SlideShare a Scribd company logo
1 of 12
Download to read offline
Masters of pharmacy, Pharmaceutical technology (Pharmaceutics)
Subject- Modern pharmaceutical analytical system (MPH-101T)
Lesion no- 4, Chromatography By- Drx JAYESH M.RAJPUT
Points:-
1. Paper chromatography: - Paper chromatography is an analytical method used to separate coloured chemicals or substances. A
paper chromatography variant, two-dimensional chromatography involves using two solvents and rotating the paper 90° in between. This is useful
for separating complex mixtures of compounds having similar polarity, for example, amino acids. The setup has three components. The mobile
phase is a solution that travels up the stationary phase, due to capillary action. The mobile phase is generally a mixture of non-polar organic
solvent, while the stationary phase is polar inorganic solvent water. Here paper is used to support the stationary phase, water. Polar water
molecules are held inside the void space of the cellulose network of the host paper. The difference between TLC and paper chromatography is
that the stationary phase in TLC is a layer of adsorbent (usually silica gel, or aluminium oxide), and the stationary phase in paper chromatography
is less absorbent paper.
Types:-
Descending
Development of the chromatogram is done by allowing the solvent to travel down the paper. Here, mobile phase is placed in solvent holder at the
top. The spot is kept at the top and solvent flows down the paper from above.
Ascending
Here the solvent travels up the chromatographic paper. Both descending and ascending paper chromatography are used for the separation of
organic and inorganic substances. The sample and solvent move upward.
Ascending-descending This is the hybrid of both of the above techniques. The upper part of ascending chromatography can be
folded over a rod in order to allow the paper to become descending after crossing the rod.
Circular chromatography
A circular filter paper is taken and the sample is deposited at the center of the paper. After drying the spot, the filter paper is tied
horizontally on a Petri dish containing solvent, so that the wick of the paper is dipped in the solvent. The solvent rises through the
wick and the components are separated into concentric rings.
Two-dimensional
In this technique a square or rectangular paper is used. Here the sample is applied to one of the corners and development is
performed at a right angle to the direction of the first run.
Factors affecting Rf value in paper chromatography
 The solvent system.
 Composition of the mobile phase.
 The working temperature of the system.
 The quality of the paper used.
 The distance through which the solvent runs.
 The quality and nature of solvents used.
 The polarity of components.
 The pH of the solvent or mobile phase.
Typical Uses of Paper Chromatography
• Separating Colored Pigments An effective technique used for separating colored pigments from a mixture. How does it work?
A few drops of the colored pigments mixture are placed on the filter paper and then it is slowly submerged into a jar of solvent.
Depending on their polarity, it dissolves the molecules present in the mixture, as the solvent rises up the paper. As the solvent
continues to rise up the filter paper, molecules of each pigment leaves the solution at different places because of different polarity.
Hence, every pigment rises up to a particular level on the chromatography paper and gets separated in the process.
It is useful in separation of plant pigments.
• Reaction Monitoring Over a period of time, the concentration of reactants decreases, whereas the concentration of production
increases in a chemical reaction. One can get a fair idea on the progress of reaction by spotting the reactants and developing the
chromatogram over different time intervals. The availability of densitometers made quantitative estimations possible, but
traditionally the technique was used for qualitative monitoring. However, as a reaction monitoring option, the rapid methods using
spectroscopic techniques are limiting the paper chromatography application.
• Qualitative Analysis To analyze or separate the different constituents of a mixture, paper chromatography is used. It is one of
the methods of qualitative analysis. We can say it as a useful tool for separating polar as well as non polar solutes. To analyze the
different compounds in drugs, most of the pharmaceutical companies use this technique.
It is used in determining the pollutants in water and testing of antibiotics.
• Isolation And Purification For components of mixture, paper chromatography has been put to use as a purification and
isolation technique. Using spectro-photometric methods, the separated components on the paper are cut, dissolved in suitable
solvents and their absorption is characterized at specific wavelengths.
• Pathology And Forensic Science For investigation of crimes, paper chromatography is useful in the field of forensic science,
as this process can be successfully carried out with even very small quantities of material. Using this technique, samples from
crime scenes are collected to be analyzed and identified.
Used in DNA and RNA fingerprinting. Moreover, to detect the presence of alcohol or chemicals in blood, pathological laboratories
use paper chromatography.
• Foods Both natural and synthetic food colors are added to foods to improve their acceptability and to make them more popular.
Paper chromatography has been primarily used for analysis of food colors in ice creams, sweets, drinks and beverages, jams and
jellies. To ensure that no non-permitted coloring agents are added to the foods, only edible colors are permitted for use. That's how
quantification and identification becomes more important.
• Analyzing Complex Mixtures Certain organic compounds such as carbohydrates and amino acids are identified or detected
from a complex mixture of organic compounds with the help of paper chromatography. It is useful in the separation of anions and
amino acids.
Ranging from forensic investigations, pharmaceuticals, environmental monitoring and foods, paper chromatography have retained
their ground in various fields. For matching the requirements of high throughput laboratories, chromatography has seen
phenomenal growth in terms of software's, applications and increased automation and separation in general.
2. Thin layer chromatography: - Thin-layer chromatography (TLC) is a chromatography technique used to
separate non-volatile mixtures.[1] Thin-layer chromatography is performed on a sheet of an inert substrate such as glass, plastic, or
aluminium foil, which is coated with a thin layer of adsorbent material, usually silica gel, aluminium oxide (alumina), or cellulose.
This layer of adsorbent is known as the stationary phase.
After the sample has been applied on the plate, a solvent or solvent mixture (known as the mobile phase) is drawn up the plate
via capillary action. Because different analytes ascend the TLC plate at different rates, separation is achieved.[2] The mobile phase
has different properties from the stationary phase. For example, with silica gel, a very polar substance, non-polar mobile phases
such as heptane are used. The mobile phase may be a mixture, allowing chemists to fine-tune the bulk properties of the mobile
phase.
Separation process and principle[edit]
Different compounds in the sample mixture travel at different rates due to the differences in their attraction to the stationary phase
and because of differences in solubility in the solvent.[6] By changing the solvent, or perhaps using a mixture, the separation of
components (measured by the Rf value) can be adjusted. Also, the separation achieved with a TLC plate can be used to estimate
the separation of a flash chromatography column. (A compound elutes from a column when the amount of solvent collected is
equal to 1/Rf.)[7] Chemists often use TLC to develop a protocol for separation by chromatography and use TLC to determine which
fractions contain the desired compounds.
Separation of compounds is based on the competition of the solute and the mobile phase for binding sites on the stationary
phase.[3] For instance, if normal-phase silica gel is used as the stationary phase, it can be considered polar. Given two compounds
that differ in polarity, the more polar compound has a stronger interaction with the silica and is, therefore, better able to displace the
mobile phase from the available binding sites. As a consequence, the less polar compound moves higher up the plate (resulting in
a higher Rf value).[6] If the mobile phase is changed to a more polar solvent or mixture of solvents, it becomes better at binding to
the polar plate and therefore displacing solutes from it, so all compounds on the TLC plate will move higher up the plate. It is
commonly said that "strong" solvents (eluents) push the analyzed compounds up the plate, whereas "weak" eluents barely move
them. The order of strength/weakness depends on the coating (stationary phase) of the TLC plate. For silica gel-coated TLC
plates, the eluent strength increases in the following order: perfluoroalkane (weakest), hexane, pentane, carbon
tetrachloride, benzene/toluene, dichloromethane, diethyl ether, ethyl acetate, acetonitrile, acetone, 2-propanol/n-
butanol, water, methanol, triethylamine, acetic acid, formic acid (strongest). For C18-coated plates the order is reverse. In other
words, when the stationary phase is polar and the mobile phase is nonpolar, the method is normal-phase as opposed to reverse-
phase. This means that if a mixture of ethyl acetate and hexane as the mobile phase is used, adding more ethyl acetate results in
higher Rf values for all compounds on the TLC plate. Changing the polarity of the mobile phase will normally not result in reversed
order of running of the compounds on the TLC plate. An eluotropic series can be used as a guide in selecting a mobile phase. If a
reversed order of running of the compounds is desired, an apolar stationary phase should be used, such as C18-functionalized
silica.
Applications
Characterization
In organic chemistry, reactions are qualitatively monitored with TLC. Spots sampled with a capillary tube are placed on the plate: a
spot of starting material, a spot from the reaction mixture, and a cross-spot with both. A small (3 by 7 cm) TLC plate takes a couple
of minutes to run. The analysis is qualitative, and it will show if the starting material has disappeared, i.e. the reaction is complete, if
any product has appeared, and how many products are generated (although this might be underestimated due to co-elution).
Unfortunately, TLCs from low-temperature reactions may give misleading results, because the sample is warmed to room
temperature in the capillary, which can alter the reaction—the warmed sample analyzed by TLC is not the same as what is in the
low-temperature flask. One such reaction is the DIBALH reduction of ester to aldehyde.
In one study TLC has been applied in the screening of organic reactions,[11] for example in the fine-tuning of BINAP synthesis
from 2-naphthol. In this method, the alcohol and catalyst solution (for instance iron(III) chloride) are placed separately on the
baseline, then reacted, and then instantly analyzed.
A special application of TLC is in the characterization of radiolabeled compounds, where it is used to determine radiochemical
purity. The TLC sheet is visualized using a sheet of photographic film or an instrument capable of measuring radioactivity. It may
be visualized using other means as well. This method is much more sensitive than the others and can be used to detect an
extremely small amount of a compound, provided that it carries a radioactive atom.
Isolation
Since different compounds will travel a different distance in the stationary phase, chromatography can be used to isolate
components of a mixture for further analysis. The separated compounds each occupying a specific area on the plate, they can be
scraped off (along with the stationary phase particles) and dissolved into an appropriate solvent. As an example, in the
chromatography of an extract of green plant material (for example spinach) shown in 7 stages of development, Carotene elutes
quickly and is only visible until step 2. Chlorophyll A and B are halfway in the final step and lutein the first compound staining
yellow. Once the chromatography is over, the carotene can be removed from the plate, extracted into a solvent and placed into
a spectrophotometer to determine its spectrum. The quantities extracted are small and a technique such as column
chromatography is preferred to separate larger amounts. However, big preparative TLC plates with thick silica gel coatings can be
used to separate more than 100 mg of material.
Examining reactions and compound stability
TLC is also used for the identification of the completion of any chemical reaction. To determine this it is observed that at the
beginning of a reaction the entire spot is occupied by the starting chemicals or materials on the plate. As the reaction starts taking
place the spot formed by the initial chemicals starts reducing and eventually replaces the whole spot of starting chemicals with a
new product present on the plate. The formation of an entirely new spot determines the completion of a reaction.[12]
Furthermore, two-dimensional TLC is frequently used as a method to check if a compound is stable in the stationary phase (such
as silica gel, which is usually slightly acidic). For this purpose, the tested compound mixture is eluted twice in a square-shaped TLC
plate, first in one direction and then rotated 90º. If the target compound appears on the diagonal of the square, it is stable in silica
gel and safe to purify. If it appears below the diagonal, it is decomposing on silica gel. If this is the case, purification can be
attempted using neutralized silica gel (with triethylamine, for example), or an alternative stationary phase such as neutral alumina.
3. Column chromatography. Column chromatography in chemistry is a chromatography method used to isolate a
single chemical compound from a mixture. Chromatography is able to separate substances based on differential adsorption of
compounds to the adsorbent; compounds move through the column at different rates, allowing them to be separated into fractions.
The technique is widely applicable, as many different adsorbents (normal phase, reversed phase, or otherwise) can be used with a
wide range of solvents. The technique can be used on scales from micrograms up to kilograms. The main advantage of column
chromatography is the relatively low cost and disposability of the stationary phase used in the process. The latter prevents cross-
contamination and stationary phase degradation due to recycling. Column chromatography can be done using gravity to move the
solvent, or using compressed gas to push the solvent through the column.
Column preparation
A column is prepared by packing a solid absorbent into a cylindrical glass or plastic tube. The size will depend on the amount of
compound being isolated. The base of the tube contains a filter, either a cotton or glass wool plug, or glass frit to hold the solid
phase in place. A solvent reservoir may be attached at the top of the column.
Two methods are generally used to prepare a column: the dry method and the wet method. For the dry method, the column is first
filled with dry stationary phase powder, followed by the addition of mobile phase, which is flushed through the column until it is
completely wet, and from this point is never allowed to run dry.[1] For the wet method, a slurry is prepared of the eluent with the
stationary phase powder and then carefully poured into the column. The top of the silica should be flat, and the top of the silica can
be protected by a layer of sand. Eluent is slowly passed through the column to advance the organic material.
The individual components are retained by the stationary phase differently and separate from each other while they are running at
different speeds through the column with the eluent. At the end of the column they elute one at a time. During the entire
chromatography process the eluent is collected in a series of fractions. Fractions can be collected automatically by means of
fraction collectors. The productivity of chromatography can be increased by running several columns at a time. In this case multi
stream collectors are used. The composition of the eluent flow can be monitored and each fraction is analyzed for dissolved
