hplc pharma 25-1-09-numbered-all print

Upload: shulalevin

Post on 07-Apr-2018

221 views

Category:

Documents


0 download

TRANSCRIPT

  • 8/4/2019 HPLC Pharma 25-1-09-Numbered-All Print

    1/19

    Created by: Dr. Shula Levin Medtechnica January 26, 2009

    [email protected] Page 1 of 19

    High Performance Liquid Chromatography

    (HPLC) in the pharmaceutical analysis

    Shulamit Levin, Medtechnica; Efal St 5 Petach Tikva

    49002

    1 Types of HPLC: Reversed Phase chromatography..2 HPLC Theory: System Suitability parameters3 The Role of HPLC in Drug Analysis

    3.1 Discovery: High throughput screening andbioanalysis

    3.2 Development: Method development andvalidation

    3.3 Manufacturing: Dissolution, content uniformity,assay, drug impurities and stability, in process

    and cleaning verification.

    4 Specialized HPLC Separations.

    4.1 Ultra-Performance Liquid Chromatography.4.2 Preparative HPLC

    4.3 Chiral Separations

    Introduction

    Drug manufacturing control requires high level andintensive analytical and chemical support of all stages to

    ensure the drug's quality and safety (1). The

    pharmacopeia constitutes a collection of recommended

    procedures for analysis and specifications for the

    determination of pharmaceutical substances, excipients,

    and dosage forms that is intended to serve as sourcematerial for reference or adaptation by anyone wishing to

    fulfill pharmaceutical requirements. The most important

    analytical technique used during the various steps of

    drug development and manufacturing is the separation

    technique: High Performance Liquid Chromatography

    (HPLC).

    The key to a proper HPLC system operation is

    knowledge of the principles of the chromatographic

    process, as well as understanding the reasons behind the

    choice of the components of the chromatographic

    systems such as column, mobile phase and detectors. A

    scheme of an HPLC system is shown in Figure 1. Ahigh pressure pump is required to force the mobile phase

    through the column at typical flow rates of 0.5-2

    ml/min. The sample to be separated is introduced into

    the mobile phase by injection device, manual or

    automatic, prior to the column. The detector usually

    contains low volume cell through which the mobile

    phase passes carrying the sample components eluting

    from the column. There are books describing the

    practicality of HPLC operation (2-11). It is expected ofany proper HPLC system that is used in the

    pharmaceutical laboratories to produce highly accurate

    and precise results, due to health related issues of

    improper measurements. Every HPLC system must be

    qualified to comply with the strict demands from health

    authorities for high quantitative performance.

    Quality standards in pharmaceutics require that all

    instruments should be adequately designed, maintained,calibrated, and tested. The approach that has been

    adopted in the environment of the analytical instrument

    has become known as the "Four Qs": design qualification(DQ), installation qualification (IQ), operational

    qualification (OQ), and performance qualification (PQ).Design qualification is performed at the vendors site,

    and it is representative of the way an instrument is

    developed and produced, usually governed by

    International Organization for Standardization (ISO)

    criteria.

    The installation qualification (IQ) process can be divided

    into two steps:pre-installationandphysical installation.

    During pre-installation, all information relevant to the

    proper installation, operation, and maintenance of the

    instrument is checked. Workers confirm the site

    requirements and the receipt of all of the parts, pieces,

    and manuals necessary to perform the installation of the

    specific HPLC unit. During physical installation, serial

    numbers are recorded and all fluidic, electrical, and

    communication connections are made for system

    components. Documentation describing how the

    instrument was installed, who performed the installation,

    and other various details are archived.

    HPLC Column

    in OvenAuto

    SamplerPump

    )flows50-500 0L/min

    Fraction

    Collector

    Waste

    Detector&Control

    DataProcessing

    1.Fucose

    2.Galactosami ne

    3.Glucosamine

    4.Galactose

    5.Glucose

    6.Mannose

    20.00Minutes5.00

    mV

    0.00

    300

    12

    34

    5

    6

    a bc d

    Or manual injector

    Figure 1: A Scheme of an HPLC System

    The operational qualification process ensures that the

    separate modules of a system (pump, injector, and

    detector) are operating according to the defined

    specifications such as accuracy, linearity, and precision.

    Specific tests are performed to verify parameters such asdetector wavelength accuracy, flow rate, or injector

    precision.

    The performance qualification (PQ) step verifies system

    performance as a whole. Performance qualification

    testing is conducted under real operating conditions inthe analytical laboratory that is going to be using the

    instrument. In practice, sometimes operational and

  • 8/4/2019 HPLC Pharma 25-1-09-Numbered-All Print

    2/19

    Created by: Dr. Shula Levin Medtechnica January 26, 2009

    [email protected] Page 2 of 19

    performance qualification blend together, particularly for

    linearity and precision (repeatability) tests, which can be

    conducted more easily at the system level.

    The performance qualification test of the HPLC systemuses a method with a well-characterized analyte mixture,

    column, and mobile phase. It incorporates type of

    measurements from the system suitability section of thegeneral chromatography chapter in the U.S.

    Pharmacopeia (12). In the end of the process proper

    documentation is archived.

    1. Modes of HPLC

    There are various modes of operation of HPLC. The

    mechanism of interaction of the solutes with the

    stationary phases determines the classification of themode of liquid chromatography. Table 1 summarizes

    the variety of modes of liquid chromatography, of which

    Reversed Phase stands out as the most widely used modein HPLC, therefore, the discussion will elaborate on this

    mode.

    Reversed Phase (13)

    Reversed phase liquid chromatography (RPLC) is

    considered as the method of choice for the analysis of

    pharmaceutical compounds for several reasons, such as

    its compatibility with aqueous and organic solutions aswell as with different detection systems and its high

    consistency and repeatability. Sensitive and accurate

    RPLC analysis, whether in the pharmaceutical or

    bioanalytical field, necessitates the use of stationaryphases which give symmetrical and efficient peaks.Therefore, manufacturers of stationary phases are

    continuously improving and introducing new RPLC

    products, and the selection of various types of reversed

    phase stationary phases is high. The needs for

    consistency as well as the globalization of the

    pharmaceutical companies require that the methods willbe transferred from site to site, using either the same

    column brands or their equivalents. Therefore, an

    extensive categorization or characterization of the rich

    selection of stationary phases has been done in recent

    years (14-21).

    The stationary phase in the Reversed Phase

    chromatographic columns is a hydrophobic support that

    is consisted mainly of porous particles of silica gel in

    various shapes (spheric or irregular) at various diameters

    (1.8, 3, 5, 7, 10 M etc.) at various pore sizes (such as60, 100, 120, 300). The surface of these particles is

    covered with various chemical entities, such as various

    hydrocarbons (C1, C6, C4, C8, C18, etc) as can be seen

    in Figure 2. There are also hydrophobic polymeric

    supports that are used as stationary phases when there is

    an extreme pH in the mobile phase. In most methods

    used currently to separate medicinal materials, C18

    columns are used, which sometimes are called ODS(octedecylsilane) or RP-18.

    The more hydrophobic are the sample components the

    longer they stay in the column thus they are separated.

    The mobile phases are mixtures of water and organic

    polar solvents mostly methanol and acetonitrile. These

    mixtures contain frequently additives such as acetate,phosphate, citrate, and/or ion-pairing substances, which

    are surface active substances such as alkylamines as ion-

    pairng of anions or alkylsulfonates, ion-pairing ofcations. The purpose of using such additives is to

    enhance efficiency and/or selectivity of the separation,

    mostly due to control of their retention.

    Reversed Phase - Stationary Phase Surface

    O

    Si

    O

    Si

    O

    Si

    O

    Si

    O

    Si

    O

    O

    O

    Si

    O

    Si

    O

    Si

    O

    Si

    O O

    Si

    O

    Si

    OO

    O

    O

    O

    O

    O

    O O O O O O O

    OH

    Si

    HO O

    OH O

    OHHO

    O O

    O

    Si

    O

    O

    OH

    R

    R

    R

    R R

    R

    NORMAL PHASE

    R=H

    REVERSED PHASE

    . R= C18; C8; C4; CN; C1 and more

    Si

    Si

    Figure 2: The surface of Reversed Phase stationary phases

    The parameters that govern the retention in Reversed

    Phase systems are the following:

    A. The chemical nature of the stationary phase

    surface(22-27).B. The type of solvents that compose the mobile

    phase and their ratio(28)

    C. The pH and ionic strength and additives of themobile phase(23, 29)

    When the effect of these parameters on the retention of

    the solutes is understood it is possible to manipulate

    them to enhance selectivity.

    A. The chemical nature of the stationary phase(16)

    The surface of the stationary phase is described in Figure2. The chemical nature is determined by the size and

    chemistry of hydrocarbon bonded on the silica gel

    surface, its bonding density (units ofmole/m2), and thepurity and quality of the silica gel support. As a rule, the

    more carbons in a bonded hydrocarbon the more itretains organic solutes (as long as similar % coverage is

    compared). The higher the bonding density the longer

    the organic solutes are retained. A column is considered

    relatively hydrophobic if its bonding density exceeds 3

    mole/m2.

    Very important modifiers of the stationary phase's

    surface are surface-active substances used as mobilephase's additives, acting as ion-pair reagents. These are

  • 8/4/2019 HPLC Pharma 25-1-09-Numbered-All Print

    3/19

    Created by: Dr. Shula Levin Medtechnica January 26, 2009

    [email protected] Page 3 of 19

    substances such as tri-ethylamine or tetrabutylamine or

    hexyl, heptyl, octyl sulfonate. They are distributed

    between the mobile phase and the hydrophobic surface

    and cover it with either positive (alkylamines) or

    negative (alkylsulfonates) charges. This change of thesurface into charged surface affects the retention

    significantly, especially on charged species in the

    sample(30).

    B. Composition of the mobile phase

    As a rule, the weakest solvent in Reversed Phase is the

    most polar one, water. The other polar organic solvents

    are considered stronger solvents, where the order of

    solvent strength follows more or less their dielectric

    properties, or polarity. The less polar the solvent added

    to the mobile phase, the stronger it gets, shortening the

    retention times.

