Simultaneous Analysis of Intact Human Insulin and Five Analogs in Human Plasma Using μelution SPE and a CORTECS UPLC Column

Similar documents
Reducing Sample Volume and Increasing Sensitivity for the Quantification of Human Insulin and 5 Analogs in Human Plasma Using ionkey/ms

Development of a High Sensitivity SPE-LC-MS/MS Assay for the Quantification of Glucagon in Human Plasma Using the ionkey/ms System

Development of a Bioanalytical Method for Quantification of Amyloid Beta Peptides in Cerebrospinal Fluid

Dr. Erin E. Chambers Waters Corporation. Presented by Dr. Diego Rodriguez Cabaleiro Waters Europe Waters Corporation 1

Analysis of Testosterone, Androstenedione, and Dehydroepiandrosterone Sulfate in Serum for Clinical Research

UPLC-MS/MS Analysis of Azole Antifungals in Serum for Clinical Research

Removal of Triton X-100 from Plasma Samples Using Mixed-Mode Solid Phase Extraction (SPE)

UPLC/MS Monitoring of Water-Soluble Vitamin Bs in Cell Culture Media in Minutes

Rapid Analysis of Water-Soluble Vitamins in Infant Formula by Standard-Addition

O O H. Robert S. Plumb and Paul D. Rainville Waters Corporation, Milford, MA, U.S. INTRODUCTION EXPERIMENTAL. LC /MS conditions

Matrix Factor Determination with the Waters Regulated Bioanalysis System Solution

A High Sensitivity UPLC/MS/MS Method for the Analysis of Clopidogrel and Clopidogrel Carboxylic Acid Metabolite in Human K 2 EDTA Plasma

Challenges in Developing an Ultra-Sensitive Bioanalytical Method for Ethinylestradiol in Human Plasma

Rapid, Simple Impurity Characterization with the Xevo TQ Mass Spectrometer

A Definitive Lipidomics Workflow for Human Plasma Utilizing Off-line Enrichment and Class Specific Separation of Phospholipids

A Novel Solution for Vitamin K₁ and K₂ Analysis in Human Plasma by LC-MS/MS

Analysis of Rosuvastatin in Dried Blood Spot and Plasma Using ACQUITY UPLC with 2D Technology

Direct Analysis of Urinary Opioids and Metabolites by Mixed-Mode µelution SPE Combined with UPLC/MS/MS for Forensic Toxicology

Performance of an ultra low elution volume 96-well plate

Method Development for the Analysis of Endogenous Steroids Using Convergence Chromatography with Mass Spectrometric Detection

Applying a Novel Glycan Tagging Reagent, RapiFluor-MS, and an Integrated UPLC-FLR/QTof MS System for Low Abundant N-Glycan Analysis

[ APPLICATION NOTE ] High Sensitivity Intact Monoclonal Antibody (mab) HRMS Quantification APPLICATION BENEFITS INTRODUCTION WATERS SOLUTIONS KEYWORDS

Rapid Analysis of Bisphenols A, B, and E in Baby Food and Infant Formula Using ACQUITY UPLC with the Xevo TQD

Quantification of Budesonide Using UPLC and Xevo TQ-S

Providing a Universal, One-step Alternative to Liquid-Liquid Extraction in Bioanalysis

[ APPLICATION NOTE ] A Generic Kit-Based Approach for LC-MS/MS Quantification of Urinary Albumin for Clinical Research APPLICATION BENEFITS

[ APPLICATION NOTE ] Oasis PRiME HLB Cartridge for Cleanup of Infant Formula Extracts Prior to UPLC-MS/MS Multiresidue Veterinary Drugs Analysis

A RAPID AND SENSITIVE UPLC/UV/MS METHOD FOR SIMVASTATIN AND LOVA S TAT IN IN SU P P O RT O F C L E A NING VA L I DAT IO N S T U DIES

Rapid Lipid Profiling of Serum by Reverse Phase UPLC-Tandem Quadrupole MS

Michael Blackburn HOW LOW CAN YOU GO: DRIVING DOWN LIMITS OF HYBRID IA-LC/MS. European Bioanalytical Forum. 19 th November 2015

Dienes Derivatization MaxSpec Kit

Meeting Challenging Requirements for the Quantitation of Regulated Growth Promoters Dexamethasone and Betamethasone in Liver and Milk

A Robustness Study for the Agilent 6470 LC-MS/MS Mass Spectrometer

[ APPLICATION NOTE ] UPLC-MS/MS Analysis of 45 Amino Acids Using the Kairos Amino Acid Kit for Biomedical Research INTRODUCTION APPLICATION BENEFITS