compounds, e.g. by analytical chromatography, UV absorption spectra, or fluorescence. Colored compounds (or fluorescent
compounds with the aid of a UV lamp) can be seen through the glass wall as moving bands.
Column Chromatography Procedure
Before starting with the Column Chromatography Experiment let us understand the different phases involved.
Mobile phase – This phase is made up of solvents and it performs the following functions:
1. It acts as a solvent – sample mixture can be introduced in the column.
2. It acts as a developing agent – helps in the separation of components in the sample to form bands.
3. It acts as an eluting agent – the components that are separated during the experiment are removed from the column
4. Some examples of solvents used as mobile phase based on their polarity are – ethanol, acetone, water, acetic acid,
pyridine, etc.
Types of Column Chromatography:
1. Adsorption column chromatography – Adsorption chromatography is a technique of separation, in which the components of the
mixture are adsorbed on the surface of the adsorbent.
2. Partition column chromatography – The stationary phase, as well as mobile phase, are liquid in partition chromatography.
3. Gel column chromatography – In this method of chromatography, the separation takes place through a column packed with gel.
The stationary phase is a solvent held in the gap of a solvent.
4. Ion exchange column chromatography – A chromatography technique in which the stationary phase is always ion exchange
resin.
Column Chromatography Applications
 Column Chromatography is used to isolate active ingredients.
 It is very helpful in Separating compound mixtures.
 It is used to determine drug estimation from drug formulations
 It is used to remove impurities.
 Used to isolation metabolites from biological fluids.
4. Gas chromatography. Gas chromatography (GC) is a common type of chromatography used in analytical
chemistry for separating and analyzing compounds that can be vaporized without decomposition. Typical uses of GC include
testing the purity of a particular substance, or separating the different components of a mixture (the relative amounts of such
components can also be determined). In some situations, GC may help in identifying a compound. In preparative chromatography,
GC can be used to prepare pure compounds from a mixture.[1][2]
In gas chromatography, the mobile phase (or "moving phase") is a carrier gas, usually an inert gas such as helium or
an unreactive gas such as nitrogen. Helium remains the most commonly used carrier gas in about 90% of instruments although
hydrogen is preferred for improved separations.[3] The stationary phase is a microscopic layer of liquid or polymer on an
inert solid support, inside a piece of glass or metal tubing called a column (a homage to the fractionating column used in
distillation). The instrument used to perform gas chromatography is called a 'gas chromatograph' (or "aerograph", "gas separator").
The gaseous compounds being analyzed interact with the walls of the column, which is coated with a stationary phase. This
causes each compound to elute at a different time, known as the 'retention time' of the compound. The comparison of retention
times is what gives GC its analytical usefulness.
Instrumentation
Autosamplers
The autosampler provides the means to introduce a sample automatically into the inlets. Manual insertion of the sample is possible
but is no longer common. Automatic insertion provides better reproducibility and time-optimization.
Different kinds of autosamplers exist. Autosamplers can be classified in relation to sample capacity (auto-injectors vs.
autosamplers, where auto-injectors can work a small number of samples), to robotic technologies (XYZ robot[9] vs. rotating robot –
the most common), or to analysis:
 Liquid
 Static head-space by syringe technology
 Dynamic head-space by transfer-line technology
 Solid phase microextraction (SPME)
Inlets (injectors)
The column inlet (or injector) provides the means to introduce a sample into a continuous flow of carrier gas. The inlet is a piece of
hardware attached to the column head.
Common inlet types are:
 S/SL (split/splitless) injector; a sample is introduced into a heated small chamber via a syringe through a septum – the heat
facilitates volatilization of the sample and sample matrix. The carrier gas then either sweeps the entirety (splitless mode) or a
portion (split mode) of the sample into the column. In split mode, a part of the sample/carrier gas mixture in the injection
chamber is exhausted through the split vent. Split injection is preferred when working with samples with high analyte
concentrations (>0.1%) whereas splitless injection is best suited for trace analysis with low amounts of analytes (<0.01%). In
splitless mode the split valve opens after a pre-set amount of time to purge heavier elements that would otherwise
contaminate the system. This pre-set (splitless) time should be optimized, the shorter time (e.g., 0.2 min) ensures less tailing
but loss in response, the longer time (2 min) increases tailing but also signal.[10]
 On-column inlet; the sample is here introduced directly into the column in its entirety without heat, or at a temperature below
the boiling point of the solvent. The low temperature condenses the sample into a narrow zone. The column and inlet can then
be heated, releasing the sample into the gas phase. This ensures the lowest possible temperature for chromatography and
keeps samples from decomposing above their boiling point.
 PTV injector; Temperature-programmed sample introduction was first described by Vogt in 1979.[citation needed] Originally Vogt
developed the technique as a method for the introduction of large sample volumes (up to 250 µL) in capillary GC. Vogt
introduced the sample into the liner at a controlled injection rate. The temperature of the liner was chosen slightly below the
boiling point of the solvent. The low-boiling solvent was continuously evaporated and vented through the split line. Based on
this technique, Poy developed the programmed temperature vaporising injector; PTV. By introducing the sample at a low initial
liner temperature many of the disadvantages of the classic hot injection techniques could be circumvented.[citation needed]
 Gas source inlet or gas switching valve; gaseous samples in collection bottles are connected to what is most commonly a six-
port switching valve. The carrier gas flow is not interrupted while a sample can be expanded into a previously evacuated
sample loop. Upon switching, the contents of the sample loop are inserted into the carrier gas stream.
 P/T (Purge-and-Trap) system; An inert gas is bubbled through an aqueous sample causing insoluble volatile chemicals to be
purged from the matrix. The volatiles are 'trapped' on an absorbent column (known as a trap or concentrator) at ambient
temperature. The trap is then heated and the volatiles are directed into the carrier gas stream. Samples requiring
preconcentration or purification can be introduced via such a system, usually hooked up to the S/SL port.
The choice of carrier gas (mobile phase) is important. Hydrogen has a range of flow rates that are comparable to helium in
efficiency. However, helium may be more efficient and provide the best separation if flow rates are optimized. Helium is non-
flammable and works with a greater number of detectors and older instruments. Therefore, helium is the most common carrier gas
used. However, the price of helium has gone up considerably over recent years, causing an increasing number of
chromatographers to switch to hydrogen gas. Historical use, rather than rational consideration, may contribute to the continued
preferential use of helium.
Detectors
Commonly used detectors are the flame ionization detector (FID) and the thermal conductivity detector (TCD). While TCDs are
beneficial in that they are non-destructive, its low detection limit for most analytes inhibits widespread use.[8] FIDs are sensitive
primarily to hydrocarbons, and are more sensitive to them than TCD.[7] FIDs cannot detect water or carbon dioxide which make
them ideal for environmental organic analyte analysis.[8] FID is two to three times more sensitive to analyte detection than TCD.[8]
Thermal conductivity detector (TCD) relies on the thermal conductivity of matter passing around a thin wire of tungsten-rhenium
with a current traveling through it.[7] In this set up helium or nitrogen serve as the carrier gas because of their relatively high thermal
conductivity which keep the filament cool and maintain uniform resistivity and electrical efficiency of the filament.[7][11] When analyte
molecules elute from the column, mixed with carrier gas, the thermal conductivity decreases while there is an increase in filament
temperature and resistivity resulting in fluctuations in voltage ultimately causing a detector response.[7][11] Detector sensitivity is
proportional to filament current while it is inversely proportional to the immediate environmental temperature of that detector as well
as flow rate of the carrier gas.[7]
In a flame ionization detector (FID), electrodes are placed adjacent to a flame fueled by hydrogen / air near the exit of the column,
and when carbon containing compounds exit the column they are pyrolyzed by the flame.[7][11] This detector works only for organic /
hydrocarbon containing compounds due to the ability of the carbons to form cations and electrons upon pyrolysis which generates
a current between the electrodes.[7][11] The increase in current is translated and appears as a peak in a chromatogram. FIDs have
low detection limits (a few picograms per second) but they are unable to generate ions from carbonyl containing carbons.[7] FID
compatible carrier gasses include helium, hydrogen, nitrogen, and argon.[7][11]
Alkali flame detector (AFD) or alkali flame ionization detector (AFID) has high sensitivity to nitrogen and phosphorus, similar to
NPD. However, the alkaline metal ions are supplied with the hydrogen gas, rather than a bead above the flame. For this reason
AFD does not suffer the "fatigue" of the NPD, but provides a constant sensitivity over long period of time. In addition, when alkali
ions are not added to the flame, AFD operates like a standard FID. A catalytic combustion detector (CCD) measures combustible
hydrocarbons and hydrogen. Discharge ionization detector (DID) uses a high-voltage electric discharge to produce ions.
The polyarc reactor is an add-on to new or existing GC-FID instruments that converts all organic compounds to methane molecules
prior to their detection by the FID. This technique can be used to improve the response of the FID and allow for the detection of
many more carbon-containing compounds.[12] The complete conversion of compounds to methane and the now equivalent
response in the detector also eliminates the need for calibrations and standards because response factors are all equivalent to
those of methane. This allows for the rapid analysis of complex mixtures that contain molecules where standards are not available.
Flame photometric detector (FPD) uses a photomultiplier tube to detect spectral lines of the compounds as they are burned in a
flame. Compounds eluting off the column are carried into a hydrogen fueled flame which excites specific elements in the
molecules, and the excited elements (P,S, Halogens, Some Metals) emit light of specific characteristic wavelengths.[11] The emitted
light is filtered and detected by a photomultiplier tube.[7][11] In particular, phosphorus emission is around 510–536 nm and sulfur
emission is at 394 nm.[7][11] With an atomic emission detector (AED), a sample eluting from a column enters a chamber which is
energized by microwaves that induce a plasma.[11] The plasma causes the analyte sample to decompose and certain elements
generate an atomic emission spectra.[11] The atomic emission spectra is diffracted by a diffraction grating and detected by a series
of photomultiplier tubes or photo diodes.[11]
Electron capture detector (ECD) uses a radioactive beta particle (electron) source to measure the degree of electron capture. ECD
are used for the detection of molecules containing electronegative / withdrawing elements and functional groups like halogens,
carbonyl, nitriles, nitro groups, and organometalics.[7][11] In this type of detector either nitrogen or 5% methane in argon is used as
the mobile phase carrier gas.[7][11] The carrier gas passes between two electrodes placed at the end of the column, and adjacent to
the cathode (negative electrode) resides a radioactive foil such as 63Ni.[7][11] The radioactive foil emits a beta particle (electron)
which collides with and ionizes the carrier gas to generate more ions resulting in a current.[7][11] When analyte molecules with
electronegative / withdrawing elements or functional groups electrons are captured which results in a decrease in current
generating a detector response.[7][11]
Nitrogen–phosphorus detector (NPD), a form of thermionic detector where nitrogen and phosphorus alter the work function on a
specially coated bead and a resulting current is measured.
Dry electrolytic conductivity detector (DELCD) uses an air phase and high temperature (v. Coulsen) to measure chlorinated
compounds.
Mass spectrometer (MS), also called GC-MS; highly effective and sensitive, even in a small quantity of sample. This detector can
be used to identify the analytes in chromatograms by their mass spectrum.[13] Some GC-MS are connected to an NMR
spectrometer which acts as a backup detector. This combination is known as GC-MS-NMR.[citation needed] Some GC-MS-NMR are
connected to an infrared spectrophotometer which acts as a backup detector. This combination is known as GC-MS-NMR-IR. It
must, however, be stressed this is very rare as most analyses needed can be concluded via purely GC-MS.[citation needed]
Vacuum ultraviolet (VUV) represents the most recent development in gas chromatography detectors. Most chemical species
absorb and have unique gas phase absorption cross sections in the approximately 120–240 nm VUV wavelength range monitored.
Where absorption cross sections are known for analytes, the VUV detector is capable of absolute determination (without
calibration) of the number of molecules present in the flow cell in the absence of chemical interferences.[14]
Other detectors include the Hall electrolytic conductivity detector (ElCD), helium ionization detector (HID), infrared
detector (IRD), photo-ionization detector (PID), pulsed discharge ionization detector (PDD), and thermionic ionization detector
(TID).[15]
Applications.
In general, substances that vaporize below 300 °C (and therefore are stable up to that temperature) can be measured
quantitatively. The samples are also required to be salt-free; they should not contain ions. Very minute amounts of a substance can
be measured, but it is often required that the sample must be measured in comparison to a sample containing the pure, suspected
substance known as a reference standard.
Various temperature programs can be used to make the readings more meaningful; for example to differentiate between
substances that behave similarly during the GC process.
Professionals working with GC analyze the content of a chemical product, for example in assuring the quality of products in the
chemical industry; or measuring toxic substances in soil, air or water. GC is very accurate if used properly and can
measure picomoles of a substance in a 1 ml liquid sample, or parts-per-billion concentrations in gaseous samples.
In practical courses at colleges, students sometimes get acquainted to the GC by studying the contents of Lavender oil or
measuring the ethylene that is secreted by Nicotiana benthamiana plants after artificially injuring their leaves. These GC analyse
hydrocarbons (C2-C40+). In a typical experiment, a packed column is used to separate the light gases, which are then detected
with a TCD. The hydrocarbons are separated using a capillary column and detected with a FID. A complication with light gas
analyses that include H2 is that He, which is the most common and most sensitive inert carrier (sensitivity is proportional to
molecular mass) has an almost identical thermal conductivity to hydrogen (it is the difference in thermal conductivity between two
separate filaments in a Wheatstone Bridge type arrangement that shows when a component has been eluted). For this reason,