    C. PH and ionic strength of the mobile phase

    When the samples contain solutes of ionizable functional

    groups, such as amines, carboxyls, phosphates,

    phosphonates, sulfates and sulfonates, it is possible to

    control their ionization degree with the help of buffers in

    the mobile phase. The carboxyl groups in the solutes

    obtain more negative charge as pH increases above their

    pKa. As a rule, the change of an ionizable molecule to

    an ion makes it more polar and less available to the

    stationary phase. For example, increasing the pH of the

    mobile phase above 4-5, which is the typical pKa of

    carboxyl groups, reduces the retention of carboxyl

    containing compounds. On the other hand, substancesthat contain amines whose pKa is around 8 will retain

    longer when the pH will be above 8. In most of the

    traditional silica-gel based stationary phases it is not

    possible to increase the mobile phases pH above 8 due

    to hydrolysis of the silica gel. During the 2000s there

    have been developed extended pH stationary phases (31,

    32)

    2. HPLC Theory: System Suitability

    Parameters

    High performance liquid chromatography is defined as a

    separation of mixtures of compounds due to differencesin their distribution equilibrium between two phases, the

    stationary phase packed inside columns and the mobile

    phase, delivered through the columns by high pressure

    pumps. Components whose distribution into the

    stationary phase is higher, are retained longer, and get

    separated from those with lower distribution into thestationary phase. The theoretical and practical

    foundations of this method were laid down at the end of

    1960s and at the beginning of 1970s. The theory of

    chromatography has been used as the basis for System-

    Suitability tests, which are set of quantitative criteria that

    test the suitability of the chromatographic system to

    identify and quantify drug related samples by HPLC atany step of the pharmaceutical analysis.

    Retention Time (tR) and Capacity Factor k' and Relative

    Retention Time (RRT)

    The time elapsed between the injection of the samplecomponents into the column and their detection is known

    as the Retention Time (tR). The retention time is longer

    when the solute has higher affinity to the stationaryphase due to its chemical nature. For example, in reverse

    phase chromatography, the more lypophilic compounds

    are retained longer. Therefore, the retention time is a

    property of the analyte that can be used for its

    identification.

    A non retained substance passes through the column at a

    time t0, called the Void Time. TheRetention Factoror

    Capacity Factor k' of an analyte is measured

    experimentally as shown in Figure 3 and Eqn 1:

    Eqn 1a

    o

    R

    tttk 0' =

    The Capacity Factordescribes the thermodynamic basis

    of the separation and its definition is the ratio of the

    amounts of the solute at the stationary and mobile phases

    within the analyte band inside the chromatographic

    column:

    Eqn 1b =m

    s

    C

    Ck'

    Where Cs

    is the concentration of the solute at the

    stationary phase and Cm is its concentration at the mobile

    phase and is the ratio of the stationary and mobilephase volumes all within the chromatographic band.

    The Retention Factor(Eqn 1a) is used to compare the

    retention of a solute between two chromatographic

    systems, normalizing it to the column's geometry and

    system flow rate. The need to determine the void time

    can be tricky sometimes, due to the instability of the

    elution time of the void time marker, t0, therefore, when

    the chromatogram is complex in nature, and one known

    component is always present at a certain retention time,

    it can be used as a retention marker for other peaks. Insuch cases the ratio between the retention time of any

    peak in the chromatogram and the retention time of themarker is used (tR (Peak)/ tR (Marker)) and referred to as the

    Relative Retention Time (RRT). RRT is also used

    instead of the capacity ratio for the identification of the

    analyte as well as to compare its extent of retention in

    two different chromatographic systems.

  • 8/4/2019 HPLC Pharma 25-1-09-Numbered-All Print

    4/19

    Created by: Dr. Shula Levin Medtechnica January 26, 2009

    [email protected] Page 4 of 19

    0

    .1

    7

    6

    0

    .7

    3

    9

    A

    U

    0.000

    0.015

    0.030

    0.045

    0.060

    Minutes

    0.00 0.05 0.10 0.15 0.20 0.25 0.30 0.35 0.40 0.45 0.50 0.55 0.60 0.65 0.70 0.75 0.80 0.85 0.90 0.95 1.00

    Figure 3: Example of Capacity factor calculation in LC. In this

    case: tR = 0.739, t0 = 0.17, therefore k' = (0.739-0.176)/0.176 = 3.20

    Efficiency: Plate Count N and Peak Capacity Pc

    Figure 4 describes a chromatogram with 4 peaks and a

    detected void peak. The parameters of the System

    Suitability are displayed in the inserted table. The

    sharpness of a peak relative to its retention time is a

    measure of the system's efficiency, calculated as N, platecount. Band-broadening phenomena in the column such

    as eddy diffusion, molecular diffusion, mass-transfer

    kinetics and extra-column effects reduce the efficiency

    of the separation (33). The sharpness of a peak isrelevant to the limit of detection and limit of

    quantification of the chromatographic system. Thesharper the peak for a specific area, the better is its

    signal-to-noise; hence the system is capable of detecting

    lower concentrations. Therefore, the efficiency of the

    chromatographic system must be established by the

    system suitability test before the analysis of low

    concentrations that requires high sensitivity of thesystem, such as the analysis of drug impurities and

    degradation products.

    The efficiency of the separation is determined by the

    Plate Count N when working at isocratic conditions,whereas it is usually measured by Peak Capacity Pc when

    working at gradient conditions (34). The following

    equation for the plate count is used by the United States

    Pharmacopoeia (USP) to calculate N (See Figure 5):

    Eqn. 22)(16

    )(Baselinew

    Rt

    N =

    Where w is measured from the baseline peak width

    calculated using lines tangent to the peak width at 50 %

    height (See Figure 5). European and Japanese

    Pharmacopoeias use the peak width at 50% of the peakheight (See Figure 5), hence the equation becomes:

    Eqn 32)(54.5

    %)50(w

    Rt

    N =

    Peak CapacityPc is defined as number of peaks that canbe separated within a retention window for a specific

    pre-determined resolution. In other words, it is the

    runtime measured in peak width units (34). It is

    assumed that peaks occur over the gradient

    chromatogram. Therefore, Peak Capacity can be

    calculated from the peak widths w in the chromatogramas follows:

    Eqn. 4:

    +

    n

    g

    wn

    t

    1

    )/1(

    1

    Where n is the number of peaks at the segment of the

    gradient selected for the calculation, tg. Thus peak

    capacity can be simply the gradient run time divided by

    the average peak width. The sharper the peaks the

    higher is the peak capacity, hence the system should be

    able to resolve more peaks at the selected run time as

    well as detect lower concentrations.

    Another measure of the column's chromatographic

    efficiency is the Height Equivalent to Theoretical Plate

    (HETP) which is calculated from the following equation:

    Eqn 5: HETP = (L/N)

    Where L is column length and N is the plate count.

    HETP is measured in micrometer. For example, in the

    peak of Figure 5 the column length was 100 mm (0.1 m)

    therefore HETP was 8.8 m at the flow rate of theexperiment.

    Sample: Resolution Solution

    Void

    Peak

    1

    Peak2

    Peak

    3

    Peak4

    AU

    0.00

    0.02

    0.04

    0.06

    0.08

    0.10

    0.12

    0.14

    0.16

    0.18

    Minutes

    0.00 0.50 1.00 1.50 2.00 2.50 3.00 3.50 4.00 4.50 5.00 5.50

    1

    2

    3

    4

    5

    Name RTArea

    (V*sec)

    Height

    (V)k'

    Signal

    To Noise

    USP

    Plate Count

    USP

    Resolution

    USP

    Tailing

    Void

    Peak 1

    Peak 2

    Peak 3

    Peak 4

    0. 665

    1. 279

    2. 818

    3. 606

    5. 120

    1780

    459570

    238940

    41372

    26102

    725

    153161

    59382

    8776

    4277

    0.00

    0.92

    3.24

    4.42

    6.70

    32

    6747

    2616

    387

    188

    1742

    4191

    11320

    13266

    15902

    8.34

    16.30

    6.69

    10.34

    1.28

    1.36

    1.21

    1.17

    1.11

    System Suitability Separation Results

    Figure 4: Example of a System Suitability Runs Result

  • 8/4/2019 HPLC Pharma 25-1-09-Numbered-All Print

    5/19

    Created by: Dr. Shula Levin Medtechnica January 26, 2009

    [email protected] Page 5 of 19

    The behavior of HETP as function of linear velocity has

    been described by various equations (35). It is

    frequently called "The Van-Deemter curve", and it is

    frequently used to describe and characterize various

    chromatographic stationary phases' performance andcompare them to each other (36-39). The lower are the

    values of HETP, the more efficient is the

    chromatographic system, enabling the detection of lowerconcentrations due to the enhanced signal-to-noise ratio

    of all the peaks in the chromatogram.

    Peak:Peak 2

    Peak2

    -2.8

    18

    AU

    0.000

    0.015

    0.030

    0.045

    0.060

    Minutes2.72 2.74 2.76 2.78 2.80 2.82 2.84 2.86 2.88 2.90 2.92 2.94 2.96 2.98

    Name Peak 2Retention Time: 2.818K Pr ime: 3.24Area: 238940Height 59382(V)USP Plate Count11320USP Resolution 16.30USP Tailing 1.21Signal To Noise 2616

    W=Width at Tangent

    W at 50%

    A B

    5% of Height

    10% of Height

    Figure 5: System Suitability measurements on a single peak

    PeakAsymmetry Factor,Af and Tailing Factor T

    The chromatographic peak is assumed to have a

    Gaussian shape under ideal conditions, describingnormal distribution of the velocity of the molecules

    populating the peak zone migrating through the

    stationary phase inside the column. Any deviation from

    the normal distribution indicates non-ideality of the

    distribution and the migration process, therefore might

    jeopardize the integrity of the peak's integration,reducing the accuracy of the quantitation. This is the

    reason why USP Tailing is a peak's system suitability

    parameter almost always measured in the system

    suitability step of the analysis.

    The deviation from symmetry is measured by the

    Asymmetry Factor, Af or Tailing Factor T. The

    calculation ofAsymmetry Factor, Af is described by the

    following equation:

    Eqn 6

    )%10(

    )%10(

    h

    h

    fB

    AA =

    WhereA andB are sections in the horizontal line parallel

    to the baseline, drawn at 10% of the peak height asshown in Figure 5.

