chapter-i - inflibnetshodhganga.inflibnet.ac.in/bitstream/10603/33340/2/chapter-i-hplc... · liquid...

55
Chapter-I 1 1.1 INTRODUCTION TO CHROMATOGRAPY Liquid chromatography (1-2 ) was defined in the early 1900s by the work of the Russian botanist, Mikhail S. Tswett. His pioneering studies focused on separating compounds [leaf pigments], extracted from plants using a solvent, in a column packed with particles. Tswett filled an open glass column with particles. Two specific materials that he found useful were powdered chalk [calcium carbonate] and alumina. He poured his sample [solvent extract of homogenized plant leaves] into the column and allowed it to pass into the particle bed. This was followed by pure solvent. As the sample passed down through the column by gravity, different colored bands could be seen separating because some components were moving faster than others. He related these separated, different-colored bands to the different compounds that were originally contained in the sample (3-4). He had created an analytical separation of these compounds based on the differing strength of each compound’s chemical attraction to the particles. The compounds that were more strongly attracted to the particles slowed down, while other compounds more strongly attracted to the solvent moved faster. This process can be described as follows: the compounds contained in the sample distribute, or partition differently between the moving solvent, called the mobile phase, and the particles, called the stationary phase. This causes each compound to move at a different speed, thus creating a separation of the compounds (5) . Tswett (6) (Figure-I.1) coined the name chromatography (7) [from the Greek words chroma, meaning color, and graph, meaning writing—literally, color writing] to describe his colorful experiment. [Curiously, the Russian name Tswett means color.] Today, liquid chromatography, in its various forms, has become one of the most powerful tools in analytical chemistry.

Upload: lecong

Post on 18-Apr-2018

227 views

Category:

Documents


2 download

TRANSCRIPT

Chapter-I

1

1.1 INTRODUCTION TO CHROMATOGRAPY

Liquid chromatography (1-2) was defined in the early 1900s by the work

of the Russian botanist, Mikhail S. Tswett. His pioneering studies focused on

separating compounds [leaf pigments], extracted from plants using a solvent, in

a column packed with particles.

Tswett filled an open glass column with particles. Two specific materials

that he found useful were powdered chalk [calcium carbonate] and alumina. He

poured his sample [solvent extract of homogenized plant leaves] into the

column and allowed it to pass into the particle bed. This was followed by pure

solvent. As the sample passed down through the column by gravity, different

colored bands could be seen separating because some components were

moving faster than others. He related these separated, different-colored bands

to the different compounds that were originally contained in the sample (3-4). He

had created an analytical separation of these compounds based on the differing

strength of each compound’s chemical attraction to the particles. The

compounds that were more strongly attracted to the particles slowed down,

while other compounds more strongly attracted to the solvent moved faster.

This process can be described as follows: the compounds contained in the

sample distribute, or partition differently between the moving solvent, called

the mobile phase, and the particles, called the stationary phase. This causes

each compound to move at a different speed, thus creating a separation of the

compounds (5).

Tswett(6) (Figure-I.1) coined the name chromatography(7) [from the

Greek words chroma, meaning color, and graph, meaning writing—literally,

color writing] to describe his colorful experiment. [Curiously, the Russian name

Tswett means color.] Today, liquid chromatography, in its various forms, has

become one of the most powerful tools in analytical chemistry.

Chapter-I

2

Figure-I.1: Tswett's Experiment

1.1.1. Liquid Chromatography (LC) Techniques

Liquid chromatography can be performed using planar or column

techniques. Column liquid chromatography is the most powerful and has the

highest capacity for sample. In all cases, the sample first must be dissolved in a

liquid that is then transported either onto, or into, the chromatographic device.

1.1.2. Thin Layer Chromatography:

The sample is spotted onto, and then flows through, a thin layer of

chromatographic particles (8-10) [stationary phase] fixed onto the surface of a

glass plate [Figure I.2]. The bottom edge of the plate is placed in a solvent. Flow

is created by capillary action as the solvent [mobile phase] diffuses into the dry

particle layer and moves up the glass plate. This technique is called thin-layer

chromatography or TLC (11-13).

Note that the black sample is a mixture of FD&C yellow, red and blue

food dyes that has been chromatographically separated (14-15).

1.1.3. Paper Chromatography:

In Figure I.3, samples are spotted onto paper [stationary phase]. Solvent

[mobile phase] is then added to the center of the spot to create an outward

radial flow. This is a form of paper chromatography. [Classic paper

chromatography is performed in a manner similar to that of TLC with linear

Chapter-I

3

flow.] In the upper image, the same black FD&C dye sample is applied to the

paper.

Figure-I.2: Thin-layer Chromatography

Figure-I.3: Paper Chromatography

Notice the difference in separation power for this particular paper when

compared to the TLC plate. The green ring indicates that the paper cannot

separate the yellow and blue dyes from each other, but it could separate those

Chapter-I

4

dyes from the red dyes. In the bottom image, a green sample, made up of the

same yellow and blue dyes, is applied to the paper. As you would predict, the

paper cannot separate the two dyes. In the middle, a purple sample, made up of

red and blue dyes, was applied to the paper. They are well separated.

1.1.4. Column Chromatography:

In this, the most powerful approach, the sample passes through a

column or a cartridge device containing appropriate particles (16) [stationary

phase]. These particles are called the chromatographic packing material.

Solvent [mobile phase] flows through the device (Figure-I.4). In solid-phase

extraction [SPE], the sample is loaded onto the cartridge and the solvent stream

carries the sample through the device. As in Tswett’s experiment, the

compounds in the sample are then separated by traveling at different

individual speeds through the device. Here the black sample is loaded onto a

cartridge. Different solvents are used in each step to create the separation (17-

18).

Figure-I.4: Column Chromatography – Solid-Phase Extraction [SPE]

When the cartridge format is utilized, there are several ways to achieve

flow. Gravity or vacuum can be used for columns that are not designed to

Chapter-I

5

withstand pressure. Typically, the particles in this case are larger in diameter [>

50 microns] so that there is less resistance to flow. Open glass columns

[Tswett’s experiment] are an example of this. In addition, small plastic columns,

typically in the shape of syringe barrels, can be filled with packing-material

particles and used to perform sample preparation. This is called solid-phase

extraction [SPE]. Here, the chromatographic device, called a cartridge, is used,

usually with vacuum-assisted flow, to clean up a very complex sample before it

is analyzed further.

Smaller particle sizes [<10 microns] are required to improve separation

power. However, smaller particles have greater resistance to flow, so higher

pressures are needed to create the desired solvent flow rate. Pumps and

columns designed to withstand high pressure are necessary. When moderate to

high pressure is used to flow the solvent through the chromatographic column,

the technique is called HPLC.

1.2 HIGH PERFORMANCE LIQUID CHROMATOGRAPHY (HPLC)

The acronym HPLC, coined by the late Prof. Csaba Horváth for his 1970

Pittcon paper, originally indicated the fact that high pressure was used to

generate the flow required for liquid chromatography in packed columns

(Figure-I.5). In the beginning, pumps only had a pressure capability of 500 psi

[35 bar]. This was called high pressure liquid chromatography, or HPLC (19-20).

The early 1970s saw a tremendous leap in technology. These new HPLC

instruments could develop up to 6,000 psi [400 bar] of pressure, and

incorporated improved injectors, detectors, and columns. HPLC really began to

take hold in the mid-to late-1970s. With continued advances in performance

during this time [smaller particles, even higher pressure], the acronym HPLC

remained the same, but the name was changed to high performance liquid

chromatography (21).

High performance liquid chromatography is now one of the most

powerful tools in analytical chemistry. It has the ability to separate, identify,

and quantitate the compounds that are present in any sample that can be

Chapter-I

6

dissolved in a liquid. Today, compounds in trace concentrations as low as parts

per trillion [ppt] may easily be identified. HPLC can be, and has been, applied to

just about any sample, such as pharmaceuticals, food, nutraceuticals, cosmetics,

environmental matrices, forensic samples, and industrial chemicals (22-23).

Figure-I.5: HPLC Column

The components of a basic high-performance liquid chromatography

[HPLC] system are shown in the simple diagram in Figure I.6.

