Improving Resolution and Column Loading Systematically in Preparative Liquid Chromatography for Isolating a Minor Component from Peppermint Extract

Similar documents
Techniques for Improving the Efficiency of Large Volume Loading in Preparative Liquid Chromatography

Encompassing Method Development, Small Scale Purification, and Post-Purification Analysis with the Investigator SFC System

Simple Techniques for Improving the Isolation of Synthetic Peptides Jo-Ann Jablonski Principal Scientist Waters Corporation

Method Development Considerations for Reversed-Phase Protein Separations

A Modular Preparative HPLC System for the Isolation of Puerarin from Kudzu Root Extracts

and for the Development of Isolation Strategies

Purification Workflow Management

Maximizing Chromatographic Resolution of Peptide Maps using UPLC with Tandem Columns

Simplifying Methods Transfer: Novel Tools for Replicating Your Established Methods on an ACQUITY Arc System

Sean M. McCarthy and Martin Gilar Waters Corporation, Milford, MA, U.S. INTRODUCTION EXPERIMENTAL RESULTS AND DISCUSSION

Chem 321 Lecture 23 - Liquid Chromatography 11/19/13

Generating Automated and Efficient LC/MS Peptide Mapping Results with the Biopharmaceutical Platform Solution with UNIFI

Bivalirudin Purification:

Illicit Drug Analysis in Urine Using 2D LC-MS/MS for Forensic Toxicology

Scale-up with the Agilent SD-1 Purification System analytical and preparative runs on a single system

Application Solutions. For Preparative-Scale Chromatography

Developing Robust and Efficient IEX Methods for Charge Variant Analysis of Biotherapeutics Using ACQUITY UPLC H-Class System and Auto Blend Plus

A Practical User Guide for the Determination of Optimal Purification Gradients for the Gilson PLC 2020

Peptide Mapping: A Quality by Design (QbD) Approach

Quality-by-Design-Based Method Development Using an Agilent 1290 Infinity II LC

Exploring Extra Sensitivity Using ionkey/ms with the Xevo G2-XS Q-Tof HRMS for Small Molecule Pharmaceutical Analysis in Human Plasma

Benefits of 2D-LC/MS/MS in Pharmaceutical Bioanalytics

Optimizing the Purification of a Standard Chiral Compound Utilizing a Benchtop, Multi-Purpose, Semi-Preparative to Preparative HPLC System

10. Validated Normal Phase HPLC Method for the Determination. Fulvestrant is primarily used in the treatment of hormone receptor

Gradient Elution. Slide 2

A Simple Rapid and Sensitive Method Development for Quantification of Quetiapine Fumarate in Bulk and Dosage Forms Using RP-HPLC

Preparative Purification of Corticosteroids by HPLC; Scalability and Loadability Using Agilent Prep C18 HPLC Columns Application

Overview of preparative HPLC. Analytical Technologies Limited

What differentiates Dynamic Extractions?

Protein-Pak Hi Res HIC Column and HIC Protein Standard

Application Note. Author. Abstract. Pharmaceuticals. Detlef Wilhelm ANATOX GmbH & Co. KG. Fuerstenwalde, Germany mau

Simultaneous in vivo Quantification and Metabolite Identification of Plasma Samples Using High Resolution QTof and Routine MS E Data Analysis

Customized systems to meet your lab s needs

Online UPLC Method for the Support of Cleaning Validation and the Routine Monitoring of Cleaning Procedures

ACQUITY Arc VERSATILITY WITHOUT COMPROMISE

BÜCHI Labortechnik AG

Introduction Ron Majors is a Senior Scientist at Agilent. Bill Champion is a chemist in Agilent s HPLC Columns tech support group.