    The calculation of Tailing Factor T, which is more

    widely used in the pharmaceutical industry, as it is

    suggested by the pharmacopeias, is described by the

    following equation:

    Eqn 7

    )%5(

    )%5()%5(

    2 h

    hh

    A

    BAT

    +=

    Where A and B are sections in the horizontal line parallel

    to the baseline, drawn at 5% of the peak height, as also

    shown in Figure 5. The USP suggests that Tailing

    Factorshould be in the range of 0.5 up to 2 to assure a

    precise and accurate quantitative measurement. The

    peaks in Figures 4 and 5 have a tailing factor within the

    USP requirements.

    Selectivity Factor andResolution Factor Rs

    The separation is a function of the thermodynamics ofthe system. Substances are separated in a

    chromatographic column when their rate of migration

    differs, due to their different distribution between the

    stationary and mobile phases. The Selectivity Factor a

    and Resolution Factor Rs measure the extent of

    separation between two adjacent peaks. Selectivity

    Factor accounts only for the ratio of the retentionfactors k' of the two peaks (k'2/k'1), whereas the

    Resolution Factor Rs accounts for the difference

    between the retention times of the two peaks relative totheir width.

    The equation that describes the experimentalmeasurement of theResolution Factor Rs is as follows:

    Eqn 8)(5.0 12

    )1()2(

    ww

    ttRs

    RR

    +

    =

    Where tR are the retention times of peak 1 and 2

    respectively and w is their respective peak widths at the

    tangents' baseline (see Figure 5). According to the

    pharmacopeias Rs should be above 2 for an accurate

    quantitative measurement. Figure 4 shows that theresolution measured between every two adjacent peaks

    in the chromatogram was above 2, therefore, it wasabove the minimum required.

    The resolution is a critical value when working with

    complex samples such as drug impurities anddegradation products, or when the formulation is

    complex and excipients might interfere with the

    quantitative measurements. Therefore, it is an essential

    part of the system suitability measurement stage before

    the quantitative work of these type of samples.

    The sample used for the measurements ofRs during the

    system suitability runs is sometimes called Resolution

    Solution, as can be seen in Figure 4 It usually containsthe components that are the most difficult to resolve.

  • 8/4/2019 HPLC Pharma 25-1-09-Numbered-All Print

    6/19

    Created by: Dr. Shula Levin Medtechnica January 26, 2009

    [email protected] Page 6 of 19

    The theoretical description of the Resolution Factor Rs

    equation is shown in Equation 9. It includes some of the

    above parameters, the plate countN, the selectivity a and

    the average of the two peaks' capacity factors k':

    Eqn 9 )1'')(1(

    4 +=

    ave

    ave

    kkNRs

    It can be clearly seen from this equation that the plate

    count is the most effecting parameter in the increase of

    the chromatographic resolution. Since the plate count

    increases with the reduction in particle diameter, the

    reason behind the reduction in particle diameter of the

    stationary phase material during the last 3 decades of

    HPLC can be understood. This is also the rational

    behind the recent trend in HPLC, the use of sub 2 micron

    particle columns and the development of a specially

    design of ultra performance HPLC systems toaccommodate such columns (36, 40-42).

    3. The Role of HPLC in Drug Analysis

    The most characteristic feature of the development in the

    methodology of pharmaceutical and biomedical analysis

    during the past 25 years is that HPLC becameundoubtedly the most important analytical method for

    identification and quantification of drugs, either in their

    active pharmaceutical ingredient or in their formulations

    during the process of their discovery, development and

    manufacturing (43-50).

    Figure 6: Steps in a drug development and manufacturing in a

    pharmaceutical company

    Figure 6 summarizes the various stages of the

    pharmaceutical streamline of drug discovery,

    development and manufacturing. Drug development

    starts with the discovery of a molecule with a therapeutic

    value (51, 52). This can be done by high throughput

    screening during which separations by fast or ultra-fast

    HPLC are performed (53). At the discovery stage there

    can be also characterizing synthetic or natural products

    (54-57). DMPK is the step where the candidate

    compounds for drug are tested for their metabolism and

    pharmacokinetics. The studies involve use of LC-MS or

    LC-MS/MS (58-62).

    The next stage is the development stage, where HPLC is

    used to characterize the products of the chemicalsynthesis, by analyzing the active pharmaceutical

    ingredients (API), their impurities (63, 64) and/or

    degradation products generated by accelerated aging (65,

    66). The development of formulation requires also

    studies of the dissolution properties of solid dosage

    forms as well as assays of the pharmaceutical

    formulations (67). Method for the verification of

    system's cleanliness during the manufacturing process

    are developed and used at this stage (68). All the HPLC

    methods that have been finalized at the developmental

    stage are validated and transferred to the manufacturing

    laboratories for a quality control analysis(69-76).

    3.1 The Discovery Stage

    The goal in the discovery stage of drug development is to

    discover a new, safe and active chemical entity (NCE)

    that will become medication for diseases. During the last

    decade parallel synthesis of potential lead compounds,

    using combinatorial chemistry, has been done (51, 77-

    79). The large numbers of products created by the

    combinatorial chemistry are then identified by fast LC-

    MS methods and screened by in-vitro bioassays and/or

    pharmacological or chemical tests to allow a selection of

    a few chosen drug candidates (53, 59, 61, 80, 81).

    When the lead compounds are selected, the next steps

    involve pharmacological studies. Due to its high

    sensitivity and selectivity, HPLC coupled with tandem

    mass spectrometry, HPLC-MS/MS, has become the

    predominant method in bioassays, and pharmacokinetic

    and metabolic studies (58-62, 82). In addition, another

    new development in this field has been the introduction

    of column's packing with ultra-fine particles (

  • 8/4/2019 HPLC Pharma 25-1-09-Numbered-All Print

    7/19

    Created by: Dr. Shula Levin Medtechnica January 26, 2009

    [email protected] Page 7 of 19

    simpler and more automated and usually involves protein

    precipitation and/or solid phase extraction techniques

    (92).

    The chromatographic method is typically a very rapidgradient or isocratic, using small columns' dimensions,

    such as 2.1x50 mm or less. A trace of a typically stable

    daughter ion signal, obtained from the fragmentation of aparent analyte is used for the detection and quantitation.

    Such a mode is called MRM (multiple reaction

    monitoring) or SRM (selective reaction monitoring),

    which is more stable, sensitive and selective than either

    the fluorescence or electrochemical signal. Samples are

    usually ordered in 96 microplates for higher throughput

    and minimizing variability of the sample vials (93). An

    internal standard is used typically to minimize assay

    variability, due to the clean-up stage and MS/MS

    response, which is still less stable than UV response

    (94).

    3.2 The Development Stage

    The development stage is the part during which some of

    the HPLC methods that will be used during the

    subsequent manufacturing stages are developed,

    validated and then transferred. The methods involve

    identification of the drug substance, its assay, impurities,

    stability, the dosage form content uniformity,

    dissolution, etc. The methods are validated first, and then

    transferred to the quality control laboratories. Details

    are given in 3.3.

    All HPLC methods used for the development ofpharmaceuticals and for the determination of theirquality have to be validated. In cases wherebymethods from the Pharmacopoeia's are used, it is not

    necessary to evaluate their suitability, provided that

    the analyses are conducted strictly according to themethods' intended use. In most other cases, especially

    in cases of modification of the drug composition, the

    scheme of synthesis, or the analytical procedure, it isnecessary to re-evaluate the suitability of the HPLC

    method to its original intended use (95).

    The parameters tested throughout the method validation

    as defined by the ICH, USP and FDA and other healthorganizations (95-97) are the following: Specificity,

    selectivity, precision (repeatability, intermediate

    precision, reproducibility or ruggedness), accuracy or

    trueness or bias, linearity range, limit of detection, limit

    of quantitation and robustness.

    The terms selectivity and specificity are often used

    interchangeably. The USP monograph defines selectivity

    of an analytical method as its ability to measure

    accurately an analyte in the presence of interference,

    such as synthetic precursors, excipients, enantiomers and

    known (or likely) degradation products that might bepresent in the sample matrix. A method whose

    selectivity is verified is a "Stability Indicating Method",

    for details please see section 3.3.

    Precision of a method is measured by injecting a series

    of standards and measuring the variability of the

    quantitative results. The measured standard deviation can

    be subdivided into three categories: repeatability,

    intermediate precision, and reproducibility (or

    ruggedness):

    Repeatability is obtained when one operatorusing one system over a relatively short time-

    span carries out the analysis in one laboratory.

    At least 5 or 6 determinations of three different

    matrices at two or three different concentrations

    should be done and the relative standard

    deviation calculated.

    Intermediate precision is a term that has beendefined by ICH (96) as the long-term variability

    of the measurement process and is determinedby comparing the results of a method run within

    a single laboratory over a number of weeks. A

    methods intermediate precision may reflect

    discrepancies in results obtained by different

    operators, from different instruments, with

    standards and reagents from different suppliers,with columns from different batches or a

    combination of these. Objective of intermediate

    precision validation is to verify that in the same

    laboratory the method will provide the same

    results once the development phase is over.

    Reproducibility (or reggedness), as defined by

    ICH represents the precision obtained betweenlaboratories. Objective is to verify that the

    method will provide the same results in

    different laboratories, preparing it for the

    transfer to other sites.

    Typical variations affecting a methods reproducibility

    are:

    Differences in room temperature and humidity;

    Operators with different experience andthoroughness;

    Equipment with different characteristics, suchas delay volume of an HPLC system;

    Variations in material and instrumentconditions, for example different protocols of

    the mobile phases preparation; changes in

    composition, pH, flow rate of mobile phase;

    Equipment and consumables of different ages;

    Columns from different suppliers or differentbatches;

    Solvents, reagents and other material withdifferent quality

    Accuracy of an analytical method is the extent to which

    test results are close to their true value. It is measured

    from the result of a quantitative determination of a well

  • 8/4/2019 HPLC Pharma 25-1-09-Numbered-All Print

    8/19

    Created by: Dr. Shula Levin Medtechnica January 26, 2009

    [email protected] Page 8 of 19

    characterized known sample. The amount measured is

    compared to the known amount.