dual TCD instruments used with a separate channel for hydrogen that uses nitrogen as a carrier are common. Argon is often used
when analysing gas phase chemistry reactions such as F-T synthesis so that a single carrier gas can be used rather than two
separate ones. The sensitivity is reduced, but this is a trade off for simplicity in the gas supply.
Gas chromatography is used extensively in forensic science. Disciplines as diverse as solid drug dose (pre-consumption form)
identification and quantification, arson investigation, paint chip analysis, and toxicology cases, employ GC to identify and quantify
various biological specimens and crime-scene evidence.
5. HPLC (high performance liquid chromatography)
High-performance liquid chromatography (HPLC), formerly referred to as high-pressure liquid chromatography, is a
technique in analytical chemistry used to separate, identify, and quantify each component in a mixture. It relies on pumps to pass a
pressurized liquid solvent containing the sample mixture through a column filled with a solid adsorbent material. Each component
in the sample interacts slightly differently with the adsorbent material, causing different flow rates for the different components and
leading to the separation of the components as they flow out of the column.
HPLC has been used for manufacturing (e.g., during the production process of pharmaceutical and biological products), legal (e.g.,
detecting performance enhancement drugs in urine), research (e.g., separating the components of a complex biological sample, or
of similar synthetic chemicals from each other), and medical (e.g., detecting vitamin D levels in blood serum) purposes.
Types
Partition chromatography
Partition chromatography was one of the first kinds of chromatography that chemists developed.[8] The partition coefficient principle
has been applied in paper chromatography, thin layer chromatography, gas phase and liquid–liquid separation applications. The
1952 Nobel Prize in chemistry was earned by Archer John Porter Martin and Richard Laurence Millington Synge for their
development of the technique, which was used for their separation of amino acids.[9] Partition chromatography uses a retained
solvent, on the surface or within the grains or fibers of an "inert" solid supporting matrix as with paper chromatography; or takes
advantage of some coulombic and/or hydrogen donor interaction with the stationary phase. Analyte molecules partition between a
liquid stationary phase and the eluent. Just as in Hydrophilic Interaction Chromatography (HILIC; a sub-technique within HPLC),
this method separates analytes based on differences in their polarity. HILIC most often uses a bonded polar stationary phase and a
mobile phase made primarily of acetonitrile with water as the strong component. Partition HPLC has been used historically on
unbonded silica or alumina supports. Each works effectively for separating analytes by relative polar differences. HILIC bonded
phases have the advantage of separating acidic, basic and neutral solutes in a single chromatographic run.
Normal–phase chromatography
Normal–phase chromatography was one of the first kinds of HPLC that chemists developed. Also known as normal-phase HPLC
(NP-HPLC) this method separates analytes based on their affinity for a polar stationary surface such as silica, hence it is based on
analyte ability to engage in polar interactions (such as hydrogen-bonding or dipole-dipole type of interactions) with the sorbent
surface. NP-HPLC uses a non-polar, non-aqueous mobile phase (e.g., Chloroform), and works effectively for separating analytes
readily soluble in non-polar solvents. The analyte associates with and is retained by the polar stationary phase. Adsorption
strengths increase with increased analyte polarity. The interaction strength depends not only on the functional groups present in
the structure of the analyte molecule, but also on steric factors. The effect of steric hindrance on interaction strength allows this
method to resolve (separate) structural isomers.
Displacement chromatography
The basic principle of displacement chromatography is: A molecule with a high affinity for the chromatography matrix (the
displacer) will compete effectively for binding sites, and thus displace all molecules with lesser affinities.[11] There are distinct
differences between displacement and elution chromatography. In elution mode, substances typically emerge from a column in
narrow, Gaussian peaks. Wide separation of peaks, preferably to baseline, is desired in order to achieve maximum purification.
The speed at which any component of a mixture travels down the column in elution mode depends on many factors. But for two
substances to travel at different speeds, and thereby be resolved, there must be substantial differences in some interaction
between the biomolecules and the chromatography matrix.
Reversed-phase chromatography (RPC)
Reversed phase HPLC (RP-HPLC) has a non-polar stationary phase and an aqueous, moderately polar mobile phase. One
common stationary phase is a silica which has been surface-modified with RMe2SiCl, where R is a straight chain alkyl group such
as C18H37 or C8H17. With such stationary phases, retention time is longer for molecules which are less polar, while polar molecules
elute more readily (early in the analysis). An investigator can increase retention times by adding more water to the mobile phase;
thereby making the affinity of the hydrophobic analyte for the hydrophobic stationary phase stronger relative to the now more
hydrophilic mobile phase. Similarly, an investigator can decrease retention time by adding more organic solvent to the eluent. RP-
HPLC is so commonly used that it is often incorrectly referred to as "HPLC" without further specification. The pharmaceutical
industry regularly employs RP-HPLC to qualify drugs before their release.
Pump pressure
Pumps vary in pressure capacity, but their performance is measured on their ability to yield a consistent and
reproducible volumetric flow rate. Pressure may reach as high as 60 MPa (6000 lbf/in2), or about 600 atmospheres. Modern HPLC
systems have been improved to work at much higher pressures, and therefore are able to use much smaller particle sizes in the
columns (<2 μm). These "ultra high performance liquid chromatography" systems or UHPLCs can work at up to 120 MPa
(17,405 lbf/in2), or about 1200 atmospheres.[18] The term "UPLC"[19] is a trademark of the Waters Corporation, but is sometimes
used to refer to the more general technique of UHPLC.
Detectors
HPLC detectors fall into two main categories: universal or selective. Universal detectors typically measure a bulk property
(e.g., refractive index) by measuring a difference of a physical property between the mobile phase and mobile phase with solute
while selective detectors measure a solute property (e.g., UV-Vis absorbance) by simply responding to the physical or chemical
property of the solute.[20] HPLC most commonly uses a UV-Vis absorbance detector, however, a wide range of
other chromatography detectors can be used. A universal detector that complements UV-Vis absorbance detection is the Charged
aerosol detector (CAD). A kind of commonly utilized detector includes refractive index detectors, which provide readings by
measuring the changes in the refractive index of the eluant as it moves through the flow cell. In certain cases, it is possible to use
multiple detectors, for example LCMS normally combines UV-Vis with a mass spectrometer.
Autosamplers
Large numbers of samples can be automatically injected onto an HPLC system, by the use of HPLC autosamplers. In addition,
HPLC autosamplers have an injection volume and technique which is exactly the same for each injection, consequently they
provide a high degree of injection volume precision. It is possible to enable sample stirring within the sampling-chamber, thus
promoting homogeneity.
Applications
Manufacturing
HPLC has many applications in both laboratory and clinical science. It is a common technique used in pharmaceutical
development, as it is a dependable way to obtain and ensure product purity.[22] While HPLC can produce extremely high quality
(pure) products, it is not always the primary method used in the production of bulk drug materials.[23] According to the European
pharmacopoeia, HPLC is used in only 15.5% of syntheses.[24] However, it plays a role in 44% of syntheses in the United States
pharmacopoeia.[25] This could possibly be due to differences in monetary and time constraints, as HPLC on a large scale can be an
expensive technique. An increase in specificity, precision, and accuracy that occurs with HPLC unfortunately corresponds to an
increase in cost.
Legal
This technique is also used for detection of illicit drugs in urine. The most common method of drug detection is an
immunoassay.[26] This method is much more convenient. However, convenience comes at the cost of specificity and coverage of a
wide range of drugs. As HPLC is a method of determining (and possibly increasing) purity, using HPLC alone in evaluating
concentrations of drugs is somewhat insufficient. With this, HPLC in this context is often performed in conjunction with mass
spectrometry.[27] Using liquid chromatography instead of gas chromatography in conjunction with MS circumvents the necessity for
derivitizing with acetylating or alkylation agents, which can be a burdensome extra step.[28] This technique has been used to detect
a variety of agents like doping agents, drug metabolites, glucuronide conjugates, amphetamines, opioids, cocaine, BZDs,
ketamine, LSD, cannabis, and pesticides.[29][30] Performing HPLC in conjunction with Mass spectrometry reduces the absolute need
for standardizing HPLC experimental runs.
Research
Similar assays can be performed for research purposes, detecting concentrations of potential clinical candidates like anti-fungal
and asthma drugs.[31] This technique is obviously useful in observing multiple species in collected samples, as well, but requires the
use of standard solutions when information about species identity is sought out. It is used as a method to confirm results of
synthesis reactions, as purity is essential in this type of research. However, mass spectrometry is still the more reliable way to
identify species.
Medical
Medical use of HPLC can include drug analysis, but falls more closely under the category of nutrient analysis. While urine is the
most common medium for analyzing drug concentrations, blood serum is the sample collected for most medical analyses with
HPLC.[32] Other methods of detection of molecules that are useful for clinical studies have been tested against HPLC, namely
immunoassays. In one example of this, competitive protein binding assays (CPBA) and HPLC were compared for sensitivity in
detection of vitamin D. Useful for diagnosing vitamin D deficiencies in children, it was found that sensitivity and specificity of this
CPBA reached only 40% and 60%, respectively, of the capacity of HPLC.[33] While an expensive tool, the accuracy of HPLC is
nearly unparalleled.
6. Affinity chromatography
Affinity chromatography is a method of separating a biomolecule from a mixture, based on a highly
specific macromolecular binding interaction between the biomolecule and another substance. The specific type of
binding interaction depends on the biomolecule of
interest; antigen and antibody, enzyme and substrate, receptor and ligand, or protein and nucleic acid[1] binding
interactions are frequently exploited for isolation of various biomolecules. Affinity chromatography is useful for its
high selectivity and resolution of separation,[2][3] compared to other chromatographic methods.
Principle[edit]
Affinity chromatography takes advantage of specific binding interactions between the analyte of interest (normally dissolved in
the mobile phase), and a binding partner or ligand (immobilized on the stationary phase). In a typical affinity chromatography
experiment, the ligand is attached to a solid, insoluble matrix--usually a polymer such as agarose or polyacrylamide--chemically
modified to introduce reactive functional groups with which the ligand can react, forming stable covalent bonds.[4] The stationary
phase is first loaded into a column to which the mobile phase is introduced. Molecules that bind to the ligand will remain associated
with the stationary phase. A wash buffer is then applied to remove non-target biomolecules by disrupting their weaker interactions
with the stationary phase, while the biomolecules of interest will remain bound. Target biomolecules may then be removed by
applying a so-called elution buffer, which disrupts interactions between the bound target biomolecules and the ligand. The target
molecule is thus recovered in the eluting solution.
Components of AC.
1. Matrix
 The matrix is an inert support to which a ligand can be directly or indirectly coupled.
 In order to for the matrix to be effective it must have certain characters:
 Matrix should be chemically and physically inert.
 It must be insoluble in solvents and buffers employed in the process
 It must be chemically and mechanically stable.
 It must be easily coupled to a ligand or spacer arm onto which the ligand can be attached.
 It must exhibit good flow properties and have a relatively large surface area for attachment.
 The most useful matrix materials are agarose and polyacrylamide.
2. Spacer arm
 It is used to improve binding between ligand and target molecule by overcoming any effects
of steric hindrance.
3. Ligand
 It refers to the molecule that binds reversibly to a specific target molecule.
 The ligand can be selected only after the nature of the macromolecule to be isolated is
known.
 When a hormone receptor protein is to be purified by affinity chromatography, the hormone
itself is an ideal candidate for the ligand.
 For antibody isolation, an antigen or hapten may be used as ligand.
 If an enzyme is to be purified,a substrate analog, inhibitor, cofactor, or effector may be used
as a the immobilized ligand.
Steps in AC.
 Affinity medium is equilibrated in binding buffer.
 Sample is applied under conditions that favor specific binding of the target molecule(s) to a
complementary binding substance (the ligand). Target substances bind specifically, but
reversibly, to the ligand and unbound material washes through the column.
 Elution is performed specifically, using a competitive ligand, or non-specifically, by changing
the pH, ionic strength or polarity. Target protein is collected in a purified, concentrated form.
 Affinity medium is re-equilibrated with binding buffer.
These events can be summarized into the following three major steps:
1. Preparation of Column
 The column is loaded with solid support such as sepharose, agarose, cellulose etc.
 Ligand is selected according to the desired isolate.
 Spacer arm is attached between the ligand and solid support.
2. Loading of Sample
 Solution containing a mixture of substances is poured into the elution column and allowed to
run at a controlled rate.
3. Elution of Ligand-Molecule Complex
 Target substance is recovered by changing conditions to favor elution of the bound
molecules.
Applications of Affinity Chromatography
 Affinity chromatography is one of the most useful methods for the separation and purification
of specific products.
 It is essentially a sample purification technique, used primarily for biological molecules such
as proteins.
Its major application includes:
 Separation of mixture of compounds.
 Removal of impurities or in purification process.
 In enzyme assays
 Detection of substrates
 Investigation of binding sites of enzymes
 In in vitro antigen-antibody reactions
 Detection of Single Nuceotide polymorphisms and mutations in nucleic acids
Advantages of Affinity Chromatography
 High specificity
 Target molecules can be obtained in a highly pure state
 Single step purification
 The matrix can be reused rapidly.
 The matrix is a solid, can be easily washed and dried.
 Give purified product with high yield.
 Affinity chromatography can also be used to remove specific contaminants, such as
proteases.
Limitations of Affinity Chromatography
 Time consuming method.
 More amounts of solvents are required which may be expensive.
 Intense labour
 Non-specific adsorption cannot be totally eliminated, it can only be minimized.
 Limited availability and high cost of immobilized ligands.
 Proteins get denatured if required pH is not adjusted.
_______________________________The end___________________________________________

