ADMISSION FOR JANUARY 2025 INTAKE IS NOW OPEN APPLY NOW DISTANCE BSC NURSING TIME –TABLES B 2,1 JULY TO DEC. 2024 IS OUT SCAM ALERT BEWARE GIDEON ROBERT UNIVERSITY AND DAARU SALAAM UNIVERSITY SIGN MOU FOR ADVANCED DEGREES GIDEON ROBERT UNIVERSITY'S COURTESY CALL TO THE HIGHER EDUCATION AUTHORITY'S HEADQUARTERS GIDEON ROBERT UNIVERSITY'S NEWLY OPENED MAPANZA CAMPUS GIDEON ROBERT UNIVERSITY RESEARCH ETHICS COMMITTEE CIVIC LEADERS DATES GIDEON ROBERT UNIVERSITY 10th GRADUATION CEREMONY CIEPUK & GIDEON ROBERT UNIVERSITY TRAINING in Zambia & Africa GIDEON ROBERT UNIVERSITY ADVERT 2019 NURSING STUDENTS CAPPING CEREMONY ABRIDGED NURSING INTRODUCED GIDEON ROBERT UNIVERSITY DOCUMETARY GIDEON ROBERT UNIVERSITY 2019 INTAKE DECEMBER 2022 GRADUATION CEREMONY NEW TUTORIALS UPLOADED ON THE BLOG OFFICIAL CONTACT DETAILS FOR GIDEON ROBERT UNIVERSITY | MTN AND ZAMTEL NETWORKS MOBILE APP LAUNCHED | JULY UPDATE | VERSION 4.4.0

PHARMACEUTICAL DRUG ANALYSIS-BACHELOR OF PHARMACY

Blog Details

PHARMACEUTICAL DRUG ANALYSIS-BACHELOR OF PHARMACY

Chapter: Pharmaceutical Drug Analysis: Pharmaceutical Chemicals: Purity and Management

Pharmaceutical Chemicals: Purity and Management

Since the Second World War a rapid development of pharmaceutical chemicals, and ultimately drugs, has made a quantum progress.

PHARMACEUTICAL CHEMICALS : PURITY AND MANAGEMENT

 

INTRODUCTION

 

Since the Second World War a rapid development of pharmaceutical chemicals, and ultimately drugs, has made a quantum progress. Medicinal chemists, pharmacologists, biochemists, analytical chemists and medical professionals have paved the way with their single goal objective to combat the sufferings of human beings. In this integrated effort the role of an analyst vis-a-vis the chemical purity of pharmaceutical substances and drugs made therefrom and finally the dosage forms that are usually available for direct patient’s usage, has become not only extremely crucial but also equally important and vital. As on date product safety has to be an integral part of all product research in pharmaceutical substances. However, the risk-beneft-ratio has got to be pegged to a bare minimum level. Therefore, it has become absolutely necessary to lay emphasis on product. safety research and development which is very crucial in all the developmental stages of a new secondary pharmaceutical product.

 

Inspite of all the qualified successes of synthetic drug research achieved in the last four decades to combat infectious diseases of the more than 80,000 different ailments, unfortunately only about one third can be treated with drugs, most of them only symptomatically. The discovery of better, effective and safer drugs is needed to fight the causes of dreadful diseases like cancer, acquired-immuno-deficiency-syndrome (AIDS), arthritis, cardio-vascular diseases, disorders of the central nervous system (CNS), such as : Alzheimer’s dis-ease and other vital infectious and metabolic diseases like rheumatoid arthritis.

 

In order to meet these challenges one needs to adopt novel approaches in pharmaceutical research. Both molecular biology and genetic engineering will be exploited duly in opening up new routes. Genetic engineering may be explored in the development of new drugs, besides, being used as a research to investigate the molecular causes of severe and dreadful diseases.

 

It is earnestly believed that towards the beginning of the new century (2001 AD), keeping in view the tremendous global technological competition, one is left with no other choice than to internationalize research and development of pharmaceutical drugs to achieve the common objective ‘better drugs for a better world’.

 

It is, however, pertinent to mention here that pharmaceutical chemicals must maintain a very high degree of chemical purity. It is quite obvious that a state of absolute purity may not be achievable, but a sincere effort must be exercised to obtain the maximum freedom from foreign substances. Bearing in mind the exorbitant operational costs to attain the ‘highest standards’ of purity, perhaps some of these processes are not economically viable. Therefore, a compromise has got to be made to strike a balance between the purity of a substance at a reasonably viable cost and at the same time its purity e.g., being fully acceptable for all pharmaceutical usages.

 

In short, a host of impurities in pharmaceutical chemicals do occur that may be partially responsible for toxicity, chemical interference and general instability.

Chapter: Pharmaceutical Drug Analysis: Pharmaceutical Chemicals: Purity and Management

Pharmaceutical Chemicals: Purity

The standards for pharmaceutical chemicals and their respective dosage forms, as laid down in, various Official Compendia fulfil broadly the following three cardinal objectives, namely : (a) Broad-based highest attainable standard, (b) Biological response versus chemical purity, and (c) Offical standards versus manufacturing standards.

PURITY

The standardization of ‘pharmaceutical chemicals’ and the dosage forms prepared therefrom plays a vital role so that the patient gets the ‘drug’ within the permissible limits of potency and tolerance.

 

The standards for pharmaceutical chemicals and their respective dosage forms, as laid down in, various

 

Official Compendia fulfil broadly the following three cardinal objectives, namely :

 

(a) Broad-based highest attainable standard,

 

(b) Biological response versus chemical purity, and

 

(c) Offical standards versus manufacturing standards.

 

1. BROAD-BASED HIGHEST ATTAINABLE STANDARD

 

Keeping in view the various methods of manufacture of a pharmaceutical substance vis-a-vis its standards of purity, types of impurity and changing pattern of stability, a broad-based highest attainable standard is always fixed. A few typical examples are stated below :



 

2. BIOLOGICAL RESPONSE VS. CHEMICAL PURITY

 

Though chemical purity is the topmost priority, yet the biological response of a pharmaceutical substance holds an equal importance. A wide variation of active ingredients ranging between 90% in one sample and 110% (± 10 per cent limit) in another sample could invariably be observed. Therefore, it has become absolutely essential to lay down definite standards so as to ensure that :

 

·              Different laboratories may produce reasonably reproducible products.

 

·              Difference in active ingredients in various lots may be minimised.

 

·              Retention of acceptable level of potency.

 

·              Freedom of toxicity during storage before use.

 

Examples :

 

(i) Substances to be stored in well-closed, light-resistant containers e.g., isoniazid, nalidixic acid, nandrolone phenylpropionate, nitrofurazone.

 

(ii) Substances to be stored under nitrogen in tightly closed, light-resistant containers at a temperature between 2° and 10°C, e.g., nandrolone decanoate, nystatin, methylergometrine maleate, human normal immunoglobulin.

 

(iii) Substances to be stored in tightly-closed, light-resistant containers in a cool place, e.g., nitrofurantoin, pancreatin, oxyphenonium bromide.

 

(iv) Substances to be stored in tightly-closed, light-resistant containers in a cool place; for parenteral administration, the container should be sterile and sealed so as to exclude micro-organisms. e.g., kanamycin sulphate, novobiocin sodium, benzylpenicillin, lincomycin hydrochloride, chloramphenicol.

 

(v) Substances to be stored in well-closed containers, at a temperature not exceeding 30°C, e.g., procaine penicillin, pepsin, menthol, erythromycin.

 

3. OFFICIAL STANDARDS VIS-A-VIS MANUFACTURING STANDARDS

 

The Offical Standards, as stipulated in the pharmacopoeias of various countries, e.g., IP BP, Eur. P., Int. P., USSRP, JP etc., of a pharmaceutical substance take cognizance of the purity, nature, methods and haz-ards of manufacture, precautions of storage and ultimately the conditions under which the product is to be used.

 

It is a well-known fact that a pharmaceutical substance can be prepared by adopting different routes of synthesis based upon the dynamic ongoing research in the field of organic-reaction-mechanisms. Relentless efforts are exerted vigorously by reputed research laboratories across the world to look for shorter routes of synthesis bearing in mind the cost-effectiveness of the final product. For instance : diclofenac sodium (an NSAID) can be manufactured by two methods, one using a bromo compound as a starting material while the other is based on a non-bromo compound. Nevertheless, the latter product is more in demand because it is completely devoid of bromine residues in the final product.

 

During the process of manufacture an unavoidable criterion is the loss of active ingredients. Therefore, all Official Standards for pharmaceutical chemicals and dosage forms should accomodate such losses caused due to loss in manufacture, unavoidable decomposition and storage under normal conditions for a stipulated period.

 

It has become an usual practice to include a ‘definite overage’ in certain dosage forms so as to compensate the noticeable losses caused either due to manufacturing or storage (anticipated decomposition), in order that the finished product may comply with the prescribed offcial standards after the stipulated duration of storage.

 

Official standards with regard to dosage form and packs, preservation and prevention from contamination in a variety of pharmaceutical products, such as eye-drops, multidose injections and antiseptic creams (external application) that may be prone to spoilage with prolonged repetitive usage should be well defined. The official standards, in general, legislate and control the presence of toxic impurities by prescribed ‘limit tests’ and also by more sophisticated analytical techniques using thin-layer chromatography (TLC), high performance thin-layer chromatography (HPTLC), gas-liquid chromatography (GLC) and high-performance liquid chromatography (HPLC).

Chapter: Pharmaceutical Drug Analysis: Pharmaceutical Chemicals: Purity and Management

Pharmaceutical Chemicals: Management

Various Official Compendia of different countries categorically specify descriptive as well as informa-tive details with regard to the pharmaceutical substances and formulated dosage forms produced therefrom.

MANAGEMENT

Various Official Compendia of different countries categorically specify descriptive as well as informa-tive details with regard to the pharmaceutical substances and formulated dosage forms produced therefrom. Hence, all pharmaceutical chemicals and finished products must rigidly conform to the laid-out standards in a particular country and are subjected to various checks at different levels either by Government/State owned drug testing laboratories or by Government/State approved drug testing laboratories.

 

Official Compendia for pharmaceutical substances usually include the following parameters, namely :

 

·              Description of the Drug or Finished Product

 

·              Identification Tests

 

·              Physical Constants

 

·              Assay of Pharmaceutical Substances

 

·              Assay of Principal Active Ingredients in Formulated Dosage Forms

 

·              Limit Test

 

·              Storage Conditions


Chapter: Pharmaceutical Drug Analysis: Pharmaceutical Chemicals: Purity and Management

Description of the Drug or Finished Product - Pharmaceutical Chemicals: Management

The description of a particular drug or finished product may essentially include the following details, namely :

DESCRIPTION OF THE DRUG OR FINISHED PRODUCT

 

The description of a particular drug or finished product may essentially include the following details, namely :

 

·              Brand Name of the Product

 

·              Name of the Active Ingredient

 

·              Strength of Active Igredient in Dosage Form

 

·              Lot/Batch Number

 

·              Date of Manufacture

 

·              Date of Expiry

 

·              Storage Conditions (if any)

 

·              Separate Dosage for Adults and Children

Chapter: Pharmaceutical Drug Analysis: Pharmaceutical Chemicals: Purity and Management

Sampling Procedures and Errors - Pharmaceutical Chemicals: Management

To collect a ‘representative sample’ forms a vital aspect of analytical chemistry, because the samples subjected to analysis are assumed to be perfectly homogeneous and truly representative.

SAMPLING PROCEDURES AND ERRORS

 

To collect a ‘representative sample’ forms a vital aspect of analytical chemistry, because the samples subjected to analysis are assumed to be perfectly homogeneous and truly representative. Thus, sampling may be considered as the most critical aspect of analysis. In other words, the accuracy and significance of measurements may be solely limited by the sampling process. Unless and until the sampling process is performed properly, it may give rise to a possible weak link in the interpretation of the analytical results. For instance, the improper handling of a blood sample both during and after sampling from a patient prior to an operation may not only pose serious complications but also may prove fatal.

 

A definite instruction with regard to the sampling of given materials have been duly put forward by a number of professional societies, namely :

 

·              Association of Official Analytical Chemists (AOAC),

 

·              American Society for Testing Materials (ASTM), and

 

·              American Public Health Association (APHA).

 

However, a good deal of the wisdom of the analyst supported by the application of statisical results and wealth of experience may go a long way in achieving reasonably accurate and reproducible results.

 

1. Sampling Procedures

 

Samples may be categorized broadly into four heads, namely :

 

(a) Gross Sample : A sample that represents the whole lot and may vary from a few grams or less to several pounds based on the nature of the bulk material.

 

(b) Sample : A sufficiently small size of the sample exclusively for the purpose of analysis and derived from the representative gross sample.

 

(c) Analysis Sample : An aliquot or portion of the ‘sample’ being subjected to actual analysis.

 

(d) Grab Sample : A single sample usually taken at random and assumed to be representative. It is considered to be the most unreliable way to sample a material.

 

1.1. For Solids

 

Sampling of solid materials are comparatively more difficult than other materials because of the follow-ing three reasons, namely :

 

(a) Variation in particle size.

 

(b) Inhomogeniety of the material.

 

(c) Variation within the particle.

 

Sampling of solids can be best accomplished by adopting the following procedures :

 

·              To take 1/50 to 1/100th of the total bulk for gross samples.

 

·              To take larger gross samples for products having larger particle size.

 

·              To sample large bodies of solid materials while they are in movement to obtain aliquots representing all portion of the bulk.

 

·              To handle tissue samples, several tiny parts of an organ may be taken and combined together.

 

1.2. For Liquids

 

Sampling of liquids may be carried out by following these procedures :

 

•      Small heterogenous liquid samples are first shaken thoroughly and then followed by immediate sampling.

 

•      Large volumes of liquids are best sampled immediately after a transfer; or if in a pipeline, after passing through a pump where it has undergone the most vigorous mixing.

 

•      Large volumes of stationary liquids are normally sampled with a ‘thief sampler’, i.e., a device for collecting aliquots at different levels.

 

•      Samples are best drawn (with a ‘thief sampler’) at various depths diagonally instead of vertically down so as to have a better cross-section of the bulk liquid.

 

•      Either separate aliquots of liquid may be analyzed individually and the results combined duly, or the various aliquots may be first combined into one gross sample and replicate analysis carried out. However, the latter method is preferred for obvious reasons since the analysis shall have a better hold on the accuracy and precision of the analysis.

 

•      For sampling of biological fluids the ‘time factor’ is of utmost importance and hence, should be performed by qualified pathologists attached to clinical laboratories under adequate supervision. A few specific examples are stated below :

 

(a) A 24 hour urine sample collections are usually more reliable than single specimens.

 

(b) A sample for blood-sugar analysis is more reliable in a fasting patient.

 

(c) A sample of cerebro-spinal-fluid (CSF) from the vertebral column by lumber puncture in patients having suspected pyogenic meningitis.

 

1.3. For Gases

 

A grab-type gas sample is usually satisfactory in certain cases. For example :

 

(a) A breathe sample may be collected by allowing the subject to blow into an evacuated bag. (Persons driving automobile under the ‘influence of alcohol’ on high-ways during festive seasons).

 

(b) Auto exhaust may also be collected in large evacuated plastic bag to monitor the pollution by vehicles run by gasoline/diesel/CNG in cities and metropolis.

 

2. Errors

 

The famous adage—‘to err is human to forgive divine’—literally means that it is natural for people to make mistakes. However, errors in analytical chemistry or more precisely in pharmaceutical drug analysis are normally of three types, namely :

 

(a) Determinate Errors

 

(b) Instrumental Errors

 

(c) Personal Errors

 

These above mentioned errors would be discussed briefly here with specific examples. It is pertinent to mention here that errors outside the range of ‘permissible errors’ in the analyses of pharmaceutical substances may cause serious problems because most of these substances are usually highly toxic, potent and used exten-sively in life-saving processes across the globe.

 

2.1. Determinate Errors

 

Errors caused due to either incorrect adoption of an assay method or an incorrect graduation read out by an analyst are termed as determinate errors. Such errors, in principle may be determined and corrected. In usual practice the determinate errors are subtle in nature and hence, not easily detected.

 

A few typical examples of determinate errors are stated below :

 

(a) Gravimetric Analysis : Where a compound is precipitated from a solution and the analyst believes that the analyte has been removed from the solution completely. Actually a small portion of the substance under investigation shall remain in solution. This sort of error is normally so insignificant that it is often neglected.

 

(b) Incomplete Chemical Reaction : Where a chemical reaction fails to attain the chemical equilibrium, thus virtually invalidating most calculations entirely based on chemical equilibrium characteristics. It may be eliminated by carrying out a detailed study of the reaction kinetics.

 

(c) Colour-change at Endpoint : Where a colour change is employed for an endpoint signal in a volumetric analysis. It may require an excess quantity of reagent to affect the colour change which ultimately shows completion of the chemical reaction between reagent and analyte. Hence, it is absolutely necessary to determine this excess amount of added reagent, otherewise the analytical results may give a positive error. Therefore, in all such analytical procedures a ‘blank titration’ is performed simultaneously to determine how much reagent is required to affect the colour change when no analyte is present.

 

2.2. Instrumnetal Errors

 

The past three decades have witnessed a quantum progress and advancement in the field of analytical chemistry. Nowadays, both microprocessor based and computer-aided analytical instruments have more or less replaced the manually operated ones in any reasonably good analytical laboratory. One of the most prevalent determinate errors is caused by analytical intruments which are found to be ‘out of calibration’. Hence, it is very essential that such instruments need to be calibrated periodically, for instance, a pH meter is calibrated using a buffer solution of known pH, say adjusting the meter to read pH = 7.00 when a buffer of pH 7.00 is measured ; a single-pan electric balance is calibrated by using standard certified weight box; an UV-spectrophotometer is calibrated using standard solutions of known substances.

 

In a similar manner, the calibration of glassware, such as : volumetric flasks, pipettes, burettes, measur-ing cylindres are duly carried out by specific methods recommended by Indian Standards Institution (ISI), British Standards Institution (BSI), National Physical Laboratory (NPL), United States Pharmacopoeia (USP) at specified temperatures.

 

2.3. Personal Errors

 

In addition to errors caused due to improper assay methods or faulty instruments, it may also be due to the analyst. A few typical examples are cited below :

 

(a) Physical Impairment : A person suffering from colour blindness may not be in a position to assess colour-changes precisely ; or if he uses bifocals he may not take the burette readings accurately.

 

(b) Learning-Curve Syndrome : An analyst must practise a new assay method employing ‘known’ samples before making an attempt to tackle an unknown sample, thereby minimising the scope of personal errors.

Chapter: Pharmaceutical Drug Analysis: Pharmaceutical Chemicals: Purity and Management

Bioavailability - Pharmaceutical Chemicals: Management

According to a biopharmaceutic expert, the term bioavailability may be defined as the rate and extent to which the ingredient is absorbed from the drug product into the body or to the site of action.

BIOAVAILABILITY

 

According to a biopharmaceutic expert, the term bioavailability may be defined as the rate and extent to which the ingredient is absorbed from the drug product into the body or to the site of action. It is measured by blood, serum or plasma levels or from urinary excretion data.

 

1. Importance

 

There are three major factors that govern the efficacy of a dosage form, namely :

 

(a) Onset of therapeutic activity.

 

(b) Intensity of the therapeutic effect.

 

(c) Duration of the therapeutic effect.

 

The above three factors are solely responsible for the rate of absorption of the drug, the distribution of the drug throughout the circulatory system and above all the elimination of the active principle from the body.

 

Official quality control methods adopted, e.g., disintegration time and dissolution rate, do not give ample therapeutic equivalence among drug products belonging to the same class. Moreover, even the products of the same manufacturer may have varying degree of bioavailability in different batches. Therefore, it has become quite necessary to introduce comparative bioavailability studies and skillfully designed fool-proof clinical tests of therapeutic equivalence as an effective true remedial measure of the ultimate performance of drug products.

 

In 1968, fifty-one patients suffered from an epidemic of anticonvulsant intoxication in Brisbane. A thorough investigation revealed that the intoxication was caused by altering one of the excipients from calcium phosphate to lactose in the drug product Phenytoin Capsule without adequate pre-testing by the manufacturer.

 

This apparent minor change of excipient was sufficient enough to bring about an appreciable major change in enhancing the bioavailability of the active principles to abnormally high levels in the affected patients.

 

2. Question of Quality

 

It has now been established beyond any reasonable doubt that quality of a drug product cannot simply be ensured by inspection or analysis, but a control system has to be built into, from the very beginning of manufac-ture of a drug. Besides effective quality control measures exercised in every aspects of production including environment, screening of raw materials, process controls, intermediate shelf-life of finished products the most important aspect is to assess the bioavailability of the active principle.

 

Difference in bioavailability, particularly in drugs with low solubilty, as ascertained by blood level attainment studies, appears to be caused by a number of formulation variables, namely : particlesize, crystalline structure, binding or disintegrating agent, excipient etc., on the release pattern of the drug in its dosage from. For example : the rate of dissolution of the drug in tablet or a capsule in the gastrointestinal fluids.

 

3. Clinical Efficacy of Drugs

 

Medical scientists mainly rely on the measurement of bioavailability of a drug as a positive indicator of therapeutic equivalence, because clinical efficacy for orally administered drugs depends on the degree of absorption and the presence of the active ingredient in the blood stream.

 

Technical information based on in vivo standards and specifications are generally incorporated in vari-ous official compendia. Hence, in order to record a legitimate assessment of bioavailability, in vivo test is an absolute necessity and the relative data obtained therefrom should form an integral part of the standard specifi-cations in the offcial standard.

 

4. Adverse Drug Reaction

 

Any dosage-form can produce adverse drug reactions. Hence, a regular feed back of relevant informa-tion on such adverse reactions from the medical practitioners to the appropriate regulatory authorities and the concerned manufacturers would not only help to intensify better safety measures but also widen the scope to improve drug-design by meticulous research scientists all over the world.

 

The following two examples convey the implications of adverse-drug reaction. They are :

 

Example 1 : Aspirin—Increased gastric damage and subsequent bleeding caused by some aspirin fomulations have been specifically attributed to the slowly dissolving aspirin particles in the stomach. However, both effervescent and highly buffered dosage forms (antacid-aspirin-tablet), which help in maintaining the aspirin in solution, have been found to minimise gastro-intestinal toxicity.

 

Example 2 : Chloramphenicol and Tetracycline—Sparingly soluble broad-spectrum antibiotics like chloramphenicol and tetracycline found to damage the gastrointestinal epithelium besides changing the normal micro-flora in the GI-tract that are required for normal good health.

Chapter: Pharmaceutical Drug Analysis: Pharmaceutical Chemicals: Purity and Management

Identification Tests - Pharmaceutical Chemicals: Management

The true identification of a drug may be accomplished in a number of ways, namely : determination of physical constants, chromatographic tests and finally the chemical tests.

IDENTIFICATION TESTS

The true identification of a drug may be accomplished in a number of ways, namely : determination of physical constants, chromatographic tests and finally the chemical tests. The physical constants essentially include the melting point, boiling point, refractive index, weight per millilitre, specific optical rotation, light absorption, viscosity, specific surface area, swelling power, infra-red absorption, and the like. The chromatographic tests include specific spot-tests by thin-layer chromatography (TLC) of pure drug or its presence in a multi-component system. However, the most specific and reliable are the chemical tests which may be categorized separately under tests for inorganic substances and organic substances. The former may be carried out by well defined general quantitative inorganic analysis and the latter by specific reactions of one or more of the functional moieties present in a drug molecule.


Chapter: Pharmaceutical Drug Analysis: Pharmaceutical Chemicals: Purity and Management

Physical Constants - Pharmaceutical Chemicals: Management

These physical constants will be discussed briefly with typical examples as under :

PHYSICAL CONSTANTS

 

A wide range of physical constants, for instance : melting point, boiling point, specific gravity, viscosity, refractive index, solubility, polymorphic forms vis-a-vis particle size, in addition to characteristic absorption features and optical rotation play a vital role in characterization of pharmaceutical chemicals and drug substances. These physical constants will be discussed briefly with typical examples as under :

 

1. Melting Point

 

It is an important criterion to know the purity of a substance ; however, it has a few limitations. The accuracy and precision of melting point is dependent on a number of factors such as—capillary size, sample size, initial temperature of heating-block and the rate of rise of temperature per unit time (minutes). Keeping in view the different manufacturing processes available for a particular drug the melting point has a definite range usually known as the melting range.


Thus the melting range takes care of the variance in manufacture together with the storage variance over a stipulated period of time.

 

2. Boiling Point

 

It is also an important parameter that establishes the purity of a substance. Depending on the various routes of synthesis available for a substance a boiling point range is usually given in different official compendia.


 

3. Refractive Index

 

It is invariably used as a standard for liquids belonging to the category of fixed oils and synthetic chemicals.


 

4. Weight Per Millilitre

 

Weight per millilitre is prevalent in the Pharmacopoeia of India for the control of liquid substances, whereas Relative Density (20°/20°) or Specific Gravity is mostly employed in the European Pharmacopoeia.


 

5. Specific Optical Rotation

 

As pharmacological activity is intimately related to molecular configuration, hence determination of specific rotation of pharmaceutical substances offer a vital means of ensuring their optical purity.


 

6. Light Absorption

 

The measurement of light absorption both in the visible and ultraviolet range is employed as an authentic means of identification of offcial pharmaceutical substances.


 

7. Viscosity

 

Viscosity measurements are employed as a method of identifing different grades of liquids.


 

8. Specific Surface Area

 

The surface area of powders is determined by subsieve-sizer which is designed for measurement of average particle sizes in the range of 0.2 to 50 microns. The relationship between average particle diameter and specific surface area (SSA) is given by the following expression :


where, SSA = Specific surface area in cm2 per g of material

 

d = Average diameter in microns

 

p = True density of material from which the powder was made in g per cm3


 

9. Swelling Power

 

The swelling power of some pharmaceutical products are well defined.

 

Examples :

 

(i) Isphagula Husk : When 1 g, agitated gently and occasionally for four hours in a 25 ml stoppered measuring cylinder filled upto the 20 ml mark with water and allowed to stand for 1 hour, it occupies a volume of not less than 20 ml and sets to a jelly.

 

(ii) Heavy Kaolin : When 2 g is titurated with 2 ml of water the mixture does not flow.

 

10. Infrared Absorption

 

Measurement and subsequent comparison of the infrared spectrum (between 4000-667 cm–1) of compounds with that of an authentic sample has recently become a versatile method for the identification of drugs having widely varying characteristics.

 

Examples : Infrared spectroscopy is employed to compare samples of chloramphenicol palmitate (biologically active form) recovered from chloramphenicol palmitate mixture vis-a-vis an artificially prepared mixture of authentic sample consisting 10 per cent of the ‘inactive polymorph’.

 

Infrared spectra of known and newly reported compounds are provided in the British Pharmacopoeia (1998) and also in ‘Sadtler Standard Spectra’ published by Sadtler Research Laboratories, Philadelphia

 

(USA) is available to check the authenticity of pure drug samples.

 

11. Miscellaneous Characteristics

 

A large number of miscellaneous characteristics are usually included in many official compendia to ascertain the purity, authenticity and identification of drugs—including : sulphated ash, loss on drying, clarity and colour of solution, presence of heavy metals and specific tests.

 

11.1. Sulphated Ash

 

Specifically for the synthetic organic compounds, the Pharmacopoeia prescribes values for sulphated ash. The sulphated ash is determined by a double ignition with concentrated sulphuric acid. Metals thus remain as sulphides that are usually stable to heat. The method is one of some precision, and provides results which are rather more reproducible than those obtained by simple ignition.


 

11.2. Loss on Drying

 

Loss on drying reflects the net weight of a pharmaceutical substance being dried at a specified tempera-ture either at an atmospheric or under reduced pressure for a stipulated duration with a specific quantity of the substance.


 

11.3. Clarity and Colour of Solution

 

When a pharmaceutical substance is made to dissolve at a known concentration in a specified solvent it gives rise to a clear solution that may be either clear or possess a definite colouration.


 

11.4. Heavy Metals

 

Various tests are prescribed in the offcial compendia to control heavy metal e.g., Ag+, Hg2+, Pb2+, Bi2+, Cu2+, As3+, , Sb3+ and Sn4+ contamination in organic pharmaceutical substances. Hence, a stringent limit is recommended for the presence of heavy metals in medicinal compounds.


 

11.5. Specific Tests

 

In fact, certain known impurities are present in a number of pharmaceutical substances. The presence of such impurities may be carried out by performing prescribed specific tests in various official compendia in order to ascertain their presence within the stipulated limits.

 



Reagents :

 

·              Dilute 1 ml N. HCl and 2.0 ml ferric ammonium sulphate soln. (10% w/v in H2O) with suffcient water to produce 100 ml.

 

·              Dissolve 50 mg cadmium acetate in a mixture of 5 ml DW and 1 ml glacial acetic acid and dilute with ethyl methyl ketone to 50 ml. Immediately before use add and dissolve suffcient Ninhydrin to produce a soln. containing 0.2% w/v.

 

·              Dissolve 10.0 g sodium tungstate and 2.5 g sodium molybdate in 80.0 ml DW in a 250 ml flask; add 5.0 ml phosphoric acid (85-90% w/w) and 10.0 ml HCl (= 11.5 N), connect to a reflux condenser and heat for 10 Hrs. Cool, add 15.0 g lithium sulphate, 5.0 ml DW and 1 drop of bromine and allow to stand for 2 Hrs. Remove the excess bromine by boiling the mixture for 15 mts. without the condenser. Cool, filter and dilute with DW to produce 100 ml.

 

Caution :

(i) The prepared soln. should be stored below 4°C, and

 

(ii) The soln. should be used within 4 months after preparation till it retains its original golden yellow colour. It must be rejected if it has a trace of green colour.


Chapter: Pharmaceutical Drug Analysis: Pharmaceutical Chemicals: Purity and Management

Limit Tests Vis-A-Vis Quantitative Determinations

In general, limit tests are quantitative or semi-quantitative tests particularly put forward to identify and control invariably small quantities of impurity that are supposed to be present in a pharmaceutical substance.

LIMIT TESTS VIS-A-VIS QUANTITATIVE DETERMINATIONS

 

In general, limit tests are quantitative or semi-quantitative tests particularly put forward to identify and control invariably small quantities of impurity that are supposed to be present in a pharmaceutical substance. Obviously the amount of any single impurity present in an official substance is usually small, and therefore, the normal visible-reaction-response to any test for that impurity is also quite small. Hence, it is necessary and important to design the individual test in such a manner so as to avoid possible errors in the hands of various analysts. It may be achieved by taking into consideration the following three cardinal factors, namely :

 

(a) Specificity of the Tests : A test employed as a limit test should imply some sort of selective reaction with the trace impurity. It has been observed that a less specific test which limits a number of possible impurities rather instantly has a positive edge over the highly specific tests.

 

Exmaple : Contamination of Pb2+ and other heavy metal impurities in Alum is precipitated by thioacetamide as their respective sulphides at pH 3.5.

 

(b) Sensitivity : The extent of sensitivity stipulated in a limit test varies widely as per the standard laid down by a pharmacopoeia. The sensitivity is governed by a number of variable factors having a common objective to yield reproducible results, for instance :

 

(i) Gravimetric Analysis : The precipitation is guided by the concentration of the solute and of the precipitating reagent, reaction time, reaction temperature and the nature and amount of other substance(s) present in solution.

 

(ii) Colour Tests : The production of visible and distinct colouration may be achieved by ascertain-ing the requisite quantities of reagents and reactants, time period and above all the stability of the colour produced.

 

(c) Personal Errors : In fact, the personal errors must be avoided as far as possible as explained.


Chapter: Pharmaceutical Drug Analysis: Pharmaceutical Chemicals: Purity and Management

Various Types of Tests for Quantitative Determinations - Pharmaceutical Chemicals: Management

A number of such tests shall be discussed here briefly with specific examples wherever possible and necessary :

VARIOUS TYPES OF TESTS FOR QUANTITATIVE DETERMINATIONS

 

In actual practice, it has been observed that different official compendia describe a number of detailed types of tests with a view to obtain a constant and regular check that might be possible to maintain the desired degree of optimum purity both in the pure pharmaceutical substances and the respective dosage-forms made therefrom.