    Linearity of an analytical method is determined by a

    series of three to six injections of five or more standards

    whose concentrations span 80-120 percent of the

    expected concentration range. The response should be

    (directly or by means of a well-defined mathematical

    calculation) proportional to the concentrations of the

    analytes. A linear regression equation, applied to the

    results, should have an intercept not significantly

    different from zero. If a significant nonzero intercept is

    obtained, it should be demonstrated that there is no effect

    on the accuracy of the method. The range of

    concentrations that an analytical method can be

    implemented on is the interval between the upper and

    lower levels (including these levels) that have been

    demonstrated to have the appropriate precision, accuracy

    and linearity. The range is normally expressed in the

    same units of the test results (e.g. percentage, parts permillion) obtained by the analytical method.

    Limit of detection: It is the lowest concentration of

    analyte in a sample that can be detected but not

    necessarily quantified. In chromatography the detection

    limit is the injected amount that results in a peak with aheight at least twice or three times as high as the baseline

    noise level.

    Limit of quantitation: It is the minimum injected

    amount that gives precise measurements. Inchromatography it typically requires peak heights of 10

    to 20 times higher than baseline noise.

    Robustness of analytical method is a measure of its

    capacity to remain unaffected by small but deliberate

    variations in method parameters and provides an

    indication of its reliability during normal usage.

    Once validated, the methods are ready for transfer to themanufacturing quality control laboratories. Method

    transfer is the last stage of the validation, whereby results

    are tested on both the development and the

    manufacturing sites. Technology transfer is especially

    important in the era of increasing globalization of thepharmaceutical companies (69, 74, 98).

    3.3 The Manufacturing Stage

    Identification

    The identification method by HPLC is aimed to confirmthe identity of the active pharmaceutical ingredient

    inside the samples of either drug substance or drug

    product. Two independent tests are needed, for example,

    one chromatographic and one spectroscopic. Therefore,

    a chromatographic run with a diode-array or MS detector

    will provide both parameters, the retention time in thechromatogram and the spectrum UV or MS of the eluting

    peak, matched against a known standard (99-103)

    Assay and Content Uniformity

    An assay of a sample containing either drug substance or

    drug product quantitatively measures the actual amountof the active ingredient compared to that expected in the

    drug substance assay, whereas in drug products the

    actual amount measured is verified against the labelclaim. The official assays are described in the

    pharmacopeias, such as United State's (USP) (12).

    When a solid dosage form such as tablet or capsule are

    tested, it is usually requested to sample a composite of

    10-20 units to avoid tablet-to-tablet variations. The API

    is extracted from the dosage form in the simplest most

    effective way to optimize recovery. A portion of the

    composite group, equivalent to an average unit dosage

    form weight is taken for the analysis (104, 105). Typical

    specifications for most drug products are 90-110% of the

    label claim.

    A content uniformity test is similar to the drug product

    assay but an individual solid dosage form is tested

    instead of a pool of the tablets/capsules as in the assay

    tests. 10 tablets are taken for the assay, and if the

    specifications of uniformity are not met, more tables are

    taken. The typical calculations are made according to a

    scheme given by the USP (106).

    Most assays and content uniformity methods involve the

    use of UV detection, no extensive sample preparation is

    needed, the standards are usually external, and highly

    qualified(107), typical specifications can be 98-102%purity on dried basis. Specifications for system

    suitability were determined by the pharmacopeia(108).

    Dissolution

    Drug absorption from a solid dosage form after oral

    administration depends on its release from the drug

    product, its dissolution or solubilization under

    physiological conditions, and the permeability across the

    gastrointestinal tract. Because of the critical nature of

    the first two of these steps, in vitro dissolution may be

    relevant to the prediction of in vivo performance. Based

    on this general consideration, in vitro dissolution tests

    for solid oral dosage forms, such as tablets and capsules,

    are used to (1) assess the lot-to-lot quality of a drug

    product; (2) guide the developers of new formulations

    and (3) ensure continuing product quality and

    performance after certain changes, such as changes in the

    formulation, the manufacturing process, the site of

    manufacture, and the scale-up of the manufacturing

    process

    Dissolution test in the pharmaceutical laboratorymeasures the release of the drug substance from its

    dosage form into a dissolution bath under standardized

    conditions, specified by the pharmacopeias (67, 109-116). The US Food and Drug Administration issued a

    guidance for the industry on 1997 (95). The tests are

  • 8/4/2019 HPLC Pharma 25-1-09-Numbered-All Print

    9/19

    Created by: Dr. Shula Levin Medtechnica January 26, 2009

    [email protected] Page 9 of 19

    performed usually on two types of bath apparatus, Type I

    is a basket method and Type II is a paddle method.

    Since most drugs have absorption in the UV-VIS range,

    their dissolution has been traditionally measured by UV-

    VIS spectrometry and HPLC-UV. The advantage ofHPLC over UV spectrometry is its separation

    capabilities, providing higher specificity and sensitivity

    as well as its applicability in formulations with multipleAPI's or very low dose.

    The method is typically isocratic and very short to

    accommodate for the high throughput needed, therefore

    the columns are typically very short. The sample is

    collected from the dissolution apparatus at certain time

    intervals, designed according to its release pattern, and

    the released substance must follow strict release

    specifications. A dissolution profile of Prednisone

    tablets, used to calibrate a dissolution bath are shown in

    Figure 7.

    Dissolution Plot : Prednisone

    Claimed Amount A: 10.00

    Q FactorA ( 30.0 , 4

    Amount

    1.40

    2.10

    2.80

    3.50

    4.20

    Transfer Time (min)

    10.00 20.00 30.00

    Figure 7: Dissolution profile of Prednisone tablets

    Drug Impurities

    Many potential impurities result from the APImanufacturing process, including starting materials,

    isomers, intermediates, reagents, solvents, catalysts and

    reaction by-products (117), therefore, impurity control in

    is essential during the process of drug development and

    manufacturing. HPLC has become the major techniquein this process (63, 64, 118-126).

    The quality of the API starting material is tested by

    HPLC to qualify it for the production of API that meets

    its specifications, as its impurities can be carried through

    directly or can participate in the reaction chemistry toproduce significant impurities in the final product, the

    API. Starting materials for the manufacturing of drug

    substances and drug products can be purchased from

    external suppliers or produced by the firm that also

    manufactures the API. It is likely that different

    synthetic routes that produce different impurities may beused by different suppliers. In some cases, the synthetic

    route is not disclosed to the API producer; therefore, a

    thorough investigation of impurities in starting materials

    must then be conducted. This investigation includes

    method development, a search for potential impurities, as

    well as identification of significant unknown impurities

    and assessment of their impact on the API quality.Results of such an investigation are used to set

    specifications for the starting material that will assure its

    suitability for use in API production (117). Similarconsiderations apply to the drug product. Strict

    international regulatory requirements have been enforced

    for several years as outlined in the International

    Conference on Harmonization (ICH) Guidelines

    Q3A(R), Q3B(R) and Q3C (127-129).

    A typical method for the measurement of impurities can

    be rather complicated and difficult, as it requires high

    resolution to distinguish between closely related

    compounds, high sensitivity to detect low concentrations

    of the impurities and wide dynamic range of the system

    to be able to detect the major components as well as their

    impurities and degradation products in one run. The

    method should have the following important attributes:

    First of all the sample preparation should be assimple as possible, to stay true to the original

    composition of the sample, either a drug

    substance or a drug product, not to lose or

    create an impurity during the process.

    The chromatographic separation should beoptimal, to resolve all the impurities from each

    other and from the API's.

    The quantitative protocol involves frequently acalibration, using well characterized impurities

    standards, and/or calibration against a dilutedstandard of the main API, taking into account

    their relative response factor. Sometimes the

    impurities are just normalized against the main

    peak using % area.

    The main peak is frequently at overloadedconcentrations and if the normalization methodof % area is used, its inclusion within the linear

    range should be established

    According to the ICH guidelines and dependingon the daily dose, the quantification limits of

    unspecified impurities can get as low as 0.05-0.1% of the main peaks. Specified known

    impurities can be reported at higher levels,provided their toxicology was determined.

    Method validation must include theestablishment of limit of detection (to be used

    for rejection of small peaks) and limit ofquantitation (which is used as the minimum

    value for linearity range specifications)

    There is a substantial effort to ensure that themain peak does not hide a co-eluted impurity by

    measuring peak purity (see next section).

    Most impurities methods involve a gradient of the

    mobile phase, to be able to resolve a complex mixture of

    all compounds in one run, polar as well as non-polar. Inrecent years the mobile phase is programmed to be mass

  • 8/4/2019 HPLC Pharma 25-1-09-Numbered-All Print

    10/19

    Created by: Dr. Shula Levin Medtechnica January 26, 2009

    [email protected] Page 10 of 19

    spectrometry compatible, i.e., the buffers used are

    volatile, so that impurities can be identified by LC-MS at

    the developmental and verification stages of the analysis.

    Drug Stability

    Stability testing of drug substance or product is

    performed to ensure that their quality does not vary withtime under the influence of a variety of environmental

    factors such as temperature, humidity, and light (130).

    In addition stability studies are typically done using

    HPLC methods, to determine the re-test period of a drug

    substance and/or the shelf life of the drug product as well

    as its recommended storage conditions. Guidelines to

    the industry are given by the ICH (129).

    An HPLC method used for stability studies is termed:

    Stability Indicating Method (SIM) (65, 66, 131-137).

    According to the FDA guidelines, a SIM is defined as a

    validated analytical procedure that accurately and

    precisely measures active ingredients (drug substance or

    drug product) free from potential interferences like

    degradation products, process impurities, excipients, or

    other potential impurities. FDA recommends that all

    assay procedures for stability studies will be stability

    indicating. A typical chromatogram of a drug substance

    in a stability program is shown in Figure 8. The results

    are shown in Table 2.

    Imp1-3.4

    64

    Imp2-6.6

    72

    Imp3-8.2

    07

    API-10.2

    40

    Imp4-13.8

    79

    14.6

    30

    AU

    0.00000

    0.00024

    0.00048

    0.00072

    0.00096

    Minutes0.00 1.00 2.00 3.00 4.00 5.00 6.00 7.00 8.00 9.00 10.00 11.00 12.00 13.00 14.00 15.00

    Figure 8: A chromatogram of an API sample at a stability program

    The HPLC system used for stability indicating methods

    development and validation must contain 3D data

    capabilities such as diode-array detectors and/or mass

    spectrometers, to be able to detect spectral non-

    homogeneity within the chromatographic peaks (99-101,

    138). Figures 9 and 10 present 3D data obtained by

    photo-diode array and mass detectors respectively. The

    modern UV-VIS diode-array technology is capable of

    evaluating specificity of the method to the analyte in

    question by collecting spectrum at each acquisition data

    point across a peak, and through software manipulations

    each such spectrum is compared to a reference spectrum,to determine spectral homogeneity, hence determine

    peak purity. Figures 11 and 12 demonstrate graphically

    how the software compares spectra collected from

    various points across the peaks to determine their

    similarity. We can see in this example that there is a

    minor interference in peaks 1 and 2, as their UV spectra

    do not perfectly overlap. The mass spectrometer'sspectra in Figure 12 confirm the suspicion and indicate

    the co-eluting interference in each of these two peaks.