Technical Overview. Author. Abstract. Edgar Naegele Agilent Technologies, Inc. Waldbronn, Germany

Fast Preparative Column Liquid Chromatography (PCLC)

for Acclaim Carbamate Column

A Comprehensive Workflow to Optimize and Execute Protein Aggregate Studies

Automated Scouting of Stationary and Mobile Phases Using the Agilent 1290 Infinity II Method Development Solution

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

Preparative HPLC is still the

Developing Purification Strategies for the Agilent 1260 Infinity II Preparative LC/MSD System

Fast and High-Resolution Reversed-Phase Separation of Synthetic Oligonucleotides

HPLC to UPLC Method Migration: An Overview of Key Considerations and Available Tools

The Theory of HPLC Gradient HPLC

UPLC/MS/MS Method for the Quantification of Trastuzumab in Human Serum at the 5-nM Level Using Xevo TQD MS and ACQUITY UPLC H-Class System

Developing Quantitative UPLC Assays with UV

Reversed-phase Separation of Intact Monoclonal Antibodies Using Agilent ZORBAX Rapid Resolution High Definition 300SB-C8 1.

Method Development of a 2D LC-HRMS Extraction and Detection Method for Organophosphorus Flame Retardants in Environmental Water Samples

Purification of oligonucleotides by anion exchange chromatography

FUTURE-PROOF SOLUTIONS FOR REGULATED LABORATORIES IN THE FACE OF CHANGING USP <621> GUIDELINES

Effective Scaling of Preparative Separations Using Axial Compression Columns

Biotherapeutic Method Development Guide

High-Throughput LC/MS Purification of Pharmaceutical Impurities

Quantification of genotoxic "Impurity D" in Atenolol by LC/ESI/MS/MS with Agilent 1200 Series RRLC and 6410B Triple Quadrupole LC/MS

Performance characteristics of the 1260 Infinity Quaternary LC system

The Use of Preparative Supercritical Fluid Chromatography for the Isolation of Cannabidiolic Acid (CBDA) From Hemp Extract

Fast Method Development Using the Agilent 1290 Infinity Quaternary LC System with Column Selection Valve

Automated QbD-Based Method Development and Validation of Oxidative Degraded Atorvastatin

Emulation of the Agilent 1100 Series LC Through Waters Empower Software Analysis of an Analgesic Mixture

Automated 2-D HPLC Using Trap Columns for the Fractionation, Isolation and Screening of Natural Products

A Sensitive and Robust Method for the Quantification of Goserelin in Plasma Using Micro-Elution Plates

DIDANOSINE ORAL POWDER Final text for addition to The International Pharmacopoeia

Biotherapeutic Method Development Guide

Rapid UHPLC Analysis of Reduced Monoclonal Antibodies using an Agilent ZORBAX Rapid Resolution High Definition (RRHD) 300SB-C8 Column

Maximizing Sample Recovery on the ACCQPrep HP125

Martin Gilar Waters Corporation, Milford, MA, U.S. Origins of synthetic oligonucleotides impurities. Lab-scale isolation options

BioTrap 500 C18/C8 INSTRUCTION MANUAL. for direct injection of up to 500 µl plasma in HPLC. Illustration of a BioTrap 500 particle

Agilent Anion-Exchange Media for Proteins

Application of Agilent AdvanceBio Desalting-RP Cartridges for LC/MS Analysis of mabs A One- and Two-dimensional LC/MS Study

Dynamic High Capacity Mustang Q Membrane Units for Scaleable Anion Exchange Chromatography Purification of Adenoviral Vectors

AdvanceBio HIC: a Hydrophobic HPLC Column for Monoclonal Antibody (mab) Variant Analysis

[ PURIFICATION SOLUTIONS ] Pure at Heart

Agilent AdvanceBio SEC Columns for Aggregate Analysis: Instrument Compatibility

Solutions Guide. MX Series II Modular Automation for Nano and Analytical Scale HPLC And Low Pressure Fluid Switching Applications

Fundamentals and Techniques of Preparative HPLC. Parto Zist Behboud Tel: 42108

Automatic Combination of Orthogonal Separation Modes for Multiple Components in Mixtures

Product Guide for LudgerSep TM N2 High Resolution Amide HPLC Columns for Glycan Analysis

ACQUITY UPC 2 BEH, CSH, and HSS Columns

Application Note. Authors. Abstract. Introduction. Pharmaceutical

Fraction Analysis of Cysteine Linked Antibody-Drug Conjugates Using Hydrophobic Interaction. chromatography. Agilent 1260 Infinity II Bio-Inert System