More Related Content

What's hot

Penetration Enhancers in Transdermal Drug Delivery System
Penetration Enhancers in Transdermal Drug Delivery SystemPenetration Enhancers in Transdermal Drug Delivery System
Penetration Enhancers in Transdermal Drug Delivery SystemSimranDhiman12
 
Compendial methods of dissolution
Compendial methods of dissolutionCompendial methods of dissolution
Compendial methods of dissolutionHemanth KG
 
Formulation and evaluation of tdds
Formulation and evaluation of tddsFormulation and evaluation of tdds
Formulation and evaluation of tddsPankaj Verma
 
Preparation methods of polymeric nanoparticles
Preparation methods of polymeric nanoparticlesPreparation methods of polymeric nanoparticles
Preparation methods of polymeric nanoparticlesAbeer Abd Elrahman
 
1 gastroretentive drug delivery systems
1 gastroretentive drug delivery systems1 gastroretentive drug delivery systems
1 gastroretentive drug delivery systemsAkash Aher
 
Protein and peptide delivery system
Protein and peptide delivery systemProtein and peptide delivery system
Protein and peptide delivery systemYamini Shah
 
thin layer chromatography
thin layer chromatographythin layer chromatography
thin layer chromatographykatta amulya
 
Sunscreen class and regulatory aspects
Sunscreen class and regulatory aspectsSunscreen class and regulatory aspects
Sunscreen class and regulatory aspectsSagugowda
 
Controlled drug delivery systems
Controlled drug delivery systemsControlled drug delivery systems
Controlled drug delivery systemsTheabhi.in
 
Gas chromatography
Gas chromatographyGas chromatography
Gas chromatographyDhruvi50
 
Affinity chromatography
Affinity chromatography Affinity chromatography
Affinity chromatography Mr. MOHD FAHAD
 
LIPOSOMES USED AS AN DRUG DELIVERY SYSTEMS
LIPOSOMES USED AS AN DRUG DELIVERY SYSTEMSLIPOSOMES USED AS AN DRUG DELIVERY SYSTEMS
LIPOSOMES USED AS AN DRUG DELIVERY SYSTEMSROHIT
 
Tumor targeting 2nd presentation
Tumor targeting 2nd presentationTumor targeting 2nd presentation
Tumor targeting 2nd presentationSayeda Salma S.A.
 