A reservoir holds the solvent [called the mobile phase, because it

moves]. A high-pressure pump [solvent delivery system or solvent manager] is

used to generate and meter a specified flow rate of mobile phase, typically

milliliters per minute. An injector [sample manager or auto sampler] is able to

introduce [inject] the sample into the continuously flowing mobile phase

stream that carries the sample into the HPLC column. The column contains the

chromatographic packing material needed to effect the separation. This packing

material is called the stationary phase because it is held in place by the column

hardware. A detector is needed to see the separated compound bands as they

elute from the HPLC column [most compounds have no color, so we cannot see

them with our eyes]. The mobile phase exits the detector and can be sent to

waste, or collected, as desired. When the mobile phase contains a separated

compound band, HPLC provides the ability to collect this fraction of the eluate

containing that purified compound for further study. This is called preparative

chromatography [discussed in the section on HPLC Scale].

Note that high-pressure tubing and fittings are used to interconnect the

pump, injector, column, and detector components to form the conduit for the

mobile phase, sample, and separated compound bands.

Chapter-I

7

Figure-I.6: High-Performance Liquid Chromatography [HPLC] System

The detector is wired to the computer data station, the HPLC system

component that records the electrical signal needed to generate the

chromatogram on its display and to identify and quantitate the concentration of

the sample constituents (Figure I.7). Since sample compound characteristics

can be very different, several types of detectors have been developed. For

example, if a compound can absorb ultraviolet light, a UV-absorbance detector

is used. If the compound fluoresces, a fluorescence detector is used. If the

compound does not have either of these characteristics, a more universal type

of detector is used, such as an evaporative-light-scattering detector [ELSD]. The

most powerful approach is the use multiple detectors in series. For example, a

UV and/or ELSD detector may be used in combination with a mass

spectrometer [MS] to analyze the results of the chromatographic separation.

This provides, from a single injection, more comprehensive information about

an analyte. The practice of coupling a mass spectrometer to an HPLC system is

called LC/MS.

Chapter-I

8

Figure-I.7: A Typical HPLC [Waters Alliance] System

1.2.1. Ultra Performance Liquid Chromatography (UPLC Technology)

In 2004, further advances in instrumentation and column technology

were made to achieve very significant increases in resolution, speed, and

sensitivity in liquid chromatography. Columns with smaller particles [1.7

micron] and instrumentation with specialized capabilities designed to deliver

mobile phase at 15,000 psi [1,000 bar] were needed to achieve a new level of

performance. A new system had to be holistically created to perform ultra-

performance liquid chromatography, now known as UPLC technology.

Basic research is being conducted today by scientists working with

columns containing even smaller 1-micron-diameter particles and

instrumentation capable of performing at 100,000 psi [6,800 bar]. This

provides a glimpse of what we may expect in the future.

Chapter-I

9

1.2.2. HPLC Operation

A simple way to understand how we achieve the separation of the

compounds contained in a sample is to view the diagram in Figure G.

Mobile phase enters the column from the left, passes through the

particle bed, and exits at the right. Flow direction is represented by green

arrows (Figure: I.8) represents the column at time zero [the moment of

injection], when the sample enters the column and begins to form a band. The

sample shown here, a mixture of yellow, red, and blue dyes, appears at the inlet

of the column as a single black band. [In reality, this sample could be anything

that can be dissolved in a solvent; typically the compounds would be colorless

and the column wall opaque, so we would need a detector to see the separated

compounds as they elute.]

1.2.3. HPLC column:

After a few minutes [lower image], during which mobile phase flows

continuously and steadily past the packing material particles, we can see that

the individual dyes have moved in separate bands at different speeds (24-26). This

is because there is a competition between the mobile phase and the stationary

phase for attracting each of the dyes or analytes. Notice that the yellow dye

band moves the fastest and is about to exit the column. The yellow dye likes [is

attracted to] the mobile phase more than the other dyes. Therefore, it moves at

a faster speed, closer to that of the mobile phase (27-28). The blue dye band likes

the packing material more than the mobile phase. Its stronger attraction to the

particles causes it to move significantly slower. In other words, it is the most

retained compound in this sample mixture. The red dye band has an

intermediate attraction for the mobile phase and therefore moves at an

intermediate speed through the column. Since each dye band moves at different

speed, we are able to separate it chromatographically.

Chapter-I

10

Figure-I.8: Understanding How a Chromatographic Column Works – Bands

A column tube and fittings must contain the chromatographic packing

material [stationary phase] that is used to effect a separation. It must withstand

backpressure created both during manufacture and in use. Also, it must provide

a well-controlled [leak-free, minimum-volume, and zero-dead-volume] flow

path for the sample at its inlet, and analyte bands at its outlet, and be

chemically inert relative to the separation system [sample, mobile, and

stationary phases]. Most columns are constructed of stainless steel for highest

pressure resistance. PEEK™ [an engineered plastic] and glass, while less

pressure tolerant, may be used when inert surfaces are required for special

chemical or biological applications. [Figure I.8].

Figure-I.9: Column Hardware Examples

Chapter-I

11

A glass column wall offers a visual advantage. In the photo in

Figure I.9 & 10, flow has been stopped while the sample bands are still in the

column (29-30). You can see that the three dyes in the injected sample mixture

have already separated in the bed; the yellow analyte, traveling fastest, is just

about to exit the column.

Figure-I.10: A Look Inside a Column

1.2.4. Detectors:

As the separated dye bands leave the column, they pass immediately

into the detector (31-32). The detector contains a flow cell that sees [detects] each

separated compound band against a background of mobile phase [Figure I.11

&12]. [In reality, solutions of many compounds at typical HPLC analytical

concentrations are colorless.] An appropriate detector has the ability to sense

the presence of a compound and send its corresponding electrical signal to a

computer data station (33-34). A choice is made among many different types of

detectors, depending upon the characteristics and concentrations of the

compounds that need to be separated and analyzed, as discussed earlier (35).

Spectroscopic Detection Ultraviolet (UV) Absorption

An ultraviolet light beam is directed through a flow cell and a sensor

measures the light passing through the cell.

If a compound elutes from the column that absorbs this light energy, it will

change the amount of light energy falling on the sensor.

The resulting change in this electrical signal is amplified and directed to a

recorder or data system.

A UV spectrum is sometimes also obtained which may aid in the

identification of a compound or series of compounds.

Chapter-I

12

Figure-I.11: Variable Wavelength Detector

Figure-I.12: Diode Array Detector

1.2.5. Chromatogram

A chromatogram is a representation of the separation that has

chemically [chromatographically] occurred in the HPLC system. A series of

peaks rising from a baseline is drawn on a time axis. Each peak represents the

Chapter-I

13

detector response for a different compound. The chromatogram is plotted by

the computer data station [Figure I.13].

Figure-I.13: How Peaks Are Created

In Figure H, the yellow band has completely passed through the detector

flow cell; the electrical signal generated has been sent to the computer data

station. The resulting chromatogram has begun to appear on screen. Note that

the chromatogram begins when the sample was first injected and starts as a

straight line set near the bottom of the screen. This is called the baseline; it

represents pure mobile phase passing through the flow cell over time. As the

yellow analyte band passes through the flow cell, a stronger signal is sent to the

computer. The line curves, first upward, and then downward, in proportion to

the concentration of the yellow dye in the sample band. This creates a peak in

the chromatogram. After the yellow band passes completely out of the detector

cell, the signal level returns to the baseline; the flow cell now has, once again,

only pure mobile phase in it. Since the yellow band moves fastest, eluting first

from the column, it is the first peak drawn.

A little while later, the red band reaches the flow cell. The signal rises up

from the baseline as the red band first enters the cell, and the peak

representing the red band begins to be drawn. In this diagram, the red band has

not fully passed through the flow cell. The diagram shows what the red band

and red peak would look like if we stopped the process at this moment. Since

Chapter-I

14

most of the red band has passed through the cell, most of the peak has been

drawn, as shown by the solid line. If we could restart, the red band would

completely pass through the flow cell and the red peak would be completed

[dotted line]. The blue band, the most strongly retained, travels at the slowest

rate and elutes after the red band. The dotted line shows you how the

completed chromatogram would appear if we had let the run continue to its

conclusion. It is interesting to note that the width of the blue peak will be the

broadest because the width of the blue analyte band, while narrowest on the

column, becomes the widest as it elutes from the column. This is because it

moves more slowly through the chromatographic packing material bed and

requires more time [and mobile phase volume] to be eluted completely. Since

mobile phase is continuously flowing at a fixed rate, this means that the blue

band widens and is more dilute. Since the detector responds in proportion to

the concentration of the band, the blue peak is lower in height, but larger in

width.