 

A number of such tests shall be discussed here briefly with specific examples wherever possible and necessary :

 

1. Limits of Insoluble Matter

 

The limits of insoluble matter present in pharmaceutical substances and stated in various official com-pendia are given below :



In the same vein, tests for clarity of solution offer another means of limiting insoluble parent drug sub-stances in their correspondingly more highly water-soluble derivatives.


 

2. Limits of Soluble Matter

 

In order to detect the presence of some very specific impurities normally present in the official substances the limits of soluble impurities have been laid down in different pharmacopoeias. Some typical examples are cited below :


 

3. Limits of Moisture, Volatile Matter and Residual Solvents

 

A good number of pharmaceutical substances usually absorb moisture on storage thereby causing deterioration. Such an anomaly can be safely restricted and limited by imposing an essential requirement for the loss in weight (Loss on Drying) when the pharmaceutical chemical is subjected to drying under specified conditions. The quantum of heat that may be applied to the substance varies widely as per the following norms :

 

(a) Nature of the substance

 

(b) Decomposition characterisics of the substance.

 

Various official compendia recommended different temperatures and duration of drying either at atmos-pheric or reduced pressure (vacuum). A few typical examples are stated below :


There are four types of hydrates which may be observed amongst the pharmaceutical chemicals, namely :

 

(a) Inorganic Salt Hydrates e.g., Magnesium Sulphate (MgSO4.7H2O) ; Sodium Sulphate (Na2SO4. 10 H2O).

 

(b) Salts of Inorganic Cations and Organic Acids e.g., Calcium Lactate, Ferrous Gluconate.

 

(c) Organic Hyrates e.g., Caffeine Hydrate, Theophylline Hydrate.

 

(d) Organic Substances e.g., Acacia, Hydroxymethyl Cellulose.

 

These substance either lose all or part of their water of crystallization on drying which sometimes attains a considerable value as could be seen in the following data :


 

3.1. Aquametry

 

It refers to the determination of water content titrimetrically with Karl Fischer Reagent (KFR). This technique has been used exclusively for the determination of water content in a number of pharmaceutical substances listed below :


Since the introduction of Gas-Liquid-Chromatography (GLC) as an essential analytical tool, it has been judiciously exploited as an useful alternative means for not only determining water content in pharmaceutical chemicals but also limiting specific volatile substances present in them. It may be expatiated with the help of the following examples :

 

Examples :

(i)  For Determination of Water Content :

Gonadorelin : (Limit NMT : 7.0 % w/v)

Procedure : Carry out the method for gas chromatography employing the following solutions

Solution (1) : Dilute 50 μ l of anhydrous methanol (internal standard) with sufficient

anhydrous propan-2-ol to produce 100 ml.

Solution (2) : Dissolve 4 mg of the sample in 1 ml of anhydrous propan-2-ol.

Solution (3) : Dissolve 4 mg of the sample in 1 ml of solution (1) above.

Solution (4) : Add 10 μ l of water to 50 μ l of solution (1).

The chromatographic procedure may be carried out by employing :

 

(a) A stainless-steel column (1 m × 2 mm) packed with porous polymer beads e.g., Chromosorb 102 (60 to 80 mesh) and maintained at 120°C.

 

(b)  Helium as the carrier gas.

 

(c) A Thermal Conducting Detector (TCD) maintained at 150°C. From the chromatograms obtained and taking into account any water detectable in solution

 

(1), calculate the percentage w/w of water taking 0.9972 as its weight per ml at 20°C.

 

(iiFor Limiting Specific Volatile Substance :

 

Orciprenaline Sulphate : (Limit of Water and Methanol : 6.0% w/w)

 

Procedure : Perform the method for gas-chromatography using the following three solutions in water containing :

 

Solution (1) : 0.50% v/v of MeOH and 0.50% v/v of EtOH (96% v/v)—as Internal Standard

 

Solution (2) : 10% w/v of the sample

 

Solution (3) : 10% w/v of the sample and 0.50% v/v of the internal standard.

 

The chromatographic procedure may be performed using a glass column (1.5 × 4 mm) packed with porous polymer beads (80 to 100 mesh) e.g., Porapack-Q and maintained at 140°C.

 

Calculate the percentage w/v of methanol taking 0.792 as its weight per ml at 20°C.

 

4. Limits of Non-Volatile Matter

 

Pharmaceutical chemicals belonging to the domain of inorganic as well as organic substances containing readily volatile matter for which the various official compendia prescribe limits of non-volatile matter. It is pertinent to mention here that the Pharmacopoeia usually makes a clear distinction between substances that are readily volatile and substances that are volatile upon strong ignition, for instance :

 

(a) Readily Volatile : e.g., Organic Substances—alcohol (95% v/v), isopropyl alcohol, chloroform, halothane, anaesthetic ether, chlorocresol and trichloroethylene ; and Inorganic substances—ammonia solution, hydrogen peroxide solution, water for injection.

 

(b) Volatile Upon Strong Ignition : e.g., hydrous wool fat (lanolin).

 

5. Limits of Residue on Ignition

 

In fact, the limits of residue on ignition are basically applicable to the following two categories of pharmaceutical substances, namely :

 

(a) Those which are completely volatile when ignited e.g., Hg.

 

(b) Those which undergo total decomposition thereby leaving a residue with a definite composition e.g., calamine—a basic zinc carbonate that gives rise to ZnO as the residue.

 

According to BP, 68.0 to 74.0% when ignited at a temperature not lower than 900°C until, after further ignition, two successive weighings do not differ, by more than 0.2% of the weight of the residue.

 

6. Limits of Loss on Ignition

 

Official compendia include the limits of ‘loss on ignition’ which is generally applied to relatively stable pharmaceutical substances that are likely to contain thermolabile impurities. A few typical examples are stated below :


 

7. Limits on Ash Value

 

The ash values usually represent the inorganic residue present in official herbal drugs and pharmaceuti-cal substances. These values are categorized into four heads, namely :

 

(a) Ash Value (Total Ash),

 

(b) Acid-Insoluble Ash,

 

(c) Sulphated Ash, and

 

(d) Water-Soluble Ash.

 

These values would be explained with the help of some typical examples stated below :

 

7.1. Ash Value (Total Ash)

 

Ash value normally designates the presence of inorganic salts e.g., calcium oxalate found naturally in the drug, as well as inorganic matter derived from external sources. The official ash values are of prime importance in examination of the purity of powdered drugs as enumerated below :

 

(i) To detect and check adulteration with exhausted drugs e.g., ginger.

 

(ii) To detect and check absence of other parts of the plant e.g., cardamom fruit.

 

(iii) To detect and check adulteration with material containing either starch or stone cells that would modify the ash values.

 

(iv) To ensure the absence of an abnormal proportion of extraneous mineral matter incorporated acciden-tally or due to follow up treatment or due to modus operandi at the time of collection e.g., soil, floor sweepings and sand.

 

The most common procedure recommended for crude drugs is described below :

 

Procedure : Incinerate 2 to 3 g of the ground drug in a tared platinum or silica dish at a temperature not exceeding 450°C until free from carbon. Cool and weigh. If a carbon-free ash cannot be obtained in this way, exhaust the charred mass with hot water (DW), collect the residue on an ashless filter paper, incinerate the residue and filter paper, add the filtrate, evaporate to dryness and ignite at a temperature not exceeding 450°C. Calculate the percentage of ash with reference to the air-dried drug.


 

7.2. Acid-Insoluble Ash

 

The method described above for ‘total ash’ present in crude drugs containing calcium oxalate has certain serious anomalies, namely :

 

·              Offers variable results upon ashing based on the conditions of ignition.

 

·              Does not detect soil present in the drug efficaciously.

 

·              The limits of excess of soil in the drug are not quite definite.

 

Hence, the treatment of the ‘total ash’ with acid virtually leaves silica exclusively and thus comparatively forms a better test to detect and limit excess of soil in the drug than does the ash.

 

The common procedure usually adopted for the determination of ‘acid insoluble ash’ is given below :

 

Procedure : Place the ash, as described earlier, in a crucible, add 15 ml DW and l0 ml hydrochloric acid ( ~– 11.5 N), cover with a watch-glass, boil for 10 minutes and allow to cool. Collect the insoluble matter on an ashless filtre paper, wash with hot DW until the filtrate is neutral, dry, ignite to dull redness, allow to cool in a desiccator and weigh. Repeat until the difference between two successive weighings is not more than l mg. Calculate the percentage of acid-insoluble ash with reference to the air-dried drug.

 

A few typical examples are listed below :


 

7.3. Sulphated Ash

 

The estimation of ‘sulphated ash’ is broadly employed in the case of :

 

(a) Unorganized drugs e.g., colophony, podophyllum resin, wool alcohols, wool fat and hydrous wool fat.

 

(b) Pharmaceutical substances contained with inorganic impurities e.g.,

 

Natural Origin : Spray-dried acacia, Frangula Bark, Activated Charcoal

 

Organic Substances : Cephalexin, Lignocaine hydrochloride, Griseofulvin, Diazoxide, Medazapam, Saccharin.

 

Inorganic Substances : Ammonium chloride, Hydroxy urea.

 

The general method for the determination of ‘sulphated ash’ is enumerated below :

 

Procedure : Heat a silica or platimum crucible to redness for 30 minutes, allow to cool in a desiccator and weigh. Place a suitable quantity of the substance being examined, accurately weighed in the crucible, add 2 ml of 1 M sulphuric acid and heat, first on a waterbath and then cautiously over a flame to about 600°C. Continue heating until all black particles have disappeared and then allow to cool. Add a few drops of 1 M sulphuric acid, heat to ignition as before and allow to cool. Add a few drops of a 16% solution of ammonium carbonate, evaporate to dryness and cautiously ignite. Cool, weigh, ignite for 15 minutes and repeat the procedure to constant weight.

 

Following are the examples to depict the ‘sulphated ash’ present in various official pharmaceutical chemicals :


 

7.4. Water-Soluble Ash

 

Water-soluble ash is specifically useful in detecting such samples which have been extracted with water.

 

A detailed procedure as per the official compendium is enumerated below :

 

Procedure : The ash as described earlier, is boiled for 5 minutes with 25 ml DW, collect the insoluble matter in a sintered-glass crucible or on an ashless filter paper, wash with hot DW and ignite for 15 minutes at a temperature not exceeding 450°. Subtract the weight of the residue thus obtained from the weight of the ash.The difference in weight represents the water-soluble ash. Now, calculate the percentage of water-soluble ash with reference to the air-dried drug.

 

A typical example of an official drug is that of ‘Ginger’, the water-soluble ash of which is found to be not more than 6.0%.


Chapter: Pharmaceutical Drug Analysis: Pharmaceutical Chemicals: Purity and Management

Limit Tests for Metallic Impurities

Limit Tests for Metallic Impurities
The official compendia lay a great deal of emphasis on the control of physiologically dangerous, cumulative poisonous and harmful impurities, such as lead, arsenic and iron present in a host of pharmaceutical chemicals.

LIMIT TESTS FOR METALLIC IMPURITIES

 

The official compendia lay a great deal of emphasis on the control of physiologically dangerous, cumulative poisonous and harmful impurities, such as lead, arsenic and iron present in a host of pharmaceutical chemicals. These impurities very often creep into the final product through a number of means stated below, namely :

 

(a) Through atmospheric pollution.

 

(b) Most frequently derived from the raw materials.

 

(c) From materials used in the process of manufacture.

 

(d) Due to solvent action on the metal of the plant in which the substance is prepared.

 

In short, all prescribed tests for impurities in the Pharmacopoeia usually fix certain limits of tolerance. For lead, arsenic and iron general quantitative or limit tests are precisely laid down which, with necessary variations and modification are rigidly applicable to pharmaceutical substances.

 

1. Limit Tests for Lead

 

Theory : The offcial test is based on the conversion of traces of lead salts present in the pharmaceutical substances to lead sulphide, which is obtained in colloidal form by the addition of sodium sulphide in an alkaline medium achieved by a fairly high concentration of ammonium acetate. The reaction may be expressed as follows :

PbCl2 + Na2S  → PbS (Dec) + 2NaCl

 

The brown colour, caused due to colloidal lead sulphide in the test solution is compared with that produced from a known amount of lead.

 

Equipment : Nessler Cylinders (or Nessler Glasses) : According to the British Standard Specification No : 612, 966—a pair of cylinders made of the same glass and having the same diameter with a graduation mark at the same height from the base in both cylinders (Figure 1).


The final comparison is made by viewing down through the solution against a light background.

Materials Required :

(i) Lead Nitrate Stock Solution : Dissolve0.1598 g of lead nitrate in 100 ml DW to which has been  added 1 ml nitric acid, then dilute with water to 1 Litre.

Note : The solution must be prepared and stored in polyethylene or glass containers free from soluble lead salts.

(ii) Standard Lead Solution : On the day of use, dilute 10.0 ml of lead nitrate stock solution with DW to 100.0 ml. Each ml of standard lead solution contains the equivalent of 10 microgrammes of lead. A control comparison solution prepared with 2.0 ml of standard lead solution contains, when compared to a solution representing 1.0 g of the substance being tested, the equivalent of 20 parts per million of lead.

(iii) Standard Solution : Into a 50 ml Nessler Cylinder, pipette 2 ml of standard lead solution and dilute with DW to 25 ml. Adjust with dilute acetic acid Sp. (IP)* or dilute ammonia solution Sp. (IP) to a pH between 3.0 and 4.0, dilute with DW to about 35 ml and mix.

(iv) Test Solution : Into a 50 ml Nessler Cylinder, place 25 ml of the solution prepared for the test as directed in the individual monograh, dissolve and dilute with DW to 25 ml the specified quantity of the substance being tested. Adjust with dilute acetic acid Sp. (IP) or dilute ammonia solution Sp. to a pH between 3.0 and 4.0, dilute with DW to about 35 ml and mix.

 

Procedure : To each of the cylinders containing the standard solution and test solution respectively, add l0 ml of freshly prepared hydrogen sulphide solution, mix, dilute with water (DW) to 50 ml, allow to stand for 5 minutes and view downwards over a white surface, the colour produced in the test solution is not darker than that produced in the standard solution.

 A few typical examples from the official compendium are given below :


 

2. Limit Test for Arsenic

 

Theory : The official process is a development of the Gutzeit Test wherein all arsenic present is duly converted into arsine gas (AsH3) by subjecting it to reduction with zinc and hydrochloric acid. Further, it depends upon the fact that when arsine comes into contact with dry paper permeated with mercuric (Hg2+) chloride it produces a yellow strain, the intensity of which is directly proportional to the quantity of arsenic present. The various chemical reactions involved may be expressed by the following equations :


The details of experimental procedure described in the Pharmacopoeia are actually based upon a paper by Hill and Collins**, but have been adequately modified from time to time in accordance with the accumu-lated and acquired experience. Explicitly, the expressions provided in the Pharmacopoeia for limits of arsenic exclusively refer to parts per million, calculated as As.

Materials Required : Arsenic limit test apparatus; HgCl2—paper : smooth white filter paper (having thickness in mm of 400 paper = weight in g per Sq. M.), soaked in a saturated solution of HgCl2, pressed to get rid of excess of soln. and dried at about 60°C in the dark ; lead acetate solution 10.0% w/v soln. of PbAc2 in CO2– free water ; KI (AsT), 1.0 g ; Zn (AsT) : l0.0 g ; Dilute Arsenic solution (AST); Standard stains, Test Solutions—are prepared according to the Indian Pharmacopoeia 1996.

Arsenic Limit Test Apparatus (Figure 2)


A wide-mouthed glass bottle capable of holding about 120 ml is fitted with a rubber bung through which passes a glass tube. The latter, made from ordinary glass tubing, has a total length of 200 mm and an internal diameter of exactly 6.5 mm (external diameter about 8 mm). It is drawn out at one end to a diameter of about 1 mm and a hole not less than 2 mm in diameter is blown in the side of the tube, near the constricted part. When the bung is inserted in the bottle containing 70 ml of liquid, the constricted end of the tube is kept above the surface of the liquid, and the hole in the side is below the bottom of the bung. The upper end of the tube is cut off square, and is either slightly rounded or ground smooth.

The rubber bungs (about 25 mm × 25 mm), each with a hole bored centrally and through exactly 6.5 mm  in diameter,are fitted with a rubber band or spring clip for holding them tightly in place.

Procedure : The glass tube is lightly packed with cotton wool, previously moistened with lead acetate solution and dried, so that the upper surface of the cotton wool is not less than 25 mm below the top of the tube. The upper end of the tube is then inserted into the narrow end of one of the pair of rubber bungs, to a depth of l0 mm (the tube must have a rounded-off end). A piece of mercuric chloride paper is placed flat on the top of the bung and the other bung placed over it and secured by means of the spring clip in such a manner that the holes of the two bungs meet to form a true tube 6.5 mm diameter interrupted by a diaphragm of mercuric chloride paper.

 

The test solution prepared as specified, is placed in the wide-mouthed bottle, 1 g of KI (AsT) and 10 g of Zn (AsT) are added, and the prepared glass tube is placed quickly in position. The reaction is allowed to proceed for 40 minutes. The yellow stain that is produced on the HgCl2 paper if As is present is compared by daylight with the standard stains obtained by performing in an identical manner with known quantities of dilute arsenic solution (AsT). The comparison of the stains is made immediately at the completion of the test.

 

By matching the intensity and depth of colour with standard stains, the proportion of arsenic in the substance may be estimated. Thus, a stain equivalent to the 1 ml standard stain obtained by performing on l0 g of a substance implies that the proportion of As is 1 part per million.

 

Cautions : 

(i) HgCl2 paper should be protected from sunlight during the test to avoid lighter or no

stain.

 

(ii) The standard and test stains must be compared immediately as they fade out on retaining.

 

(iii) The reaction may be expedited by the application of heat and 40°C is considered to be the most ideal temperature.

 

(iv) The tube should be washed with HCl (AsT), rinsed with DW, and dried between successive tests.

 

Special Techniques : The special techniques are usually applicable to a host of pharmaceutical sub-stances before the normal test can be performed. A few typical examples would be discussed briefly here, namely :

 

(i) Free Acids : They are first converted to their respective sodium salts with Na2CO3 and As3+ oxi-dised to As5+ by evaporating the solution with Br2. The residue is ignited carefully until carbonised to destroy organic matter, while As is kept as non-volatile sodium arsenate. The resulting residue is dissolved in brominated HCl and the test carried out in the normal manner.

 

Examples : Aspirin, Saccharin, Sodium Salicylate, Sodium Aminosalicylate.

 

(ii) Substances Reacting Vigorously with HCl : The As is readily converted to AsCl3 which being volatile in nature is also carried off along with relatively large volumes of CO2 (generated by the substance and HCl).

 

Examples : Magnesium Carbonate, Light Magnesium Oxide, Calcium Hydroxide, Chalk, KOH, NaOH.

 

(iii) Insoluble Substances : These substances, as those that do not interfere with the solution of As and its subsequent reduction to AsH3 (arsine). Such substances are suspended in water along with stannated-HCl, and the normal test is performed.

 

Examples : Magnesium Trisilicate, Bentonite, Barium Sulphate, Light and Heavy Kaolin.

 

(iv) Metals Interfering with Normal Reaction

 

(a) Iron : It gets deposited on the surface of Zn thereby depressing the intensity of reaction between Zn and HCl to produce H2.

 

Remedy : The sample is dissolved in H2O and stannated HCl to allow conversion of all As to As3+ and finally as AsCl3. The latter being volatile in nature can be separated by distillation from remaining metallic salts and the distillate examined in the normal manner.

 

Example : Ferrous Sulphate.

 

(b) Antimony : Sb-compounds are also reduced simultaneously by Zn/HCl to yeild SbH3 (stilbine) that reacts with HgCl2 paper to give a stain. Therefore, the sample is first distilled with HCl to yield a distillate containing all the As as AsC3 (volatile), but yields only a fraction of Sb as SbCl3 (non-volatile). A repeated distillation obviously gets rid of even the last traces of Sb.

 

Examples : Antimony Potassiun Tartrate, Antimony Sodium Tartrate.

 

A few typical examples are cited below from the official compendium.


 

3. Limit Test for Iron

 

Theory : The limit test for Iron is based on the reaction between iron and thioglycollic acid in a medium buffered with ammonium citrate to give a purple colour, which is subsequently compared with the standard colour obtained with a known amount of iron (0.04 mg of Fe). Ferrous thioglycollate is a co-ordination compound that attributes the purple colour ; besides thioglycollic acid converts the entire Fe3+ into Fe2+. The reactions involved may be expressed as follows :


 

Materials Required

Nessler cylinder : 1 pair ; Ferric ammonium sulphate : 1.726 g ; Sulphuric acid (0. 1 N) : 10.0 ml ; Iron-free citric acid (20% w/v) : 2.0 ml ; Thioglycollic acid : 0.1 ml; Iron-free ammonia solution : 20 ml.

 

Standard Iron Solution : Weigh accurately 0.1726 g of ferric ammonium sulphate and dissolve in 10 ml of 0.1 N sulphuric acid and suffcient water to produce 1 Litre. Each ml of this solution contains 0.02 mg of Fe.

 

Standard Colour : Dilute 2.0 ml of standard iron solution with 40 ml DW in a Nessler cylinder. Add 2 ml of a 20% w/v solution of iron-free citric acid and 0.1 ml of thioglycollic acid, mix, make alkaline with iron-free ammonia solution, dilute to 50 ml with DW and allow to stand for 5 minutes.

 

Procedure : Dissolve the specified quantity of the substance being examined in 40 ml DW, and trans-fer to a Nessler cylinder. Add to it 2 ml iron-free citric acid solution and 0.1 ml thioglycollic acid, mix, make alkaline with iron-free ammonia solution, dilute to 50 ml with DW and allow to stand for 5 minutes. Any colour produced is not more intense than the standard colour.

 

Some examples of this test for pharmaceutical substances are listed below :



 

Chapter: Pharmaceutical Drug Analysis: Pharmaceutical Chemicals: Purity and Management

Limit Tests for Lead

Limit Tests for Lead
Theory : The offcial test is based on the conversion of traces of lead salts present in the pharmaceutical substances to lead sulphide, which is obtained in colloidal form by the addition of sodium sulphide in an alkaline medium achieved by a fairly high concentration of ammonium acetate.

Limit Tests for Lead

 

Theory : The offcial test is based on the conversion of traces of lead salts present in the pharmaceutical substances to lead sulphide, which is obtained in colloidal form by the addition of sodium sulphide in an alkaline medium achieved by a fairly high concentration of ammonium acetate. The reaction may be expressed as follows :

PbCl2 + Na2S  → PbS (Dec) + 2NaCl

 

The brown colour, caused due to colloidal lead sulphide in the test solution is compared with that produced from a known amount of lead.

 

Equipment : Nessler Cylinders (or Nessler Glasses) : According to the British Standard Specification No : 612, 966—a pair of cylinders made of the same glass and having the same diameter with a graduation mark at the same height from the base in both cylinders (Figure 1).


The final comparison is made by viewing down through the solution against a light background.

Materials Required :

(i) Lead Nitrate Stock Solution : Dissolve0.1598 g of lead nitrate in 100 ml DW to which has been  added 1 ml nitric acid, then dilute with water to 1 Litre.

Note : The solution must be prepared and stored in polyethylene or glass containers free from soluble lead salts.

(ii) Standard Lead Solution : On the day of use, dilute 10.0 ml of lead nitrate stock solution with DW to 100.0 ml. Each ml of standard lead solution contains the equivalent of 10 microgrammes of lead. A control comparison solution prepared with 2.0 ml of standard lead solution contains, when compared to a solution representing 1.0 g of the substance being tested, the equivalent of 20 parts per million of lead.

(iii) Standard Solution : Into a 50 ml Nessler Cylinder, pipette 2 ml of standard lead solution and dilute with DW to 25 ml. Adjust with dilute acetic acid Sp. (IP)* or dilute ammonia solution Sp. (IP) to a pH between 3.0 and 4.0, dilute with DW to about 35 ml and mix.

(iv) Test Solution : Into a 50 ml Nessler Cylinder, place 25 ml of the solution prepared for the test as directed in the individual monograh, dissolve and dilute with DW to 25 ml the specified quantity of the substance being tested. Adjust with dilute acetic acid Sp. (IP) or dilute ammonia solution Sp. to a pH between 3.0 and 4.0, dilute with DW to about 35 ml and mix.

 

Procedure : To each of the cylinders containing the standard solution and test solution respectively, add l0 ml of freshly prepared hydrogen sulphide solution, mix, dilute with water (DW) to 50 ml, allow to stand for 5 minutes and view downwards over a white surface, the colour produced in the test solution is not darker than that produced in the standard solution.

 A few typical examples from the official compendium are given below :


 

Chapter: Pharmaceutical Drug Analysis: Pharmaceutical Chemicals: Purity and Management

Limit Test for Arsenic

Limit Test for Arsenic
Theory : The official process is a development of the Gutzeit Test wherein all arsenic present is duly converted into arsine gas (AsH3) by subjecting it to reduction with zinc and hydrochloric acid.

Limit Test for Arsenic

 

Theory : The official process is a development of the Gutzeit Test wherein all arsenic present is duly converted into arsine gas (AsH3) by subjecting it to reduction with zinc and hydrochloric acid. Further, it depends upon the fact that when arsine comes into contact with dry paper permeated with mercuric (Hg2+) chloride it produces a yellow strain, the intensity of which is directly proportional to the quantity of arsenic present. The various chemical reactions involved may be expressed by the following equations :


The details of experimental procedure described in the Pharmacopoeia are actually based upon a paper by Hill and Collins**, but have been adequately modified from time to time in accordance with the accumu-lated and acquired experience. Explicitly, the expressions provided in the Pharmacopoeia for limits of arsenic exclusively refer to parts per million, calculated as As.

Materials Required : Arsenic limit test apparatus; HgCl2—paper : smooth white filter paper (having thickness in mm of 400 paper = weight in g per Sq. M.), soaked in a saturated solution of HgCl2, pressed to get rid of excess of soln. and dried at about 60°C in the dark ; lead acetate solution 10.0% w/v soln. of PbAc2 in CO2– free water ; KI (AsT), 1.0 g ; Zn (AsT) : l0.0 g ; Dilute Arsenic solution (AST); Standard stains, Test Solutions—are prepared according to the Indian Pharmacopoeia 1996.

Arsenic Limit Test Apparatus (Figure 2)


A wide-mouthed glass bottle capable of holding about 120 ml is fitted with a rubber bung through which passes a glass tube. The latter, made from ordinary glass tubing, has a total length of 200 mm and an internal diameter of exactly 6.5 mm (external diameter about 8 mm). It is drawn out at one end to a diameter of about 1 mm and a hole not less than 2 mm in diameter is blown in the side of the tube, near the constricted part. When the bung is inserted in the bottle containing 70 ml of liquid, the constricted end of the tube is kept above the surface of the liquid, and the hole in the side is below the bottom of the bung. The upper end of the tube is cut off square, and is either slightly rounded or ground smooth.

The rubber bungs (about 25 mm × 25 mm), each with a hole bored centrally and through exactly 6.5 mm  in diameter,are fitted with a rubber band or spring clip for holding them tightly in place.

Procedure : The glass tube is lightly packed with cotton wool, previously moistened with lead acetate solution and dried, so that the upper surface of the cotton wool is not less than 25 mm below the top of the tube. The upper end of the tube is then inserted into the narrow end of one of the pair of rubber bungs, to a depth of l0 mm (the tube must have a rounded-off end). A piece of mercuric chloride paper is placed flat on the top of the bung and the other bung placed over it and secured by means of the spring clip in such a manner that the holes of the two bungs meet to form a true tube 6.5 mm diameter interrupted by a diaphragm of mercuric chloride paper.

 

The test solution prepared as specified, is placed in the wide-mouthed bottle, 1 g of KI (AsT) and 10 g of Zn (AsT) are added, and the prepared glass tube is placed quickly in position. The reaction is allowed to proceed for 40 minutes. The yellow stain that is produced on the HgCl2 paper if As is present is compared by daylight with the standard stains obtained by performing in an identical manner with known quantities of dilute arsenic solution (AsT). The comparison of the stains is made immediately at the completion of the test.

 

By matching the intensity and depth of colour with standard stains, the proportion of arsenic in the substance may be estimated. Thus, a stain equivalent to the 1 ml standard stain obtained by performing on l0 g of a substance implies that the proportion of As is 1 part per million.

 

Cautions : 

(i) HgCl2 paper should be protected from sunlight during the test to avoid lighter or no

stain.

 

(ii) The standard and test stains must be compared immediately as they fade out on retaining.

 

(iii) The reaction may be expedited by the application of heat and 40°C is considered to be the most ideal temperature.

 

(iv) The tube should be washed with HCl (AsT), rinsed with DW, and dried between successive tests.

 

Special Techniques : The special techniques are usually applicable to a host of pharmaceutical sub-stances before the normal test can be performed. A few typical examples would be discussed briefly here, namely :

 

(i) Free Acids : They are first converted to their respective sodium salts with Na2CO3 and As3+ oxi-dised to As5+ by evaporating the solution with Br2. The residue is ignited carefully until carbonised to destroy organic matter, while As is kept as non-volatile sodium arsenate. The resulting residue is dissolved in brominated HCl and the test carried out in the normal manner.

 

Examples : Aspirin, Saccharin, Sodium Salicylate, Sodium Aminosalicylate.

 

(ii) Substances Reacting Vigorously with HCl : The As is readily converted to AsCl3 which being volatile in nature is also carried off along with relatively large volumes of CO2 (generated by the substance and HCl).

 

Examples : Magnesium Carbonate, Light Magnesium Oxide, Calcium Hydroxide, Chalk, KOH, NaOH.

 

(iii) Insoluble Substances : These substances, as those that do not interfere with the solution of As and its subsequent reduction to AsH3 (arsine). Such substances are suspended in water along with stannated-HCl, and the normal test is performed.

 

Examples : Magnesium Trisilicate, Bentonite, Barium Sulphate, Light and Heavy Kaolin.

 

(iv) Metals Interfering with Normal Reaction

 

(a) Iron : It gets deposited on the surface of Zn thereby depressing the intensity of reaction between Zn and HCl to produce H2.

 

Remedy : The sample is dissolved in H2O and stannated HCl to allow conversion of all As to As3+ and finally as AsCl3. The latter being volatile in nature can be separated by distillation from remaining metallic salts and the distillate examined in the normal manner.

 

Example : Ferrous Sulphate.

 

(b) Antimony : Sb-compounds are also reduced simultaneously by Zn/HCl to yeild SbH3 (stilbine) that reacts with HgCl2 paper to give a stain. Therefore, the sample is first distilled with HCl to yield a distillate containing all the As as AsC3 (volatile), but yields only a fraction of Sb as SbCl3 (non-volatile). A repeated distillation obviously gets rid of even the last traces of Sb.

 

Examples : Antimony Potassiun Tartrate, Antimony Sodium Tartrate.

 

A few typical examples are cited below from the official compendium.


 

Limit Test for Iron

 

Theory : The limit test for Iron is based on the reaction between iron and thioglycollic acid in a medium buffered with ammonium citrate to give a purple colour, which is subsequently compared with the standard colour obtained with a known amount of iron (0.04 mg of Fe). Ferrous thioglycollate is a co-ordination compound that attributes the purple colour ; besides thioglycollic acid converts the entire Fe3+ into Fe2+. The reactions involved may be expressed as follows :


 

Materials Required

Nessler cylinder : 1 pair ; Ferric ammonium sulphate : 1.726 g ; Sulphuric acid (0. 1 N) : 10.0 ml ; Iron-free citric acid (20% w/v) : 2.0 ml ; Thioglycollic acid : 0.1 ml; Iron-free ammonia solution : 20 ml.

 

Standard Iron Solution : Weigh accurately 0.1726 g of ferric ammonium sulphate and dissolve in 10 ml of 0.1 N sulphuric acid and suffcient water to produce 1 Litre. Each ml of this solution contains 0.02 mg of Fe.

