installed in the vacuum line. Another Swagelok® welding fitting with the inner diameter Of l/4-in was installed to insert the ion transport capillary tube cO-axially with the skimmer. The capillary tube was electrically isolated from the flange by using a Vespcl® ferrule (ID l/l6-in enlarged to 3/16-in, OD l/4-in). The original electrical feedthroughs were used without modifications. E. Design and Fabrication Of the Ion Transport Capillary A vital part Of the atmospheric pressure mass spectrometry is to transfer ions produced at atmospheric pressure into the high vacuum region Of the mass spectrometer. In ESI/MS, this is achieved by restricting the flow of ions and gas into the mass spectrometer using small diameter skimmers, nozzles, or capillary tubes. The ESI produced ions are generally solvated clusters. The solvated clusters must be desolvated to be mass analyzed. Several researchers Observed no signal fiom an analyte without 59 external assistance for the desolvation. On the other hand, expansion Of gas and solvent vapor molecules into a vacuum creates a strong cooling. The resulting condensation Of polar neutrals (notably water vapor) on analyte ions produces cluster ions having a mass far beyond the range Of most mass analyzers (13). In this study, a stainless steel capillary tube (0.5 mm 1. D., 1/16" 0. D.) was used to transport ions produced at atmospheric pressure or outside the mass spectrometer into the high vacuum region Of the mass spectrometer. The declustering was achieved by heating the capillary tube while ions and gas were passing through it. A capillary tube with 0.5 mm inner diameter was used to transport ions produced in atmospheric pressure into the ESI source region (several torr). The inner diameter Of the capillary tube controls the flow Of gas and ions tO Obtain the adequate degree of vacuum inside the mass analyzer using the present vacuum system. The ends Of a stainless steel capillary tube (0.5 mm ID, 1/16" OD and 25 cm long) were electropolished tO remove the sharp ends. A heating wire (1/16" OD) was wrapped around the capillary tube tO control heating Of the capillary tube. The capillary tube and the heating wire were placed inside another stainless steel tube (3/16” OD, 1/8” ID) as shown in Figure 2-6. In this design, the ion transport capillary has to be placed inside another tube because a 25-cm long capillary tube can be bent or deformed by heating and cooling. The deformation Of the capillary tube can reduce the ion transmission emciency. One end Of the capillary tube was silver soldered to one end Of the outer stainless steel tube to Obtain a vacuum seal. The whole length Of capillary tube can be heated by this way tO achieve complete desolvation Of ions. A controlled voltage was applied tO the heating wire using a voltage regulator. A Cr—Ni thermocouple was attached tO the outer walls Of the capillary tube and temperature Of the tube was monitored using a thermocouple gauge. The capillary tube was connected to a voltage supply (SOV-ISOV) (Hewlett Packard 6207B DC power supply) to apply voltage to the ion transport capillary. 60 skimmer (0.5mm ID.) in A: ._ ._:::_:.:._1$?:-:-;12-':-:-: ; .:v:.:::-;;~‘-; W -‘.'.'.'.‘.-.-'.-.-.-.‘.‘.'.~.‘;-.‘.'.'.' Heated Ion Transport Capillary Tube 1" pumping line Insulator O-ring Figure 2-5. The ESI source designed for the HP 5985 mass spectrometer. 61 Alternatively, the ion transport capillary tube was resistively heated by applying DC current through the Opposite ends of the capillary tube using a power supply (Sorensen Power Supplies, Raytheon CO. SRL 20-25). The desolvation was achieved by both methods separately. It is important tO have a very good vacuum seal between the stainless steel capillary tube and the vacuum flange during repeated heating and cooling processes. The vacuum seal was made between the 3/16" tube and the vacuum flange using a stainless steel Swagelok fitting and a Vespel fen'ule. For this system, a capillary tube was used to transport ions into the vacuum system. Therefore, it can be assumed that the nozzle diameter is that Of the capillary tube. The pressure inside the ESI chamber is about 1.5 torr. The upstream pressure Of the capillary tube is 760 torr. The silent zone exists up to 7.5 mm from the end Of the capillary tube inside the ESI chamber (from eqn 2-3). For efiicient ion sampling, the skimmer should be kept inside the silent zone (13). In this system the skimmer is kept at about 3.4 mm fiom the end of the capillary tube. If the skimmer is kept very close to the capillary tube, the pressure inside the source housing cannot be maintained properly. That is mainly because Of most Of ions and gases emerging from the capillary tube through the skimmer into the source region. Pressure Inside the Mass Analyzer: when heated capillary inlet closed ..................................... 5 X 10'7 to“ with capillary inlet Open to atmospheric air --------------------- ~5 x 10'6 torr (at 100°C) 62 58888092 v.88 3% mm 05 com gammmov 33 ace—=98 team—:5 com 2F .orm 82mm 538m ems; > .2; , .6 an: own; same b b m _> k ' ’ ’ l 'l ’r ' ' ' ’ ’ 1" 1" lllrl a ‘v 1" 1" 1' ‘ ll: t‘v‘"" .. a 0330688.; 63 F. Electrospray Needle Assembly (with nebulizer) In an ESI interface, an analyte solution is electrosprayed into the atmosphere through a metal capillary tube (150 pm) that is held at a potential of several kilovolts relative to the counter electrode. The buildup of charges at the liquid surface at the tip of the capillary tube creates such an instability in the liquid such that coulomb repulsion forces are suflicient to overcome the surface tension so that small charged droplets separate fiom the liquid emerging from the capillary tube. The technique works best with flow rates in the range of 5-10 uL/min (2). At higher flow rates and/or higher percentages of water, the electrosprayer produced a stream of larger droplets together with the desired fog (3). An increased voltage applied to the spray capillary dispersed the liquid droplets, but at the same time initiated a corona discharge (23, 24) that substantially lowered the sensitivity for samples ionized in solution. The combination of pneumatic nebulization and electric field tolerates higher eluent flow rates and a higher percentage of water in the formation of a spray of charged droplets (6). An electrospray needle assembly was constructed in house with a nebulizer as shown in Figure 2-7. The construction of ESI needle assembly was canied out using standard fittings and other components. The ESI needle is a lO-cm long stainless steel capillary tube and it has an inner diameter of 150 um and outer diameter Of 250 pm. The capillary tube was placed inside another stainless steel capillary tube with an inner diameter of 275 um (outer diameter of 1/16”). The outer capillary tube was secured to a stainless steel union Tee (Swagelok®) using a Vespel® ferrule (I. D. = 1/16”). The small capillary tube protrudes about 0.2 mm from the outer capillary tube. The small capillary tube was sent through the union Tee and tightened with another Vespel ferrule (I. D. = 250 pm) at the other end of the Tee. The other end of the stainless steel capillary tube was connected to a fused silica capillary tube fi'om the liquid chromatographic system. A 64 butt connector was placed in between two pieces Of firsed silica capillaries to avoid a leakage of high voltage into the HPLC pump. A Teflon tube (0. D. = 1/16") carrying N2 gas was connected to the remaining outlet of the union Tee. N2 at 35 psig was used for pneumatic nebulization. The stainless steel union was connected to a high voltage (0 - 5 kV) supply (Bertan Associate, Inc, Model 603-50P). The whole needle assembly was secured on a custom-made polycarbonate holder. The holder and the needle assembly were placed inside a Plexiglas cylinder that is front-side Open, for safety purposes. The Plexiglas cylinder was mounted on a movable rail to adjust the height and the position relative to the ion transport capillary. G. Electropolishing Of the Capillary Tubes The sharp edges of the ion transport capillary and the ESI needle were removed by electrOpolishing both capillary tubes: the ESI needle and the ion transport capillary. A mixture of glyceroVH3PO4/HZO (1:1 :1) was used as the electrolyte solution. A stainless steel electrode having a larger surface area was used as a cathode. The capillary tube was used as the anode. A current of 50 mA was passed through the circuit for about 2 hr or until the desired smoothness was Obtained. The capillary tubes were thoroughly cleaned with water and methanol before use. 65 new bum—.502 505$» .5 55 33 on :8 38m 05. 6:802 xmuamoboflm firm 0.53m cougfinoz e8 mew NZ 33 coach l > o_ a - c + 3 .H a: om: 23 526 85 seamen one rezone our 825% .o ._ :9: wmqtvmrrw ”r: .' 95a 0A 88% 66 uoaoEobooam .932 3% mm 0.,: co coerce: moisten—m 829:8 2:. .w-m 05mm 9 so: menace - ..H a mm H m — 5:38 tonnage :8 «.882 Hmm / _ m \ ///////////// E a _.l BASED 8588/ \’ 67 V. RESULTS AND DISCUSSION Several configurations were considered for the construction Of the ESI source for the HP 5985 single quadrupole mass spectrometer. The Vestec corporation implemented an ESI source for a single quadrupole mass spectrometer and in this design, a heated ESI source was used to achieve declustering of solvated ions. Three skimmers in series were used to sample the flow of ions and gas molecules into the mass spectrometer. The introduction of two stages of differential pumping and two pumping lines before the mass analyzer is not easily provided in our mass spectrometer, because the source housing and the flange must be modified to introduce two additional pumping lines and the probe. Heating gas has been employed to achieve declustering of solvated ions in some designs. These configurations need high pumping capacity pumps to evacuate the source housing and mass analyzer regions (Fenn et al. used 1000L/s and 3000Us pumps (3); Henion et al. cryo surfaces at 20-30K in the source housing (5), Smith et al. 30,000Us and 500113 (5a). Chowdhury, Katta, and Chait (7) reported an ESI/MS interface that uses a heated capillary tube to transfer ions produced at atmospheric pressure into the mass spectrometer and to desolvate the ions passing through it. The advantages of this design are: after the first adjustment of the position of the heated capillary tube no further adjustments are needed, the spray plume can be optimized by visually Observing the spray, no need for heating gas, and therefore no need for very high pumping capacity. Chait's design was compatible with our modifications because of the simplicity of the interface. Simulation studies show that the best ion transmission can be Obtained when the ion transport capillary, the ESI needle, the skimmer, and the mass analyzer are placed co-axially. Therefore, a new source, an ion transport capillary, and an ESI needle were 68 designed and built as shown in Figures 2-4, 2-6, 2-7. The vacuum flange was modified to accommodate the ion transport capillary and the pumping line for the mechanical pump (Figure 2-5). The ion transport capillary was installed and isolated from the source and the flange by a Teflon spacer and a Vespel ferrule, respectively. The outside end of the heated capillary tube was closed with a Teflon cap when the system is not in use. The complete system is shown in Figure 2-8. The pressure inside the mass analyzer was monitored using a vacuum gauge located at the base of the analyzer chamber. The pressure inside the mass analyzer was found to be 5 x 10'7 torr (capillary end closed) and 5 x 10'5 torr (capillary Open to the atmosphere) when the capillary is at 100°C. Back streaming of difi‘usion pump Oil was observed at the source region. Since one end of the heated capillary tube is Open to the atmospheric air, it has no influence on the flow rate of the liquid inlet system provided a stable spray. This feature is very important when combining liquid separation techniques with the ESI/MS. The ESI needle is capable of producing a stable spray plume at high flow rates (>300 uL/min) with the assistance Of nebulizing gas. The ESI needle was kept a few mm Off-axis to the heated capillary tube to avoid sampling of larger droplets. The heated capillary can be cleaned with a stainless steel capillary tube (350 um ID) when it is plugged with residual solids. The distance between the capillary tube and the ESI needle is about 1cm. The position of the needle relative to the capillary tube was optimized by monitoring the current emitted by the ESI spray. The analyte solution is electrosprayed fi'om the ESI needle that is maintained at 3kV relative to the ion transport capillary tube through which droplets, ions, and gases enter the mass spectrometer. A flow rate of 10 uL/min was maintained by a Waters HPLC pump with a splitter (split ratio 1:9). The distance between the electrospray needle tip and the capillary tube is typically 1cm. The quality Of the mass spectrum is strongly dependent on the quality of the spray emitting from the needle. In the present design, the 69 spray can rapidly be optimized by direct Observation from outside the vacuum housing and by monitoring the current emitted fi'om the needle. The position Of the stainless steel capillary tube was adjusted to give a fine symmetrical plume. Electrospraying of the analyte solution produces fine, highly charged droplets. These droplets attempt to follow the electric field lines and migrate towards the metal capillary tube that projects into the first vacuum stage of the mass spectrometer. The first vacuum region is evacuated by rotary pump (Edwards E8, ). A fi'action of migrating droplets enters the long stainless steel capillary tube assisted by the strong flow of gas that results fiom the large pressure difference between the extremities Of the tube. Droplets entering the tube tend to be focused toward its axis by this strong gas flow (25) and are thus transported through the tube (7). The droplets were continuously desolvated inside the capillary tube by controlling the heat applied to the capillary tube. A fi'action Of the material that emerges from the capillary tube passes into a second vacuum chamber through a coaxial, 0.5 mm diameter orifice in a skimmer situated about 3.4 mm from the end of the tube. The solvent vapor emerging from the heated capillary tube was removed by the rotary pumping stage before the skimmer. The ions that pass through the skimmer are focused by a set of ion focusing lenses into the mass analyzer. Further desolvation was carried out in the skimmer region by collisional activation due to the voltage difference between capillary and skimmer (26, 27). A Data Collection After the successful modification of the ESI source and the vacuum system, the ESI interface was assessed for its ability to make electrosprayed ions. It is known that molecules having pre-forrned charges show very good response (6) in electrospray mass spectrometry. Tetraphenylphosphonium iodide (molecular weight of tetraphenylphosphonium cation is 339 u) was selected as the model compound for tuning the mass spectrometer. 70 Tetraphenylphosphonium bromide dissolved in 4% acetic acid in methanol/H20 (SO/46M % v/v/v) was continuously infused into the ESI interface. A continuous flow rate of lOuL/min was maintained by a Waters HPLC pump (model 410) with a split ratio of 1:9. The Waters HPLC pump has the minimum flow rate of lOOuL/min and increments of lOOuUmin. A voltage of +3.9kV was applied to the ESI needle. The temperature of the ion transport capillary was elevated to about 85°C as measured by a Cr/Nr thermocouple attached to the outside wall Of the ion transport capillary. The voltage on the capillary tube was held at 100V. The skimmer was kept at 15V. The ESI current between the ESI needle and the ion transport capillary was Optimized by looking at an oscilloscope connected to a home-made picoammeter. Figure 2-9 shows the circuit diagram used for the ESI current measurement. The distance between the capillary tube and the needle was adjusted to obtain the optimum current. The ESI needle was kept at about 1cm away fiom the capillary tube and a few mm ofl'-axis. A high frequency oscilloscope was connected to the mass spectrometer output using an extension to monitor the detector signal. The mass analyzer was continuously scanned between 330 and 345u. The voltages on the lenses were varied while continuously scanning the mass analyzer. Once the signal for the tetraphenylphosphonium cation (TPP+ m/z = 339) was observed on the oscilloscope, it was easy to optimize the signal by changing lens voltages, skimmer voltage, capillary voltage, capillary temperature etc.,. Further, the signal was Optimized by moving the position of capillary tube relative to the skimmer. Afier optimizing the system with the oscilloscope, the mass spectrometer was re-tuned with the data system. The present PIP-5985 data system has no capability to calibrate the mass spectrometer without the E1 source. Therefore, the mass spectrometer could not be calibrated with the ESI interface. When the E81 needle was very close to the heated capillary tube, a discharge could occur and a glow (a discharge between the needle and the heated capillary) could be observed if the surrounding was dark enough. The discharge could be heard if the 71 surroundings were quiet. When a discharge occurs, the major ion in the spectrum was due to H+ and no other major ion was observed. The change in capillary voltage with the ion intensity of stanozolol (M. W. = 328) in methanol/acetic acid (1%) containing very basic pyrazole ring in the molecule was studied. The intensity of the signal did not change significantly when the capillary voltage was varied between 75-125V. Outside this range a drop in signal intensity was observed. As the skimmer voltage falls below 10V, the signal intensity started to drop. When the skimmer voltage was above 20V, the intensity of the peak becomes weak. The temperature of the capillary also can be varied without adversely affecting the signal. Next, an arginine (50 pmol/uL) solution was introduced into the ESI interface at a flow rate of 10 uL/min. The voltage on the ESI needle was at 3kV, heated capillary was at 100V and 100°C, the skimmer was at 15V, and lenses were maintained at the same values after tuning. Arginine (M. W. = 174) was detected as a protonated molecule under the above conditions. The spectrum contains a single peak representing the protonated molecule of arginine in solution. No fragmentation was Observed for this species. The capability Of forming multiple charges was assessed by introducing a solution of tri-peptide lys-tyr-lys (10 pmol/uL) (M. W. = 437). It contains two lysine residues (two fi'ee side chain amino groups) and a terminal amino group. The spectrum of tri- peptide contains two major peaks at m/z 438 and 220 corresponding to the protonated molecule and the doubly charged molecule, respectively. The doubly charged molecule has the m/z value of 219.5, but the data system presents it as 220. Figure 2-10 shows the ESI/MS mass spectrum of the tri-peptide lys-tyr-lys. Afier establishing spectra of low molecular weight compounds, higher molecular weight peptides were tried on the ESI interface. A seven-residue peptide, Val4 Angiotensin III, (arg-val-tyr-val-his-pro-phe) (M. W. = 917) was successfirlly tried on the 72 ESI/MS interface. The spectrum (Figure 2-11) shows the protonated molecule at m/z = 918 and the doubly charged molecule at m/z 459. The Val4 Angiotensin III peptide has arg, his, and a terminal amino group as possible charging sites; but the doubly charged ion was observed as the highest charge state. The spectrum of bradykinin (M. W. = 1060) having the sequence of arg-prO-pro-gly-phe-ser-prO-phe-arg was also acquired. This molecule has two possible charging sites: a fi'ee amino terminal and the amine group on the arginine side chain. Only doubly charged molecule was Observed in its spectrum, but the singly charged molecule has an m/z value above the limit of the mass analyzer which is 1000 Daltons. These molecules did not show any fragmentation under these conditions. The main goal of using the modified ESI interface is for the analysis of small molecules. Therefore, further analyses Of large molecules were not attempted with the interface. VI. Design and Construction of Computer Interface A. Introduction The HP 5985 mass spectrometer uses a HP 21MX-E series computer with the Tektronix 4012 graphics display terminal to control the mass spectrometer. The data are transferred to an Old fashioned cartridge or PDP-ll system for storage. The data system on the HP 5985 mass spectrometer is no longer reliable for continuous operation. At the same time it did not provide several desired capabilities that are required for modern mass spectrometry such as fast data acquisition, profile data collection, etc.,. The calibration of the mass spectrometer can only be done with the E1 source. For the calibration, the ESI source had to be replaced with the E1 source. The scanning rate of the instrument cannot be fully controlled by the user. 929:3 Hmm manage .8.“ 86me “Soho 05. .mrm Saw—m 2.823 3 7 3 ¢ >c_m-o my «2 _ S596 tease :3 H‘ BE “.10.. . : "33 Ill” .38wa use >2+ . /18_|_|» one. "5 Jlllwu a: .e / >8- m _\# _/_ 7 J\ >2 + >2- 74‘ Relative Intensity M210“ 0 130 170 210 220 250 290 (Mr-H)“ 438 330 370 Figure 2-10. ESI/MS mass spectrum of Lys-Tyr-Lys (M.W. = 437). 570 75 459 (M+2H)2+ 90 130 170 210 250 290 330 370 410 Relative Intensity 810 850 890 930 970 1010 1050 570 610 650 690 730 770 Figure 2-11. ESI/MS mass spectrum of Angiotensin III (M. W. = 917). Flow rate,= lOuUmin. ESI needle at 3.9kV. Heated capillary at 100°C and 100V and the Skimmer at 15V. 76 B. Computer Interface An inexpensive interface was designed and constructed in house using readily available components. The 21MX E-series computer on the HP 5985 mass spectrometer was replaced with a 486 DX, 33MHz personal computer (Gateway 2000, S. Dakota). Since the 486 computer is not directly compatible with the electronics of the HP 5985 mass spectrometer, an communication interface between the 486 computer and the mass spectrometer has been developed. A block diagram of the digital interface for the HP 5985 mass spectrometer is shown in Figure 2-12. The HP 5985 mass spectrometer has an asynchronous digital interface consisting of separate l6-bit input and output ports with a write strobe to initiate commands to the HP 5985 and a status flag from the HP 5985 indicating data validity for read and write operations. The host computer uses an enhanced version of the industry standard D1024, a CIO—DIO48 to drive this interface. The CIO-DIO48 digital I/O board (Computer Products, Inc., Mansfield, MA) has 24 digital I/O lines and 24 high current (60 mA) output lines. Figure 2-13 shows a schematic diagram of the CIO-DIO48 computer board (28). The board is based upon the industry workhorse 82C55 Programmable Peripheral Interface chip. The 82C55 has 2- byte wide bi-directional data ports (A and B), two 4-bit bi-directional data or control ports bi (C high and C low), and a control/status register used to configure data direction of ports and mode Of Operation. The first 82C55 has ports A and B configured as two 8- bit strobed inputs and is used to receive 16-bit data fiom the 5895 via connector P1. The second 82C55 on board has high current drivers bufl‘ering its cmos level signals. It is configured as two 8-bit strobed output ports and is used to send 16-bit commands to the HP 5985 through P2. The PC/XT/AT bus interface on the board was plugged into an extra expansion slot in the system board of the computer. All signals are connected to the 77 HP 5985 through two 2-meter long ribbon cables and a custom adapter board as shown in Figure 2-14. The adapter board simplifies routing the 2 cables from the 37D connectors P1 and P2 on the CIO-DIO48 to the single connector on the rear of the HP 5985. The digital interface on the mass spectrometer uses a 48 contact, 0.156" on center, edge connector. A standard 50-contact connector, IS, on the adapter has 50 pins and care is required to insure proper alignment of contacts. The top two pins of the J 5 adapter (pin numbers 49 and 50) were not connected. Figure 2-15 shows the schematic diagram Of the adapter. The P1 male connector of the CIO-DIO is connected to a female connector, 11, on the ribbon cable. The connector, J3, at the other end Of the cable is connected to the P3 connector on the adapter. The P2 connector on the board was connected to the 12 connector on another 2 meter ribbon cable and J4 connector at the other end was connected to the P4 connector on the adapter. The RF-fi'equency driver and the other electronic components of the HP 5985 mass spectrometer were used without modifications. C. Data Acquisition and Processing Program A window-based computer program to control the mass spectrometer and to acquire data was developed in the laboratory with the assistance of a computer programmer. This program allows a user to interact with the mass spectrometer using a DX-486, 33MI-Iz computer. Figure 2-16 shows the control panel Of the program. The voltages on difl'erent lenses can be changed with the sliding bars associated with the variables. The resolution of mass spectrometer was adjusted by changing the Ofi‘set and the Gain of the mass axis. The program displays the real-tirne data, the total ion chromatogram, and a mass spectrum on the computer screen. Figure 2-17 shows the ESI/MS mass spectrum of stanozolol (M. W.