    0.00

    0.02

    0.04

    0.06

    0.08

    0.10

    0.12

    0.14

    0.16

    0.18

    0.20

    0.22

    0.24

    AU

    1.15 1.20 1.25 1.30 1.35 1.40 1.45 1.50 1.55 1.60 1.65 1.70 1.75 1.80 1.85

    Minutes

    260.00280.00

    300.00

    320.00

    340.00

    360.00

    380.00

    Wavelength

    (nm)

    Figure 9: 3 dimentional plot of a chromatographic run using a diode-

    array detector

    Since 2006 it has been recommended that a peak purity

    test, based upon photo-diode-array (PDA) detection, will

    also be based on mass spectrometry (MS) to be more

    definite (99, 101, 138). As already was noticed inFigures 9-12 and discussed in the previous section, the

    spectroscopic data obtained by the MS detector is much

    more reliable in evaluating the interferences co-elutingwith the major chromatogram's peaks. Moreover, the

    UV-VIS diode-array detectors might be limited in

    evaluating peak purity in cases whereby the UV-VISspectra of the main peak and its impurities are similar

    and/or the noise in the system is relatively high(raising the detection limit of the impurities).

    0.000e+000

    1.000e+007

    2.000e+007

    3.000e+007

    4.000e+007

    5.000e+007

    6.000e+007

    7.000e+007

    8.000e+007

    9.000e+007

    1.000e+008

    1.100e+008

    Intensity

    1.00 1.05 1.10 1.15 1.20 1.25 1.30 1.35 1.40 1.45 1.50 1.55 1.60 1.65 1.70 1.75 1.80

    Minutes

    200.00

    400.00

    600.00

    800.00

    1000.00

    m/z

    Figure 10: 3 dimentional plot of a chromatographic runusing a mass spectrometer detector

  • 8/4/2019 HPLC Pharma 25-1-09-Numbered-All Print

    11/19

    Created by: Dr. Shula Levin Medtechnica January 26, 2009

    [email protected] Page 11 of 19

    Three conditions must be fulfilled for the major

    components and their related compounds to be effective

    in the peak purity determination by diode-arraydetectors:

    They must have a UV chromophore, or someabsorbance in the wavelength range selected

    There should be a minimal degree ofchromatographic resolution between the main

    peak and its co-eluting impurities

    (Chromatographic resolution between 0.3 to

    0.7).

    There must be some degree of spectraldifference between the analyte peak and the co-

    eluting impurity.

    Stress testing is carried out on a single batch of the drug

    substance and/or the drug product, using forceddegradation of the drug substance (65). The results of

    such studies can help identify the likely degradation

    products, which can in turn help establish the

    degradation pathways and the chemical stability of the

    molecule and to validate the stability indicatingcapabilities of the analytical procedures used.

    nm250.00

    300.00 350.00

    1.19

    1.19

    1.20

    1.18

    1.21

    Peak1

    nm250.00

    300.00 350.00

    1.32

    1.31

    1.33

    1.31

    1.34

    Peak 2

    nm250.00

    300.00 350.00

    1.75

    1.74

    1.76

    1.74

    1.77

    Peak 3

    AU

    0.00

    0.05

    0.10

    0.15

    0.20

    0.25

    Minutes

    1.00 1.10 1.20 1.30 1.40 1.50 1.60 1.70 1.80 1.90 2.00

    AU

    0.00

    0.05

    0.10

    0.15

    0.20

    0.25

    Minutes

    1.00 1.20 1.40 1.60 1.80 2.00

    Peak1 Peak 2 Peak 3

    Figure 11: Chromatogram of three sulfa drugs at 270 nm and a plot

    of UV Spectra collected from each peak's Apex,

    Inflection and two offset points, to demonstrate

    comparison of spectra collected at various points across

    the peak.

    The nature of the stress testing will depend on the

    individual drug substance and the type of drug product

    involved. It usually includes the effect of temperatures,

    humidity, oxidation, and photolysis on the drug

    substance. The testing also evaluates the vulnerability ofthe drug substance to hydrolysis across a wide range of

    pH values when in solution or suspension. Photo-

    stability testing is also an integral part of a stress testing(65, 66, 124, 129, 131, 135, 137).

    SIR Chromatogram

    265.1

    279.1

    311.1

    Intensity

    0.0

    2.0x106

    4.0x106

    6.0x106

    8.0x106

    1.0x107

    1.2x107

    Intensity

    0

    1x107

    2x107

    3x107

    4x107

    5x107

    Intensity

    0

    2x107

    4x107

    6x107

    8x107

    Minutes

    1 .1 0 1. 20 1. 30 1 .4 0 1 .5 0 1 .6 0 1 .7 0 1 .8 0

    Peak 1

    Peak 2

    Peak 3

    Peak 1

    265.1

    266.1267.1 529.2

    530.1

    Intensity

    0.0

    5.0x106

    1.0x107

    1.5x107

    279.1

    280.2

    281.1 557.2

    Intensity

    0

    2x107

    4x107

    6x107

    311.1

    312.2313.1

    Intensity

    0

    2x107

    4x107

    6x107

    8x107

    m/z

    20 0. 00 3 00 .0 0 40 0. 00 500 .00 6 00. 00

    Peak 2

    Peak 3

    MS Spectra

    Figure 12: SIR chromatograms of the same sulfa drugs as in Figure

    6 and their corresponding mass spectra, obtained by

    combining spectra across the entire peak.

    Cleaning Validation

    Cleaning validation tests are performed to assure the

    cleanliness of the pharmaceutical manufacturing

    equipment, such as blender, tablet press, etc. There are

    strict specifications for the maximum allowable amounts

    of the residues, according to their toxicology and daily

    dosage (139). The analytical methods must be extremely

    sensitive, to be able to track the residues of the analytes,

    their degradants or impurities, other contaminations orthe cleaning reagents themselves (85, 140-147). The

    methods must be very sensitive and short, as the entire

    manufacturing process depends on the approval of the

    cleanliness of apparatus. Due to the enhanced sensitivity

    as well as speed of analysis, the use of ultra performance

    liquid chromatography has immediately adopted in the

    cleaning validation tests (85).

    In-Process Control

    In-process control method monitors the progress in the

    manufacturing of an active pharmaceutical ingredient or

    its formulation. High performance liquidchromatography is frequently employed for the in-

    process assay of active pharmaceutical ingredient

    synthesis (68, 72, 148, 149). The results signal the

    production chemist/pharmacist whether to proceed with a

    subsequent unit operation. The decision about whether touse an in-process control test in a manufacturing is

    established during process development and is based on

    scientific judgment. In recent years the FDA issued a

    recommendations for process analytical technology

    (PAT) (150). The guidance intended to describe a

    regulatory framework that will encourage the voluntarydevelopment and implementation of innovative

    pharmaceutical development, manufacturing, and qualityassurance. The new LC ultra performance/rapid

  • 8/4/2019 HPLC Pharma 25-1-09-Numbered-All Print

    12/19

    Created by: Dr. Shula Levin Medtechnica January 26, 2009

    [email protected] Page 12 of 19

    technologies are emerging as the technology of choice

    for the in-process control assay due to their extended

    efficiency, sensitivity and speed of the analysis (68,

    151).

    4. Special Topics

    4.1 Ultra High Performance Liquid Chromatography

    In order to enhance chromatographic performances in

    terms of efficiency and speed, LC has recently evolvedthanks to the development of short columns packed with

    small particles (sub-2 m), working at high pressures(Above 1000 bar) (40, 152-155). This approach has

    been described 30 years ago according to the

    fundamental chromatographic equations (156). However,

    systems and columns compatible with such high

    pressures have been introduced into the market during

    2004 only. Advantages of small particles working at high

    pressure have been vividly discussed in the literature, interms of sensitivity, efficiency, resolution, and analysis

    time, showing how systems working at a maximum

    pressure of 1000 bar give reliable and reproducible

    results. The implementation of the new technology in thepharmaceutical and life sciences is soaring and it

    becomes the next benchmark technology for the near

    future (41, 42, 84, 85, 152, 157).

    There are important experimental parameters oneshould be aware of while reducing the particle size

    (158). First and foremost is the column pressure.

    Equation 10 shows the dependence of column head

    pressure on a number of experimental parametersincluding the particle size. As can be seen in Eqn 10,the pressure is inversely proportional to the square of

    the particle size.

    Eqn 10:2100 pd

    LP

    =

    Where P is the pressure drop, is the flow resistance

    parameter, is viscosity (mPa/s), L iscolumn length(mm), - is the linear velocity (mm/s) and dpparticle

    size (m). So when the particle size is halved, thepressure goes up by a factor of four. However, oftenfor fast separations, the column length is also reduced,

    so the pressure increase is not nearly as high as onewould expect, because pressure is proportional tocolumn length.

    If longer columns such as 100 or 150 mm are required

    to achieve higher resolution and sensitivity, then

    higher pressure pumps are required. Currently, thereare commercial HPLC systems with upper pressure

    limits as high as 15,000 psi. It should be noted that the

    total pressure that the HPLC system experiences is thesum of the column and the instrument backpressures.The latter results when small internal diameter

    capillaries are used in the flow paths to reduce extra

    column effects and minimize the gradient delayvolume (33). As the flow rate increases, the back

    pressure due to these capillaries increasesproportionally.

    An experimental parameter that requires the mostcareful consideration when reducing the columnlength and especially internal diameter is the

    extracolumn band broadening. Some of the modernultra-fast LC columns are only 1.5-cm long with an

    internal diameter of 2.1 mm. Such a column has a

    total void volume of around 33 L. Manyconventional HPLC instruments were developed for

    typical 15 and 25 cm x 4.6 mm analytical columns,where the total column void volumes are 1.6 and 2.6

    mL, respectively. A 1.5 cm x 2.1 mm column packedwith 1.8-m particles often produces a few microliterspeaks, which requires that extra-column band

    broadening will be minimized to accommodate for thetrue advantages of these small columns. Somemodern liquid chromatographs have been designed or

    modified to minimize the extracolumn effects (157).For others, upgrade kits are available to modify some

    older HPLC instruments to work satisfactorily with

    sub-2-m columns.