 

Standard Colour : Dilute 2.0 ml of standard iron solution with 40 ml DW in a Nessler cylinder. Add 2 ml of a 20% w/v solution of iron-free citric acid and 0.1 ml of thioglycollic acid, mix, make alkaline with iron-free ammonia solution, dilute to 50 ml with DW and allow to stand for 5 minutes.

 

Procedure : Dissolve the specified quantity of the substance being examined in 40 ml DW, and trans-fer to a Nessler cylinder. Add to it 2 ml iron-free citric acid solution and 0.1 ml thioglycollic acid, mix, make alkaline with iron-free ammonia solution, dilute to 50 ml with DW and allow to stand for 5 minutes. Any colour produced is not more intense than the standard colour.

 

Some examples of this test for pharmaceutical substances are listed below :

 

Chapter: Pharmaceutical Drug Analysis: Pharmaceutical Chemicals: Purity and Management

Limit Test’s for Acid Radical Impurities

Acid radical impurities constitute a serious but unavoidable source of impurities in a large number of pharmaceutical chemicals.

LIMIT TEST’S FOR ACID RADICAL IMPURITIES

 

Acid radical impurities constitute a serious but unavoidable source of impurities in a large number of pharmaceutical chemicals. However, the two most commonly found acid radical impurities are chloride (Cl ) and sulphate (SO42–) that evidently arise from the inevitable use of raw tap-water in various manufacturing operations. As these two acid radical impurities are found in abundance due to contamination, the Pharmaco-poeia categorically stipulates limit tests for them which after due minor modifications are applicable to a number of pharmaceutical substances.

 

In addition to the above two commonly found impurities, there are a number of other acid radical impurities which exist in pharmaceutical substances, namely : arsenate, carbonate, cyanide, nitrate, oxalate, phosphate and silicate.

 

All these acid radical impurities shall be discussed briefly as under :

 

1. Limit Test for Chlorides

 

The limit test for chlorides is based on its precipitation with silver nitrate in the presence of dilute HNO3, and comparing the opalescence produced due to the formation of AgCl with a standard opalescence achieved with a known quantity of Cl ions.

The equation may be expressed as :


Materials Required : Nessler cylinder 1 pair ; dilute nitric acid (10% w/w of HNO3) 10.0 ml ; silver nitrate solution (5.0% w/v in DW) 1.0 ml.

 

Standard Opalescence : Place 1.0 ml of a 0.05845% w/v solution of NaCI in 10 ml of dilute HNO3 in a Nessler cylinder. Dilute to 50 ml with DW and add 1 ml of AgNO3 solution. Stir immediately with a glass rod and allow to stand for 5 minutes.

 

Procedure : Dissolve the specified quantity for the substance in DW, or prepare a solution as directed in the text and transfer to a Nessler cylinder. Add 10 ml of dilute nitric acid, except when it is used in the preparation of the solution, dilute to 50 ml with DW, and add 1 ml of AgNO3 solution. Stir immediately with a glass rod and allow to stand for 5 minutes. The opalescence produced is not greater than the standard opalescence, when viewed transversely.

 

A few typical examples of this test representing a wide spectrum of pharmaceutical substances are enumerated below :



 

2. Limit Test for Sulphates

 

Theory : The limit test for sulphates is based upon its precipitation as barium sulphate in the presence of barium chloride, hydrochloric acid and traces of barium sulphate. In this combination, hydrochloric acid exerts its common ion effect whereas traces of BaSO4 aids in the rapid and complete precipitation by seeding. Thus, the opalescence caused by the sample is compared immediately with a standard turbidity produced with a known amount of the SO42– ion.

 

The main objective of this test is to provide a rigid control of sulphate as an impurity present primarily in inorganic pharmaceutical substances.

 

Materials Required : Nessler cylinders 1 pair ; dilute hydrochloric acid (10% w/v of HCl) 2.0 ml.

 

Barium Sulphate Reagent : Mix 15 ml of 0.5 M barium chloride, 55 ml of DW, and 20 ml of sulphate free alcohol, add 5 ml of a 0.0181% w/v soln. potassium sulphate dilute to 100 ml with DW, and mix. It should always be prepared fresh.

 

0.5 M Barium Chloride : BaCl2 dissolved in DW to contain in 1 Litre 122.1 g of BaCl2. 2H2O.

Standard Turbidity : Place 1.0 ml of a 0.1089% w/v soln. of K2SO4 and 2 ml of dilute HCl in a Nessler cylinder, dilute to 45 ml with DW, add 5 ml BaSO4 reagent, stir immediately with a glass rod and allow to stand for 5 minutes.

 

Procedure : Dissolve the specified quantity of the substance in DW, transfer to a Nessler cylinder, and the preparation of the solution. Dilute to 45 ml with DW, add 5 ml barium sulphate reagent, stir immediately with a glass rod, and allow to stand for 5 minutes. The turbidity is not greater than the standard turbidity, when viewed transversely.

 

A few examples of this test consisting of a cross-section of pharmaceutical substances are stated below :



 

3. Limit Test for Arsenate

 

Acetarsol : An organic arsenic compound, being therapeutically active when administered orally, that might be of value in the treatment of spirochaetal or protozoal diseases, for instance : syphilis, yaws, relapsing fever, sleeping sickness and amoebic dysentry.

 

It is made from p-hydroxyphenylarsonic acid, which may be prepared either by straight forward meth-ods from phenol or from p-aminophenylarsonic acid. The resulting compound obtained from either of these reactions is nitrated, reduced and the base is finally acetylated to afford acetarsol.


Inorganic arsenates are found to be extremely toxic in nature and hence careful control is maintained by the addition of magnesium ammonio-sulphate solution to an aqueous solution of the sample, thereby producing an instant white precipitate.

 

4. Limit Test for Carbonate

 

Carbonate impurity in pharmaceutical chemicals usually arise from contamination with atmospheric CO2.

Examples of a few official compounds subject to this test from the Pharmacopoeia are given below :


 

5. Limit Test for Cyanide

 

Cyanide present in Edetate Disodium is assayed by titration with AgNO3 in neutral solution employing dimethylaminobenzylidenerhodamine as an adsorption indicator with a colour change from yellow to orange.

 

A few typical examples are illustrated below :

 

A. Edetate Disodium

 

Materials Required : Edetate disodium 30.0 g ; sodium hydroxide solution (20% w/v in DW) 35.0 ml ; dimethylaminobenzylidenerhodamine solution (0.02% w/v in acetone) 1.0 ml ; 0.01 N AgNO3 solution (1.699 g in 1 litre of DW) 100 ml.

 

Procedure : Dissolve 30.0 g in a mixture of 100 ml DW and 35 ml NaOH solution, add 1 ml dimethylaminobenzylidenerhodamine and titrate with 0.01N silver nitrate until the colour of the solution changes from yellow to orange. Repeat the operation without the disodium edetate. The difference between the titrations is not more than 1.25 ml.

 

B. Iodine

 

Materials Required : Iodine 3.5 g ; zinc powder 10 g ; ferrous sulphate solution (2.0% w/v in boiled and cooled DW) 1.0 ml ; sodium hydroxide solution (20% w/v in DW) 1 ml ; hydrochloric acid (~ 11.5 N) 20 ml.

 

Procedure : Triturate 3.5 g thoroughly with 35 ml DW, filter and decolorise the filtrate by the addition of a little zinc powder. To 5.0 ml of the filtrate add a few drops of ferrous sulphate solution and 1 ml NaOH solution ; warm gently and acidify with HCl, no blue colour or green colour is produced.

 

C. Potassium Iodide

 

Materials Required : Potassium iodide 0.5 g ; ferrous sulphate solution (2.0% w/v in boiled and cooled DW) 1 drop ; NaOH solution (20% w/v in DW) 0.5 ml ; HCl 20.0 ml.

 

Procedure : Dissolve 0.5 g in 5 ml warm DW, add 1 drop of ferrous sulphate solution and 0.5 ml NaOH solution and acidify with HCl, no blue colour is produced.

 

6. Limit Test for Nitrate

 

Basic nitrate is usually found as an impurity in bismuth salts (e.g., bismuth subcarbonate), very often due to the mode of preparation from the metal via bismuth nitrate.

 

BP (1914) first described a limit test, based upon the production of coloured nitro-compounds by the interaction of traces of nitrates with phenol-2, 4-disulphonic acid, and the conversion of these subsequently into dark-yellow ammonium salts. However, this test has a serious disadvantage of correctly matching the yellow colours with great difficulty.

 

BP (1932) put forward a more reliable test for nitrate based upon the oxidation of indigocarmine to colourless substances by the action of traces of nitrates in presence of hot and fairly concentrated sulphuric acid, and the reaction may be expressed as follows :


The quantities as specifed in the Pharmacopoeia allow an official limit of nitrate equivalent to about 0.29% BiONO3.

 

A few typical instances of pharmaceutical substances are enumerated below :


 

7. Limit Test for Oxalate

 

Oxalate is found to be a frequent impurity in pharmaceutical substances belonging to the category of either organic acids e.g., anhydrous citric acid, tartaric acid; or salts of organic acids e.g., ferrous gluconate, sodium citrate, potassium citrate and sodium cromoglycate. The presence of this impurity is due to the following two prime factors, namely :

 

(a) The use of oxalic acid to get rid of Ca2+ during various manufacturing processes.

(b) The use of oxalic acid in the isolation and purification of organic bases e.g., ephedrine (thereby resulting into the formation of well defined crystalline oxalates).

 

A few typical examples are cited below :


 

8. Limit Test for Phosphate

 

The limit test for phosphate is based upon the formation of a yellow colour reaction with molybdovanadic reagent (combination of ammonium vanadate and ammonium molybdate) in an acidic medium. However, the exact composition of the molybdovanadophosphoric acid complex is yet to be established.

Three typical examples of pharmaceutical substances are stated below :


 

Molybdovanadic Reagent : Suspend 4.0 g of finely powdered ammonium molybdate and 0.1 g of finely powdered ammonium metavanadate in 70 ml DW and grind until dissolved. Add 20 ml of HNO3 and dilute to 100 ml with DW.

 

Phosphate Standard Solution (5 ppm PO4) : Dilute 0.5 ml of a 0.143% w/v soln. of potassium dihydrogen orthophosphate (KH2PO4) to 100 ml with DW.

 

Sulphomolybdic Solution : Dissolve with heating, 25 ml ammonium molybdate in 200 ml DW. Separately, with care, add 280 ml H2SO4 to 500 ml DW. Cool and mix the two solutions and dilute to 1 Litre with DW.

 

Methylaminophenol-sulphite Solution : Dissolve 0.1 g of 4-methylaminophenol sulphate, 20 g sodium metabisulphite and 0.5 g anhydrous sodium sulphite in sufficient DW to produce 100 ml.


Chapter: Pharmaceutical Drug Analysis: Pharmaceutical Chemicals: Purity and Management

Limit Tests for Non-Metallic Impurities

A few typical examples are described below which essentially contains the above cited nonmetallic impurities :

LIMIT TESTS FOR NON-METALLIC IMPURITIES

 

Non-metallic impurities, such as boron, free halogens (I2, Br2 and Cl2) and selenium in pharmaceutical substances usually contribute untoward reactions, skin manifestations and are found to be toxic to healthy tissues.

 

A few typical examples are described below which essentially contains the above cited nonmetallic impurities :

 

1. Boron

 

A. Salbutamol Sulphate : Boron shows its presence in the above compound as a result of the use of sodium borohydride (NaBH4) in the manufacturing process. The estimation depends upon the conversion of boron to borate and the organic matter is subsequently destroyed by ignition with anhydrous sodium carbonate. The quantity of boron is finally determined by colorimetric assay.

 

Materials Required : Salbutamol sulphate 50 mg ; solution of an equimolar mixture of anhydrous sodium carbonate and potassium carbonate (3% w/v in DW) 5.0 ml ; Solution of curcumin (0.125% w/v in glacial acetic acid) 3.0 ml ; mixture of H2SO4 and glacial CH3COOH (5 ml : 5 ml) 3.0 ml ; ethanol (96%) 100 ml ; solution of boric acid (dissolve 5 g of boric acid in a mixture of 20 ml DW and 20 ml absolute ethanol and dilute to 250 ml with absolute ethanol) : 100 ml.

 

Procedure : To 50 mg of substance add 5 ml of a 3% w/v solution of an equimolar mixture of anhydrous Na2CO3 and K2CO3, evaporate to dryness on a water-bath and dry at 120°C. Ignite the residue rapidly until the organic matter has been destroyed, allow to cool and add 0.5 ml DW and 3 ml freshly prepared 0.125% w/v soln. of curcumin in glacial acetic acid. Warm gently to effect solution, allow to cool and add 3 ml of a mixture of H2SO4, with stirring, to 5 ml of glacial acetic acid. Mix and allow to stand for 30 minutes. Add sufficient ethanol (96%) to produce 100 ml, filter and measure the absorbance of the filtrate at the maximum of 555 nm. Calculate the content of boron from a reference curve prepared from the absorbance obtained by treating suit-able aliquots of a solution of boric acid in the same manner.

 

Prescribed Limits : Not more than 50 ppm.

 

2. Free Halogens

 

A few typical examples of pharmaceutical chemicals in which free halogens like Iodine, Bromine, Fluo-rine and Chlorine are present as non-metallic impurities are given below.

 

A. Clioquinol : (Free Iodine)

 

Materials Required : Clioquinol 1.0 g ; potassium iodide 1.0 g ; H2SO4 (1 M) 1.0 ml ; chloroform 2.0 ml ; sodium thiosulphate (0.005 M) 0.1 ml.

 

Procedure : Shake 1.0 g with a solution of 1 g potassium iodide in 20 ml DW for 30 seconds, allow to stand for 5 minutes and filter. To 10 ml of the filtrate add 1 ml 1 M H2SO4 and 2 ml chloroform and shake.

 

Prescribed Limits : Any colour in the chloroform layer is discharged on the addition of 0.1 ml of 0.005 M sodium thiosulphate.

 

B. Diethylpropion Hydrochloride : (Free Bromine)

 

Test : Place 0.05 ml of a 10% w/v solution on starch-iodide paper.

 

Prescribed Limit : No colour is produced.

 

C. Doxycycline Hydrochloride : (Free Fluorine)

 

Materials Required : Doxycyline Hydrochloride : 0.30 g ; oxygen-combustion flask ; 1 L capacity;

 

Nessler cylinder 100 ml ; zirconyl alizarin solution* : 5.0 ml ; fluoride standard solution (10 ppm F) (dilute 5.0 ml of a 0.0442 % w/v soln. of sodium fluoride, previously dried at 300°C for 12 hours, to 100 ml with DW) : 3.0 ml.

 

Procedure : Burn 0.30 g, in three equal portions, by the method for oxygen-flask combustion (BP), using a 1 Litre flask and a separate 20 ml portion of DW as the absorbing liquid for each combustion, shaking the flask vigorously for about 15 minutes and transferring to the same 100 ml Nessler cylinder. Add 5 ml of acid zirconyl alizarin solution to the combined liquids, adjust the volume to 100 ml with DW and allow to stand for 1 hour.

 

Prescribed Limit : The colour of the resulting solution is greater than that obtained by repeating the operation with no substance enclosed in the successive portions of filter paper burnt in the method for oxygen flask combustion, but adding 3.0 ml of fluoride standard solution (10 ppm F) to the combined absorption liquids before adding the acid zirconyl alizarin solution.

 

D. Chloroform : (Free Chlorine)

 

Materials Required : Chloroform 10.0 ml ; cadmium iodide solution (5.0% w/v in DW) 1.0 ml ; starch mucilage 0.1 ml.

 

Procedure : Shake 10 ml of chloroform with 20 ml of freshly boiled and cooled DW for 3 minutes and allow to separate. To the aqueous layer add 1 ml cadmium iodide soln. and 0.1 ml of 10 ml of starch mucilage.

 

Prescribed Limit : No blue colour is produced.

 

E. Tetrachloroethylene (Free Chlorine)

 

Perform the limit test as stated under chloroform. No blue colour is produced.

 

3. Selenium

 

A. Selenium Sulphide

 

Theory : Selenium is very toxic and its contamination is usually controlled by an absorptiometric method after destruction of the organic compound with fuming nitric acid. The latter converts selenium (Se) as selenous acid (H2SeO3), which on subsequent treatment with 3,3-diaminobenzidine under controlled experimental pa-rameters, results into the formation of a highly coloured compound known as 3,4-diaminophenylpiazselenol. The latter is consequently extracted with toluene after making the aqueous solution alkaline, and the colour compared with a standard prepared likewise from a known amount of selenium. The various reactions involved may be expressed as follows :


Materials Required : Selenium sulphide : 10.0 g ; formic acid (2.5 M) : 2.0 ml ; 3,3-diaminobenzedine tetrahydrochloride solution (0.5% w/v in DW) : 2.0 ml ; ammonia (5 M) : 20 ml ; selenium standard solution (1 ppm Se) (Dilute 2.5 ml of a 0.00654% w/v solution of selenous acid to 100 ml with DW) : 5.0 ml.

 

Procedure : To 10 g of selenium sulphide add 100 ml DW, mix well, allow to stand for 1 hour with frequent shaking and filter. To 10 ml of the filtrate, add 2 ml of 2.5 M formic acid, dilute to 50 ml with DW, adjust the pH to 2.0 to 3.0 with 2.5 M formic acid, add 2.0 ml of a 3,3-diaminobenzidine tetrahydrochloride in DW, allow to stand for 45 minutes and adjust the pH to 6.0 to 7.0 with 5 M ammonia. Shake the solution for 1 minute with 10 ml of toluene and allow to separate. Measure the absorbance at 420 nm.

 

Prescribed Limit : The measured absorbance at 420 nm is not greater than that of a solution prepared by treating 5 ml of selenium standard solution (1 ppm Se) in the same manner (5 ppm, calculated as Se).

 

Chapter: Pharmaceutical Drug Analysis: Theory and Technique of Quantitative Analysis

Theory and Technique of Quantitative Analysis

The ‘technique of quantitative analysis’ is broadly based on the following three major heads, namely : (a) Technique of Volumetric Analysis, (b) Technique of Gravimetric Analysis, and (c) Biomedical Analytical Chemistry.

THEORY AND TECHNIQUE OF QUANTITATIVE ANALYSIS

 

INTRODUCTION

 

The ‘technique of quantitative analysis’ is broadly based on the following three major heads, namely :

 

(a) Technique of Volumetric Analysis,

 

(b) Technique of Gravimetric Analysis, and

 

(c) Biomedical Analytical Chemistry.

 

Volumetric analysis essentially comprises of the most precise and accurate measurement of interacting solutions or reagents. It makes use of a number of graduated apparatus, such as : graduated (volumetric) flasks, burettes, pipettes and measuring cylinder of different capacities (volumes).

 

However, it is pertinent to mention here that quite a few techniques related to measurement of pharmaceutical substances and reagents involved is more or less common to both gravimetric and volumetric analysis. Besides, in gravimetric analysis, some more additional techniques play a vital role, namely : precipitation, filtration, washing of the precipitate and ignition of the precipitate.

 

Biomedical analytical chemistry happens to be one of the latest disciplines which essentially embraces the principles and techniques of both analytical chemistry and biochemistry. It has often been known as ‘clinical chemistry’. This particular aspect of analytical chemistry has gained significant cognizance in the recent past by virtue of certain important techniques being included very much within its scope of analysis, namely : colorimetric assays, enzymic assays, radioimmunoassays and automated methods of clinical analysis.

 

It is, however, important to mention here that certain other routine procedures also carried out in a clinical laboratory fall beyond the scope of biomedical analytical chemistry, narnely : microbiological assays, heamatological assays, serum analysis, urine analysis and assays of other body fluids.

It will be very much within the scope of this to discuss briefly the various important details, with specific examples wherever necessary, of volumetric analysis, gravimetric analysis and biomedical analytical chemistry


Chapter: Pharmaceutical Drug Analysis: Theory and Technique of Quantitative Analysis

Volumetric Analysis

Volumetric analysis may be broadly defined as those analytical methods whereby the exact volume of a solution of known concentration actually consumed during the course of an analysis is considered as a measure of the amount of active constituent in a given sample under determination (assay).

VOLUMETRIC ANALYSIS

Volumetric analysis may be broadly defined as those analytical methods whereby the exact volume of a solution of known concentration actually consumed during the course of an analysis is considered as a measure of the amount of active constituent in a given sample under determination (assay).

 

1. THEORY

 

According to the official method of analysis, hydrochloric acid can be determined by first weighing a given sample accurately, and secondly, by adding carefully a solution of known strength of sodium hydroxide in the presence of an appropriate indicator unless and until the exact equivalent amounts of HCl and NaOH have undergone the following chemical reaction :


Analyte (or Active Constituent) is the chemical entity under assay e.g., HCl.

 

Titrant is the solution of known strength (or concentration) employed in the assay e.g., NaOH.

 

Titration is the process of adding and then actually measuring the volume of titrant consumed in the assay. This volume is usually measured by the help of a calibrated burette.

 

Indicator is a chemical substance sensitive enough to display an apparent change in colour very close to the point in the ongoing titration process at which equivalent quantities of analyte and titrant have almost virtually reacted with each other.

 

Equivalence Point (or Stoichiometric Point) is the point at which there appears an abrupt change in certain characteristic of the prevailing reaction mixture—a change that is either ascertained electrometrically or is visibly spotted by the use of indicators.

 

In usual practice, the volumetric titrations may be accomplished either by direct titration method e.g., assay of HCl employing NaOH as the titrant, or by residual titration method e.g., assay of ZnO in which case a known-excess-measured volume of standardised solution of H2SO4, more than the actual amount chemically equivalent to ZnO, is added to the sample ; thereupon, the H2SO4 which remain unreacted with ZnO is subsequently titrated (sometimes referred to as back titration or residual titration in the text) employing standardized NaOH solution.

 

Thus, we have :

 

Known amount of H2SO4 consumed Known amount of NaOH + Unknown amount of ZnO Most official compendia usually record the results of drug assays in terms of % w/v, % w/w and % v/v.

 

2. DEFINITIONS

 

In order to have a clear-cut understanding of the various calculations involving volumetric assays through-out this book one needs to gain an in-depth knowledge of the various terms related to ‘equivalents’. They are :

 

(a) Gram-equivalent Weight (GEW) : It is the weight in grams that is chemically equivalent to 1 gram-atom of hydrogen (1.0079 g).

 

It is also sometimes simply referred to as the ‘gram-equivalent’. However, GEW has two distinct definitions for neutralization as well as as oxidation-reduction reactions as stated below :

 

(i)          For Neutralization Reactions : GEW is defined as that weight of a substance in grams which contains, furnishes, reacts directly or indirectly and replaces 1 gram-atom or ion of hydrogen.

(ii) For Oxidation—Reduction Reactions

 

Explanation : A reaction usually takes place by the combination of oxidizing and reducing agents and this may be considered as the basis for the quantitative measurement of one of the reactants. For instance, FeSO4 can be determined quantitatively by its reaction with ceric sulphate [Ce(SO4)2] as expressed by the following equation :

                        .......(a)

Equation (a) can be split into two half-equations as shown below thereby depicting the loss of elec-trons by the Fe2+ ion [Eq. (b)] and the gain of electrons by the Ce4+ [Eq. (c)] :

                      ...........(b)

                  .................(c)

From Eq. (a) it is evident that each molecule of FeSO4, upon oxidation, happens to lose one electron. Hence, one mole of FeSO4 loses 6.02 × 10 23 electrons which is equivalent to 1 Faraday or 96,500 C. Thus, in electrochemical determination of equivalence point the quantity of electricity is almost identical with that required to reduce 1 mole of Ce(SO4)2. It follows from here that 1 mole of FeSO4 and 1 mole of Ce(SO4)2 are chemical equivalents. In other words, 1 g of H, acting as a reducing agent, loses electrons equivalent to 96,500 C.

 

(b) Equivalent Weight of a Reducing Agent is that weight which loses electrons equivalent to 96,500 C.

 

It may be calculated by dividing the gram-molecular weight by the number of electrons lost by each molecule, for instance :


hence, the equivalent weight of FeSO4 oxidizing to Fe2(SO4)3 comes out to be 151.919 [FeSO4 :

molecular weight = 151.91] or 1 gram-molecular weight.

 

(c) Equivalent Weight of an Oxidizing Agent is that weight which gains electrons equivalent to 1 Faraday, or to the electrons gained by 1 gram-ion of H+ ions (2H+ + 2e H2 ).

 

It may be calculated by dividing the gram-molecular weight by the number of electrons gained by each molecule, for example :

(a)   Ce4+ + e   → Ce3+      (cerous ion)

hence, the equivalent weight of ceric sulphate is 1 gram-molecular weight 332.24 g [Ce(SO4)2 : molecular weight = 332.24]

(b) MnO4– + 5e  → Mn2+   (manganous ion)

hence, the equivalent weight of potassium permanganate is 1/5th gram-molecular weight 31.61 g.

 

(KMnO4 : 1/5 × 158.05 = 31.61)

(c)   Cr2O72– + 6e —  2Cr3+                    (chromous ion)

 

hence, the equivalent weight of potassium dichromate is l/6 gram-molecular weight 49.03 g. (K2Cr2O7 : 1/6 × 294.18 = 49.03)

(d) I2 + 2e  → 2I–  (iodide ion)

hence, the equivalent weight of iodine is 1 gram-molecular weight 126.90 g. (I2 : Molecular Weight = 126.90)

(e)    BrO3 + 6e   → Br                            (bromide ion)

 

hence, the equivalent weight of potassium bromate is 1/6 gram-molecular weight 27.83 g.

 

(KBrO3 : 1/6 × 167.01 = 27.83)

 

(d) Gram-milliequivalent Weight (GmEW) is nothing but GEW/1000. This term is very much used in all types of volumetric calculations.

 

(e) Equivalent (equiv) is the number of gram-equivalents involved in a quantitative method.

 

(f) Milliequivalent (meq) is the number of gram-milliequivalents involved in a quantitative method.

 

However, meq is used more frequently than equiv in quantitative procedures.

 

(g) Standard Solution is a solution of known (pre-determined) normality or molarity.

 

(h) Normality (expression of concentration) is the number of equivalents of solute per litre (equiv/lire) or milliequivalents per ml. (meg/ml) solution.

 

(i) Molarity is the expression of the concentration of a solution in terms of moles per litre.

 

(j) Standardization is the actual determination of either the normality or the molarity of a solution.

 

(k) Primary Standard is the substance of known purity (‘AnalaR’-grade reagents) whose carefully weighed quantity helps in the standardization of an unknown solution (normality or molarity).

 

(l) Secondary Standard is another standard solution that is used for standardization of an unknown solution.

 

Example : An unknown solution of HCl may be standardized volumetrically in two ways, namely :

 

(i) by the help of ‘AnalaR’-grade Na2CO3 i.e., purity is known-‘Primary Standard’, and

 

(ii) by the help of another standard solution of NaOH—‘Secondary Standard’.

 

(m) Titer : is the weight of a substance chemically equivalent to 1 ml of a standard solution.

 

Example : 1 ml of 1 N HCl contains 0.03646 g (i.e., 0.001 equiv or 1 meq) of HCl and hence is chemically equivalent to 0.04000 g (i.e., 0.001 equiv or 1 meq) of NaOH.

 

Thus, most calculations in volumetric determinations (titrimetry) are enormously facilitated by using titer values.

 

For instance, in the offcial procedure for the assay of tartaric acid, it is stated that ‘Each millilitre of 1 N sodium hydroxide is equivalent to 75.04 mg of C4H6O6’. The C4H6O6 titer of 1 N sodium hydroxide is, therefore, 75.04 mg/ml, a value that may be calculated as follows :

 

An examination of the equation indicates that 1 mole or 150.09 g of 


is 2 equiv, and the equivalent weight of H2C4H4O6 is 75.04 g. Hence, each millilitre of 1 NaOH contains 0.001 equiv of NaOH and is equivalent to 0.001 equiv or 0.001 × 75.04 = 0.07504 g or 75.04 mg of H6C4O6.


Chapter: Pharmaceutical Drug Analysis: Theory and Technique of Quantitative Analysis

Volumetric Apparatus

In the broader sense, volumetric apparatus may be divided into two categories, namely : (a) To deliver a definite volume of liquid, and (b) To contain a definite volume of liquid.

VOLUMETRIC APPARATUS

 

As we have seen that the volumetric analysis essentially requires the precise and accurate measurement of weights and volumes of interacting solutions. However, the weights are measured upto the fourth place of decimal by using a manually operated good analytical balance or a single-pan electrical balance that need to be calibrated periodically with the help of a standard weight box.

 

In the broader sense, volumetric apparatus may be divided into two categories, namely :

 

(a) To deliver a definite volume of liquid, and

 

(b) To contain a definite volume of liquid.

 

1. Volumetric Apparatus Meant to Deliver a Definite Volume of Liquid

 

The two specific volumetric apparatus meant to deliver a defnite volume of liquid are burettes and pipettes which will be discussed very briefly below :

 

1.1. Burettes

 

Various official compendia specifies a standard temperature (°C) for glass volumetric apparatus as mentioned hereunder :

 

Pharmacopoeia of India (IP) : 27°C ;

 

United States Pharmacopoeia (USP) and National Formulatory (NF) : 25°C ;

 

National Bureau of Standards (NBS) : 20°C.

 

A burette is a graduated glass tube of uniform bore throughout the entire length, used for the accurate delivery and measurement of variable volumes of liquids. Burettes are graduated into millilitres (ml) and 1/10 millilitres (0.1 ml) and are made of varying capacity ranging from 1 ml to 100 ml ; however, the most common size is the 50 ml burette that is used invariably and conveniently for most volumetric titrations. They are usually closed at the bottom either by a Teflon or glass stopcock to monitor and control the outflow of liquid.

 

Specifications : The design, construction and capacity of volumetric glassware must be in accordance with those laid down by the Indian Standards Institution (ISI). The tolerances on capacity for burettes, as speci-fied in the relevant Indian Standards Institution, specifications are given in Table 2.1.


British Standards Institution (B.S. 846 : 1962) has laid down specifications for burettes and these are produced to either Class ‘A’ or Class ‘B’ specifications. All Class ‘A’ and a few of Class ‘B’ burettes have graduations that extend right round the barrel (or stem) of the burette to minimise errors due to parallax while taking the exact burette reading. It may be noted that Class ‘B’ burettes are normally graduated on one side only. Permitted tolerances on capacity for burettes used in common practice are stated in Table 2.2.


In fact, the tolerance actually represents the maximum error allowed at any point and also the maximum difference allowed between the errors at any two points. For instance, a tolerance of ± 0.05 ml signifies that the burette may have an error at any point by ± 0.05 ml, provided that the difference between the errors at any two given points does not exceed 0.05 ml.

Burettes calibrated at 20°C and 25°C deliver different weights of water for each 10 ml, when weighed with standard brass weights in air at 50% relative humidity (RH) at standard atmospheric pressure, as given below :


Hence, the true volume for each 10 ml segment of the burette can be calculated from the weights obtained and recorded on a convenient chart.

 

Leakage : A burette must be tested for any sort of leakage before putting it into operation. Teflon stopcocks are usually adjusted by a knurled nut for perfect use. Glass stopcocks may require a small quantity of a special type of grease or lubricant to allow both ease of operation and to check leakage.

 

Outlet Tip : From a practical point of view the outlet tip of either types of burette, i.e., having Teflon or glass stopcocks, must be of such diameter and taper as to allow the delivery of a single drop whose volume is significantly less than that which can be held between any two finest graduations of the scale with which the burette is calibrated.