=328) that .was collected using new program. 78 5380.50on m8:— mwam mm 05 com 3a.t8£ 3&6 05 .«o :5qu 3003 oi. .NTN ocswfi 2 Al mlfil 2 3538 x83 3 AIS. AI E .8 3 4| El: 3 8282800 nu / mm mm fl mm .8833 “M W ”.... mam 8338 u / .. B E a 13:41:43] : a m a. u o no a mm fl mm mew. E 2 mm mm / m a u. m m" fl "m m 1 .l. m fl "u , 2 2 E wwounromu NH 038 coca 79' ounce coarse $05-06 cede cameo assess. are .m 3 seem 020 >+ 2w m. z. 5 ho F0 um pm b< —< E — mam 8.3332:an mmmuuam (.50 5 mo 0 0 mg C Q. as mam <20 80 38888on 82: 3% mm 2: :0 Been 3:va 2: SE ow 8 bacon ovaE 8830 05 .«o .53an 23828 2:. .Erm 23E N a v n O m 0 5 0a 0 Na «a Ca Ha 04 fin .4 ha ON 04 QFDH‘OFUH" .h'n-fl HHHHHNNNNN‘d4-9‘OFUH" .soaueccou so... use. one... ..an on (0:. 37.219... ca Cuzco on as)... 0.30 .LOvuoccou out. ax unattacolcoc (0.: now a Orvooootcou ate... a co: 0.. O’N- ‘CN so «1.0 (20¢ v «.0& d 0.0t Du ‘OLOIESQ it. It.” IVflOMO CO 0’“. «it‘s 50 n).- .huv «.0; d v.0; 0v DOvSOL ca 0483 "moeoz 81 coatoacm 88886on made 3% mm 05 2 Boos $05.05 Set $33 05.. «TN oeawfi IQNOuO co aha-0v « 0» .nrt OOOFt! o N a a a a a O h 0 H a IVNO abnoo (0 0h 82 Figure 2-18 shows the reconstructed total ion current chromatogram obtained by injecting 100pg of testosterone hydrazone into a HPLC system on the ESI mass spectrometer interface. The scan rate was 1 scan/sec (3 50-450u) and the flow rate of the liquid stream was 40uL/min. A C13 column (15cm x 800nm) having 3-pm particles (LC packings) was used. The mobile phase was CH3CN/1120I'I‘FA (40/59.9/0.1 %v/v). The data system is still under development. The data system has no ability to process data on the same program. Therefore, the data were transferred into itr-2, another data system that was developed for a time-of-flight mass spectrometer after transforming into a compatible format. The development of the data system is not complete at this stage. The goal of the development of the data system is to collect and process data on the same computer without transferring into another computer. Usually, ESI/MS data is noisier than EI data and therefore, capabilities such as averaging and smoothing are a very important part of the ESI/MS data processing and development. VII. Triple Quadrupole Mass Spectrometer (TQMS) In this research project, a triple quadrupole mass spectrometer (TQMS) fitted with a Finnigan electrospray source was also used to study the fiagmentation patterns and its other modes of Operations. The Finnigan electrospray source that replaced their earlier source (from Analytica Branford) has some similar features to the ESI source developed at this laboratory. The main different features are the incorporation of a tube lens and the octapole rod assembly to focus ions emerging fi'om the skimmer. Figure 2-19 shows the Finnigan ESI source. 83 38888on 88: 3% mm 05 new 8898 83 3538 05 me $53 35:8 2F OTN ouswfi 4.52 a 8.88. e “E E 8.8. o 83> 2m ......an 8.8 fin M.,... ..ocw coboom cod @ . . .n omd— . :o_mm_Em 8.8- g g 8.8 mmmrx 8.33m Home: .8 96m 83 8:5 825 .9550 84 . mm)" 329 .arc50242 l -r * - . . . - , .l . .1 100 300 Figure 2-17. The ESI/MS mass spectrum of stanozolol acquired using the new data system. Testosterone hydrazone ‘ M+=402 at, -. . .I 0 200 400 scan# Figure 2-18. The reconstructed total ion chromatogram of a LC/ESI/MS analysis of (100pg) testosterone hydrazone. C-18 (15cm x 800 m, 3pm particles) column. Flow rate 40uL/min Of ACN/HZO/TFA (40/59.9/0.1 %v/v). Scan rate 350-450 in 1 sec. 85 The principles and modes of Operation of TQMS will be briefly discussed here. The triple-quadrupole mass spectrometer (TQMS) is a tandem arrangement, in which the first and third quadrupoles are mass-selective and the second quadrupole serves as collision cell (29,30). A schematic of a triple quadrupole mass spectrometer is shown in Figure 2-20. Following ionization in ion source, the ions are focused and passed into the first mass analyzer, Q1. After the first stage of mass analysis, the ions continue their flight path into the second quadrupole, Q2, which is a collision cell or a transmission device. Introduction of a reagent gas into this region (Q2) facilitates the interaction of mass- selected ions and the reagent gas. The reaction products generated in Q2 may then pass to the third quadrupole Q3, which is the second mass analyzer. This technique is referred to as tandem mass spectrometry, mass spectrometry/mass spectrometry, or simply MS/MS. The technique provides a powerful tool to study the ion-molecule reactions and collisionally activated dissociation processes of selected ions. A. MS/MS Scan Modes with TQMS There are four difl‘erent MS/MS scan modes available with a triple quadrupole mass spectrometer, the daughter ion scan, parent ion scan, functional relationship scan and the selected reaction monitoring (SRM) mode (31,32). The TQMS may also be Operated as a single stage quadrupole mass spectrometer by scanning only one of the two mass analyzers while the other two quadrupole rod assemblies in the RF -0nly mode, firnction as ion transmission devices. Thefirnction of the quadrupoles in the different scan modes is illustrated in Table 2-2. 1. Daughter Ion Scan The most familiar and commonly used MS/MS scan mode is the daughter ion scan also called product ion scan. In this mode, the first mass analyzer, Q1, is set to pass ion 86 current of a particular mass-to-charge (m/z) ratio which is referred to as the precursor ion, parent ion, or reactant ion. Precursor ions selected by Q1 interact with the collision gas in Q2 generating product ions or daughter ions. These fragment ions formed in the collision cell enter the third quadrupole which is scanned to Obtain a product (or daughter) ion mass spectrum. 2. Parent Ion Sean A parent ion (also called precursor ion) mass spectrum is obtained if Q3 is set to pass a selected product ion, while the first mass analyzer, Q1, is scanned sequentially passing parent ions with different mass into Q2. The resulting spectrum will indicate all those ions generated in the source which upon reaction with collision gas in Q2, yield a specific m/z value as chosen by the second mass analyzer, Q3. 3. Functional Relationship Scan In a functional relationship scan, both Q1 and Q3 are scanned with a mass offset. The most familiar functional relationship scan is a neutral loss or neutral gain scan, where the mass offset remains constant. Neutral loss scans are used when examining dissociation reactions in the collision cell. A neutral gain scan may be utilized if a reactive collision gas is introduced into the collision cell for associative ion/molecule reactions. In a neutral loss/gain experiment, ions will be detected provided that the parent ion eliminates (or gains) a neutral moiety prior to entering the second stage of mass analysis, the mass lost or gained being equal to the constant mass offset of Q1 and Q3. V 87 Table 2-2: Function of the quadrupoles in various TQMS scan modes. Q1 Q2 Q3 Mode Scan RF-Only RF-Only MS RF-0nly RF-Only Scan MS Fixed RF-0nly RF-Only SIM (MS) RF-0nly RF-Only Fixed SIM (MS) Fixed RF-Only Scan Daughter Ion Scan (MS/MS) Scan RF-Only Fixed Parent Ion Scan (MS/MS) Scan RF-Only Scan Functional Relationship (MS/MS) Fixed RF-Only Fixed SRM (MS/MS) MS = single stage analysis SIM = selected ion monitoring MS/MS = tandem analysis SRM selected reaction monitoring 4. Selected Reaction Monitoring (SRM) Selected reaction monitoring refers to the MS/MS mode in which Q1 and Q3 are set to pass ions Of only one m/z value. Neither of the mass analyzers is scanned, a feature which provides greater sensitivity at the expense of flexibility. Selected reaction monitoring (SRM) is analogous to a selected ion monitoring (SIM) experiment which is 88 employed with a single mass analyzer, and is more selective than SIM experiment. The SRM and SIM techniques are Often used to detect low levels of a selected analyte. 89 0088003 «32 con 0mm. 05 :0 00%85 $538005 comm—BE 0.3. .34.. 08mg 855 09.5.“. 95.5 3082002 82 3 - 08 8.8a €an 3 05805 0t0fimo§< ‘1 06:65 i. { U 4 l— a 0:802 Hmm Own. 83 23300 909 + r 8 See a -8 be as no 9O 08080Q 08088809." 0008 06020000 033 02. 0mg. 05. .oN-N 0.5mE 3980000 00.58 :8 H 00380000 0004 n 0003000 225.6020 vcooom n 3980000 0.80000 3:0-va 009805 comma—00 N 0003000 0:58.009: «00E H 91 VIII. References 10. ll. 12. 13. 14. 15. 16. Smith, R D.; Olivares, J. A.; Nguyen, N. T.; Udseth, H. R. Anal. Chem. 1988, 60, 436 Aebersold, R.; Bures, E. J.; Namchuk, M.; Goghari, M. H.; Shusan, B.; Covey, 'l‘. C. Protein Science 1992, 1, 494-503 Yamashita, M.; Fenn, J. B. J. Phys. Chem. 1984, 88, 4451 Yamashita, M.; Fenn, J. B. J. Phys. Chem. 1984, 88, 4471 Olivares, J. A.; Nguyen, N. T.; Yonker, C. R.; Smith, R. D. Anal. Chem. 1987, 59, 1232 Bruins, A. P.; Covey, T. R.; Henion, J. D. Anal. Chem. 1987, 59, 2642. Chowhury, S. K.; Katta, V.; Chait, B. T. Rcmid Commun. Mass Spectrom. 1990, 4, 81 Allen, M. A.; Vestal, M. L. J. Am. Soc. Mass Spectrom. 1992, 3, 18 Dawson, P. H. Quadrupole Mass Spetrometiy and Its Applications. Elsevier, New York, 197 6 Dawson, P. H.; Yu, B. Int. J. Mass Spectrom. Ion Processes, 1984, 56, 25 Miller, P. E. Denton, M. B. J. Chem. Educat. 1986, 63, 617 Miiler, P. E. Denton, M. B. Int. J. Mass Spectrom. Ion Processes 1986, 72, 223 Bruins, A. P. Mass Spec. Rev. 1991, 10, 53-77 Anderson, I. B.; Andres, R. P.; Fenn, J. B. In "Advavced Atomic Molecular Physics” Bates, D. R.; Estermann, I., Eds; Academic Press, New York, 1965, Chapter 8 Camparque, R. J. Phys. Chem. 1984, 88, 4466-4474 Beijerinck, H. C. W.; van Gerwen, R. J. F.; Kerstel, E. R. T.; Martens, J. F. M.; van Vlembergen, E. J. W.; Smits, M. R. Th.; Kaasheok, G. H. Chem. Phys. 1985, 96, 153 17. 18. 19. 20. 21. 22. 23. 24. 25. 26. 27. 28. 29. 30. 31. 32. 92 Gray, A L. J. Anal. At. Spectrom. 1989, 4, 371 Seraukas, R. V.; Brown, G. R.; Pertel, R. Int. J. Mass. Spectrom. Ion. Phys. 1975, 16, 69-87 Alexander, A. J .; Kebarle, P. Anal. Chem. 1986, 58, 471 Arpino, P. Mass Spectrometry Rev. 1990, 9, 631 SIMION, Dahl, D. A.; Delmore, J. E. Idaho National Engineering Laboratory, Idaho Falls. O'Hanlon, J. F. A User Guide to Vacuum Technology, Wiley-Interscience, New York, 1989, pp 27 Fenn, J. B.; Mann, M.; Meng, C. K.; Wong, S. F.; Whitehouse, C. H. Mass Spectrometry Rev. 1990, 9, 37 Smith, R D.; Loo, J. A.; Loo, R. R. 0.; Busman, M.; Udseth, H. R Mass Spectrometry Rev. 1991, 10,359 Anderson, J. B.; Andres, R. P.; Fenn, J. B. Adv. Chem. Phys. 1966, 10, 275 L00, J. A.; Udseth, H. R.; Smith, R. D. Anal. Biochem. 1989, 179, 404 ‘ Loo, J. A.; Udseth, H. R.; Smith, R. D. Rapid Commun. Mass Spectrom. 1988, 2, 207 CIO-DIO user's manual, Rev. 3.0, Computer Boards, Inc., Mansfield, MA Yost, R A.; Enke, C. G. Anal. Chem. 1979, 51, 1251A Yost, R. A.; Enke, C. G. McGilvery, D. C.; Smith, D.; Morrison, J. D. Int. J. Mass Spectrom. Ion Phys. 1979, 30, 127 Vincenti, M.; Schwartz, J. C.; Cooks, R. G.; Wade, A. P.; Enke, C. G. Org. Mass Spectrom. 1988, 23, 579 Schwartz, J. C.; Wade, A. P.; Enke, C. G.; Cooks, R. G. Anal. Chem. 1990, 62, 1809 CHAPTER 3 PEPTIDE SEQUEN CIN G I. INTRODUCTION Many proteins of biOlogical interest are often only available in sub-picomole quantities, and isolation and purification techniques reduce this amount further. Consequently, there is a demand for an increase in sensitivity of current sequencing methods or for developing new sequencing techniques. Several chemical methods have been developed for determining the sequences of proteins and peptides. All involve sequential degradation of the molecule fiom one end or the other, removing and identifying the amino acids one at a time. Sequencing peptides from the C-terminal is possible by a variety of chemical methods (1). All suffer from poor yields, which limit the number of residues that can be determined and most involve complex, multistep chemistry. Recent improvements in the thiohydantoin method (2) show promise for making C-terminal sequencing more practical. More often carboxypeptidases are used to remove amino acids one at a time fi'om the C- terminus. The released amino acids are then identified. The enzymatic approach to C- terminal sequencing sufi‘ers fiom varying rates of hydrolysis for different amino acid, making its use less than ideal in practice. The most accepted N-tenninal sequencing method is that first proposed by Edman (3). This approach involves reaction of the amino-terminus of the peptide with phenylisothiocyanate to form phenylthiocarbamayl derivative. The N-tenninal amino acid 93 94 residue is then cleaved, converted to the more stable phenylthiohydantoin, identified, and quantitated. The remaining peptide is put through subsequent cycles of degradation. Although amino terminal sequencing is sensitive and, as a result of automation, can be left unattended for long periods of time, the process is relatively slow, normally taking about lb per residue. The most critical limitation of the Edman sequencing of peptides and proteins is the unavailability of a free amino terminus for coupling in N-terminal blocked peptides and proteins. Microsequencing can yield uncertainty in certain residue assignments, especially when the quantity is small or mixtures contribute overlapping signals. As sequencing extends towards the C-terminus of a peptide, residue signals drop off gradually owing to imperfect yields of the repetitive chemical and physical process. Many post-translational modifications are not readily identified by Edman chemistry methods because of the harsh conditions employed. The chromatographic identification of the modified PTH-amino acids could be misassigned as another amino acid with identical retention time, or simply taken as an unknown residue, especially when low levels are sequenced. When a mass spectrometer is used for detection in a peptide sequanator, reliable identification of the thiohydantoin (TH) amino acids can be accomplished by their molecular weight information combined with HPLC retention times. A Edman Degradation The preferred procedure for determining the sequence of amino acids in a peptide has involved the use of Edman degradation chemistry first published in 1950 (3). The Edman procedure determines the sequence of amino acids in a peptide starting at the amino terminus. The methodology employs phenylisothiocyanate (PITC) that attaches to the amino terminus of the peptide as shown in Figure 3-1 to form the (phenylthiocarbamyl) FTC-peptide under alkaline Conditions. Treatment of the FTC-peptide with anhydrous trifluoroacetic acid (TFA) causes cleavage of the N-temiinal amino acid as a FTC-amino 95 acid in equilibrium with the anilinothiazolinone (ATZ) of the amino acid, which upon exposure to aqueous TFA is converted to the phenylthiohydantoin (PTH) derivative of the amino acid. The reaction solution is extracted under conditions to isolate the cyclized phenylthiohydantoin derivative of the amino acid. Identifying the cyclized amino acid PTH derivative by retention time on HPLC thus determines the amino acid at the amino termimrs of the original peptide. The cycle is then repeated to determine the second amino acid fi'om the amino temrinus, etc. The validity of an analysis conducted with a conventional amino acid sequenator rests on the stability of a set of parameters that affect the HPLC chromatogram. That is, the conventional sequenator relies on a single channel of information, namely UV absorbance versus time as a means of ”identifying” various amino acid derivatives that are injected and eluted from an HPLC column automatically. Because the conventional automated sequenator ”identifies” an amino acid solely by retention time, any operational parameter that affects the elution profile or order of amino acids can seriously affect the validity of the analysis. For example, in a typical HPLC protocol, histidine and arginine are especially sensitive to ionic strength of the bufi‘er solution in the mobile phase and, to some extent, are influenced by the condition of the end-capping of the column (4). Thus, slight changes in the bufi'er solution from those used in analysis of the calibration sample or the gradual deterioration of the column can cause either one of these two amino acid derivatives to wander into time windows for other amino acids and give false-positive responses. Another operational problem associated with HPLC and the use of chemical reagents for derivatization prior to analysis by HPLC is the potential for interference from the breakdown products of the reagents or compounds associated with the sample. For example, one problem commonly encountered during low-level sequence analysis is the potential interference of phenylthiourea (a breakdown product fi'om exposure of the reagents to oxygen during sample application to the sequenator) which will co-elute with 96 tryptophan as the phenylthiohydantoin derivative (5). In this case, especially during the first four to five cycles in a given assay, it is possible for phenylthiourea to cause a false positive for tryptophan or to cause such a high chemical background that reliable detection of tryptophan is not possible. A third problem in the use of the conventional automated sequenator is evident in an analysis when any of the 20 common amino acids has been modified. In these circumstances, the modified amino acid as the phenylthiohydantoin derivative will elute at a non-standardized time window and, thus, will either be ”missed" or it will elute in some other time window and will be misidentified (6). The latter circumstance is frequently realized when hydroxyproline is present in the peptide; the PTH derivative of hydroxyproline typically has the same retention time as the PTH derivative of alanine or histidine, depending on the buffer . This is a common problem with sequenators as indicated in a recent survey by the American Biomolecular Resource Service Facilities, which showed that in a survey (7) of some 60 peptide facilities, 54% of them misidentified hydroxyproline as being histidine. Several factors limit the sensitivity of the Edman degradation procedure. The limit of detection for the phenylthiohydantoin derivative of the amino acid (usually by UV detection) during HPLC analysis establishes the minimum sample requirement for the Edman degradation procedure for amino acid sequencing. The most common means of detection is based on UV absorption of the phenylthiohydantoin derivative in a flow- through detector at the exit of the HPLC instrument. Typically, five picomoles of the phenylthiohydantoin derivative of the amino acid can be detected by UV absorption with an acceptable signal-to-background ratio (8). Background in the HPLC eflluent either fi'om biological sources or from chemical sources plays a significant role in the detection limit issue. Under extraordinary conditions of using ultra-clean solvents and careful cleaning of HPLC columns, it has been possible to achieve detection limits closer to 200 fmols (9) with UV detection ,of products from the Edman degradation procedure. 97 R1 0 R2 0 R.... ll N=C =8 HzN—CH—C—NH—CH—C—NH—CH Coupling 1p}! 8-10 H R] o a; o a... l I II I ll N—C—NH — CH —c—'—-NH—CH—c —'NH — CH l Anhydrous J CF3COOH Cleavage R2 0 R3 0 K... NH \c’N\ l l I II | \ /CH-—R1 HzN—CH —C—NH—CH—C —NH — cu S -—C O l CH3COOH/H20 Conversion 0 l / N \ CH—Rr PTH-AA Ll. / s Figure 3-1. Chemical Scheme for Edman Degradation of Peptide . 