    Some other experimental parameters that must betaken into account when running fast and ultrafastseparations in LC are as follows (86, 159):

    Detector time constant: Peaks can be only a secondor two wide when short narrow-bore columns run at

    high flow rates. A time constant that is too slowwill make the peaks artificially broad because the

    detector cannot keep up with the rapidly changingsignal.

    Data sampling (acquisition) rate: Data systemneeds enough data points, minimum of 20 across apeak to define the peak as proper for integration of

    area, determine retention time, etc.

    Autosampler cycle time: Separations whose runtime is less than a minute or two, throughput can be

    slowed down by a slow autosampler.Gradient delay volume: If the volume between the

    points, where the gradient forms (mixer), to the

    column head is too large, gradient profiles will becompromised because gradient has not reached

    column in time.

    When transferring methods from HPLC to UPLC there is

    a scale-down process of the sample load, flow-rate, and

    gradient times (86, 159). Sample load must be reduced

    proportionally to the columns volumes, a typical factor

    can be from ~1.5 mL (4.6x150 mm column) to ~0.15 mL(2.1x100 mm column). Equation 11 describes the

    calculations required for such a scale-down.

  • 8/4/2019 HPLC Pharma 25-1-09-Numbered-All Print

    13/19

    Created by: Dr. Shula Levin Medtechnica January 26, 2009

    [email protected] Page 13 of 19

    Eqn 11:

    HPLC

    Uplc

    HPLC

    Uplc

    HPLCUplcID

    ID

    L

    LMM =

    Where M is the sample load in mg, L is column's length

    and ID is internal column's diameter.

    It should be noted that the separation mechanisms inUPLC compared to HPLC are still the same;

    chromatographic principles are maintained while speed,

    sensitivity and resolution is improved. This all supports

    easier method transfer from HPLC to UPLC and its

    revalidation. The main advantages are significant

    reduction of analysis time, which means also reduction

    in solvent consumption, enhanced peak capacity and

    sensitivity, which means that complex analytical

    determination of pharmaceutical preparations can be

    accomplished much easier within a reasonable time.

    Moreover, time spent with new method development,

    optimization and validation is significantly saved.

    A typical chromatogram obtained in the UPLC system is

    shown in Figure 13. A mixture of alkylphonone is

    chromatographed on a 2.1x50 mm Acquity BEH C18

    column packed with 1.7M particles at roomtemperature using a gradient of 5% MeCN to 95%Acetonitril within 3 minutes at flow rate of 0.5 mL/min .

    2-acetylfuran-0.6

    19

    acetanilide

    -0.7

    37

    acetophenone-1.1

    45

    propiophenone-1.5

    04

    butylparaben

    -1.7

    05

    benzophenone-1.9

    12

    valerophenone

    -2.0

    67

    AU

    0.00

    0.05

    0.10

    0.15

    0.20

    0.25

    0.30

    0.35

    0.40

    0.45

    0.50

    Minutes0.00 0.20 0.40 0.60 0.80 1.00 1.20 1.40 1.60 1.80 2.00 2.20

    Figure 13: Mixture of alkylphenones injected into WatersAcquity UPLC using Acquity BEH C18 2.1x50 mm

    1.7 M

    4.2 Preparative Chromatography

    Preparative chromatography is used to isolate and purify

    compounds (6, 73, 160-166). Typically this type of

    separations requires larger columns and the

    chromatograms generally display broader peaks, due to

    the increased column load. The separation is optimizedfor higher throughput (weight/time), then sample load is

    maximized on small analytical columns, and finallyquantities are scaled up to the preparative column

    according to the desired purity and recovery.

    The parameters that will govern the efficiency of the

    preparative separation are the plate count, the capacity

    factor k' and the selectivity between the solutes (see

    section 2). As selectivity, a, between two adjacentpeaks of interest is improved, a better separation between

    them at higher sample loads is obtained, getting better

    product purity. Mobile phase changes cause this changein selectivity, as well as selecting the appropriate

    stationary phase.

    Peak shape is no longer a concern, only the collected

    product's purity. The scaling up of the mass load can be

    done according to the column's volume ratio as is shown

    in Eqn 11, but instead of ratio between UPLC and

    HPLC, the ratio is between the analytical column and the

    preparative one.

    A rational approach to scaling up HPLC runs, using

    adsorption isotherms measurements and calculations has

    been described (167). In the pharmaceutical work

    preparative chromatography is used mostly to isolate

    drug impurities for the purpose of their identification

    and/or using them as working standards for impurities

    determination (168). Purification of compounds during

    drug development and manufacturing has been done in

    recent years using UV in line with mass spectrometer, to

    achieve a mass-directed purification, whereby the mass

    spectrometer is used to determine the cutoff of the

    purified compound and direct the pure compound into its

    collection vessel (169).

    4.3 Chiral Chromatography

    The biological activity of chiral substances often

    depends upon their stereochemistry, since the living

    body is a highly chiral environment (170-172). A large

    percentage of commercial and investigationalpharmaceutical compounds are enantiomers, and many

    of them show significant enantioselective differences in

    their pharmacokinetics and pharmacodynamics. The

    importance of chirality of drugs has been long

    recognized, and the consequence of using them as

    racemates or as enantiomers has been frequentlydiscussed in the pharmaceutical literature (173-175).

    With increasing evidence of problems related tostereoselectivity in drug action, chiral chromatography

    has become the major tool in their analysis (171, 173-

    184). Great efforts have been devoted to the

    development of better methodology for enantioselectivechromatography during the past two decades, and have

    resulted in variety of chiral stationary phases. Chiral

    agents were derivatized and immobilized on the surface

    of the support (silica gel mostly), and served as the in

    situ chiral discriminators during the chromatographicprocess.

    The chiral stationary phases can be categorized into the

    following classes:

  • 8/4/2019 HPLC Pharma 25-1-09-Numbered-All Print

    14/19

    Created by: Dr. Shula Levin Medtechnica January 26, 2009

    [email protected] Page 14 of 19

    A. Chiral affinity by proteins (serum albumin, a1-acid glycoprotein, ovomucoid and

    chymotrypsin) (185).

    B. Stereoselective access to helical chiral polymers(derivatized or free polysaccharides) (186, 187).

    C. Steric interactions between p-Donor p-acceptortype of chiral aromatic amide groups (Pirkle)

    (188).D. Host-guest interactions inside chiral cavities

    (cyclodextrins, crown ethers, macrocyclic

    antibiotics and imprinted polymers) (189-192).

    Major developments in chiral stationary phases have led

    to dramatic advances in enantioselective chromatography

    (175). Chromatographic enantioseparation on chiral

    stationary phases results from energy differences

    between transient diastereomeric complexes formed by

    the solute with the chiral agent on the surface of the

    chiral stationary phase interactions. These differential

    interactions are directly influenced by the mobile phaseenvironment. The solvents in the mobile phase can be

    either normal phase, or aqueous reversed phase's (193) or

    polar organic (187).

    During recent years the trend in the pharmaceutical

    manufacturing of drugs containing chiral centers is todevelop them as single enantiomers rather than

    racemates due to the complexity of the tests andresearch involved in the use of racemates as API's

    (64).

    Conclusion

    HPLC technology has matured to the extent that almost

    any existing organic compound can be analyzed by an

    existing method that can be found in the analytical

    literature, such as professional journals, protocol books

    such as Pharmacopeia's or AOAC manuals. The mostremarkable change in the pharmacopoeias in the past

    25 years has been the increasing importance of HPLCtechnology in the analysis of all aspects of drug

    development and manufacturing.

    References1. Velagaleti, R., Burns, P., and Gill, M. (2003)

    Drug Information Journal37, 407-438.2. Dong, M. W. (2006) Modern HPLC for

    practicing scientists, John Wiley & Sons, New

    Jersey.

    3. Rochet, J. C. (2006)American Journal of

    Pharmaceutical Education70.

    4. Katz, E. (1998) Handbook of HPLC, CRCPress.

    5. Farb, D., Luttrell, A., and Kirsch, R. (2005)

    Pharmaceutical Quality Control Lab Guide

    book, University Of Health Care.

    6. Wellings, D. (2006) A Practical Handbook ofPreparative HPLC, Elsevier Science.

    7. Winslow, P., and Meyer, R. (2004) Compliance

    Handbook for Pharmaceuticals, Medical

    Devices, and Biologics.

    8. Hage, D. S. (2006) Handbook of Affinity

    Chromatography, Taylor & Francis Group.9. Walker, J. M. (2002) The Protein Protocols

    Handbook, Humana Pr Inc.

    10. Ahuja, S., and Dong, M. W. (2005) Handbookof pharmaceutical analysis by HPLC, Elsevier

    Academic Press.

    11. Ahuja, S., and Scypinski, S. (2001) Handbook

    of Modern Pharmaceutical Analysis, Academic

    Press.

    12. USP-NF (2005) United State Pharmacopeia

    National Formulary, United State

    Pharmacopeial Convention, Inc.

    13. Levin, S. (2004)Journal of Liquid

    Chromatography & Related Technologies27,

    1353-1376.

    14. Stella, C., Rudaz, S., Gauvrit, J. Y., Lantri, P.,

    Huteau, A., Tchapla, A., and Veuthey, J. L.

    (2007)Journal of Pharmaceutical andBiomedical Analysis43, 89-98.

    15. Claessens, H. A., and van Straten, M. A. (2004)

    Journal of Chromatography A1060, 23-41.

    16. Marchand, D. H., Snyder, L. R., and Dolan, J.

    W. (2008)Journal of Chromatography A1191,

    2-20.

    17. Luo, H., Ma, L., Zhang, Y., and Carr, P. W.

    (2007)Journal of Chromatography A.

    18. Euerby, M. R., and Petersson, P. (2003)Journal

    of Chromatography A994, 13-36.

    19. Haghedooren, E., Diana, J., Noszal, B.,Hoogmartens, J., and Adams, E. (2007) Talanta

    71, 31-37.