 

Use of the Burette : The following steps are usually observed while operating a burette, namely :

 

(i) Burette tap is neatly lubricated with a thin-film of grease,

 

(ii) Rinse the burette, before putting it into operation, at least twice with small volumes of the solution (titrant), say about 5.0 ml, carefully draining out the solution between the addition of each portion,

 

(iii) Pour the solution into the burette until the former is little above the zero mark,

 

(iv) Open the burette tap slowly to fill up the tip of the burette and to expel all air bubbles,

 

(v) With the zero at eye-level carefully, drain out the liquid until the lower part of the meniscus is either at level or just below the Zero mark,

 

(vi) Remove the drop on the tip of the burette by just touching rapidly against the inner-neck of a flask or a porcelain tile,

 

(vii) The Class ‘B’ burettes should be read at level so as to avoid errors due to parallax,

 

(viii) To assist easy and accurate observation of the meniscus (lower for colourless solutions and upper for coloured solutions) it is always advisable to hold a piece of white paper behind the burette at the appropriate level,

 

(ix) Burette readings may be recorded to the nearest 0.02 ml, and

 

(x) Once a titration is completed, 15 seconds duration should be allowed to elapse before the final read-ing is made, to allow for drainage.

 

1.2. Pipettes

 

The pipette is the second volumetric apparatus that is meant to deliver a definite volume of liquid. Pipettes are of two types, namely :

 

(i) Transfer Pipettes : They have only one specific mark engraved on them and are specifically employed to deliver (or transfer) a definite volume of liquid under certain specified conditions, and

 

(ii) Graduated Pipettes : They have graduated stems and are used to deliver different small volumes as needed. However, they are not normally used for measuring very exact volumes of liquids.

 

The tolerances on capacity for pipettes, as specified by the Indian Standards Institution (ISI), are stated in Tables 2.3 and 2.4.


The British Standards Institution (BSI) has laid down the permitted tolerances and delivery times for commonly used bulb transfer pipettes as shown in Table 2.5.


The USP specifies the following tolerances accepted by the National Bureau of Standards for transfer pipettes :


The salient features of single-graduation mark transfer pipettes are :

 

(a) Capacity, temperature at which it was graduated (Ex) and reference to delivery time in seconds is stated on the bulb e.g., BOROSIL 1552 25 secs ‘A’ Ex 20 ml 20°C BS 1583.

 

(b) Class ‘A’ pipettes do mention the delivery time,

 

(c) Drainage time is specified, though an additional waiting time of 3 seconds after apparent cessation of flow is still important.

 

Note : The stated times apply only for water and aqueous solutions.

 

Use of the Transfer Pipette : The following steps mentioned sequentially must be followed while making use of a transfer pipette :

 

·              Always rinse the pipette with DW before use and allow it to drain as completely as possible,

 

·              Droplets of water remaining in the tip must be removed by touching against filter paper; and at the same time wipe out the outer surface of the pipette to prevent dilution of the solution to be pipetted,

 

·              Rinse the pipette with 2 to 3 small portions (5 ml) of the solution and drain out the liquid completely,

 

·              Gently suck the liquid up into the pipette a little above the single graduation mark and quickly shut the upper end of the pipette with the tip of the index finger. Now, remove the pipette from the stock solution and carefully wipe out the outer surface of the stem free from any liquid adhering to it. Hold the pipette vertically and keeping the graduation mark at the eye-level, slowly release the pressure on the index finger until the bottom of the meniscus just coincides with the graduation mark. Maintain sufficient pressure on the index-fnger so as to check any escape of liquid from the pipette, and quickly get rid of the drop attached to the tip by gently touching against a porcelain tile. Put the pipette into the receiving container, and permit the liquid to drain out with the tip of the pipette touching the inside of the container at an angle of 60°, taking care that the tip must not be dipping into the deliv-ered liquid. After all the solution has drained out, hold the pipette in this position for at least 3 seconds (waiting time), and then remove the pipette.

 

Note :

·              The National Physical Laboratory (NPL) describes a method of reading meniscus in graduated glassware, viz., a dark horizontal line on a white background is placed 1 mm below the meniscus. A slight adjustment of the position of the dark line causes the meniscus to stand out sharply against the white background,

 

·              The small drop of liquid that remains in the tip of the emptied pipette is taken into account while doing the calibration, and hence, it must not be added to the delivered liquid by blowing down the pipette.

 

·              Liquids having more viscosity and much larger surface tension than water must be provided with adequate draining time e.g., strong solution of iodine.

 

·              Presently, many analysts make use of pipette filler for sucking in and draining out of liquids from the transfer pipettes for obvious reasons.

 

Automatic Pipettes (Transfer Pipettes) : Automatic pipettes are always preferred to ordinary transfer pipettes because of their ability to handle corrosive and toxic liquids in routine analytical laboratories, e.g., determination of Iodine Value in edible oils by iodine-monochloride (ICl) solution.

 

The automatic pipette (Figure 2.1) dispenses a stated volume of liquid when filled with liquid used in the assay from tip (B) to tip (C) and is allowed to drain out in the normal manner. D is connected to an aspirator which is placed above the pipette so as to enable the solution to flow under gravity.


 

Operation of the Automatic Pipette : The automatic pipette may be operated by observing the following steps in a sequential manner :

                   

·              Turn the two-way tap clockwise to open so that the solution starts flowing into the pipette.

 

·              After about 5.0 ml has run into the pipette, turn A clockwise through 180°, so that solution now flows from the pipette to fill the delivery tube B.

 

·              As soon as B is full upto the tip, again turn A clockwise through 180°, so that the body of the pipette is filled completely to the top-tip.

 

·              Close the tap A by turning clockwise through 90° when the solution starts to overflow at C.

 

·              The pipette is now full from the lower-tip to the upper-tip and is ready for operation.

 

·              Remove the drop of solution from tip B, run out and drain for 15 seconds in the usual way.

 

2. Volumetric Apparatus Meant to Contain a Definite Volume of Liquid

 

The two particular volumetric apparatus meant to contain a definite volume of liquid are volumetric flasks (also known as measuring or graduated flasks) and measuring cylinders (also known as graduated cylin-ders) which will be discussed here briefly :

 

2.1. Volumetric Flasks (Syn. Measuring Flasks or Graduated Flasks)

 

Volumetric flasks are normally round or pear-shaped, flat-bottomed; having a long-neck, which possesses a single graduation mark round the neck.

 

Flasks bearing one graduation mark, are meant to contain specified volume of liquid at 20°C, when the lower part of the meniscus coincides with the mark and are known as volumetric flasks.

 

The long and narrow neck of uniform diameter affords as a measure of accurate adjustment, since the height of the liquid is sensitive enough to small variations of volume.

 

Units of Capacity

 

Litre—is defined as ‘the volume occupied by one kilogram of water at its temperature of maximum density (4°C) and subjected to normal atmospheric pressure’. The litre is considered as the standard unit of volume for all volumetric measurements.

 

The cubic centimetre is the volume occupied by a cube of which each side is 1 cm in length, and thus, 1 litre equals 1000.028 c.c. Therefore, it follows from here that the millilitre and cubic centimetre are not the same, though the difference is quite negligible. Hence, all volumetric apparatus is universally standardized in millilitres.

 

Standard Calibrations

 

The Indian Standard Institution (ISI) has laid down the tolerances on capacity of volumetric flasks (with different capacities) calibrated at 27°C as stated in Table 2.6.


The United States Pharmacopoeia (USP) requirements for volumetric flasks calibrated to contain the indicated volume at 25°C are given in Table 2.7.


The British Standards Institution (BSI) and the National Physical Loaboratory (NPL) have laid down the tolerances in the capacity of volumetric flasks (i.e., measuring flask) at 20°C by two sets of toler-ances viz., Grade ‘A’ and Grade ‘B’ respectively, evidently to indicate the class of accuracy to which the flask has been subjected to for graduation, followed by the manufacturer’s name and finally the BS standard number. However, the permitted tolerances for volumetric flasks commonly used in analytical laboratories are de-picted in Table 2.8.


Preparation of Standard Solutions

 

A reasonably well established analytical laboratory requires a number of standard solutions for its routine as well as specific assays. Therefore, it necessitates to know the intricacies of preparing the standard solutions as detailed in the following steps :

 

·              Transfer the requisite quantity of the accurately weighed pharmaceutical substances or solid quantitatively into a beaker and dissolve it in either distilled water (DW) or other specified solvent,

 

·              Pour the resulting solution quantitatively, into the funnel placed in the mouth of the volumetric flask with the help of a glass rod and a sharp jet of water from a wash-bottle by holding the beaker with the right hand and the guiding rod with the left hand,

 

·              Wash down the contents of the beaker through the funnel by means of the glass rod and the jet of DW. Repeat the process several times till the flask is 2/3rd full,

 

·              Remove the funnel, swirl the contents of the volumetric flask and make up the volume upto the mark,

 

·              Final adjustment of the volume must be made with the help of a teat pipette by adding DW/solvent dropwise. In doing so, adequate care should be taken to allow suffcient time for water/solvent to drain-down the inside of the neck of the flask, and

 

·              Finally shake the contents of the flask thoroughly for 2 to 3 minutes to obtain a perfect homogeneous solution.

 

Note :

 

(i) For precise work, the temperature of the solution must be adjusted to 20°C before making the volume upto the mark,

 

(ii) Standard solutions are usually stored in stock-bottles,

 

(iii) Ensure before any transfer is actually affected that the receiving vessel must be rinsed with at least 2 to 3 successive small quantities of the solution, and

 

(iv) When a standard solution is used a while after preparation, the contents of the stockbottle must be shaken thoroughly before any solution is withdrawn, thereby the condensed droplets of water collected on the inside neck of the container gets mixed with the main bulk of the solution.

 

2.2. Graduated Cylinders

 

The graduated cylinders are also referred to as the measuring cylinders among volumetric apparatus meant to contain a definite volume of liquid. Measuring cylinders are containers either unstoppered or stoppered having a wide range of capacities varying from 5 ml upto 2000 ml (2 Litres). In usual practice, the smaller cylinders upto 100 ml are normally graduated either in fractions of a millimitre or in millilitres. On the contrary, the large cylinders are graduated in units of 2, 5, 10, 20, or 50 ml, as per their specific size and volume. However, it is pertinent to mention here that measuring cylinders are used in a broader sense for measuring volumes of solution when only approximate volumes are needed.

Chapter: Pharmaceutical Drug Analysis: Theory and Technique of Quantitative Analysis

Volumetric Analysis: General Cosiderations

(a) Cleaning of volumetric apparatus, (b) Calibration of volumetric apparatus, and (c) Effect of temperature on volumetric measurement.

GENERAL COSIDERATIONS

 

Volumetric apparatus invariably used in titrimetric assays, meant either to deliver a definite volume of liquid viz., burettes and pipettes, or to contain a definite volume of liquid viz., volumetric flasks and measuring cylinders, have essentially the following three cardinal general considerations, namely :

 

(a) Cleaning of volumetric apparatus,

 

(b) Calibration of volumetric apparatus, and

 

(c) Effect of temperature on volumetric measurement.

 

These three aspects will be discussed briefly hereunder :

 

1. Cleaning of Volumetric Apparatus

 

New as well as used volumetric apparatus, namely : burettes, pipettes, volumetric flasks and measuring cylinders etc., employed in carrying out most of the pharmacopoeial assays should be extremely clean. It is particularly of great importance where small volumes of liquids are measured.

 

A positive evidence for a dirty apparatus may be sought by observing the adherence of droplets to the walls of a burette or pipette. However, in a clean volumetric apparatus, the liquid drains down quite uniformly thereby wetting the walls so that no droplets are visible to the naked eye.

 

A few very effective cleaning fluids that are used in good analytical laboratories are, namely :

 

(i) Chromic Acid Mixture,

 

(ii) Synthetic Detergent Solutions (or Alkaline Cleansing Agents), and

 

(iii) Teepol.

 

1.1. Chromic Acid Mixture

 

Materials Required : Sodium dichromate : 200 g ; Sulphuric acid : 1500 ml.

 

Procedure : Weigh 200 g sodium dichromate and transfer to a 2 Litre hard-boroslicate glass beaker. Dissolve it in 100 ml of water and cool in an ice-bath to about 10-15°C. Now, add to it 1500 ml of sulphuric acid (36 N) in small bits at intervals with constant stirring. Chromic acid mixture is extremely corrosive and hygroscopic and must be stored in closed glass-stoppered bottles.

 

Precautions :

 

(i) Chromate solution should be chilled before addition of H2SO4,

 

(ii) Safety goggles should be worn during the addition of the acid,

 

(iii) In case, a green colour develops, discharge the mixture into a sink with continuously flowing water,

 

(iv) Chromic acid must not be used for cleaning calibrated containers employed for optical measure-ments,

 

(v) Glass apparatus washed with chromic acid mixture must be subjected to adequate prolonged rinsing because glass (silicates and borosilicates) have a tendency to absorb the chromic acid,

 

(vi) Hot solutions should be avoided when cleaning accurately calibrated apparatus, due to the production of a permanent change in volume caused by heat known as thermal aftereffect,

 

(vii) All volumetric glasswares must be finally rinsed with purified water (distilled water) before use for analytical purposes.

 

1.2. Synthetic Detergent Solutions (or Alkaline Cleansing Agents)

 

Detergents are synthetic cleansing agents used with water. The most commonly used anionic surfactants containing carboxylate ions are known as soaps which are generally prepared by the saponification of natural fatty acid glycerides in alkaline solution. Usually a 2 to 5% (w/v) solution of a good detergent powder in water serves as a reasonably effective cleansing agent.

 

1.3. Teepol (or Gardinol Type Detergents)

 

It is a mixture of the sodium salts of sulphated fatty alcohols made by reducing the mixed fatty acids of coconut oil or cottonseed oil, and fish oils. Sometimes natural waxes such as spermaceti, wool fat and bees wax are sulphated directly.

 

A 1 to 3% (w/v) solution of Teepol in water may also serve as a good cleansing agent for the removal of stubborn deposits and stains present in glass apparatus.


Chapter: Pharmaceutical Drug Analysis: Theory and Technique of Quantitative Analysis

Technique of Volumetric Analysis

Following are the various steps that need to be observed carefully so as to achieve reasonably correct and reproducible results in the volumetric titrations :

TECHNIQUE OF VOLUMETRIC ANALYSIS

 

Following are the various steps that need to be observed carefully so as to achieve reasonably correct and reproducible results in the volumetric titrations :

 

1)          Conical flasks are considered to be the most suitable vessels meant for volumetric titrations because the mixing can be performed quite rapidly, easily and safely by gently swirling the contents of the flask during the titration,

 

2)          Beakers are not usually preferred, but in case they are to be used in volumetric analysis, following two provisions may have to be made for stirring :

 

·              use of a magnetic stirrer with a magnetic guide for the solution, and

 

·              use of a stirring rod,

 

3)          The titration container or vessel must always be kept polished so as to view the end point vividly,

 

4)          The solution under titration is normally viewed against a white background e.g., white tile or white paper,

 

5)          When the end point is being approached it is always advisible to have the drops of titrant split. It can be accomplished by opening the stopcock of the burette in such a manner that only a fraction of a drop flows out and remains adhered to the tip of the burette. Touch of the liquid against the inside of the flask and wash it down into the main bulk of the liquid with a fine jet of DW (from a wash-bottle),

 

6)          In a situation, where the colour-change at the end-point is rather gradual and not abrupt, it is always useful to have a comparison-solution readily available,

 

Example : Methyl orange offers a gradual end-point. Hence, two flasks containing the same volume of solution having approximately the same composition as the liquid being titrated may be prepared; first, slightly acidic—Red solution, second, slightly basic—Yellow solution.

 

7)          In fact, these carefully-prepared comparison solutions would ultimately help in deciding the colour change thereby confirming the actual end-point without any controversy, whatsoever,

 

8)          All titrations must be carried out in triplicate and the results of two concurrent readings (i.e., whose difference falls within 0.05 ml-based on a 20 ml titration) may be taken into consideration,

 

9)          Remainder solution in the burette, after titrations have been performed must be rejected and should not be put back to the stock-bottle for obvious reasons of contamination. The burette in operation is then washed thoroughly with DW and allowed to drain by placing it up-side down on a burette stand.


Chapter: Pharmaceutical Drug Analysis: Theory and Technique of Quantitative Analysis

Biomedical Analytical Chemistry

This particular aspect of analytical chemistry is the outcome of the unique amalgamation of the principles and techniques of analytical chemistry and biochemistry and was initially termed as ‘clinical chemistry’.

BIOMEDICAL ANALYTICAL CHEMISTRY

This particular aspect of analytical chemistry is the outcome of the unique amalgamation of the principles and techniques of analytical chemistry and biochemistry and was initially termed as ‘clinical chemistry’ but is more recently and more descriptively known as ‘biomedical analytical chemistry’.

 

Presently, both serum and urine assays are being used extensively in diagnostic medicine which evidently signifies that the pharmacist of today should be fully conversant with the ever-increasingly important techniques of biomedical analytical chemistry. It is, however, necessary to make a passing reference to microbiological assays and haematological assays, also being carried out in a clinical laboratory, though it should not be treated under this topic since these methods are outside the scope of biomedical analytical chemistry.

 

Classical example of SGOT-PAS episodes : Patients suffering from tuberculosis (TB) when diagnosed with para-aminosalicylic acid (PAS) invariably showed elevated serum levels of the intracellular enzyme serum-glutamic-oxaloacetic-transaminase (SGOT) which was initially considered and treated as a drug-induced hepatic toxicity. Later, an extensive and intensive studies revealed this to be an absolutely false diagnosis. In fact, the apparent enhanced SGOT levels were actually caused on account of the interference of PAS in the SGOT assay.

 

In the same vein, such analytical and biochemical interferences with respect to drug interference in various biomedical assays are being profusely cited in current scientific and research journals, such as the American Journal of Hospital Pharmacy and Clinical Chemistry.

 

It has been established beyond any doubt that analytical interferences can only take place when a drug or its resulting metabolite happens to interfere with the analytical method adopted for the assay.

 

In order to have a comprehensive account on the various aspects of ‘Biomedical Analytical. Chemistry’, we may have to study the following four methods of assay with specific emphasis on their principle and applications, namely :

 

(a) Colorimetric Assays,

 

(b) Enzymatic Assays,

 

(c) Radioimmunoassays, and

 

(d) Automated Methods of Clinical Analysis.


Chapter: Pharmaceutical Drug Analysis: Theory and Technique of Quantitative Analysis

Biomedical Analytical Chemistry: Colorimetric Assays

Theory : In fact, two fundamental laws actually govern the practice of colorimeteric assays of photometry.

COLORIMETRIC ASSAYS

 

A. Theory : In fact, two fundamental laws actually govern the practice of colorimeteric assays of photometry.

 

First Law : Bougner’s (1729) or Lambert’s (1760) Law defines that—“when a beam of monochromatic light, previously rendered plane-parallel, enters an absorbing medium at right angles to the plane-parallel surfaces of the medium, the rate of decrease in radiant power with the length of light path through the absorbing medium `b’ is directly proportional to the radiant power of the beam, i.e., the light will be diminished in geometric (not arithmetic) or exponential progression”.

 

Alternatively, it may be explained that if a particular thickness absorbs half the light, the thickness which follows the first half and is equal to it will not absorb the entire second half, but instead only half of this half and will consequently reduce it to one-quarter. Thus, we have :


Upon integration and changing to logarithms of base 10, and substituting P = P0 when b = 0, we may get :

2.303 log (P0/P) = kb  ... (b)

In other words, the radiant power of the unabsorbed light decreases exponentially as the thickness of the absorbing medium increases arithmetically,

P = P0 e–kb = P0 10–0.43 kb...(c)

Second Law : Bernard’s (1852) or Beer’s (1852) Law defines that—‘the radiant power of a beam of parallel monochromatic radiation decreases in a similar manner as the concentration of the light-absorbing constituent increases”. Thus we have :

 

2.303 log (P0/P) = k C   ... (d)

where, C = concentration of substance, and

 

k = constant of proportionality.

 

Therefore, from Eq. (b) and Eq. (d), the two Laws may be combined and expressed with a single constant as follows :

 

log (P0/P) = abc    ... (e)

or P = P0 10abc     ... (f)

where, a = absorptivity constant*.

 

[* and not to be tenned as absorbancy index, extinction coeffcient or specific extinction.]

 

In fact, the absorptivity constant ‘a’ is dependent upon the wavelength of the radiation as well as the nature of the absorbing material, whose concentration ‘C’ is usually expressed in grams per litre.

 

Molar Absorptivity () : It is the product of the molecular weight of the substance and its absorptivity and is designated by the symbol .

 

Beer’s Law (or Beer-Lambert’s Law) : The combined law is invariably referred to as ‘Beer’s Law’, while some texts refer to this as ‘Beer-Lambert’s Law’.

 

Eq. (f) is mostly expressed as shown below :

 

          A = abc       ...(g)

where, A = absorbance,

 

a = absorptivity,

 

b = optical path length, and

 

c = analyte concentration.

 

The term A1%1cm designates the absorbance of a 1 cm layer of solution that essentially contains 1% by weight of absorbing solute.

 

It is pertinent to mention here that most of the pure pharmaceutical substances (RS) do possess a definite characteristic absorbance (i.e., A1%1cn ) that forms the basis of their assay vis-a-vis the unknown sample.

 

Beer’s Plot : It is a plot of the absorbance value (along Y-axis) against a series of unknown solute concentrations in g/litre (along X-axis) thereby yielding a straight line passing through the origin.

 

Therefore, the solute-concentration present in an unknown solution can be estimated conveniently from the Beer’s Plot or sometimes referred to as the Standard Curve, merely by measuring the absorbance value of the solution and then finding the concentration value that corresponds to the measured absorbance value as is illustrated in the following Figure 2.2.


The colorimetric assay of sulphadiazine is based on the acid-catalysed equilibrium reaction that occurs between vanillin (an aldehyde) and sulphadiazine (an arylamine). The chemical species that forms as shown below is known as the Schiff’s Base and is yellow in colour.


Transmittance. The relationship between per cent transmittance and concentration is shown in Figure 2.3.


From Figure 2.3, it is quite evident that at lower concentrations the per cent trasmission is high and is vice varsa at higher concentrations.

 

However, a direct relationship between per cent transmittance and absorbance is illustrated in Figure 2.4.



B. Applications in Biomedical Analytical Chemistry Colorimetric assays have a wide spectrum of applications in biomedical analytical chemistry which may be categorized under the following four heads, namely :

 

(i) Colorimetric Assays of Biochemicals,

 

(ii) Colorimetric Assays Involving Complexation Reactions,

 

(iii) Colorimetric Assays Involving Redox Reactions, and

 

(iv) Colorimetric Assays of Enzyme Levels.

 

All these four categories of colorimetric assays shall be discussed briefly with appropriate examples, wherever necessary, to have an indepth knowledge and better understanding of the practical aspects.

 

1. Colorimetric Assays of Biochemicals

 

In this context, the discussion shall be restricted to the colorimetric assays of urea (BUN), bilirubin and cholesterol. However, the clinical significance of these substances and the extent to which they are present in biological fluids; besides the various drugs that usually interfere with their assay are also described adequately in the following pages :

 

1.1. Urea (BUN)

 

The extent of urea (BUN) present in biological fluids is normally determined in many Auto Analyzers by the following method :

 

The quantity of substance having an unknown structure is determined at 520 nm spectrophotometrically, while the normal BUN level is determined by averaging the BUN levels of a number of normal subjects.


Profile of BUN-levels

·              normal BUN level is 10-15 mg per 100 ml,

 

·              Enhanced BUN levels clearly signify a renal dysfunction, for instance urinary tract obstruction and nephritis i.e., inflammation of the kidney.

 

·              Increased incidence of BUN is also found in subjects suffering from diabetes, hepatic disorders and gastrointestinal disturbances,

 

·              Decreased BUN level is usually indicative of acute hepatic dysfunction and excessive dehydration,

 

·              A few important drugs, namely : thiazide diuretics (e.g., chlorothiazide, hydroflumethiazide, bendroflumethiazide, benzthiazide, cyclothiazide etc.), neomycin, tetracyclines, methyldopa etc., help in enhancing the BUN levels perhaps due to interference with normal renal function,

 

·              Phenothiazines (e.g., promethazine, chlorpromazine, ethopropazine etc.) on the contrary causes a significant decrease in BUN levels due to lowering of urea production from the liver, and

 

·              Substances that are inherently present in the serum and absorb at 520 nm shall interfere with these measurements, and therefore, necessary corrections for these materials have got to be made adequately.

 

1.2. Bilirubin

 

Bilirubin is diazotized with para-sulphonyl benzene diazonium compound and the absorbance of the resulting azobilirubin is measured at 600 nm to determine bilirubin level in the biological fluid e.g., blood serum. In usual practice, a serum blank is run simultaneously by reacting the serum with caffeine, sulphanilic acid and tartaric acid, and the absorbance of the blank is measured at 600 nm which is subsequently subtracted from the azobilirubin absorbance initially obtained before the bilirubin level is finally determined.


Profile of Bilirubin Levels

 

·              Normal bilirubin level ranges between 0-1.5 mg per 100 ml,

 

·              Enhanced bilirubin level may suggest drug toxicity, bile-tract obstruction, hepatitis and hepatic dysfunction,

 

·              As normal bilirubin level commences from zero, hence conditions responsible for its decrease are practically non-existent,

 

·              Increased bilirubin levels are caused due to the intake of large doses of such drugs as : chloroquine, vitamin K, sulpha-drugs, tetracyclines, paracetamol, nicotinic acid and monoamine oxidase inhibi-tors (e.g., iproniazid RP 1.0 ; nialamide RP 1.8 ; isocarboxazid RP 3.1 ; phenelzine RP 18 ; pheniprazine RP3l ; and tranylcypromine RP 45), where RP designates the ‘Relative Potency’ based on the tryptamine potentiation test. The elevated levels are due to hepatic injury, and

 

·              Drugs that interfere with the assay are, namely : (a) phenylazopyridine hydrochloride—a coloured drug, (b) azo-compound forming medicinals, and (c) degradation product of novobiocin.

 

1.3. Cholesterol

 

Cholesterol interacts with glacial acetic acid and acetic anhydride to result into the formation of a coloured product whose absorption is measured at 630 nm and this is found to be directly proportional to the level of cholesterol present in the serum. The reaction may be expressed as follows :


The above reactions is also referred to as the Libermann’s Reaction.

 

Profile of Cholesterol Levels

 

·              Normal total cholesterol level is 200 mg per 100 ml,

 

·              Increased cholesterol levels in serum are found in patients suffering from chronic hepatitis, atherosclerosis (deposit of fat in arteries of heart) and hypothyroidism,

 

·              Decreased cholesterol levels in serum is indicative of liver ailment and hyperthyroidism,

 

·              Corticosteroids (i.e., steroidal compounds) found in urine that possess biological properties resembling those of adrenal cortical extract, either in the increase or decrease of cholesterols levels,

 

·              Oestrogens, for instance : estrone, estriol, estradiol etc., are found to lower the cholesterol levels,

 

·              The broad-spectrum antibiotic chlorotetracycline and the aminoglycoside antibiotic kanamycin are observed to lower the cholesterol levels by forming salts with bile acids (e.g., cholic acid, deoxycholic acid and chenodeoxycholic acid) in the intestinal canal,

 

·              Likewise, the antoconvulsant phenytoin sodium and neomycin—an aminoglycoside antibiotic also decrease the cholesterol levels, and

 

·              Interestingly, penicillamine—a degradation product of penicillin and phenothiazines—the histamine H1—receptor antagonists, such as : promethazine teoclate, methadilazine hydrochloride, trimeprazine tartrate are found to increase the cholesterol levels.


Chapter: Pharmaceutical Drug Analysis: Theory and Technique of Quantitative Analysis

Biomedical Analytical Chemistry: Enzymatic Assays

Theory : All colorimetric enzymatic assays essentially involve the measurement of the activity of an ezyme under the following two circumstances, namely : (a) When substrate is in large excess, and (b) When enzyme concentration is in large excess.

ENZYMATIC ASSAYS

 

A. Theory : 

All colorimetric enzymatic assays essentially involve the measurement of the activity of an ezyme under the following two circumstances, namely :

 

(a) When substrate is in large excess, and

 

(b) When enzyme concentration is in large excess.

 

A.1. Substrate Present in Large Excess : 

In reality, an enzyme reaction is nothing but a special kind of generalized reaction that may best be expressed as follows :

                     .....................(a)

 

Where, E = Enzyme,

            S = Substrate

          ES = Enzyme-substrate complex, and

          P = Product.

From Eq. (a), we have,

Rate of Product Formation = Vmax [S]/Km + [S]                                          ...(b)

 

Where, Km = (k2 + k3) / k1,

Vmax = Max. rate of reaction

Assuming, [S] to be in large excess [S] >> Km,

 

From Eq. (b) we have :

 

Rate of Reaction = Vmax [S]/[S]

or    Rate of Reaction = Vmax   ...(c)

 

Example : In order to measure the activity of an enzyme E, such as creatine phosphokinase (CPK), the concentration of the substrate S, for instance creatine, should be in large excesses so that the products measured shall be in the linear portion of the curve (Part ‘A’) in Figure 2.5.


Therefore, with a view to obtaining the best results, the two experimental parameters, namely : the temperature (constant-temperature-water-bath) and the time (phaser) should always be kept constant in order that the rate of reaction, as determined by the amount of product formed, specially designates the activity of the enzyme under assay, and devoid of the influence of any other variables on the reaction rate.

 

A.2. Enzyme Concentration in Large Excess

 

In order to analyze the quantity of substrate (S) present in a biological sample glucose oxidase is added in excess of the actual amount needed for the complete conversion of all the substrate to product ; and to achieve this object the reaction is allowed to run for a fairly long duration (i.e., to complete the reaction). It can be seen evidently in Part ‘B’ of Figure 2.5, wherein the sepecific reaction time the substrate (S) has been consumed completely and consequently, the concentration of the product achieves a maximum value.

 

1. Assay Methods

 

A few typical examples of colorimetric assay of enzyme levels will be discussed briefly hereunder :

 

1.1. Alkaline Phosphatase (AP)

 

Theory : 

Alkaline phosphatase is responsible for the cleavage of O-P bonds. It is found to be relatively non-specific and this characteristic permits the AP level to be assayed based on the fact that p-nitrophenylphosphate ion gets converted to p-nitrophenolate anion at pH 10.5; as expressed in the following reaction.


In actual practice, p-nitrophenylphosphate is present in large excesses, and the reaction is carried out at 38°C for 30 minutes. The resulting amount of p-nitrophenolate ion is estimated by the help of an usual standard curve employing known concentrations of p-nitrophenolate prepared from p-nitrophenol.

 

Bessey-Lowry Activity : 

One unit of activity may be defined as the amount of enzyme present in 1 millilitre of serum that liberates 1 μ mol of p-nitrophenol (0.1391 mg)* per hour at pH 10.5 after 30 minutes at 38°C.

 

Elimination of Interference due to Coloured Drugs

 

p-Nitrophenol is colourless, whereas the phenolate ion under basic conditions is yellow in appeanace. Therefore, the elimination of interference due to coloured drugs present in the serum is accomplished effectively by first, measuring the absorbance of the serum under basic conditions, and secondly, under acidic conditions. Thus we have :

Ap-nitrophenolate = Abasic – Aacidic

 

Profile of AP-levels

 

·              Normal AP-levels in adults range between 0.8 to 2.3 Bessey-Lowry units and in children between 2.8 to 6.7,

 

·              Increased AP-levels are observed in patients suffering from liver diseases, hyperparathyroidism and in rickets,

 

·              Decreased AP-levels could be seen in patients suffering from hypoparathyroidism and pernicious anaemia (i.e., an anaemia tending to be a fatal issue).

 

Interference due to Bilirubin

 

Bilirubin is eliminated by dializing the incubated p-nitrophenolate ion (at pH 10.5, and maintaining at 38°C for 30 minutes) into 2-amino-2-methyl-1-propanol, without carrying out the blank determination stated earlier.

 

There are a few medicinals that cause increased bilirubin levels which ultimately enhances AP-levels ; unless and until a corrective measure is taken in the respective procedure one may be left with false AP-level enhancement. Some typical examples are, namely : amitriptyline, chloropropamide, erythromycin, phenylbutazone, sulpha-drugs and tetracyclines.