1.2.6. Identification and Quantification of Compounds

In Figure H, three dye compounds are represented by three peaks

separated in time in the chromatogram. Each elutes at a specific location,

measured by the elapsed time between the moment of injection [time zero] and

the time when the peak maximum elutes. By comparing each peak’s retention

time [tR] with that of injected reference standards in the same

chromatographic system [same mobile and stationary phase], a

chromatographer may be able to identify each compound.

Figure-I.14: Identification

Chapter-I

15

In the chromatogram shown in Figure I-14, the chromatographer knew

that, under these LC system conditions, the analyte, acrylamide, would be

separated and elute from the column at 2.85 minutes [retention time].

Whenever a new sample, which happened to contain acrylamide, was injected

into the LC system under the same conditions, a peak would be present at 2.85

minutes [Sample B in Figure I.15].

Once identity is established, the next piece of important information is

how much of each compound was present in the sample. The chromatogram

and the related data from the detector help us calculate the concentration of

each compound. The detector basically responds to the concentration of the

compound band as it passes through the flow cell. The more concentrated it is,

the stronger the signal; this is seen as a greater peak height above the baseline.

Figure-I.15: Identification and Quantitation

In Figure I.15, chromatograms for Samples A and B, on the same time

scale, are stacked one above the other. The same volume of sample was injected

in both runs. Both chromatograms display a peak at a retention time [tR] of 2.85

minutes, indicating that each sample contains acrylamide. However, Sample A

displays a much bigger peak for acrylamide. The area under a peak [peak area

count] is a measure of the concentration of the compound it represents. This

Chapter-I

16

area value is integrated and calculated automatically by the computer data

station. In this example, the peak for acrylamide in Sample A has 10 times the

area of that for Sample B. Using reference standards, it can be determined that

Sample A contains 10 picograms of acrylamide, which is ten times the amount

in Sample B [1 picogram]. Note there is another peak [not identified] that elutes

at 1.8 minutes in both samples. Since the area counts for this peak in both

samples are about the same, this unknown compound may have the same

concentration in both samples.

1.2.7. Isocratic and Gradient program

Two basic elution modes are used in HPLC. The first is called isocratic

elution. In this mode, the mobile phase, either a pure solvent or a mixture,

remains the same throughout the run. A typical system is outlined in Figure I.16.

Figure-I.16: Isocratic LC System

The second type is called gradient elution, wherein, as its name implies,

the mobile phase composition changes during the separation. This mode is useful

for samples that contain compounds that span a wide range of

chromatographic polarity [section on HPLC Separation Modes]. As the

separation proceeds, the elution strength of the mobile phase is increased to

elute the more strongly retained sample components.

Chapter-I

17

Figure-I.17: High-Pressure-Gradient System

In the simplest case, shown in Figure I.17, there are two bottles of

solvents and two pumps. The speed of each pump is managed by the gradient

controller to deliver more or less of each solvent over the course of the

separation. The two streams are combined in the mixer to create the actual

mobile phase composition that is delivered to the column over time. At the

beginning, the mobile phase contains a higher proportion of the weaker solvent

[Solvent A]. Over time, the proportion of the stronger solvent [Solvent B] is

increased, according to a predetermined timetable. Note that in Figure I.18, the

mixer is downstream of the pumps; thus the gradient is created under high

pressure. Other HPLC systems are designed to mix multiple streams of solvents

under low pressure, ahead of a single pump. A gradient proportioning valve

selects from the four solvent bottles, changing the strength of the mobile phase

over time [Figure I.18].

Chapter-I

18

Figure-I.18: Low-Pressure-Gradient System

1.2.8. HPLC Scale [Analytical, Preparative, and Process]

We have discussed how HPLC provides analytical data that can be used

both to identify and to quantify compounds present in a sample. However,

HPLC can also be used to purify and collect desired amounts of each compound,

using a fraction collector downstream of the detector flow cell. This process is

called preparative chromatography [Figure I.19].

In preparative chromatography, the scientist is able to collect the

individual analytes as they elute from the column [e.g., in this example: yellow,

then red, then blue].

Figure-I.19: HPLC System for Purification: Preparative Chromatography

Chapter-I

19

The fraction collector selectively collects the eluate that now contains a

purified analyte, for a specified length of time. The vessels are moved so that

each collects only a single analyte peak.

A scientist determines goals for purity level and amount. Coupled with

knowledge of the complexity of the sample and the nature and concentration of

the desired analytes relative to that of the matrix constituents, these goals, in

turn, determine the amount of sample that needs to be processed and the

required capacity of the HPLC system. In general, as the sample size increases,

the size of the HPLC column will become larger and the pump will need higher

volume-flow-rate capacity. Determining the capacity of an HPLC system is

called selecting the HPLC scale. Table I.1 lists various HPLC scales and their

chromatographic objectives.

Table-I.1: Chromatography Scale

The ability to maximize selectivity with a specific combination of HPLC

stationary and mobile phases—achieving the largest possible separation

between two sample components of interest—is critical in determining the

requirements for scaling up a separation [discussion on HPLC Separation

Modes]. Capacity then becomes a matter of scaling the column volume [Vc] to

the amount of sample to be injected and choosing an appropriate particle size

[determines pressure and efficiency; discussion of Separation Power]. Column

volume, a function of bed length [L] and internal diameter [i.d.], determines the

amount of packing material [particles] that can be contained (Figure I.20).

Chapter-I

20

Figure-I.20: HPLC Column Dimensions

In general, HPLC columns range from 20 mm to 500 mm in length [L]

and 1 mm to 100 mm in internal diameter [i.d.]. As the scale of chromatography

increases, so do column dimensions, especially the cross-sectional area. To

optimize throughput, mobile phase flow rates must increase in proportion to

cross-sectional area. If a smaller particle size is desirable for more separation

power, pumps must then be designed to sustain higher mobile-phase-volume

flow rates at high backpressure. Table I.2 presents some simple guidelines on

selecting the column i.d. and particle size range recommended for each scale of

chromatography.

For example, a semi-preparative-scale application [red X] would use a

column with an internal diameter of 10–40 mm containing 5–15 micron

particles. Column length could then be calculated based on how much purified

compound needs to be processed during each run and on how much separation

power is required.

Table –I.2: Chromatography Scale vs. Column Diameter and Particle Size

Chapter-I

21

1.2.9. Separation Performance – Resolution

The degree to which two compounds are separated is called

chromatographic resolution [RS]. Two principal factors that determine the

overall separation power or resolution that can be achieved by an HPLC column

are: mechanical separation power, created by the column length, particle size,

and packed-bed uniformity, and chemical separation power, created by the

physicochemical competition for compounds between the packing material and

the mobile phase. Efficiency is a measure of mechanical separation power,

while selectivity is a measure of chemical separation power.

1.2.10. Mechanical Separation Power – Efficiency

If a column bed is stable and uniformly packed, its mechanical

separation power is determined by the column length and the particle size.

Mechanical separation power, also called efficiency, is often measured and

compared by a plate number [symbol = N]. Smaller-particle chromatographic

beds have higher efficiency and higher backpressure. For a given particle size,

more mechanical separation power is gained by increasing column length.

However, the trade-offs are longer chromatographic run times, greater solvent

consumption, and higher backpressure. Shorter column lengths minimize all

these variables but also reduce mechanical separation power, as shown in

Figure I.21.

Chapter-I

22

Column Length and Mechanical Separating Power [Same Particle Size]

Figure-I.21: Particle Size and Mechanical Separating Power [Same Column

Length]

For a given particle chemistry, mobile phase, and flow rate, as shown in

Figure I.21, a column of the same length and i.d., but with a smaller particle size,

will deliver more mechanical separation power in the same time. However, its

backpressure will be much higher.