Monoclonal Antibody Analysis on a Reversed-Phase C4 Polymer Monolith Column

An Automated System for At-Line Process Analysis of Biopharmaceutical Fermentation Reactions

Optimizing Purified Sample Recovery on Reverse-Phase HPLC Systems

High Throughput Sub-4 Minute Separation of Antibodies using Size Exclusion Chromatography

[ WHITE PAPER ] High Throughput Microflow LC-MS: Sensitivity Gains on a Practical Timescale ABSTRACT INTRODUCTION

HIC as a Complementary, Confirmatory Tool to SEC for the Analysis of mab Aggregates

Hillary B. Hewitson, Thomas E. Wheat, Diane M. Diehl INTRODUCTION

Fast mass transfer Fast separations High throughput and improved productivity Long column lifetime Outstanding reproducibility Low carryover

High-resolution Analysis of Charge Heterogeneity in Monoclonal Antibodies Using ph-gradient Cation Exchange Chromatography

High-Resolution Separation of Intact Monoclonal Antibody Isoforms

HPLC Columns HIC PH-814 MANUAL. Shodex HPLC Columns Europe, Middle East, Africa, Russia. For technical support please use contact details shown below:

Reducing Cycle Time for Charge Variant Analysis of Monoclonal Antibodies

Technical Overview. Author. Abstract. A.G.Huesgen Agilent Technologies, Inc. Waldbronn, Germany

Automated fraction re-analysis does it really make sense? Application. Udo Huber. Abstract

Preparative HPLC: Factors and Parameters that Directly Affect Recovery of Collected Fractions

Automated alternating column regeneration on the Agilent 1290 Infinity LC

Transcription:

Improving Resolution and Column Loading Systematically in Preparative Liquid Chromatography for Isolating a Minor Component from Peppermint Extract Jo-Ann M. Jablonski and Rui Chen Waters Corporation, Milford, MA, USA A P P L I C AT ION B E N E F I T S Focused gradients improve the resolution of closely eluting components, thereby increasing the column loading for more efficient target compound purification. At-column dilution alleviates the peak distortion and loss of resolution attributed to the injection of large volumes of strong solvent, leading to improved resolution, column loading, and overall productivity in natural product isolation. WAT E R S SO LU T IONS AutoPurification System XSelect CSH Column XSelect C 18 Prep OBD Column K E Y W O R D S Preparative liquid chromatography, natural product isolation, focused gradients, at-column dilution INT RO DU C T ION Natural products are widely used in the pharmaceutical, food supplement, nutraceutical, and alternative medicine industries. 1-4 Chromatography has long been an integral part of natural product research, including chemical fingerprinting, structural elucidation, and isolation of bioactive compounds on the preparative scale. Since natural product extracts are usually complex mixtures comprised of many different compound classes with a variety of functional groups, acid-base properties, and molecular sizes, reversed-phase liquid chromatography (RPLC) often lends itself as the technique of choice for the analysis and purification of natural products, largely due to its general applicability. The use of preparative high performance liquid chromatography (prep HPLC) has become a mainstay in the isolation of most classes of natural products over the last ten years. 4 In target compound purification, adequate resolution between target analytes and their adjacent interference peaks is a prerequisite for successful preparative chromatography. Typical approaches for improving resolution include the following: evaluating different stationary phases, mobile phases, and modifiers; changing the temperature of the separation; and varying the gradient slope. However, the ultimate objective for prep chromatography is to efficiently collect target compounds of desired purity. Consequently, experimental parameters such as sample diluents and injection techniques and their impact on solvent consumption and productivity should also be considered in the overall method development strategy. 5 This is particularly important for natural product isolation, since the desired compounds often exist at low concentrations within very complex matrices. To that end, at-column dilution (ACD) has proven to be a viable alternative to conventional injection techniques. ACD allows for injections of large volumes of sample in strong solvents while preserving chromatographic integrity and resolution, thereby improving overall purification productivity. 6 This application note uses peppermint extract 7 to demonstrate a typical prep HPLC method development workflow, systematically improving resolution and column loading for the isolation of a minor component in a natural product. 1