SOLID LIPID NANOPARTICLE ppt
SOLID LIPID NANOPARTICLE ppt SOLID LIPID NANOPARTICLE ppt
SOLID LIPID NANOPARTICLE ppt Anwesha Dandapath
 

What's hot (20)

Liposomes
LiposomesLiposomes
Liposomes
 
Nasopulmonary Drug Delivery System
Nasopulmonary Drug Delivery SystemNasopulmonary Drug Delivery System
Nasopulmonary Drug Delivery System
 
Penetration Enhancers in Transdermal Drug Delivery System
Penetration Enhancers in Transdermal Drug Delivery SystemPenetration Enhancers in Transdermal Drug Delivery System
Penetration Enhancers in Transdermal Drug Delivery System
 
Compendial methods of dissolution
Compendial methods of dissolutionCompendial methods of dissolution
Compendial methods of dissolution
 
Column Chromatography
Column ChromatographyColumn Chromatography
Column Chromatography
 
Formulation and evaluation of tdds
Formulation and evaluation of tddsFormulation and evaluation of tdds
Formulation and evaluation of tdds
 
Preparation methods of polymeric nanoparticles
Preparation methods of polymeric nanoparticlesPreparation methods of polymeric nanoparticles
Preparation methods of polymeric nanoparticles
 
1 gastroretentive drug delivery systems
1 gastroretentive drug delivery systems1 gastroretentive drug delivery systems
1 gastroretentive drug delivery systems
 
Protein and peptide delivery system
Protein and peptide delivery systemProtein and peptide delivery system
Protein and peptide delivery system
 
thin layer chromatography
thin layer chromatographythin layer chromatography
thin layer chromatography
 
Sunscreen class and regulatory aspects
Sunscreen class and regulatory aspectsSunscreen class and regulatory aspects
Sunscreen class and regulatory aspects
 
Controlled drug delivery systems
Controlled drug delivery systemsControlled drug delivery systems
Controlled drug delivery systems
 
Gas chromatography
Gas chromatographyGas chromatography
Gas chromatography
 
Contraversial ingredientts
Contraversial ingredienttsContraversial ingredientts
Contraversial ingredientts
 
Microspheres
MicrospheresMicrospheres
Microspheres
 
Affinity chromatography
Affinity chromatography Affinity chromatography
Affinity chromatography
 
LIPOSOMES USED AS AN DRUG DELIVERY SYSTEMS
LIPOSOMES USED AS AN DRUG DELIVERY SYSTEMSLIPOSOMES USED AS AN DRUG DELIVERY SYSTEMS
LIPOSOMES USED AS AN DRUG DELIVERY SYSTEMS
 
Tumor targeting 2nd presentation
Tumor targeting 2nd presentationTumor targeting 2nd presentation
Tumor targeting 2nd presentation
 
SOLID LIPID NANOPARTICLE ppt
SOLID LIPID NANOPARTICLE ppt SOLID LIPID NANOPARTICLE ppt
SOLID LIPID NANOPARTICLE ppt
 
Targeted drug delivery system
Targeted drug delivery systemTargeted drug delivery system
Targeted drug delivery system
 

Similar to CHROMATOGRAPHY AND ITS TYPES

Paper chromatography
Paper chromatographyPaper chromatography
Paper chromatographySadia RaNa
 
Chromatography (Physical Chemistry Report)
Chromatography (Physical Chemistry Report)Chromatography (Physical Chemistry Report)
Chromatography (Physical Chemistry Report)Muhammad Mamdouh
 
Chromatography ankit
Chromatography ankitChromatography ankit
Chromatography ankitAmanRathore54
 
Chromatography by narayan sarkar and simi baruah
Chromatography by  narayan sarkar and simi baruahChromatography by  narayan sarkar and simi baruah
Chromatography by narayan sarkar and simi baruahNarayanSarkar6
 
CHROMATOGRAPHY - PRINCIPLE,APPLICATIONS.
CHROMATOGRAPHY - PRINCIPLE,APPLICATIONS.CHROMATOGRAPHY - PRINCIPLE,APPLICATIONS.
CHROMATOGRAPHY - PRINCIPLE,APPLICATIONS.PiyashiDas
 
Paper chromatography.pdf
Paper chromatography.pdfPaper chromatography.pdf
Paper chromatography.pdfDeepaksahu1000
 
Method Validation Is Used To Analyze The Specific And...
Method Validation Is Used To Analyze The Specific And...Method Validation Is Used To Analyze The Specific And...
Method Validation Is Used To Analyze The Specific And...Laura Benitez
 
Thin layer chromatography Assignment
Thin layer chromatography AssignmentThin layer chromatography Assignment
Thin layer chromatography Assignmentkannan M
 
Chromatography- Paper chromatography pdf
Chromatography- Paper chromatography pdfChromatography- Paper chromatography pdf
Chromatography- Paper chromatography pdfDubey Ankit
 
Types of Chromatography(Stationary Phase).pptx
Types of  Chromatography(Stationary Phase).pptxTypes of  Chromatography(Stationary Phase).pptx
Types of Chromatography(Stationary Phase).pptxPriyaDixit46
 

Similar to CHROMATOGRAPHY AND ITS TYPES (20)

Paper chromatography by ck
Paper chromatography by ckPaper chromatography by ck
Paper chromatography by ck
 
Paper chromatography
Paper chromatographyPaper chromatography
Paper chromatography
 
Chromatography
ChromatographyChromatography
Chromatography
 
Paper chromatography
Paper chromatographyPaper chromatography
Paper chromatography
 
Paper chromatography
Paper chromatographyPaper chromatography
Paper chromatography
 
Paper chromatography
Paper chromatographyPaper chromatography
Paper chromatography
 
Chromatography (Physical Chemistry Report)
Chromatography (Physical Chemistry Report)Chromatography (Physical Chemistry Report)
Chromatography (Physical Chemistry Report)
 
Chromatography
ChromatographyChromatography
Chromatography
 
CHROMATOGRAPHY.pdf
CHROMATOGRAPHY.pdfCHROMATOGRAPHY.pdf
CHROMATOGRAPHY.pdf
 
Chromatography ankit
Chromatography ankitChromatography ankit
Chromatography ankit
 
Chromatography by narayan sarkar and simi baruah
Chromatography by  narayan sarkar and simi baruahChromatography by  narayan sarkar and simi baruah
Chromatography by narayan sarkar and simi baruah
 
CHROMATOGRAPHY - PRINCIPLE,APPLICATIONS.
CHROMATOGRAPHY - PRINCIPLE,APPLICATIONS.CHROMATOGRAPHY - PRINCIPLE,APPLICATIONS.
CHROMATOGRAPHY - PRINCIPLE,APPLICATIONS.
 
Paper chromatography.pdf
Paper chromatography.pdfPaper chromatography.pdf
Paper chromatography.pdf
 
paper chromatography
paper chromatographypaper chromatography
paper chromatography
 
Chromatography
ChromatographyChromatography
Chromatography
 
Method Validation Is Used To Analyze The Specific And...
Method Validation Is Used To Analyze The Specific And...Method Validation Is Used To Analyze The Specific And...
Method Validation Is Used To Analyze The Specific And...
 
Thin layer chromatography Assignment
Thin layer chromatography AssignmentThin layer chromatography Assignment
Thin layer chromatography Assignment
 
TLC.pptx
TLC.pptxTLC.pptx
TLC.pptx
 
Chromatography- Paper chromatography pdf
Chromatography- Paper chromatography pdfChromatography- Paper chromatography pdf
Chromatography- Paper chromatography pdf
 
Types of Chromatography(Stationary Phase).pptx
Types of  Chromatography(Stationary Phase).pptxTypes of  Chromatography(Stationary Phase).pptx
Types of Chromatography(Stationary Phase).pptx
 

More from JayeshRajput7

STUDY OF CONSOLIDATION PARAMETERS
STUDY OF CONSOLIDATION PARAMETERSSTUDY OF CONSOLIDATION PARAMETERS
STUDY OF CONSOLIDATION PARAMETERSJayeshRajput7
 
COMPRESSION AND COMPACTION
COMPRESSION AND COMPACTION COMPRESSION AND COMPACTION
COMPRESSION AND COMPACTION JayeshRajput7
 
PREFORMULATION CONCEPTS AND OPTIMIZATION IN PHARMACEUTICAL FORMULATION
PREFORMULATION CONCEPTS AND OPTIMIZATION IN PHARMACEUTICAL FORMULATIONPREFORMULATION CONCEPTS AND OPTIMIZATION IN PHARMACEUTICAL FORMULATION
PREFORMULATION CONCEPTS AND OPTIMIZATION IN PHARMACEUTICAL FORMULATIONJayeshRajput7
 
cGMP AND INDUSTRIAL MANAGEMENT
cGMP AND INDUSTRIAL MANAGEMENTcGMP AND INDUSTRIAL MANAGEMENT
cGMP AND INDUSTRIAL MANAGEMENTJayeshRajput7
 
PHARMACEUTICAL VALIDATION
PHARMACEUTICAL VALIDATIONPHARMACEUTICAL VALIDATION
PHARMACEUTICAL VALIDATIONJayeshRajput7
 
SUSTAINED RELEASE (SR) AND CONTROLLED RELEASE (CR) DRUG DELIVERY SYSTEMS.
SUSTAINED RELEASE (SR) AND CONTROLLED RELEASE (CR) DRUG DELIVERY SYSTEMS.SUSTAINED RELEASE (SR) AND CONTROLLED RELEASE (CR) DRUG DELIVERY SYSTEMS.
SUSTAINED RELEASE (SR) AND CONTROLLED RELEASE (CR) DRUG DELIVERY SYSTEMS.JayeshRajput7
 
ELECTROPHORESIS AND ITS TYPES
ELECTROPHORESIS AND ITS TYPESELECTROPHORESIS AND ITS TYPES
ELECTROPHORESIS AND ITS TYPESJayeshRajput7
 
GASTRO RETENTIVE DRUG DELIVERY SYSTEM (GRDDS)
GASTRO RETENTIVE DRUG DELIVERY SYSTEM (GRDDS)GASTRO RETENTIVE DRUG DELIVERY SYSTEM (GRDDS)
GASTRO RETENTIVE DRUG DELIVERY SYSTEM (GRDDS)JayeshRajput7
 
Documentation in pharaceutical industry
Documentation in pharaceutical industryDocumentation in pharaceutical industry
Documentation in pharaceutical industryJayeshRajput7
 
Transdermal drug delivery system
Transdermal drug delivery systemTransdermal drug delivery system
Transdermal drug delivery systemJayeshRajput7
 
ocular drug delivery
ocular drug deliveryocular drug delivery
ocular drug deliveryJayeshRajput7
 
CMC, post approval regulatory affairs, etc
CMC, post approval regulatory affairs, etcCMC, post approval regulatory affairs, etc
CMC, post approval regulatory affairs, etcJayeshRajput7
 
Vaccine delivery systems
Vaccine delivery systemsVaccine delivery systems
Vaccine delivery systemsJayeshRajput7
 
Non-clinical drug development
Non-clinical drug developmentNon-clinical drug development
Non-clinical drug developmentJayeshRajput7
 
immunological assays
immunological assaysimmunological assays
immunological assaysJayeshRajput7
 
Biostatics with reference to bioassay
Biostatics with reference to bioassayBiostatics with reference to bioassay
Biostatics with reference to bioassayJayeshRajput7
 
Radioimmunoassay and related immunoassay techniques
Radioimmunoassay and related immunoassay techniquesRadioimmunoassay and related immunoassay techniques
Radioimmunoassay and related immunoassay techniquesJayeshRajput7
 