1.2.11. Chemical Separation Power – Selectivity

The choice of a combination of particle chemistry [stationary phase] and

mobile-phase composition—the separation system—will determine the degree

of chemical separation power [how we change the speed of each analyte].

Optimizing selectivity is the most powerful means of creating a separation; this

may obviate the need for the brute force of the highest possible mechanical

efficiency. To create a separation of any two specified compounds, a scientist

may choose among a multiplicity of phase combinations [stationary phase and

Chapter-I

23

mobile phase] and retention mechanisms [modes of chromatography]. These

are discussed in the next section.

1.2.12. HPLC Separation Modes

In general, three primary characteristics of chemical compounds can be

used to create HPLC separations. They are:

• Polarity

• Electrical Charge

• Molecular Size

First, let’s consider polarity and the two primary separation modes that

exploit this characteristic: normal phase and reversed-phase chromatography.

1.2.12.a. Separations Based on Polarity

A molecule’s structure, activity, and physicochemical characteristics are

determined by the arrangement of its constituent atoms and the bonds

between them. Within a molecule, a specific arrangement of certain atoms that

is responsible for special properties and predictable chemical reactions is

called a functional group. This structure often determines whether the

molecule is polar or non-polar. Organic molecules are sorted into classes

according to the principal functional group(s) each contains. Using a separation

mode based on polarity, the relative chromatographic retention of different

kinds of molecules is largely determined by the nature and location of these

functional groups. As shown in Figure P, classes of molecules can be ordered by

their relative retention into a range or spectrum of chromatographic polarity

from highly polar to highly non-polar.

Figure-I.22: Chromatographic Polarity Spectrum by Analyte Functional Group

Chapter-I

24

Water [a small molecule with a high dipole moment] is a polar

compound. Benzene [an aromatic hydrocarbon] is a non-polar compound.

Molecules with similar chromatographic polarity tend to be attracted to each

other; those with dissimilar polarity exhibit much weaker attraction, if any, and

may even repel one another. This becomes the basis for chromatographic

separation modes based on polarity.

Another way to think of this is by the familiar analogy: oil [non-polar] and

water [polar] don’t mix. Unlike in magnetism where opposite poles attract each

other, chromatographic separations based on polarity depend upon the

stronger attraction between likes and the weaker attraction between opposites.

Remember, “Like attracts like” in polarity-based chromatography.

Figure-I.23: Proper Combination of Mobile and Stationary Phases Effects

Separation Based on Polarity

To design a chromatographic separation system [Figure I.23], we create

competition for the various compounds contained in the sample by choosing a

mobile phase and a stationary phase with different polarities. Then, compounds

in the sample that are similar in polarity to the stationary phase [column

packing material] will be delayed because they are more strongly attracted to

the particles. Compounds whose polarity is similar to that of the mobile phase

will be preferentially attracted to it and move faster.

In this way, based upon differences in the relative attraction of each

compound for each phase, a separation is created by changing the speeds of the

analytes.

Figures I.24, 25, and 26 display typical chromatographic polarity ranges

for mobile phases, stationary phases, and sample analytes, respectively. Let’s

consider each in turn to see how a chromatographer chooses the appropriate

Chapter-I

25

phases to develop the attraction competition needed to achieve a polarity-

based HPLC separation.

Figure-I.24: Mobile Phase Chromatographic Polarity Spectrum

A scale, such as that shown in Figure R-1, upon which some common

solvents are placed in order of relative chromatographic polarity is called an

eluotropic series. Mobile phase molecules that compete effectively with analyte

molecules for the attractive stationary phase sites displace these analytes,

causing them to move faster through the column [weakly retained]. Water is at

the polar end of mobile-phase-solvent scale, while hexane, an aliphatic

hydrocarbon, is at the non-polar end. In between, single solvents, as well as

miscible-solvent mixtures [blended in proportions appropriate to meet specific

separation requirements], can be placed in order of elution strength. Which end

of the scale represents the ‘strongest’ mobile phase depends upon the nature of

the stationary phase surface where the competition for the analyte molecules

occurs.

Figure-I.25: Stationary Phase Particle Chromatographic Polarity Spectrum

Silica has an active, hydrophilic [water-loving] surface containing acidic

silanol [silicon-containing analog of alcohol] functional groups. Consequently, it

falls at the polar end of the stationary-phase scale shown in Figure R-2. The

Chapter-I

26

activity or polarity of the silica surface may be modified selectively by

chemically bonding to it less polar functional groups [bonded phase]. Examples

shown here include, in order of decreasing polarity, cyanopropylsilyl- [CN], n-

octylsilyl- [C8], and n-octadecylsilyl- [C18, ODS] moieties on silica. The latter is

a hydrophobic [water-hating], very non-polar packing.

Figure-I.26: Compound/Analyte Chromatographic Polarity Spectrum

After considering the polarity of both phases, then, for a given stationary

phase, a chromatographer must choose a mobile phase in which the analytes of

interest are retained, but not so strongly that they cannot be eluted. Among

solvents of similar strength, the chromatographer considers which phase

combination may best exploit the more subtle differences in analyte polarity

and solubility to maximize the selectivity of the chromatographic system. Like

attracts like, but, as you probably can imagine from the discussion so far,

creating a separation based upon polarity involves knowledge of the sample

and experience with various kinds of analytes and retention modes. To

summarize, the chromatographer will choose the best combination of a mobile

phase and particle stationary phase with appropriately opposite polarities.

Then, as the sample analytes move through the column, the rule like attracts

like will determine which analytes slow down and which proceed at a faster

speed.

1.2.12.b. Normal-Phase HPLC

In his separations of plant extracts, Tswett was successful using a polar

stationary phase [chalk in a glass column; Figure I.27] with a much less polar

Chapter-I

27

[non-polar] mobile phase. This classical mode of chromatography became

known as normal phase.

Figure-I.27: Normal-Phase Chromatography

Figure I.27 represents a normal-phase chromatographic separation of

our three-dye test mixture. The stationary phase is polar and retains the polar

yellow dye most strongly. The relatively non-polar blue dye is won in the

retention competition by the mobile phase, a non-polar solvent, and elutes

quickly. Since the blue dye is most like the mobile phase [both are non-polar], it

moves faster. It is typical for normal-phase chromatography on silica that the

mobile phase is 100% organic; no water is used.

1.2.12.c. Reversed-Phase HPLC

The term reversed-phase describes the chromatography mode that is

just the opposite of normal phase, namely the use of a polar mobile phase and a

non-polar [hydrophobic] stationary phase. Figure I.28 illustrates the black

three-dye mixture being separated using such a protocol.

Figure-I.28: Reversed-Phase Chromatography

Now the most strongly retained compound is the more non-polar blue

dye, as its attraction to the non-polar stationary phase is greatest. The polar

yellow dye, being weakly retained, is won in competition by the polar, aqueous

Chapter-I

28

mobile phase, moves the fastest through the bed, and elutes earliest like

attracts like.

Today, because it is more reproducible and has broad applicability,

reversed-phase chromatography is used for approximately 75% of all HPLC

methods. Most of these protocols use as the mobile phase an aqueous blend of

water with a miscible, polar organic solvent, such as acetonitrile or methanol.

This typically ensures the proper interaction of analytes with the non-polar,

hydrophobic particle surface. A C18–bonded silica [sometimes called ODS] is

the most popular type of reversed-phase HPLC packing.

Table I.3 presents a summary of the phase characteristics for the two

principal HPLC separation modes based upon polarity. Remember, for these

polarity-based modes, like attracts like.

Table-I.3: Phase Characteristics for Separations Based on Polarity

1.2.13. Hydrophilic-Interaction Chromatography [HILIC]

HILIC may be viewed as a variant of normal-phase chromatography. In

normal-phase chromatography, the mobile phase is 100% organic. Only traces

of water are present in the mobile phase and in the pores of the polar packing

particles. Polar analytes bind strongly to the polar stationary phase and may

not elute.