 

Materials Required : 

0.01 M p-Nitrophenol (dissolve 140 mg of p-nitrophenol in 100 ml of DW) : 1.0 ml ; 0.02 N NaOH (dissolve 160 mg in 200 ml DW) : 200 ml ; 5 ml of alkaline-buffered substrate (l M p-nitrophenylphosphate) (dissolve 7.5 g glycine, 0.095 g anhydrous MgCl2 and 85 ml of 1 N NaOH to 1 litre with DW ; and mixing with an equal volume of a solution prepared by dissolving 0. l0 g of p-nitro-phenylphosphate in 25 ml of water) ; temperature bath previously set at 38°C ; alkaline phosphatase for unknowns (commercial source) ; working standard [dilute 0.50 ml of a solution of p-nitrophenol (10.0 mol/ litre, 0.139 g/100 ml) to 100 ml with 0.02 N NaOH].

 

Procedure :

 

(1) First of all prepare a standard calibration curve as per Table 2.9.


(2) Plot a graph of absorbance A Vs units of alkaline phosphatase per millilitre.

 

(3) Proceed for the assay of AP in the serum sample sequentially as follows :

 

(i) Pipette 1.0 ml of alkaline—buffered substrate into each of two test tubes and keep in a water-bath preset at 38°C,

 

(ii) When both the test tubes have attained the temperature equlibrium, add 0.10 ml of serum and water to these tubes separately. The one with water serves as a reagent blank and is always needed per set of unknowns. Now, put the two tubes for incubation for exactly 30 minutes period,

 

(iii) Enzyme activity is arrested by adding 10.0 ml of 0.02 N NaOH to each tube. Remove them from the water-bath and mix the contents thoroughly,

 

(iv) Read out the absorbance of the unknown tube at 410 nm against the ‘reagent blank’ tube,

 

(v) Transfer the contents from the cuvets to the respective test-tubes and add 0.1 ml of HCl ( ~ 11.5 N to each tube and mix the contents carefully. This operation removes the colour developed due to p-nitrophenol,

 

(vi) Again read out the absorbance of the serum sample against the reagent blank tube at 410 nm. This gives the colour due to the serum itself,

 

(vii) Now, the corrected reading is achieved by subtracting the reading obtained in step (vi) from the reading in step (v). The alkaline-phosphatase activity of the serum as Bessey-Lowery units is obtained from the calibration-curve step (i). Under these experimental parameters, we have :

 

1 Bessey-Lowry Unit = 5 × 10 –8 mol of p-Nitrophenolate anion.

 

Thus, one unit of phosphatase activity liberated 1 μ mol of p-nitrophenol (l μ mol = 0.1391 mg) per hour per millilitre of serum under specified conditions.

 

Note : In case, a value more than 10 Bessey-Lowry Units is obtained, it is always advisable to repeat the process either with a smaller volume of serum or a shorter incubation period, and then finally adjust the calculations accordingly.

 

(4) Report the concentration of AP in units per millilitre.

 

1.2. Lactate Dehydrogenase (LDH)

 
Theory : 

The method of LDH assay is based on kinetic analysis. In a kinetic enzymatic assay a unit of enzyme activity is defined as ‘the quantity of enzyme that brings about a certain absorbance increase in 30 seconds or 1 minute at a fixed temperature (for instance 25 ± 0.2°C) ’.

 

The kinetic assay of LDH is based on the conversion of lactic acid to pyruvic acid, in the presence of nicotinamide adenine dinucleotide (NAD), and is closely monitored at intervals of 30 seconds or 1 minute by measuring the increase in absorbance at 340 nm. In this particular instance lactic acid available in an excess to ensure that the increase in pyruvic acid is linear with time, i.e., directly proportional to time. The reaction involved may be expressed as follows :


The liberated nicotinamide-adenine-dinucleotide hydrogenase (NADH) has an absorption maxima at 340 nm, whereas lactic acid. NAD+ and pyruvic acid do not absorb at all at this wavelenath.

Temperature Correction Factor : 

The rate of the above reaction is temperature dependent. Hence, if the temperature (experimental) is higher or lower than that used to define a unit of activity, a definite correction factor should be applied as per Table 2.10.


From Table 2.10 it may be observed that :

 

(a) At a temperature beyond 25°C (Tf = 1.0), the absorbance increases at a faster rate than at 25°C due to enhanced rate of reaction and enhanced formation of NADH, thereby lowering the correction factor from 1.0 e.g., 0.80 at 28°C,

 

(b) At a temperature lower than 25°C the rate of reaction is slower than at 25°C, thereby increasing the correction factor from 1.0 e.g., 1.24 at 24°C, and

 

(c) Rule of thumb suggests that for each 10°C rise in temperature the reaction rate is almost doubled and the correction factor is halved, for example : at 35°C the correction factor is 0.47 (or 1.0/2 ~ 0.47).

 

Profile of LDH-levels :

 

1)          Normal LDH levels are as follows : Absorbance Units per ml : 42 to 130, International Units per ml : 0.20 to 0.063

 

2)          LDH level in serum is found to be increased in 8 to 10 hours after a myocardial infarction (i.e., development or presence of an infarct in the heart) ; obviously the heart muscle is destroyed and consequently the enzymes leak into the serum,

 

3)          Increased LDH levels are found in patients suffering from diseases related to liver and renal func-tions, cancer and pulmonary infarction,

 

4)          Drugs like codeine and morphine help in enhancing LDH levels.

 

Materials Required : 

Dermatube LDH provided by Worthington Biochemical, USA.

 

Procedure : 

The following steps need to be followed in a sequential manner :

 

1)          Dissolve the contents of Dermatube LDH (containing NADH and lactic acid) with 2.8 ml of DW,

 

2)          Put this solution in a cuvette and then insert it in a colorimeter previously warmed up to 25°C. Set the wavelength at 340 nm. Carefully adjust the absorbance of this solution to 0.1 by making use of the proper variable control as explained earlier,

 

3)          Remove the cuvette and add to it 0.2 ml of serum. Mix the contents of the cuvette and replace it quickly in position. Carefully record the absorbance exactly at intervals of 30 seconds for 2 to 3 minutes. In case, the absorbance happens to rise very rapidly, repeat step 3 by diluting 0.1 ml of the serum to 0.2 ml with DW,

 

4)          From the foregoing measurement of absorbances calculate an average A/min,

 

5)          Note the temperature at which the reaction is carried out accurately and then find out Tf from Table 2.10.

 

6)          Report the LDH concentration as follows :


Chapter: Pharmaceutical Drug Analysis: Theory and Technique of Quantitative Analysis

Radioimmunoassays (RIAS)

An assay method based on immunological antibody-hapten (Ab-Ha) reaction that makes use of a radioactive tracer is usually known as radioimmunoassay.

RADIOIMMUNOASSAYS (RIAS)

 

An assay method based on immunological antibody-hapten (Ab-Ha) reaction that makes use of a radioactive tracer is usually known as radioimmunoassay. A hapten (or haptene) is a small molecule that represents the portion of an antigenic molecule or complex which determines its immunologic specificity, for instance : cortisol ; whereas an antibody is a relatively large protein that is specific for certain haptens. An antibody is generated by binding the hapten to a protein, resulting into the formation of an antigen that specifically stimulates the immune system to produce antibodies specific for the hapten.

 

The assays that utilize protein instead of antibody are normally termed as competitive protein bind-ing assays. As an antibody is also a protein, therefore, a radioimmunoassay may be looked upon as a type of competitive protein binding assay.

 

Theory : 

Generally, a radioimmunoassay makes use of a radioactive hapten and subsequently the percent of radioactivity bound to the antibody is measured. The radioactivity is determined by the help of a Geiger-Müller Counter or Geiger-Counter or G-M Tube and sometime by a Scintillation Counter.

 

First of all, a ‘Standard Curve’ or a ‘Calibration Curve’ is plotted between the reciprocal value (i.e., 1 × % –1 radioactivity bound to the antibody) versus the amount of standard for a series of unknowns. Thus, the amount of hapten present in the unknown sample is measured from the plotted calibration curve conveniently.

 

The radioimmunoassay is based on the evolved competition between the combination of radioactive (Ha+) and nonradioactive (Ha) hapten to the antibody as represented below :


Let us assume that the binding constants for Ha+ and Ha are equal ; now, for a fixed quantity of Ha+ but an increased concentration of Ha. The ultimate impact would be that lesser Ha+ shall be bound. In actual practice, however, the use of Tritium (H3) or Carbon-14 (C14), which helps to render the Ha radioactive, ulti-mately maintains the equality of these binding constants, namely : KHa+ and KHa . It also confirms that the chemical properties of both radioactive (Ha+) and nonradioactive (Ha) entities are more or less the same as far as the antibody is concerned.

 

Salient Features of Radioimmunoassays

 

·              They belong to a class of extremely sensitive methods of analysis,

 

·              Sample required for assay is usually very small e.g., 1 ml of serum,

 

·              Concentrations upto the nanogram range i.e., 10–9 g can be measured accurately,

 

·              A large number of hormones and drugs which find their abundant usage in a bad way, namely :

 

·              cortisol (17-hydroxycorticosterone or hydrocortisone), insulin, morphine, barbiturates (sedatives), vitamin B12, digoxin and human growth hormones, such as : somatotropin (elaborated in the placenta),

 

·              Incidence of interferences observed in the radioimmunoassays are fairly insignificant by virtue of the highly specific hapten-antibody complexation reaction, and

 

·              Exceptions do occur when two 5-substituted barbiturates present together cannot be assayed by this method, obviously due to interference.

 

1. Cortisol (In Plasma)

 

Theory : 

Cortisol (or hydrocortisone) was introduced in the year 1951, for the treatment of rheumatoid arthritis. It has a significant effect on protein metabolism. It also exerts widespread effects on carbohydrates, lipid and protein synthesis (or anabolism). The cardinal side effects such as excessive potassium excretion and sodium retention, enhanced gastric acidity, oedema, psychosis and negative nitogen balance are some of the exaggerated manifestations of the normal metabolite functions of cortisol.

 

Most importantly, the determination of cortisol levels is considered useful in the diagnosis and treatment of various ailments, namely : Addison’s Disease i.e., pernicious anaemia—a condition whereby the maturation of the red cells may not proceed beyond the stage of megaloblasts; Cushing’s Syndrome.

 

Adrenal Tumours : 

The assay-method is entirely based on the Schwartz-Mann Kit. According to this method, cortisol is first extracted from the plasma using CH2Cl2 (methylene chloride). In the actual radioimmunoassay the cortisol present in the extract competes with Cortisol-H3 (i.e., the radioactive tracer) for the common binding sites on transcortin, which is incidently not an antibody but a cortisol-binding protein. Now, the free cortisol is quantitatively removed by adsorption on dextran-coated charcoal from the one bound to the transcortin. Finally, the bound radioactivity (due to Cortisol-H3) is measured which is then employed to calculate exactly the amount of cortisol present in the sample by the help of a Standard Curve (or Calibration Curve).

 

Materials Required : 

Schwartz-Mann-H3 Cortisol RIA-Kit ; liquid scintillation counter, centrifuge.

 

Procedure : 

The various steps to be followed sequentially for the assay of cortisol in plasma are as follows :

 

·              The cortisol is usually extracted from the samples drawn from the patients, as follows :

 

Place 100 μ l of plasma in each of two tubes and add 2.5 ml of methylene chloride. Shake the contents of the tube vigorously for 10 minutes and transfer 0.5 ml of clear extract (i.e., the lower layer) to another tube. Evaporate the methylene chloride either at 35°C in an oven or in a stream of N2. The extract thus obtained is employed in the following step.

·              The following steps viz., Step 1 to Step 15, related to the procedure for the assay and the calibration curves must be performed simultaneously :



·              Results : Average the counts per minute in vials 3 and 4. This is the blank value. Now, subtract the blank from all other counts per minute to obtain the actual counts per minute and average the counts per minute for vials 1 to 2 to find the total count per minute. The percent bound may be calculated using the following expression :


Finally, plot the percent bound Vs nanograms (ng) per tube of cortisol standard either on linear or on semilog paper and make use of this Standard Curve to calculate the amount of cortisol present in the unkown samples.


Chapter: Pharmaceutical Drug Analysis: Theory and Technique of Quantitative Analysis

Automated Methods of Clinical Analysis

Theory : An ‘Autoanalyzer’ serves as the most versatile and important instrument in a well-equipped ‘clinical laboratory’ that caters for the rapid screening of serum levels for upto forty (40) important chemical substances in the field of diagnostic medicine.

AUTOMATED METHODS OF CLINICAL ANALYSIS

 

Theory : An ‘Autoanalyzer’ serves as the most versatile and important instrument in a well-equipped clinical laboratory’ that caters for the rapid screening of serum levels for upto forty (40) important chemical substances in the field of diagnostic medicine. These autoanalyzers may be either ‘Single Channel’ i.e., per-forming only one determination on each sample or Multichannel’ i.e., carrying out several different determinations on each sample.

 

A few important substances that are routinely analyzed in a clinical laboratory with the aid of an ‘Autoanalyzer’ are, namely : serum-glutamic-oxaloacetic transaminase (SGOT) ; creatine-phophokinase (CPK); alkaline-phosphatase (AP) belonging to the class of enzymes ; and a host of biochemical substances, for instance : bilirubin, serum albumin, blood urea nitrogen (BUN), uric acid, creatinine, total protein, glucose, cholesterol, besides a few common inorganic ions, such as : Cl, Ca2+, K+, Na+.

 

The basic principles underlying both automated and unautomated methods of analysis are more or less the same. Out of the broad-spectrum of biological samples blood analysis is the most common one. There exists a number of parameters which may be assayed, and spectrophotometry is ideally suited for nearly all of them, a few typical examples are cited in Table 2.11.


Explanation : Glucose (having an aldehyde functional moiety) reduces Cu2+ to Cu2O (i.e., Cu+) as per the following reaction :


As some other sugars are also present in blood sample, and besides the above reaction not being abso-lutely stoichiometric, it has become necessary in actual practice to establish an emperical calibration curve using known concentrations of glucose. The above reaction is allowed to proceed for exactly 8 minutes at 100°C. To the resulting solution phosphomolybdic acid is added, which is subsequently reduced by Cu2O to give rise to an intensely coloured ‘molybdenum blue’ that is measured at 420 nm accurately.

 

Alternatively, glucose forms a specific complex with o-toluidine according to the following reaction that forms the basis of the colorimetric assay :


The diagnostic green colour is usually developed for exactly 10 minutes at 100°C and measured subse-quently at 635 nm.

 

1. Instrumentation

 

The schematic diagram of an Auto Analyser is shown in Figure 2.6. The major component parts com-prise of the various important sections namely : the preparation section, the reaction section and the analysis section which will be discussed briefly here.


 

1.1. Preparation Section

 

This particular section of the Auto Analyzer consists mainly of the sampler, proportioning pumps, and programmer. First, the sampler introduces a fixed quantity of serum sample into the ‘analysis train’, which varies from one instrument to another instrument supplied by different manufacturers. For instance, the SMA-12 Survey Auto Analyzer possesses 12 analysis trains or streams as illustrated in Figure 2.7.


 

The proportioning pump controls the rate of advancement, viz 10 inch/minute, of each sample through the analysis stream. Hence, a fixed length of tubing is equivalent to a fixed amount of time. Each analysis stream is made of transparent plastic flexible tubing, and each patient-sample is separated from one another by an air-bubble.

 

1.2. Reaction Section

 

The reaction section essentially comprises of the dialyzer, heat bath and phaser, and obviously the reaction takes place in this zone. Let us consider the following generalized reaction :


Where , [C]c = Molar concentration of substance C raised to the cth power,

       A = Component in serum (e.g., cholesterol), and

       B = Reactant that reacts with A to give a coloured product.

Evidently, B is added always in excess to ensure :

(a)       rapid reaction, and

(b) complete reaction by forcing the reaction to the right in accordance to the Le Chatelier’s principle.

Now, the rate of forward reaction = k [A]a [B]b

Hence, the rate constant may be expressed as follows :

k  = Ae–Ea/RT             ...............(c)

where , R = Gas constant ( 1.99 cal/K-mol),

T = Temperature, and

Ea = Activation energy of the reaction as depicted in Figure 2.8.


From Eq. (c) it may observed that as the temperature T is enhanced then the rate of reaction also enhances simultaneously because a higher value of T offers a smaller negative exponent of e or a larger number. Therefore, in actual experimental operations temperature is increased by the aid of a heat-bath so as to accelerate the reaction which in turn allows the reaction to attain equilibrium state as rapidly as possible.

 

Naturally at a very high temperature there is every possibility for decomposition of either the products or the reactants.

 

1.3. Analysis Section

 

The recent advancement in the field of computer technology and anlytical instrumentation it has become very easy and convenient to have the analyical data from a series of biological samples processed at high speed as digital readouts or on computerized recorders. Many hospitals round the globe make extensive use of advanced computer softwares for data processing as stated beiow :

 

·              Uptodate listing of various laboratory tests,

 

·              Listing of drugs and metabolites that cause interference both biochemically and analytically,

 

·              Storing of levels of biologically important compounds for various disease states, and

 

·              A tentative diagnosis for a patient based on his serum sample under investiation together with the drugs and dosages being administered and the levels of biologically important compounds.

 

Caution : Nevertheless, the concerned physician or pharmacist must exercise his or her own expertise and knowledge while prescribing drug(s) to a patient along with these computerized data informations.


Chapter: Pharmaceutical Drug Analysis: Errors In Pharmaceutical Analysis and Statistical Validation

Errors in Pharmaceutical Analysis

The terminology ‘error’ invariably refers to the difference in the numerical values between a measured value and the true value.

Errors in Pharmaceutical divided into two different portions, namely : (a) Errors in Pharmaceutical Analysis, and (b) Statistical Validation, which will be discussed individually in the following sections :

 

ERRORS IN PHARMACEUTICAL ANALYSIS

 

INTRODUCTION

 

The skill, knowledge, expertise and above all the degree of confidence involved in the ultimate result of an analyst is solely governed by the extent of accuracy and precision achieved by the analytical procedure vis-a-vis the possible sources of error that may be incorporated inadvertently. In fact, the quantitative pharmaceutical analysis is not merely confined to just taking a random sample, performing a single assay quickly, and finally making a loud claim that the result so obtained cannot be challenged. Truly speaking an ideal analyst must have a total in-depth knowledge of the chemistry involved along with the pros and cons of interferences that may be caused due to the host of compounds, elements and ions besides adequate exposure and hands-on experience of the statistical distribution of values.

 

The terminology ‘error’ invariably refers to the difference in the numerical values between a measured value and the true value. It has become universally accepted in methods of comparison that the percentage composition of a ‘standard sample’ provided and certified by the National Institute of Standards and Technology (NIST) or the British Pharmacopoea Chemical Reference Substance (BPCRS) or the European Pharmacopoea Chemical Reference Substance (EPCRS) must be regarded and treated as absolutely correct, pure and authentic while evaluating a new analytical method. Consequently, the differences thus obtained between the standard values and those by the new analytical methods are then treated as ‘errors’ in the latest procedure.


Chapter: Pharmaceutical Drug Analysis: Errors In Pharmaceutical Analysis and Statistical Validation

Classification of Errors in Pharmaceutical Analysis

The numerous uncertainties usually encountered in a chemical analysis give rise to a host of ‘errors’ that may be broadly categorised into two heads, namely : (i) Determinate (systematic) Errors, and (ii) Indeterminate (random) Errors.

CLASSIFICATION OF ERRORS

The numerous uncertainties usually encountered in a chemical analysis give rise to a host of ‘errors’ that may be broadly categorised into two heads, namely :

 

(i) Determinate (systematic) Errors, and

 

(ii) Indeterminate (random) Errors.

 

It is pertinent to mention here that it becomes rather difficult at times to place a particular ‘error’ into one of the above mentioned categories ; however, the classification may prove to be beneficial with regard to study of the various analytical errors that crop up in the course of routine analysis.

 

1. DETERMINATE (SYSTEMATIC) ERRORS

 

These are errors that possess a definite value together with a reasonable assignable cause; however, in principle these avoidable errors may be measured and accounted for coveniently. The most important errors belonging to this particular class are :

 

(a) Personal Errors : They are exclusively caused due to ‘personal equation’ of an analyst and have no bearing whatsoever either on the prescribed procedure or methodology involved.

 

(b) Instrumental Errors : These are invariably caused due to faulty and uncalibrated instruments, such as : pH meters, single pan electric balances, uv-spectrophotometers, potentiometers etc.

 

(c) Reagent Errors : The errors that are solely introduced by virtue of the individual reagents, for instance : impurities inherently present in reagents ; high temperature volatalization of platinum (Pt) ; unwanted introduction of ‘foreign substances’ caused by the action of reagents on either porcelain or glass apparatus.

 

(d) Constant Errors : They are observed to be rather independent of the magnitude of the measured amount ; and turn out to be relatively less significant as the magnitude enhances.

 

Example : Assuming a constant equivalence—point error of 0.10 ml is introduced in a series of titrations, hence for a specific titration needing only 10.0 ml of titrant shall represent a relative error of 1% and only 0.2% for a corresponding 50 ml of titrant consumed.

 

(e) Proportional Errors : The absolute value of this kind of error changes with the size of the sample in such a fashion that the relative error remains constant. It is usually incorporated by a material that directly interferes in an analytical procedure.

 

Example : Estimation of ‘chlorate’—an oxidant by iodometric determination. In this particular instance two things may happen, namely :

 

(i) Presence of ‘Bromate’—another oxidizing agent would give rise to positively higher results, and hence, it must be duly corrected for, and

 

(ii) Absolute error might increase while dealing with large samples, whereas the relative error would remain more or less constant if the sample is perfectly homogenous,

 

(f) Errors due to Methodology : Both improper (incorrect) sampling and incompleteness of a reaction often lead to serious errors. A few typical examples invariably encountered in titrimetric and gravimetric analysis are cited below :


 

(g) Additive Errors : It has been observed that the additive errors are independent of the quantum of the substances actually present in the assay.

 

Examples : (i) Errors caused due to weights, and

 

(ii) Loss in weight of a crucible in which a precipitate is incenerated.

 

Detection of this error is ascertained by taking samples of different weights.

 

2. INDETERMINATE (RANDOM) ERRORS

 

As the name suggests, indeterminate errors cannot be pin-pointed to any specific well-defined reasons. They are usually manifested due to the minute variations which take place inadvertently in several successive measurements performed by the same analyst, using utmost care, under almost identical experimental parameters. These errors are mostly random in nature and ultimately give rise to high as well as low results with equal probability. They can neither be corrected nor eliminated, and therefore, form the ‘ultimate limitation’ on the specific measurements. It has been observed that by performing repeated measurement of the same variable, the subsequent statistical treatment of the results would have a positive impact of ‘reducing their importance’ to a considerable extent.

 

Example : Figure 3.1, represents the absolute errors in nitrogen analysis by means of micro Kjeldahl’s Method*. Here, each vertical line labelled ( x’1 xt) designates the absolute deviation of the mean of the set from the true value. In Figure 3.1, A represents ( x’1xt) the absolute error obtained by ‘analyst-1’ for the assay of benzyl-iso-thioureahydrochloride, whereas B represents ( x’2xt) the absolute error obtained by ‘analyst-2’ for the assay of the same compound.


Thus, it is evident from Figure 3.1, that the broad spread of individual errors centres around the mean values (x’nx ) thereby affording a direct indication of indeterminate type uncertainties. Hence, larger indeterminate errors seem to be linked with the performance of ‘analyst-2’ than with that of ‘analyst-1’.

 

Salient Features of Indeterminate Errors

 

The various salient features of indeterminate errors are enumerated below :

 

1)          Repeated mesurement of the same variable several times and subsequent refinement to the extent where it is simply a coincidence if the corresponding replicates eventually agree to the last digit,

 

2)          Both unpredictable and imperceptible factors are unavoidably incorporated in the results what generally appear to be ‘random fluctuations’ in the measured quantity,

 

3)          Recognition of specific definite variables which are beyond anyone’s control lying very close to the performance limit of an instrument, such as : temperature variations, noise as well as drift from an electronic circuit, and vibrations caused to a building by heavy vehicular-traffic,

 

4)          A variation that may be regarded as random by a slipshod analyst may at the same time prove to be quite evident and manageable by a careful observer, and

 

5)          The average of a number of fine observations having random scatter is definitely more accurate, precise and, hence, more cogent than coarse data that appear to agree perfectly.

 

3. ACCURACY

 

In connexion with the scientific data the two terminologies ‘accuracy’ and ‘precision’ are invariably practised synonymously, but there exists a clear distinction between them as discussed below :

 

In usual practice an accurate result is the one which matches very nearly with true value of a measured amount. The comparison is normally done with regard to the ‘error’; and the accuracy is inversely propor-tional to it i.e., the greater the accuracy, the smaller is the error. ‘Absolute error’ is the difference between the experimental value and the true value.

 

Example : An analyst determines a value of 70.55% cineole in a fresh sample of Eucalyptus Oil that actually contains 70.25%, the absolute error is given by :

 

70.55 – 70.25 = 0.30%

 

The error thus obtained is invariably stated with regard to the actual size of the measured quantity i.e., either in percent (%) or in parts per thousand (ppt). Therefore, the relative error is given by :


 

4. PRECISION

 

It may be defined as—‘the agreement amongst a cluster of experimental results ; however, it does not imply anything with respect to their relation to the ‘true value’ ’. Precision designates ‘reproducibility’ of a measurement, whereas accuracy the correctness of a measurement. Precision invariably forms an integral part of accuracy, but ironically a high degree of precision may not necessarily suggest accuracy.

 

Example : A sample of pure Peppermint Oil is known to contain 30.10 ± 0.03 per cent of Menthone. The results obtained by two Analysts-1 and 2 employing the same sample of peppermint oil and making use of the same analytical reagents and procedure are as stated below :



The arithmetic mean is 31.44% and the results vary between 31.40% to 31.46%

The ultimate results of the analysis put forward by the Analysts-1 and 2 may be summarized as under :

 

(i) The results achieved by Analyst-1 are fairly accurate i.e., in close proximity to the correct result ; however, the precision stands at an inferior level to the results obtained by Analyst-2. The results accomplished by Analyst-2 are indeed extremely precise but fail in accuracy,

 

(ii) The results of Analyst-1 lie on either sides of the average value as shown by two ‘cross-signs’ on each side which might have been caused due to ‘random errors’ discussed earlier. It is quite evi-dent that there exists a constant (determinate) error in the results obtained by the Analyst-2, and

 

(iii) In case, Analyst-3 had performed the estimations on the very same day in quick succession i.e., one after the other, this type of analysis could be termed as ‘repeatable analysis’. If the estimations had been carried out on two separate days altogether, thereby facing different laboratory conditions then the results so obtained would be known as ‘reproducible analysis’.

 

In short, there exists a marked and pronounced distinction between a within-run precision (i.e., repeatability) and an in-between-run precision (i.e., reproducibility).

 

5. MINIMISING SYSTEMATIC ERRORS

 

Systematic errors may be reduced substantially and significantly by adopting one of the following procedures rigidly, such as :

 

(i) Calibration of Instruments, Apparatus and Applying Necessary Corections

 

Most of the instruments, commonly used in an analytical laboratory, such as : UV-Spectrophoto-meter, IR-Spectrophotometer, single—pan electric balance, pH-meter, turbidimeter and nephelometer, polarimeter, refractometer and the like must be calibrated duly, before use so as to eliminate any possible errors. In the same manner all apparatus, namely : pipettes, burettes, volu-metric flasks, thermometers, weights etc., must be calibrated duly, and the necessary corrections incorporated to the original measurements

In some specific instances where an error just cannot be avoided it may be convenient to enforce an appropriate correction for the effect that it ultimately causes ; for instance : the inherent impu-rity present in a weighed precipitate can be estimated first and then deducted duly from its weight.

 

(ii) Performing a Parallel Control Determination

 

It essentially comprises of performing an altogether separate estimation under almost identical experimental parameters with a quantity of a standard substance that consists of exactly the same weight of the component as is present in the unknown sample. Thus, the weight of the component present in the unknown sample may be calculated with the help of the following expression :


where,             X = Weight of the component present in the Unknown Sample.

 

Note :             A good number of Standard Samples, including primary standards, such as : arsenic trioxide, benzoic acid, potassium hydrogen phthalate, sodium oxalate, are available as :

 

BPCRS = British Pharmacopoeia Chemical Reference Substance,

 

EPCRS = European Pharmacopoeia Chemical Reference Substance,

 

CRM = BCS—Certified Reference Materials,

 

ECRM = EURONORM—Certified Reference Materials.

 

(iii) Blank Determination :

 

In order to ascertain the effect of the impurities present in the reagents employed and reaction vessels used ; besides establishing exactly the extent to which an excess of standard solution required to locate the exact end-point under the prevailing experimental parameters of the unknown sample—a blank determination is an absolute necessity. It may be accomplished by performing a separate parallel estimation, without using the sample at all, and under identical experimental parmeters as employed in the actual analysis of the given sample.

 

Note : Always avoid using an appreciably large blank correction which gives rise to a vague and uncertain ‘exact value’ thereby minimising the precision of the analysis.

 

(iv) Cross-checking Results by Different Methods of Analysis

 

In certain specific cases tha accuracy of a result may be cross-checked by performing another analysis of the same substance by an altogether radically different method.

 

Examples :

 

(a) HCl-Solution : It may be assayed either by titration with a standard solution of a strong alkali (NaOH), or by precipitation and weighing as AgCl ; and

 

(b) Fe3+ : It may be assayed either by gravimetric method as Fe(III) hydroxide after getting rid of the interfering elements and igniting the precipitate to Fe(III) oxide, or by titrimetric method i.e., first reducing to the Fe(II) state and then titrating with a suitable oxidizing agent, for instance Ce(IV) sulphate, K2Cr2O7. In short, the results thus obtained by the two fundamen-tally different techniques must be concordant thereby justifying and ascertaining the fact that the values obtained are fairly small limits of error.

 

(v) Method of Standard Addition

 

Here, a small known quantity of the component under estimation is added to the sample, which is subsequently subjected to analysis for the total amount of component present. The actual differ-ence in the quantity of components present in samples with or without the added component ultimately gives the recovery of the quantum added component. A good satisfactory recovery builds up the confidence in the accuracy of the method of analysis.

 

Note : The method of ‘standard addition’ is particularly useful to physicochemical techniques

 

of analysis, for instance : spectrophotometry, turbidimetry.

 

(vi) Method of Internal Standards

 

The specific method is of immense value both in chromatographic as well as spectroscopic determinations. Here, a fixed quantity of a reference substance (i.e., the ‘internal standard’) is added to a series of known concentrations of the material to be assayed.

 

A graph is plotted between the concentration values and the ratios obtained from the physical value (i.e., peak area of absorption) of the ‘internal standard’ and the series of known concentra-tions, thereby producing a straight line. Any unknown concentration may be determined effec-tively by adding the same amount of ‘internal standard’ and locating exactly where the ratio obtained falls on the concentration scale.

 

Chapter: Pharmaceutical Drug Analysis: Errors In Pharmaceutical Analysis and Statistical Validation

Pharmaceutical Statistical Validation

After accomplishing the thorough investigation of various aspects of possible ‘determinate errors’ and having applied the relevant corrections, it has been observed that the data thus generated not only show fluctuations but also are found to be random in nature.

STATISTICAL VALIDATION

 

INTRODUCTION

After accomplishing the thorough investigation of various aspects of possible ‘determinate errors’ and having applied the relevant corrections, it has been observed that the data thus generated not only show fluctuations but also are found to be random in nature. The powerful and effective technique of statistics may render such results, which scatter in a random manner, into a better form that may be employed intelligently. Besides, the specific statistical treatment of the calibration data, aided by pre-programmable calculators and micro-computers, very often yields a fairly accurate and more presentable determination of the graphs between absorbance and concentration than those produced manually.

 

STATISTICAL VALIDATION

The statistical validation of analytical results will be discussed with regard to the following six as-pects individually, along with appropriate examples wherever possible, in the sections that follow :

 

(i) Statistical treatment of finite samples,

 

(ii) Distribution of random errors,

 

(iii) Significant errors,

 

(iv) Comparison of results,

 

(v) Method of least squares, and

 

(vi) Criteria for rejection of an observation.