SEPARATION OF BRANCHED PFOS ISOMERS BY UPLC WITH MS/MS DETECTION

A RAPID AND SENSITIVE ANALYSIS METHOD OF SUDAN RED I, II, III & IV IN TOMATO SAUCE USING ULTRA PERFORMANCE LC MS/MS

LC-MS/MS Method for the Determination of Tenofovir from Plasma

Robust extraction, separation, and quantitation of structural isomer steroids from human plasma by SPE-UHPLC-MS/MS

Analysis of the Non-Ionic Surfactant Triton-X Using UltraPerformance Convergence Chromatography (UPC 2 ) with MS and UV Detection

Rapid and Accurate LC-MS/MS Analysis of Nicotine and Related Compounds in Urine Using Raptor Biphenyl LC Columns and MS-Friendly Mobile Phases

Detection of Cotinine and 3- hydroxycotine in Smokers Urine

The setup and validation of an LC-MS/MS assay of Androstenedione & Testosterone a labs experience

Vitamin D Metabolite Analysis in Biological Samples Using Agilent Captiva EMR Lipid

Analysis of anti-epileptic drugs in human serum using an Agilent Ultivo LC/TQ

Determination of Clarithromycin in Human Plasma by LC-EI Tandem Mass Spectrometry: Application to Bioequivalence Study

[ APPLICATION NOTE ] APPLICATION BENEFITS INTRODUCTION WATERS SOLUTIONS KEYWORDS

Determination of 6-Chloropicolinic Acid (6-CPA) in Crops by Liquid Chromatography with Tandem Mass Spectrometry Detection. EPL-BAS Method No.

SPE-LC-MS/MS Method for the Determination of Nicotine, Cotinine, and Trans-3-hydroxycotinine in Urine

Profiling Flavonoid Isomers in Highly Complex Citrus Juice Samples Using UPLC Ion Mobility Time-of-Flight Mass Spectrometry

Comprehensive Study of SLE as a Sample. Preparation Tool for Bioanalysis

PosterREPRINT SIMULTANEOUS QUANTIFICATION OF PSYCHOTHERAPEUTIC DRUGS IN HUMAN PLASMA BY TANDEM MASS SPECTROMETRY

LC-MS/MS analysis of Chlorates in Milk and Whey Powder using the Agilent 6470 QQQ

John Haselden 1, Gordon Dear 1, Jennifer H. Granger 2, and Robert S. Plumb 2. 1GlaxoSmithKline, Ware, UK; 2 Waters Corporation, Milford, MA, USA

Determination of Amantadine Residues in Chicken by LCMS-8040

Determination of β2-agonists in Pork Using Agilent SampliQ SCX Solid-Phase Extraction Cartridges and Liquid Chromatography-Tandem Mass Spectrometry

Validation of a Reliable LC-MS/MS Method for Analysis of Simultaneously in Clinical Laboratory

LC-MS/MS for the quantification of Peptide biomarker and mixture of closely related Protein in formulation

Extraction of 25-hydroxy Vitamin D from Serum Using ISOLUTE. PLD+ Prior to LC-MS/MS Analysis

Supporting information

Neosolaniol. [Methods listed in the Feed Analysis Standards]

Jose Castro-Perez, Henry Shion, Kate Yu, John Shockcor, Emma Marsden-Edwards, Jeff Goshawk Waters Corporation, Milford, MA, U.S. and Manchester, UK

4.5 Minute Analysis of Benzodiazepines in Urine and Whole Blood Using LC/MS/MS and an Ultra Biphenyl Column

Extraction of a Comprehensive Steroid Panel from Human Serum Using ISOLUTE. SLE+ Prior to LC/MS-MS Analysis

Comparison of a UPLC Method across Multiple UHPLC Systems

Dry eye disease commonly known as atopic keratoconjunctivitis is an autoimmune disease of

Abstract. Introduction

Reduced Ion Suppression and Improved LC/MS Sensitivity with Agilent Bond Elut Plexa

The Raptor HILIC-Si Column

Using Hydrophilic Interaction Chromatography (HILIC) for the Retention of Highly Polar Analytes

The Comparison of High Resolution MS with Triple Quadrupole MS for the Analysis of Oligonucleotides

A Comprehensive Screening of Illicit and Pain Management Drugs from Whole Blood Using SPE and LC/MS/MS

Finnur Freyr Eiríksson

Author. Introduction. Abstract

Paul D. Rainville Ph.D. Health Sciences Group Waters Corporation Waters Corporation 1