Adding some water [< 20%] to the organic mobile phase [typically an

aprotic solvent like acetonitrile] makes it possible to separate and elute polar

compounds that are strongly retained in the normal-phase mode [or weakly

retained in the reversed-phase mode]. Water, a very polar solvent, competes

effectively with polar analytes for the stationary phase. HILIC may be run in

either isocratic or gradient elution modes. Polar compounds that are initially

attracted to the polar packing material particles can be eluted as the polarity

[strength] of the mobile phase is increased [by adding more water]. Analytes

Chapter-I

29

are eluted in order of increasing hydrophilicity [chromatographic polarity

relative to water]. Buffers or salts may be added to the mobile phase to keep

ionizable analytes in a single form.

1.2.14. Hydrophobic-Interaction Chromatography [HIC]

HIC is a type of reversed-phase chromatography that is used to separate

large biomolecules, such as proteins. It is usually desirable to maintain these

molecules intact in an aqueous solution, avoiding contact with organic solvents

or surfaces that might denature them. HIC takes advantage of the hydrophobic

interaction of large molecules with a moderately hydrophobic stationary phase,

e.g., butyl-bonded [C4], rather than octadecyl-bonded [C18], silica. Initially,

higher salt concentrations in water will encourage the proteins to be retained

[salted out] on the packing. Gradient separations are typically run by decreasing

salt concentration. In this way, biomolecules are eluted in order of increasing

hydrophobicity.

1.2.15. Separations Based on Charge: Ion-Exchange Chromatography [IEC]

For separations based on polarity, like is attracted to like and opposites

may be repelled. In ion-exchange chromatography and other separations based

upon electrical charge, the rule is reversed. Likes may repel, while opposites are

attracted to each other. Stationary phases for ion-exchange separations are

characterized by the nature and strength of the acidic or basic functions on

their surfaces and the types of ions that they attract and retain. Cation exchange

is used to retain and separate positively charged ions on a negative surface.

Conversely, anion exchange is used to retain and separate negatively charged

ions on a positive surface [Figure I.29]. With each type of ion exchange, there

are at least two general approaches for separation and elution.

Chapter-I

30

Figure-I.29: Ion-Exchange Chromatography

Strong ion exchangers bear functional groups [e.g., quaternary amines or

sulfonic acids] that are always ionized. They are typically used to retain and

separate weak ions. These weak ions may be eluted by displacement with a

mobile phase containing ions that are more strongly attracted to the stationary

phase sites. Alternately, weak ions may be retained on the column, then

neutralized by in situ changing the pH of the mobile phase, causing them to lose

their attraction and elute.

Weak ion exchangers [e.g., with secondary-amine or carboxylic-acid

functions] may be neutralized above or below a certain pH value and lose their

ability to retain ions by charge. When charged, they are used to retain and

separate strong ions. If these ions cannot be eluted by displacement, then the

stationary phase exchange sites may be neutralized, shutting off the ionic

attraction, and permitting elution of the charged analytes.

Chapter-I

31

Table-I.4: Ion-Exchange Guidelines

When weak ion exchangers are neutralized, they may retain and

separate species by hydrophobic [reversed-phase] or hydrophilic [normal-

phase] interactions; in these cases, elution strength is determined by the

polarity of the mobile phase. Thus, weak ion exchangers may be used for

mixed-mode separations [separations based on both polarity and charge].

Table D outlines guidelines for the principal categories of ion exchange.

For example, to retain a strongly basic analyte [always positively charged], use a

weak-cation-exchange stationary phase particle at pH > 7; this assures a

negatively charged particle surface. To release or elute the strong base, lower

the pH of the mobile phase below 3; this removes the surface charge and shuts

off the ion-exchange retention mechanism.

Note that a pKa is the pH value at which 50% of the functional group is

ionized and 50% is neutral. To assure an essentially neutral, or a fully charged,

analyte or particle surface, the pH must be adjusted to a value at least 2 units

beyond the pKa, as appropriate [indicated in Table I.4].

Chapter-I

32

Do not use a strong-cation exchanger to retain a strong base; both

remain charged and strongly attracted to each other, making the base nearly

impossible to elute. It can only be removed by swamping the strong cation

exchanger with a competing base that exhibits even stronger retention and

displaces the compound of interest by winning the competition for the active

exchange sites. This approach is rarely practical, or safe, in HPLC and SPE. [Very

strong acids and bases are dangerous to work with, and they may be corrosive

to materials of construction used in HPLC fluidics!]

1.2.16. Separations Based on Size: Size-Exclusion Chromatography [SEC] –

Gel-Permeation Chromatography [GPC]

In the 1950s, Porath and Flodin discovered that biomolecules could be

separated based on their size, rather than on their charge or polarity, by

passing, or filtering, them through a controlled-porosity, hydrophilic dextran

polymer. This process was termed gel filtration. Later, an analogous scheme

was used to separate synthetic oligomers and polymers using organic-polymer

packings with specific pore-size ranges. This process was called gel-permeation

chromatography [GPC]. Similar separations done using controlled-porosity

silica packings were called size-exclusion chromatography [SEC]. Introduced in

1963, the first commercial HPLC instruments were designed for GPC

applications .

All of these techniques are typically done on stationary phases that have

been synthesized with a pore-size distribution over a range that permits the

analytes of interest to enter, or to be excluded from, more or less of the pore

volume of the packing. Smaller molecules penetrate more of the pores on their

passage through the bed. Larger molecules may only penetrate pores above a

certain size so they spend less time in the bed. The biggest molecules may be

totally excluded from pores and pass only between the particles, eluting very

quickly in a small volume. Mobile phases are chosen for two reasons: first, they

are good solvents for the analytes; and, second, they may prevent any

interactions [based on polarity or charge] between the analytes and the

stationary phase surface. In this way, the larger molecules elute first, while the

Chapter-I

33

smaller molecules travel slower [because they move into and out of more of the

pores] and elute later, in decreasing order of their size in solution. Hence the

simple rule: Big ones come out first.

Since it is possible to correlate the molecular weight of a polymer with its size

in solution, GPC revolutionized measurement of the molecular-weight

distribution of polymers that, in turn, determines the physical characteristics

that may enhance, or detract from, polymer processing, quality, and

performance [how to tell good from bad polymer].

1.3 DISSOLUTION

Drug absorption from a solid oral dosage depends on the release

of the drug substance from the drug product (dissolution), the solubility and

the permeability across the gastrointestinal tract (36-37). The first aspect of the

tablets is determined by the manufacture of the product. It is the process by

which a solid or liquid forms a solution in a solvent. In solids this can be

explained as the breakdown of the crystal lattice into individual ions, atoms or

molecules and their transport into the solvent. Dissolution testing is widely

used in the pharmaceutical industry for optimization of formulation (38-39).

A scientific framework for classifying drug substances based on their

aqueous solubility and intestinal permeability (40-43). According to the BCS

(biopharmaceutical classification system) the drug substances are classified as

follows,

Class-1: High Solubility-High Permeability

Class-2: Low Solubility-High Permeability

Class-3: High Solubility- Low Permeability

Class-4: Low Solubility-Low Permeability

1.3.1 Dissolution apparatus

Dissolution profiling methods can be developed based on drug

solubility, dosage form solubility, application of the drug/dosage form. Mainly

the dissolution method development is depends on the selection of dissolution

medium, apparatus, RPM (round per min) and dissolution profile time.

Chapter-I

34

Figure-I.30 shows the Lab India manufactured dissolution apparatus.

Based on application dissolution apparatus are seven types;

Apparatus 1: Basket (37º)

Apparatus 2: Paddle (37º)

Apparatus 3: Reciprocating Cylinder (37º)

Apparatus 4: Flow-Through Cell (37º)

Apparatus 5: Paddle over Disk (32º), Transdermal Delivery System, use

paddle and vessel from Apparatus 2 with a stainless steel disk assembly to hold

the transdermal on the bottom of vessel.

Apparatus 6: Cylinder (32º), Transdermal Delivery System, use

Apparatus 1 except replace the basket shaft with a stainless steel cylinder

element.

Apparatus 7: Reciprocating Holder, for transdermal delivery systems

and also a variety of dosage forms

Figure-I.30: Lab India dissolution apparatus.