    20. Van Gyseghem, E., Jimidar, M., Sneyers, R.,

    Redlich, D., Verhoeven, E., Massart, D. L., and

    Vander Heyden, Y. (2004)Journal of

    Chromatography A1042, 69-80.

    21. Dehouck, P., Visky, D., Heyden, Y. V., Adams,

    E., Kovacs, Z., Noszl, B., Massart, D. L., and

    Hoogmartens, J. (2004)Journal of

    Chromatography A1025, 189-200.

    22. O'Gara, J. E., Alden, B. A., Walter, T. H.,

    Petersen, J. S., Niederlaender, C. L., and Neue,

    U. D. (1995)Analytical Chemistry67, 3809-

    3813.

    23. Neue, U. D., Walter, T. H., Alden, B. A., Jiang,

    Z., Fisk, R. P., Cook, J. T., Glose, K. H.,

    Carmody, J. L., Grassi, J. M., and Cheng, Y.

    (1999)AMERICAN LABORATORY 31, 36-39.

    24. Neue, U. D., Tran, K. V., Mndez, A., and Carr,

    P. W. (2005)Journal of Chromatography A

    1063, 35-45.

    25. Neue, U. D. (2007)JOURNAL OF

    SEPARATION SCIENCE30, 1611.26. Neue, U. D., OGara, J. E., and Mndez, A.

    (2006)Journal of Chromatography A1127,

    161-174.27. Cheng, Y. F., Walter, T. H., Lu, Z., Iraneta, P.,

    Alden, B. A., Gendreau, C., Neue, U. D.,

  • 8/4/2019 HPLC Pharma 25-1-09-Numbered-All Print

    15/19

    Created by: Dr. Shula Levin Medtechnica January 26, 2009

    [email protected] Page 15 of 19

    Grassi, J. M., Carmody, J. L., and O Gara, J. E.

    (2000)LC GC18, 1162-1173.

    28. Neue, U. D., Phoebe, C. H., Tran, K., Cheng, Y.

    F., and Lu, Z. (2001)Journal of

    Chromatography A925, 49-67.29. Neue, U. D., Mendez, A., Iran, K. V., and

    Diehl, D. M. (2006)HPLC Made to Measure: A

    Practical Handbook for Optimization.30. Gilar, M., Fountain, K. J., Budman, Y., Neue,

    U. D., Yardley, K. R., Rainville, P. D., Russell

    Ii, R. J., and Gebler, J. C. (2002)Journal of

    Chromatography A958, 167-182.

    31. Jenkins, K., Diehl, D., Morrison, D., and

    Mazzeo, J. (2005)LC GC North America, 92-

    93.

    32. Fountain, K. J., Morrison, D., Diehl, D. M., and

    Martin, J. (2007)LC GC North America, 64-65.

    33. Kirkland, J. J., Yau, W. W., Stoklosa, H. J., and

    Dilks Jr, C. H. (1977)J Chromatogr Sci15,

    303-16.

    34. Neue, U. D. (2005)Journal of Chromatography

    A1079, 153-161.

    35. Usher, K. M., Simmons, C. R., and Dorsey, J.

    G. (2008)Journal of Chromatography A.

    36. Wren, S. A. C., and Tchelitcheff, P. (2006)

    Journal of Chromatography A1119, 140-146.

    37. Neue, U. D., and Kele, M. (2007)Journal of

    Chromatography A1149, 236-244.

    38. Jones, M. D., and Plumb, R. S. (2006)J. Sep.

    Sci29, 2409-2420.

    39. Gritti, F., and Guiochon, G. (2006)Journal of

    Chromatography A1128, 45-60.

    40. Swartz, M. (2005)Journal of Liquid

    Chromatography & Related Technologies28,

    1253-1263.

    41. Novakova, L., Matysova, L., and Solich, P.

    (2006) Talanta68, 908-918.

    42. King, S., Peter, J., Stoffolano, E. R., Eichhold,

    T. E., Ii, S. H. H., Baker, T. R., Richardson, E.

    C., and Wehmeyer, K. R. (2005)LC GC North

    America, 36-39.

    43. Schirmer, R. (1991) Modern Methods of

    Pharmaceutical Analysis, CRC Press.

    44. McGill, J., and Holly, D. (1996)Am J Pharm

    Educ60, 370-4.

    45. Tang, L., Fitch, W., Alexander, M., and Dolan,

    J. (2000)Anal. Chem72, 5211-5218.

    46. Lee, D., and Webb, M. (2003) Pharmaceutical

    Analysis, Blackwell Science Ltd.

    47. Lunn, G., and Wiley, J. (2005) HPLC methods

    for recently approved pharmaceuticals, Wiley-

    Interscience.

    48. Gorog, S. (2006) Trends in Analytical

    Chemistry25, 755-757.

    49. Gorog, S. (2007) Trends in Analytical

    Chemistry26, 12-17.50. Kazakevich, Y. V., and LoBrutto, R. (2007)

    HPLC for Pharmaceutical Scientists, Wiley-

    Interscience.

    51. Bleicher, K., Bohm, H., Muller, K., and

    Alanine, A. (2003)Nature Reviews Drug

    Discovery2, 369-378.

    52. Daniel, B. (2007)HPLC for Pharmaceutical

    Scientists.53. Castro-Perez, J., Plumb, R., Granger, J. H.,

    Beattie, I., Joncour, K., and Wright, A. (2005)

    Rapid Commun. Mass Spectrom19, 843848.54. Butler, M. S. (2004)Journal of Natural

    Products67, 2141-2153.

    55. Smyth, W. F., McClean, S., Hack, C. J.,

    Ramachandran, V. N., Doherty, B., Joyce, C.,

    ODonnell, F., Smyth, T. J., OKane, E., and

    Brooks, P. (2006) Trends in Analytical

    Chemistry25, 572-582.

    56. Butler, M. S. (2005)Natural Product Reports22, 162-195.

    57. Lam, K. S. (2007) Trends in Microbiology15,

    279-289.

    58. Chu, I., and Anomeir, A. (2006) Current Drug

    Metabolism7, 467-477.

    59. Kassel, D. B. (2005) Using Mass Spectrometryfor Drug Metabolism Studies.

    60. Lappin, G., and Garner, R. C. (2005)Expert

    Opin. Drug Metab. Toxicol.1, 23-31.

    61. Kassel, D. B. (2004) Current Opinion in

    Chemical Biology8, 339-345.

    62. Wang, J., and Urban, L. (2004)DrugDiscovery, 73.

    63. Ng, L., Lunn, G., and Faustino, P. (2007)

    Analysis of Drug Impurities.

    64. Argentine, M., Owens, P., and Olsen, B. (2007)

    Advanced Drug Delivery Reviews59, 12-28.65. Bakshi, M., and Singh, S. (2002)Journal of

    Pharmaceutical and Biomedical Analysis28,

    1011-1040.

    66. Consultant, P., and Raleigh, N. (2000)Drug

    Stability: Principles and Practices.

    67. Rosanske, T. W., Brown, C. K., and

    Christopher, M. R. a. T. W. R. (1996) in

    Progress in Pharmaceutical and Biomedical

    Analysis, Vol. Volume 3, pp. 169, Elsevier.

    68. Gomis, D. B., Nunez, N. S., Enguita, P. B.,

    Abrodo, P. A., and Alvarez, M. D. G. (2006)

    Journal of Liquid Chromatography & Related

    Technologies29, 931-948.

    69. Rozet, E., Dew, W., Morello, R., Chiap, P.,

    Lecomte, F., Ziemons, E., Boos, K. S.,

    Boulanger, B., Crommen, J., and Hubert, P.

    (2008)Journal of Chromatography A1189, 32-

    41.

    70. Kataoka, H. (2003) Trends in Analytical

    Chemistry22, 232-244.

    71. Koh, H. L., Yau, W. P., Ong, P. S., and Hegde,

    A. (2003)Drug Discovery Today8, 889-897.

    72. Wrezel, P. W., Chion, I., and Hussain, M. S.(2004)LC GC NORTH AMERICA22, 1006-

    1009.

    73. Brandt, A., AG, S., Berlin, G., and Kueppers, S.(2002)LC GC Europe March, 2.

  • 8/4/2019 HPLC Pharma 25-1-09-Numbered-All Print

    16/19

    Created by: Dr. Shula Levin Medtechnica January 26, 2009

    [email protected] Page 16 of 19

    74. Varma, S. (2001) The Quality Assurance

    Journal5, 85-92.

    75. Jimidar, M. I., and De Smet, M. (2007)HPLC

    Method Development for Pharmaceuticals.

    76. Gibson, M. (2005) Technology TrandferIntroduction and Objectives, PDA.

    77. Gardner, C. R., Almarsson, O., Chen, H.,

    Morissette, S., Peterson, M., Zhang, Z., Wang,S., Lemmo, A., Gonzalez-Zugasti, J., and

    Monagle, J. (2004) Computers and ChemicalEngineering28, 943-953.

    78. Maurer, H. (2000) Combinatorial Chemistry &

    High Throughput Screening3, 467-480.

    79. Schultz, L., Garr, C. D., Cameron, L. M., and

    Bukowski, J. (1998)Bioorganic & Medicinal

    Chemistry Letters8, 2409.

    80. Corti, G., Maestrelli, F., Cirri, M., Zerrouk, N.,

    and Mura, P. (2006)European Journal of

    Pharmaceutical Sciences27, 354-362.

    81. Alsenz, J., and Kansy, M. (2007)Advanced

    Drug Delivery Reviews59, 546-567.

    82. Korfmacher, W. A. (2005)Drug DiscoveryToday10, 1357-1367.

    83. Plumb, R., Jones, M., Rainville, P., and

    Nicholson, J. (2008)Journal of

    Chromatographic Science46, 193-198.

    84. Pedraglio, S., Rozio, M. G., Misiano, P., Reali,

    V., Dondio, G., and Bigogno, C. (2007)Journalof Pharmaceutical and Biomedical Analysis44,

    665-673.

    85. Fountain, K. J., Wingerden, M., and Diehl, D.

    M. (2007)LC GC Magazine-North America-

    Solutions for Separation Scientists37, 66-67.86. de Villiers, A., Lestremau, F., Szucs, R.,

    Glbart, S., David, F., and Sandra, P. (2006)

    Journal of Chromatography A1127, 60-69.