98 A third factor limiting the sample size requirement in the Edman degradation procedure is the cumulative efi‘ect of successive losses. (10-12) suffered by the residual peptide as one amino acid after another is cleaved away and the peptide is repeatedly exposed to extraction conditions for isolating the phenylthiohydantoin derivative of the newly cleaved amino acid. II. Analysis of Thiohydantoin Amino Acids by Mass Spectrometry There have been several detection methods employed to increase the sensitivity and the reliability of PTH detection. Mass spectrometry has been used to analyze PTH amino acids reliably and accurately. The mass spectrometric techniques were able to detect PTH derivatives successfully, but earlier investigators could not achieve the desired sensitivity required for peptide sequencing. In this chapter, the use of mass spectrometry in pm detection and peptide sequencing will be summarized . ' A. PTH Analysis by Electron Impact (E1) Mass Spectrometry Pisano and Homing have separated the PTH and DNP derivatives of thirteen amino acids by gas chromatography (13). Melvas separated five PTH amino acids by gas chromatography and detected them by EI mass spectrometry (14). He used the PTH amino acids of ala, pro, val, leu, and ile as model compounds. He was able to observe very strong molecular ion peaks as well as extensive fragrnentations for the five PTH amino acids. Fairwell and Lovins (15) studied the suitability of ATZ amino acids for the detection of the end product of Edman degradation chemistry. They compared the EI mass spectra of several ATZ amino acids which are especially very fragile derivatives with those of corresponding PTH amino acids. They observed that both ATZ and PTH amino acids have very similar mass spectra. They demonstrated the ability to use ATZ amino 99 acids for mass spectral detection by sequencing the first five residues of Bovine Ribonuclease. They concluded that during sample introduction to the mass spectrometer through the direct inlet probe, ATZ amino acids spontaneously undergo thermal rearrangement to the more stable corresponding PTH amino acid. Sun and Lovins (16) studied the methyl thiohydantoin (MTH) and PTH derivatives by EI mass spectrometry with low ionizing voltages. They obtained mass spectra of eighteen MTH derivatives and thirteen er derivatives with ionizing voltages of 70eV (usual. ionizing voltage), 20eV and 11eV. It could be readily observed that the spectra using 11eV energy are much less complex than those obtained at higher electron voltages. At the lower electron energies most if not all of the fi'agment ions begin to disappear and the spectra contain in a number of cases only the molecular ion. With the exception of arg, lys, and thr, the mass spectra of all derivatives investigated contained the molecular ion as the base peak of the spectrum with only one or two and in some cases no fiagment iom of significant intensity. The spectra of PTH derivatives of amino acids: arg, lys, and thr gave an abundant fiagment ion and a small number of less abundant fragment ions in their individual spectra at low ionizing voltages. They recorded the abundance of some ions with the ionizing voltages and observed that the maximum sensitivity was observed at about 20eV electron energy. The complexity of spectra greatly decreased at below 20eV, but the sensitivity also decreased. B. PTH Analysis by Chemical Ionization (CD-Mass Spectrometry Fales et al. have analyzed PTH amino acids by CI/MS in order to achieve the high sensitivity required by the Edman degradation (1?). The protonated molecule appeared as the major peak in the CI spectrum in 16 PTH amino acids. The PTH derivatives of arg, lys, cys, and asp gave fragment ions as the major peak in' the spectrum. This indicates the stability of 16 PTH amino acids under the CI conditions. However under the CI 100 conditions, PTH-arg decomposes at high temperature (at 220°C), lys forms an unstable 8- phenylthiocarbamyl PTH, PTH-cys undergoes extensive fragrnentations, and PTH-asp loses CH202. The loss of CH202 can be observed with PTH-asp and PTH-glu that is uncommon with the Other carboxyl amino acids under the CI conditions. Such an efi‘ect had been observed previously with free amino acids under the CI conditions (18, 19). These researchers compared the sensitivity of the PTH amino acids with E1 and CI mass spectrometry and they reported that Cl is 100 times more sensitive than El because of fewer fi'agmentations under CI conditions (20). C. PTH-Amino Acid Analysis by TwooStep Laser Desorption/Multiphoton Ionization Engelke, Hahn, Henke and Zare reported their work on PTH-amino acid analysis using two-step laser desorption/multiphoton ionization (21). In this experiment, the sample was loaded onto a glass cup and spread out as a thin film over the inner surface. The thickness of the sample varied fiom hundreds of monolayers to submonolayers. One monolayer of the sample molecules on the glass substrate corresponds to about 10 picomoles in the desorbing C02 laser area of 0.01cm2. After the sample is introduced into the vacuum system, as a first step, a C02 laser beam is directed to the thin film of the sample. Neutral molecules are generated in a fast desorption process and expand into the high vacuum between two electrodes that form the acceleration region of a simple linear TOF mass spectrometer. In the second step, these molecules are ionized by resonance-enhanced multiphoton ionization (REMPI) with an UV laser (NszAG laser at 266 nm). The main features of the spectra are: (l) a high yield of the molecular ion, (2) a small degree of fiagmentation, and (3) considerable reduction of background. They 1 0 1 demonstrated the simplicity of the method by analyzing an equimolar mixture of five PTH- arnino acids. D. PTH Analysis by Thermospray Mass Spectrometry Thermospray mass spectrometry (TS/MS) has been used to identify the PTH aminoacidsbyPramaniketat. (22). Intheirearlywork, theyacquiredTSmassspectra for 19 synthetic PTH-amino acids corresponding to those commonly observed during N- terminal amino acid sequencing. All the PTH amino acids including PTH(PTC) lys, in which the amino side chain additionally bears a PTC moiety, gave a prominent protonated molecule. Ions corresponding to the addition of ammonium ions (M+NH4+) were not observed. In most cases, a peak for the protonated molecule was the base peak in the spectrum, although for PTH-thr and PTH(PTC)-lys, one or more fiagrnent ions were always more abundant than the protonated molecule. Preliminary investigations of the sensitivity in the full scanning mode (150 - 410 u, entire mass range covering all the PTH amino acids), different quantities of standard PTH amino acids ranging fi'om 1 - 150 pmol per component were subjected to LC/MS. All the PTH amino acids could be identified down to the 50—pmol level. Six pm amino acids could be identified at the l-pmol level with S/N ratio 10:1 or better (22). TS/MS has been used to determine the composition of amino acids hydrolysates after reaction with PITC to form PTC-amino acid (23). All the PTC-amino acids except PTC-cys could be detected at 250-pmol level with S/N ratio 80:1. In the PTC-cys analysis 1 nmol was required, due to extensive fragmentation (23). These higher detection limits of PTC-amino acids may be due to more extensive fiagmentation than those of PTH- amino acids. l 02 E. PTH Analysis by Atmospheric Pressure Chemical Ionization Mass Spectrometry Sakari and Kambara (24) analyzed PTH amino acids using atmospheric pressure chemical ionization (APCI) mass spectrometry. Samples and solvent molecules were ionized by corona discharge and protonated molecules were observed with only a few fi’agment ions in APCI mass spectra. They were able to detect PTH amino acids at the 10- 100 pg level using the selected ion monitoring mode. F. Thiohydantoin Detection by ECNI/MS Several reagents were introduced to increase the electron capturing properties of the sequencing reagent in the Edman chemistry. The reagent pentafluorophenylisothiocyanate (PFPITC) was proposed as a sequencing reagent and the resulting PTH derivatives were analyzed by gas chromatography-electron capture detection (25), or gas chromatography-negative ion chemical ionization mass spectrometry (NICI-MS) (26, 27). The PFPTH-asn amino acid was observed at the 155- frnol level with S/N ratio of 80 using NICI-MS. Andregg et al. (28) investigated the use of 4-nitro phenylisothiocyanate (NPITC) that was first introduced by Tarr (29) to detect the thiohydantoin amino acids by NICI-MS. They detected NPTH amino acids at the 50- frnol level using selected ion monitoring (28). G. Modified Thiohydantoin Detection by Pneumatically Assisted Electrospray Mass Spectrometry While we were pursuing this project to find modified Edman reagents for sensitive detection of thiohydantoin amino acids by electrospray mass spectrometry, Aebersold et 103 al. introduced a new reagent for Edman degradation (30). The new reagent carries a quaternary ammonium moiety separated by two methylene groups from the conventional Edman reagent, PITC. They demonstrated detection limits for the thiohydantoin amino acids at the attomol level. The reagent shows comparable coupling and cleavage rates to that of PITC. Because of the quaternary group of the new reagent, it has a very polar nature. This reagent is compatible with use only in a solid phase peptide sequenator. The recently developed methods for solid phase sequencing has reduced the sample requirement to picomole or subpicomole quantities (31-34). The advantages of very low detection limits may not be obtained successfully with solid phase peptide sequenators. Recently Basic et al. (3 5) introduced another reagent for Edman sequencing followed by ESI/MS detection. This reagent features a N, Nedimethyl aminopropyl group attached to the isothiocyanate firnctionality. III. Other Modifications to Detect Thiohydantoin Amino Acids A Fluorescent and Colored Derivatives PITC is by far the most important reagent for N-terminal sequential degradation, but the detection of PTH amino acids at low levels is difficult. Several fluorescent reagents were developed to increase the detectability of TH amino acids. On the whole the claimed advantages, mainly the ease of detection of the TH derivatives, are outweighed by disadvantages such as low solubility in the coupling buffer, low reaction yields, chemical stability and background contamination. Therefore, these reagents failed to translate improved detectability into improved sequencing sensitivity. In this section, some important reagents are briefly discussed. 104 l.'DABIT C (dimethylaminoazobenzene-isothiocyanate) Methods The sequencing reagent DABITC has been widely applied for manual liquid and solid phase rnicrosequencing (3 6). The reagent was described by Chang and Creaser in 1976 (3 7) and has several advantages over the usual degradation with PITC; firstly, it releases the thiohydantoins as red-colored derivatives which are visible to the eye in picomole quantities on polyarnide sheets; and secondly, the side product of the Edman chemistry forms blue-colored compounds whose color easily difi’erentiates them fiom the red color thiohydantoin amino acids. ' However, the coupling yield is lower than that of the PITC, due to the bulky side group of the DABITC reagent. Therefore, a second coupling step with the PITC was introduced to complete the reaction (38). Since the introduction of this DABITC/PITC double coupling procedure this method has found wide application. The identification of TH amino acids can most quickly and simply be performed on a stamp-sized polyamide thin-layer sheet (3 9). HPLC systems for additional confirmation of TH amino acids and quatitation have been developed (40-42). 2. Dansylamino-PITC Method Jin et al. (43) introduced a new Edman reagent, Dansylamino-PITC, having the dansylarnino functionality. The dansylarnino pm amino acids were detected at the 1-5 pmol level on TLC and at the 200-fmol level by HPLC (44). The dansylamino-PITC carries a bulky naphthyl ring and gives a coupling yield about 75-95% (44). However, the coupling with dansylamino-PITC is not complete even if the coupling reaction is performed for 60 min at 52°C (44). The increase in coupling time causes an increase in the by-products. Hirano and Wittmann-Liebold (45) improved the method by incorporating a PITC coupling step after the coupling with dansylamino-PITC, but before 105 the cleavage. The resulting dansylamino-PTH amino acids were detected on TLC at 100- pmol level. B. Radioactive Derivatives The use of a radioactively labeled coupling reagent in the Edman degradation was first reported by Cherbuliez (47) and by Laver (48) who used [358] phenylisothiocyanate in the end group determination of various proteins. Although a detection limit of 0.1nmol was obtained, sequential analysis of protein was not attempted. Laursen (49), in 1969, reported the use of 3H-labeled phenylisothiocyanate in a solid-phase sequenator. [14C] PITC was introduced by Silver and Hood (50) in conjunction with an automated sequenator. Jacobs and Niall (51), used 35S-labeled PITC in- an automated peptide sequenator. The thiohydantoin amino acids which were generated at each cycle of the degradation were identified by TLC and autoradiography at the lO—pmol level. Tsugita et al. (52), described a method to form a radioactive derivative fi'om the intermediate obtained from the Edman degradation instead of the usual conversion to the more stable PTH amino acid. The ATZ amino acid was quantitatively reacted with [12511iodohistatnine to form a sensitized phenylthiocarbamyl derivative. The derivative was detectable at the finol level. Because the ATZ amino acid is used for the reaction, the kinetics of the reaction were not affected by this derivatization. Although these reagents improve the sensitivity compared with conventional PITC, radioactively labeled reagents undergo autoradiodegradation, resulting in decreasing product yields and increasing proportions of labeled by-products (52). Handling of highly radioactive materials is not convenient for routine analysis. 106 >~H~=~~Cs 4-N, Ndimethylaminoazobenzene-4'-isothiocyanate (DABITC) N ©© son‘s. 4-(N-1-dimethylarrinonaphthalene-5—sdponylamino) phenylisothiocyanate Figure 3-2. Structures of proposed fluorescent reagents 107 IV. Peptide and Protein Sequencing by Automated Sequenator The manual peptide sequencing by Edman degradation is a time and labor consuming process. In 1967, Edman and Begg (53) introduced the automated peptide sequenator based on Edman chemistry to improve the peptide sequencing methodology. Their sequenator was based on liquid-phase mobilization of peptide and called the spinning cup sequenator. In the spinning cup sequenator, short peptides tend to be washed out during the solvent extraction steps. Several research groups have improved this technique in order to overcome this difficulty. Another two types of sequenators, solid-phase sequenator (54,55), and gas-phase sequenator (56-58) were developed to reduce the possibility of loses due to the extraction solvents. The intention of this part is to provide a brief introduction of the development of automated peptide sequencing and current instrumentation available for this technique. Advantages of the liquid-phase sequenator includes the increased speed of sequencing compared to that of manual methods, a barrier for atmospheric 02 that decreases by-product formation, the effective extraction steps, and the improved sensitivity by precise extraction procedures and miniaturizing the reaction vessel. A. Liquid Phase Peptide (Spinning Cup) Sequenator The liquid-phase automated peptide sequenators are widely used in determining peptide sequence fiom the N-terminal of a peptide or protein by Edman degradation. A short account is given here on liquid-phase peptide sequencing. The description of automated methods were reviewed in detail by Nlall (51). . 108 A Edman and Begg (53) introduced the first automated peptide sequenator to perform peptide sequencing. This automated sequenator was able to carry out the coupling and cleavage steps independently as instructed by a computer. The spinning cup sequenator has encountered great difficulty making the required discrimination with short peptides, particularly hydrophobic ones, because the extraction solvents in combination with residual sequencing reagents cause the peptide to wash out of the cup. In order to overcome this problem, a volatile bufi‘er (trimethylamine) was introduce (59) to reduce the by-products and extensive extractions. Subsequently, several protein carriers were introduced (60, 61, 62). The major breakthrough came with the introduction of polybrene to spinning cup technology (61, 62). This substance efl‘ectiVely anchors small quantities of both proteins and peptides in the cup and allows sequencing of even short hydrophobic peptides to completion. Hunkapillar et al. (63) combined two important developments in sequencing chemistry, polybrene for non-covalent attachment and HPLC for PTH separation followed by a UV-Vls detector for identification. The instrumentation was fidrther improved by introducing a miniaturized glass disk as a sample support in place of a spinning cup (64). B-. Solid Phase Peptide Sequenator The solid phase approach to sequence analysis of proteins was introduced by Laursen in 1971 (54). The essence of the procedure is that proteins and peptides are covalently attached to an insoluble support. This is then placed in a glass column and the reagents for Edman degradation are introduced sequentially by an arrangement of pumps and valves as instructed by an electro-mechanical or electronic control device. A major advantage of this approach is that because the protein is covalently attached, by-products and excess reagents can be removed by solvent washes without concomitant risk of loss of sample. Several. resins have been developed to attach proteins in solid phase sequencing . 109 to improve extraction and sequencing emciency. They include porous glass beads, polystyrene resins, glass fiber sheets with activated isothiocyanate surface (65 -67), and polyvinyldene difluoride (PVDF) membranes (68). C. Gas Phase Peptide Sequenator In 1984, Hewick et al. (69) introduced a new sequenator which employed the coupling and cleavage reagents in gaseous phase rather than liquid phases. Only sufiicient PITC solution to completely wet the glass fiber disc is delivered (~20uL) to the sample holder. The heptane (solvent used to dissolve PITC) is removed by briefly flushing the cartridge with argon. The coupling stage is effected by slowly bleeding TMA/HZO vapor through the cartridge to the waste. Likewise, cleavage is effected by slowly bleeding trifluoroacetic acid vapor through the cartridge to the waste. The advantages of the new miniaturized gas-liquid sequenator includes: high sensitivity sequencing, very low reagent and solvent consumption, a much shorter degradative cycle, and low maintenance and running costs (69). V. Mass Spectrometric Methods to Determine the Sequence of Peptides and Proteins Mass spectrometric methods are gaining wide acceptance in peptide sequencing in recent years. The Edman degradation chemistry cannot be used when the N-terminus of the peptide is blocked. Mass spectrometry plays a significant roll in sequencing such peptides. In this part, three major mass spectrometric techniques to get sequence information will be discussed. l 10 A. Fast Atom Bombardment Mass Spectrometry The development of fast atom bombardment (FAB) ionization technique along with mass spectrometry provides a method of ionizing non-volatile samples. This development created growth in peptide sequencing using mass spectrometry (70). FAB/MS was used to obtain molecular weight information (71-74) and tandem mass spectrometry (MS/MS) were used to obtain fragrnentations of selected peptide ions. The mass spectrometry is a very useful technique to get peptide sequence information of N-terminal blocked peptides which cannot be sequenced by Edman degradation. During FAB/MS, energy deposited on a peptide molecule is usually not enough to induce complete fiagmentation pattern necessary to deduce the sequence. Collisionally induced dissociation has been used to enhance the fi'agmentation and it provides several advantages: all fiagment ion peaks are directly related to the peptide structure or the selected ion, matrix peaks are excluded fi'om the spectrum, and mixtures can be analyzed without purification. The amino acid sequence can be deduced by interpreting the fi'agment ion peaks formed by the (M-i-H)+ of a peptide. There are three different bonds that can fragment along the backbone of a peptide. The first nomenclature for peptide fragmentation (Figure 3-3) was proposed by Roepstorfl‘ and Fohlmann (75). The letters A, B, and C denote the N-terminal fiagment (ions that contain N-terminus) while X, Y, and Z represent carboxyl terminal fragments (ions that contain C-terminus). The assumed structures are shown in Figure 3-4. Since then, the nomenclature has undergone revisions (76). Biemann used lower case letters to distinguish from the single letter code amino acids, irnmonium ions, or the loss of side chain residues from (M+H)+ ions. There are two types of strategies are used to induce fiagmentation, low-energy and high-energy collisionally induced dissociation. This distinction is based on the kinetic . 1 11 energies of the selected beam, being either in the order of 50V for quadrupole instruments, or several kilo-volts in the sector instruments- The low-energy experiment deposited lower internal energy (estimated to be 2-3 eV) deposited into the selected beam, thereby, resulting in the induction of the fi'agmentation in the peptide backbone. In the high-energy experiments (estimated to be lO-l 5eV deposited), fragmentation in amino acid side chains and other moieties, and an array of irnmonium ions were observed in addition to the peptide backbone fiagmentation (77). Derivatization schemes have been developed to increase the fragmentation efl'lciency and to control the fragmentation patterns during ionization. There are two types of derivatives have been prepared for this purpose, charged or hydrophorbic uncharged derivatives. A permanent charge has been deposited on the peptide molecule to obtain complete sequence information. The derivatization reactions include ethyltriphenylphosphonium reagents for N- and C-termini (7 8-80), and a quaternary ammonium moieties for N terminus (81-83). An on-line HPLC technique called "continuous flow FAB/MS" has been developed for FAB mass spectrometric analysis (84, 85). Continuous flow FAB/MS has two advantages with respect to the conventional FAB, suppression of matrix background ions (86) and reduction of competitive ionization that is due to hydrophobicity difl‘erences in peptide mixtures (87). B. Electrospray Mass Spectrometry Tandem mass spectrometry combined with ESI has proven to be a usefill technique to obtain sequence data of proteins and peptides. Several research groups discussed tandem mass spectral data obtained from low-energy CID studies (88). High-energy CID studies were also reported (89). 112 X3 Y3 Z. X2 Y2 Z. X. Y. Z [— F — '— — — T F— i— i‘ ‘3. It“ u i3 ‘3. i‘ ii HgN—C--C--N--C- C- III-'04 C--N—-C-C—OH H H H H H H H HERRERA—15.5... Figure 3-3. Fragmentation of the peptide backbone and nomenclature proposed by Roepstorfi‘ and Fohlmann. 113 N —terminal Ions + 0,: H—(NH—CHR—C0).- t—NH=CHR. or H. CR;R: H—ENH—CHR—coi‘.-.—NH—CH H. R, a, + l: H—lNH-CHR—COJPt—NH—CH- b,: H—(NH—CHR—CO).-.—NH—CHR.—-C-=—O‘ H. c,: H-TNH—CHR—CO)‘.—NH: a H' CR. . r— 1 u d,. H—(NH—CHR—CO).-t—NH-—CH C-terminal Ion Types l H. 0,: HN=CH—CO—(NH—CHR—CO).-.—OH CR... H’ 0,: CH—CO—lNH—CHR—COJM—OH x,: ‘OEC—NH—CHR.—CO—(NH—CHR——CO)..-t—OH or H. 0=c=N—'CHR.—co—(NH—CHR—CO)‘.-.—0H H. y,: H-(‘FH—CHR—coil—on "O y. — 2: 'HN=CR,—co—(NH—CHR—c0)‘.-.—0H can: H. 2,: CH—CO—lNH—CHR—COIH-OH HO 2, + l: -CHR,—co-+'NH—CHR—c0i‘.-.—0H Figure 3-4. Structures of the common ions encountered in FAB-CAD-MS/MS and their nomenclature as revised by Biemann. l 14 Fragmentation of the peptide backbone was induced by increasing the skimmer voltage of an ESI source on a single quadrupole mass spectrometer (90). Another method to induce the fi’agmentation on the peptide backbone involves increasing the temperature of the metal capillary interface (91). The authors induced thermal fiagmentation of large multiply charged ions (melittin and myoglobin). The spectra were comprised of multiple charged fiagrnent ions. Microseparation techniques are always very valuable in the biological sample purification and analysis. On-line separation techniques with the mass spectrometry provide the sensitivity and speed necessary to characterize peptides and proteins. HPLC/ESI/MS/MS has been used to sequence peptide mixtures (tryptic digests). Numerous examples were given to illustrate the use of mass spectrometry to sequence blocked peptides, define N- and C- terminal heterogeneity, locate and correct errors in DNA- and cDNA- deduced protein sequences, and identify sites of modifications such as deamidation, isoaspartyl formation, phosphorylation, oxidation, disulfide bond formation, and glycosylation (88, 92). C. Matrix Assisted Laser Desorption Mass Spectrometry Matrix assisted laser desorption ionization (MALDI) mass spectrometry is a soft ionization technique that can be used to determine the accurate molecular weights of peptides and proteins. Because of the softness of the technique, only singly charged or doubly charged ions are frequently observed in the MALDI mass spectra of proteins. Structural characterization of proteins cannot be done on MALDI due to the lack of fragmentation. A tandem time-of-flight mass spectrometer is still under development (93- 95). l 15 Spengler et al. (96, 97) studied the metastable decay of laser-desorbed ions in the first field-flee drift region of a reflectron time-of-flight mass spectrometer. Fragment ions fiom metastable decay were mass analyzed and a complete structural analysis has been performed for several peptides and proteins. The postsource decay fi'agments of a protein (M.W. = 12500) were compared with the ions generated by a tandem four sector mass spectrometer (98). 