E X P E R IM E N TA L Sample description A total of 3.3 g dried peppermint was extracted with a 20 ml 80:20 methanol/water mixture for six hours at room temperature. 7 The supernatant was filtered with an Acrodisc Syringe Filter with GHP Membrane, 25 mm, 0.45 μm LC conditions System: AutoPurification Columns: XSelect CSH C 18 4.6 x 100 mm, 5 µm; Mobile phase A: Mobile phase B: UV wavelength: Flow rate: XSelect CSH Phenyl-Hexyl 4.6 x 100 mm, 5 µm; XSelect CSH Fluoro-Phenyl 4.6 x 100 mm, 5 µm; XSelect C 18 Prep OBD 19 x 100 mm, 5 µm 0.1% trifluoroacetic acid (TFA) in water 0.1% TFA in acetonitrile 220 nm 1.46 ml/min for analytical and 25.0 ml/min for preparative experiments The analytical and preparative gradients used in this study are summarized in Table 1. For the ACD injections, a separate ACD pump delivered a constant 1.3 ml/min acetonitrile (5% of the total flow rate) directly to the injection valve while the gradient pump delivered the gradient at a flow of 23.7 ml/min. The two flow streams were combined at the head of the column. The number of column volumes (CV) per gradient segment was constant for all three methods, ensuring that the chromatography at the prep scale was identical to the chromatography at the analytical scale. Other key experimental parameters are listed in the respective figure captions. (min) Analytical %B (min) Prep conventional injection %B (min) Prep ACD 5.0 5.0 17.4 0.4 5.0 4.3 11.7 25.4 1.4 17.4 5.3 12.4 12.2 95.0 12.2 25.4 16.1 20.4 17.2 95.0 12.6 95.0 16.5 9 17.4 5.0 17.6 95.0 2 9 25.4 5.0 17.8 5.0 21.7 2-mL loop, system volume = 6.3 ml %B 25.8 5.0 29.7 5-mL loop, system volume = 9.3 ml; ACD pump flows at 1.3 ml/min, total flow rate was 25.0 ml/min Table 1. Gradients used in the study. The analytical flow rate was 1.46 ml/min and the preparative flow rate was 25.0 ml/min. 2

R E SU LT S A N D D IS C U S S ION Column screening and focused gradients Prep chromatography shares many basic principles with its analytical counterpart. As a result, preparative HPLC method development often starts with an analytical LC followed by geometric scale-up to prep. LC/UV chromatograms of the peppermint extract using a generic gradient on three different columns is shown in Figure 1. The target compound, as well as other minor components in the crude extract, was best resolved on the XSelect CSH C 18 Column, as shown in Figure 1A. The XSelect CSH C 18 Column chemistry was, therefore, chosen for all ensuing experiments. (A) XSelect CSH C 18 3.50 4.00 4.50 5.00 5.50 6.00 6.50 7.00 (B) XSelect CSH Phenyl-Hexyl 3.50 4.00 4.50 5.00 5.50 6.00 6.50 7.00 (C) XSelect CSH Fluoro-Phenyl 3.50 4.00 4.50 5.00 5.50 6.00 6.50 7.00 Figure 1. LC/UV chromatograms of the peppermint extract obtained on three different columns: (A) XSelect CSH C 18 ; (B) XSelect CSH Phenyl-Hexyl; and (C) XSelect CSH Fluoro-Phenyl. The asterisk denotes the target compound peak. All column dimensions were 4.6 x 100 mm with 5-µm particles. The injection volume was 10 µl. A generic gradient from 5% to 95%B in 12 minutes was used, and the total run time was 20 minutes. 3