1.3.2. Dissolution medium:

Dissolution medium can be selected based on the drug release profile

and application of the drug application in to the body. Generally used

dissolution mediums are, 0.1N (pH ~1.2) or 0.01N (pH ~2.0) hydrochloric acid,

0.05 M sodium acetate buffer, typically at pH 4.5, 0.05 M potassium phosphate

Chapter-I

35

buffer, typically at pH 6.0, 6.8, 7.0 or 7.5, Simulated Gastric Fluid, Simulated

Intestinal Fluid, 0.05 M sodium citrate buffer at pH 2.0 or 3.0 and sodium

borate buffer at pH 8-10. Dissolution profile calculations and acceptance

criteria mentioned in appendix.

1.4 . HPLC METHOD DEVELOPMENT

Reverse phase HPLC is the most commonly used form of HPLC. Recent

days a wide range of improvement has been observed in HPLC instrumentation

and method development (44-48). This is especially true for chromatographs and

detectors (MASS, NMR and IR). Isocratic and gradient reverse-phase HPLC have

evolved as the primary techniques for the analysis of non-volatile drug

substances and impurities. The HPLC detector of choice for many types of

methods development is the photodiode array (PDA) detector because it can be

used for both quantitative and qualitative analysis. The use of a PDA detector to

determine peak purity of the substances in stressed samples greatly facilitates

the development of stability-indicating assays. This trend will continue with the

need to better define degradation pathways. The ultraviolet (UV) absorbance

detector remains the most common HPLC detector for potency and impurity

analysis. Once specificity has been demonstrated, the PDA detector is replaced

with a variable wavelength detector and HPLC effluent is monitored at fixed

wavelengths.

Stability-indicating and impurity methods often are required to measure

analytes within a wide concentration range. Recent food and drugs

administration (FDA) and international conference on harmonization (ICH)

guidance about chiral drug products and impurities has posed new challenges

for methods development scientists (49-53). However, recent advances in the use

of chiral HPLC columns have greatly facilitated progress in this area. Advances

in laboratory robotics and automation are to be applied to method

development and validation. Development teams are using laboratory robotics

to develop automated methods for high-volume tests.

Chapter-I

36

Development steps:

Mainly the method development process consists of different

steps (54-58):

Define method objectives and understand the chemistry (10%):

Determine the goals for method development (e.g. purpose of method

like impurity profile, assay, dissolution and content uniformity) and to

understand the chemistry (UV absorbance, polarity, pH, pKa and solubility) of

the analytes and the drug product.

Initial HPLC conditions (20%):

Develop preliminary HPLC conditions to achieve minimally acceptable

separations, based molecule chemistry like polarity, pH of the molecule, UV

absorbance, selection of suitable buffers, organic modifiers, mobile pH, and

diluent for samples preparation.

Sample preparation procedure (10%):

Develop a suitable sample preparation scheme for the test sample based

on polarity, sample nature (solubility, stability, pH sensitivity and precipitate

property). If it requires heating then clearly monitor the time and temperature

of water bath.

Standardization (10%):

Determine the appropriate standardization method and the use of

relative response factors in calculations. Establish and standardize the sample

solution stability, mobile phase sensitivity (pH and ratio), relative retention

time (RRT), relative response factor (RRF), system suitability conditions

(theoretical plates, resolution and tailing factor).

Final method optimization/robustness (20%):

Identify the “weaknesses” of the method and optimize the method

through experimental design. Understand the method performance with

different conditions, different instrument set ups and different samples.

Method validation (30%):

Perform the complete method validation according to international

conference on harmonization (ICH) guidelines with precision, specificity,

Chapter-I

37

linearity, accuracy, limit of detection (LOD), limit of quantification (LOQ),

robustness and ruggedness.

Selection of chromatographic conditions:

Selection of chromatography:

Selection of chromatography is the primary step in method development

like reverse phase, normal phase and chiral chromatography. This can be

happened from molecule polarity, chiral centers, functional groups present in

the parent moiety.

Selection of detector:

Based on the molecule property UV activity, refractive index,

fluorescence and volatile nature, the detector can be selected. Generally used

detectors are UV/Visible, RI detector, fluorescence, evaporative light scattering

(ELS) detector, conductivity, mass (MS) detector, electro chemical and IR

detector.

Selection of buffer:

Buffer selection is the critical parameter in method development. Buffer

selection depends on molecule retention time, peak shape, column suitability

and resolution. Generally used buffer salts are phosphates, acetates, citrates,

formats, perchlorates, ammonium salts and metal alkyl sulphonates.

Selection of HPLC column:

In HPLC, the column selection is the major factor for better separation

and reproducible results. Column has selected based on the analyte elution,

peak shape and response. For column selection we need to know the molecule

chemistry then only the column selection is happened.

Selection of diluent:

Diluent plays a key role in solution stability. Select the suitable diluents

for standard and test solutions which are easily soluble without forming the

precipitate. Some samples soluble in acids or basic solutions at this time select

the acid or base up to soluble volume only then add neutral solutions as diluent

because this will give good peak shape and increase the life time of the column.

Chapter-I

38

Selection of run time and flow rate:

Run time selection depends on the elution of the analytes, un-retained

peaks. Before finalization of the method run the samples for up to three times

of the main peak elution or late eluted peak. Generally, flow rate range is

between used 0.5mL per min to 2.0mL per min.

Selection of standard and sample solution concentration:

Sample concentrations are selected based on the response of the

analytes at selected detector settings, mobile phase, limit of detection and limit

of quantification levels, resolution of the method, impurities limit for products,

strength of the dosage form and application of the method like dissolution,

assay, content uniformity, related substances.

Selection of mobile phase composition:

Mobile phase optimization depends on, type of modifier (acetonitrile,

methanol or tetra-hydro furan), solvent strength so that k' for all solutes is in

the range of 1-20 and perform stepwise isocratic study in 20% steps starting at

100% organic and gradient determination of % organic.

Selection of injection volume:

Generally used injection volume is 20µL per injection. Sometimes this is

not applicable due to lower strength of the dosage forms or non-coincides of

the combination ingredients at this time individual injection volumes can be

selected.

System suitability:

As per ICH guidelines system suitability parameters are theoretical

plates, resolution and tailing factor (59-62).

1.5 HPLC METHOD VALIDATION

Validation of analytical methods plays a major role in judging the

quality, reliability and consistency of analytical data, which is an integral part of

any good analytical practice. Validation of analytical methods is required by

most regulations and quality standards that impact laboratories (63-65).

Chapter-I

39

The analytical procedure validation defines as "Validation of an

analytical method is the process by which it is established by laboratory

studies, that the performance characteristics of the method meet the

requirements for the intended analytical application".

Typical analytical parameters used in method validation include (Table I.5):

1. Precision

2. Accuracy

3. Linearity

4. Range

5. Ruggedness

6. Limit of detection

7. Limit of quantitation

8. Selectivity/Specificity

9. Robustness

Table-I.5: Method validation parameters and Limits

METHOD VALIDATION PARAMETERS AND ACCEPTANCE CRITERIA Validation Parameter Acceptance Criteria

System suitability

Blank chromatogram shall not have any peak at the retention times of the all peaks (impurities and drug product) with an area more than 0.1%.

The tailing factor for drug product peak from first injection of diluted standard solution should be not more than 2.0.

The peak area ratio of two injections of standard preparation for drug product peak should be not less than 0.9 and not more than 1.1.

The resolution between each peaks like impurities and drug product from system suitability solution should be not less than 2.0

Specificity

Blank interference

No peak shall be detected in the blank at >0.1% of the average standard area response of standard solution at the RT of all impurities and drug product peak.

Placebo Interference

Placebo chromatogram shall not have any peak at the retention times of all peaks (impurities and drug product) with an area

Chapter-I

40

more than 0.1%. Peak purity of drug product shall pass.

Peaks shall not have any flag in purity results table.

Peak purity for each known impurity shall pass for spiked placebo preparation. Peaks shall not have any flag in purity results table.

Known Impurities

There shall be no interference from known impurities to drug product and any unknown impurities.

Impurities shall be adequately resolved from each other and from drug product as well as unknown impurities also.

Peak purity of impurities and drug product shall pass. All peaks shall not have any flag in purity results table.

Degradation Studies (Acid,

Base, Peroxide, Thermal, Humidity,

Neutral, UV light, Visible

light)

Peaks of degradation products shall be adequately resolved from each other and from drug product and known impurities.