1. Peptide Ladder by MALDI ‘ Chait and co-workers (99) have described a method to sequence a protein by using multiple steps of wet chemistry with a final, single-step mass spectrometric read out of the amino acid sequence. A sequence defining a concatenated set of peptide fragments, each difi‘ering from the next by a single residue, is chemically generated in a controlled fashion. This was achieved by carrying out rapid step-wise Edman degradation in the presence of a small amount of terminating reagent, phenyl thiocyanate. A small proportion of peptide chain blocked at the amino terminus is generated at each cycle. A predetermined number of cycles is performed without intermediate separation or analysis of the released amino acid derivatives. The resulting mixture is read out in a single operation by matrix-assisted laser desorption mass spectrometry (MALDI). The mass spectrum contains molecule ions corresponding to each terminated polypeptide species present. The mass differences between consecutive peaks each correspond to an amino acid residue, and their order of occurrence in the data set defines the sequence of amino acids in the original peptide chain. 116 VI. List of References 10. 11. 12. 13. 14. 15. 16. 17. Inglis, A. S. Anal. Biochem. 1991, 195, 183 Bailey, J. M.; Nikfarjam, F.; Shenoy, N. R; Shively, J. E. Protein Science, 1992, 1, 1622 Edman, P., Acta. Chem. Scand. 1956, 10: 761, J6mvall, H.; HOOg, J .-O.; Gustavsson, A. -M. Methods in Protein Sequence Analysis, Birkhuser, Basel, 1991 Burdon, R. H.; Kmippenberg, P. H. Van (Eds), Laboratory Techniques in Biochemistry and Molecular Biology, 1989, Elsevier, New York, 295 Mische, S., et. al. (Eds), Techniques in Protoin Chonustry IV, 1993, 453 Calacay, J., Rusnak, M., and Shively, J.E., Anal. Biochem. 1991, 192: 23 Joe Leykam, Manager of MSU Peptide Facility, personal communication Walker, J.E., Feamley, I.M., and Blows, R.A, J. Biochem. 1986, 237, 73 Hunkapiller, M., Kent, 8., Caruthers, M., Dreyer, W., Firca, J., Gimn, C ., Horvath, S., Hunkapiller, T., Tempst, P., and Hood, L., Nature 1984, 310, 105 Vandekerckhove, J ., Bauw, G., Puype, M., Van Darnme, J ., and Van Montagu, M., in onooooo Methodo in Protein Mimmoengo Analysio; B. Wittmann- Liebold, J. Salnikov, and V. Erdmann, eds., Springer-Verlag, Berlin, 1986,179 Pisano, J. J.; VandenHeuvel, W. J. A.; Homing, E. C. Biochem. Biophys. Res. Commun. 1962, 7, 82 Melvas, B. W. Acta. Chem. Scand 1969, 23 (No. 5) 1679 Fairwell, T.; Lovins, R. E. Biochem. Biophys. Res. Commun. 1971, 43 (No. 6), 1280 Sun, T.; Lovins, R. E. Anal. Biochem. 1972, 45, 176 Fales, H. M.; Nagai, Y.; Milne, W. A.; Brewer, H. B.; Bronzert, T. J. Jr.; Pisano, J. J ;Anal. Biochem. 1971, 43, 288 18. 19. 20. 21. 22. 23. 24. 25. 26. 27. 28. 29. 30. 31. 32. 33. 34. Richards, F. F.; Barnes, W. T.; Lolviri7s, R E.; Salomone, R; Waterfeild, M. D. Nature 1969, 221, 1241 Milane, G. W. A; Axenrod, T.; Fales, H. M.; J. Am. Chem. Soc. 1970, 97, 5170 Pisano J. J, Bronzert T. J J. Biol. Chem. 1969, 244, 5597 Engelke, F.; Hahn,J. H.; Henke, W.; Zare ,R NAnal. Chem. 1987, 59, 909 Pramanik, B. C.; Hinton, S. M.; Millington, D. S.; Dourdeville, T. A; Slaughter, C. A. Anal. Biochem. 1988, 175, 305 Pramanik, B. C.; Moomaw, C. R; Evans, C. T.; Cohen, S. A; Slaughter, C. A. Anal. Biochem. 1989, 176, 269 Sakari. M.; Kambara. H. Anal. Chem. 1988, 60, 774 Lequin, R. M.; Niall, H. D. Biochem. Biophys. Acta. 1972, 252, 76 Tomqvist, M.; Mowrer, J .; Jensen, S.; Ehrenberg, L Anal. Biochem. 1986, 154, 255 Gosh, A; Andregg, R. Proceedings of the 37th ASMS Conference on Mass Spectrometry and Allied Topics, Miami Beach, FL, May 21 - 26, 1989, 917 Waidanatha, S.; Andregg, R. J. Proceedings of the 37th ASMS Conference on Mass Spectrometry and Allied Topics, Miami Beach, FL, May 21- 26, 1989, 881 Tarr, G ”Methods of Protein Microcharacterization" Ed: Shiverly, J 1986 Humana Press, Clifton, N. J. pp 156 Aebersold, R; Bures, E. J.; Namchuk, M.; Goghari, M. H.; Shusan, B.; Covey, T. C. Protein Science 1992, I, 494-503 Aebersold, R; Pipes, G.; Nika, H.; Hood, L. E.; Kent, S. B. H., Biochemistry ' 1988, 27, 6860 Pappin, D. J. C.; Coull, J. M.; Koester, H. In Current Research in Protein Chemistry (Villafranca, J. ;Ed) pp 191-202 Academic Press Sandiego Aebersold, R; Pipes, G.; Wettenhall, R. E. H.; Nika, H.; Hood, L. E. Anal. Biochem. 1990 187, 56 Coull, J. M.; Papin, D. J. C.; Mark. J.; Aebersold, R;Koester, H. Anal. Biochem. 1991 I94, 110 35. 36. 37. 38. 39. 40. 41. 42. 43. 45. 46. 47. 48. 49. 50. 51. 52. 53. 1 18 Basic, C.; Bailey, J. M.; Shively, J. E.; Lee, T. D. Proceedings of the 41st ASMS Conference on Mass Spectrometry and Allied Topics, San Francisco, CA, May 31- June 4, 1993 “frttman-Liebold, B.; Kimura, M. 1984 in Methods in Molecular Biology, vol 1, Walker, J. M. ed; Humana press, Clifton, NJ, pp 221 Chang, J. Y.; Creaser, E. H.; Bently, K. W. Biochem. J. 1976, 153, 607 Chang, J. Y.; Brauer, D,; Wittman-Liebold, B.; FEBS Lett. 1978, 93, 205 Chang, J. Y.; Creaser, E. H. J. Chromtogr. 1977, 132, 303 Lehmann, A; Wittman-Liebold, B. FEBS Lett. 1984, 176, 360 Chang, J. Y.; Lehmann, A; Wittman-Liebold, B. Anal. Biochem. 1980, 102, 380 Prieto, J. A,; Collar, C.; de Barber, C. B. J. Chromatogr. Sci. 1991, 29, 205 Tsuga, A.;Kamo, M.; Jone, C.; Shikarna, N. J. Biochem. 1989, 106 60 Jill, S-W.; Chen, G-X.; Palacz, Z.; Wittmann-Liebold, B. FEBS Lett. 1986, 198, 150' Salnikov, J .;Palacz, Z.; Wittmann-Liebold, B. in Methods in Protein Sequence Analysis, (Walsh ed;), Humana Press, Crifion, NJ, 247 Hirano, H.; Wittmann-Liebold, B.; Tsukuba, 1.; in Methods in Protein Sequence Analysis, (VVlttmann-Liebold, B ed;), Springer-Verlag Berlin, 1989, 42 Cherbuliez, E.; Sussman, A. R; Rabinowitz, J. Helv. Chem. Acta. 1966, 44, 319 Laver, W. G.; Biochem. Biophys. Acta. 1961, 53, 469 Laursen, R. A. Eur. J. Biochem. 1971, 20, 89 Silver, J.; Hood, L. Anal. Biochem. 1972, 67, 392 Jacobs, J. W.; Niall, H. D. J. Biol. Chem. 1975, 250, 3629 Tsuga, A; Arai, I.; Kamo, M.; Jone, C. J. Biochem. 1988, 103, 399 Edman, P.; Begg, G. EuropeanJ. Biochem. 1967, 1, 80 54. 55. 56. 57. 58. 59. 61. 62. 63. 65. 68. 69. 70. 71. 72. 73. 119 Laursen, R A. Eur. J. Biochem. 1971, 20, 89—102 Laursen, R. A; Bonner, A. G.; Horn, M. J. 1975 in Instrumentation in Amino Acid Sequence Detamination (Perharn, R. N. ed.) pp73-110, Academic Press, London. Hewick, R M.; Hunkapillar, M. W.; Hood, L. E.; Dreyer, W. J. J. Biol. Chem. 1981, 256, 7990 ' Hunkpiller, M. W.; Lujan, E.; Ostrander,F.; Hood, L. E. Methods Enzymol. 1983, 91, 227 Hunkpiller, M. W.; Hewick, R. M.; Dreyer, W. J .; Hood, L. E. Methods Enzymol. 1983, 91, 399 Hewick, R. M.; Hunkpiller, M. W.; Hood, L. E.; Dreyer, W. J., J. Biol. Chem. 1981, 256, 7990 ~ Tarr, G. E.; Beecher, J. E; Bell, M.; McKean, D. J. Anal. Biochem. 1978, 84, 622 Klapper, D. G.; Wilde, C. E.; Capra, J. D. Anal. Biochem. 1978, 85, 126 Hunkapillar, M. W.; Hood, L. E. Biochemistry, 1978, 17, 2124 Shively, J. E.; Miller, P.; Ronk, M. Anal. Biochem. 1987, 163, 517 Aebersold, R; Pipes, G.; Nika, H.; Hood, L. E.; Kent, S. B. H. Biochemistry, 1988, 27, 6860 Pappin, D. J. C.; Coull, J.; Koester, H. Anal. Biochem. 1990, 187, 10 Hewick, R M.; Hunkapillar, M. W.; Hood, L. E.; Dreyer, W. J. J. Biol. Chem. 1981, 256, 7990 Cotrell, J. S.; Greathead, R. J. Mass Spectrom. Rev. 1986, 5, 215 Barber, M.; Bordoli, R. S.; Sedgewick, R. D.; Tyler, A. N. J. Chem. Soc. Chem. Commun. 1981, 325 Barber, M.; Bordoli, R. S.; Sedgewick, R. D.; Tyler, A. N. Nature, 1981, 293, 270 Barber, M.; Bordoli, R. S.; Sedgewick, R. D.; Tyler, A. N.; Whalley, E. T. Biomed Mass Spectrom. 1981, 8, 337 74. 75. 76. 77. 78. 79. 80. 81. 82. 83. 85. 86. 87. 88. 89. 91. Morris, H. R; Panico. M.; Barber, MioBordoli, R S.; Sedgewick, R D.; Tyler, A. N. Biochem. Biophys. Res. Commun. 1981, 101, 623 Roepstofi‘, P.; Fohlman, J. Biomed Mass Spectrom. 1984, 11, 601 Biemann, K. Biomed Environ. Mass Spectrom. 1988, 16, 99 Burlingarne, A. L.; Baille, T. A; Russell, D. H. Anal. Chem. 1992, 64, 467R Wagner, D. S.; Salari, A; Gage, D.A; Leykam, J.; Fetter, J.; Hollingsworth, R, Watson, J. T. Biol. Mass Spectrom. 1991, 20, 419 Watson, J. T.; Wagner, D. S.; Chang, Y-S.; Strahler, J. R; Hanash, S. M.; Gage, D. A Int. J. Mass Spectrom. Ion Processes, 1991, 111, 191 Wagner, D. S.; Allison, J. Fragmentation of Peptides Cationed with Metal Ions; Proceedings of the 40th ASMS Conference on Mass Spectrometry and Allied Topics, Washington DC, May 31-June 5, 1992 Kidwell, D. A; Ross, M. M.; Colton, R. J. .I. Am. Chem. Soc. 1984, 106, 2219 Renner, D.; Spiteller, G. Biomed Environ. Mass Spectrom. 1986, 13, 405 Stults, J. T.; Lai, J .; McCune, S.; Wetzel, R. Anal. Chem. 1993, 65, 1703 Ito, Y.; Takeuchi, T.; Ishii, D.; Goto, M. J. Chromatogr. 1985, 346, 161 Caprioli, R. M.; Fan, T.; Cottrell, J. S.'Anal Chem. 1986, 58, 2949 Caprioli, R M.; Fan, T. Biochem. Biophys. Res. Commun. 1986, 141, 1058 Caprioli, R. M.; Moore, W. T.; Fan, T. Rapid Commun. Mass Spectrom. 1987, 1, 15 ' Carr, S. A; Hemling, M. E.; Bean, M. F.; Roberts, G. D. Anal Chem. 1991, 63, 2802 Murata, H.; Takao, T.; Shimonishi, Y.; Rapid Commun. Mass Spectrom. 1994, 8, 205 Katta, V.; Chodhury, S. K.; Chait, B. T. Anal. Chem. 1991, 63, 174 Rockwood, A L.; Busman, M.; Udseth, H. R; Smith, R. D. Rapid Commun. Mass Spectrom. 1991, 5, 582 92. 93. 94. 95. 96. 97. 98. 121 Michael, H.; Dickinson, T. A; Shabanowitz, J.; Davis, T. A; Cox, A. L.; Hunt, D. F. Proceedings of the 40th ASMS Conference on Mass Spectrometry and Allied Topics, Washington DC, May 31- June 5, 1992 Seeterlin, M. A; Vlasak, P. R; Beussmarl, D. J.; McLane, R. D.; Enke, C. G. .1. Am. Soc. Mass Spectrom. 1993, 4, 751 Cornish, T. J .; Cotter, R. J. Rapid Commun. Mass Spectrom. 1993, 7, 1037 Gardner, 13.; Enke, c. 6.; Watson, 1. T. Proceedings of the 44th ASMS Conference on Mass Spectrometry and Allied Topics, Chicago, IL , May 29-June 3, 1994 Spengler, B.; Kirsch, D.; Kaufrnann, R; Rapid Commun. Mass Spectrom. 1992, 6, 105 Spengler, 13.; Kirsch, D.; Kaufmanrl, R; J. Phys. Chem. 1992, 96, 9678 Spengler, B.; Kirsch, D.; Kaufrnann, R; Rapid Commun. Mass Spectrom. 1993, 7, 902 Chait, B. T.; Wang, R; Beavis, R. C.; Kent, S. B. H. Science, 1993, 262, 89 CHAPTER 4 Development of Edman Reagents for the ESI/MS Detection I. INTRODUCTION Protein sequencing is a highly sensitive technique that has been invaluable for providing critical primary structural data on isolated proteins. The isolation of low picomolar amounts of purified proteins and peptides has become a common occurrence, due largely to substantial advances in the techniques available for sample purification. As a result, there is an increased need for routine protein sequence analysis at low picomolar levels. The Edman sequencing chemistry remains the most popular method for the protein and peptide sequencing (1). During the Edman sequencing, the peptide is cleaved one amino at a time and the resulting thiohydantoin (TH) amino acids are detected by a UV detector after the separation by HPLC (see Figure 3-1). The identification of TH amino acids is done solely by using the retention time on the HPLC chromatogram. Recently, the Association of Biomolecular Resource Facilities (ABRF) Sequencing Committee evaluated the sequencing reliability among 75 facilities using an identical peptide containing two modified amino acids. The report says that identification of the post- translationally modified amino acids represents a challenge in sequence analysis (2). Mass spectrometry can be used to identify the TH amino acids by molecular weight that is more specific for the compound, rather than retention time. The previous attempts to use El (3), CI (4) and thermospray (TS) (5) mass spectrometry for the determination of thiohydantoin amino acid were not successful. These mass spectrometric techniques required a fairly high level of samples. Under these mass spectrometric conditions, the PTH amino acids undergo extensive fragrnentations resulting low 122 123 Aebersold et al. (6) described a modified reagent with very high sensitivity but due to its high polarity, a solid phase sequanator has to be used. Basic et al. (7) characterized a series of alkyl amine thiohydantoin derivatives by ESI/MS. Figures 4-lc and (1 show the structures of the reagents proposed by Aebersold et al (Figure 4-1a) and Basic et al. (Figure 4-1b) for ESI/MS. In order to improve the response of TH amino acids towards ESI mass spectrometry, a new class of modified TH amino acids was synthesized and characterized by mass spectrometry. The detectability of the modified amino acids was assessed on ESI/MS. ' 11. Alternative Edman Reagents The PTH amino acids were analyzed by ESI/MS. The detectability of PTH derivatives was poor in ESI/MS except for the amino acids containing basic side groups such as arginine and histidine. In order to detect all the amino acids at the same sensitivity, another fimctional group that is readily amenable to ESI/MS, must be introduced. In this context, three reagents were investigated. One reagent is t-BOC aminomethylphenylisothiocyanate (BAMPITC) that was introduced to improve the detectability of TH amino acids by. forming fluorescent derivatives after the HPLC separation. Another two reagents were proposed and their properties were investigated. The TH derivatives were characterized by mass spectrometry. A. t-BOC aminomethylphenylisothiocyanate (BAMPITC) 1. Introduction Reagent I, t-BOC aminomethylphenylisothiocyanate (BAMPITC) (Figure 4-2a) was developed for post column derivatization of the modified PTH amino acids for sensitive detection (8). This reagent carries a primary amino group blocked by a t-BOC 124 group and during the acid cleavage step of the sequencing process, a fi'ee primary anrino group is generated (Figure 4-2b). In the original process, the free amino group was subjected to fluorescent labeling at the time of detection to improve the detectability of the modified thiohydantoin amino acids. It was reported that the coupling rate is comparable to that of PITC. The authors claimed the repetitive yield is over 90% for the automated solid phase sequencing. i / C N \CH —R s/C—NH (a). Phenylisothiocyanate (PITC) (b). Phenylthiohydantoin (TH) amino acid /./NCS (CH3)3N+—CH2CH2 : (c). 3—(4'ethy1ene-N,N,N-trimethylamino)pheny1-2-isothiocyanate H3 /N —CH2 —CH2 —CH2 -NCS H3C (d). Dirnethylaminopropylisothiocyanate Figure 4-1. The structures of conventional Edman reagent (a), phenylthiohydantoin derivative (b), reagents proposed by Aebersold et al. (c), and Basic et al. (d). N—C — S i (CH3)3C— o— C-NH— c 2 (a). t-BOC aminomethylphenylisothiocyante i /C\ H—R H2N- CH21\ f /—NH S (b). Aminomethylphenylthiohydantoin amino acid Figure 4-2. Structures of t-BAMPITC and aminomethylphenylthiohydantoin amino acid 2. Experimental The TH amino acids were synthesized using the BAMPITC reagent in pyridine/water (1 : 1) and the pH was adjusted to 9 with triethylamine. The reaction mixture was incubated at 50°C for 30 minutes. The reaction mixture was evaporated to dryness and excess reagent and the by-products were extracted with 2-mL portions of toluene three times. The residue was dried and incubated at 50°C for 10 minutes with anhydrous TFA This cleaves the t-BOC group from the amino terminus as well as forms the unstable anilinothioazolinine (ATZ) amino acid. The ATZ-amino acid is converted to the stable TH amino acid with aqueous TFA (25%). 126 Analysis by ESI/MS was undertaken on the TSQ 700 triple quadrupole mass spectrometer fitted with a Finnigan ESI source. The sample was introduced at a flow rate of 4uL/min. The AMTH derivatives were analyzed by ESI/MS using CH3OH/HgO/HOAC (50/49.9/0.1 %v/v) as the solvent system. In all the experiments, needle was held at 4.5kV and the heated capillary at 200°C. 3. Results and Discussion Since the reagent generates a free amino group during the acid cleavage step (Figure 4-2b), it will improve the sensitivity of ESI/MS. Therefore, the detectability of modified TH amino acids were investigated using ESI/MS. The t-BOC group on the N atom cleaves when treated with anhydrous TFA and the cleavage is essentially complete in several minutes (9). Due to the free amino group on the TH derivative afier the acid cleavage, the AMTH amino acids could be observed as the protonated molecules. The TH derivatives of lys and arg were observed as doubly charged ions in the ESI/MS spectrum. All the other TH derivatives were observed as singly charged species. All the AMTH amino acids could be detected at the low femtomol level using ESI/MS. The TH amino acids with acidic residues are expected to give a low response in positive ESI/MS due to the -COOH group in the side chain because the -COOH group of the TH amino acid can be deprotonated in solution to form an electrically neutral species. But the asp- AMTH was observed at 200 finol level. Wittmann-Liebold et al. (9) discussed the synthesis and use of BAMPITC in microsequencing in detail. They compared the efficiency of manual sequencing and concluded that the repetitive yield was similar to that of PITC, but it needed longer reaction times. In the solid phase sequencer, the yield was about 48% for the second cycle 127 of the insulin B chain (oxidized). Since the reported repetitive yield was low for the automated sequencer, two new reagents were synthesized and characterized by mass spectrometry. B. Designing New Reagents In order to improve the ESI/MS response of TH amino acids, a basic or a pre- formed charged group must be introduced into the reagent. Aebersold et al. introduced a quartenary ammonium group to the PITC reagent. The reagent cannot be used in the liquid phase sequanator due to its high polarity. The work described here uses a pyridyl group as a basic residue to improve the ESI/MS response (Figure 4-3 a and b). Two new reagents were synthesized and characterized. The isothiocyanate group was separated hour the pyridyl ring in one reagent. Because of the pyridyl group, they are suitable for the liquid phase sequanator and the aromatic properties desired for the Edman reagent are retained. @N=C=S gab—N=C=S N N (a). Pyridylisothiocyanate (b). 3-Pyridylmethylisothiocyanate Figure 4-3. The modified Edman reagents. IH. Preparation of Isothiocyanate Several methods have been reported for the synthesis of isothiocynates. Among them, most reactions gave low reaction yields (25-7 5%) (10-13). Two methods have been selected to synthesize modified isothiocynates using their corresponding amines (14, 15). 128 These two methods gave reasonable reaction yields (over 90%) and relatively simple purification procedures. A Method 1. The method proposed by Makaiyama et al. was adapted (14) for the synthesis of isothiocyanates fiom the corresponding amine. Pyridylisothiocyanate was synthesized using pyridylarnine, C82, and 2-chloro-l-methylpyridinium iodide (Figure 4-4). In a round bottom flask, cooled by an ice-salt bath, 1.1 mmol of C82 and 2.1 mmol of triethylammine were placed. The mixture was stirred with a magnetic stirrer and aminopyridine ( 1 mmol) was slowly added to the cooled reaction mixture. The stirring was continued for thirty minutes after all the amine had been added, and then the reaction mixture was allowed to stand for another thirty minute. During this time a heavy precipitate of triethylammonuim pyridyldithiocarbarnate (Reaction 4-1) was formed. A solution of triethylanrine (111 mg, 1.1 mmol) in dichlromethane (4mL) was slowly added at room temperature under an argon atmosphere to a mixture of triethylaminium pyridylditiocarbamate (304 mg, 1 mmol) and 2-chloro- l -methylpyridinium iodide (281 mg, 1.1 mmol), and then the reaction mixture was stirred at room temperature for an additional two hours. After the removal of solvent, the residue was separated by the preparative thin layer chromatography or the column chromatography. The reaction yield was not optimized. 3-pyridylmethy1isothiocyanate was prepared using 3-aminomethyl pyridine by the same method. B. Method 11 In this method, isothiocyanates were synthesized by reacting an amine with a thiocarbanyl transfer reagent: di-2-pyridyl thiocarbonate (DPT). This method seems to be 1 29 simple and is easy to clean up after the reaction. In the first step, DPT was synthesized . and purified (Figure 4—5). At the second step the amine was reacted with DPT in CH2C12 at room temperature to get corresponding isothiocyanate (Figure 4-6) (15). 1. Preparation of DPT. In a round bottom flask surrounded by an ice bath, 2 mol of 2-hydroxypyridine and 2 mol of triethylamine were dissolved in 50 mL of CHzClz. 1 mol of thiophosgene was slowly added to the reaction mixture with stirring under a hood. The reaction mixture was stirred for another 2 hours at room temperature. The reaction mixture was neutralized with. an anion exchange resin and filtered through a Buchner flame]. The residue was washed with distilled water to remove the unreacted starting materials and triethylamine. The residue was recrystalized with CH2C12. 2. Preparation of Pyridylisothiocyanate using DPT 3-aminopylidine (0.1 mol) was dissolved in 25 mL of CH2C12, in a round bottom flask using a magnetic stirrer. The thiocarbamyl transfer reagent, DPT (0.1 mol) was added to the above mixture and stirred for about 2 hours. Figure 4-6 shows the reaction scheme involved in synthesis of isothiocyanate using DPT. The 2-hydroxypyridine was precipitated fi'om the reaction mixture by adding diethyl ether. About 80% of the 2- hydroxypyridine could be recovered by this method. The reaction mixture was filtered and dried over anhydrous Na2SO4 overnight. The filtrate was concentrated on a rotovap and separated on a preparative thin layer chromatographic plate. 13o R—NH2 + 052 + H31»: —» R—NHCSZ- l~:t3NH+ R—NHcsz' Et3NH+ + @\ c1 CH3 1 CH2CI2 ©S — C — NHR + Et3NH+ Y. x' II I s CH3 Et3NH+Y' + Q + R—N=C=S S xv=mml I one Figure 4-4. The reaction scheme of isothiocyanate formation by Makayarna Method 131 S .. +2©1 —-» (,1in /C\ "CH o/\ thiophogene hydroxypyfim di-Z-pyridyl thiocarbonate (DPT) Figure 4-5. Reaction scheme for the synthesis of DPT NH2 g @0/:\0/© Pyridyl anine DPT lance ©n=c=s + 2 @on Pyridyl lsotlnocyanate Figure 4-6. Scheme of isothiocyanate formation using DPT isothiocyanate transfer reagent. 