Chapter: Pharmaceutical Drug Analysis: Errors In Pharmaceutical Analysis and Statistical Validation

Statistical Treatment of Finite Samples

The various techniques by which one may effectively treat the scientific data normally obtained in actual analytical procedures are enumerated below :

STATISTICAL TREATMENT OF FINITE SAMPLES

 

The various techniques by which one may effectively treat the scientific data normally obtained in actual analytical procedures are enumerated below :

 

1. Mean

 

It is the average of a series of results. The mean of a finite number of measurements, xl, x2, x3, x4, ........, xn, is commonly represented as x’. It may be calculated by taking the average of individual results as shown below :


It is evident that the mean of n results is  n times more reliable than any one of the individual results. Therefore, there exists a diminishing return from accumulating more and more replicate meaurements. In other words, the mean of 9 results is 3 times as reliable as 1 result in measuring central tendency (i.e., the value about which the individual results tend to cluster) ; the mean of 16 results is 4 times as reliable etc.

 

2. Median

 

The median of an even number of results is nothing but the average of the ‘two middle values’ pro-vided the results are listed in order ; whereas for an odd number of results the median is the ‘middle value’ itself. However, the ‘mean’ and the ‘median’ are exactly identical in the case of a truly symmetrical distribu-tion. In short, median is an useful measure specifically when dealing with very small samples.

 

3. Average Deviation (or Mean Deviation)

 

It is the average of the differences between the individual results and the mean. It is regarded as a measure of variability. In the case of a small number of observations the average deviation is found to be not quite significant statistically. The average or mean distribution may be calculated by adopting the following steps, namely :

 

(i) To find the differences between individual results and the mean, without considering the +ve or –ve sign,

 

(ii) To add these individual deviations, and

 

(iii) To divide by the number of results (i.e., n).

 

Hence, an ‘average deviation’ may be expressed as :


 

4. Standard Deviation

 

It is the distance from the mean to the point of inflexion of the normal distribution curve. In compari-son to the average deviation the ‘standard deviation’ is usually considered to be much more useful and meaningful statistically. For a finite number of values it is normally symbolised as ‘S’, and may be expressed as follows :


In a situation, where ‘n’ is fairly large, say to the extent of 50 or more, it hardly matters whether the denominator in the above expression is either n – 1 or n; however, the former (i.e., n – 1) is strictly correct.

 

5. Coefficient of Variation (ν)

 

The coefficient of variation (ν) is simply the standard deviation(s) expressed as a percentage of the mean ( x’ ) as stated below :


 

6. Variance (s2)

 

The variance is the square of the standard deviation(s) i.e., s2. However, the former is fundamentally more important in statistics than the latter, whereas the latter is employed more frequently in the treatment of chemical data.

 

7. Calculations of Fundamental Statistical Parameters

 

Example : The normality of a solution of sodium hydroxide as determined by an ‘analyst’ by FOUR different titrations are found to be 0.5038 ; 0.5049 ; 0.5042 ; and 0.5039. Calculate the mean, median, average deviation, standard deviation and coefficient of variation.



Chapter: Pharmaceutical Drug Analysis: Errors In Pharmaceutical Analysis and Statistical Validation

Distribution of Random Numbers

Distribution of Random Numbers
Results obtained from a given set of measurements that scatter in a random manner are adequately treated by most logical methods of statistics.

DISTRIBUTION OF RANDOM NUMBERS

 

Results obtained from a given set of measurements that scatter in a random manner are adequately treated by most logical methods of statistics.

 

In a situation whereby a large number of replicate readings, not less than 50, are observed of a titrimetric equivalence point (continuous variable), the results thus generated shall normally be distributed around the mean in a more or less symmetrical fashion. Thus, the mathematical model which not only fits into but also satisfies such a distribution of random errors is termed as the Normal or Gaussian distribution curve. It is a bell-shaped curve which is noted to be symmetrical about the mean as depicted in Figure 3.2.


 

The equation of the normal curve may be expressed as given below :


where, y = Relative frequency with which random sampling of the infinite population shall bring forth a specific value x,

 

 σ= Standard deviation, and

 

μ= Mean.

 

From the Normal distribution curve (Figure 3.2) it may be observed that 68.26% of results shall fall within one standard deviation on either side of the mean, 95.46% shall fall within two standard deviations, and 99.74% within three standard deviations.


Chapter: Pharmaceutical Drug Analysis: Errors In Pharmaceutical Analysis and Statistical Validation

Significant Figures

Generally, significant figures may be defined as—“All digits* that are certain plus one which contains some uncertainty are said to be significant figures”.

SIGNIFICANT FIGURES

 

Generally, significant figures may be defined as—“All digits* that are certain plus one which contains some uncertainty are said to be significant figures”.

 

Examples :

(a) Burette Reading : Burettes are mostly graduated with the smallest graduation as 0.1 ml ; hence, while taking the burette reading the figures 6.3 ml can be read off with ample certainty. However, the second place of the decimal is normally estimated by arbitrarily sub-dividing the smallest division into 10 equal parts. Consequently, the final burette reading of 6.32 ml essentially contains three significant figures, of which two are certain, and one with some uncertainty.

 

(b) Measuring Weights : In the two measured quantities : 4.7350 g and 4.0082 g the zero is a significant figure ; whereas, in the quantity 0.0065 kg the zeros are not significant figures. Thus, in the latter instance the zeros only serve to locate the decimal point and, therefore, may be eliminated completely by proper choice of units, e.g., 6.5 g. Moreover, the first two numbers do have five significant figures, whilst 0.0065 only has two significant figures.

 

1. Computation Rules

 

The following computation rules are advocated to make sure that a calculated result, arrived at either by addition and subtraction or multiplication and division essentially contains only the number of ‘digits’ duly justified by the experimental data.

 

(a) Addition and Subtraction

 

In addition and subtraction, retain only as many decimal places as appear in the number that has the fewest decimals.

 

Example : Add algebraically the numbers given : 16.48 + 9.375 – 3.5450 + 118.9.

 

Following three steps are to be carried out sequentially :

 

(i) All numbers are required to be rounded up preliminarily to two decimal places,

 

(ii) Add the rounded numbers, and 

(iii) Final result is then rounded to one decimal place.


 

(b) Multiplication and Division

 

In multiplication or division, retain in each term one more signifcant figure than is contained in the term with the largest uncertainty. However, the percentage precision of product cannot be greater than the percentage precision of the least precise term entering the calculation. Hence, the multi-plication : 2.64 × 3.126 × 0.8524 × 32.9453 must be accomplished using the values

 

2.64 × 3.126 × 0.852 × 32.95

 

which is equal to 231.6796. Thus, the result obtained may be expressed to five significant figures as 231.68.

 

(c) Rounding Numbers

 

In rounding numbers, always drop the last digit in case it is less than 5, e.g., 8.62 will become 8.6. If the last digit is more than 5, always increase the preceeding digit by one i.e., 9.38 will become 9.4. In case, the digit to be dropped is 5, always round up the preceding digit to the nearest even number i.e., 8.75 will become 8.8 ; and 8.65 will become 8.6. Evidently, this method avoids a tendency to round up numbers in one direction only.

 

In rounding off quantities to the nearest correct number of significant figures, add one to the last figure retained provided the following figure is either 5 or over. Hence, the average of 0.6526, 0.6521, and 0.6524 is 0.6525 (0.65237).

 

(d) Always retain as many significant figures in a result as will yield only one uncertain figure.

 

Examples : (i) A volume read off from a burette reading that lies between 15.6 ml and 15.8 ml must be recorded as 15.7 ml, but not as 15.70 ml, because the latter would indicate that the reading lies between 15.69 and 15.71 ml.

 

(ii) A weight, to the nearest 0.1 mg, is recorded as 2.4500 g ; and it must not be written as either 2.450 g or 2.45 g, because in the latter instance an accuracy of a centigram is emphasized whereas in the former a milligram.


Chapter: Pharmaceutical Drug Analysis: Errors In Pharmaceutical Analysis and Statistical Validation

Comparison of Results

In fact there are two frequently employed methods that may be used to compare the results, namely : (a) Student’s t-Test, and (b) Variance-Ratio Test (or F-Test).

COMPARISON OF RESULTS

 

In a situation where the same sample has been analysed by two separate techniques altogether, each of them repeated several times, and that the mean values obtained are not the same ; statistically it may be possible to ascertain whether the analytical procedure adopted has been either accurate and precise or if it is superior to one of the two methods.

 

In fact there are two frequently employed methods that may be used to compare the results, namely :

 

(a) Student’s t-Test, and

 

(b) Variance-Ratio Test (or F-Test).

 

In order to perform these two tests one should have a clear understanding of the statistical term ‘the number of degrees of freedom’.

 

Degrees of Freedom : It is the number of individual observations which can be allowed to vary under conditions that the mean ( x ) and standard deviation(s), once determined, be held constant.

Thus, a sample having n values have n degrees of freedom, whereas the sum Σ(xx’ )2 is considered to have n – 1 degrees of freedom, because for any defined value of the mean, x’ , only n – 1 value can be assigned freely, as the nth is being defined from the other values automatically.

1. Student’s t-Test

 

It is usually employed for small samples only. It serves two main objectives, namely :

 

(i) It is employed to test the difference between the means of two sets of data x1  and x2 , and

 

(ii) It is used to compare the mean obtained from a sample having certain standard value and to express certain degree of confidence in the significance of the comparison.

 

The value of t may be obtained from the following expression :

                 ........................(i)

where,         x’  = Mean value,

μ = True value,

 

s = Standard deviation, and

 

n = Degree of Freedom.

 

Example : If x the mean of 12 determinations = 9.59, and µ the true value = 9.03, find out whether or not this result is significant provided the standard deviation(s) is 0.16.

 

From the above Eq. (i) we have :


Now, from the t-tables for (n – 1 i.e., 12 – 1 =) 11 degrees of freedom we have :


Evidently, the calculated value for t is 12.11 and the result is highly significant. Besides, the t-table also gives the information that the probability of obtaining the difference of 0.56 between the experimental and the true result comes out to be less than 1 in 100, which obviously is indicative of the fact that some kind of bias does exist in the laboratory method adopted.

 

2. Variance-Ratio Test (or F-Test)

 

A test that makes use of the ratio of the variances of two sets of results to determine if the standard deviations (s) are significantly different. Its application may also be extended to compare precisely the results obtained either from two different laboratories or from two different analytical procedures.

 

It is simply calculated from the following equation :

                                  ...................(ii)

where, s1 and s2 = Standard deviations of two sets of results.

 

Various steps invoived to determine F-Test are

 

(i) Find the ratio from Eq. (ii),

 

(ii) Place the larger s-value in the numerator so that F > 1,

 

(iii) Check for its significance against values in the F-table, and

 

(iv) If the F-value in the table is less than the calculated F-value, then the two standard deviations are significantly different ; otherwise, they are not.

 

Example : A sample of anhydrous sodium carbonate (Na2CO3) is analysed by two different methods which give the results for the percentage of Na2CO3 as follows :


Is there any significant difference between the precision of these two sets of results ?

 

Applying the variance-ratio or F-Test from Eq. (ii) we have :


From the standard table having F-values at the 95% probability level, under column n – 1 = 4 (since s2 >sl ) and row n – 1 = 5, find F = 6.39. Because, 6.39 > 1.69, the standard deviations are not significantly different [see step (iv) above].


Chapter: Pharmaceutical Drug Analysis: Errors In Pharmaceutical Analysis and Statistical Validation

Method of Least Squares

Method of Least Squares
A number of pharmaceutical analytical methods are solely based on instrumental measurements of an absolutely physical nature, such as : measuring peak areas with the help of a gas-chromatograph (GC), and measuring absorbance of a solution using a spectrophotometer (UV).

METHOD OF LEAST SQUARES

 

A number of pharmaceutical analytical methods are solely based on instrumental measurements of an absolutely physical nature, such as : measuring peak areas with the help of a gas-chromatograph (GC), and measuring absorbance of a solution using a spectrophotometer (UV). In both these instances, the physical characteristics are directly proportional to the concentration of the analyte under examination. In usual prac-tice, a number of solutions having known concentrations is prepared and the response of the instrument is subsequently measured for each standard solution. Finally, a standard curve or calibration curve is plotted between the observed response Vs concentration, which invariably gives rise to straight line. It has been noticed, that the experimental points rarely fall exactly upon a straight line by virtue of the indeterminate errors caused by the instrument readings. At this juncture, an analyst is confronted with the tedious problem to obtain the ‘best’ straight line for the standard curve based on the observed points so that the error in estimating the concentration of the unknown sample is brought down to the least possible extent. At this stage, instead of deciding to draw the line merely on an analyst’s judgement, statistics comes to the rescue by providing a mathematical relationship whereby the analyst not only may calculate the slope objectively but also can obtain the ‘best’ straight line. The statistical process involved is termed as the method of least squares.

 

Example : The results obtained from the determination of concentration of the standard solutions and measurements of corresponding peak areas with a GC are recorded in Table 3.1 and plotted in Figure 3.3 ; where the former is represented along the x-axis and the latter along the y-axis. How to draw the’ ‘best’ straight line through all these points ?


Considering that the relationship between the concentration and the observed peak areas is a linear one, the equation for a straight line may be expressed as :

 

y = mx + b

 

where, m = Slope of the line, and

 

b = Intercept on the y-axis.

 

It may also be assumed that values of x are free of any error.


Presumably, the indeterminate errors caused by the instrument readings, y, are responsible for not allowing the ‘data points’ to fall exactly on the line. Therefore, the sum of the squares of the deviations obtained from the real instrument readings with respect to the correct values are minimized coinsiderably by adjusting adequately the values of the slope, m, and the intercept, b.

 

Table 3.1, comprises the values of x and y to enable plot of the graph in Figure 3.3, besides values of x2, y2 and xy and also the sums of all these terms.

Statistically, the slope (m) and intercept (b) of the straight line may be obtained by the help of the following equations :


Therefore, the equation of the line is

 

y = 1.99x + 1.02

 

Thus, the standard deviation of the y values, Sy, is given by :


The number of degrees of freedom in the above expression is n – 2, because two degrees have already been consumed while calculating the values of m and b earlier.

 

The standard deviation of the slope, Sm, is given by :


At this point, let us suppose that the ‘calibration curve’ is used to find out the concentration of the ‘unknown’. Assuming that three determinations have been carried out separately, thereby giving three y values of 5.85, 5.88, 5.91, or an average value, yu , of 5.88. Thus, using the expression : y = mx + b, we have


The standard deviation (Su) in this result is obtained from the expression :


where, nu = Number of determination of unknown,

n = Number of points in the calibration graph, and

y’ = Average of the y-values in the calibration graph (i.e., 34.94/5 = 6.99)


In case, the above statistical analysis has been based on a single determination, for instance : y = 5.88, the value of Su shall come out to be :




Chapter: Pharmaceutical Drug Analysis: Errors In Pharmaceutical Analysis and Statistical Validation

Recommendations for Criteria of Rejecting an Observation

Two situations often arise, namely : (i) Number of replicates being small, and (ii) Number of replicates being large.

RECOMMENDATIONS FOR CRITERIA OF REJECTING AN OBSERVATION

 

An analyst, while carrying out a series of measurements, invariably comes across with ONE specific result in a set of replicates that obviously appears to be quite ‘out of place’ with the others, and at this juncture he should take an appropriate decision whether to discard (or expunge) this result from any further consideration. Thus, two situations often arise, namely :

 

(i) Number of replicates being small, and

 

(ii) Number of replicates being large.

 

A. Number of Replicates being Small

 

An analyst in the true sense encounters a serious problem when the number of replicates at his disposal is SMALL. Firstly, the divergent result shows a distinct and significant effect upon the mean value ( x’) ; and secondly, the prevailing scanty available data does not permit getting at the real statistical analysis of the status of the suspected result.

 

B. Number of Replicates being Large

 

In this instance, the analyst has the privilege of rejecting one value (i.e., the ‘out-of place’ value) as it is not an important one by virtue of the following two main reasons :

 

Firstly, a single value shall exert merely a small effect upon the mean value ( x ) ; and secondly, the treatment of data with the real statistical analysis would certainly reveal vividly the probability that the suspected ‘out of place’ result is a bonafide member of the same population as the others.

Blaedel et al.* (1951), Wilson** (1952) and Laitinen*** (1960) have put forward more broadly accepted and recommended criteria of rejecting an observation.

 

1. Rules Based on the Average Deviation

 

Both ‘2.5d’ and ‘4d’ rules are quite familiar to analysts. They may be applied in a sequential manner as follows :

 

(i) Calculate the mean ( x’ ) and average deviation ( d’ ) of the ‘good’ results,

 

(ii) Determine the deviation of the ‘suspected’ result from the mean of the ‘good’ results,

 

(iii) In case, the deviation of the suspected result was found to be either 2.5 times the average deviation of the good results (i.e., ‘2.5d’ rule) or 4 times the average deviation of the good results (i.e.4d’ rule) the suspected result was rejected out right ; otherwise the result was duly retained.

 

Note : The ‘limit for rejection’ seems to be too low for both the said rules.

 

2. Rules Based on the Range

 

The Q test, suggested by Dean and Dixon**** (1951) is statistically correct and valid, and it may be applied easily as stated below :

 

(i) Calculate the range of the results,

 

(ii) Determine the difference between the suspected result and its closest neighbour,

 

(iii) Divide the difference obtained in (ii) above by the range from (i) to arrive at the rejection Quotient Q,

 

(iv) Finally, consult a table of Q-values. In case, the computed value of Q is found to be greater than the value given in the table, the result in question can be rejected outright with 90% confidence that it was perhaps subject to some factor or the other which never affected the other results.

 

Table 3.2, records some of the Q-values as given below :


Example : Five determinations of the ampicillin content in capsules of a marketed product gave the following results : 0.248, 0.245, 0.265, 0.249 and 0.250 mg per capsule. Apply the Q-test to find out if the 0.265 value can be rejected.

 

The value of Q is :


The value in Table 3.2, at n = 5 is Q = 0.64. Because, the determined value 0.75 > 0.64, according to ‘rule based on the range’ the result i.e., 0.265 can be rejected.

 

Note : The Q-test administers excellent justification for the outright rejection of abnormally erroneous values ; however, it fails to eliminate the problem with less deviant suspicious values.


Chapter: Pharmaceutical Drug Analysis: Errors In Pharmaceutical Analysis and Statistical Validation

Sampling Statistics

The errors that are solely attributed to sampling, specifically in the instance of heterogeneous solids, usually give rise to the most important source of uncertainty in carrying out analysis of pharma-ceutical substances.

SAMPLING STATISTICS

 

The errors that are solely attributed to sampling, specifically in the instance of heterogeneous solids, usually give rise to the most important source of uncertainty in carrying out analysis of pharma-ceutical substances.

 

Thus, the overall standard deviation, sT, i.e., the total error is given by the following expression :

              ......................(i)

where, SS = Standard deviation of the sampling procedure (i.e., the sampling error), and SA = Standard deviation of the analytical procedures (i.e., the analytical error).

 

The Eq. (i) may also be expressed as :


where  VS = Variance due to sampling, and

VA = Variance due to analytical method(s).

 

The individual determination of VS and VA may be accomplished by using the method described under variance.

 

Example : If the sampling error is ± 2.8% and the analytical errors by two different analysts come out to be ± 0.9% and ± 0.1% respectively, we may have :


From Eqs. (a) and (b) it is quite evident that the actual contribution of the analytical error viz., ± 0.9% and ± 0.1%, to the total error (S T) is more or less insignificant.

 

Note : Youden* (1967) suggested that once the analytical error is reduced to 1/3rd of the sampling error, further reduction of the former is not required anymore.

 

In order to have a meaningful ‘sampling plan’ the following points should be taken into consideration**, namely :

 

·              Number of samples to be taken :

 

·              Size of the sample, and

 

·              Should separate samples be analysed or should a sample made up of two or more increments (i.e., composite sample) be prepared.

 

Unknown Bulk Material : 

A container-load of Paracetamol (10 MT) arrives at a raw-material stores and the composition of the bulk material is unknown, it will be a sensible and logical practice to carry out first and foremost a preliminary investigation by collecting a large number of samples and assaying the analyte of interest.

 

Thus, the confidence limits are given by the following expression :

           ............................(c)

where,      µ = Estimate of the true mean,

 x’ = Mean of the analytical results,

t = Parameter depending upon the number of degrees of freedom (ν) and the confidence level required,

SS = Standard deviation of individual sample, and

n = Number of samples taken.

 

Example : The estimate in variability of Paracetamol in a consignment of 10 MT, based on 20 determinations, was found to be ± 1.4%. How many samples must be taken to give (at 95% confidence level) a sampling error of less than 0.5% paracetamol ?

 

The 0.5% value, in reality, represents the difference between the sample mean x and the actual value µ. If this value is designated by E, then Eq. (c) may be expressed as :


From the tables [Percentage Points of the t-Distribution] the value of t for (n – 1 ), 19 degrees of freedom at 95% confidence level is 2.09.


Conclusion : 

From this test it has been established that at least 34 samples are required if the specifi-cations provided in the above cited example are to be fulfilled adequately.

 

Sample Size : 

Another major problem associated with the sampling process is that of the sample size.

 

In fact, the sample size withdrawn from a heterogeneous material is solely guided by two factors, namely :

 

(a) Variation in particle size, and

 

(b) Precision required in the results of the analysis.

 

The sampling variance, V, is inversely proportional to the actual number of sampling increments (n) and may be expressed as :


where,  k = Constant entirely dependent on the size of the increment and variation within the bulk material.

 

The following points with regard to sampling may be observed carefully :

 

·              A major source of error in sampling may be incorporated from the actual process of taking increments from the bulk material,

 

·              The accuracy of the sample is determined by its total size (based on Random Sampling Theory), and

 

·              The number of increments taken shall directly influence the sampling accuracy provided the bulk material comprises of varying particle sizes.


Chapter: Pharmaceutical Drug Analysis: Aqueous Titrations

Aqueous Titrations

1. Lowry and Bronsted’s Theory of Acids and Bases 2. Lewis’s Theory 3. Usanovich Theory 4. Lux-Flood Concept

AQUEOUS TITRATIONS

INTRODUCTION

 

Arrhenius’ definition of an acid is—‘a substance which yields hydrogen ion (H+) in an aqueous medium’; and that of a base is—‘a substance which yields hydroxy ions (OH) in an aqueous medium’.

However, these definitions have two serious short-comings, they are :

 

(a) they lack explanation of the behaviour of acids and bases in non-aqueous media, and

 

(b) acidity is associated with hydrogen ion—a relatively simple particle ; whereas, basicity is associated with hydroxyl ion—a relatively complex entity.

 

 

1. LOWRY AND BRONSTED’S THEORY OF ACIDS AND BASES

 

Just after the First World War in 1923, Bronsted and Bjerrum in Denmark and Lowry in Great Britain jointly put forward a more acceptable and satisfactory theory of acids and bases which is devoid of objec-tions earlier raised in Arrhenius’ definition.

 

According to Lowry and Bronsted’s theory—‘an acid is a substance capable of yielding a proton (hydrogen ion), while a base is a substance capable of accepting a proton’. Thus, a complementary relationship exists between an acid and a base that may be expressed in a generalized fashion as below :


 

1.1. Conjugate Acid-Base Pair

 

The pair of substances which by virtue of their mutual ability either gain or lose a proton is called a conjugate acid-base pair. A few typical examples of such pairs are :


It is quite evident from the above examples that not only molecules but also anions and cations can act as acids and bases.

 

In an acid-base titration, the acid will not release a proton unless the base capable of accepting it is simultaneously present ; in other words, in a situation where actual acid-base behaviour exists then an interaction should involve two sets of conjugate acid-base pairs, represented as :

 


Some other examples include :


In short, the species which essentially differ from each other by one proton only, are known as conjugate base and acid respectively. Sometimes, such a reaction is termed as protolytic reaction or protolysis, where A1 and B1 make the first conjugate acid-base pair and A2 and B2 the other pair.

 

1.2. Merits of Lowry-Bronsted Theory

 

It has two points of merit, which are :

 

(a)       hydrochloric acid on being dissolved in water undergoes a protolytic reaction, thus :


It may be observed that H3O+, known as hydronium or oxonium ion is invariably formed when an acid is dissolved in water.

Likewise, ammonia on being dissolved in water is also subjected to protolysis, thus :


(b)       all proton-transfer reactions may be handled, thus :


 

1.3. Demerits of Lowry-Bronsted Theory

 

It does not hold good for nonprotonic solvents, for instance : BF3, POCl3 and SO2.

 

2. LEWIS’S THEORY

 

Lewis (1923) put forward another definition of acids and bases solely dependent on giving or taking of an electron pair. According to Lewis—‘an acid is an electron pair acceptor, whereas a base is an electron pair donor’. Therefore, it is obvious that whenever any neutralization occurs the formation of an altogether new coordinate covalent bond between the electron pair donor and acceptor atoms take place.

 

Thus, Lewis’s definition is a much broader definition that includes coordination compound formation as acid-base reactions, besides Arrhenius and Lowry-Bronsted acids and bases. Examples :


The reaction of borontrifluoride (acid) with ammonia (base) results into a stable octet configuration between mutual sharing of a pair of electrons of latter (donor) and former (acceptor). 


The reaction of ammonia (base) with Ag+ (acid) results into a stable configuration due to the mutual sharing of a pair of electrons of latter (donor) and former (acceptor).

 

3. USANOVICH THEORY

 

Usanovich (1934) modified the Lewis concept of acid and base by removing the restriction of either donation or acceptance of the electron pair in a more generalized fashion. According to him :

 

Acid : It is a chemical species that reacts with a base thereby giving up cations or accepting anions or electrons.

 

Base : It is a chemical species that reacts with an acid thereby giving up anions or electrons or combines with cations.

 

Unlike Arrhenius, Lowry-Bronsted and Lewis acids and bases, the Usanovich’s concept in a much broader sense includes all the oxidizing agents as acids and the reducing agents as bases, e.g.,


In the Iron (II)—Iron (III) system, the ferric ion (III) acts as an oxidizing agent and is an acid ; while the ferrous ion (II) acts as a reducing agent and is a base.


Similarly, in the Cerous (III)—Ceric (IV) system, the ceric ion (IV) behaves as an oxidizing agent and acts as an acid ; while the cerous ion (III) behaves as a reducing agents and acts as a base.

 

4. LUX-FLOOD CONCEPT

 

The concept of acid-base reactions with respect to the oxide ion was first introduced by Lux (1929) and supported by Flood (1947). According to the Lux-Flood concept—‘an acid is the oxide-ion acceptor while a base is the oxide donor’. Examples :


In the above reactions both MgO and CaO are the oxide ion donor and hence act as bases, whereas SiO2 and SO3 are the oxide-ion acceptor and hence act as acids. Ultimately, the Lux-Flood acid and base react to form magnesium silicate (MgSiO3) and calcium sulphate (CaSO4) salts respectively.


Chapter: Pharmaceutical Drug Analysis: Aqueous Titrations

Theory of Acidimetry

1. Direction Titration Method 2. Residual Titration Method

THEORY OF ACIDIMETRY

Acidimetry, essentially involves the direct or residual titrimetric analysis of alkaline substances (bases) employing an aliquot of acid and is provided usually in the analytical control of a large number of substances included in the various official compendia. Examples :

 

(a) Organic substances : urea, sodium salicylate, diphenhydramine, emetine hydrochloride, meprobamate, paramethadione, pyrazinamide etc., and

 

(b) Inorganic substances : sodium bicarbonate, milk of magnesia, ammonium chloride, calcium hydroxide, lithium carbonate, zinc oxide etc.

 

The two methods, namely : direct titration method and residual titration method are briefly discussed as under :

 

1. DIRECT TITRATION METHOD

 

It is an usual practice that when a solid substance is to be assayed, an aliquot quantity of the same may be weighed accurately and dissolved in sufficient water so that the resulting solution should have more or less the same equivalent concentration as that of the acid used in the titration. Methyl orange (pH range = 3.0 to 4.4) is the indicator of choice for obvious reasons, as phenolphthalein and most other indicators are instantly affected by the carbonic acid (H2CO3) generated in the reaction which ultimately cause a change in colour even before the reaction attains completion.

 

2. RESIDUAL TITRATION METHOD

 

Residual titration or back titration is normally employed in the following two situations, namely :

 

Case I :      when a chemical reaction proceeds rather slowly or sluggishly, and

 

Case II : when the substance under determination fails to give a sharp and distinctly visible end-point with an indicator by direct titration.

 

In usual practice, the residual titration is accomplished by allowing to dissolve the substance under estimation in an accurately measured quantity of a standard solution of known strength present in excess and subsequently titrating the excess of the latter with another previously standardized solution. A good number of examples of this particular method shall be discussed in subsequent exercises.

Chapter: Pharmaceutical Drug Analysis: Aqueous Titrations

Aqueous Titrations: Assay of Drugs

A few typical examples of acidimetric titrations, employing ‘direct titration method’ (DTM) and ‘residual titration method’ (RTM) from the ‘Pharmacopoeia of India’ are described here :

ASSAY OF DRUGS

A few typical examples of acidimetric titrations, employing ‘direct titration method’ (DTM) and ‘residual titration method’ (RTM) from the ‘Pharmacopoeia of India’ are described here :

 

1. DIRECT TITRATION METHOD

 

1.1. Sodium Carbonate (DTM)

 

Materials Required : 1 g of sodium carbonate ; 0.5 N sulphuric acid.

 

Procedure : Weigh accurately about 1 g, dissolve in 20 ml of water (DW) and titrate with 0.5 N sulphuric acid, using methyl orange solution as indicator. Each ml of 0.5 N sulphuric acid is equivalent to 0.42 g of NaHCO3.

 

Equation :

 

2NaHCO3 + H2SO4   → Na2SO4 + 2H2O + 2CO2

2(84.01)

 

It is evident from the above equation that 2 ml of NaHCO3 is equivalent to 1 ml of H2SO4. Hence, 1 ml of NaHCO3 is 1 equivalent and the equivalent weight is equal to the gram-molecular weight i.e., 84.01/ 2 = 42.0 g. One millilitre of 0.05 N sulphuric acid or 1 milliequivalent is equivalent to 42.0 mg or 1 meq of NaHCO3.

Thus, the purity of the sample assayed may be calculated as follows :


 

1.2. Sodium Hydroxide (DTM)

 

Materials Required : 1.5 g of sodium hydroxide ; 1 N sulphuric acid.

 

Procedure : Weigh accurately about 1.5 g of sodium hydroxide and dissolve in about 40 ml of carbon-dioxide free distilled water (i.e., boiled and cooled DW). Cool and titrate with 1 N sulphuric acid using phenolphthalein solution as indicator. When the pink colour of the solution is discharged record the volume of acid solution required.

 

Equations : Thus, the end-point obtained with phenolphthalein as an indicator designates complete neutralization of all the NaOH as shown by the equation :

    ........................(a)

However, in a cold solution, with phenolphthalein as an indicator, the end-point of titration of sodium carbonate with 1 N sulphuric acid is exhibited when the sodium carbonate is fully transformed into sodium carbonate, thus :

                      ...............................(b)

At this juncture, add methyl orange solution and proceed ahead with the titration until a persistant pink colour is produced. Each millilitre of 1 N sulphuric acid is equivalent to 0.040 g (or 40 mg) of total alkali, calculated as NaOH and each millilitre of acid consumed in the titration with methyl orange is equiva-lent to 0.106 g of Na2CO3.

 

Explanation

 

1. Titration to a phenolphthalein end-point serves two purposes :

(a) Neutralization of sodium hydroxide, and

(b) Conversion of Na2CO3 to NaHCO3.

2. When the Na2co3 is converted to NaHCO3 ( CO32-+H+ < == > HCO3- )the H+ remains low because the CO 2– is strongly basic, thereby the pH of the resultng mixture ranges between 8 to 9.8. This is when phenolphthalein changes colour till the conversion of Na2CO3 to NaHCO3 is complete.