Quantitative Analysis of Vit D Metabolites in Human Plasma using Exactive System

Application Note. Author. Abstract. Introduction. Food Safety

Supplementary Information

Integration of steroids analysis in serum using LC-MS/MS with full-automated sample preparation

Analyze Barbiturates in Urine with Agilent 6430 LC/MS/MS and Poroshell 120 EC-C18

High-Throughput, Cost-Efficient LC-MS/MS Forensic Method for Measuring Buprenorphine and Norbuprenorphine in Urine

Quantification of lovastatin in human plasma by LC/ESI/MS/MS using the Agilent 6410 Triple Quadrupole LC/MS system

Author. Introduction. Small Molecule Pharmaceuticals & Generics

Lipid Class Separation Using UPC 2 /MS

Analysis of Food Sugars in Various Matrices Using UPLC with Refractive Index (RI) Detection

DETERMINATION OF CANNABINOIDS, THC AND THC-COOH, IN ORAL FLUID USING AN AGILENT 6490 TRIPLE QUADRUPOLE LC/MS

Extraction of Multiple Mycotoxins From Grain Using ISOLUTE Myco prior to LC-MS/MS Analysis

Extraction of Multiple Mycotoxins From Nuts Using ISOLUTE Myco prior to LC-MS/MS Analysis

Jody Dunstan, 1 Antonietta Gledhill, 1 Ailsa Hall, 2 Patrick Miller, 2 Christian Ramp 3. Waters Corporation, Manchester, UK 2

INTRODUCTION CH 3 CH CH 3 3. C 37 H 48 N 6 O 5 S 2, molecular weight Figure 1. The Xevo QTof MS System.

Determination of Benzodiazepines in Urine by CE-MS/MS

Ultra Performance Liquid Chromatography Coupled to Orthogonal Quadrupole TOF MS(MS) for Metabolite Identification

Analysis of N-Linked Glycans from Coagulation Factor IX, Recombinant and Plasma Derived, Using HILIC UPLC/FLR/QTof MS

High Throughput Extraction of Opiates from Urine and Analysis by GC/MS or LC/MS/MS)

LC/MS/MS of Trichothecenes and Zearalenone in Wheat Using Different Sample Prep Methods

Paul Silcock 1, Anna Karrman 2, and Bert van Bavel 2. Aim. Introduction. Sample preparation

Analysis of Cholesterol-Lowering Drugs (Statins) Using Dried Matrix Spot Technology

Fast and simultaneous analysis of ethanol metabolites and barbiturates using the QTRAP 4500 LC-MS/MS system

Rapid and Robust Detection of THC and Its Metabolites in Blood

Transcription:

Simultaneous Analysis of Intact Human Insulin and Five Analogs in Human Plasma Using μelution SPE and a CORTECS UPLC Column Erin E. Chambers and Kenneth J. Fountain Waters Corporation, Milford, MA, USA APPLICATION BENEFITS CORTECS UPLC Column for highest sensitivity and narrowest peaks widths Selective, fast extraction without timeconsuming affinity purification Quantitative accuracy; more accurate and precise than traditional LBA methods High sensitivity Specificity, including differentiation between human insulin and insulin lispro Simultaneous quantification of human insulin, glargine, lispro, aspart, glulisine, and detemir Quantification without digestion Excellent asset utilization; allows the use of current LC/MS instrumentation WATERS SOLUTIONS ACQUITY UPLC I-Class System with 2D Technology CORTECS UPLC C 18 + Columns Oasis MAX 96-well µelution Plates Xevo TQ-S Mass Spectrometer MassLynx Software KEY WORDS Non-specific binding, large peptides, Oasis, sample preparation, bioanalysis, quantification, insulin, glargine, lispro, UPLC, CORTECS, plasma INTRODUCTION Insulin is perhaps one of the best known and earliest peptide therapeutics. Multiple long and fast-acting analogs have also been developed, and a patient may often be prescribed one of each for diabetes control. Quantification of biologics, such as insulins, has historically been performed using ligand binding assays (LBAs) such as ELISAS. LC/MS/MS, however, has certain advantages over LBAs, such as shorter development times, higher accuracy and precision, the ability to multiplex, no cross-reactivity, and the ability to readily distinguish between closely related insulins. Intact insulins are particularly difficult to analyze by LC/MS/MS, as MS sensitivity is low due to poor transfer into the gas phase, and poor fragmentation patterns exist due to the presence of multiple stabilizing disulfide bonds. In addition, insulin and its analogs suffer from nonspecific binding and poor solubility, making LC and sample preparation method development difficult. A few LC/MS/MS methods do exist; however, most of those methods involve time-consuming and laborious immunoaffinity purification and/or nano-flow LC. Distinguishing between human insulin and insulin lispro (Humalog) is a very specific challenge for quantifying insulins, as they differ by a simple reversal in the position of two amino acids. Only a single low-molecular weight fragment differentiates the two, making selective sample preparation and chromatography critical. This study takes advantage of mixed-mode solid-phase extraction (SPE) and a high-efficiency, solid-core particle column that contains a low-level positive surface charge to eliminate interferences while facilitating high sensitivity quantification. Furthermore, selectivity studies show that the presence of high levels of human insulin, such as one might encounter in type II diabetic patients, does not interfere with quantification of lispro or any of the other analogs. This work provides a single, simple method for the simultaneous, direct quantification of intact human insulin and multiple insulin analogs in human plasma (Figure 1), achieving LODs of 50 to 200 pg/ml for each. Average accuracy for standard curve points was 99% to %. Average inter- and intra-day accuracies for QC samples were 98% and 94%, respectively. Average inter- and intra-day precisions for QC samples were 7.5% and 5.3%, respectively. Matrix factors for all analogs were calculated in six sources of human plasma and CVs of matrix factors were <15% in all cases, further supporting the selectivity of the method. 1