132 C. Separation of Reaction Mixture (i). Preparative thin layer Chromatography (PTLC) The products synthesized by the methods 1 and H were analyzed by thin-layer chromatography (TLC) prior to separation of the mixture by preparative thin-layer chromatography (PTLC) or flash column chromatography (16). TLC separation was performed with Whatman® (Whatman paper Ltd; Maidstone, Kent, England) pro-made TLC plates that were coated with 250-um thickness of silica Gel and fluorescent indicator on flexible aluminum plates. The plates were cut into 2.5 x lO-cm strips and spotted by a micropipet with solution containing the product to be analyzed. The sample spot was 0.2 mm in diameter and made 1.0 cm above the bottom of the TLC plate. For sample development, the spotted plate was placed in a solvent chamber containing the mobile phase at a depth of about 0.5 cm. The plate was removed from the chamber when the mobile phase migrated to within approximately 1.0 cm of the top of the plate. The plate was allowed to air dry in a hood. When dry, the plate was observed under UV light. The position of the migrated dr0plets appeared as purple spots on the plate. The positions of the spots were marked and their Rf values were calculated. Several solvent combinations were investigated for optimum separation. The reaction mixture was applied onto a silica gel preparative thin layer chromatographic (PTLC) plate with fluorescent indicator (2-mm thickness, S-um particles, 20 x 20 cm) as a band at 1 cm from the bottom of the plate- It was kept under hood for several minutes to evaporate the solvent. The PTLC plate was developed in a closed tank with CHCl3/CH2C12 (50/50 v/v%) mixture. After the solvent front was 1 cm away fi'om the top of the plate, removed fi'orn the tank, and air dried under the hood. The plate was observed in a viewing chamber by fluorescence at 254 nm to identify the separated bands. The bands were marked, cut and collected for extraction. The silica gel fi'actions containing products were extracted by using CH2C12 as the solvent. The 133 presence of the reagent was confirmed by FAB-MS, EI-MS or the characteristic colorimetric reaction with the sodium azide and the dimethylforrnamide (17). 2. Column Chromatography Large scale preparative separations are traditionally carried out by tedious long column chromatography. Although the results are somewhat satisfactory, the technique is always time consuming and frequently gives poor recovery due to band tailing. In recent years, several preparative systems have evolved which reduce separation times to 1-3 hr and allow the resolution of components having 11sz 0.05 on analytical TLC (15, 17). Recently Skill et al. developed a substantially faster technique for the routine purification of reaction products and called the flash chromatography (16). Flash chromatography is basically an air pressure driven hybrid of medium pressure and short column chromatography that has been optimized for rapid separations. Optimal separation by flash chromatography of a mixture is accomplished in a reasonable length of time when the components have Rf value of about 0.35 and are separated by ARf value of 0.1. Separation of the products was performed with flash chromatography. A 2-inch diameter column with a stopcock and flow controller was chosen to carry out the separation. A small plug of glass wool was placed in the tube connecting the stopcock to the column body. A smooth 1/8-inch layer of 50-100 mesh sand is added to cover the bottom of the column and dry 40-63 um diameter silica gel (Sigma Chem. Co.; St. Louis, MO) was poured into the column in a single portion to give depth of about 6-inches. With the stopcock open the column was gently tapped vertically on the bench top to pack the silica gel. Next a 1/8-inch layer of sand was carefillly placed on the flat top of the dry gel bed. The column was filled with the 50:50 (v/v) CH2C12 : CHC13 solvent mixture that produced the TLC separation and pressure was applied to rapidly push all the air from the 134 silica gel. One gram of the reaction mixture dissolved in the solvent mixture was applied to the column and the flow controller was briefly placed on the column to push the sample into the silica gel. The column was refilled with solvent mixture and eluted at a flow rate of 2-inches per minute (approximately 75 mL/min). A rack containing forty 20 x 150 mm test tubes was used to collect the eluted fractions. Small fi'actions were collected at the start of the run and larger fractions were collected at the end of the run. Composition of the collected fractions was determined by TLC and the fractions containing the same compound were combined and concentrated by rotoevaporation. The presence of the isothiocyanate was confirmed by EI-MS, FAB-MS, and a characteristic color change with sodium azide and dimethylformarnide. 3. Results and Discussion Pyridyl and pyridylmethyl isothiocyanates were prepared by both methods. In the method 1, several reaction products were observed on the TLC plate. The Rfvalues of the isothiocyanate and the nearest compound were 0.52 and 0.44 respectively for the PMITC reaction mixture. On the PTLC plate, two bands were partially overlapped and bands have to be selected carefillly. About 80% of hydroxypyridine was precipitated using diethyl ether. This makes the separation less complex owing to less reaction products. Three major spots on the TLC plate were observed for the reaction mixture. D. Characterization of Isothiocyanates 1. Colorimetric Detection Mass spectrometric techniques can provide the molecular weight information and fragrnentations necessary to identify a particular compound. Mass spectrometry is not very good in distinguishing isomeric compounds. Therefore, a chemical method has to be 135 used to identify the correct firnctional group. The isothiocyanate (-NC 8) can exist in another isomeric form as thiocyanate (-CN S). Therefore, a chemical method was used to chemically characterize the isothiocyanate reagents. The isothiocyanate group of the reagent reacts with sodium azide in the presence of dirnethylforrnamide to evolve C02 gas and a blue (or green) colored solution (18). This is a characteristic chemical reaction for the presence of -NCS filnctional group in the molecule. 2. Mass Spectrometric Detection i. EI/MS EI mass spectrometry was carried out on a PIP-5980 MSD mass spectrometer. The samples were introduced through a gas-chromatographic inlet. The injector temperature was held at 200°C and the transfer line at 220°C. The temperature of the column was held at 100°C for 2 minutes and increased at a rate of 10°C/min to 250°C. The EI mass spectrum of pyridylisothiocyanate (M.W. = 136 Daltons) shows the molecular ion at m/z 136. The EI mass spectrum of 3-pylidylmethylisothiocyanate (PMITC) shows a peak at m/z 150 that corresponds to the molecular weight. An intense peak was observed at m/z 92 that represents a stable fragment C6H6N+ that has an aromatic character. ii. FAB/MS FAB mass spectrometric data were collected on a JOEL I-IX 110 double focusing high resolution mass spectrometer having EB geometry. The samples were ionized by a high energy Xe beam (6 keV). A portion of 2uL of sample was placed onto a FAB probe tip containing a viscous matrix usually glycerol. The probe with sample was introduced into the ion source chamber and evacuated for ionization. The ions were accelerated to 10 keV before going through the analyzers. The data were recorded on a DA 5000 data 136 system The reagent PyITC shows the protonated molecule at m/z 137 and 3-PMITC shows an intense peak at m/z 151 in their FAB mass spectra representing (M+H)+ ion. IV. Thiobydantoin amino acids A. Thiohydantoin Synthesis Representatives from each group of amino acids containing acidic, neutral, and basic side chains were selected to synthesize the thiohydantoin derivatives of amino acids. The following amino acids were selected as representative amino acids: asp, val, phe, leu, ile, gly, ala, ser, pro, asn, and lys. An amino acid (2 mmol each) was dissolved in H20/pyridine (1 : 1) mixture in two separate vials and the pH of the solutions was adjusted to 9 with triethylamine. The reaction mixtures were flushed with N2 gas for about 10 seconds. The PyITC reagent (2%) in acetonitrile (25 uL) was added to one vial containing the amino acid and coupling buffer. A portion of 25 uL of PMITC (2%) in acetonitrile was added to the other vial. The two vials were incubated for 30 minutes at 50°C for PyITC and at 60°C for PMITC. The solvent was evaporated in a Speedvac® and the excess reagent and by products were extracted with toluene. The dried residue was treated with anhydrous trifluoroacetic acid (TFA) to form anilinothioazolinone (ATZ) derivatives. The ATZ amino acids were converted to the more stable thiohydantoin (TH) amino acid by treating with 25% TFA at 50°C for 10 minutes. The resulting reaction mixture was injected onto a pre-equilibrated reversed phase HPLC system for separation. The TH derivatives were collected and dried under vacuum. The derivatives were stored under nitrogen at -20°C until use. B. Extraction of TH Derivatives The extraction of TH amino acids fi'om the reaction mixture was evaluated using FAB/MS. All the TH amino acids (PyTH and PMTH) were dissolved in ethyl acetate. In l 37 the presence of a water layer, the derivatives were observed in both layers and F AB/MS indicates that the solubility in water is greater than that in ethyl acetate. Afier the cleavage of ACP-PyTC and the ACP-PMTC (lOOpmol) with anhydrous TFA, dried residue was extracted with ethyl acetate and analyzed by ESI/MS. The ACP peptide has the sequence of VQAADYING. The TH derivative of val was observed in the extract. This indicates the possibility of extraction using ethyl acetate as the solvent. This extraction solvent is compatible with the liquid phase peptide sequanator. Therefore, both reagents are compatible with the liquid phase peptide sequenator. C. Mass Spectral Analysis of Thiohydantoins The PyTH and PMTH derivatives of amino acids were subjected to various mass spectrometric analysis techniques. Amino acids with non-polar side chains were studied by GC/MS using EI and CI mode of ionization. In the E1 mode, extensive fiagmentations were observed for all the derivatives with a reasonably intense peak for the molecular ion. In the CI mode, a major (M+H)+ was observed for all the non polar TH amino acids. The fi'agmentation was minimal for the TH derivatives of amino acids containing non-polar side chains. The derivatives with polar side chains were not analyzed by GC/MS. FAB/MS analysis was undertaken on the TH derivatives of both reagents. All the TH amino acids showed intense peaks for (M+H)+. FAB/CAD/MS/MS studies showed the fi'gmentations necessary to identify the TH amino acids in their mass spectra. Figure 4.7 shows the FAB/CAD/MS/MS mass spectrum of PMTH-val. 138 a b . +§1 +H ‘1 I” : ’1 IC : /Sl’ : @V “‘2' “-c a r‘ ~-- -- ,r O ’ / I’l’ HC__CH3 5 1 Fl CH3 A0 9 . I l “ 1 v C e 31 a 207 Q as . b u 2. n . [M+H-Val]+ d 1 1 a . n . c 1 e . 0 100 150 200 “320 Figure 4-7. FAB/CAD/MS/MS spectrum of val-PMTH. 1. Detection by ESI/MS The purified TH-amino acids were subjected to ESI/MS on a TSQ 700 TQMS fitted with a Finnigan ESI source. The capillary was heated to 200°C and the ESI needle was at 4kV. The nebulizing gas pressure was at 35 psi. The TH-amino acid sample was 139 introduced into the source through a flowing stream of flow at a rate of 100 ul/ min. The liquid system used was a mixture of CH3OH/HzO/HOAC (50/49.9/0.1%v/v/v). The total flow was directed to the ESI source without a splitter. Under the above conditions, ESI/MS mass spectra of TH-amino acids gave single peaks representing the (M+H)+- The basic nature of the pyridyl ring provides protonated molecules in solution. The ions are converted into the gas phase during the ESI process. The TH-amino acids did not show significant fi'agmentation under these conditions. Figure 4-8 shows the ESI/MS mass spectrum of val-PyTH (5pmol). The amino acids containing acidic, basic and neutral side chain groups show a similar response due to the pyridyl group in the Edman reagent. The TH derivative of lys shows the attachment of an additional PyNCS group to the side chain amino group, but it did not show a strong doubly charged ion in ESI/MS. The lack of fiagmentation improves the sensitivity for detecting the TH analogs by ESI/MS. This simplifies the detection technique for TH- amino acids for peptide sequencing. To evaluate the detection limits of the TH amino acids, increasing amounts of purified phe-PyTH were injected to a stream of CH3OH/H20/HOAC (50/49.9/0. 1% v/v/v) at a flow rate of lOOuL/min. Figure 4-9 shows the profile obtained for selected ion monitoring of the ion at m/z 284 (M+H)+. This shows the detection limit of phe-PyTH at the 5-finol level. Since the PMTH derivatives contain a pyridyl ring external to the thiohydantoin ring structure, it does not conjugate with the unsaturated functional groups in the TH ring. The pyridyl ring is fi'eely available for protonation during the ESI/MS process. The PMTH derivatives of amino acids gave only a single peak in the ESI/MS mass spectrum. Figure 4-10 shows the ESI/MS mass spectrum of PMTH val (5pmol). The PMTH derivatives of all the amino acids studied gave similar response during the ESI/MS. The I40 detectability of phe-PMI'H was evaluated on the selected ion monitoring mode. Figure 4- 11 shows the profile obtained for the replicate injections of the PM-thiohydantoin amino acid. The conditions were maintained as in the previous experiment. The phe-PMTH derivatives can be detected at the low femtomol level using the selected ion monitoring mode. (M+H) 236.0 100- 80* ON 0 Relative Intensity b O 204 100 150 200 250 300 350 450 m/z Figure 4-8. Portion of the ESI/MS mass spectrum of PyTH-val (5pmol). Sample was injected into a stream of CH3OH/HzO/HOAC (50/49.9/0.1 %v/v/v) at a flow rate of lOOuL/min. 141 500 finol 5 Pm°1 F1 40: "i E 0 0 % 20~ a: 50 fmol Sfmol wr'wvvv 200 "'466" 6256"" 366771666 tenai'i‘oarsoo scan # Figure 4-9. The mass spectral profile obtained by injecting increasing amounts of purified PyTH-phe derivative into a stream of CH3 OH/HzO/HOAC (50/49.9/0.1%v/v/v) at a flow rate of IOOuL/min. 142 + (M+H) 250 100 so .5 60- § .5 _':‘>’ 33 32 4o 20~ 100 150 200 250 300 350 400 m/z Figure 4-10. Portion of the ESI/MS mass spectrum of PMTH-val (5pmol). Sample was injected into a stream of CH3OI-I/H20/HOAC (50/49.9/0.1 %v/v/v) at a flow rate of lOOuL/min. 143 '1 T 50 pmol 40" .29 E .5 0 .2 L3 O :4 20‘ 5 pmol '5 fmol 50 frnol 500 fmol I ' iolto' ' ' fl ‘ ' ' islto """" iooo """" isbo Scan # Figure 4-11. The profile obtained for the replicate injections of the PM-thiohydantoin amino acid in selected ion (m/z 298) monitoring mode. The conditions were maintained as in the previous experiment. 144 D. ESI/CID/MS/MS In order to characterize the thiohydantoin amino acids, tandem mass-spectrometry of TH amino acids was undertaken on a TQMS. Solutions of the TH-arnino acids were infilsed into the ESI source at a rate of 3ul/min. The concentration of the samples were 1pmol/ul. The ESI needle was held at 4kV, capillary at 200°C. The collision chamber (Q2) pressure was at 3 mtorr and energy was at 30eV (lab). The protonated molecule was subjected to CID and MS/MS mass spectrum was collected at the Q3. Figures 4-13 shows the ESI/ClD/MS/MS mass spectra of lys-PyTH. The proposed fragmentation patterns are indicated in Figure 4-12. Peaks representing a loss of the side chain group and the reagent were observed in all the derivatives studied. The fragmentation pattern helps to characterize the TH amino acid derivatives. Figure 4-14 shows the MS/MS spectrum of PMTH-asp and the proposed fragmentation pattern. c ,c + H ~~~~~ ‘ O (1” | ll 5 s / C\ i ll 1 ©/ N\ /CH—(CH2)4— NH-l—C—t-NH O l : N N C+ N/H ’1' I} S/ + H” _ H Figure 4-12. Proposed fragmentation patterns for PyTH-lys under ESI/CID/MS/MS. 145 b 265 roe C 137 80‘ £9 60* E + ,5 (M+H) 3 a 401 "7' 307 3 4e 95 129 147 204 204 283 M “W.” U W W W .... W ... M .... 100 200 300 400 m/z Figure 4-13. ESI/MS/MS mass spectrum of PyTH-lys. Collision gas pressure was at 3mtorr and energy was at 30eV. Sample was infused at a flow rate of 3 uL/min. Mass spectrometer was scanned 100 - 450 Daltons in 0.5 sec. b +H a cs ‘\ O -H (a) s‘ \‘ ‘ 1 i, ll : I I ' l l ©‘CH2T1‘\/‘\ fH —§—C Hz—COOH I l | r P‘ 4/gc+ ‘ hfli i s \ (M+H)+ 266 uxn 80y 601 40‘ b 151 C a 92 206 20‘ 99 225 169 m fi’ ‘Tfi V v v v v v v v v v v v v v r V T T I I '50 100 150 200 250 3b0 m/z Figure 4-14. ESI/CID/MS/MS spectrum of PMTH-asp. Collision energy at 30eV and pressure at 3 mtorr and the proposed fragrnentations. 147 Using mass spectrometry, isomass compounds cannot be distinguished easily. This is a drawback of the mass spectrometric techniques when used to detect the isomass thiohydantoin amino acids without HPLC separation. Detection of Ian and ile is very important in peptide sequencing to get the correct sequence information. The TH derivatives of leu and ile were subjected to tandem mass spectrometry to evaluate the possibility of distinguishing leu and ile based on their fi'agmentation pattern. Figures 4-15 and 16 show the MS/MS mass spectra of PyTI-I-leu and ile respectively. The spectra are very similar in nature. Both PyTH derivatives of leu and ile amino acids show a loss of side chain that has a similar mass. A strong peak for a (leu-PyTH - leu + H)+ was observed. According to these results, leu and ile derivatives cannot be separated based on the MS/MS data along. In order to identify ile and leu separately, a short HPLC column has to be used. b a +H . 0 ‘~ \“ ‘ I : $113 I I ©://1‘\:\/;H -:r—CH2— on I I b 137 100 80- 60- 40‘ + (M+H) 250 20* 121 86 m M Figure 4-15. ESI/CID/MS/MS mass spectrum of PyTH-leu and the proposed fragmentation pattern. Collision energy 30eV and pressure 3 mtorr. 149 b + ”0‘ as, s“ W \‘ t‘ : Ac 5 @/ 1‘\ fH —:r—CI H—CHZCH3 I N /C—-‘~“— NH E ('3H3 I s \ b 137 1007 sod E? .... 60.. 3 .5 .2 3E 52 4o~ + (14+ED 20~ so 250 121 163 205 so 100 150 206 'ffizso ...360 m/z Figure 4-16 ESI/MS/MS mass spectrum of PyTH-ile amino acid derivative and the proposed fiagmentation pattern. Conditions were similar to those of Figure 4-15. 150 V. Studies on Reaction Completion A Experimental The coupling reaction is very important for Edman degradation chemistry for successful repetitive cycles. The coupling reaction efficiency was assessed with bradykinin (RPPGFSPFR) peptide under conditions typically used in manual or automated sequencing. Aliquots of lOpmol of bradykinin in water were placed in microfirge tubes, supplemented with 50uL of coupling buffer (HzO/methanol/N-ethylmorpholine 25/70/5 % v/v) and with 10 pL of reagent dissolved in acetonitrile. The atmospheric air was removed by passing dry nitrogen through the solution for about 10 seconds. The samples were mixed by vortexing and incubated at 45°C, 50°C, and 60°C respectively for times ranging ham 1 minute to 10 minutes in a water bath. After incubation, the whole reaction mixture was immediately injected into a pre-equilibrated reversed phase HPLC system. The native peptide and the derivatized peptide was monitored using a UV-VIS detector. The reaction products were analyzed on a C18-column (4.6mm x 25 cm) at a flow rate of 2mL/min. A chromatographic column was developed on a gradient program of 20%B to 100% B. The solvent A was H20/0.1% TFA and the solvent B was acetonitrile/0.1% TFA The eluents fi'om the column were monitored at 214nm using a Kratos Spectrflow 747 variable absorbance detector. B. Results and Discussion 1. Coupling reaction with PyITC Figure 4-17 shows the variation of coupling reaction completion (%) with time at various temperatures. The coupling reaction is clearly temperature and time dependent as shown in Figure 4-17. The reaction yield with the reagent concentration (1% - 10%) was also assessed for the completeness. The coupling reaction seems to be faster than that of 1 5 1 PITC under similar conditions. At 50°C, 2% PyITC seems to be reasonable for the reaction. The use of low reagent concentrations helps to reduce amounts of by-products formed during the coupling reaction. The formation of by-products is always a problem with Edman sequencing and interferes with TH detection. In an automated sequenator, a reducing agent such as dithiothreitol is added to the solvents to prevent formation of by- products usually the oxidation products. 2. Coupling reaction with PMITC Figure 4-18 shows the variation of reaction completeness with temperature and time. It clearly demonstrates the dependence of reaction yield on temperature and time. The percent reaction completeness of PMITC is lower than that of PyITC at 50°C, but at 60°C it reaches completion. C. Cleavage Reaction of TC-peptides VGVAPG, and ACP (V QAADYING) peptides (40 nmol) were derivatized with the reagents PyITC, PMITC, and PITC, respectively. The products were purified by RP- HPLC and the fi'actions were manually collected. The pooled solutions were equally divided into four effendorf tubes and dried under vacuum to dryness. The dried products were incubated with 20 11].. of anhydrous TFA in a water bath at 50°C time ranging fi'om 1 minute to 10 minute. The products were immediately analyzed by FAB/MS. Alternatively, the incubated products were injected into a pre-equilibrated RP- HPLC system and developed as before. The disappearance of TC-peptide and the appearance of the shortened peptide was monitored on a UV-Vls detector. The peaks were manually collected and confirmed by FAB/MS. % Conversion 100 i 804 60$ 405 20— 152 +45°C -¢-5¢c -* ”60°C Time/min Figure 4-17. Kinetic assessment of coupling reaction between pyridyl isothiocyanate and bradykinin (RPPGFSPFR). The peptide was treated with 2.5% PyITC in a buffer solution and the coupling reaction was assessed by analyzing the reaction mixture by RP-HPLC. % Conversion 100.0— 1 80.0; 60.03 40.05 20.01 .1 + 60°C + 50°C 0.0 ‘ 0 d -l .1 Time/min Figure 4-18. Kinetic assessment of coupling reaction between pyridylmethyl isothiocyanate and bradykinin (RPPGFSPFR). The peptide was treated with 2.5% PMITC in a bufl'er solution and the coupling reaction was assessed as in Figure 4-17. 153 D. Results and Discussion The anhydrous TFA acid cleaves the thiocarbamyl peptide to give an ATZ amino acid and a truncated peptide. The unstable ATZ amino acid is converted to the more stable PTH amino acid before HPLC separation followed by UV detection. The cleavage step is an important part of the peptide sequencing procedure. FAB/MS mass spectrometry is a convenient technique to study the reaction products quickly by using the direct insertion probe. Figure 4-19 shows that the TC-peptides undergo cleavage to give unstable ATZ amino acids and truncated peptides in the presence of anhydrous TFA at 50°C. F AB/MS data shows that the PyTC-VGVAPG peptide underwent cleavage at 50° C within 10 minutes (Figure 4-19a). This reaction is somewhat slower than the cleavage of the other derivatives. It cleaved the PyTC-ACP peptide to nearly the same extent at 10 minutes. Figure 4-19b shows the cleavage of PMTC-VGVAPG when incubated with anhydrous TFA at 50°C for 2 minutes. The cleavage reaction of PMTC-VGVAPG is complete within 2 minutes and faster than that of the PyTC-peptide. Figure 4-19c shows the extent of cleavage of PTC-VGVAPG at 50°C after 5 minutes. After 5 minutes, the PTC-peptide was almost disappeared in the mass spectrum. The cleavage reaction for PMTC-peptides is comparable to that of the PTC-peptide. PyTC-peptide cleavage is slower than that of the PTC-peptide. But within 10 minutes, both TC-peptides undergo complete cleavage. The progress of the cleavage reaction was assessed by monitoring the appearance of the earlier eluting truncated peptide and the disappearance of the original peptide. HPLC analysis indicates that cleavage of PyTC- and PMTC- peptides with anhydrous TFA is essentially complete within 10 minutes. 154 1- R 4 . l 1 . . . 80‘ O ,t j 369 'V « (a) e 60* A b :40- (Gvapcetrn . g . 4°° e. l 0 “2g (VGVAPG—PyTC + H)+ c 553 . ’ l owl—VA l 4% A'f A L Y L I A A JV it 350 400 450 500 550 600 650 100 M12 100 R J c l t .801 t 4 i . “ 9 (cvapc + 10+ "9 v 1 0 so 335 400 1 A 1 b 1 “40— II 1 . d . . 4 461 “20‘ ' (VGVAPG-PMTC + Hij+ c . 