3. The HCO –, is weakly basic in nature due to : HCO + H+  + < == >  H CO , and the NaHCO3 thus formed remains unneutralised even though H+ has been increased to the point where phenolphthalein affords a change in colour.

4. The neutralization of the generated NaHCO3 is complete only when H+ has been enhanced by further addition of acid, as observed by the change in colour of methyl orange at pH 3.2 to 4.4.

 

The reaction is represented by the equation :


Calculations : The total volume of 1 N sulphuric acid consumed in the titration was required to neutralize NaOH and Na2CO3, thereby converting the latter first to NaHCO3 at the phenolphthalein end-point and then to H2CO3 at the methyl orange end-point.

 

From Eq. (a), it may be observed that the equivalent weight of NaOH is 40.00 g. Hence, each millili-tre of the total amount of 1 N sulphuric acid consumed is equivalent to 40.00 mg or 1 meq of NaOH. Thus, the total alkalinity calculated as NaOH is therefore :


The volume of 1 N sulphuric acid i.e., the difference between the acid consumed to a methyl orange end-point and the acid consumed to a phenolphthalein end-point, required to neutralize the NaHCO3 as in Eq. (c) is equal to the volume needed to generate the NaHCO3 from Na2CO3 as in Eq. (b). Thus, from Eq. (b) it may be calculated that each millilitre of 1 N sulphuric acid is equivalent to 106.0 mg of Na2CO3. Hence, the quantity (%) of Na2CO3 present in the sample is given by :


 

1.3. Cognate Assays

Sodium bicarbonate ; sodium salicylate tablets


 

2. RESIDUAL TITRATION METHOD

 

2.1. Zinc Oxide (RTM)

 

Materials Required : 1.5 g of zinc oxide ; 1 N sulphuric acid ; 1 N sodium hydroxide ; 2.5 g ammo-nium chloride.

 

Procedure : 1.5 g of freshly ignited and cooled zinc oxide is accurately weighed and dissolved with 2.5 g of ammonium chloride in 50 ml of 1 N sulphuric acid with the help of gentle heating. After complete dissolution, add methyl orange and titrate the excess of sulphuric acid with 1 N sodium hydoxide. Each millilitre of 1 N sulphuric acid is equivalent to 40.6 mg of ZnO.

 

Equation :


The requisite quantity of ZnO gets dissolved in the sulphuric acid thereby neutralizing an equivalent amount as shown by the above equation. Thus, the amount of sulphuric acid neutralized by the ZnO is estimated by subtracting, from the total amount of sulphuric acid utilized, the quantity neutralized by the standard NaOH in the back titration. The equivalent weight of ZnO, as shown in the above equation comes out to be 40.69 g (i.e., 81.38/2 = 40.69). Hence, each millilitre of 1 N sulphuric acid, 1 meq neautralized by the ZnO, is equivalent to 40.68 mg or 1 meq of ZnO.

 

Thus, the percentage of zinc oxide present in the sample may be calculated as follows :


 

2.2. Cognate Assays

 

Calamine ; Ephedrine ; Lithium carbonate ; Milk of Magnesia ; Magnesium stearate ; Sodium lactate Injection.


Chapter: Pharmaceutical Drug Analysis: Aqueous Titrations

Theory of Alkalimetry

Acidic substances are usually determined quantitatively by methods similar to those used for the quantitative determinations of bases.

THEORY OF ALKALIMETRY

Acidic substances are usually determined quantitatively by methods similar to those used for the quantitative determinations of bases. However, two methods are generally adopted for the assay of acidic substances, namely :

 

(a) Direct Titration Methods : It is accomplished by directly titrating an exact quantity of the acid, acid salt or other acidic substance with standard alkali solutions.

 

(b) Residual Titration Methods : It is carried out by the addition of an excess of the standard alkali solution and subsequently determining the amount in excess by residual titration with standard acid solution.

 

As a general principle, the following guidelines may be observed carefully, namely :

 

(i) the normality of the solution obtained by dissolving the acidic substance must be approximately the same as that of the titrant,

 

(ii) the liquid acidic substance to be titrated must be brought to room temperature (25°C) before titration, because many indicators offer different values at different temperatures, and

 

(iii) the quantity of acid to be taken should be calculated in such a manner that approximately 30 to 40 ml of the previously standardized base shall be utilized for the assay.

 

Inorganic Acids—for these either methyl red or phenolphthalein may be employed as indicators and the alkali must be standardized with the particular indicator used.

 

Organic acids—for these phenolphthalein is invariably used, but bromothymol blue, thymol blue and thymolphthalein are also employed as per specific requirements.

 

Besides, the aforesaid visual methods of assay i.e., observing the change in colour of indicators used, alternative instrumental methods such as : potentiometric, amperometric, polarographic, conducto-metric methods are also employed in determining the end-point.

 

 

1. DIRECT TITRATION METHOD (DTM)

 

1.1. Tartaric Acid

 

Materials Required : 2 g of Tartaric acid ; 1 N sodium hydroxide.

 

Procedure : Place 2 g of previously dried and accurately weighed sample of tartaric acid in a conical flask. Dissolve it in 40 ml of DW, add a few drops of phenolphthalein indicator and titrate with standardized 1 N sodium hydroxide. Each millilitre of 1 N sodium hydroxide is equivalent to 75.04 mg of C4H6O6.

 

Equation :

 

H2C4H4O6 + 2NaOH Na2C4H4O6 + 2H2O

(150.09)

 

From the above equation it is evident that two moles of sodium hydroxide is needed to neutralize one mole of tartaric acid, therefore, the equivalent weight of tartaric acid is 75.04 g. Hence, each millilitre of 1 N sodium hydroxide is equivalent to 0.07504 g (i.e., 1 meq) of tartaric acid.

 

Thus, the percentage of tartaric acid present in the sample is given by :


 

1.2. Busulphan

 

Materials Required : 0.25 g of Busulphan ; 0.1 N sodium hydroxide.

 

Procedure : Weigh accurately about 0.25 g of busulphan, add 25 ml of DW and boil gently under a reflux condenser for 30 minutes. Wash the condenser with a small quantity of DW, cool and titrate with 0.1 N sodium hydroxide using phenolphthalein solution as indicator. Each millilitre of 0.1 N sodium hydrox-ide is equivalent to 0.01232 g of C6H14O6S2.


Busulphan is first hydrolyzed by refluxing it with water and two moles methanesulphonic acid (from one mole of Busulphan) thus generated, titrated with 0.1 N sodium hydroxide employing phenolphthalein as indicator. Hence, the equivalent weight of busulphan is 123.145 g. Therefore, each millilitre of 0.1 N sodium hydroxide is equivalent to 0.01232 g of busulphan.

 

Thus, the percentage of busulphan present in the sample may be calculated as under :


Caution : Busulphan is extremely poisonous. Great care should be taken to avoid inhaling the particles of busulphan or exposing the skin to it.

 

1.3. Cognate Assays

 

Benzoic acid ; cellulose acetate phthalate ; chlorpropamide ; ibuprofen ; indomethacin ; nicotinic acid ; oxyphenbutazone ; phosphoric acid ; phenylbutazone and salicylic acid.


 

2. RESIDUAL TITRATION METHODS (RTM)

 

This method is mostly applicable to official compounds belonging to the class of esters, acid anhydrides, aldehydes and acid chlorides. In practice this method applies to such substances that normally react too slowly with the titrant because of their poor solubility which may be accomplished either by a heating process or by a precipitation method so as to convert the substance capable for reaction with the standard base.

 

2.1. Aspirin Tablets

 

Materials Required : 20 Aspirin Tablets ; 0.5 N sodium hydroxide ; 0.5 N HCl.

 

Procedure : Weigh and powder 20 tablets. Accurately weigh a quantity of the powder equivalent to about 0.5 g of aspirin, add 30.0 ml of 0.5 N sodium hydroxide boil gently for 10 minutes and titrate with 0.5 N hydrochloric acid using phenol red solution as an indicator. Repeat the operation without the substance being examined, the difference between the titrations represents the amount of 0.5 N sodium hydroxide required by the aspirin. Each ml of 0.5 N sodium hydroxide is equivalent to 0.04504 g of C9H8O4.

 

Equations : The aspirin is titrated with sodium hydroxide so as to neutralize any free acid formed by hydrolysis of the acetylsalicylic acid as shown by the following equation :


The carbonyl group present in acetylsalicylic acid is subsequently neutralized with NaOH to yield :


Further reaction of aspirin with excess of standard NaOH added followed by heating results in the saponification of the sodium acetylsalicylate as shown below :


 

2.2. Cognate Assays

 

Lactic acid ; Methyl salicylate ; Nicoumalone.


Chapter: Pharmaceutical Drug Analysis: Non-Aqueous Titrations

Non-Aqueous Titrations

During the past four decades a plethora of newer complex organic medicinal compounds have taken cognizance in the therapeutic armamentarium.

NON-AQUEOUS TITRATIONS

 

INTRODUCTION

During the past four decades a plethora of newer complex organic medicinal compounds have taken cognizance in the therapeutic armamentarium. Evidently, these compounds posed two vital problems of quality control, both in pure and dosage forms by virtue of their inherent characteristics, namely :

 

(a) poor solubility, and

 

(b) weak reactivity in aqueous medium.

 

Initially, the above two problems were usually circumvented in the following manner :

 

Example 1 : Amine salts—It is first changed to the water-soluble free base, extracted with an appro-priate organic solvent and treated with an excess volume of standard acid ; subsequently, the solvent was evaporated, and the remaining acid determined with a standard base.

 

Example 2 : Sodium salts—It is first acidified to release the water-insoluble organic acid, extracted with a suitable organic solvent, the solvent was removed and the residue was subsequently dried and weighed.

 

Example 3 : Nitrogen containing compounds—They are estimated by micro Kjeldahl’s Method. Nevertheless, such specific quantitative methods gave rise to certain serious anomalies and drawbacks.

 

In order to overcome these shortcomings the non-aqueous titrations were introduced.

 

Non-aqueous titrations have the following advantages, namely :

 

·              Elimination of poor solubility of substances,

 

·              Enhancement of weak reactivity of substances,

 

·              Selective titration by using suitable solvent and titrant of acidic/basic components of physiologi-cally active moiety of a salt,

 

·              Maintenance of speed, precision, accuracy and simplicity at par with classical methods of analysis, and

 

·              Weak bases which have Kb values less than 10–6 can be titrated satisfactorily by non-aqueous titrations. The reason being that in aqueous medium and at higher Kb values (> 10–6) the solvent water competes progressively with the basic species in solution for the proton of the solvent.


Chapter: Pharmaceutical Drug Analysis: Non-Aqueous Titrations

Non-Aqueous Titrations: Theory

The concepts of the Lowry-Bronsted theory may explain the various reactions that take place during many non-aqueous titrations.

THEORY

The concepts of the Lowry-Bronsted theory may explain the various reactions that take place during many non-aqueous titrations. Thus, an acid is a proton donor and a base is a proton acceptor. Therefore, when an acid HA undergoes dissociation it gives rise to a proton and the conjugate base A of the acid :


In other words, the liberated base A shall unite with a proton to give the corresponding conjugate acid HA of the base A because every base has its conjugate acid and vice versa.

 

Hence, from the above definitions it may be implied that :

 

(a) an acid : could be either an electrically neutral molecule e.g., HNO3 ; or a negatively charged anion e.g., HSO4 ; or a positively charged cation e.g., C6H5NH2+ , H3O ;

 

(b) a base : could be either an electrically neutral molecule e.g., C6H5NH2 ; or an anion e.g., Cl, NO3.

 

1. SOLVENTS

 

These are of three types and they will be discussed briefly here :

 

(a) Protophillic Solvents : They are essentially basic in nature and normally react with acids to form solvated protons :

 

Example :


Perchloric acid displays more strongly acidic characteristics than a weak acid, for instance : acetic acid when dissolved in a weakly basic solvent.

 

(b) Protogenic Solvents : They are acidic in nature and character e.g., sulphuric acid. They exert a ‘levelling effect’ on bases i.e., they become indistinguishable in strength when dissolved in strongly basic solvents due to their enhanced affinity of strong bases for protons.

 

(c) Amphiprotic Solvents : They possess both protophillic and protogenic characteristics.

 

Examples : Acetic acid, water and alcohols.

 

They undergo dissociation to a very less extent. Acetic acid is mostly employed as a solvent for the titration of basic substances and its dissociation can be depicted as shown below :

 

CH3COOH   < == >  H+  +  CH3COO

 

In the above instance acetic acid is behaving as an acid.

 

Perchloric Acid : It is a very strong acid and when it is made to dissolve in acetic acid, the latter can behave as a base and forms an ‘onium ion’ after combining with protons donated by the perchloric acid. Thus, we have :


As the CH3COOH2+ ion can instantly donate its proton to a base, therefore, a solution of perchloric acid in glacial acetic acid, behaves as a strongly acidic solution.

 

Pyridine, a weak base, when dissolved in acetic acid, the latter exerts its levelling effect and subsequently increases the basic characteristics of the pyridine. Therefore, it is practically feasible to titrate a solution of a weak base in acetic acid against a mixture of perchloric acid in acetic acid. Thus, a sharp end point is achieved which otherwise cannot be obtained when the titration is performed in an aqueous medium.

The various reactions with perchloric acid, acetic acid and pyridine are summarized below :


Acetonitrile, acetone and dimethylformamide—these non-aqueous solvents exert a greater differen-tial in the protophillic properties of many substances than in the corresponding aqueous solutions, due to the levelling effect of water in the latter solutions. Hence, the most acidic substance in aqueous solutions of a number of acids is the formation of the hydronium ion as shown below :


It is pertinent to observe here that the following inorganic acids almost exhibit equal strength in aqueous solutions, whereas in non-aqueous solvents, their ‘acidity’ retards in the following order :


In glacial acetic acid (an acidic solvent) and in dioxane (a neutral solvent), the perchloric acid (HClO4) behaves as more acidic (i.e., less protophyllic) than HCl; and, therefore, many base-hydrochlorides (i.e., chlorides) may be titrated with standard HClO4, just as carbonates may be titrated in aqueous solution with standard HCl.

 

In short, it is possible to titrate mixtures of two or three components selectively with a single titration by wisdom of the right choice of solvent for the non-aqueous titrations.


Chapter: Pharmaceutical Drug Analysis: Non-Aqueous Titrations

Non-Aqueous Titrations: Methodology

For non-aqueous titrations, the following four steps are usually taken into consideration, namely : (i) Preparation of 0.1 N Perchloric acid, (ii) Standardization of 0.1 N Perchloric Acid, (iii) Choice of Indicators, and (iv) Effect of Temperature on Assays.

METHODOLOGY

For non-aqueous titrations, the following four steps are usually taken into consideration, namely :

 

(i) Preparation of 0.1 N Perchloric acid,

 

(ii) Standardization of 0.1 N Perchloric Acid,

 

(iii) Choice of Indicators, and

 

(iv) Effect of Temperature on Assays.

 

1. PREPARATION OF 0.1 N PERCHLORIC ACID

 

Materials Required : 8.5 ml of perchloric acid (70.0 to 72.0%) ; 1 Litre of glacial acetic acid ; 30 ml of acetic anhydride.

Procedure : Gradually mix 8.5 ml of perchloric acid to 900 ml of glacial acetic acid with vigorous and continuous stirring. Now add 30 ml acetic anhydride and make up the volume to 1 litre with glacial acetic acid and allow to stand for 24 hours before use.

 

The acetic anhydride reacts with the water (approx. 30%) in perchloric acid and some traces in glacial acetic acid thereby making the resulting mixture practically anhydrous. Thus, we have :


Precautions : The following precautions must be observed :

 

(a) Perchloric acid is usually available as a 70 to 72% mixture with water (sp. gr. 1.6). It usually undergoes a spontaneous explosive decomposition and, therefore, it is available always in the form of a solution.

 

(b) Conversion of acetic anhydride to acetic acid requires 40-45 minutes for its completion. It being an exothermic reaction, the solution must be allowed to cool to room temperature before adding glacial acetic acid to volume,

 

(c)       Avoid adding an excess of acetic anhydride especially when primary and secondary amines are to be assayed, because these may be converted rapidly to their corresponding acetylated non-basic products :


(d) Perchloric acid is not only a powerful oxidising agent but also a strong acid. Hence, it must be handled very carefully.

 

Perchloric acid has a molecular weight of 100.46 and 1 L of 0.1 N solution shall contain 1 /10th the equivalent weight or 10.046 g. To prepare 1 L of standard perchloric acid solution, it requires 8.5 ml (sp. gr. 1.6) volume and a purity of 72% which will calculate out as 9.792 g of HClO4.

 

2. STANDARDIZATION OF 0.1 N PERCHLORIC ACID

 

Alkaline earth (e.g., Mg, Ca, Ba), and alkali (e.g., Na, K, Rb), salts of organic acids behave as bases in acetic acid solution :


In usual practice, potassium hydrogen phthalate (or potassium biphthalate, KHC8H4O4) is employed as a standardizing agent for acetous perchloric acid. The reaction may be expressed as follows :


Procedure : Weigh accurately about 0.5 g of potassium hydrogen phthalate in a 100 ml conical flask. Add 25 ml of glacial acetic acid and attach a reflux condenser fitted with a silica-gel drying tube. Warm until the salt gets dissolved completely. Cool and titrate with 0.1 N perchloric acid by making use of either of the following two indicators :

 

(a) acetous crystal violet-2 drops, end point Blue to Blue-Green (0.5% w/v)

 

(a)       acetous oracet blue B-2 drops, end point Blue to Pink.

 

3. CHOICE OF INDICATORS

A number of indicators stated below are commonly used in non-aqueous titrations. It is, however, necessary to mention here that the same indicator must be used throughout for carrying out the standardiza-tion, titration and neutralization of mercuric acetate solution.


 

4. EFFECT OF TEMPERATURE ON ASSAYS

 

Generally, most non-aqueous solvents possess greater coeffcients of expansion as compared to water, which is why small differences in temperature may afford significant and appreciable errors that can be eliminated by the application of appropriate correction factors. Hence, it is always advisable to carry out standardization and titration preferably at the same temperature. In a situation where these temperature parameters cannot be achieved, the volume of titrant may be corrected by the application of the following formula :

 

Vc = V [1 + 0.001 (tl + t2)]

where, Vc = Corrected volume of titrant,

 

V = Volume of titrant measured,

 

tl = Temperature at which titrant was standardized, and

t2 = Temperature at which titration was performed.


Chapter: Pharmaceutical Drug Analysis: Non-Aqueous Titrations

Assay by Non-Aqueous Titrations

(a) Acidimetry in Non-aqueous Titrations—It can be further sub-divided into two heads, namely : (i) Titration of primary, secondary and tertiary amines, and (ii) Titration of halogen acid salts of bases. (b) Alkalimetry in Non-aqueous Titrations—i.e., titration of acidic substances.

ASSAY BY NON-AQUEOUS TITRATIONS

Assays of various pharmaceutical substances either in pure form or in dosage form may be assayed successfully by non-aqueous titrations. For the sake of convenience these typical titrations can be catego-rized into two broad groups, namely :

 

(a) Acidimetry in Non-aqueous Titrations—It can be further sub-divided into two heads, namely :

 

(i) Titration of primary, secondary and tertiary amines, and

 

(ii) Titration of halogen acid salts of bases.

 

(b) Alkalimetry in Non-aqueous Titrationsi.e., titration of acidic substances.

 

1. ACIDIMETRY IN NON-AQUEOUS TITRATIONS

 

In order to perform feasible titrations of weak bases, the solvent system should be selected specifically in such a fashion so as to eliminate as far as possible the competing reaction of water for the proton besides enhancing the strength of the basic species.

 

1.1. Titration of primary, secondary and tertiary amines

 

1.1.1.  Methlyldopa

 

In general, the reaction taking place between a primary amine and perchloric acid may be expressed as follows :


The specific reaction between methyldopa and perchloric acid is expressed by the following equation :


Materials Required : Methyldopa 0.2 g ; anhydrous formic acid : 15 ml ; glacial acetic acid : 30 ml ; dioxane : 30 ml ; 0.1 N perchloric acid and crystal violet solution.

 

Procedure : Weigh accurately about 0.2 g and dissolve in 15 ml of anhydrous formic acid, 30 ml of glacial acetic acid and 30 ml of dioxane. Add 0.1 ml of crystal violet solution and titrate with 0.1 N perchloric acid. Perform a blank determination and make any necessary correction. Each ml of 0.1 N perchloric acid is equivalent to 0.02112 g of C10H13NO4.

 

Calculations : The percentage of methyldopa present in the sample is given by :


 

1.1.2.  Methacholine Clloride

Materials Required : Methacholine chloride : 0.4 g ; glacial acetic acid : 50 ml ; mercuric acetate solution : 10 ml ; 0.1 N perchloric acid and crystal violet solution.

 

Procedure : Weigh accurately about 0.4 g, previously dried and stored in a vacuum desiccator, and dissolve in 50 ml of glacial acetic acid, add 10 ml of mercuric acetate solution, one drop of crystal violet solution and titrate with 0.1 N perchloric acid to a blue-green end point. Perform a blank determination and make any necessary correction. Each ml of 0.1 N perchloric acid is equivalent to 0.01957 g of C8Hl8ClNO2.

 

Equation :


Mercuric acetate : It is essentially added to prevent the interference of the hydrochloric acid dis-placed through the formation of the relatively un-ionized HgCl2, thereby making a predominant shift in the equilibrium so that the titrimetric reaction is quantitative.

 

Blank Titration : It is usually carried out to account for the possible reaction of atmospheric moisture with the titrant perchloric acid and also to check the titrant being employed to bring about the blue-green end-point.

 

Calculations : The percentage of methacholine chloride in the sample may be calculated by the following expression :


 

1.1.3.  Cognate Assays

Table 5.1, enlists the various cognate determinations using different indicators but employing the same titrant i.e., 0.1 N perchloric acid.



 

1.1.4.  Potentiometric Titrations

 

These non-aqueous titrations may also be carried out with the help of potentiometric titrations which technique shall be discussed at length elsewhere in this book.

 

It is always preferred to first ascertain the equivalence point of a given neutralization reaction potentiometrically (i.e., an instrumental method of analysis) ; and secondly, by selecting an appropriate indicator that will ensure the sharpest colour change for the least increment of volume of titrant added near the equivalence point.

 

In actual practice, however, there are quite a number of non-aqueous titrations of pharmaceutical substances either in pure or in dosage forms that can be successfully performed potentiometrically.


 

1.2. Titration of Halogen Acid Salts of Bases

 

In general, the halide ions, namely : chloride, bromide and iodide are very weakly basic in character so much so that they cannot react quantitatively with acetous perchloric acid. In order to overcome this problem, mercuric acetate is usually added (it remains undissociated in acetic acid solution) to a halide salt thereby causing the replacement of halide ion by an equivalent amount of acetate ion, which serves as a strong base in acetic acid as shown below :


 

1.2.A. Amitriptyline Hydrochloride

 

Materials Required : Amitriptyline hydrochloride : 1.0 g ; mercuric acetate ; crystal violet ; 0.1 N perchloric acid ; glacial acetic acid.

 

Procedure : Weigh accurately about 1.0 g and dissolve in 50 ml of glacial acetic acid, warming slightly, if necessary, to affect the solution. Cool, add 10 ml of mercuric acetate solution, two drops of crystal violet solution and titrate with 0.1 N perchloric acid to a green end-point. Perform a blank determination and make any necessary correction. Each ml of 0.1 N perchloric acid is equivalent to 0.03139 g of C20H23N. HCl.

 

Equations :


Calculations :


 

1.3. Cognate Assays

 

The following estimations of various pharmaceutical substances can also be carried out by the aforesaid procedure (Table 5.3) :



 

2. ALKALIMETRY IN NON-AQUEOUS TITRATIONS

 

A plethora of weakly acidic pharmaceutical substances may be titrated effectively by making use of a suitable non-aqueous solvent with a sharp end-point. The wide spectrum of such organic compounds in-clude : anhydrides, acids, amino acids, acid halides, enols (viz., barbiturates), xanthines, sulphonamides, phenols, imides and lastly the organic salts of inorganic acids.

 

However, a weak inorganic acid e.g., boric acid, can be estimated conveniently employing ethylenediamine as the non-aqueous solvent.

 

2.1. Preparation of 0.1 N Potassium Methoxide in Toluene-Methanol

 

Materials Required : Absolute methanol : 40 ml ; dry toluene : 50 ml ; potassium metal : 4 g.

 

Procedure : Add into a dry flask, a mixture of methanol (40 ml) and dry toluene (50 ml) and cover it loosely. Carefully add freshly cut pieces of potassium metal to the above mixture gradually with constant shaking. After complete dissolution of potassium metal, add enough absolute methanol to yield a clear solution. Toluene 50 ml is added with constant shaking until the mixture turns hazy in appearance. The process is repeated by the alternate addition of methanol and benzene until 1 litre of solution is obtained, taking care to add a minimum volume of methanol to give a visible clear solution.

 

2.1.1. Preparation of 0.1 N Sodiun Methoxide

 

It is prepared exactly in a similar manner as for 0.1 N Potassium Methoxide, using 2.3 g of freshly-cut sodium in place of potassium.

 

2.1.2. Preparation of 0.1 N Lithium Methoxide

 

It is prepared as for 0.1 N Potassium Methoxide, but using 0.7 g of lithium in place of potassium.

 

2.2. Standardization of 0.1 N Methoxide Solution

 

Materials Required : Dimethylformamide (DMF) : 10 ml ; thymol blue (0.3% in MeOH) ; 0.1 N lithium methoxide in toluene-methanol ; benzoic acid : 0.6 g.

 

Procedure : The apparatus shown in Figure 5.1, is employed for the standardization of 0.1 N methoxide solution. Transfer 10 ml of DMF in a conical flask and add to it 3 to 4 drops of thymol blue and first neutralize the acidic impurities present in DMF by titrating with 0.1 N lithium methoxide in toluene-methanol. Quickly introduce 0.06 g of benzoic acid and titrate immediately with methoxide in toluene-methanol.


 

Caution : Care must be taken to avoid contamination of neutralized liquid with atmospheric carbon dioxide.

Equations :  The various equations involved in the above operations are summarized as stated below:


Interaction between sodium metal and methanol is an exothermic reaction and hence, special care must be taken while adding the metal into the dry solvent in small lots at intervals with adequate cooling so as to keep the reaction well under control.


The clear solution of sodium methoxide must be kept away from moisture and atmospheric CO2 as far as possible so as to avoid the above two chemical reactions that might ultimately result into the formation of turbidity.


Step 1 : It shows the solution of benzoic acid (primary standard) in DMF,

 

Step 2 : It depicts ionization of sodium methoxide,

 

Step 3 : It illustrates the interaction between the solvated proton and the methylated ion.

 

In summing up, the net reaction in the process of standardization has been expressed. The interaction between the water in the solvent (DMF) and the titrant is equivalent to the volume of sodium methoxide consumed by DMF or may be considered as a blank determination.

 

2.2.1. Ethosuximide

 

Materials Required : Ethosuximide : 0.2 g ; dimethylformamide : 50 ml ; azo-violet (0.1% w/v in DMF) : 2 drops ; sodium methoxide 0.1 N.

 

Procedure : Weigh accurately about 0.2 g, dissolve in 50 ml of dimethylformamide, add 2 drops of azo-violet solution and tirate with 0.1 N sodium methoxide to a deep blue end point, taking precautions to prevent absorption of atmospheric carbon dioxide. Perform a blank determination and make any necessary correction. Each ml of 0.1 N sodium methoxide is equivalent to 0.01412 g of C7H11NO2.

 

Equations :


 

2.3. Cognate Assays

 

The following determinations as stated in Table 5.4 may be carried out effectively by using 0.1 N sodium hydroxide either titrimetrically using an appropriate indicator or potentiometrically :


 

2.4. Tetrabutylammonium Hydroxide

 

The alkalimetry in non-aqueous titrations may also be carried out efficiently by using tetrabutylammonium hydroxide along with an appropriate indicatior.

 

2.4.1. Preparation of 0.1 N Tetrabutylammonium Hydroxide in Toluene-Methanol

 

Materials Required : Tetrabutylammonium iodide : 40 g ; absolute methanol : 90 ml ; silver oxide : 25 g ; dry toluene : 150 ml.

 

Procedure : Carefully dissolve 40 g of tetrabutylammonium iodide (Bu4NI) in 90 ml of absolute methanol, add to it 20 g of finely powdered purified silver oxide and finally shake the mixture thoroughly for 1 hour. Centrifuge about 2-3 ml of the resultant mixture and test for iodide in the supernatant liquid. In case, it gives a positive test, add about 2 g more of silver oxide and shake for an additional period of 30 minutes. The said method may be repeated until the supernatant liquid obtained is completely free from iodide. The mixture thus obtained is filtered through a fine sintered glass filter and finally rinse the container with 3 portions, each of 50 ml of dry toluene. These washings may be added to the filtrate and the final volume is made upto 1 litre with dry toluene. The clear solution may be flushed with CO2-free nitrogen for at least five minutes and duly protected from both CO2 and moisture during storage.

Equation :


 

2.4.2. Standardization of 0.1 N Tetrabutylammonium Hydroxide

 

Materials Required : Benzoic acid : 60 mg ; dimethylbromide : 10 ml ; thymol blue solution (0.3% w/v in methanol) ; 0.1 N tetrabutylammonium hydroxide.

 

Procedure : Accurately weigh about 60 mg of benzoic acid into 10 ml of previously neutralized dimethyl formamide to the blue colour of thymol blue (3 drops) by titration against 0.1 N tetrabutylammonium hydroxide. Allow the benzoic acid to dissolve gradually and completely and titrate with 0.1 N tetrabutylammonium hydroxide preferably in an atmosphere of CO2-free nitroaen.

Calculations :


 

2.4.3.  Chlorthalidone

 

Materials Required : Chlorthalidone : 0.3 g ; pyridine (dehydrated) : 50 ml ; 0.1 N tetrabutylammonium hydroxide.

 

Procedure : Weigh accurately about 0.3 g and dissolve in 50 ml of dehydrated pyridine. Titrate with 0.1 N tetrabutylammonium hydroxide, determining the end point potentiometrically and protecting the solution and titrant from atmospheric carbon dioxide throughout the determination. Perform a blank determination and make any necessary correction. Each ml of 0.1 N tetrabutylammonium hydroxide is equivalent to 0.03388 g of Cl4H1lClN2O4S.

 

Equations :


 

2.4.4.  Cognate Assays

 

The following pharmaceutical substances may be assayed by employing tetrabutylammonium hydroxide either by using a suitable indicator titrimetrically or potentiometrically as given in Table 5.5.


The assay of the aforesaid pharmaceutical substances with tetrabutylammonium hydroxide is on a mole-for-mole basis. As these are monobasic acids in character, therefore, they react quantitatively in a non-aqueous media with the base titrant, employing typical acid-base indicators to detect the end-points.


Chapter: Pharmaceutical Drug Analysis: Non-Aqueous Titrations

Acidimetry in Non-Aqueous Titrations

In order to perform feasible titrations of weak bases, the solvent system should be selected specifically in such a fashion so as to eliminate as far as possible the competing reaction of water for the proton besides enhancing the strength of the basic species.

ACIDIMETRY IN NON-AQUEOUS TITRATIONS

 

In order to perform feasible titrations of weak bases, the solvent system should be selected specifically in such a fashion so as to eliminate as far as possible the competing reaction of water for the proton besides enhancing the strength of the basic species.

 

1. Titration of primary, secondary and tertiary amines

 

1.1.  Methlyldopa

 

In general, the reaction taking place between a primary amine and perchloric acid may be expressed as follows :


The specific reaction between methyldopa and perchloric acid is expressed by the following equation :


Materials Required : Methyldopa 0.2 g ; anhydrous formic acid : 15 ml ; glacial acetic acid : 30 ml ; dioxane : 30 ml ; 0.1 N perchloric acid and crystal violet solution.