Since the analyte of interest eluted at ~22% B in the initial generic gradient, shown in Figure 2A, focused gradients ranging from 17% to 25% B were employed to further improve the resolution, as shown in Figures 2B and 2C. Focused gradients increase the residence time of closely eluting compounds on the column for better partition, improving the selectivity (α) between compounds with minute polarity differences. 8 However, decreased gradient slope also increases retentivity (k ), which in turn leads to broader peaks, reduced peak heights, prolonged run time, and greater solvent cost for prep chromatography. Therefore, caution should be exercised when using focused gradients to ensure the balance between resolution and run time. In the current study, at 0.72 %B/CV, shown in Figure 2C, the target peak was clearly baseline resolved from all adjacent peaks with a total run time of 25 minutes. For a shorter run time, the method could be terminated immediately after target peak collection with column washing steps to follow. (A) 7.17% B/CV 5% to 95% B in 12.00 min 2.00 3.00 4.00 5.00 6.00 7.00 8.00 9.00 10 10 12.00 (B) 1.43% B/CV 17% to 25% B in 5.36 min 2.00 3.00 4.00 5.00 6.00 7.00 8.00 9.00 10 10 12.00 (C) 0.72% B/CV 17% to 25% B in 10.73 min 2.00 3.00 4.00 5.00 6.00 7.00 8.00 9.00 10 10 12.00 Figure 2. LC/UV chromatograms of the peppermint extract on an XSelect CSH C 18 4.6 x 100 mm, 5 µm Column using three different gradients. Scale-up Proper scaling from analytical to prep requires the gradient slope (change in %B/CV) to be maintained for each step of the chromatography. 9 The flow rate and the injection volume are scaled geometrically. Table 2 summarizes the flow rate and injection volumes used when the chromatography was scaled from the analytical column to the preparative column. The properly scaled focused gradient methods are shown in Table 1. 4

Analytical Prep Column dimension 4.6 x 100 mm 19 x 100 mm Column volume 1.4 ml 23.8 ml Flow rate 1.46 ml/min 25.0 ml/min Injection volume 30 µl 512 µl Injection volume 40 µl 682 µl Table 2. Summary of scale-up parameters. A loading study (not shown) performed on the XSelect CSH C 18 4.6 x 100 mm Column showed that 30 µl was the maximum volume that could be injected without losing resolution between the target compound and the impurity. Geometrically, an injection volume of 30 µl on the analytical column scales to 512 µl on a 19 x 100 mm prep column. In the conventional injection technique, the target peak is resolved from its closely eluting neighbors, as shown in Figure 3A. Further increasing the injection volume, such as the 682-µL injection in Table 2, shown in Figure 3B, resulted in decreased resolution between the peak of interest and its closely eluting neighbors. For the peppermint extract in this study, a sensible injection volume for a 19 x 100 mm column was, therefore, limited to 512 µl using the conventional injection technique. The observed resolution loss was partially due to the strong solvent used as the sample diluent. Natural products sometimes require the use of strong organic solvents, such as methanol, ethanol, and acetone, to extract them from the sample matrix. However, large injection volumes of strong solvents often distort chromatography and result in a loss of chromatographic resolution. Sample molecules entering the column in a strong solvent do not retain. Instead, they move through the column until the strong solvent is diluted sufficiently by the initial-strength mobile phase to promote retention. As a result, the samples retain on the column as wide bands. The samples elute from the column as broad, poorly resolved peaks, thereby limiting the chromatographic efficiency and overall productivity. e-1 (A) 512 µl e-1 5.0e-2 2.00 4.00 6.00 8.00 10 12.00 14.00 16.00 18.00 e-1 (B) 682 µl e-1 5.0e-2 2.00 4.00 6.00 8.00 10 12.00 14.00 16.00 18.00 Figure 3. 512-µL and 682-µL injections of peppermint extract on an XSelect C 18 Prep OBD 19 x 100, 5 µm Column with the Waters AutoPurification System plumbed in the conventional mode. 5