153 . t 1... AAA; '7 AA A1 _A. ‘1: ..L_._ 0350 400 450 500 650 600 650 M12700 100 R 1 O l l r :30 (e) i 1 O " ‘ 3 9 (GVAPG + 10+ ° so. if 400 A t b “40 ll ‘ g d . a 461 3” 1 '3 (vchc-erc + H)+ ‘ 1 i at M.;. Jul- ..L ... . i l; A ' AL .1. L'L‘ ._ .L‘ A 400 4E1 560 650 600 550 M12700 Figure 4-19. FAB/MS mass spectra showing the cleavage of thiocabamyl peptide (VGVAPG) with anhydrous TFA at 50°C. (a) PyTC-peptide after 10 min, (b). PMTC- peptide afier 2min., and (c) PTC-peptide alter 5 min. 1 5 5 VI. Peptide Sequencing The ACP peptide (V QAADYING) was manually sequenced using both reagents. The peptide (lOOpmol) was incubated in coupling bufi‘er (200uL) with the reagent for 30 minutes at 50°C for PyITC and 60°C for PMITC. The solvents were evaporated and excess reagent and by-products were extracted with ethyl acetate twice. The dried residue was incubated with anhydrous TFA (20uL) for 10 minutes at 50°C and dried. The ATZ derivative was extracted with ethyl acetate and dried in vacuo. The residue was converted to the more stable thiohydantoin derivative with 25% TFA The TH amino acids were injected into a stream of CH3OH/HzO/HOAC (50/49.9/0.1 %v/v) at a flow rate Iota/min and analyzed by ESI/MS. The first five residues were manually sequenced and could be detected at lOOpmol using ESI/MS. The overlap between residues were minimal. In this experiment long coupling times (30 minutes) were used (typical times for manual sequencing). A fairly high amount of material (lOOpmol) was used in this experiment; and the amount can be decreased by using automated peptide sequanator. The detection limit of the sequencing process was not assessed. In manual sequencing there can be sample loses due to incomplete extractions, wash out, or adsorption to walls of the tubes. In the automated sequanator, the sample loses are minimized by non-covalently immobilizing the sample on a polybrene membrane to avoid wash outs, precise extraction steps, and using a micro reaction chamber. Therefore, low-levels can be sequenced using the automated sequanator. The thiohydantoin amino acids can be analyzed at the low femtomol level as shown earlier. These reagents have the potential to prepare suitable derivatives of peptides for low-level sequencing capabilities and detection by ESI/MS. 156 VH.. Post-translationally Modified Amino Acids One of the great interests in protein sequencing today is post-translationally modified amino acids. It is very important to know the correct sequence without any ambiguity. In the peptide sequencer, the PTH amino acids are separated by HPLC and detected by UV absorption of the eluent. The PTH amino acids are identified by their retention time in the HPLC chromatogram. The retention times are standardized using the common PTH amino acids. Post-translationally modified amino acids are missed or unidentified by their retention times, if they are not included in the retention standard. The results of the Association of Biomolecular Resource Facilities Sequencing Committee survey shows that hydroxyproline and O-phosphoserine are diflicult to identify during peptide sequencing for reliable confirmation (19). In this survey, 75 facilities participated and only 46% facilities identified hydroxyproline correctly. The most difficult amino acid to identify was phosphoserine. In this part, mass spectrometry will be used to detect modified TH amino acids by their molecular weight information. Hydroxyproline is a common modified amino acid present in the peptides. During the HPLC separation and detection technique, there are two peaks present in the chromatogram as shown in Figure 4-20. A peak lies close to the peak for PTH alanine (20). Usually, this will be identified as alanine unless the accompanying peak is taken into consideration. Therefore, hydroxyproline was used as a modified amino acid to study the suitability of mass spectrometry with the new reagents. The thiohydantoin derivatives of hydroxyproline were prepared as described earlier using both reagents PyITC and PMITC. The purified TH amino acids were analyzed by ESI/MS. The derivatives (5pmol) were injected to a stream of CH3OH/HzO/TF A (50/49.9/O.1 %v/v) at a flow rate of . 157 lOOuI/min. Figure 4-21 shows the ESI/MS mass spectrum obtained for the PMTH- hydroxyproline. This can be identified without ambiguity using the molecular weight. Another common modified amino acid present in proteins is O-phosphoserine. O- phosphoserine was derivatized with the modified Edman reagents and analyzed by ESI/MS. Figure 4-22 shows the mass spectrum obtained for PM'I'H-O-phosphoserine (5pmol). The pyridyl group in the modified reagent makes it amenable to ESI/MS. The O-phosphoserine derivatives can be identified using ESI/MS to determine its molecular weight and HPLC its retention time. "RHYDROXYPROUN! soon D N «triob D i Y M 14 r v D E 1' P W a U A P T b O U 4000 s c P It T H r? r L senor D r r 1 Lil 8' Al one " AAiJJ_ A _- .. .l A I 1 l 1 l l 1 so so 100 12.0 14.0 use no 220 240 200 20.0 Figure 4-20. HPLC chromatogram for the PTH amino acid standard containing hydroxyproline. Hydroxyproline eluted as two peaks. 158 100- O /c\ Cflz—N CH—CHz O c .../1 \ . _ CH H so / ‘ \ / 2—0 8 C142 MW.-263 so- >5 3:: ii H .5 0 .g .9; o 40‘ d. 20‘ 150 Mini LA -- -11 T—‘ re ' r j l H H* r r 150 200 250 300 350 400 450 m/z Figure 4-21. ESI/MS mass spectrum of hydroxyproline (5pmol). Sample was injected into a stream of stream of CH30H/I-120/HOAC (50/49.9/O.1 %v/v/v) at a flow rate of lOOuL/min 159 318.1 i /C\ CH2—N CH—CHz—O—P03H O \. .4 / S MW.-317 60" a? an t: 3 .5 to- 0 ..>.. E O a: 20‘ {112" 192.0 29?.9 150‘ 200 250 300 .350 400 450 Scan# Figure 4-22. The ESI/MS mass spectrum of PMTH-O-phosphoserine (5pmol). Sample was injected to a stream of CH3OH/HzO/HOAC (50l49.9/0.1 %v/v/v) at a flow rate of 100nL/min. 160 VIII. Conclusion The responses of the modified amino acid derivatives were evaluated for ESI/MS. They were detectable at the low femtomol level using selected ion monitoring. Percent completeness of the coupling reaction at 50°C indicates that the PyITC reagent reacts very fast with the fi'ee amino terminus of the peptide. The cleavage rate is little slower compared to that of PITC reagent. The PMITC reagent reacts slowly with the free amino terminus of the peptide at 50°C and the reaction goes to near completion at 60°C by 10 minutes. PMTC-peptide cleaves very quickly with anhydrous TFA. The PMTH derivatives can be detected at the low femtomol level using ESI/MS. Both reagents were evaluated for their capacity to use as Edman reagents in peptide sequencing. They both can be used in sequencing and the resulting thiohydantoins can be detected by ESI/MS. In order to completely evaluate the reagents for low level sequencing, the automated sequencer must be used. 16] IX. References 10. 11. 12. 13. 14. 15. 16. 17. Edman, P. Acta. Chem. Scand 1956, 10, 761 Mische, S.M.; Yfiksel, K. 0.; Mende-Mueller, L. M.; Matsudaira, P.; Crimmins, D. L.; Andrews, P. C. Techniques in Protein Chemistry IV, 1993, 453 Melvas, B. W. Acta. Chem. Scand. 1969, 23, 1679 Fales, H. M.; Nagai, Y.; Milne, W. A.; Brewer, H. B.; Bronzert, T. J.; Jr.; Pisano, J. J. Anal. Biochem. 1971, 43, 288 Pramanik, B. C.; Hinton, S. M.; Millington, D. S. Dourdeville, T. A.; Slaughter, C. A Anal. Biochem. 1988, 175, 305 Aebersold, R.; Bures, E. J.; Namchuk, M.; Goghari, M. H.; Shushan, B.; Covey, T. C. Protein Science, 1992, I, 494 Basic, C.; Bailey, J. M.; Shively, J. E.; Lee, T. D. Proceeding of 41st ASA/IS Conference on Mass Spaectromerry and Allied Topics, San Francisco, CA, May 31-June 4, 1993 L'Italien, J. J.; Kent, S. B. H. J. Chromatogr. 1984, 283, 149 1111, S-W.; Chen, G-X.; Palacz, Z.; Wittmann-Liebold, B. FEBS, 1986, 198, 150 More, M. L.; Crossley, F. S. Org. Syn. 21 81 Dains, F. B.; Brewster, R. Q.; Olander, C. P. Org. Syn. Call 1, 447 Dyson, G. M. Org. Syn. Call! 165 L'abbe, G. Synthesis 1990 525 Shibanuma, T.; Shiono, M.; Mukaiyama, T , Chem. Lett., 1977, 573 Kim, 8.; Yr, K. Y., Tetrahedron Lett. 1985, 26, (13), 1661 Still, W. C.; Kahn, M.; Mitra, A. J. Org. Chem. 1978, 43 (19), 2923 In ”The Chemistry of Cyanates and Their Thio Derivatives”, Patai, 8 (Ed) 1977 John Wiley & Sons, New York 18. 19. 20. 162 Hunt, B. J.; Rigby, W. Chem. Ind. (London) , 1967,1868 Hihara, J. Protein Analysis Renaissance, Applied Biosystems, 1993/94, 15 Sengoku, Y.; Shionoya, Y.; Hagiwara, K.; Kirsher, 8.; Yuan, P.-M Protein Analysis Renaissance, Applied Biosystems, 1993/94, 25 CHAPTER 5 MODIFICATION OF ANABOLIC STEROIDS FOR ELECTROSPRAY MASS SPECTROMETRY I. INTRODUCTION The illegal use of anabolic steroids to affect the outcome of both human and equine sport events is of major concern and has been well documented (1,2). A concise summary of the controversial literature on the effect of anabolic steroids on athletic performance and their deleterious side effects has appeared (3). Most anabolic steroids are synthetic derivatives of the male sex hormone testosterone, with high anabolic potential and minimized androgenic activity. Due to extensive metabolism, confirmation of the unlawfiil use of this group of doping agents requires an understanding of their metabolic fate in the species and the development of more efficient analytical methods for unambiguously detecting these steroids and their metabolites in biological fluids (4). Several different analytical techniques have been employed to detect anabolic steroids in biological fluids. The most common techniques are radioimuimnnoassay (5) (RIA), high performance liquid chromatography (HPLC) (5), gas chromatography/electron impact mass spectrometry (GC/EI-MS) (6), gas chromatography/electron capture negative ionization mass spectrometry (GC/ECNI- MS)(7), and more recently liquid chromatography/mass spectrometry (8-12). The major advantage of RIA is its sensitivity, but it is not routinely used in screening because of disadvantages such as cross-reactivity, difiiculty in measuring the testosterone-to- epitestosterone ratio, and difficulty in detecting methenolone (13). HPLC has been used mainly for the analysis of anabolics in the veterinary, pharmaceutical, and forensic fields, rather than in human urine screening. The major problem with HPLC is absolute confirmation of the analyte. 1 63 164 By far the most common analytical method for anabolic steroid analysis is GC/MS. Steroids must be derivatized to protect fimctional groups and to enhance volatility. (6, 14, 15). Methoxime-trimethylsilyl derivatives were among the first to be used in the analysis of anabolic steroids. The methoxime moiety protects the carbonyl functionality while the trimethylsilyl reacts with the hydroxyl groups. Reports of detection limits for individual steroids or metabolites range from 3 ng/mL to 20ng/mL of urine (16-18). Trimethylsilyl (TMS) derivatives are now more widely accepted than methoxime-TMS derivatives (19- 22). Selective derivatization can be accomplished by selecting the silanizing reagent and catalyst. I In order to detect trace levels of anabolic steroids long after administration, analytical techniques with better sensitivity and selectivity are needed. Electron capture negative ionization (ECNI) mass spectrometry has the capacity to meet some of these analytical needs. But ECNI has not been as widely used as had been anticipated, this is partially due to problems associated with finding proper derivatives that are amenable to the ionization technique. Chemical oxidation followed by GC/ECNI-MS has been developed in this laboratory as a sensitive and selective detection technique for dexarnethasone and prednisolone (23, 24). In this method, steroid molecules are chemically oxidized to a highly electrophilic species while not significantly afl'ecting the electrophilic character of the biological matrix, thereby pemritting selective and sensitive detection of the analyte. Efl'ect of structure on the ECNI mass spectrometric response of steroids has been reported (24). It was determined that the key to enhanced ECNI-MS response of ketosteroids is extensive or, B-unsaturation. The atmospheric pressure chemical ionization (APCI) (11) and thermospray (9,10) approaches are the most widely used in LC/MS techniques to detect steroids and their conjugates. The detection limits of low ng/uL to pg/uL has been reported for steroids using thermospray mass spectrometry (8). 165 Usually, the forms of metabolites are conjugation with glucuronic acid or sulfate. Since drug conjugates are not amenable to determination by GC/MS, owing to their low volatility and thermal instability, they are hydrolyzed before analysis by GC/MS. Henion et al. reported the detection of anabolic steroid sulfate conjugates by using negative electrospray mass spectrometry (ESI/MS) with excellent detection limits (11). Subsequently, several research groups have reported the use of negative ESI/MS to detect steroid conjugates in biological-fluids (12). Figure 5-1 shows the generic steroid structure and the numbering system used in steroid nomenclature. The use of anabolic steroids to enhance the performance of athletes are forbidden by the International Olympic Committee (10C). The structures of banned anabolic steroids are given in Figure 5-2. The objective of this part of the research project is to develop analytical methodology to detect trace levels of anabolic steroids present in biological samples using electrospray mass spectrometry with high specificity. II. Modification of Steroids for ESI/MS The anabolic steroids are a very important class of molecules with similar structural characteristics. All the steroids have similar steroid skeleton except stanozolol. The analysis of stanozolol is discussed later in this chapter. The unconjugated steroids have no functional group to form ions readily in solution. The structures of common anabolic steroids have a keto functionality in common as shown in Figure 5-2. The keto group was modified using Girard's T reagent or hydroxylammine hydrochloride to improve the sensitivity by ESI/MS. The Girard's T reagent forms a hydrazone derivative with the keto group of the steroid and it carries a quartenaiy ammonium group which has a pre-formed charge. The reagent hydroxyl amine hydrochloride forms an oxime 166 derivative with the keto functionality and can be protonated easily in acidic solution. Formation of both derivatives with steroids will be discussed in detail later in this chapter. 111. Hydrazone Derivative Formation The Girard's T reagent first introduced by Girard and Sandulesco (25, 26) reacts with the keto functionality of steroid ketones to form a water soluble hydrazone. The hydrazone derivative carries a quaternary ammonium moiety that is amenable to positive electrospray mass spectrometry. The ESI/MS response of quartenary ammonium groups is well documented to be in the low femtogram range (27). In this study, several testosterone analogs (Table 5-1), were used as representative model compounds. The reaction, shown in Figure 5-3, with testosterone, proceeded readily when 10 nmol of steroid in methanol was mixed with a 20-fold excess of Girard's T reagent dissolved in methanol. The solution was brought to a final acid concentration of 1M HOAC by adding glacial acetic acid. The reaction mixture was incubated in a water bath at 50°C for 30 minutes. The reaction mixture was injected directly into a pre-equilibrated RP-HPLC system for separation. A Mass Spectral Analysis In recent years, electrospray mass spectrometry has gained wide applicability in biological mass spectrometry due to its high sensitivity and capacity to form multiply charged ions in determining the molecular weight of polymers that cannot be determined by conventional mass spectrometry. In this study, ESI/MS has been investigated for analysis of anabolic steroids. 167 21 22 24 26 o 18 12 23 25 27 11 17 13 1 19 16 2 8 14 15 3 5 7 A = androstane, C19 skeleton P = pregnane, C21 skeleton C = cholestone, C27 skeleton ene = double bond 01 = hydroxy group one = carbonyl group or = Stbstituents erdendingintothe plane ofthe pay [3 = Stbstituents extendingoutofthe plane ofthe page Figure 5-1. Nomenclature and numbering system for the steroid carbon skeleton 168 as“ as“ as: Testosterone Nandrolone Methyltestosterone OH OH on M“ yogi-m3 1 H Orandrolone Bolasterone Boldenone 0H OH OH 0'13 ' (113 I I I l '> Cl Fluoryrmsterone Mesterolone Costebol OH OH 01-1 M. 013 X'GES (fi- 013 Mcthand'cnone Methenolone Dehydrochbromethyl testosterone OH OH OH OHC Stanozolol Norethandrolone Fomyklienolone 0H 0H 0H - -CH3 «EL/95 CH3 11% “(MW Oxyrmsterone Drannstanolone Oxyrmtholone Figure 5-2. The Structures of Anabolic Steroids banned By IOC 169 It has previously been shown that quaternary ammonium moieties and strongly acidic groups such as sulfates or sulfonates are detectable at low femtomole levels by pneumaticatically assisted electrospray mass spectrometry (27, 28). Hydroxylated compounds have been analyzed by positive electrospray mass spectrometry as a sodiated molecule (M+Na)+ (29). We investigated a method to improve the detection limits of anabolic ketosteroids by forming hydrazone derivatives with Girard's T reagent. The mass spectral analysis was undertaken on the home-built electrospray interface for the preliminary investigations. Other investigations were undertaken on a TSQ 700 triple quadrupole mass spectrometer fitted with a Finnigan ESI source. The ESI needle was held at 4.5kV and the heated capillary tube was at 200°C. Samples were introduced into the ESI source through an infusion pump or loop injection. The ESI mass spectra were acquired by directly loop-injecting a SuL portion of the reaction mixture containing 100ng 11L of each hydrazone into a stream of mobile phase (CH3OH/HzO/CH3COOH 50:49.9:0.1 % v/v/v). Hydrazone derivatives were prepared with several steroids (Table 5-1) and the ESI/MS mass spectra were acquired. All the hydrazone chlorides show a strong peak for the molecular cation in the ESI/MS mass spectra. The electrospray mass spectra of the hydrazone derivatives of model compounds showed little or no fragmentation under normal ESI conditions as illustrated in Figure 5-4. The lack of fragmentation coupled with the fixed charge on the quaternary ammonium moiety facilitate efficient detection of these steroid derivatives by electrospray mass spectrometry. 170 Table 5- 1. The steroids used to prepare hydrazone derivatives Testosterone Boldenone Methyltestosterone Bolasterone Nandrolone Mesterolone Fluoxymesterone Methandrostenolone 1,2 Dehydro 17a methyltestosterone B. Detection Limits of Hydrazones The strong response of quaternary ammonium functional groups using electrospray mass spectrometry is well documented and detection limits in the femtogram range have been reported (27, 28). To determine the detection limits for the hydrazone derivatives of testosterone analogs, increasing amounts of methyltestosterone hydrazone chloride were injected into the ion source of the triple quadrupole mass spectrometer. A volume of SuL was flow injected into a stream of methanol/water/acetic acid (SO/49.9/0. 1%v/v) at a flow rate of lOOuIJmin. Figure 5-5 shows the mass spectral response during selected reaction (m/z 416 to m/z 357) monitoring in the triple quadrupole mass spectrometer. Detection limits into the low femtogram level were achieved. 171 H or 3 ... H3C— N— CH2CONH— NH2 + CH3 Girard's T reagent Testosterone Cl' H3 + , + H20 H3C-- N— CH2CONH— N ' CH3 Testosterone Hydramne Chloride Figure 5-3. The reaction scheme for the formation of steroid hydrazone. The Girard's T reagent reacts with the keto group of the steroid to form hydrazone derivative. 172 M+ 450.2 100 i 80‘ .5? 60 3 .5 3 3 0 M 40. 20‘ . it, JILL; Al... .1“ 11.111 1‘1“] hrtlerll 1...: ALL“... [.11 360 380 400 420 440 460 480 500 m/z Figure 5-4. Portion of the ESI/MS mass spectrum of fluoxymesterone hydrazone chloride (molecular weight of fluoxymesterone hydrazone cation = 450) 173 IM‘ hang A Rehdhn:hflen§ty 2119 2019 200., 2m 2099 20°” \J L t a v T—v—v Vrfi unur# Fig. 5-5. Detection limits for the methyltestosterone hydrazone molecular cation. The samples were injected into a stream of CH3OH/HZO/HOAC (SO/49.9/O. 1% v/v) flowing at a rate of lOOuL/min in increasing concentration. The injection volume was 5 1.1L. The amounts are indicated above the corresponding peak. l 74 C. MS/MS Studies of Hydrazones by ESI/MS Tandem mass spectrometry (MS/MS), since its development in the 1970s, has been gaining acceptance as a rapid, sensitive and selective analytical method for the analysis of complex biological samples (3 0-33). The advantages gained by MS/MS often complement those gained by extensive sample clean-up procedures and lengthy chromatographic separations (34-3 6). Other capabilities of tandem mass spectrometry such as selected reaction monitoring and constant neutral-loss scanning make this mode of operation attractive in analyzing mixtures to obtain selectivity. It has been investigated the capacity of MS/MS to reduce chemical background during an analysis by using constant neutral-loss scanning with the triple quadrupole mass spectrometer. The collision-induced fragmentation of hydrazones was studied with the aim of discriminating between secondary ions generated by fiagmentation of the primary ion and chemical noise. Such discrimination can be used as the basis for increasing the signal-to- background ratio by selective electronic filtering. These experiments were of particular importance as they addressed one of the major technical problems in highly sensitive analyses of complex biological samples, chemical noise obscuring low-level signals from the analyte. The experiment was undertaken on a triple quadrupole mass spectrometer fitted with a F innigan ESI source. The salient fragmentation pathways comprised a stepwise loss of neutral N(CH3 )3 (59u) followed by another loss of 28u. The fi’agmentation of testosterone, methyltestosterone, nandrolone, fluoxymesterone, and bolasterone hydrazones provided analogous spectra. The hydrazone derivatives of boldenone, methandrostenolone, and mesterolone gave strong peaks at (059) , (C-8 7), and additional peaks at (C-15), (C-102), and (C-117) in their MS/MS spectra where C indicates the molecular cation of the derivative. Table 5-2 shows the list of peaks observed in the tandem mass spectrometric studies. The collision energy was maintained 175 at 40eV relative to the laboratory frame. An increase in collision energy beyond 40eV did not yield new fragments, but abundances of product ions increased with collision energy. During the MS/MS study, the samples (100pg/uL) were infused at a flow rate of 3Wmin with a syringe pump (Cambridge, MA ) while the mass spectrometer was scanned from lOu to 500u in 0.5sec and the pressure inside the collision cell was maintained at 3.0 mtorr. Figure 5-6 shows the MS/MS spectrum of methyltestosterone hydrazone after averaging data acquired over one minute. Therefore, all the hydrazone derivatives show a common loss of 59u in their MS/MS spctra. D. Study of Fragmentations with Offset Energy and Pressure When MS/MS experiments are carried out on a triple quadrupole mass spectrometer, two major variables affect the abundance of representative product ions. One variable is the collision energy (offset energy) applied to the RF -only quadrupole. Another variable is the pressure inside the collision cell. The effect of both variables were studied while infirsing the sample at a constant flow rate of 3 uL/min. The ESI needle was at 4.5kV and capillary tube was heated to 200°C. The nebulizing gas (N2) pressure was at 35psi. All the experiments were conducted under similar conditions and data acquired after averaging over one minute. The abundances of product ions were studied as a function of collision energy fi'om 20eV to 40eV. Figure 5-7 shows the product ion abundances for the testosterone hydrazone derivative with the ofi'set energy at the collision pressure of 3mtror (Ar). The absolute abundances of three characteristic ions of MS/MS spectra of testosterone hydrazone (collision gas pressure at 3 mtorr) were plotted against the Vub. Figure 5-6 shows that the m/z 343 (M-59)+ ion has the highest abundance at offset energy of 30eV. When examining the respective spectra, the spectrum at 40eV has more 176 abundant fiagments than that of at 30eV and the ion current at M+ has distributed over the large number of fragment ions at 40eV. At 30eV, the product spectrum has only a few fiagment ions and the most abundant fragment ion is the (M-59)"' ion. More intense fiagmentation can be induced at 40eV collision energy. The same pattern was observed at the collision gas pressures l, 1.5, 2.0, 2.5, 3.0, and 3.5 mtorr. At 40eV collision offset energy, as the gas pressure increases, the abundances of m/z 343 and 163 increase and the abundance of the parent ion decreases as shown in Figure 5-8. In order to do neutral loss scanning or selected reaction monitoring (SRM), the energy of 30eV at 3 mtorr is appropriate for hydrazone derivatives. Table 5-2. A list of peaks observed in MS/MS studies of hydrazone molecular cation M+. (where C = M") Steroid Derivative Major Peaks Observed in MS/MS spectrum (C-15) (C-59) (C-87) (C-102) (C-1 17) Methyltestosterone + + Nandrolone + + + Fluoxymesterone + + Bolasterone + + Boldenone + + + + + Mesterolone + + + 177 (M-59)+ 357.3 1001 80* g 60. a 0 ,> g (M-87)+ 9‘ 40* 329.3 + M 416.3 20, 163.1 134.9 .41 l--- , W., .1 , - 100 200 300 400 m/z Fig. 5-6. ESI/MS/MS spectrum of the molecular cation of methyltestosterone hydrazone (M"’). The spectrum was obtained by infirsing the sample at a flow rate of 3 uL/min. and at a concentration of 100 pg/ 11L. The mass spectrometer was scanned at a rate of 980u/sec and spectra were averaged for one minute. The collision energy was set at 40eV (lab). Ar was used as the collision gas and the collision cell pressure was 3 mtorr. 