E X P E R IM E N TA L Sample preparation Step 1: Protein precipitation (PPT) 25 µl of bovine insulin (internal standard, final concentration 10 ng/ml) was added to 250 µl human plasma and mixed. Samples were precipitated with 250 µl 1:1 methanol/acetonitrile containing 1% acetic acid, and centrifuged for 10 minutes at 13,000 rcf. The supernatant was transferred to a 2-mL 96-well plate containing 900 µl of 5% concentrated NH 4 OH in water (v:v) and mixed. Step 2: SPE using an Oasis MAX µelution 96-well Plate Condition: 200 µl methanol Human insulin Humalog (insulin lispro) Lantus (insulin glargine) Insulin B chain Insulin B chain Equilibrate: Load sample: Wash: 200 µl water Entire diluted PPT supernatant was loaded onto the extraction plate in two steps of approximately 700 µl each 200 µl 5% NH 4 OH in water Novalog (insulin aspart) Insulin B Chain Wash: 200 µl 5% methanol + 1% acetic acid Elute: 2 X 25 µl 60:30:10 methanol/water/acetic acid Dilute: 50 µl water Inject: 30 µl Levemir (insulin detemir) Insulin B Chain Insulin B chain Apidra (insulin glulisine) Insulin B chain Figure 1. Structures for human insulin and the insulin analogs quantified in this application. 2

UPLC conditions System: ACQUITY UPLC I-Class with 2D Technology, configured for at-column dilution with trap and back elution Analytical column: CORTECS UPLC C 18 + 1.6 µm, 2.1 x 50 mm (p/n 186007114) Trap column: XBridge C 18 IS 3.5 µm, 2.1 x 20 mm (p/n 186003019) Elution mobile Phase A: 0.1% HCOOH in water Elution mobile Phase B: 0.1% HCOOH in acetonitrile Loading solvent: 85:15 mobile phase A:B; 0.1 ml/min for first two minutes, reverse valve, then clean trap column with ramp from 4 to 9 B Dilution solvent: % mobile phase A, 0.3 ml/min for first two minutes Valve position: Initial position one (forward loading of trap), switch to position two at two minutes (back elute of trap onto analytical column), back to position one at 6.5 minutes Gradient: Load for two minutes; switch valve and back elute from trap column onto analytical column with a linear gradient from 15% to 4 B over four minutes MS conditions Mass spectrometer: Ionization mode: Capillary voltage: Xevo TQ-S ESI positive 3.0 kv Desolvation temp.: 600 C Cone gas flow: Desolvation gas flow: Collision cell pressure: Collision energy: Cone voltage: Specific insulin MRM transition 150 L/h 0 L/h 3.8 X 10 (-3) mbar Optimized by component, see Table 1 Optimized by component, see Table 1 Cone voltage (V) Collision energy (ev) Ion type Glargine 1011 1179 60 25 Lispro 1162 217 50 40 Detemir 1184 454.4 60 20 y2 Aspart 971.8 660.8 60 18 y11 Glulisine 1165 1370 14 22 Bovine (IS) 956.6 1121.2 60 18 Human insulin 1162 226 50 40 Table 1. MS conditions for human insulin, insulin analogs, and the internal standard bovine insulin. Elution flow rate: 0.25 ml/min Column temp.: 60 C Sample temp.: 15 C Injection volume: 30 µl Data management Chromatography software: Quantification software: MassLynx TargetLynx Run time: Collection plates: 8.5 minutes Waters 1-mL ACQUITY collection plates 3