At-column dilution ACD, an alternative injection technique, permits the injection of large volumes of strong solvents and concurrently improves sample solubility, column loading, and resolution. 6 With ACD, the chromatographic system is plumbed so that the sample in strong solvent is diluted at the head of the column with aqueous mobile phase. The sample is deposited on the column and the strong solvent flushes from the column before sample elution begins. Once the gradient is initiated, the sample components elute as narrow, sharply resolved bands, as shown in Figure 4. The strong solvent effect is effectively alleviated and the resolution is preserved. Furthermore, because the sample is continually surrounded by organic solvent until the point of dilution at the head of the column, no sample precipitation occurs. Sample enters column in dilute solvent and retains as a narrow band at column head. Strong solvent flushes from column before the gradient begins sample elution. Samples elute as narrow, sharply resolved bands in the solvent gradient. Sample S band Solvent gradient Sample band Strong solvent Strong solvent Figure 4. Schematic of at-column dilution. Figures 5A and 5B show the chromatograms using the conventional injection technique and ACD with the same 682-µL injection volume. Clearly, the one with the ACD, as shown in Figure 5B, provides improved resolution of the target compound from the closely eluting neighboring peaks. With ACD, a maximum injection volume of 2.7 ml was possible without the loss of resolution, as shown in Figure 5C. This represents a five-fold increase in column loading compared to the 512-µL injection volume using the conventional injection technique. It is important to note that the initial hold at the beginning of the ACD method ensures a complete sweeping of the sample loop. For example, a 5-mL loop was used for the 2.7-mL sample injection in Figure 5C, so an extra four minutes was added to the initial hold at 1.3 ml/min, as shown in Table 1. 6

2.0 (A) Conventional 682 µl 5.00 10 15.00 20 2.0 (B) ACD 682 µl 5.00 10 15.00 20 2.0 (C) ACD 2.7 ml 5.00 10 15.00 20 Figure 5. Comparison of prep LC/UV chromatograms with 682-µL and 2.7-mL injections of peppermint extract on an, XSelect C 18 Prep OBD 19 x 100, 5 µm Column with the AutoPurification System plumbed in conventional and at-column dilution modes. The target minor component was successfully isolated from the 2.7-mL sample load in a fraction with a purity of 94% by UV analysis, as shown in Figure 6. 5.5e-1 4.5e-1 4.0e-1 3.5e-1 3.0e-1 2.5e-1 2.0e-1 e-1 e-1 5.0e-2 3.00 4.00 5.00 6.00 7.00 8.00 9.00 10 Figure 6. Purity analysis by UV of the fraction from a 2.7-mL injection with ACD. 7

C O N C LUSIONS This application note illustrated a systematic preparative HPLC method development to isolate a minor component from peppermint extract using an AutoPurification System. The overall workflow included screening different column chemistries, applying focused gradients, scaling up, and employing an ACD injection scheme. With proper scale-up from an optimized analytical chromatographic condition, employing ACD increased the sample loading by five-fold while maintaining the resolution on the preparative scale. The techniques demonstrated in this case study have general applicability for laboratories routinely performing natural product isolation using preparative HPLC. References 1. Harvey AL. Strategies for discovering drugs from previously unexplored natural products. Drug Discovery Today. 2000; 5 (7):294-300. 2. Harvey AL. Natural products in drug discovery. Drug Discovery Today. 2008; 13 (19/20): 894-901. 3. Li JWH, Vederas JC. Drug Discovery and natural products: end of an era or endless frontier? Science. 2009; 325(10):161-165. 4. Latif Z, Sarker SD. Isolation of natural products by preparative high performance liquid chromatography (prep-hplc). Methods Mol Biol. 2012; 864: 255-74. 5. Rathore AS, Velayudhan A. An overview of scale-up in preparative chromatography in Scale-up and optimization in preparative chromatography: principles and biopharmaceutical applications, Eds. Rathore AS, Velayudhan A, Marcel Dekker, Inc. 2003. 6. Thomas Wheat, et al. At-Column Dilution Application Notes. Waters Application Note 71500078010rA. 2003. 7. Fecka I, Turek S. Determination of Water-Soluble Polyphenolic Compounds in Commercial Herbal Teas from Lamiaceae: Peppermint, Melissa, and Sage. J. Agric. Food Chem. 2007; 55: 10908-10917. 8. Jablonski JM, Wheat TE, Diehl DM. Developing Focused Gradients for Isolation and Purification. Waters Application Note 720002955EN. 2009 September. 9. Aubin A, Cleary R. Analytical HPLC to Preparative HPLC: Scale-Up Techniques using a Natural Product Extract. Waters Application Note 720003120EN. 2009 June. Waters and XSelect are registered trademarks of Waters Corporation. AutoPurification, CSH, OBD, 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. May 2013 720004672EN AG-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