1 78 E. F AB/MS Analysis of Hydrazones FAB/MS analysis was undertaken on a JOEL HX 110 high performance double focusing mass spectrometer with forward geometry (often denoted as an EB instrument because the electric sector, E, precedes the magnetic sector, B). The hydrazone samples were dissolved in methanol and dispersed on a few microliters of matrix that was glycerol on a probe tip. The probe was inserted to an ion source of the mass spectrometer and evacuated. An analyte ion beam was generated by bombardment of high energy Xe atoms. The ion beam was accelerated to lOkeV and mass analyzed by the FAB/MS. The data were collected on a DASOOO data system. Figure 5-9 shows the FAB mass spectrum of methyltestoterone hydrazone (1ng). The asterisks indicate the matrix peaks. The FAB mass spectrum of the steroid hydrazone gave a strong peak representing the molecular cation and several peaks for the glycerol adduct ions. The matrix peaks of the FAB mass spectrum make the detection of low level of analytes difficult in most cases. Comparison of Figures 5-4 and 5-9 clearly shows the simplicity of the ESI mass spectrometry of hydrazone. 179 51¢— 41¢— €31¢~ m 3 =21&4 H i 11$ 010°“....q.Le-.~.-.','.':T.*?T .... .... 15 20 25 so 35 4o 45 Collision Energy (eV) Figure 5-7. Variation of product ion abundances with collision ofi‘set energy at the collision cell gas pressure of 3mtorr. +402 +343 l l l I I I O .5 1 1 .5 2 2.5 3 3 .5 4 Pressure (mtorr) Figure 5-8. The variation of product ion abundances with collision cell gas pressure at the collision offset energy of 40eV. 180 F. High Energy CID on FAB/MS The molecular cation was subjected to the high-energy FAB/CAD/MS/MS analysis. The M+ was selected and fragmented with Ar gas. Figure 5-10 shows the FAB/CAD/MS/MS spectrum of methyltestosterone chloride. G. Comparison of ESI/CID/MS/MS with F AB/CID/MS/MS The ESI/MS/MS spectra and the FAB/MS/MS spectra of the hydrazone derivatives are significantly different. The energy deposited in the ions are different in each case. The ESI/MS/MS spectrum on the TQMS shows more fragmentation at the lower-mass end of the spectrum and the high-energy FAB/MS/MS spectra shows more fragmentation at the higher-mass end of the spectra. Both spectra show the loss of 59u and 87u fi'om the molecular cation. In the FAB/MS mass spectrum of the testosterone hydrazone, no significant fragments were observed below 200u. But in the TQMS, there were several strong peaks below 200u due to ring cleavages of the steroid skeleton. 181 M4- 185-0 416.0 100 - . R . . 4 l . a 80 . t . i V ‘ 0 a so . . 277 A . b ‘ 359.0 11 40 l n . d 4 ‘ 4 n 20 « C o 1 302.1 1 245-0 477.9 10° 150 200 250 300 350 400 450 m 500 Figure 5-9. The F AB/MS spectrum of methyltestosterone hydrazone chloride (1 ng). Glycerol was used as the matrix and matrix peaks are indicated by an asterisk. M+ .<-o.-—.w 8 H OI A l A A A A 4 b I u 10 - n 4 d 4 ‘ I n 5.. c ‘ 237.1 C 68.0 o l l 1 1 111111 c fi‘r I Y I V 7 Y Y I Y fi fr T r I' V I Y I V Y~r Tfi Y 50 100 150 200 250 300 350 m Figure 5-10. The FAB/CAD/MS/MS spectrum of methyltestosterone hydrazone chloride. l 82 H. Neutral Loss Scanning Studies of Hydrazone Derivatives Because all hydrazone derivatives fragmented with a neutral loss of 59u, corresponding to the elimination of trimethylarnine, a method of MS/MS scanning for this class of compounds could be implemented by scanning both quadrupole mass analyzers in tandem with a mass difference of 59 u. In this mode of operation, termed as MS/MS constant neutral loss scanning, only those compounds that undergo a neutral loss of 5% will appear in the final analysis, therefore permitting a means of detecting these derivatives in the presence of significant levels of potentially interfering contaminants. A derivative of model compound nandrolone hydrazone was subjected to neutral loss scanning of 59u and the following spectrum (Figure 5-11) was obtained. The neutral loss spectrum of nandrolone hydrazone contains only a single peak and therefore, hydrazones that lose 59u in an MS/MS experiment, are responsive to this mode of scanning. The nandrolone hydrazone (lOOpg/uL) was infirsed at a flow rate of 311L/min into the ESI source. The mass spectrometer was scanned from 10-500u in 0.5 sec. The collision gas pressure was at 3 mtror and energy was at 40eV. An application of the neutral loss scanning mode is presented later in the analysis of a spiked urine sample. 1. LC/MS Analysis Isolation and separation of trace quantities of biologically important materials is always a primary concern of an analyst. Sample losses during purification and extraction steps are unavoidable. By using on-line separation techniques, the losses due to purification and extraction steps can be minimized. By carefully selecting the correct separation technique, the signals from trace materials can be maximized. In order to evaluate on-line HPLC with ESI/MS, a mixture of hydrazones of model compounds was 183 analyzed by using a C13 column with acetonitrile and water mixture containing 0.1% TFA and gradient of 30% to 60% acetonitrile over 20 min. The total flow of 200pL/min was introduced into the ESI interface and the individual derivatives were identified as shown in Figure 5-12 by scanning Q1 between 350 and 500 u in 0.5 sec. The reconstructed total ion current (RITC) and mass chromatograms show that all the four derivatives can be identified after HPLC separation. Also, the chromatograms (Figure 5-12) show that the derivatives form isomeric compounds that have slightly difl‘erent retention times. The sensitivity and separation can be improved by using small diameter (800 pm or smaller) columns. 184 + at 100 388 80 ~ §§c114 is? .8 33 $3 404 204 200" .-256.V.Sdo,. 5507' '400 4361 VYSBO m/z Figure 5-11. The neutral loss scan mass spectrum of nandrolone hydrazone derivative. 185 100« (e).m/z 388 so . 100, m 501 (d). m/z 402 g 100« m E (c).m/z416 o 50‘ ,> 33. 0 9‘ 100‘ 50, (b). m/z 450 100 50‘ (a). RTIC VVVVVVV 500- 10001 1500 Fig. 5-12. Total ion current (RTIC) chromatogram for LC/MS analysis of hydrazone derivatives of model compounds (8) and mass chromatograms for (b) fluoxymesterone hydrazone (m/z 450), (c) methyltestosterone hydrazone (m/z 416), (d) testosterone hydrazone (m/z 402) and (e) nandrolone hydrazone (m/z 388). 186 IV. Feasibility study of Steroid Detection in Urine Sample preparation methods and extraction procedures have been well documented in the literature (5, 20-22). Samples were prepared by spiking 5 mL of urine (obtained from a male volunteer with no previous history of anabolic steroid use) with a standard solution containing the model compounds, nandrolone, methyltestosterone, and fluoxymesterone. A blank urine samples and fortified urine samples (10ng/mL) were extracted using C13 Sep-pak® cartridges (Waters, Millipore Corp., Milford, MA) previously washed with 5 mL of methanol and 5 mL of water. The urine sample was passed through the cartridge followed by 5 mL of water to remove water-soluble urinary components. Steroids were then eluted with 2 mL of methanol and evaporated to dryness under a nitrogen stream. The residue was dissolved in methanol (0.5 mL) and derivatized with the Girard's T reagent. The reaction mixture was subjected to analysis by electrospray mass spectrometry afier HPLC separation. The reconstructed mass chromatograms (Fig 5-13) for the model compounds (lOng/mL) show the presence of all the steroids added to the spiked urine sample. 187 100 - (d) m/z 388 50 . 3.. 100 1 “ m/ 416 '5‘, 50« (c) z .> 3 a W 100 ~ (b) m/z 450 50‘ 100 ' (a) RTIC 50‘ "”566 1666 {566 I scan# Fig. 5-13. Total ion current chromatogram (RTIC) for LC/MS analysis of hydrazone derivatives of urine extract (a) and mass chromatograms for (b) fluoxymesterone hydrazone (m/z 450), (c) methyltestosterone hydrazone (m/z 416), and (d) nandrolone hydrazone (m/z 388). 1 88 A. MS/MS Studies of the Derivatized Urine Extract The neutral-loss scanning mode of operation discriminated against the matrices which do not lose a fragment corresponds to the offset mass between Q1 and Q3. This method is very useful in analyzing hydrazone derivatives because all the hydrazones lose 59u in their MS/MS spectra. If the components in the urine matrix do not lose 59u in the tandem mass spectra, they will not appear in the final spectrum. In order to evaluate the neutral loss scanning mode of operation for urine analysis, a spiked urine sample was prepared with methyltestosterone, extracted and derivatized with the Girard's T reagent. A urine sample was spiked with methyltestosterone (lOng/mL) and allowed 1hr to equilibrate at room temperature. A 5mL portion of the urine sample was extracted and derivatized as described in earlier section. The derivatized reaction mixture was evaporated to dryness and reconstituted in SOOuL of methanol. The final concentration of the hydrazone in solution was lOOpg/uL. The sample was infirsed at a flow rate of SuIJmin and data were collected in the full scanning mode over one minute. The signal due to the steroid hydrazone was very small and signals fi'om other compounds dominated the normal ESI mass spectrum that averaged over one minute (Figure 5-14). The same sample was subjected to neutral loss scanning of 59u and the spectrum on Figure 5-15 was obtained. This clearly demonstrates the possibility of avoiding interference fi'om complex matrices present in the biological samples by selecting a suitable scanning mode of operation of the triple quadrupole mass spectrometer. In both cases the sample was infirsed into the ESI ion source at a rate of 511L/min. The collision offset energy for the neutral loss scanning experiment was set at 40eV and the pressure was at 3mtorr. 189 377.1 100“ 80* 481.4 g 60- E “:3 3.3 o . d 40 M+ 416.4 20. l ' 3160* 380 460' 420 440 4160' 480' 500 m/z Fig. 5-14 Portion of the electrospray mass spectrum of extracted urine sample after spiking with methyltestosterone (1 Ong/mL) followed by derivatization with Girard's T reagent. The sample was infirsed at a flow rate of 511L/min without separation. The spectrum is dominated by signals from the impurities in the urine sample. 190 + M 416.3 100 a 80- .? 604 E .'.>: .13 £2 40« 20- 360 380 400 420 440 460 480 500 m/z Figure 5-15. The simplified mass spectrum of the sample used to obtain figure 5-14, by neutral loss scanning (59 u) of the quadrupoles (Q1 and Q3). The collision energy was set to 40eV and collision gas pressure at 3mtorr. 191 V. Kinetic Studies of the Hydrazone Formation Reaction The coupling reaction between the keto group of the steroid and the Girard's T reagent is very important part of the reaction for successful analysis by the methodology described in this chapter. The Girard's T reagent reacts with the ketone in the presence of an acid; usually acetic acid. The completeness of the reaction was monitored at different temperatures and acid concentrations. A. Dependence of Conversion (%) with Temperature The coupling reaction rate of hydrazone formation was studied using testosterone. Aliquots of 10 nmol of steroid in methanol were placed in a microfuge tube, supplemented with 20-fold excess of Girard's T reagent and acidified with glacial acetic acid to 1M. The samples were mixed by vortexing and incubated at 30°C, 40°C, and 50°C, respectively, for times ranging from 15 min to 1 hour in a water bath. After incubation, the whole reaction mixture was immediately injected into a pre-equilibrated RP-HPLC system. Progression of the reaction was quantitated by monitoring the disappearance of testosterone and the appearance of a new peak corresponding to the expected product as characterized by the UV spectrum and elution time. Testosterone hydrazone has a maximum at 280nm in the UV absorption spectrum. Figure 5-16 shows the dependence of percent reaction completeness with time and temperature. As the temperature increases, the rate of the hydrazone formation increases. As expected, the rate of reaction increases with time to a constant value. 192 B. Dependence of Reaction Conversion (%) with Acid Concentration The dependence of acid concentration on the reaction rate also was investigated. Aliquots of 10 nmol of testosterone were placed in microfirge tubes and a 20-fold excess of Girard's T reagent was added to each. The reaction mixture was acidified with glacial acetic acid to bring the solution to 0.1M, 0.2M, 0.5M, 1M, 2M, or SM, respectively. The samples were incubated at 37°C or 50°C for 30 min. The reaction mixture was injected into a pre-equilibrated RP-HPLC system. Progress of the reaction was monitored as described earlier. The rate of reaction greatly increases in the presence of an acid. As shown in the Fig. 5-17, the rate of reaction becomes almost constant at acid concentrations 2 1M. Wheeler (20) investigated the rate of reaction of a series of steroid ketones in 0.2M acetic acid/methanol at 25°C and established the order of 3 > 6 >> A4’3 ~ 7 ~ 17 > 20 > 12 >> 11, where the position of the ketone is indicated by number. The rates of hydrolysis are in the order of 114-3 ~ 3 < 6 < 17 ~ 20 < 7 ~ 12 ~ 11 (26). Previous workers indicated the possibility of increasing the rate of hydrolysis at high acid concentrations (> 1M), but the hydrolysis constants for A4'3 steroids (most of the anabolic steroids are A4'3) are very low as shown earlier by Wheeler (26). By considering the above factors, 1M HOAC in the reaction mixture for 30 minutes is suitable for the formation of hydrazones with testosterone analogs. VI. Source CID during ESI/MS Collisionally induced dissociation in ESI/MS can be carried out at the source by decreasing the electric field (in positive mode) between the skimmer and the nozzle in single quadrupole mass spectrometers. This technique allows the researchers to do CID 193 on a single quadrupole mass spectrometer fitted with an ESI source. In a Finnigan TQMS, source CID can be induced by applying an additional potential to the RF-only octapole. At the octapole region, the pressure is in the mtorr region and is suitable for CID. Source CID can be induced by using an additional offset potential on the octapole to accelerate ions in this high pressure region to undergo CID. In order to mass analyze the source-induced fi'agments, the potentials on the entire Methyltestosterone hydrazone was infused into the ESI source of the TQMS which was operated in the single quadrupole mode. The ESI needle was at 4.5kV and the heated capillary was at 200°C. The sample (lOng/uL) was introduced at a rate of SuIJmin. The other voltages on the lenses were kept at normal tuned values. The potential on the RF-only octapole was decreased from -3V to -43V and mass spectra were acquired. The mass spectrometer was scanned from 100 to 50011 in 0.5s and data averaged for one minute. Figure 5-18 shows the mass spectrum resulting fi'om the source- CID of the methyltestosterone at the octapole region of the triple quadrupole mass spectrometer. It provides the characteristic peaks observed in MS/MS experiment; loss of 59u and 87u. Therefore, source-CID can be used to induce fi'agmentation in a single quadrupole mass spectrometer in order to obtain more structural characterization combined with ESI. A. MS/MS of the (M-59)+ Ion Derived fi'om Methyltestosterone Hydrazone Source CID induces strong peaks representing loss of 59u (C1159) and 87u (0"- 87) fi'om the molecular cation of methyltestosterone hydrazone in TQMS instrument operated in the single quadrupole mode. This gave a valuable information necessary to characterize the analyte using a single quadrupole mass spectrometer. The source-CID of 194 electrosprayed ions adds another dimension to the capability of the TQMS. Therefore the ions induced by source-CID can be used to generate another set of ions at the Q2 which canbemassanalyzed in the Q3. In this experiment source CID was induced by decreasing the voltage on the RF- only octapole and a selected ion (m/z 357) was allowed to pass through Q1. At Q2, collisionally activated dissociation was done via collisions with Ar gas (3mtorr). The ofl‘set energy of the Q2 was held at 40eV. The Q3 was scanned from 100-500 11 in 0.5s. The MS/MS spectrum of the ion at m/z 357 is shown in Figure 5-19. B. MS/MS of the (INA-87)+ Ion Derived from Methyltestosterone Hydrazone In a second experiment, a peak representing the ion at m/z 329 was allowed to pass through Q1 for collection of the MS/MS spectrum of the ion at m/z 329 (see Figure 5-20). The ESI interface and mass spectrometer parameters were kept as earlier experiment. The two spectra from the ions at m/z 357 and 329 are remarkably different from each other. The MS/MS spectrum of m/z 357 did not give a peak at m/z 329. Therefore, ions represented by the peak at m/z 357 have no contribution toward ions represented by the peak at m/z 329. This technique provides valuable information regarding the structure of the molecule and the selected ions. From this study, it can be concluded that on a TQMS instrument, MS/MS/MS studies can be done on ESI produced ions effectively. The MS/MS/MS data of methyltestosterone hydrazone chloride indicates that ions corresponding to (M-59)+ and (M-87)+ derive fiom two different fiagmentation pathways. These two ions have no common fragments and they undergo fi'agmentation giving different product ions. The ion at m/z 329 (M-87)+ may be due to a rearrangement product. The MS/MS spectra of all the steroid hydrazones show a strong peak at (M-87)+, therefore the rearrangement reaction may be common to all hydrazones. 195 100— % Reaction Conversion o IIIIIUI'IIUITi‘ITUIT1IIUIIIIITI] 1 O 2 0 3 0 4 O 5 O 6 0 7 0 Reaction time/min Figure 5-16. Variation of percent reaction conversion with time and temperature for hydrazone formation 100— - .A —'—' — —_ — —A 8 3 A/ ' L3 80‘: -Jo-———I/- Q,‘ . .. 8 605 l G . —V-— 50°C :8 40- / +3700 0 .1 cu . . -/ m 20.4 , s l o l'U'rl'U'II'V'Il'l'rliii'lllel 0 1 2 3 4 5 6 HoAc concentration! M Figure 5-17. Effect of HOAC concentration on percent reaction conversion 196i + (Ni-59) 357 100 i 80‘ E 60‘ 35:” 132 o + + .5 M g (M'37) and) 40* 416 329 20 ~ 151 299 100m“"26'6”"W'soofi'wfidofiflr m/z Figure 5-18. ESI/MS mass spectrum of methyltestosterone hydrazone chloride obtained by inducing CID at the octapole. The octapole ofl‘set was set to 40eV. 100- 804 60- Relative Intensity 4o~ 20~ 93 .7 197 162.8 150.9 108.1 177.1 vvvvv m/z 416 (molecular cation) l-S9u 357.4 Figure 5-19. The MS/MS spectrum of the ion at m/z (M-59)"' derived fi'om methyltestosterone hydrazone chloride (MW of cation is 416u) by source C1D. 198 135.0 100 ~ 123.0 80 ~ 3‘ g 60 « L5 0 .2 g m/z 416 (molecular cation) a: 40 d l -87u 93.9 148.9 329.1 20 - 174.8 79.9 1 186.8 160. ' 260 ' 360 ' 460 m/z Figure 5-20. The MS/MS spectrum of the ion at m/z (M-87)+ derived from methyltestosterone hydrazone cation (m/z 416) by source CID. 1 99 VII. Oxime Derivative Formation The formation of oxime derivatives to protect the keto functionality of steroids is a well known reaction (16-18). In GC-MS analysis, methoxime derivatives are widely used. The oximes are Schiff bases that can be protonated easily in acidic solution. The ESI/MS is well known technique that ions in solution transfers to the gas phase. Therefore, neutral steroids are not easily detected during ESI/MS because they do not exist as ions. After forming oxime derivatives with hydroxylammine hydrocloride, they can exist as protonated molecules in acidic solution and thus give a good response to ESI/MS. In this reaction, shown in Figure 5-21 with testosterone, 10 nmol of steroid in methanol was mixed with 200 uL hydroxylamine hydrochloride in pyridine (1mg/mL). The reaction mixture was incubated at 70°C for 30 minutes and then dried under vacuum using a SpeedVac® centrifuge concentrator. The residue was dissolved in water and extracted with ethyl acetate. The excess reagent is water soluble. The extract was injected into a pre-equilibrated HPLC system for further purification. A. Results and Discussion Oxime derivatives have been widely studied and used in GC/MS methods to protect the keto functionality of steroids. Theonot et al,. studied the reaction kinetics of oxime formation with oxymethalonone (3 6). The oximes are Schilf bases and can easily be protonated in acidic solution. Under normal ESI conditions, when the steroid oxime was introduced in CH3OI-I, a peak representing the protonated molecule was observed with very little or no fiagmentation. Figure 5-22 shows the electrospray mass spectrum of testosterone oxime in methanol (100pg injected). When oxime derivatives of the model compounds were dissolved in acetonitrile and injected into a stream of 200 acetonitrile/HzO/HOAC (50/49.9/0.1%), a peak representing (M+CH3CN+H)+ was observed in addition to a peak for the protonated molecule. A significant peak for methanol adduct was not observed when methanol was used as the ESI solvent. Figure 5- 23 shows the ESI mass spectrum of testosterone oxime in CH3CN/HzO/HOAC (50/49.9/0.l % v/v). ESI/MS spectra of several anabolic steroid oximes were acquired in acetonitrile/HZO/HOAC. It was found that all the oximes show (M+CH3 CN+I-I)+ in addition to the (M+H)+. The intensities of peaks representing (M+CH3 CN+H)+ and (M’rl-I)+ ions varies with variables such as flow rates, concentrations, etc. This indicates that the oxime molecules can form an adducts with acetonitrile molecule. Therefore, this process leads to loss of sensitivity when ESI/MS is carried out in CH3CN/I-120 as the solvent system. At the same time, this process can be used to achieve additional selectivity by selecting proper solvents. Therefore, methanol is a better choice for the analysis of oximes by ESI/MS to obtain (M+H)"' as the major signal. The MS/MS analysis of (M+CH3CN+H)"' at a collision cell pressure of 3mtorr shows dissociation to the protonated molecule at a very low offset energy of 5eV. A significant fraction of acetonitrile solvent adduct can be dissociated in the presence of only collision gas without an offset energy. Figure 5-24 shows the ESI/CID mass spectrum of (M+CH3 CN+H)+ at 5eV offset energy and 3mtorr collision gas pressure. The CID studies show that the oxime derivatives form a weak interaction with CH3CN, but one strong enough to go through the capillary tube heated to 200°C. The adduct can be dissociated into the protonated molecule using a very low collision energy applied to it. Therefore, source C11) can help to decompose adduct ions formed with ACN. By selecting proper voltages for source CID, only the protonated molecule can be generated in the source region of the ESI/MS instrument. By this method, the abundance of the protonated molecule can be increased. 