RESULTS AND DISCUSSION Mass spectrometry Several multiple charged precursors were observed for each of the analogs, typically the two most abundant were selected for CID, and either one or two diagnostic fragments monitored during method development. Representative MS/MS spectra at the optimal collision energies for the primary transitions chosen for quantification are shown in Figures 2A and 2B. In several cases, higher intensity MRM transitions existed than the one that was ultimately chosen. Specifically, most insulins will produce very intense immonium ion fragments. Glargine, for example, produces a tyrosine immonium ion fragment at 136. However, tests in extracted plasma demonstrated that the use of higher precursor and fragment pairs (Table 1) yielded significantly improved specificity in matrix, outweighing any apparent sensitivity difference (Figure 3). This facilitates the use of analytical scale LC and traditional SPE methodologies versus nano-scale chromatography and affinity-based purification schemes previously reported. Human insulin and lispro, however, share almost complete overlap in their fragmentation patterns due to a simple reversal in the positions of amino acids 28 and 29 in the B chain (Figure 1). A single low-molecular weight fragment differentiates the two, with human insulin yielding a fragment at 226 and lispro producing a fragment at 217 (Figure 2B), both arising from the last two amino acids in the B chain. Without baseline chromatographic separation, the MS must rely on these non-specific fragments for quantification, making both selective sample preparation and high efficiency chromatography crucial. % 0 Insulin glulisine MSMS of 5+ 1165 1161 1161 1162 1370 1369 1370 1370 1162 346 1161 1369 1426 346 1165 1344 328 347 1160 1166 1371 1427 301 447 1046 512 961 1092 1470 1689 1739 200 300 400 500 600 700 800 900 0 1 1200 1300 1400 1500 1600 1700 1800 % 973 969 136 968 1139 661 925 5 1123 1263 1301 1403 0 200 300 400 500 600 700 800 900 0 1 1200 1300 1400 1500 1600 1700 1800 1180 % 0 % 454 Insulin detemir MSMS of 5+ 1184 971 972 972 200 300 400 500 600 700 800 900 0 1 1200 1300 1400 1500 1600 1700 1800 1011 143 8 983 1164 1179 0 200 300 400 500 600 700 800 900 0 1 1200 1300 1400 1500 1600 1700 1800 1184 Insulin aspart MSMS of 6+ 972 1362 1366 Insulin glargine MSMS of 5+ 1011 Figure 2A. MS/MS spectra from CID of the optimal precursors for insulin glulisine, aspart, detemir, and glargine. Asterisks indicate the fragments that were chosen for quantification. 4

226 Human insulin MSMS of 5+ 1163 345 345 219 292 227 327 652 248 201 346 652 446 653 808 405 609 711753 758 217 217 927 960 877 1129 1065 1130 1331 1217 1241 1356 1159 1400 15171571 200 300 400 500 600 700 800 900 0 1 1200 1300 1400 1500 1600 1700 1800 Insulin lispro MSMS of 5+ 1163 213 219 1162 1159 1163 230 1159 1391 1410 270 292 652 1114 1331 345 361 1065 1185 1421 1298 446 948 1497 200 300 400 500 600 700 800 900 0 1 1200 1300 1400 1500 1600 1700 1800 Figure 2B. MS/MS spectra from 5+ precursors of human insulin and insulin lispro. Asterisks indicate the fragments that were chosen for quantification. 0.99 867 > 136 (Lantus) 9.40e5 0.56 0.99 1.20 1.30 1.49 1.15 1.40 1.58 1.68 867 136 (tyrosine immonium ion) Lack of specificity 0.20 0.40 0.60 0.80 1.00 1.20 1.40 1.60 1.80 2.00 2.20 2.40 2.60 2.80 3.00 3.20 3.40 1: MRM of 4 channels ES+ 867 > 984 (Lantus) 8.97e5 867 984 0.20 0.40 0.60 0.80 1.00 1.20 1.40 1.60 1.80 2.00 2.20 2.40 2.60 2.80 3.00 3.20 3.40 Time Figure 3. Extracted ion chromatograms from two different transitions for insulin glargine. Use of the immonium ion fragment (top panel) shows a lack of specificity relative to the use of a higher precursor and fragment pair (bottom panel.) 5