More from JayeshRajput7 (20)

POSTER ON COVID-19
POSTER ON COVID-19POSTER ON COVID-19
POSTER ON COVID-19
 
STUDY OF CONSOLIDATION PARAMETERS
STUDY OF CONSOLIDATION PARAMETERSSTUDY OF CONSOLIDATION PARAMETERS
STUDY OF CONSOLIDATION PARAMETERS
 
COMPRESSION AND COMPACTION
COMPRESSION AND COMPACTION COMPRESSION AND COMPACTION
COMPRESSION AND COMPACTION
 
PREFORMULATION CONCEPTS AND OPTIMIZATION IN PHARMACEUTICAL FORMULATION
PREFORMULATION CONCEPTS AND OPTIMIZATION IN PHARMACEUTICAL FORMULATIONPREFORMULATION CONCEPTS AND OPTIMIZATION IN PHARMACEUTICAL FORMULATION
PREFORMULATION CONCEPTS AND OPTIMIZATION IN PHARMACEUTICAL FORMULATION
 
cGMP AND INDUSTRIAL MANAGEMENT
cGMP AND INDUSTRIAL MANAGEMENTcGMP AND INDUSTRIAL MANAGEMENT
cGMP AND INDUSTRIAL MANAGEMENT
 
PHARMACEUTICAL VALIDATION
PHARMACEUTICAL VALIDATIONPHARMACEUTICAL VALIDATION
PHARMACEUTICAL VALIDATION
 
SUSTAINED RELEASE (SR) AND CONTROLLED RELEASE (CR) DRUG DELIVERY SYSTEMS.
SUSTAINED RELEASE (SR) AND CONTROLLED RELEASE (CR) DRUG DELIVERY SYSTEMS.SUSTAINED RELEASE (SR) AND CONTROLLED RELEASE (CR) DRUG DELIVERY SYSTEMS.
SUSTAINED RELEASE (SR) AND CONTROLLED RELEASE (CR) DRUG DELIVERY SYSTEMS.
 
ELECTROPHORESIS AND ITS TYPES
ELECTROPHORESIS AND ITS TYPESELECTROPHORESIS AND ITS TYPES
ELECTROPHORESIS AND ITS TYPES
 
GASTRO RETENTIVE DRUG DELIVERY SYSTEM (GRDDS)
GASTRO RETENTIVE DRUG DELIVERY SYSTEM (GRDDS)GASTRO RETENTIVE DRUG DELIVERY SYSTEM (GRDDS)
GASTRO RETENTIVE DRUG DELIVERY SYSTEM (GRDDS)
 
Documentation in pharaceutical industry
Documentation in pharaceutical industryDocumentation in pharaceutical industry
Documentation in pharaceutical industry
 
Transdermal drug delivery system
Transdermal drug delivery systemTransdermal drug delivery system
Transdermal drug delivery system
 
ocular drug delivery
ocular drug deliveryocular drug delivery
ocular drug delivery
 
CMC, post approval regulatory affairs, etc
CMC, post approval regulatory affairs, etcCMC, post approval regulatory affairs, etc
CMC, post approval regulatory affairs, etc
 
Vaccine delivery systems
Vaccine delivery systemsVaccine delivery systems
Vaccine delivery systems
 
Non-clinical drug development
Non-clinical drug developmentNon-clinical drug development
Non-clinical drug development
 
immunological assays
immunological assaysimmunological assays
immunological assays
 
clinical trials
clinical trialsclinical trials
clinical trials
 
Quality assuarance
Quality assuaranceQuality assuarance
Quality assuarance
 
Biostatics with reference to bioassay
Biostatics with reference to bioassayBiostatics with reference to bioassay
Biostatics with reference to bioassay
 
Radioimmunoassay and related immunoassay techniques
Radioimmunoassay and related immunoassay techniquesRadioimmunoassay and related immunoassay techniques
Radioimmunoassay and related immunoassay techniques
 

Recently uploaded

Unraveling Hypertext_ Analyzing Postmodern Elements in Literature.pptx
Unraveling Hypertext_ Analyzing  Postmodern Elements in  Literature.pptxUnraveling Hypertext_ Analyzing  Postmodern Elements in  Literature.pptx
Unraveling Hypertext_ Analyzing Postmodern Elements in Literature.pptxDhatriParmar
 
4.16.24 Poverty and Precarity--Desmond.pptx
4.16.24 Poverty and Precarity--Desmond.pptx4.16.24 Poverty and Precarity--Desmond.pptx
4.16.24 Poverty and Precarity--Desmond.pptxmary850239
 
4.16.24 21st Century Movements for Black Lives.pptx
4.16.24 21st Century Movements for Black Lives.pptx4.16.24 21st Century Movements for Black Lives.pptx
4.16.24 21st Century Movements for Black Lives.pptxmary850239
 
4.11.24 Poverty and Inequality in America.pptx
4.11.24 Poverty and Inequality in America.pptx4.11.24 Poverty and Inequality in America.pptx
4.11.24 Poverty and Inequality in America.pptxmary850239
 
Congestive Cardiac Failure..presentation
Congestive Cardiac Failure..presentationCongestive Cardiac Failure..presentation
Congestive Cardiac Failure..presentationdeepaannamalai16
 
ICS2208 Lecture6 Notes for SL spaces.pdf
ICS2208 Lecture6 Notes for SL spaces.pdfICS2208 Lecture6 Notes for SL spaces.pdf
ICS2208 Lecture6 Notes for SL spaces.pdfVanessa Camilleri
 
BIOCHEMISTRY-CARBOHYDRATE METABOLISM CHAPTER 2.pptx
BIOCHEMISTRY-CARBOHYDRATE METABOLISM CHAPTER 2.pptxBIOCHEMISTRY-CARBOHYDRATE METABOLISM CHAPTER 2.pptx
BIOCHEMISTRY-CARBOHYDRATE METABOLISM CHAPTER 2.pptxSayali Powar
 
ARTERIAL BLOOD GAS ANALYSIS........pptx
ARTERIAL BLOOD  GAS ANALYSIS........pptxARTERIAL BLOOD  GAS ANALYSIS........pptx
ARTERIAL BLOOD GAS ANALYSIS........pptxAneriPatwari
 
Using Grammatical Signals Suitable to Patterns of Idea Development
Using Grammatical Signals Suitable to Patterns of Idea DevelopmentUsing Grammatical Signals Suitable to Patterns of Idea Development
Using Grammatical Signals Suitable to Patterns of Idea Developmentchesterberbo7
 
Active Learning Strategies (in short ALS).pdf
Active Learning Strategies (in short ALS).pdfActive Learning Strategies (in short ALS).pdf
Active Learning Strategies (in short ALS).pdfPatidar M
 
MS4 level being good citizen -imperative- (1) (1).pdf
MS4 level   being good citizen -imperative- (1) (1).pdfMS4 level   being good citizen -imperative- (1) (1).pdf
MS4 level being good citizen -imperative- (1) (1).pdfMr Bounab Samir
 
Concurrency Control in Database Management system
Concurrency Control in Database Management systemConcurrency Control in Database Management system
Concurrency Control in Database Management systemChristalin Nelson
 
Scientific Writing :Research Discourse
Scientific  Writing :Research  DiscourseScientific  Writing :Research  Discourse
Scientific Writing :Research DiscourseAnita GoswamiGiri
 
Q-Factor HISPOL Quiz-6th April 2024, Quiz Club NITW
Q-Factor HISPOL Quiz-6th April 2024, Quiz Club NITWQ-Factor HISPOL Quiz-6th April 2024, Quiz Club NITW
Q-Factor HISPOL Quiz-6th April 2024, Quiz Club NITWQuiz Club NITW
 
4.11.24 Mass Incarceration and the New Jim Crow.pptx
4.11.24 Mass Incarceration and the New Jim Crow.pptx4.11.24 Mass Incarceration and the New Jim Crow.pptx
4.11.24 Mass Incarceration and the New Jim Crow.pptxmary850239
 
ICS 2208 Lecture Slide Notes for Topic 6
ICS 2208 Lecture Slide Notes for Topic 6ICS 2208 Lecture Slide Notes for Topic 6
ICS 2208 Lecture Slide Notes for Topic 6Vanessa Camilleri
 
Blowin' in the Wind of Caste_ Bob Dylan's Song as a Catalyst for Social Justi...
Blowin' in the Wind of Caste_ Bob Dylan's Song as a Catalyst for Social Justi...Blowin' in the Wind of Caste_ Bob Dylan's Song as a Catalyst for Social Justi...
Blowin' in the Wind of Caste_ Bob Dylan's Song as a Catalyst for Social Justi...DhatriParmar
 
CLASSIFICATION OF ANTI - CANCER DRUGS.pptx
CLASSIFICATION OF ANTI - CANCER DRUGS.pptxCLASSIFICATION OF ANTI - CANCER DRUGS.pptx
CLASSIFICATION OF ANTI - CANCER DRUGS.pptxAnupam32727
 
Beauty Amidst the Bytes_ Unearthing Unexpected Advantages of the Digital Wast...
Beauty Amidst the Bytes_ Unearthing Unexpected Advantages of the Digital Wast...Beauty Amidst the Bytes_ Unearthing Unexpected Advantages of the Digital Wast...
Beauty Amidst the Bytes_ Unearthing Unexpected Advantages of the Digital Wast...DhatriParmar
 

Recently uploaded (20)

Unraveling Hypertext_ Analyzing Postmodern Elements in Literature.pptx
Unraveling Hypertext_ Analyzing  Postmodern Elements in  Literature.pptxUnraveling Hypertext_ Analyzing  Postmodern Elements in  Literature.pptx
Unraveling Hypertext_ Analyzing Postmodern Elements in Literature.pptx
 
Faculty Profile prashantha K EEE dept Sri Sairam college of Engineering
Faculty Profile prashantha K EEE dept Sri Sairam college of EngineeringFaculty Profile prashantha K EEE dept Sri Sairam college of Engineering
Faculty Profile prashantha K EEE dept Sri Sairam college of Engineering
 
4.16.24 Poverty and Precarity--Desmond.pptx
4.16.24 Poverty and Precarity--Desmond.pptx4.16.24 Poverty and Precarity--Desmond.pptx
4.16.24 Poverty and Precarity--Desmond.pptx
 
4.16.24 21st Century Movements for Black Lives.pptx
4.16.24 21st Century Movements for Black Lives.pptx4.16.24 21st Century Movements for Black Lives.pptx
4.16.24 21st Century Movements for Black Lives.pptx
 
4.11.24 Poverty and Inequality in America.pptx
4.11.24 Poverty and Inequality in America.pptx4.11.24 Poverty and Inequality in America.pptx
4.11.24 Poverty and Inequality in America.pptx
 
Congestive Cardiac Failure..presentation
Congestive Cardiac Failure..presentationCongestive Cardiac Failure..presentation
Congestive Cardiac Failure..presentation
 
ICS2208 Lecture6 Notes for SL spaces.pdf
ICS2208 Lecture6 Notes for SL spaces.pdfICS2208 Lecture6 Notes for SL spaces.pdf
ICS2208 Lecture6 Notes for SL spaces.pdf
 
BIOCHEMISTRY-CARBOHYDRATE METABOLISM CHAPTER 2.pptx
BIOCHEMISTRY-CARBOHYDRATE METABOLISM CHAPTER 2.pptxBIOCHEMISTRY-CARBOHYDRATE METABOLISM CHAPTER 2.pptx
BIOCHEMISTRY-CARBOHYDRATE METABOLISM CHAPTER 2.pptx
 
ARTERIAL BLOOD GAS ANALYSIS........pptx
ARTERIAL BLOOD  GAS ANALYSIS........pptxARTERIAL BLOOD  GAS ANALYSIS........pptx
ARTERIAL BLOOD GAS ANALYSIS........pptx
 
Using Grammatical Signals Suitable to Patterns of Idea Development
Using Grammatical Signals Suitable to Patterns of Idea DevelopmentUsing Grammatical Signals Suitable to Patterns of Idea Development
Using Grammatical Signals Suitable to Patterns of Idea Development
 
Active Learning Strategies (in short ALS).pdf
Active Learning Strategies (in short ALS).pdfActive Learning Strategies (in short ALS).pdf
Active Learning Strategies (in short ALS).pdf
 
MS4 level being good citizen -imperative- (1) (1).pdf
MS4 level   being good citizen -imperative- (1) (1).pdfMS4 level   being good citizen -imperative- (1) (1).pdf
MS4 level being good citizen -imperative- (1) (1).pdf
 
Concurrency Control in Database Management system
Concurrency Control in Database Management systemConcurrency Control in Database Management system
Concurrency Control in Database Management system
 
Scientific Writing :Research Discourse
Scientific  Writing :Research  DiscourseScientific  Writing :Research  Discourse
Scientific Writing :Research Discourse
 
Q-Factor HISPOL Quiz-6th April 2024, Quiz Club NITW
Q-Factor HISPOL Quiz-6th April 2024, Quiz Club NITWQ-Factor HISPOL Quiz-6th April 2024, Quiz Club NITW
Q-Factor HISPOL Quiz-6th April 2024, Quiz Club NITW
 
4.11.24 Mass Incarceration and the New Jim Crow.pptx
4.11.24 Mass Incarceration and the New Jim Crow.pptx4.11.24 Mass Incarceration and the New Jim Crow.pptx
4.11.24 Mass Incarceration and the New Jim Crow.pptx
 
ICS 2208 Lecture Slide Notes for Topic 6
ICS 2208 Lecture Slide Notes for Topic 6ICS 2208 Lecture Slide Notes for Topic 6
ICS 2208 Lecture Slide Notes for Topic 6
 
Blowin' in the Wind of Caste_ Bob Dylan's Song as a Catalyst for Social Justi...
Blowin' in the Wind of Caste_ Bob Dylan's Song as a Catalyst for Social Justi...Blowin' in the Wind of Caste_ Bob Dylan's Song as a Catalyst for Social Justi...
Blowin' in the Wind of Caste_ Bob Dylan's Song as a Catalyst for Social Justi...
 