Peak purity for drug product shall pass. Peaks shall not have any flag in purity results table.

Peak purity for each known impurities and drug product shall pass. Peaks shall not have any flag in purity results table.

Linearity

Correlation coefficient shall be ≥ 0.999 for each known impurity and drug product (specific cases the limit is 0.997).

Bias at 100% level (i.e., at target concentration) response shall be ≤ 5.0%.

If multiple strengths in the drug product then consider the concentration of samples and standards for linearity levels.

Precision %RSD of percent individual impurity from

6 replicate preparations shall be NMT 15.0%.

Intermediate Precision

%RSD of percent individual impurity from 6 replicate sample preparations of analyst 2 shall be NMT 15.0%.

Cumulative %RSD of assay from 12 replicate sample preparations (analyst 1 and analyst 2 combined) shall be NMT 15.0.

Accuracy If multiple strengths in the drug product,

then consider the standard and sample

Chapter-I

41

solution concentrations for both lower and higher strengths.

For Lower and higher levels need to perform the six preparations.

Individual percent recovery of each analyte at each spike level shall be 85.0% to 115.0%.

Limit of Detection (LOD) and Limit of Quantification (LOQ)

For LOQ, a signal to noise ratio of 9.0 to 11.4 shall be obtained.

For LOD, a signal to noise ratio of 2.0 to 3.4 shall be obtained.

The %RSD of percent individual impurity from 6 replicate preparations at LOQ shall be NMT 15.0%.

The mean recovery of each known impurity from 6 replicate preparations at LOQ shall be 85.0% to 115.0%.

Range

For linearity, correlation coefficient shall be ≥0.999 and bias at 100% response shall be not more than 5%

For precision, %RSD from 6 replicate preparations made in accuracy section (higher level) and at LOQ level shall be NMT 15.0%

For accuracy, individual recovery of each analyte at each spike level shall be 85.0% to 115.0%

Ruggedness (perform the

stability study for 1 and 2 days interval)

Solution stability at

Room Temperature

Standard solution-1 and 2 peak areas similarity factor shall be 0.90 to 1.10.

Difference in percent individual impurity at each time point compared to initial shall be ± 20%.

Difference in percent total impurities at each time point compared to initial shall be ±15%.

If the room temperature stability study results are satisfactory then refrigerator stability is not required.

Solution stability at

Refrigerator

Stability of system

suitability solution

System suitability parameters are within the limit as specified in the test method.

Chapter-I

42

Mobile phase at Room

Temperature

System suitability shall pass at each analysis.

If the room temperature stability study results are satisfactory then no need to perform the refrigerator stability.

Difference in percent individual impurity at each time point compared to initial shall be ± 20%.

Difference in percent total impurities at each time point compared to initial shall be ±15%.

Mobile phase at

Refrigerator

Robustness

(Variations: Mobile phase composition ratio, mobile

phase pH, Flow rate,

Column oven temperature)

System suitability criteria of the method shall be met for all experiments.

Adequate resolution shall be obtained for all impurities from each other and from drug product peak.

Peak purity shall pass for drug product. Peaks shall not have any flag in purity results table.

Peak purity shall pass for each known impurity. Peaks shall not have any flag in purity results table.

Filter validation

Filter validation should be performed with centrifuged, PVDF, NYLON type of filter samples.

Percent difference of each individual impurity for each filtered sample compared to unfiltered sample shall be ±20%.

Percent difference in total impurities of each filtered sample compared to unfiltered sample shall be ±15%.

1.6 CHROMATOGRAPHIC CALCULATIONS

Theoretical Plates (half-height method):

Chapter-I

43

Where:

N = Number of theoretical plates

Ve = Elution volume or retention time (mL, sec, or cm)

h = Peak height

w1/2 = width of the peak at half peak height (mL, sec, or cm)

Theoretical Plates (USP method):

Where:

N = Number of theoretical plates

Ve = Elution volume, retention time or retention distance (mL, sec, or

cm)

h = Peak height

wb = Width of the peak at the base line (mL, sec, or cm)

Peak Asymmetry Factor:

Where:

As = Peak asymmetry factor

b = Distance from the point at peak midpoint to the trailing edge (at

10% of peak height)

a = Distance from the leading edge of of peak to the midpoint (at 10%

of peak height)

Chapter-I

44

Tailing Factor (USP method):

Where:

T = Tailing factor (measured at 5% of peak height)

b = Distance from the point at peak midpoint to the trailing edge

a = Distance from the leading edge of the peak to the midpoint

Height Equivalent to a Theoretical Plate (HETP):

Where:

H = Height equivalent of a theoretical plate

L = Length of the column

N = Number of theoretical plates

Dimensions: when using HPLC or UPLC columns, H is usually expressed in µm.

Resolution:

Chapter-I

45

1.7 PHARMACEUTICAL PRODUCTS

1.7.1 INTRODUCTION

Pharmaceutical products – more commonly known as medicines or drugs – are

a fundamental component of both modern and traditional medicine. It is

essential that such products are safe, effective, and of good quality, and are

prescribed and used rationally.

1.7.2 FLUINDIONE TABLETS

Structure:

Chemical Name: 2-(p-fluorophenyl)-1,3-indandione

Dosage form: Tablets

Maximum Daily Dose: 20 mg

Therapeutic activity: Prevents coagulation (clotting) of blood

Therap-Category: Anticoagulant.

Impurities related to Fluindione:

Three degradants are identified and reported below.

Impurity P1 (2-(4-Fluoro-phenyl)-2-hydroperoxy-indan-1,3-dione)

Chapter-I

46

Impurity P2 (2-(4-Fluoro-phenyl)-2-hydroxy-indan-1,3-dione)

Impurity A (2-phenyl-1,3-indanedione)

1.7.3 CALCIUM PANTOTHENATE, L-CYSTINE, PARA AMINO BENZOIC ACID

AND THIAMINE MONONITRATE CAPSULES

1.7.3.1 Calcium Pantothenate

Structure:

Chapter-I

47

Chemical Name: b-Alanine,N-(2,4-dihydroxy-3,3-dimethyl-1-

oxobutyl)-,calcium salt (2:1),(R)

Dosage form: Capsules

Maximum Daily Dose: 5-10 mg

Therapeutic activity: Pantothenic acid is important for our bodies to

properly use carbohydrates, proteins, and lipids

and for healthy skin

Therap-Category: Nutritional supplement (vitamin)

1.7.3.2 L-Cystine

Structure:

ChemicalName : 3,3’-Disulfanediylbis (2R)-2-aminopropanoic acid

Dosage form : Capsules

Maximum Daily Dose: 0.5 to 1.5 gm

Therapeutic activity: Reducing symptoms of hair pulling.

Cysteine is able to fulfil several important functions

in the human body due to its specific structure

including sulfur and contributes significantly to the

general well-being.

Therap-Category: L-cysteine has anti-inflammatory properties that it

can protect against various toxins, due to its

essential amino acid ability to neutralise toxic

heavy metals in the body.

1.7.3.3 Para amino benzoic acid

Structure:

Chapter-I

48

Chemical Name : 4-Aminobenzoic acid.

Dosage form: Capsules

Maximum Daily Dose: 300-400 mg

Therapeutic activity: PABA is used to improve the protein used in the

body, it relates to red blood cell formation as well

as assisting the manufacture of folic acid in the

intestines. Para-aminobenzoic acid is used in

sunscreen preparations since it can help protect

the skin against ultra-violet radiation.

Therap-Category: PABA has been suggested as a treatment

for Peyronie's disease & scleroderma ( that affects

the connective tissues of the skin and various

organs)

1.7.3.4 Thiamine mononitrate

Structure:

Chemical Name : Thiazolium,3-[(4-amino-2-methyl-5-

pyrimidinyl)methyl]-5-(2-hydroxyethyl)-4-methyl-

,nitrate

Dosage form: Capsules

Maximum Daily Dose: 300 mg

Therapeutic activity: Preventing kidney disease in people with type 2

diabetes.