Liquid chromatography Unlike small molecules, larger peptides and small proteins, i.e., insulins, suffer from poor mass transfer in and out of fully-porous particles. Thus, using a column packed with solid-core particles allows for sharper peak shapes at the higher flow rates typically needed for bioanalytical studies. 1,2 Specifically for insulins, it is documented that using a column packed with particles containing a low-level positive surface charge gives superior peak shape and resolution to other columns. 3 CORTECS C 18 + Columns combine the benefits of solid-core particle technology and a low-level positive surface charge to obtain a new level of separation performance for this application. With CORTECS C 18 +, insulin peaks are typically 4.0 to 4.5 seconds wide with 2X higher area counts than state-of-the-art, fully-porous particle columns. Representative chromatograms are shown in Figure 4. The use of multidimensional chromatography facilitated higher injection volumes, through the use of at-column dilution (ACD), while providing additional sample cleanup by employing a trap and back-elute strategy. The combination of these chromatographic system elements allowed us to inject up to 40 µl without experiencing insulin breakthrough. Chromatographic breakthrough was evident on a 1D system with injection volumes >10 µl (data not shown). Insulin detemir RT 5.52 min 4.29 4.30 3.60 5.52 Insulin glulisine RT 4.29 min 5.26 Human insulin RT 4.30 min 5.26 5.46 Time 3.60 4.28 4.13 4.27 5.81 5.28 5.73 4.04 5.78 4.23 3.21 5.19 Insulin glulisine RT 4.28 min Insulin glargine RT 4.13 min Insulin aspart RT 4.27 min Bovine insulin (IS) RT 4.23 min Time Figure 4. UPLC/MS/MS chromatogram for human insulin, five-insulin analogs, and bovine insulin (IS). 6

Sample preparation A proof-of-concept study published earlier in 2013 3 used reversed-phase only SPE to extract four insulin analogs. Although the method clearly demonstrated feasibility, endogenous background was high in the transitions for human insulin and lispro, with average LLOQs of 200 to 500 pg/ml reported for the four insulins, making accurate low-level quantification and distinction between the lispro and human insulin impossible. This application presents a significantly more selective two-step extraction utilizing a protein precipitation (PPT) pre-treatment step and a strong anion-exchange mixed-mode SPE device to effectively reduce endogenous background, as well as improve specificity and detection limits. During optimization of protein precipitation conditions for insulin, various ratios of organic solvent to plasma, as well as different precipitation solvents were tested. Protein precipitation (PPT) with a 1:1 ratio of various solvents (ACN or ACN modified with one of the following: 5% NH 4 OH, 1% acetic acid, or 2% formic acid) resulted in >9 recovery of the six insulins without precipitating the peptides themselves. Protein precipitation with higher ratios of organic resulted in peptide loss due to undesired precipitation of the insulins. The PPT pre-treatment effectively eliminated protein binding and reduced endogenous interferences from large proteins such as albumin. The PPT supernatants were then diluted with 5% concentrated NH 4 OH to bring the ph to ~11. At this ph, insulin and its analogs will carry a net negative charge, putting them in the proper charge state to bind to Oasis MAX by ion exchange. A 2007 study 4 determined that mixed-mode SPE (especially in its ion-exchange elution) was significantly more selective than reversed-phase only SPE. For this assay specifically, where the use of low fragments for human insulin and lispro was necessary, it was imperative to have the most selective sample preparation possible. The diluted supernatant was then applied to conditioned SPE plates, and analytes were well retained on the SPE sorbent during the basic ph load step. Optimization of the elution solution was critical to fully elute the insulins, maintain solubility, and minimize interferences from the plasma matrix. The optimum elution solution was 6 organic with 1 acetic acid. The final eluate was diluted with an equal volume of water. 7