CLASSIFICATION OF ANTI - CANCER DRUGS.pptx
CLASSIFICATION OF ANTI - CANCER DRUGS.pptxCLASSIFICATION OF ANTI - CANCER DRUGS.pptx
CLASSIFICATION OF ANTI - CANCER DRUGS.pptx
 
Beauty Amidst the Bytes_ Unearthing Unexpected Advantages of the Digital Wast...
Beauty Amidst the Bytes_ Unearthing Unexpected Advantages of the Digital Wast...Beauty Amidst the Bytes_ Unearthing Unexpected Advantages of the Digital Wast...
Beauty Amidst the Bytes_ Unearthing Unexpected Advantages of the Digital Wast...
 

CHROMATOGRAPHY AND ITS TYPES

  • 1. Masters of pharmacy, Pharmaceutical technology (Pharmaceutics) Subject- Modern pharmaceutical analytical system (MPH-101T) Lesion no- 4, Chromatography By- Drx JAYESH M.RAJPUT Points:- 1. Paper chromatography: - Paper chromatography is an analytical method used to separate coloured chemicals or substances. A paper chromatography variant, two-dimensional chromatography involves using two solvents and rotating the paper 90° in between. This is useful for separating complex mixtures of compounds having similar polarity, for example, amino acids. The setup has three components. The mobile phase is a solution that travels up the stationary phase, due to capillary action. The mobile phase is generally a mixture of non-polar organic solvent, while the stationary phase is polar inorganic solvent water. Here paper is used to support the stationary phase, water. Polar water molecules are held inside the void space of the cellulose network of the host paper. The difference between TLC and paper chromatography is that the stationary phase in TLC is a layer of adsorbent (usually silica gel, or aluminium oxide), and the stationary phase in paper chromatography is less absorbent paper. Types:- Descending Development of the chromatogram is done by allowing the solvent to travel down the paper. Here, mobile phase is placed in solvent holder at the top. The spot is kept at the top and solvent flows down the paper from above. Ascending Here the solvent travels up the chromatographic paper. Both descending and ascending paper chromatography are used for the separation of organic and inorganic substances. The sample and solvent move upward. Ascending-descending This is the hybrid of both of the above techniques. The upper part of ascending chromatography can be folded over a rod in order to allow the paper to become descending after crossing the rod.
  • 2. Circular chromatography A circular filter paper is taken and the sample is deposited at the center of the paper. After drying the spot, the filter paper is tied horizontally on a Petri dish containing solvent, so that the wick of the paper is dipped in the solvent. The solvent rises through the wick and the components are separated into concentric rings. Two-dimensional In this technique a square or rectangular paper is used. Here the sample is applied to one of the corners and development is performed at a right angle to the direction of the first run. Factors affecting Rf value in paper chromatography  The solvent system.  Composition of the mobile phase.  The working temperature of the system.  The quality of the paper used.  The distance through which the solvent runs.  The quality and nature of solvents used.  The polarity of components.  The pH of the solvent or mobile phase. Typical Uses of Paper Chromatography • Separating Colored Pigments An effective technique used for separating colored pigments from a mixture. How does it work? A few drops of the colored pigments mixture are placed on the filter paper and then it is slowly submerged into a jar of solvent. Depending on their polarity, it dissolves the molecules present in the mixture, as the solvent rises up the paper. As the solvent continues to rise up the filter paper, molecules of each pigment leaves the solution at different places because of different polarity. Hence, every pigment rises up to a particular level on the chromatography paper and gets separated in the process. It is useful in separation of plant pigments. • Reaction Monitoring Over a period of time, the concentration of reactants decreases, whereas the concentration of production increases in a chemical reaction. One can get a fair idea on the progress of reaction by spotting the reactants and developing the chromatogram over different time intervals. The availability of densitometers made quantitative estimations possible, but traditionally the technique was used for qualitative monitoring. However, as a reaction monitoring option, the rapid methods using spectroscopic techniques are limiting the paper chromatography application.
  • 3. • Qualitative Analysis To analyze or separate the different constituents of a mixture, paper chromatography is used. It is one of the methods of qualitative analysis. We can say it as a useful tool for separating polar as well as non polar solutes. To analyze the different compounds in drugs, most of the pharmaceutical companies use this technique. It is used in determining the pollutants in water and testing of antibiotics. • Isolation And Purification For components of mixture, paper chromatography has been put to use as a purification and isolation technique. Using spectro-photometric methods, the separated components on the paper are cut, dissolved in suitable solvents and their absorption is characterized at specific wavelengths. • Pathology And Forensic Science For investigation of crimes, paper chromatography is useful in the field of forensic science, as this process can be successfully carried out with even very small quantities of material. Using this technique, samples from crime scenes are collected to be analyzed and identified. Used in DNA and RNA fingerprinting. Moreover, to detect the presence of alcohol or chemicals in blood, pathological laboratories use paper chromatography. • Foods Both natural and synthetic food colors are added to foods to improve their acceptability and to make them more popular. Paper chromatography has been primarily used for analysis of food colors in ice creams, sweets, drinks and beverages, jams and jellies. To ensure that no non-permitted coloring agents are added to the foods, only edible colors are permitted for use. That's how quantification and identification becomes more important. • Analyzing Complex Mixtures Certain organic compounds such as carbohydrates and amino acids are identified or detected from a complex mixture of organic compounds with the help of paper chromatography. It is useful in the separation of anions and amino acids. Ranging from forensic investigations, pharmaceuticals, environmental monitoring and foods, paper chromatography have retained their ground in various fields. For matching the requirements of high throughput laboratories, chromatography has seen phenomenal growth in terms of software's, applications and increased automation and separation in general. 2. Thin layer chromatography: - Thin-layer chromatography (TLC) is a chromatography technique used to separate non-volatile mixtures.[1] Thin-layer chromatography is performed on a sheet of an inert substrate such as glass, plastic, or aluminium foil, which is coated with a thin layer of adsorbent material, usually silica gel, aluminium oxide (alumina), or cellulose. This layer of adsorbent is known as the stationary phase. After the sample has been applied on the plate, a solvent or solvent mixture (known as the mobile phase) is drawn up the plate via capillary action. Because different analytes ascend the TLC plate at different rates, separation is achieved.[2] The mobile phase has different properties from the stationary phase. For example, with silica gel, a very polar substance, non-polar mobile phases such as heptane are used. The mobile phase may be a mixture, allowing chemists to fine-tune the bulk properties of the mobile phase.
  • 4. Separation process and principle[edit] Different compounds in the sample mixture travel at different rates due to the differences in their attraction to the stationary phase and because of differences in solubility in the solvent.[6] By changing the solvent, or perhaps using a mixture, the separation of components (measured by the Rf value) can be adjusted. Also, the separation achieved with a TLC plate can be used to estimate the separation of a flash chromatography column. (A compound elutes from a column when the amount of solvent collected is equal to 1/Rf.)[7] Chemists often use TLC to develop a protocol for separation by chromatography and use TLC to determine which fractions contain the desired compounds. Separation of compounds is based on the competition of the solute and the mobile phase for binding sites on the stationary phase.[3] For instance, if normal-phase silica gel is used as the stationary phase, it can be considered polar. Given two compounds that differ in polarity, the more polar compound has a stronger interaction with the silica and is, therefore, better able to displace the mobile phase from the available binding sites. As a consequence, the less polar compound moves higher up the plate (resulting in a higher Rf value).[6] If the mobile phase is changed to a more polar solvent or mixture of solvents, it becomes better at binding to the polar plate and therefore displacing solutes from it, so all compounds on the TLC plate will move higher up the plate. It is commonly said that "strong" solvents (eluents) push the analyzed compounds up the plate, whereas "weak" eluents barely move them. The order of strength/weakness depends on the coating (stationary phase) of the TLC plate. For silica gel-coated TLC plates, the eluent strength increases in the following order: perfluoroalkane (weakest), hexane, pentane, carbon tetrachloride, benzene/toluene, dichloromethane, diethyl ether, ethyl acetate, acetonitrile, acetone, 2-propanol/n- butanol, water, methanol, triethylamine, acetic acid, formic acid (strongest). For C18-coated plates the order is reverse. In other words, when the stationary phase is polar and the mobile phase is nonpolar, the method is normal-phase as opposed to reverse- phase. This means that if a mixture of ethyl acetate and hexane as the mobile phase is used, adding more ethyl acetate results in higher Rf values for all compounds on the TLC plate. Changing the polarity of the mobile phase will normally not result in reversed order of running of the compounds on the TLC plate. An eluotropic series can be used as a guide in selecting a mobile phase. If a reversed order of running of the compounds is desired, an apolar stationary phase should be used, such as C18-functionalized silica. Applications Characterization In organic chemistry, reactions are qualitatively monitored with TLC. Spots sampled with a capillary tube are placed on the plate: a spot of starting material, a spot from the reaction mixture, and a cross-spot with both. A small (3 by 7 cm) TLC plate takes a couple of minutes to run. The analysis is qualitative, and it will show if the starting material has disappeared, i.e. the reaction is complete, if any product has appeared, and how many products are generated (although this might be underestimated due to co-elution). Unfortunately, TLCs from low-temperature reactions may give misleading results, because the sample is warmed to room temperature in the capillary, which can alter the reaction—the warmed sample analyzed by TLC is not the same as what is in the low-temperature flask. One such reaction is the DIBALH reduction of ester to aldehyde. In one study TLC has been applied in the screening of organic reactions,[11] for example in the fine-tuning of BINAP synthesis from 2-naphthol. In this method, the alcohol and catalyst solution (for instance iron(III) chloride) are placed separately on the baseline, then reacted, and then instantly analyzed. A special application of TLC is in the characterization of radiolabeled compounds, where it is used to determine radiochemical purity. The TLC sheet is visualized using a sheet of photographic film or an instrument capable of measuring radioactivity. It may be visualized using other means as well. This method is much more sensitive than the others and can be used to detect an extremely small amount of a compound, provided that it carries a radioactive atom. Isolation Since different compounds will travel a different distance in the stationary phase, chromatography can be used to isolate components of a mixture for further analysis. The separated compounds each occupying a specific area on the plate, they can be scraped off (along with the stationary phase particles) and dissolved into an appropriate solvent. As an example, in the chromatography of an extract of green plant material (for example spinach) shown in 7 stages of development, Carotene elutes quickly and is only visible until step 2. Chlorophyll A and B are halfway in the final step and lutein the first compound staining yellow. Once the chromatography is over, the carotene can be removed from the plate, extracted into a solvent and placed into a spectrophotometer to determine its spectrum. The quantities extracted are small and a technique such as column chromatography is preferred to separate larger amounts. However, big preparative TLC plates with thick silica gel coatings can be used to separate more than 100 mg of material. Examining reactions and compound stability TLC is also used for the identification of the completion of any chemical reaction. To determine this it is observed that at the beginning of a reaction the entire spot is occupied by the starting chemicals or materials on the plate. As the reaction starts taking place the spot formed by the initial chemicals starts reducing and eventually replaces the whole spot of starting chemicals with a new product present on the plate. The formation of an entirely new spot determines the completion of a reaction.[12] Furthermore, two-dimensional TLC is frequently used as a method to check if a compound is stable in the stationary phase (such as silica gel, which is usually slightly acidic). For this purpose, the tested compound mixture is eluted twice in a square-shaped TLC plate, first in one direction and then rotated 90º. If the target compound appears on the diagonal of the square, it is stable in silica gel and safe to purify. If it appears below the diagonal, it is decomposing on silica gel. If this is the case, purification can be attempted using neutralized silica gel (with triethylamine, for example), or an alternative stationary phase such as neutral alumina.
  • 5. 3. Column chromatography. Column chromatography in chemistry is a chromatography method used to isolate a single chemical compound from a mixture. Chromatography is able to separate substances based on differential adsorption of compounds to the adsorbent; compounds move through the column at different rates, allowing them to be separated into fractions. The technique is widely applicable, as many different adsorbents (normal phase, reversed phase, or otherwise) can be used with a wide range of solvents. The technique can be used on scales from micrograms up to kilograms. The main advantage of column chromatography is the relatively low cost and disposability of the stationary phase used in the process. The latter prevents cross- contamination and stationary phase degradation due to recycling. Column chromatography can be done using gravity to move the solvent, or using compressed gas to push the solvent through the column. Column preparation A column is prepared by packing a solid absorbent into a cylindrical glass or plastic tube. The size will depend on the amount of compound being isolated. The base of the tube contains a filter, either a cotton or glass wool plug, or glass frit to hold the solid phase in place. A solvent reservoir may be attached at the top of the column. Two methods are generally used to prepare a column: the dry method and the wet method. For the dry method, the column is first filled with dry stationary phase powder, followed by the addition of mobile phase, which is flushed through the column until it is completely wet, and from this point is never allowed to run dry.[1] For the wet method, a slurry is prepared of the eluent with the stationary phase powder and then carefully poured into the column. The top of the silica should be flat, and the top of the silica can be protected by a layer of sand. Eluent is slowly passed through the column to advance the organic material. The individual components are retained by the stationary phase differently and separate from each other while they are running at different speeds through the column with the eluent. At the end of the column they elute one at a time. During the entire chromatography process the eluent is collected in a series of fractions. Fractions can be collected automatically by means of fraction collectors. The productivity of chromatography can be increased by running several columns at a time. In this case multi stream collectors are used. The composition of the eluent flow can be monitored and each fraction is analyzed for dissolved compounds, e.g. by analytical chromatography, UV absorption spectra, or fluorescence. Colored compounds (or fluorescent compounds with the aid of a UV lamp) can be seen through the glass wall as moving bands. Column Chromatography Procedure Before starting with the Column Chromatography Experiment let us understand the different phases involved. Mobile phase – This phase is made up of solvents and it performs the following functions: 1. It acts as a solvent – sample mixture can be introduced in the column. 2. It acts as a developing agent – helps in the separation of components in the sample to form bands. 3. It acts as an eluting agent – the components that are separated during the experiment are removed from the column 4. Some examples of solvents used as mobile phase based on their polarity are – ethanol, acetone, water, acetic acid, pyridine, etc. Types of Column Chromatography: 1. Adsorption column chromatography – Adsorption chromatography is a technique of separation, in which the components of the mixture are adsorbed on the surface of the adsorbent. 2. Partition column chromatography – The stationary phase, as well as mobile phase, are liquid in partition chromatography. 3. Gel column chromatography – In this method of chromatography, the separation takes place through a column packed with gel. The stationary phase is a solvent held in the gap of a solvent. 4. Ion exchange column chromatography – A chromatography technique in which the stationary phase is always ion exchange resin.
  • 6. Column Chromatography Applications  Column Chromatography is used to isolate active ingredients.  It is very helpful in Separating compound mixtures.  It is used to determine drug estimation from drug formulations  It is used to remove impurities.  Used to isolation metabolites from biological fluids. 4. Gas chromatography. Gas chromatography (GC) is a common type of chromatography used in analytical chemistry for separating and analyzing compounds that can be vaporized without decomposition. Typical uses of GC include testing the purity of a particular substance, or separating the different components of a mixture (the relative amounts of such components can also be determined). In some situations, GC may help in identifying a compound. In preparative chromatography, GC can be used to prepare pure compounds from a mixture.[1][2] In gas chromatography, the mobile phase (or "moving phase") is a carrier gas, usually an inert gas such as helium or an unreactive gas such as nitrogen. Helium remains the most commonly used carrier gas in about 90% of instruments although hydrogen is preferred for improved separations.[3] The stationary phase is a microscopic layer of liquid or polymer on an inert solid support, inside a piece of glass or metal tubing called a column (a homage to the fractionating column used in distillation). The instrument used to perform gas chromatography is called a 'gas chromatograph' (or "aerograph", "gas separator"). The gaseous compounds being analyzed interact with the walls of the column, which is coated with a stationary phase. This causes each compound to elute at a different time, known as the 'retention time' of the compound. The comparison of retention times is what gives GC its analytical usefulness. Instrumentation Autosamplers The autosampler provides the means to introduce a sample automatically into the inlets. Manual insertion of the sample is possible but is no longer common. Automatic insertion provides better reproducibility and time-optimization. Different kinds of autosamplers exist. Autosamplers can be classified in relation to sample capacity (auto-injectors vs. autosamplers, where auto-injectors can work a small number of samples), to robotic technologies (XYZ robot[9] vs. rotating robot – the most common), or to analysis:  Liquid  Static head-space by syringe technology  Dynamic head-space by transfer-line technology  Solid phase microextraction (SPME) Inlets (injectors) The column inlet (or injector) provides the means to introduce a sample into a continuous flow of carrier gas. The inlet is a piece of hardware attached to the column head. Common inlet types are:  S/SL (split/splitless) injector; a sample is introduced into a heated small chamber via a syringe through a septum – the heat facilitates volatilization of the sample and sample matrix. The carrier gas then either sweeps the entirety (splitless mode) or a portion (split mode) of the sample into the column. In split mode, a part of the sample/carrier gas mixture in the injection chamber is exhausted through the split vent. Split injection is preferred when working with samples with high analyte concentrations (>0.1%) whereas splitless injection is best suited for trace analysis with low amounts of analytes (<0.01%). In splitless mode the split valve opens after a pre-set amount of time to purge heavier elements that would otherwise