    87. Nardo, C. (2005).

    88. Johnson, K. A., and Plumb, R. (2005)Journal

    of Pharmaceutical and Biomedical Analysis39,

    805-810.

    89. Korfmacher, W. A., Cox, K. A., Bryant, M. S.,

    Veals, J., Ng, K., Lin, C., and Watkins, R.

    (1997)Drug Discovery Today2, 532-537.

    90. Wickremsinhe, E. R., Singh, G., Ackermann, B.

    L., Gillespie, T. A., and Chaudhary, A. K.

    (2006) Current Drug Metabolism7, 913-928.

    91. Kamel, A., and Prakash, C. (2006) Current

    Drug Metabolism7, 837-852.

    92. Alnouti, Y., Csanaky, I. L., and Klaassen, C. D.

    (2008)Journal of Chromatography B873, 209.

    93. Mallet, C. R., Lu, Z., Fisk, R., Mazzeo, J. R.,

    and Neue, U. D. (2003)Rapid communications

    in mass spectrometry17, 163-170.

    94. Li, R., Dong, L., and Huang, J. (2005)Analytica

    Chimica Acta546, 167-173.

    95. (CDER), C. f. D. E. a. R. (1997), U.S.Department of Health and Human Services,

    Food and Drug Administration.

    96. ICH-Q2B (1996), Geneva.97. USP-26-NF21 (2003).

    98. Gibson, D. V., and Williams, F. (1990)

    Technology transfer, Sage Publications

    Newbury Park, Calif.

    99. Li, W., and Hu, C. (2008)Journal of

    Chromatography A.100. Gilliard, J. A., and Ritter, C. (1997)Journal of

    Chromatography A786, 1-11.

    101. Lindholm, J., Westerlund, D., Karlsson, K.,Caldwell, K., and Fornstedt, T. (2003)Journal

    of Chromatography A992, 85-100.

    102. Swartz, M. E. (1996) CHIRALITY867, 76.

    103. Rivier, L. (2006)Applications of LC-MS in

    Toxicology.

    104. Lambropoulos, J., Spanos, G. A., and Lazaridis,

    N. V. (1999)Journal of Pharmaceutical and

    Biomedical Analysis19, 793-802.

    105. Berman, J., and Planchard, J. A. (1995)Drug

    Development and Industrial Pharmacy21,

    1257-1283.

    106.

    http://www.usp.org/USPNF/compendi

    alTools.html.

    107. Williams, R. L. (2006)Journal of

    Pharmaceutical and Biomedical Analysis40, 3.

    108. Renger, B. (2000)Journal of Chromatography

    B: Biomedical Sciences and Applications745,

    167.

    109. Aiache, J. M. (1990)Journal of Pharmaceuticaland Biomedical Analysis8, 499.

    110. Zahirul, M., and Khan, I. (1996)International

    Journal of Pharmaceutics140, 131.

    111. QURESHI, S. A. (1996)Drug Information

    Journal30, 1055-1061.

    112. SHIU, G. K. (1996)Drug Information Journal

    30, 10451054.

    113. Castellano, P., Vignaduzzo, S., Maggio, R., and

    Kaufman, T. (2005)Analytical and

    Bioanalytical Chemistry382, 1711-1714.

    114. Bateman, S., Paul, W., Alan, T., and Colin, P.

    (2005) in Encyclopedia of Analytical Science,

    pp. 101, Elsevier, Oxford.

    115. Azarmi, S., Roa, W., and Lbenberg, R. (2007)

    International Journal of Pharmaceutics328,

    12.

    116. Deng, G., Ashley, A., Brown, W., Eaton, J.,

    Hauck, W., Kikwai, L., Liddell, M., Manning,R., Munoz, J., and Nithyanandan, P. (2008)

    Pharmaceutical Research25, 1100-1109.

    117. Gavin, P. F., Olsen, B. A., Wirth, D. D., and

    Lorenz, K. T. (2006)Journal of Pharmaceutical

    and Biomedical Analysis41, 1251-1259.

    118. Smith, R. J., and Webb, M. L. (2007) Analysis

    of Drug Impurities, Blackwell Publishing.

    119. Ahuja, S. (2007)Advanced Drug Delivery

    Reviews59, 3-11.

    120. Kovaleski, J., Kraut, B., Mattiuz, A.,

    Giangiulio, M., Brobst, G., Cagno, W.,

    Kulkarni, P., and Rauch, T. (2007)Advanced

    Drug Delivery Reviews59, 56-63.121. Basak, A., Raw, A., Al Hakim, A., Furness, S.,

    Samaan, N., Gill, D., Patel, H., Powers, R., and

  • 8/4/2019 HPLC Pharma 25-1-09-Numbered-All Print

    17/19

    Created by: Dr. Shula Levin Medtechnica January 26, 2009

    [email protected] Page 17 of 19

    Yu, L. (2007)Advanced Drug Delivery Reviews

    59, 64-72.

    122. Qiu, F. (2007)Journal of Liquid

    Chromatography & Related Technologies30,

    877-935.123. Olsen, B., Castle, B., and Myers, D. (2006)

    Trends in Analytical Chemistry25, 796-805.

    124. Baertschi, S. (2006) Trends in AnalyticalChemistry25, 758-767.

    125. Nageswara Rao, R., and Nagaraju, V. (2003)

    Journal of Pharmaceutical and Biomedical

    Analysis33, 335-377.

    126. Roy, J. (2002)AAPS PharmSciTech3, 1-8.

    127. ICH (2006)INTERNATIONAL CONFERENCE

    ON HARMONISATION OF TECHNICAL

    REQUIREMENTS FOR REGISTRATION OF

    PHARMACEUTICALS FOR HUMAN USE.

    128. ICH (2005)INTERNATIONAL CONFERENCE

    ON HARMONISATION OF TECHNICAL

    REQUIREMENTS FOR REGISTRATION OF

    PHARMACEUTICALS FOR HUMAN USE.

    129. ICH (2003)INTERNATIONAL CONFERENCEON HARMONISATION OF TECHNICAL

    REQUIREMENTS FOR REGISTRATION OF

    PHARMACEUTICALS FOR HUMAN USE.

    130. Carstensen, J., and Rhodes, C. (2000) Drug

    Stability: Principles and Practices, Informa

    Healthcare.

    131. Kumar, V., Bhutani, H., and Singh, S. (2007)

    Journal of Pharmaceutical and Biomedical

    Analysis43, 769-773.

    132. Mohammadi, A., Mehramizi, A., Moghaddam,

    F., jabarian, L., pourfarzib, M., and Kashani, H.(2007)Journal of Chromatography B854, 152-

    157.

    133. Shah, R., Bryant, A., Collier, J., Habib, M., and

    Khan, M. (2008)International Journal of

    Pharmaceutics360, 77-82.

    134. Mohammadi, A., Rezanour, N., Ansari

    Dogaheh, M., Ghorbani Bidkorbeh, F., Hashem,

    M., and Walker, R. (2007)Journal of

    Chromatography B846, 215-221.

    135. Xu, Q. A., Trissel, L. A., and American

    Pharmacists, A. (1999) Stability-indicating

    HPLC Methods for Drug Analysis, American

    Pharmaceutical Association.

    136. Chan, E. C. Y., Wee, P. Y., and Ho, P. C.

    (1999) Clinica Chimica Acta288, 4753.

    137. Swartz, M., and Krull, I. (2005)

    Chromatography Online.

    138. Fourneron, J. D. (2001)Journal of

    Chromatographic Science39.

    139. APIC (1999), Active Pharmaceutical

    Ingredients Committee.

    140. Yang, P., Burson, K., Feder, D., and

    Macdonald, F. (2005) PharmaceuticalTechnology.

    141. Schmidt, A. H. (2006)JOURNAL OF LIQUID

    CHROMATOGRAPHY AND RELATEDTECHNOLOGIES29, 1663.

    142. Zayas, J., Coln, H., Garced, O., and Ramos, L.

    M. (2006)Journal of Pharmaceutical and

    Biomedical Analysis41, 589-593.

    143. Forsatz, B., and Snow, N. (2007)LC GC

    NORTH AMERICA25, 960.144. Kathiresan, K., Kannan, K., Valliappan, K.,

    Karar, P. K., and Manavalan, R. (2007)ASIAN

    JOURNAL OF CHEMISTRY19, 5034.145. Baran, P. A., Bielejewska, A., Glice, M. M.,

    Beczkowicz, H., Maruszak, W., Kosmacinska,

    B., and Golebiewski, P. (2007) PRZEMYSL

    CHEMICZNY86, 747.

    146. Kathiresan, K., Kannan, K., Kannappan, N.,

    Venkatesan, P., Shenoy, K. R. P., and

    Manavalan, R. (2007)ASIAN JOURNAL OF

    CHEMISTRY19, 4438.

    147. Resto, W., Hernndez, D., Rey, R., Coln, H.,

    and Zayas, J. (2007)Journal of Pharmaceutical

    and Biomedical Analysis44, 265-269.

    148. Laviana, L., Llorente, I., Bayod, M., and

    Blanco, D. (2003)Journal of Pharmaceutical

    and Biomedical Analysis32, 167-174.

    149. Moes, J. J., Ruijken, M. M., Gout, E., Frijlink,

    H. W., and Ugwoke, M. I. (2008)International

    Journal of Pharmaceutics357, 108.

    150. Guidance, F. (2004) ((CDER), C. f. D. E. a. R.,

    Ed.), U.S. Department of Health and Human

    Services, Food and Drug Administration.

    151. Anurag S. Rathore, M. Y. S. Y. A. S. (2008)

    Biotechnology and Bioengineering100, 306-

    316.

    152. Swartz, M. E. (2005)LC-GC23, 8-14.

    153. Neue, U. D., Smith, B., and Johnson, K. A.(2007)J. Sep. Sci30, 1158-1166.

    154. Mazzeo, J. R., Neue, U. D., Kele, M., and

    Plumb, R. S. (2005)Analytical

    chemistry(Washington, DC)77, 460-467.

    155. Diehl, D. M., Tran, K., Grumbach, E. S., Neue,

    U. D., and Mazzeo, J. R. (2003)LC GC North

    America, 67-68.

    156. J.C. Giddings, P., (1965).

    157. Jerkovich, A. D., LoBrutto, R., and Vivilecchia,