Therap-Category: Nutritional supplement (vitamin)

Chapter-I

49

1.7.4 GLUCOSAMINE AND CHONDROITIN TABLETS

1.7.4.1 Glucosamine Hydrochloride

Structure:

Chemical Name: (3R,4R,5S)-3-Amino-6-(hydroxymethyl)oxane-

2,4,5-triol

Dosage form: Tablets

Maximum Daily Dose: 1.5 gm

Therapeutic activity: Treatment of osteoarthritis symptoms (knee

degenerative osteoarthritis)

Therap-Category: Bone Modulating

Impurities related to Glucosamine Hydrochloride :

Chemical name Structure

N – Acetyl –

Glucosamine

Pyrazine

5 – Hydroxymethyl – 2

– Furaldehyde

Chapter-I

50

2 – Furaldehyde

Pyrrole – 2 –

Carboxaldehyde

1.7.4.2 Chondroitin

Structure:

Chemical Name: Chondroitin-4-sulfate: R1=H; R2= SO3H; R3=H.

Chondroitin-6-sulfate; R1= SO3H; R2,R3= H

Dosage form: Tablets

Maximum Daily Dose: 800 - 1200 mg

Therapeutic activity:

Treatment of osteoarthritis symptoms (In of osteoarthritis, cartilage in the

joints breaks down. Taking chondroitin sulfate, one of the building blocks of

cartilage, might slow this breakdown). Therap-Category: Bone Modulating

1.8 SCOPE OF THE RESEARCH WORK

The scope of this study is to develop novel stability indicating HPLC

method & validate to know the complete performance of the method for the

below mentioned pharmaceutical products.

Chapter-I

51

1) Fluindione tablets.

2) Kercavit capsules (Calcium pantothenate 60 mg, L-Cystine 200 mg, Para

amino benzoic acid 30 mg & Thiamine mononitrate 60 mg).

3) Glucosamine HCl 500 mg and Chondroitin sulfate 400 mg SR tablets.

The present study is to develop & validate

Assay, Related substances, dissolution, and antioxidant content in

Fluindione tablets.

Single analytical stability indicating assay method for multivitamins in

capsule. ( Brand name: kercavit )

Quantification of Glucosamine HCl impurities in Glucosamine HCl 500

mg and Chondroitin sulfate 400 mg SR tablets.

Chapter-I

52

1.9 REFERENCES

1. http://www.cem.msu.edu/~cem333/Week16.pdf

2. http://www.und.edu/dept/chromatography/Docs/Chromatography.pdf

3. http://faculty.uml.edu/david_ryan/84.314/Instrumental%20Lecture%

2018.pdf

4. http://www.ecs.umass.edu/cee/reckhow/courses/772/772l20/772l20

print.pdf

5. http://www.javeriana.edu.co/Facultades/Ciencias/neurobioquimica/lib

ros/celular/programacell_archivos/Liquid%20Chromatography.pdf

6. Mikhail Tswett., "chromatography" Die Adsorption." 24, 316–326, 1906

7. http://www.chem.uwec.edu/Chem455_S05/Pages/Manuals/LC_Rubins

on.pdf

8. http://www.chem.umass.edu/~samal/269/tlc.pdf

9. http://infohost.nmt.edu/~jaltig/TLC.pdf

10. http://courses.chem.psu.edu/chem36/Experiments/PDF's_for_techniqu

es/TLC.pdf

11. http://www.chem.wisc.edu/courses/342/Fall2004/TLC.pdf

12. http://ocw.mit.edu/courses/chemistry/5-301-chemistry-laboratory-

techniques-january-iap-2012/labs/MIT5_301IAP12_TLC_Handout.pdf

13. http://www.chromsource.com/books/Milestones-TLC.pdf

14. http://sitesmedia.s3.amazonaws.com/chem/files/2012/08/TLC_Primer

.pdf

15. http://www.watrex.cz/watrex/cz/dwn_ds.php?id=MACHEREY_NAGEL_

TLC.pdf

16. http://web.uvic.ca/~berryde/techniques/column%20chrom.pdf

17. http://www.gsgis.k12.va.us/facultyweb/ashore/chemstuff/handouts/c

hromatographypacket.pdf

18. http://courses.chem.psu.edu/chem36/Experiments/PDF's_for_techniqu

es/CC.pdf

19. http://polymer.ustc.edu.cn/xwxx_20/xw/201109/P020110906263097

048536.pdf

Chapter-I

53

20. http://www.ux1.eiu.edu/~cfjpb/teaching/ia/iaprojects/hplc.pdf

21. http://www.arlok.com/articles/High-

Performance%20Liquid%20Chromatography.pdf

22. http://www.hopkinsmedicine.org/mams/mams/middleframe_files/tea

ching_files/me330.884/2005/ms2005-lecture-4-chromatography.pdf

23. http://web.campbell.edu/faculty/bryan/CHEM441/JournalArticles/cer

eal.pdf

24. http://www.discoverysciences.com/uploadedFiles/Home/HPLCCols_Se

lectionInfo_p24to33.pdf

25. http://www.mac-mod.com/pdf/technical-report/036-

ColumnComparisonGuide.pdf

26. http://www.nestgrp.com/pdf/colcare.pdf

27. http://www.teknolab.no/pdf/Restek_p290-329_HPLCcolumns.pdf

28. http://www.chromtech.com/Catalog/ProductLine/brochures/Zorbax_G

uide.pdf

29. http://www.chromatographyonline.com/lcgc/data/articlestandard/lcg

ceurope/262003/61538/article.pdf

30. http://scholar.lib.vt.edu/theses/available/etd-12062002-

051420/unrestricted/JS_thesis.pdf

31. http://lipidlibrary.aocs.org/topics/detect92/file.pdf

32. http://faculty.ksu.edu.sa/Dr.almajed/Documents/LC%20Detector.pdf

33. http://www.colorado.edu/chemistry/chem5181/Lectures/C6_Detector

s.pdf

34. http://www.metabolomics.ca/Project/Presentations/Evaporative%20L

ight%20Scattering%20Detection%20in%20Application%20to%20Lipid

%20Separation%20by%20HPLC.pdf

35. http://www.forumsci.co.il/HPLC/Detectors_handouts.pdf

36. http://www.dissolutiontech.com/DTresour/0503art/DT0503art1.pdf

37. http://mediaserver.aaps.org/meetings/09_PPB/Wed/Track_I/Om_Anan

d.pdf

Chapter-I

54

38. http://www.globalresearchonline.net/journalcontents/volume2issue1/

Article%20008.pdf

39. http://www.dissolutiontech.com/DTresour/200505Articles/DT200505

_A01.pdf

40. http://cpb.pharm.or.jp/cpb/200007/c07_0951.pdf

41. http://www.dissolutiontech.com/DTresour/201211Articles/DT201211

_A05.pdf

42. http://www.usp.org/sites/default/files/usp_pdf/EN/USPNF/2011-02-

25711DISSOLUTION.pdf

43. http://www.fda.gov/downloads/Drugs/GuidanceComplianceRegulator

yInformation/Guidances/ucm070237.pdf

44. http://www.restek.com/pdfs/pres-2003-lc-md.pdf

45. http://www.chem.agilent.com/Library/eseminars/Public/Microsoft%2

0PowerPoint%20- 20rapid%20HPLC%20Method%20Development.pdf

46. http://www.cvg.ca/images/HPLC_Method_Development.pdf

47. http://www.uspnf.com/uspnf/login

48. http://www.edqm.eu/en/european-pharmacopoeia-7th-edition-

1401.html

49. http://www.pharmacopoeia.co.uk/

50. http://www.edqm.eu/en/edqm-homepage-628.html

51. http://www.ipc.gov.in/

52. http://www.fda.gov/

53. Q2 (R1) Validation of Analytical Procedures: Methodology

54. Q3A (R2) & Q3B (R2) Impurities in new drug substances and products

55. Q8 (R2) Pharmaceutical Development

56. Q6A Specifications; Test procedures and acceptance Criteria for new

drug substances and New drug products.

57. WHO The International Pharmacopoeia

58. http://www.fda.gov/downloads/Drugs/Guidances/UCM134409.pdf

59. http://www.cvg.ca/images/SystemStabilityTests.pdf

60. http://www.pharmacopeia.cn/v29240/usp29nf24s0_c621s12.html