  • 7. contaminate the system. This pre-set (splitless) time should be optimized, the shorter time (e.g., 0.2 min) ensures less tailing but loss in response, the longer time (2 min) increases tailing but also signal.[10]  On-column inlet; the sample is here introduced directly into the column in its entirety without heat, or at a temperature below the boiling point of the solvent. The low temperature condenses the sample into a narrow zone. The column and inlet can then be heated, releasing the sample into the gas phase. This ensures the lowest possible temperature for chromatography and keeps samples from decomposing above their boiling point.  PTV injector; Temperature-programmed sample introduction was first described by Vogt in 1979.[citation needed] Originally Vogt developed the technique as a method for the introduction of large sample volumes (up to 250 µL) in capillary GC. Vogt introduced the sample into the liner at a controlled injection rate. The temperature of the liner was chosen slightly below the boiling point of the solvent. The low-boiling solvent was continuously evaporated and vented through the split line. Based on this technique, Poy developed the programmed temperature vaporising injector; PTV. By introducing the sample at a low initial liner temperature many of the disadvantages of the classic hot injection techniques could be circumvented.[citation needed]  Gas source inlet or gas switching valve; gaseous samples in collection bottles are connected to what is most commonly a six- port switching valve. The carrier gas flow is not interrupted while a sample can be expanded into a previously evacuated sample loop. Upon switching, the contents of the sample loop are inserted into the carrier gas stream.  P/T (Purge-and-Trap) system; An inert gas is bubbled through an aqueous sample causing insoluble volatile chemicals to be purged from the matrix. The volatiles are 'trapped' on an absorbent column (known as a trap or concentrator) at ambient temperature. The trap is then heated and the volatiles are directed into the carrier gas stream. Samples requiring preconcentration or purification can be introduced via such a system, usually hooked up to the S/SL port. The choice of carrier gas (mobile phase) is important. Hydrogen has a range of flow rates that are comparable to helium in efficiency. However, helium may be more efficient and provide the best separation if flow rates are optimized. Helium is non- flammable and works with a greater number of detectors and older instruments. Therefore, helium is the most common carrier gas used. However, the price of helium has gone up considerably over recent years, causing an increasing number of chromatographers to switch to hydrogen gas. Historical use, rather than rational consideration, may contribute to the continued preferential use of helium. Detectors Commonly used detectors are the flame ionization detector (FID) and the thermal conductivity detector (TCD). While TCDs are beneficial in that they are non-destructive, its low detection limit for most analytes inhibits widespread use.[8] FIDs are sensitive primarily to hydrocarbons, and are more sensitive to them than TCD.[7] FIDs cannot detect water or carbon dioxide which make them ideal for environmental organic analyte analysis.[8] FID is two to three times more sensitive to analyte detection than TCD.[8] Thermal conductivity detector (TCD) relies on the thermal conductivity of matter passing around a thin wire of tungsten-rhenium with a current traveling through it.[7] In this set up helium or nitrogen serve as the carrier gas because of their relatively high thermal conductivity which keep the filament cool and maintain uniform resistivity and electrical efficiency of the filament.[7][11] When analyte molecules elute from the column, mixed with carrier gas, the thermal conductivity decreases while there is an increase in filament temperature and resistivity resulting in fluctuations in voltage ultimately causing a detector response.[7][11] Detector sensitivity is proportional to filament current while it is inversely proportional to the immediate environmental temperature of that detector as well as flow rate of the carrier gas.[7] In a flame ionization detector (FID), electrodes are placed adjacent to a flame fueled by hydrogen / air near the exit of the column, and when carbon containing compounds exit the column they are pyrolyzed by the flame.[7][11] This detector works only for organic / hydrocarbon containing compounds due to the ability of the carbons to form cations and electrons upon pyrolysis which generates a current between the electrodes.[7][11] The increase in current is translated and appears as a peak in a chromatogram. FIDs have low detection limits (a few picograms per second) but they are unable to generate ions from carbonyl containing carbons.[7] FID compatible carrier gasses include helium, hydrogen, nitrogen, and argon.[7][11] Alkali flame detector (AFD) or alkali flame ionization detector (AFID) has high sensitivity to nitrogen and phosphorus, similar to NPD. However, the alkaline metal ions are supplied with the hydrogen gas, rather than a bead above the flame. For this reason AFD does not suffer the "fatigue" of the NPD, but provides a constant sensitivity over long period of time. In addition, when alkali ions are not added to the flame, AFD operates like a standard FID. A catalytic combustion detector (CCD) measures combustible hydrocarbons and hydrogen. Discharge ionization detector (DID) uses a high-voltage electric discharge to produce ions. The polyarc reactor is an add-on to new or existing GC-FID instruments that converts all organic compounds to methane molecules prior to their detection by the FID. This technique can be used to improve the response of the FID and allow for the detection of many more carbon-containing compounds.[12] The complete conversion of compounds to methane and the now equivalent response in the detector also eliminates the need for calibrations and standards because response factors are all equivalent to those of methane. This allows for the rapid analysis of complex mixtures that contain molecules where standards are not available. Flame photometric detector (FPD) uses a photomultiplier tube to detect spectral lines of the compounds as they are burned in a flame. Compounds eluting off the column are carried into a hydrogen fueled flame which excites specific elements in the molecules, and the excited elements (P,S, Halogens, Some Metals) emit light of specific characteristic wavelengths.[11] The emitted light is filtered and detected by a photomultiplier tube.[7][11] In particular, phosphorus emission is around 510–536 nm and sulfur emission is at 394 nm.[7][11] With an atomic emission detector (AED), a sample eluting from a column enters a chamber which is energized by microwaves that induce a plasma.[11] The plasma causes the analyte sample to decompose and certain elements generate an atomic emission spectra.[11] The atomic emission spectra is diffracted by a diffraction grating and detected by a series of photomultiplier tubes or photo diodes.[11] Electron capture detector (ECD) uses a radioactive beta particle (electron) source to measure the degree of electron capture. ECD are used for the detection of molecules containing electronegative / withdrawing elements and functional groups like halogens, carbonyl, nitriles, nitro groups, and organometalics.[7][11] In this type of detector either nitrogen or 5% methane in argon is used as the mobile phase carrier gas.[7][11] The carrier gas passes between two electrodes placed at the end of the column, and adjacent to the cathode (negative electrode) resides a radioactive foil such as 63Ni.[7][11] The radioactive foil emits a beta particle (electron)
  • 8. which collides with and ionizes the carrier gas to generate more ions resulting in a current.[7][11] When analyte molecules with electronegative / withdrawing elements or functional groups electrons are captured which results in a decrease in current generating a detector response.[7][11] Nitrogen–phosphorus detector (NPD), a form of thermionic detector where nitrogen and phosphorus alter the work function on a specially coated bead and a resulting current is measured. Dry electrolytic conductivity detector (DELCD) uses an air phase and high temperature (v. Coulsen) to measure chlorinated compounds. Mass spectrometer (MS), also called GC-MS; highly effective and sensitive, even in a small quantity of sample. This detector can be used to identify the analytes in chromatograms by their mass spectrum.[13] Some GC-MS are connected to an NMR spectrometer which acts as a backup detector. This combination is known as GC-MS-NMR.[citation needed] Some GC-MS-NMR are connected to an infrared spectrophotometer which acts as a backup detector. This combination is known as GC-MS-NMR-IR. It must, however, be stressed this is very rare as most analyses needed can be concluded via purely GC-MS.[citation needed] Vacuum ultraviolet (VUV) represents the most recent development in gas chromatography detectors. Most chemical species absorb and have unique gas phase absorption cross sections in the approximately 120–240 nm VUV wavelength range monitored. Where absorption cross sections are known for analytes, the VUV detector is capable of absolute determination (without calibration) of the number of molecules present in the flow cell in the absence of chemical interferences.[14] Other detectors include the Hall electrolytic conductivity detector (ElCD), helium ionization detector (HID), infrared detector (IRD), photo-ionization detector (PID), pulsed discharge ionization detector (PDD), and thermionic ionization detector (TID).[15] Applications. In general, substances that vaporize below 300 °C (and therefore are stable up to that temperature) can be measured quantitatively. The samples are also required to be salt-free; they should not contain ions. Very minute amounts of a substance can be measured, but it is often required that the sample must be measured in comparison to a sample containing the pure, suspected substance known as a reference standard. Various temperature programs can be used to make the readings more meaningful; for example to differentiate between substances that behave similarly during the GC process. Professionals working with GC analyze the content of a chemical product, for example in assuring the quality of products in the chemical industry; or measuring toxic substances in soil, air or water. GC is very accurate if used properly and can measure picomoles of a substance in a 1 ml liquid sample, or parts-per-billion concentrations in gaseous samples. In practical courses at colleges, students sometimes get acquainted to the GC by studying the contents of Lavender oil or measuring the ethylene that is secreted by Nicotiana benthamiana plants after artificially injuring their leaves. These GC analyse hydrocarbons (C2-C40+). In a typical experiment, a packed column is used to separate the light gases, which are then detected with a TCD. The hydrocarbons are separated using a capillary column and detected with a FID. A complication with light gas analyses that include H2 is that He, which is the most common and most sensitive inert carrier (sensitivity is proportional to molecular mass) has an almost identical thermal conductivity to hydrogen (it is the difference in thermal conductivity between two separate filaments in a Wheatstone Bridge type arrangement that shows when a component has been eluted). For this reason, dual TCD instruments used with a separate channel for hydrogen that uses nitrogen as a carrier are common. Argon is often used when analysing gas phase chemistry reactions such as F-T synthesis so that a single carrier gas can be used rather than two separate ones. The sensitivity is reduced, but this is a trade off for simplicity in the gas supply. Gas chromatography is used extensively in forensic science. Disciplines as diverse as solid drug dose (pre-consumption form) identification and quantification, arson investigation, paint chip analysis, and toxicology cases, employ GC to identify and quantify various biological specimens and crime-scene evidence. 5. HPLC (high performance liquid chromatography) High-performance liquid chromatography (HPLC), formerly referred to as high-pressure liquid chromatography, is a technique in analytical chemistry used to separate, identify, and quantify each component in a mixture. It relies on pumps to pass a pressurized liquid solvent containing the sample mixture through a column filled with a solid adsorbent material. Each component in the sample interacts slightly differently with the adsorbent material, causing different flow rates for the different components and leading to the separation of the components as they flow out of the column. HPLC has been used for manufacturing (e.g., during the production process of pharmaceutical and biological products), legal (e.g., detecting performance enhancement drugs in urine), research (e.g., separating the components of a complex biological sample, or of similar synthetic chemicals from each other), and medical (e.g., detecting vitamin D levels in blood serum) purposes. Types Partition chromatography Partition chromatography was one of the first kinds of chromatography that chemists developed.[8] The partition coefficient principle has been applied in paper chromatography, thin layer chromatography, gas phase and liquid–liquid separation applications. The 1952 Nobel Prize in chemistry was earned by Archer John Porter Martin and Richard Laurence Millington Synge for their
  • 9. development of the technique, which was used for their separation of amino acids.[9] Partition chromatography uses a retained solvent, on the surface or within the grains or fibers of an "inert" solid supporting matrix as with paper chromatography; or takes advantage of some coulombic and/or hydrogen donor interaction with the stationary phase. Analyte molecules partition between a liquid stationary phase and the eluent. Just as in Hydrophilic Interaction Chromatography (HILIC; a sub-technique within HPLC), this method separates analytes based on differences in their polarity. HILIC most often uses a bonded polar stationary phase and a mobile phase made primarily of acetonitrile with water as the strong component. Partition HPLC has been used historically on unbonded silica or alumina supports. Each works effectively for separating analytes by relative polar differences. HILIC bonded phases have the advantage of separating acidic, basic and neutral solutes in a single chromatographic run. Normal–phase chromatography Normal–phase chromatography was one of the first kinds of HPLC that chemists developed. Also known as normal-phase HPLC (NP-HPLC) this method separates analytes based on their affinity for a polar stationary surface such as silica, hence it is based on analyte ability to engage in polar interactions (such as hydrogen-bonding or dipole-dipole type of interactions) with the sorbent surface. NP-HPLC uses a non-polar, non-aqueous mobile phase (e.g., Chloroform), and works effectively for separating analytes readily soluble in non-polar solvents. The analyte associates with and is retained by the polar stationary phase. Adsorption strengths increase with increased analyte polarity. The interaction strength depends not only on the functional groups present in the structure of the analyte molecule, but also on steric factors. The effect of steric hindrance on interaction strength allows this method to resolve (separate) structural isomers. Displacement chromatography The basic principle of displacement chromatography is: A molecule with a high affinity for the chromatography matrix (the displacer) will compete effectively for binding sites, and thus displace all molecules with lesser affinities.[11] There are distinct differences between displacement and elution chromatography. In elution mode, substances typically emerge from a column in narrow, Gaussian peaks. Wide separation of peaks, preferably to baseline, is desired in order to achieve maximum purification. The speed at which any component of a mixture travels down the column in elution mode depends on many factors. But for two substances to travel at different speeds, and thereby be resolved, there must be substantial differences in some interaction between the biomolecules and the chromatography matrix. Reversed-phase chromatography (RPC) Reversed phase HPLC (RP-HPLC) has a non-polar stationary phase and an aqueous, moderately polar mobile phase. One common stationary phase is a silica which has been surface-modified with RMe2SiCl, where R is a straight chain alkyl group such as C18H37 or C8H17. With such stationary phases, retention time is longer for molecules which are less polar, while polar molecules elute more readily (early in the analysis). An investigator can increase retention times by adding more water to the mobile phase; thereby making the affinity of the hydrophobic analyte for the hydrophobic stationary phase stronger relative to the now more hydrophilic mobile phase. Similarly, an investigator can decrease retention time by adding more organic solvent to the eluent. RP- HPLC is so commonly used that it is often incorrectly referred to as "HPLC" without further specification. The pharmaceutical industry regularly employs RP-HPLC to qualify drugs before their release. Pump pressure Pumps vary in pressure capacity, but their performance is measured on their ability to yield a consistent and reproducible volumetric flow rate. Pressure may reach as high as 60 MPa (6000 lbf/in2), or about 600 atmospheres. Modern HPLC systems have been improved to work at much higher pressures, and therefore are able to use much smaller particle sizes in the columns (<2 μm). These "ultra high performance liquid chromatography" systems or UHPLCs can work at up to 120 MPa (17,405 lbf/in2), or about 1200 atmospheres.[18] The term "UPLC"[19] is a trademark of the Waters Corporation, but is sometimes used to refer to the more general technique of UHPLC. Detectors HPLC detectors fall into two main categories: universal or selective. Universal detectors typically measure a bulk property (e.g., refractive index) by measuring a difference of a physical property between the mobile phase and mobile phase with solute while selective detectors measure a solute property (e.g., UV-Vis absorbance) by simply responding to the physical or chemical property of the solute.[20] HPLC most commonly uses a UV-Vis absorbance detector, however, a wide range of other chromatography detectors can be used. A universal detector that complements UV-Vis absorbance detection is the Charged aerosol detector (CAD). A kind of commonly utilized detector includes refractive index detectors, which provide readings by
  • 10. measuring the changes in the refractive index of the eluant as it moves through the flow cell. In certain cases, it is possible to use multiple detectors, for example LCMS normally combines UV-Vis with a mass spectrometer. Autosamplers Large numbers of samples can be automatically injected onto an HPLC system, by the use of HPLC autosamplers. In addition, HPLC autosamplers have an injection volume and technique which is exactly the same for each injection, consequently they provide a high degree of injection volume precision. It is possible to enable sample stirring within the sampling-chamber, thus promoting homogeneity. Applications Manufacturing HPLC has many applications in both laboratory and clinical science. It is a common technique used in pharmaceutical development, as it is a dependable way to obtain and ensure product purity.[22] While HPLC can produce extremely high quality (pure) products, it is not always the primary method used in the production of bulk drug materials.[23] According to the European pharmacopoeia, HPLC is used in only 15.5% of syntheses.[24] However, it plays a role in 44% of syntheses in the United States pharmacopoeia.[25] This could possibly be due to differences in monetary and time constraints, as HPLC on a large scale can be an expensive technique. An increase in specificity, precision, and accuracy that occurs with HPLC unfortunately corresponds to an increase in cost. Legal This technique is also used for detection of illicit drugs in urine. The most common method of drug detection is an immunoassay.[26] This method is much more convenient. However, convenience comes at the cost of specificity and coverage of a wide range of drugs. As HPLC is a method of determining (and possibly increasing) purity, using HPLC alone in evaluating concentrations of drugs is somewhat insufficient. With this, HPLC in this context is often performed in conjunction with mass spectrometry.[27] Using liquid chromatography instead of gas chromatography in conjunction with MS circumvents the necessity for derivitizing with acetylating or alkylation agents, which can be a burdensome extra step.[28] This technique has been used to detect a variety of agents like doping agents, drug metabolites, glucuronide conjugates, amphetamines, opioids, cocaine, BZDs, ketamine, LSD, cannabis, and pesticides.[29][30] Performing HPLC in conjunction with Mass spectrometry reduces the absolute need for standardizing HPLC experimental runs. Research Similar assays can be performed for research purposes, detecting concentrations of potential clinical candidates like anti-fungal and asthma drugs.[31] This technique is obviously useful in observing multiple species in collected samples, as well, but requires the use of standard solutions when information about species identity is sought out. It is used as a method to confirm results of synthesis reactions, as purity is essential in this type of research. However, mass spectrometry is still the more reliable way to identify species. Medical Medical use of HPLC can include drug analysis, but falls more closely under the category of nutrient analysis. While urine is the most common medium for analyzing drug concentrations, blood serum is the sample collected for most medical analyses with HPLC.[32] Other methods of detection of molecules that are useful for clinical studies have been tested against HPLC, namely immunoassays. In one example of this, competitive protein binding assays (CPBA) and HPLC were compared for sensitivity in detection of vitamin D. Useful for diagnosing vitamin D deficiencies in children, it was found that sensitivity and specificity of this CPBA reached only 40% and 60%, respectively, of the capacity of HPLC.[33] While an expensive tool, the accuracy of HPLC is nearly unparalleled. 6. Affinity chromatography Affinity chromatography is a method of separating a biomolecule from a mixture, based on a highly specific macromolecular binding interaction between the biomolecule and another substance. The specific type of binding interaction depends on the biomolecule of interest; antigen and antibody, enzyme and substrate, receptor and ligand, or protein and nucleic acid[1] binding interactions are frequently exploited for isolation of various biomolecules. Affinity chromatography is useful for its high selectivity and resolution of separation,[2][3] compared to other chromatographic methods.
  • 11. Principle[edit] Affinity chromatography takes advantage of specific binding interactions between the analyte of interest (normally dissolved in the mobile phase), and a binding partner or ligand (immobilized on the stationary phase). In a typical affinity chromatography experiment, the ligand is attached to a solid, insoluble matrix--usually a polymer such as agarose or polyacrylamide--chemically modified to introduce reactive functional groups with which the ligand can react, forming stable covalent bonds.[4] The stationary phase is first loaded into a column to which the mobile phase is introduced. Molecules that bind to the ligand will remain associated with the stationary phase. A wash buffer is then applied to remove non-target biomolecules by disrupting their weaker interactions with the stationary phase, while the biomolecules of interest will remain bound. Target biomolecules may then be removed by applying a so-called elution buffer, which disrupts interactions between the bound target biomolecules and the ligand. The target molecule is thus recovered in the eluting solution. Components of AC. 1. Matrix  The matrix is an inert support to which a ligand can be directly or indirectly coupled.  In order to for the matrix to be effective it must have certain characters:  Matrix should be chemically and physically inert.  It must be insoluble in solvents and buffers employed in the process  It must be chemically and mechanically stable.  It must be easily coupled to a ligand or spacer arm onto which the ligand can be attached.  It must exhibit good flow properties and have a relatively large surface area for attachment.  The most useful matrix materials are agarose and polyacrylamide. 2. Spacer arm  It is used to improve binding between ligand and target molecule by overcoming any effects of steric hindrance. 3. Ligand  It refers to the molecule that binds reversibly to a specific target molecule.  The ligand can be selected only after the nature of the macromolecule to be isolated is known.  When a hormone receptor protein is to be purified by affinity chromatography, the hormone itself is an ideal candidate for the ligand.  For antibody isolation, an antigen or hapten may be used as ligand.  If an enzyme is to be purified,a substrate analog, inhibitor, cofactor, or effector may be used as a the immobilized ligand. Steps in AC.  Affinity medium is equilibrated in binding buffer.  Sample is applied under conditions that favor specific binding of the target molecule(s) to a complementary binding substance (the ligand). Target substances bind specifically, but reversibly, to the ligand and unbound material washes through the column.  Elution is performed specifically, using a competitive ligand, or non-specifically, by changing the pH, ionic strength or polarity. Target protein is collected in a purified, concentrated form.  Affinity medium is re-equilibrated with binding buffer. These events can be summarized into the following three major steps: 1. Preparation of Column  The column is loaded with solid support such as sepharose, agarose, cellulose etc.  Ligand is selected according to the desired isolate.  Spacer arm is attached between the ligand and solid support. 2. Loading of Sample  Solution containing a mixture of substances is poured into the elution column and allowed to run at a controlled rate. 3. Elution of Ligand-Molecule Complex  Target substance is recovered by changing conditions to favor elution of the bound molecules. Applications of Affinity Chromatography  Affinity chromatography is one of the most useful methods for the separation and purification of specific products.  It is essentially a sample purification technique, used primarily for biological molecules such as proteins. Its major application includes:
  • 12.  Separation of mixture of compounds.  Removal of impurities or in purification process.  In enzyme assays  Detection of substrates  Investigation of binding sites of enzymes  In in vitro antigen-antibody reactions  Detection of Single Nuceotide polymorphisms and mutations in nucleic acids Advantages of Affinity Chromatography  High specificity  Target molecules can be obtained in a highly pure state  Single step purification  The matrix can be reused rapidly.  The matrix is a solid, can be easily washed and dried.  Give purified product with high yield.  Affinity chromatography can also be used to remove specific contaminants, such as proteases. Limitations of Affinity Chromatography  Time consuming method.  More amounts of solvents are required which may be expensive.  Intense labour  Non-specific adsorption cannot be totally eliminated, it can only be minimized.  Limited availability and high cost of immobilized ligands.  Proteins get denatured if required pH is not adjusted. _______________________________The end___________________________________________