Linearity, accuracy, and precision To generate standard curves, human plasma was fortified with a mixture of the insulins at the following final concentrations: 50,, 200, 500, 0, 2000, 5000, and 10,000 pg/ml. Quality control (QC) samples were prepared in human plasma at the following concentrations: 150, 750, 2500, and 7500 pg/ml. Bovine insulin (final concentration 10 ng/ml) was used as the internal standard (IS) for all insulins. Peak area ratios (PARs) of the analyte peak area to the IS peak were calculated. Calibration curves, prepared in human plasma, were constructed using PARs of the calibration samples by applying a one/concentration weighted linear regression model. All QC sample concentrations were then calculated from their PARs against their respective calibration lines. All curves were linear using the 1/x regression. A summary of standard curve performance for all insulins is shown in Table 2. For human insulin, the basal concentration in pooled or individual control plasma was determined by calculating the x-intercept. The basal level of human insulin (average = 1937 pg/ml) was then added to the spiked concentration for all standard curve and QC samples to enable accurate quantification. A representative standard curve for insulin lispro is shown in Figure 5. Analyte Std. curve range pg/ml Std. curve range fmol/ml r 2, linear fit, 1/x weighting Mean % accuracy of all points Insulin lispro 50 to 10,000 8.6 to 1720 0.998 99.99 Insulin glargine 50 to 10,000 8.3 to 1650 0.996 99.98 Human insulin 50 to 10,000 8.6 to 1720 0.996.00 Insulin detemir 200 to 10,000 33.8 to 1690 0.998 96.40 Insulin glulisine 50 to 10,000 8.6 to 1720 0.995.00 Insulin aspart to 10,000 17.2 to 1716 0.995.00 Table 2. Standard curve ranges, r 2 values, and mean accuracy for curve points for all compounds. Compound name: Humalog Correlation coefficient: r = 0.998354, r^2 = 0.996710 Calibration curve: 0.00024951 x + 0.000753141 Response type: Internal std ( Ref 2 ), Area ( IS conc. / IS area ) Curve type: Linear, Origin: Exclude, Weighting: 1/x, Axis trans: None 10.0 Residual 0.0-10.0 Conc Response 2.00 1.00-0.00-0 0 2000 3000 4000 5000 6000 7000 8000 9000 10,000 Conc Figure 5. Representative standard curve for insulin lispro. 8

Inter- and intra-day accuracy and precision were calculated for all QC samples. Representative summary statistics for insulin glargine, lispro, and human insulin can be found in Tables 3, 4, and 5. All other analogs exhibited similar performance. Insulin Glargine Inter-day n=9 150 150.1 18.7 12.4 102.7 750 718.4 47.3 6.6 95.8 2500 2369.3 131.2 5.5 94.8 7500 7648.5 511.3 6.7 102.0 Intra-day n=3 150 167.4 16.6 9.9 111.6 750 757.7 62.4 8.2 101.1 2500 2378.0 184.9 7.8 95.1 7500 7949.5 257.9 3.2 106.0 Table 3. Inter- and intra-day accuracy and precision for QC samples for insulin glargine. Insulin Lispro Inter-day n=9 150 144.0 17.5 12.2 96.0 750 721.8 32.3 4.5 96.2 2500 2447.1 202.9 8.3 97.9 7500 7697.5 634.8 8.2 102.6 Intra-day n=3 150 164.6 14.9 9.1 109.8 750 748.2 19.8 2.6 99.8 2500 2417.6 230.4 9.5 96.7 7500 8215.4 243.1 3.0 109.5 Table 4. Inter- and intra-day accuracy and precision for QC samples for insulin lispro. Human Insulin Inter-day n=9 (Avg basal level was 1937 pg/ml) 150 1915.1 125.4 6.5 92.0 750 2542.5 141.0 5.5 94.8 2500 4326.0 146.7 3.4 97.6 7500 9819.0 960.3 9.8 104.0 Intra-day n=3 (Basal level was 1872 pg/ml) 150 2056.5 16.7 0.8 90.2 750 2506.3 46.6 1.9 99.3 2500 4269.8 206.4 4.8 101.3 7500 10,233.2 265.2 2.6.3 Table 5. Inter- and intra-day accuracy and precision for QC samples for human insulin. 9