201 HO — NH3+ C1' + Hydroxylam'ne Hydrochloride Testosterone + HCl + H20 HO—‘N Testosterone Oxime Figure 5-21. Reaction scheme for the formation of testosterone oxime from testosterone and hydroxylamine hydrochloride. 202 + (M+H) 304.3 100 - 80— g 60. § 5 o .2. E 32 40- 20- 50 100 150 200 250 300 350 m/z Figure 5-22. ESI mass spectrum of methyltestosterone oxime in CH3 OH/I-120/I-IOAC (50/49.9/0.l % v/v/v) 203 + (M+CH3CN+H) 345.1 100 - + (M+H) 304.2 80 ~ 60 -1 i=1 0 ,> .8 34’ 4o — 20 ~ Figure 5-23. Portion of the ESI mass spectrum of methyltestosterone oxime in CH3CN/1120/I-IOAC (50/49.9/0.1 % v/v/v) 204 + (M+H) 304.3 100 ~ 80 g 60- ‘5’ ,3 E 34’ 40 201 + (M+CH 3CN+H) 345.1 “2 1‘, €4-1VL- T - f; . ,At‘?‘f‘;“: - .A‘- :Ma-L, L-kaf 9%,— 100 200 300 400 m/z Figure 5-24. ESI/CID/MS/MS spectrum of (M+CH3 CN+I-I)+ for testosterone oxime. The ofl‘set energy was set at 5eV and the collision energy at 3 mtorr (Ar) 205 B. Tandem Mass Spectrometric Studies of Oximes The protonated molecule of the testosterone oxime in CH3OH was subjected to collisionally induced dissociation to obtain the fragmentation pattern shown in Figure 5- 25. The MS/MS spectrum of testosterone oxime in CH3 OH shows several peaks related to the steroid skeleton that are common to the MS/MS spectra of oxime derivatives of testosterone, methyltestosterone, and nandrolone. The fragmentation pattern of the oximes did not yield much structural information that could distinguish homologous steroids. C. Detection Limits of Steroid Oximes The purified testosterone oxime derivative was dissolved in methanol and increasing amounts of derivative were injected into a stream of CH3 OH/HzO/HOAC (50l49.9/0.1 % v/v) at a flow rate of 10 uL/min. The detection limits in the low femtogram range (100 fg injected) were achieved in the selected ion monitoring mode of the TQMS. . 206 123.9 100 - 80, 111.91 13 60. t: 8 .s O .> 3 £2 404 (M+H)+ 304.1 20‘ 137.9 VAWVL‘AAA’AA- rfiféll Vfiw v T 50 "166' ' '1‘566 ' 266' ' 250 306' ' 330 m/z Figure 5-25. ESI/CID/MS/MS spectrum of testosterone oxime 207 VIII. Investigation of Stanozolol by ESI/MS A Introduction Although the synthetic anabolic steroid stanozolol (ST) has been available to the medical profession since 1961 and has been widely used owing to its reputation for very high anabolic to androgenic properties(3 7), the available literature covering analytical methodologies for its determination is limited. Reported methods for the determination of stanozolol in pharmaceutical formulations include gas chromatography (GC) (3 8), colorimetry (3 9), liquid chromatography/ultraviolet (LC/UV) (40), gas chromatography/mass spectrometry (GC/MS) (41-43 ), isotope dilution mass spectrometry (44), and high performance liquid chromatography/tandem mass spectrometry using atmospheric pressure chemical ionization (APCI) (45). The standard procedure for doping control in urine samples, based on enzymatic hydrolysis before derivatization and subsequent analysis by GC/MS, has proven difficult for ST. Since ST is structurally difi‘erent fi'om most anabolic steroids, its GC behavior is poor and its detection in urine is relatively difficult. Its pyrazole ring condensed into the androstane ring system induces difi‘ering GC behavior from that of related compounds. Another reason for the difficulty in its detection is due to its low excretion in urine. When the excretion of stanozolol was investigated with radio-labeled drug, only 16% of the radioactivity was excreted in the urine during the first day, while 40-60% was excreted in the feces (46). Derivatization techniques for GC/MS include preparation of N,O-bis, trimethylsilyl (N, O-bis-TMS) ether derivatives (47), the trimethylsilylether (OTMS) (46), the heptafluorobutyrarnide (NHFB) (46) and methylation (48). Masse et al. (49) and Donike et al. (43) extensively studied stanozolol metabolism and identified eleven metabolites in human urine samples. All the metabolites were identified as mono- or di-hydroxylated stanozolol analogs, most of which are excreted in 208 urine in the form of conjugates (49). The structure of stanozolol and its metabolites are shown in figure 5-26. Recently, Masse et al. reviewed the analysis of anabolic steroids including stanozolol using GC/MS. In this part of the dissertation, the analysis of stanozolol using positive electrospray mass spectrometry is described. The HPLC behavior, the extraction of a stanozolol- spiked urine sample, and its analysis by ESI/MS are discussed. B. Mass Spectral Analysis Stanozolol (ST) (Sigma chemical Co. St. Louis, MO) was dissolved in methanol (lOOpg/uL) and introduced into the ESI source. The ESI needle was held at 4.5kV and the heated capillary tube at 200°C. The sample (SuL) was flow injected into a stream of CH3OH/HZO/HOAC (50/49.9/0.1 % v/v/v) at a flow rate of 100 uL/min. ST produces a strong signal at m/z 329 that corresponds to the protonated molecule. The ST molecule has a basic site in its structure that causes difficulties during analysis by GC/MS. Due to the basic N atoms in the pyrazole ring, ST undergoes protonation easily under acidic conditions. The protonated molecule in solution is transferred to gas phase by electrospraying the solution. Figure 5-27 shows the electrospray mass spectrum of stanozolol that is an average of 5 scans. The mass spectrometer was scanned 200-500u in lsec. Under normal ESI conditions stanozolol undergoes little or no fragmentation. The ion current due to the ST is concentrated at (M+H)+, hence creating a sensitive and selective detection method for ST. Since the stanozolol molecules can be selectively detected by ESI/MS without forming derivatives, it will be very advantageous to use ESI/MS to detect stanozolol in complex biological samples. 209 OH “\s CH3 . 17 ““\ 11 \ C D 16 <——— 31 HN B 6 —>A>.— F igne 5-26. Structure of ambolic steroid stanozolol, and preferred metabolic hydroxylation sites indicated by an arrow. 210 + (Mm) 329 100~ 80‘ i? (it 3 .5 .3 E. ,8 40~ 20~ ~.‘ A fifi e '40:.L- fa“. .A‘.+Ae+ A A 200' '230 300 350 400 450 500 m/z Figure 5-27. ESI/MS mass spectrum of stanozolol in CH3OH and injected to a stream of CH3OH/H20/HOAC (50/49.9/0.1 % v/v/v) The capillary at 200°C and needle at 4.5kV. 211 C. Tandem Mass Spectrometry of Stanozolol Tandem mass spectrometry is a very useful technique for structural characterization of various molecules. The stanozolol metabolites are structurally similar compounds containing different numbers of hydroxyl groups at difl‘erent positions (49, 50). Henion et al. reported MS/MS properties of stanozolol metabolites by atmospheric pressure chemical ionization (APCI) mass spectrometry on a TQMS (45). The authors studied the fragmentation of stanozolol and three other metabolites and observed different CID tandem mass spectra. The tandem mass spectrometry technique can be used to identify difi‘erent metabolites in urine samples. In this study, standard solutions of stanozolol were used to study the ESI/MS/MS properties. The sample solution (lOpg/uL) was infirsed into the ESI source at a rate of 4p.L/min. The collision gas pressure was adjusted to 3 mtorr and the ofi‘set energy to 55eV (Lab). The mass spectrum obtained by ESI/MS/MS of stanozolol is given in Figure 5-28. The suggested fragmentation pathways are also given in Figure 5-29 (45). The fragrnentations are similar to those obtained by APCI mass spectrometry on the TQMS. The fi'agment ion at m/z 81, resulting from cleavage of the A-ring between carbon atoms C1/C2 and C3/C4 (possibly C1/C2 and C4/C5), dominates the tandem mass spectrum of stanozolol. Increasing the collision gas pressure or acceleration energy led only to changes in the abundances of the fragment ion at m/z 81 and parent ion at m/z 329; an increase of relative abundances for the other fi'agment ions was not accomplished with these changes. Henion et al. (45) compared the fragmentation patterns for 17- deuteromethyl stanozolol and unlabelled stanozolol to determine the ring cleavage positions, but they were not able to unambiguously assign the position of cleavage to give a specific fi'agment. 212 81 100 ‘ 80‘ g 60 § 5 95 .3 ... + % 107 (M+H) M 40. 121 329 20‘ 135 Figure 5-28. ESI/MS/MS mass spectrum of stanozolol. Collision energy was 55eV and the gas pressure was 3mtorr. 213 950 13.213). 109 (127-18) . 1--- 1 1 1 1 1 1 ‘ 1 1 1 1 1 1 1 1 a 1-- L43 (58+2H-16) 71(72-11) H+ : ' “1 163 : 1‘ 121 (163-42) : 135 ~, ---J : 93 (135-42) 43 --J 95 n}, 67 93 ('ZH) 149 ‘59 (57"2H) 107 (149-42) Figure 5-29. The possible fragmentation pathways of stanozolol molecule D. Comparison of ESI/MS/MS with FAB/MS/MS The high-energy F AB/CAD linked scan mass spectra were acquired on a JOEL HX 110 double focusing mass spectrometer. The CID energy was lOkV and He was used as the collision gas. The FAB/CAD/MS/MS mass spectrum (figure 5-30) shows a loss of 16 and 18 mass units from the protonated molecule, while ESI/MS/MS on the TQMS did not give an intense peak for a loss of H20. The higher mass end of the spectrum is significantly different from each other. ESI/MS/MS on the TQMS gave very weak peaks at high mass whereas the FAB/CAD/MS/MS spectrum shows a series of intense peaks at high mass. In the ESI/MS/MS spectrum of stanozolol from the TQMS, there is a series of intense peaks at the lower mass end of the spectrum. 214 (M+H)+ 10" R r 32.3 e 4 1 4 a 8‘ t 4 1 ‘ (NI‘l'I'I-I‘12('))+ v 4 e 6" A 4 b 4 U ‘d n 4 d l ' I 4 n 2. 257.3 c 110.9 ‘ J‘ 13 .9 9 J 1 .. 1 . ‘ l7 '0 ‘l' ‘ v r ' ' 1 fi' ' 50 100 150 200 250 300 “/2350 Figure 5-30. FAB/CAD/MS/MS mass spectrum of stanozolol 215 The ESI/MS/MS spectrum of stanozolol is similar to that obtained by APCI mass spectrometry on TQMS, but different from the FAB/CAD/MS/MS spectrum obtained on the double focusing mass spectrometer. These differences are due to the different energies employed in the collisionally induced dissociation process. B. Effect of Solvent Composition When stanozolol is dissolved in methanol and analyzed by ESI/MS, the major peak in the mass spectrum is due to (M+H)+. A significant peak for (M+CH3 OH+H)+ was not observed as shown in figure 5-27. When the mobile phase was changed to CH3CN/HzO/HOAC (50/49.9/O.1 % v/v) system, there were two major peaks observed in the ESI mass spectrum of ST, corresponding to (M+H)+ and (M+CH3CN+H)‘*' as shown in figure 5-31. The association of acetonitrile (CH3CN) as an adduct ion is weak but stable at the ESI/MS conditions used in this experiment. F. ESI/MS/MS of CH3CN Adduct Ion of ST The CH3CN adduct ion of ST was isolated at the Q1 and collected the CID mass spectrum at the Q3. The solvent adduct ion dissociates into the protonated molecule in the presence of 2 mtorr Ar gas in the collision cell (Q2), even with a small collision offset energy (5eV) as shown in figure 5-32. Even without the collision energy, a significant fiaction of (M+CH3 CN+H)+ ion current dissociates into the (M+H)+ ion. The adduct ion also can be dissociated into the protonated molecule by increasing the ofi‘set voltage by 20V on the octapole of the TQMS. Therefore, it is possible to form only (M+I-I)"' as the 216 major ion in the ESI/MS mass spectrum by inducing source-CID in the region before the skimmer in a single quadrupole mass spectrometer. + (M+ACN+H) 370 100‘ 80‘ g 60- r: o E Q) .2 E 0 a: 40‘ + 20* (101+H) 329 ,1, fl ,,,,,,,,,,, 11L,” 1 L 1 150 200 250 300 350 400 Figure 5-31. Portion of the ESI/MS mass spectrum of stanozolol in CH3OH as injected into a stream of CH3CN/I-IZO/HOAC (SO/49.9/0.1 % v/v/v)- The capillary was at 200°C and needle at 4.5kV. 217 + (M+H) 329 100 801 g 601 E 32’ E g" 40 20+ + (M+CH), CN+H) L 370 '''''' 1'66" ”H.20OTVYHH3IO071H40O m/z Figure 5-32. The ESI/MS/MS mass spectrum of the CH3CN adduct ion of ST. The ofl'set energy was at 5eV and the collision cell pressure was at 2 mtorr. 21 8 G. HPLC Analysis of Stanozolol The screening of stanozolol was carried out with a 4.6mm id x 15cm column packed with C13 particles (All tech, Deerfield, IL). The elution profile was monitored at 220nm. Two solvent systems were tested for reasonable peak shape. The methanol/water/TF A (50/49.9/0.1 % v/v/v) solvent system at a rate of lmI/min gave a broad peak for ST whereas CH3CN/HzO/TFA (50/49.9/0.l % v/v/v) at a rate of lmL/min gave a good peak at retention time of 4min. Therefore the CH3CN solvent system is better choice for a reasonable peak shape and resolution. H. LC/MS of Stanozolol Biological samples are mixtures of various compounds. Separation techniques coupled with mass spectrometry provides the necessary selectivity, sensitivity, and reliability required to identify mixtures. HPLC coupled with mass spectrometry provides the capability to analyze thermally labile compounds without extensive derivatization. A Vydac C13 column (2 mm x 25 cm.) was used in LC/MS analysis of stanozolol. A portion of 10 11L of stanozolol (100ng 11L) was injected and an isocratic run of CH3CN/I~120/'I'FA (40/59.9/0.1 % v/v) at a rate of 200 al./min was used to develop the chromatogram. Figure 5-33 shows the mass chromatograms for m/z 329 (M+I-I)+ and m/z 370 (M+CH3CN+H)+. The both peaks were present in the chromatogram and the intensity of the peak at m/z 370 is higher than that at m/z 329 in CH3CN. A tailing of the elution profile was observed due to the basic nature of the pyrazole ring of the stanozolol molecule. The sensitivity of stanozolol on the ESI/LC/MS was reduced using acetonitrile as the solvent due to the formation of adduct ion with ST. However, this ACN protocol 2 19 can be used to obtain additional selectivity by monitoring ions representing both (M+I-I)+ and (M+CH3CN+I-I)+ in the ESI/MS mass spectrum. 100 m/z 329 50 ' '3 t: 0 H G LA MA; A A 1—1 ._- -.. A L. 2 100 .15 34’ m/z 370 50‘ fi* """" s """"" s """""" s """"" s """"" I" Figure 5-33. The reconstructed mass chromatograms for (M-l-I-I)+ (m/z 329) and (M+CH3CN+H)+ (m/z 370) for stanozolol (1ng injected) during an LC/MS run. 220 I. Urine Extract A sample of urine from a male volunteer was collected and a 5mL of portion was spiked with stanozolol standard (Sng/mL). The sample was allowed to equilibrate for about one hour before extraction. A C13 cartridge (Waters, Milford, MA) was activated using 5 mL of methanol; and excess methanol was washed with 2 mL of water. The sample (5 mL) was passed through the cartridge at a rate of 20 mL/min and washed with 5 mL of water to remove water soluble urinary components. The cartridge was washed with 0.1M NH40H to improve the extraction efficiency as described by Massé et al (49). During elution of urine on the Sep-pak C13 cartridge, some of these compounds would be partially retained or adsorbed owing to specific interactions with the silica core of the stationary phase. Higher recoveries were obtained when treated with 0.1M NH40H after passing urine through the column and before elution of stanozolol (49). The stanozolol was eluted with 5 mL of methanol. The extract was evaporated to dryness under a stream of N2 and reconstituted in 500 uL of methanol. The urine extract was subjected to selected reaction monitoring (SRM) on the TQMS. The SRM is a selective technique to identify compounds based on specific fragment ions. The selected product ions can be monitored in the SRM mode of the TQMS. In this study, the most abunth product ion, m/z 81 was chosen for SRM. The following reactions were monitored in the SRM mode of the TQMS to detect stanozolol in the spiked urine sample; m/z 329 —> 329, and 329 —-> 81. Figure 5-34 shows the SRM profile of the above reactions for the urine extract. A portion of lOuL of urine extract was injected into a stream of CH3OH/H20/HOAC (50/49.9/O.1 %v/v) at a rate of 100 uUmin. The SRM mass spectrum of urine extract indicates the presence of stanozolol in the urine extract. The complete MS/MS mass spectrum of stanozolol was collected using the urine extract. The appearance of the spectrum was similar to that of 221 the standard sample. Next, the detection limits of ST were determined using positive electrospray mass spectrometry. It was observed that ST can be detected at the low femtogram level using selected reaction monitoring mode. IX. Conclusions In this report, the feasibility of forming oxime or hydrazone derivatives of anabolic ketosteroids for electrospray detection has been described. Under normal ESI conditions, hydrazone derivatives of ketosteroids gave only a single peak in the mass spectrum representing the intact molecule. The feasibility has been demonstrated for detecting these derivatives at the low-femtogram level in biological fluids. The hydrazone formation reaction was investigated in detail because it is a promising candidate for method development in determining anabolic ketosteroids in biological fluids. Tandem mass spectrometry (MS/MS) combined with ESI/HPLC/MS is a valuable tool for identifying the steroids present in biological fluids by combining fragmentation patterns with retention time and molecular weight information. Collisionally induced dissociation studies of hydrazone derivatives demonstrated a major loss of 59u fiom the molecular ions of model compounds. The characteristic loss of 59u can be used in the neutral loss scanning mode to obtain additional selectivity for the analyte. The neutral loss scanning mode can be used to discriminate against the biological matrix as shown above. This method has a high potential for detecting low levels of anabolic ketosteroids present in biological samples. The oxime derivatives of anabolic steroids show an enhanced response in the ESI/MS analysis. The selection of CH3CN for the solvent system for oxime analysis is very important since they form an adduct with acetonitrile. The source CID can be used to decompose the adduct ions to obtain (M+I-I)+ as the major ion. 222 The stanozolol molecule has a very basic pyrazole ring which makes analysis by GC/MS difficult. In ESI/MS, the pyrazole ring can be protonated in acidic solution and transferred to the gas phase without significant decomposition. Stanozolol can be detected with high sensitivity using ESI/MS. Detection limits at the low femtogram level were achieved with the selected reaction monitoring mode. Since stanozolol forms an adduct with acetonitrile, care must be taken to select the correct solvents for analysis. Tandem mass spectrometry combined with liquid chromatography can be used to confirm the presence of stanozolol in biological samples. 223 g SRM329 ——>31 0 .5 50. O .2. 1.5 0 ad 50 100 150 200 250 300 100 H ,5; SRM329 —> 329- ,3, 50« E 0 ..>. .‘é’ & so 100 150 200 250 300 scan # Figure 5-34. Selected reaction monitoring profile of stanozolol in urine extract (lOuL). The solvent system was CH3OH/HzO/HOAC (50/49.9/O.l %v/v/v) at a rate of lOOuL/min. 224 X. References 10. ll. 12. l3. 14. R H. Strauss, Drugs and Performance in Sports, W. B. Saunders, Philadelphia (1987) T. Tobin, Drugs and Performance in Horse, Charles C. Thomas, Springfield, Illinois (1981) H. A. Haupt and G. D. Rovere, Am. J. Sports Med 1984, 12, 469 W. M. Muck and J. D. Henion Biomed Environ. Mass Spectrom. 1990, 19, 37 Sample, R. H. B. and Baenziger, J. C. In Analytical Aspects of Drug Testing, D Deutch, ed., Wiley, New York, pp 247-272 (1988) Goudreault, D.; Masse, R. J. Steroid Biochem. Molec. Biol. 1990, 3 7, 137 Gaskell, S. J.; "Analysis of Steroids" In Methods of Biochemical Analysis, Vol. 29, D. Glick, ed., Wiley (interscience), New York, NY, pp 385-434 (1983) Curcuruto, 0.; Franchi, D.; Hamdan, M.; Favretto, D.; Traldi, P. Rapid Commun. Mass Spectrom. 1993, 7, 673 Shacketon, C. H. J. Steroid Biochem. Mol. Biol. 1993, 45, 127 Watson, D.; Taylor, G. W.; Murray, S. Biomed Environ. Mass Spectrom. 1986, 13, 65 Weidolf, L. O. G.; Henion, J. D. Biomed Environ. Mass. Spectrom. 1988, 15, 283 Kletke, C.; Roitman, E.; Bitsch, P.; Shaldeton, C. Proceeding of 40th ASMS Conference on Mass Spectrometry and Allied Topics, Washington DC, May 31- June 5, 1992 Hatton, C. K.; Catlin, D. H. Clin. Lab. Med, 1987, 7, 655-668 D. Goudreault; R. Masse J. Steroid Biochem. Molec. Biol. 1990, 37, 137 15. 16. 17. l8. 19. 20. 21. 22. 23. 24. 25. 26. 27. 28. 29. 30. 31. 32. 33. 225 B. Chung; H-Y. P. Choo; T. Kim; K. Born; 0. Kwon; J. Suh; J. Yang; J. Park J. Anal. Toxicol. 1990, 14, 91 Honour, J. W.; Brooks, C. J. W.; Shackleton, C. H. L. Biomed Mass Spectrom. 1982, 9, 505 Bjorkhem, 1.; Ek, H. J. steroid Biochem. 1983, 18, 481 Bjorkhem, 1.; Lantoo, 0.; Lof, A. J. steroid Biochem. 1980, 13, 481 Moss, M. S.; Cowan, D. A , in Clarke's Isolation and Identification of Drugs, 2nd Ed, ed. by Mofi‘at, A. C, Pharmaceutical Press, London 1986 Masse, R.; Ayotte, Dugal, R. J. Chromatogr.1989, 489, 23 Shakleton, C. H. L.; Whitney, J. 0. Clin. Chim. Acta 1980, 107, 231 Kruger, T. L.; Litton, R. W.; Kondrat, R. W. Cooks, R. G. Anal. Chem. 1976, 48, 2113 K. Kayganich; J. T. Watson; C. Kilts; J. Richie Biomed Environ. Mass. Spectrom., 1990, 19, 341 H. K. Mayer; W. Reusch; J. T. Watson Org. Mass Spec. 1992, 27, 560 Girard, A.; Sandulesco, G. Helv. Chim. Acta. 1936, 19, 1095 Wheeler, 0. H. J. Chem. Edu. 1968, 45, (6)435 Bruins, A. P.; Covey, T. R.; Henion, J. D. Anal. Chem. 1987, 59, 2642 Conboy, J. J.; Henion, J. D.; Martin, M. W.; Zweigenbaun, J. A. A. Anal. Chem. 1990, 62, 800 Fenn, J. B.; Mann, M.; Meng, C. K; Wong, S. F.; Whitehouse, C. M. Mass Spectrometry Reviews, 1990, 9, 37 Kondradt, R. W.;Cooks, R. G. Anal. Chem. 1978, 50, 81A McLarferty, F. W.; Bockhofl‘, F. W. Anal. Chem. 1976, 50, 69 Yost, R. A.; Enke, C. G. Anal. Chem. 1979, 5], 1251A Glish, G. L.; Shaddock, V. M.; Harmon, K.; Cooks, R. G. Anal. Chem. 1980, 52, 165 34. 35. 36. 37. 38. 39. 40. 41. 42. 43. 45. 46. 47. 48. 226 Brotherton, H. 0.; Yost, R. A Anal. Chem. 1983, 55, 549 Johnson, J. V.; Yost, R. A Anal. Chem. 1985, 5 7, 758A Thenot, J-P.; Homing, E. C. Anal. Lett. 1972, 5(1), 21 Bates. P. C.; Chew. L. P.; Milward. D. J. Endocrinol. 1987, 114, 373 Magin. D. F. J. Chromatogr. 1975, 115, 687 Shingbal. D. M.; Barad. U. G.; J. Assoc. Ofii Chem, 1985, 68, 98, Cavrini. V.; Pietra. M.; Raggi. M. A; Mioli. M. G. Analyst, 1987, 112, 1671 Rossi, S.-A.; Johnson, J. V.; Yost, R. A Biol. Mass Spectrom. 1992, 21, 420 Schanzer, W.; Donike, M. Anal. Chim. Acta. 1993, 23, 275 Schnzer, W.; Opfermann, G.; Donike, M. .1. Steroid Biochem. 1990, 153, 36 Lantto, 0.; Bjorhem, 1.; Ek. H.; Johnson, D. J. Steroid Biochem. 1981, 14, 721 Muck. W. M.; Henion. J. D Biomed Environ. Mass Spectrum. 1990, 19, 37 Choo, H-Y. P.; Kwon, O-S.; Park, J. J. Anal. Toxicol. 1990, 14, 109 Masse. R.; Ayotte. C.; Bi. H.; Dugal. R J. Chromatogr. 1989, 497, 17 Schanzer, W.; Opfermann, G.; Donike, M. Proceedings of 36th ASMS Conference on Mass Spectrometry and Allied Topics, San Francisco, CA, June 5- June 10, 1988, 397