Specificity Matrix factors and CVs of matrix factors for all analogs were calculated in six individual sources of human plasma, as outlined in the 2007 AAPS white paper. 5 The % CV of the matrix factors were 12.3, 11.6, 11.8, 9.0, and 7.7 for insulin detemir, glargine, aspart, glulisine, and lispro, respectively. All easily met the recommended criteria of <15% CV. In addition, a study was performed to assess the impact of high levels of human insulin, expected to be found in type II diabetic patients, on the assay specificity. This is particularly important for two reasons. First, not all analytes are chromatographically resolved from one and another. Second, due to the use of a nominal mass instrument (triple quadrupole) and the high degree of sequence homology between the analogs themselves and with human insulin, the possibility of MS overlap in some of the transitions exists. For this test, plasma samples were fortified to a final concentration of 5 ng/ml with all of the analogs. A subset of these samples was also spiked with human insulin at 200X higher concentration, to a final concentration of 1 µg/ml human insulin. All samples were then pre-treated, extracted, and quantified. There was no significant change in area counts for any of the analogs when human insulin was present in high concentration. CONCLUSIONS Using analytical scale chromatography and simple 96-well SPE sample preparation, this method reaches detection limits for intact insulins, comparable to those previously achieved using immuno-precipitation and nano-scale chromatography. 6-11 Development of this assay was challenging due to a high degree of non-specific binding (NSB), protein binding, and difficulty maintaining peptide solubility throughout the SPE extraction and elution process. Sample pretreatment prior to SPE proved to be critical in improving recovery and specificity. Protein precipitation with 1% acetic acid 1:1 acetonitrile/methanol resulted in 8 to % recovery without precipitating the peptides themselves. Mixed-mode strong anion SPE provided an additional layer of selectivity, and facilitated the use of the low MS fragments that were necessary to distinguish human insulin from lispro. The use of the CORTECS C 18 + Column provided significantly improved sensitivity and peak shape for insulin and its analogs versus charged-surface fully-porous columns and traditional C 18 solid-core columns. This led to an improvement in detection limits of approximately 2X, when combined with proper MS fragment choice, and selective SPE cleanup enabled quantification limits of 50 to 200 pg/ml for the six insulins. All FDA criteria 12 for accuracy and precision of the method were easily met. Average accuracies for standard curve points and QC samples were >92%, with most close to 99%. Inter- and intra-day precision for all QC samples was greater than 7.5%. CVs of matrix factors across six lots of human plasma were <15%, further supporting the selectivity of the method. 10

References 1. Wagner BM, Schuster SA, Boyes BE, Kirkland JJ. Superficially porous silica particles with wide pores for biomacromolecular separations. Journal of Chromatography A. 2012; 1264(0):22-30. 2. Kirkland JJ, Schuster SA, Johnson WL, Boyes BE. Fused-Core Particle Technology in High-Performance Liquid Chromatography: An Overview. Journal of Pharmaceutical Analysis. http://dx.doi.org/10.1016/j.jpha.2013.02.005 3. Chambers E, Legido-Quigley C, Smith N, Fountain KJ. Development of a fast method for direct analysis of intact synthetic insulins in human plasma: the large peptide challenge. Bioanalysis. 2012; 5(1):65-81. 4. Chambers E, Wagrowski-Diehl DM, Lu Z, Mazzeo JR. Systematic and comprehensive strategy for reducing matrix effects in LC/MS/MS analyses. Journal of Chromatography B. 2007; 852(1-2):22-34. 5. Bansal S, Destefano A. Key elements of bioanalytical method validation for small molecules. The AAPS Journal. 2007; 9(1):E109-E114. 6. Thevis M, Thomas A, Delahaut P, Bosseloir A, Schänzer W. Doping Control Analysis of Intact Rapid-Acting Insulin Analogues in Human Urine by Liquid Chromatography-Tandem Mass Spectrometry. Analytical Chemistry. 2006; 78(6):1897-1903. 7. Thevis M, Thomas A, Schänzer W. Mass spectrometric determination of insulins and their degradation products in sports drug testing. Mass Spectrometry Reviews. 2008; 27(1):35-50. 8. Thevis M, Thomas A, Schänzer W, Östman P, Ojanperä I. Measuring insulin in human vitreous humour using LC-MS/MS. Drug Testing and Analysis. 2012; 4(1):53-56. 9. Thomas A, Schänzer W, Delahaut P, Thevis M. Sensitive and fast identification of urinary human, synthetic and animal insulin by means of nano-uplc coupled with high-resolution/high-accuracy mass spectrometry. Drug Testing and Analysis. 2009; 1(5):219-227. 10. Thomas A, Schänzer W, Delahaut P, Thevis M. Immunoaffinity purification of peptide hormones prior to liquid chromatography-mass spectrometry in doping controls. Methods. 2012; 56(2):230-235. 11. Thomas A, Thevis M, Delahaut P, Bosseloir A, Schänzer W. Mass Spectrometric Identification of Degradation Products of Insulin and Its Long-Acting Analogues in Human Urine for Doping Control Purposes. Analytical Chemistry. 2007; 79(6):2518-2524. 12. Guidance for Industry Bioanalytical Method Validation. 2001. Waters, ACQUITY UPLC, Oasis, UPLC, ACQUITY, Xevo, and MassLynx are registered trademarks of Waters Corporation. CORTECS, IS, XBridge, TargetLynx, and The Science of What s Possible are trademarks of Waters Corporation. All other trademarks are the property of their respective owners. 2013 Waters Corporation. Produced in the U.S.A. June 2013 720004727EN LL-PDF Waters Corporation 34 Maple Street Milford, MA 01757 U.S.A. T: 1 508 478 2000 F: 1 508 872 1990 www.waters.com