 

Procedure : Weigh accurately about 0.2 g and dissolve in 15 ml of anhydrous formic acid, 30 ml of glacial acetic acid and 30 ml of dioxane. Add 0.1 ml of crystal violet solution and titrate with 0.1 N perchloric acid. Perform a blank determination and make any necessary correction. Each ml of 0.1 N perchloric acid is equivalent to 0.02112 g of C10H13NO4.

 

Calculations : The percentage of methyldopa present in the sample is given by :


 

1.2.  Methacholine Clloride

Materials Required : Methacholine chloride : 0.4 g ; glacial acetic acid : 50 ml ; mercuric acetate solution : 10 ml ; 0.1 N perchloric acid and crystal violet solution.

 

Procedure : Weigh accurately about 0.4 g, previously dried and stored in a vacuum desiccator, and dissolve in 50 ml of glacial acetic acid, add 10 ml of mercuric acetate solution, one drop of crystal violet solution and titrate with 0.1 N perchloric acid to a blue-green end point. Perform a blank determination and make any necessary correction. Each ml of 0.1 N perchloric acid is equivalent to 0.01957 g of C8Hl8ClNO2.

 

Equation :


Mercuric acetate : It is essentially added to prevent the interference of the hydrochloric acid dis-placed through the formation of the relatively un-ionized HgCl2, thereby making a predominant shift in the equilibrium so that the titrimetric reaction is quantitative.

 

Blank Titration : It is usually carried out to account for the possible reaction of atmospheric moisture with the titrant perchloric acid and also to check the titrant being employed to bring about the blue-green end-point.

 

Calculations : The percentage of methacholine chloride in the sample may be calculated by the following expression :


 

1.3.  Cognate Assays

Table 5.1, enlists the various cognate determinations using different indicators but employing the same titrant i.e., 0.1 N perchloric acid.


 

1.4.  Potentiometric Titrations

 

These non-aqueous titrations may also be carried out with the help of potentiometric titrations which technique shall be discussed at length elsewhere in this book.

 

It is always preferred to first ascertain the equivalence point of a given neutralization reaction potentiometrically (i.e., an instrumental method of analysis) ; and secondly, by selecting an appropriate indicator that will ensure the sharpest colour change for the least increment of volume of titrant added near the equivalence point.

 

In actual practice, however, there are quite a number of non-aqueous titrations of pharmaceutical substances either in pure or in dosage forms that can be successfully performed potentiometrically.


 

2. Titration of Halogen Acid Salts of Bases

 

In general, the halide ions, namely : chloride, bromide and iodide are very weakly basic in character so much so that they cannot react quantitatively with acetous perchloric acid. In order to overcome this problem, mercuric acetate is usually added (it remains undissociated in acetic acid solution) to a halide salt thereby causing the replacement of halide ion by an equivalent amount of acetate ion, which serves as a strong base in acetic acid as shown below :


 

2.A. Amitriptyline Hydrochloride

 

Materials Required : Amitriptyline hydrochloride : 1.0 g ; mercuric acetate ; crystal violet ; 0.1 N perchloric acid ; glacial acetic acid.

 

Procedure : Weigh accurately about 1.0 g and dissolve in 50 ml of glacial acetic acid, warming slightly, if necessary, to affect the solution. Cool, add 10 ml of mercuric acetate solution, two drops of crystal violet solution and titrate with 0.1 N perchloric acid to a green end-point. Perform a blank determination and make any necessary correction. Each ml of 0.1 N perchloric acid is equivalent to 0.03139 g of C20H23N. HCl.

 

Equations :


Calculations :


 

3. Cognate Assays

 

The following estimations of various pharmaceutical substances can also be carried out by the aforesaid procedure (Table 5.3) :



 

Chapter: Pharmaceutical Drug Analysis: Non-Aqueous Titrations

Alkalimetry in Non-Aqueous Titrations

1. Preparation of 0.1 N Potassium Methoxide in Toluene-Methanol 2. Standardization of 0.1 N Methoxide Solution 3. Cognate Assays 4. Tetrabutylammonium Hydroxide

ALKALIMETRY IN NON-AQUEOUS TITRATIONS

 

A plethora of weakly acidic pharmaceutical substances may be titrated effectively by making use of a suitable non-aqueous solvent with a sharp end-point. The wide spectrum of such organic compounds in-clude : anhydrides, acids, amino acids, acid halides, enols (viz., barbiturates), xanthines, sulphonamides, phenols, imides and lastly the organic salts of inorganic acids.

 

However, a weak inorganic acid e.g., boric acid, can be estimated conveniently employing ethylenediamine as the non-aqueous solvent.

 

1. Preparation of 0.1 N Potassium Methoxide in Toluene-Methanol

 

Materials Required : Absolute methanol : 40 ml ; dry toluene : 50 ml ; potassium metal : 4 g.

 

Procedure : Add into a dry flask, a mixture of methanol (40 ml) and dry toluene (50 ml) and cover it loosely. Carefully add freshly cut pieces of potassium metal to the above mixture gradually with constant shaking. After complete dissolution of potassium metal, add enough absolute methanol to yield a clear solution. Toluene 50 ml is added with constant shaking until the mixture turns hazy in appearance. The process is repeated by the alternate addition of methanol and benzene until 1 litre of solution is obtained, taking care to add a minimum volume of methanol to give a visible clear solution.

 

1.1. Preparation of 0.1 N Sodiun Methoxide

 

It is prepared exactly in a similar manner as for 0.1 N Potassium Methoxide, using 2.3 g of freshly-cut sodium in place of potassium.

 

1.2. Preparation of 0.1 N Lithium Methoxide

 

It is prepared as for 0.1 N Potassium Methoxide, but using 0.7 g of lithium in place of potassium.

 

2. Standardization of 0.1 N Methoxide Solution

 

Materials Required : Dimethylformamide (DMF) : 10 ml ; thymol blue (0.3% in MeOH) ; 0.1 N lithium methoxide in toluene-methanol ; benzoic acid : 0.6 g.

 

Procedure : The apparatus shown in Figure 5.1, is employed for the standardization of 0.1 N methoxide solution. Transfer 10 ml of DMF in a conical flask and add to it 3 to 4 drops of thymol blue and first neutralize the acidic impurities present in DMF by titrating with 0.1 N lithium methoxide in toluene-methanol. Quickly introduce 0.06 g of benzoic acid and titrate immediately with methoxide in toluene-methanol.


 

Caution : Care must be taken to avoid contamination of neutralized liquid with atmospheric carbon dioxide.


Equations :  The various equations involved in the above operations are summarized as stated below:


Interaction between sodium metal and methanol is an exothermic reaction and hence, special care must be taken while adding the metal into the dry solvent in small lots at intervals with adequate cooling so as to keep the reaction well under control.


The clear solution of sodium methoxide must be kept away from moisture and atmospheric CO2 as far as possible so as to avoid the above two chemical reactions that might ultimately result into the formation of turbidity.


Step 1 : It shows the solution of benzoic acid (primary standard) in DMF,

 

Step 2 : It depicts ionization of sodium methoxide,

 

Step 3 : It illustrates the interaction between the solvated proton and the methylated ion.

 

In summing up, the net reaction in the process of standardization has been expressed. The interaction between the water in the solvent (DMF) and the titrant is equivalent to the volume of sodium methoxide consumed by DMF or may be considered as a blank determination.

 

2.1. Ethosuximide

 

Materials Required : Ethosuximide : 0.2 g ; dimethylformamide : 50 ml ; azo-violet (0.1% w/v in DMF) : 2 drops ; sodium methoxide 0.1 N.

 

Procedure : Weigh accurately about 0.2 g, dissolve in 50 ml of dimethylformamide, add 2 drops of azo-violet solution and tirate with 0.1 N sodium methoxide to a deep blue end point, taking precautions to prevent absorption of atmospheric carbon dioxide. Perform a blank determination and make any necessary correction. Each ml of 0.1 N sodium methoxide is equivalent to 0.01412 g of C7H11NO2.

 

Equations :


 

3. Cognate Assays

 

The following determinations as stated in Table 5.4 may be carried out effectively by using 0.1 N sodium hydroxide either titrimetrically using an appropriate indicator or potentiometrically :


 

4. Tetrabutylammonium Hydroxide

 

The alkalimetry in non-aqueous titrations may also be carried out efficiently by using tetrabutylammonium hydroxide along with an appropriate indicatior.

 

4.1. Preparation of 0.1 N Tetrabutylammonium Hydroxide in Toluene-Methanol

 

Materials Required : Tetrabutylammonium iodide : 40 g ; absolute methanol : 90 ml ; silver oxide : 25 g ; dry toluene : 150 ml.

 

Procedure : Carefully dissolve 40 g of tetrabutylammonium iodide (Bu4NI) in 90 ml of absolute methanol, add to it 20 g of finely powdered purified silver oxide and finally shake the mixture thoroughly for 1 hour. Centrifuge about 2-3 ml of the resultant mixture and test for iodide in the supernatant liquid. In case, it gives a positive test, add about 2 g more of silver oxide and shake for an additional period of 30 minutes. The said method may be repeated until the supernatant liquid obtained is completely free from iodide. The mixture thus obtained is filtered through a fine sintered glass filter and finally rinse the container with 3 portions, each of 50 ml of dry toluene. These washings may be added to the filtrate and the final volume is made upto 1 litre with dry toluene. The clear solution may be flushed with CO2-free nitrogen for at least five minutes and duly protected from both CO2 and moisture during storage.

Equation :


 

4.2. Standardization of 0.1 N Tetrabutylammonium Hydroxide

 

Materials Required : Benzoic acid : 60 mg ; dimethylbromide : 10 ml ; thymol blue solution (0.3% w/v in methanol) ; 0.1 N tetrabutylammonium hydroxide.

 

Procedure : Accurately weigh about 60 mg of benzoic acid into 10 ml of previously neutralized dimethyl formamide to the blue colour of thymol blue (3 drops) by titration against 0.1 N tetrabutylammonium hydroxide. Allow the benzoic acid to dissolve gradually and completely and titrate with 0.1 N tetrabutylammonium hydroxide preferably in an atmosphere of CO2-free nitroaen.

Calculations :


 

4.3.  Chlorthalidone

 

Materials Required : Chlorthalidone : 0.3 g ; pyridine (dehydrated) : 50 ml ; 0.1 N tetrabutylammonium hydroxide.

 

Procedure : Weigh accurately about 0.3 g and dissolve in 50 ml of dehydrated pyridine. Titrate with 0.1 N tetrabutylammonium hydroxide, determining the end point potentiometrically and protecting the solution and titrant from atmospheric carbon dioxide throughout the determination. Perform a blank determination and make any necessary correction. Each ml of 0.1 N tetrabutylammonium hydroxide is equivalent to 0.03388 g of Cl4H1lClN2O4S.

 

Equations :


 

4.4.  Cognate Assays

 

The following pharmaceutical substances may be assayed by employing tetrabutylammonium hydroxide either by using a suitable indicator titrimetrically or potentiometrically as given in Table 5.5.


The assay of the aforesaid pharmaceutical substances with tetrabutylammonium hydroxide is on a mole-for-mole basis. As these are monobasic acids in character, therefore, they react quantitatively in a non-aqueous media with the base titrant, employing typical acid-base indicators to detect the end-points.


Chapter: Pharmaceutical Drug Analysis: Permanganate, Dichromate and Ceric Sulphate Titration Methods

Permanganate, Dichromate and Ceric Sulphate Titration Methods

The oxidation and reduction processes essentially take place simultaneously in a reaction, thus one entity gets reduced in the process of oxidizing the second.

PERMANGANATE, DICHROMATE AND CERIC SULPHATE TITRATION METHODS

INTRODUCTION

The oxidation and reduction processes essentially take place simultaneously in a reaction, thus one entity gets reduced in the process of oxidizing the second. ‘Redox’—is the abbreviated form of reduction— oxidation systems. In the oxidation—reduction methods of analysis a change in valence of the reacting products is a must which is contrary to precipitation and neutralization methods of analysis where no change in valence occur. The major oxidizing agents normally employed in volumetric titrations include, potassium permanganate, potassium dichromate, and ceric sulphate.

Chapter: Pharmaceutical Drug Analysis: Permanganate, Dichromate and Ceric Sulphate Titration Methods

Theory - Permanganate, Dichromate and Ceric Sulphate Titration Methods

As a number of elements are capable of exhibiting more than one oxidation state, hence volumetric titration methods based on redox reactions are usually employed widely.

THEORY

As a number of elements are capable of exhibiting more than one oxidation state, hence volumetric titration methods based on redox reactions are usually employed widely.

 

The phenomenon of oxidation may be explained in the following manner :

 

(i)          addition of oxygen :

Example : 

(ii)       removal of hydrogen :

Example :  

(iii) enhancement in the ratio of electronegative to the electropositive portion of the molecule :

 

Examples :  


In the same vein, the process of reduction may also be explained as stated below :

(i) addition of hydrogen :


Example :


(ii) removal of oxygen :

 

Example :


(iii) enhancement in the ratio of electropositive to electronegative portion of the molecule :

Example : [same as under oxidation (iii) above]

It is quite evident from the above cited examples that reduction need not always imply a reaction involving hydrogen, since HgCl2 is reduced to Hg2Cl2, and that oxidation may not essentially suggest a reaction involving oxygen, since Fe2+ is oxidized by Cl2 to Fe3+. It is, therefore, pertinent to observe here that whenever one entity undergoes oxidation, definitely some other entity undergoes reduction correspondingly and vice-versa. In other words, there always exists a transfer of electrons in oxidation-reduction reactions, because in every such reaction the charge gained or lost by one substance must essentially be lost or gained by another.

A reducing agent is the reactant that loses electrons in an oxidation-reduction reaction :


Thus, the reactant containing a constituent atom or atoms are converted to a higher state of oxidation.

 

An oxidizing agent is the reactant that gains electrons in an oxidation-reduction reaction :


Thus, the reactant containing a constituent atom or atoms are converted to a lower state of oxidation.

 

The quantitative measurement of one of the reactants may be accomplished by the reaction derived from the combination of oxidizing and reducing agents, for instance


and hence, ferrous sulphate can be estimated quantitatively by its reaction with ceric sulphate.


Chapter: Pharmaceutical Drug Analysis: Permanganate, Dichromate and Ceric Sulphate Titration Methods

Assay Methods - Permanganate, Dichromate and Ceric Sulphate Titration Methods

The quantitative estimations of a number of pharmaceutical substances may be carried out by using a variety of potential oxidizing agents as stated below :

ASSAY METHODS

The quantitative estimations of a number of pharmaceutical substances may be carried out by using a variety of potential oxidizing agents as stated below :

 

(i) Permanganate Methods :

 

(a) Direct Titration Methods,

 

(b) Indirect Titration Methods, and

 

(c) Residual Titration Methods.

 

(ii) Dichromate Methods :

 

Direct titrations with Potassium Dichromate.

 

(iii) Ceric Sulphate Titration Methods :

 

Direct Titrations with Ceric Sulphate


Chapter: Pharmaceutical Drug Analysis: Permanganate, Dichromate and Ceric Sulphate Titration Methods

Permanganate Titration Methods

The vital application of potassium permanganate as a potential oxidizing agent in an acidic medium mainly rests on the reactions designated.

PERMANGANATE METHODS

 

The vital application of potassium permanganate as a potential oxidizing agent in an acidic medium mainly rests on the reactions designated by the following equations :

 

Chemically we have :


Ionically we have :


 

1. Preparation of 0.1 N Potassium Permanganate Solution

 

Materials Required : Potassium permanganate : 3.5 g.

 

Procedure : Weigh accurately about 3.2 g of potassium permanganate on a watch-glass. Transfer the contents to a 250 ml beaker containing cold water and stir vigorously with a glass rod to effect rapid dissolution. Decant the solution through a small plug of glass wool supported by a funnel, into a 1 litre volumetric flask thereby leaving the undissolved residues in the beaker. Add more DW to the beaker and repeat the above process till all the potassium permanganate gets dissolved. Finally make up the volume to the graduated mark and shake well so as to effect uniform mixing.

 

Note : (i) KMnO4 must be weighed on a watch-glass and not on any kind of paper since cellu-lose fibers are corrosively attacked by it,

 

(ii) Likewise, filtration of KMnO4 solution must be done though cleaned glass wool and not cotton wool, and

 

(iii) Avoid heat in the preparation of KMnO4 solution because traces of grease or other pos-sible contaminants on the glass vessels used can catalyse its decomposition.

 

2. Standardization of 0.1 N Potassium Permanganate Solution

 

Materials Required : Oxalic acid : 6.3 g ; sulphuric acid concentrated : 5 ml.

 

Theory : The standardization of potassium permanganate solution is based upon the following equations :


Procedure : Weigh accurately about 6.3 g of pure oxalic acid (AnalaR-Grade) into a 1 litre volumet-ric flask, dissolve in suffcient DW and make up the volume upto the mark. Pipette out 25 ml of this solution, add to it 5 ml of concentrated sulphuric acid along the side of the flask, swirl the contents carefully and warm upto 70°C. Titrate this against the potassium permanganate solution from the burette till the pink colour persists for about 20 seconds.

 

Precautions :

 

(i) Sufficient acid must be present, otherwise formation of a brown colour during titration may be observed,

 

(ii) Similar brown colouration can also be observed by using too high a temperature or by using a dirty flask, and

 

(i)          To avoid such anomalies always rinse the flask with solution of H2O2 and dilute H2SO4 before performing the titrations.

 

3. Direct Titration Methods

 

Hydrogen peroxide solution and potassium bromide are two pharmaceutical substances that may be estimated by employing 0.1 N potassium permanganate solution and adopting the direct titration method.

 

3.1. Hydrogen Peroxide Solution

 

Materials Required : Hydrogen peroxide solution : 10 ml ; 5 N sulphuric acid : 5 ml ; 0.1 N potas-sium permanganate.

 

Procedure : Dilute 10 ml of hydrogen peroxide solution to 250 ml with DW in a volumetric flask. To 25.0 ml of this solution add 5 ml of 5 N sulphuric acid and titrate with 0.1 N KMnO4 to a permanent pink end-point. Each ml of 0.1 N potassium permanganate is equivalent to 0.001701 g of H2O2.

 

Equations :

Chemically, we have :


Ionically we have :


Calculations : (For % w/v of H2O2)

 

The ‘volume strength’ of the hydrogen peroxide solution is the number of ml of oxygen at NTP* which may be produced by the complete thermal decomposition of 1 ml of H2O2 solution. Hence, decompo-sition takes place as designated by the following equation :


The IP limit of H2O2 solution is 5-7% w/v.

Now, let us consider a sample which contains 6.25 per cent w/v H2O2 :


 

3.2.  Potassium Bromide

 

Materials Required : Potassium bromide : 1.2 g ; sulphuric acid (36 N) ; 10 ml ; 0.1 N KMnO4.

Procedure : Weigh accurately about 1.2 g of potassium bromide and dissolve in DW and make up the volume to 1 litre mark with water in a volumetric flask. To 10.0 ml of the solution, add 100 ml of DW and 10 ml of (36 N) sulphuric acid along the side of the flask and a few glass beads (to avoid bumping of solution). Heat to boiling and while the solution is still boiling, titrate with 0.1 N KMnO4 added dropwise until the pink colour just persists. Each ml of 0.1 N KMnO4 is equivalent to 0.01190 g of KBr.

 

Equations :

 

The Br– is oxidised to bromine by acidified KMnO4, thus :


 

4. Indirect Titration Methods

 

In the indirect method of permanganate oxidation certain compounds are first converted by means of chemical reactions to an equivalent amount of oxalate which is then subsequently oxidized quantitatively by permanganate.

 

4.1. Assay of Cherry Juice for Malic Acid

 

In this particular assay the malic acid present in the cherry juice is estimated by the following three steps sequentially :

 

Step 1 : Conversion of malic acid to an equivalent amount of calcium salt,

 

Step 2 : Conversion of calcium salt to corresponding insoluble calcium oxalate, and

 

Step 3 : Liberation of oxalate and subsequent oxidation with permanganate.

 

Materials Required : Cherry juice : 10 ml ; calcium carbonate : 1.0 g ; ammonia TS : 1 ml ; ammonium oxalate TS : 15 ml ; diluted ammonia (1 in 49) : 25 ml ; diluted sulphuric acid (1 in 3 ; approximately 9 N) : 30 ml ; potassium permanganate 0.1 N.

 

Procedure : Place 10 ml of precisely measured cherry juice in a 125 ml flask and add to it 1 g of calcium carbonate. Heat the contents on a water-bath for 15 minutes while swirling periodically and filter. Wash the filter 2 to 3 times with 5 ml portions of DW. Add to the combined filtrate and washings 1 ml of ammonia TS followed by 15 ml of ammonium oxalate TS. Warm the contents on a water-bath for 15 minutes, filter through filter paper and wash the filter with 5 ml portions of a solution previously made by mixing 1 ml of ammonia TS with 49 ml of DW. Perforate the filter paper and wash the precipitate into the same flask with hot DW and followed by 30 ml of diluted sulphuric acid. The resulting solution is heated to 80°C and finally titrated with 0.1 N KMnO4. Each ml of 0.1 N KMnO4 is equivalent to 6.704 g of C4H6O5.

 

Equations : Malic acid first reacts with CaCO3 to yield the soluble calcium malate that goes into the filtrate, whereas the insoluble calcium carbonate is filtered off and rejected. Thus,


The interaction between calcium malate and ammonium oxalate results into an equivalent quantity of calcium oxalate by displacement mechanism which is subsequently precipitated :



 

5. Residual Titration Methods

 

The residual titration method for pharmaceutical substances using potassium permanganate solution are mainly of two categories, namely :

 

(i) titration wherein an excess of standard oxalic acid is added to the substance and then the excess of oxalic acid is back titrated with KMnO4, and

 

(ii) titration wherein an excess of standard KMnO4 solution is used to oxidize the product, and then the amount in excess is estimated by reduction with either :

 

(a) excess ferrous ammonium sulphate and back titrated with more of standard KMnO4, or

(b) excess standard oxalic acid.

 

5A. Assay of Sodium Nitrite

 

Materials Required : Sodium nitrite : 1.0 g ; 0.1 N potassium permanganate : 50 ml ; sulphuric acid (conc.) : 5 ml ; 0.1 N oxalic acid.

 

Procedure : Weigh accurately about 1 g of sodium nitrite and dissolve it in DW to make 100 ml in a volumetric flask. Transfer 10 ml of this solution into a mixture of 50 ml of 0.1 N KMnO4, 100 ml of water and add 5 ml of sulphuric acid along the side of the flask. Heat the contents to 40°C, allow it to stand for 5 minutes and add 25 ml of 0.1 N oxalic acid. Warm the resulting mixture to about 80°C on a steam-bath and titrate with 0.1 N KMnO4 solution. Each ml of 0.1 N potassium permanganate is equivalent to 3.450 mg of NaNO2.

Precautions : While adding NaNO2 solution

 

(i) Care should be taken to immerse the tip of the pipette beneath the surface of the permanganate mixture, otherwise the nitrous acid (volatile) generated by NaNO2 and H2SO4, would be lost, and

 

(ii) Oxidation of nitrous acid (HNO2) to nitric acid (HNO3) takes place sluggishly at ambient temperature and hence, it is necessary to warm it upto 40°C for 5 minutes to expedite completion of reaction.

Examples : Chemically we have :


i.e., each molecule of sodium nitrite loses two electrons.

Calculations :



Chapter: Pharmaceutical Drug Analysis: Permanganate, Dichromate and Ceric Sulphate Titration Methods

Dichromate Titration Methods

1. Preparation of 0.1 N Potassium Dichromate Solution 2. Standardization of 0.1 N Potassium Dichromate Solution

DICHROMATE METHODS

 

Potassium dischromate (K2Cr2O7) is a strong oxidizing agent, quite comparable to KMnO4 that nor-mally shows only one pertinent reduced oxidation state : Thus, chemically we have :


Ionically we have :


Potassium dichromate exhibits much greater stability in aqueous solution in comparison to potassium permanganate. Potassium dichromate possesses an inherent oranage colour that is not intense enough to serve its own end-point signal, specifically in the presence of the green Cr3+ ion, which is supposed to be present at the end-point. Hence, redox indicators are usually employed to locate the exact end-point e.g., barium diphenylamine sulphonate.

 

1. Preparation of 0.1 N Potassium Dichromate Solution

 

Materials Required : Potassium dichromate : 4.930 g.

Procedure : Weigh accurately 4.93 g of potassium dichromate previously powdered and dried at 20°C for 4 hours and dissolve in sufficient DW to produce 1 litre in a volumetric flask.

 

Note : Potassium dichromate can be obtained as a primary standard reagent and hence, standard solu-tions may be prepared determinately and stored for long periods of time.

Equations : Chemically we have :


Ionically we have :


From this equation it follows that the equivalent weight of potassium dichromate is 1/6th of the molecular weight i.e., 294.22/6 or 49.03 g.

 

2. Standardization of 0.1 N Potassium Dichromate Solution

 

It can be achieved by following these steps, namely :

 

(a) Preparation of Standard Solution of Mohr’s Salt FeSO4(NH4)2.SO4.6H2O :

 

Materials Required : Mohr’s salt : 4.9 g ; dilute sulphuric acid (1 in 3, approx. 9 N) : 20 ml.

 

Procedure : Weigh accurately about 4.9 g of pure sample of Mohr’s salt and transfer it to a 250 ml volumetric flask. Add 20 ml of dilute sulphuric acid and make up the volume to the mark with DW and finally mix the contents of the flask thoroughly.

 

Calculations : The quantity of Mohr’s salt required for 250 ml of the solution having a normality of 0.05 N can be calculated as follows :


(b) Standardization of 0.1 N K2Cr2O7 Solution :

Materials Required : Standard solution of Mohr’s salt (0.05 N) : 250 ml, sulphuric acid (2 N) : 20 ml ; potassium dichromate solution (0.1 N) : 1 litre.

 

Procedure : Transfer 20 ml of the primary standard solution (Mohr’s salt) to the titration flask and add 20 ml of 2 N sulphuric acid. Take the potassium dichromate solution in the burette. Put drops of freshly prepared potassium ferricyanide, K3[Fe(CN)6], solution in the grooves of a porcelain tile. Now, proceed with the titration of Mohr’s salt solution against K2Cr2O7 solution. Transfer drops of the titrated solution by means of a glass rod and mix with drops of the indicator, already taken in the groove-tile. Alternatively, pre-soaked and dried filter paper with K3[Fe(CN)6] solution can also be used in place of the groove-tile method.

 

In order to arrive at the exact end-point the above titration may be carried out at three stages, namely :

 

Stage 1 : Spot tests are carried out at intervals of 1-2 ml until a blue colour is no longer produced with K3[Fe(CN)6], which provides an altogether rough estimate of the K2Cr2O7 solution required for the titration,

 

Stage 2 : Spot tests are only performed near the approach of the end of titration at intervals of 0.1-0.2 ml, and

 

Stage 3 : Spot tests are finally done only at the end-point.

 

The above sequential steps give fairly accurate results because the error caused by the removal of part of the solution for the spot tests is made negligibly small. However, the titration is repeated to get a set of concordant readings.

 

By applying the relationship between N1V1 (K2Cr2O7) and N2V2 (Mohr’s salt), the normality of the former may be calculated.

 

2.1. Iron Ore

 

Materials Required : Iron ore : 0. 1 g ; hydrochloric acid (conc.) : 15 ml ; diphenylamine (1% w/v in conc. H2SO4) ; zinc metal (granulated) : 4 g ; ammonium thiocyanate solution (0.1% in water) ; mixture of sulphuric acid and phosphoric acid [dissolve 15 ml of H2SO4 (sp. gr. 1.84) in 50 ml of DW, cool and add 15 ml of H3PO4 (sp. gr. 1.70) and make the volume to 100 ml with DW] : 25 ml.

 

Procedure :

 

(a) Preparation of Standard K2Cr2O7 Solution : Instead of using solutions having definite normal-ity, routine industrial laboratories make use of ‘emperical solution’ which is normally expressed in terms of ‘titer for the substance determined’. For this assay, let us prepare an emperical K2Cr2O7 solution (250 ml) of such a concentration that 1 ml of the same exactly correspond to 0.0025 g Fe.

 

Calculations :


By Law of Equivalence, we have :

1 gram-equivalent of K2Cr2O7 (49.03 g) 1 gram-equivalent of Fe (55.85 g)


Therefore, weigh accurately 0.5488 g of pure K2Cr2O7 and transfer it quantitatively into a 250 ml volumetric flask, dissolve in DW, make up the volume and mix thoroughly.

Hence, the ‘iron titer’ of this solution is :


 

(b) Preparation of Ore Solution : Weigh accurately 0.1 g of powdered and dried ore on a clean watch glass and transfer it quantitatively into a 100 ml-volumetric flask. Add 15 ml of concen-trated hydrochloric acid, warm the contents of the flask carefully over a sand-bath until most of the dark grains of ore get dissolved completely and only a whitish silica precipitate settles at the bottom of the flask.

(c) Reduction of Fe3+ to F2+ in the Ore Solution : Introduce carefully a few pieces of granulated pure zinc metal into the flask, place a funnel in the neck of the flask to avoid splashes and boil the solution gently until the yellow colour has disappeared completely, thereby ascertaining that com-plete reduction of Fe3+ to Fe2+ is affected.

 

Note : It may be further confirmed by doing a spot test with NH4CNS solution which only shows a blood-red colour with Fe3+.

 

The contents of the flask is cooled, filtered through cotton wool, washings done with DW and the filtrate diluted to about 350 ml with DW. This dilution is a must so as to avoid any interference caused by its inherent green colour with the estimation of the equivalence point in the titration as per the following chemical reaction :

 

K2Cr2O7  +  6FeCl2  +  14HCl   →     2CrCl3  +  2KCl  +  6FeCl3  + 7H2O

 

(d) Final Titration : The 350 ml solution obtained in (c) above is now quantitatively titrated against K2Cr2O7 solution employing diphenylamine as an internal indicator. Add 25 ml of a mixture of sulphuric acid and phosphoric acid to the solution along with 2 drops of diphenylamine indicator and titrate the solution with K2Cr2O7 solution carefully, by adding small lots at intervals with constant shaking, until a persistant blue-violet colour appears.

 

Note : (a) The acidity of the solution must be maintained fairly high which can be achieved by adding orthophosphoric acid, H3PO4,

 

(b) The quantity of diphenylamine must not exceed 2 drops by virtue of the fact that at higher concentration with lower acidity during very slow titration, the indicator undergoes an altogether different type of chemical change that ultimately gives a green colour instead of the desired blue-violet colour.

 

(e) Calculations : Multiply the number of millilitres of K2Cr2O7 Solution consumed in the titration by the ‘iron titer’ and therefrom determine the amount of iron present in the sample. Finally, the percentage of iron present in the ore may be calculated.

Chapter: Pharmaceutical Drug Analysis: Permanganate, Dichromate and Ceric Sulphate Titration Methods

Ceric Sulphate Titration Methods

1. Preparation of 0.1 N Ammonium Ceric Sulphate Solution 2. Stadardization of 0.1 N Ammonium Ceric Sulphate Solution

CERIC SULPHATE TITRATION METHODS

 

Ammonium ceric sulphate serves as a powerful oxidizing agent in an acidic medium. The salt has a bright yellow colour and so its solution. On reduction, the resulting cerous salt obtained is colourless in appearance and, therefore, strong solutions may be considered as self-indicating. In general practice, 0.05 N solutions are employed invariably for estimations. As this concentration is very dilute for observation of the respective end-point, hence the inclusion of an appropriate indicator becomes necessary. The oxidation reac-tion involved may be expressed as follows :


It is interesting to observe that the solutions of ammoniurn ceric sulphate possess a number of advantages over permanganate and dichromate methods discussed earlier, viz.,

 

(i) solutions remain fairly stable even when boiled,

 

(ii) solutions quantitatively react with either arsenite (AsO33–) or oxalate [(COO)2]2– ion, and there-fore, either arsenic trioxide or sodium oxalate may be employed as a primary standard,

 

(iii) cerous ion Ce3+ is colourless and hence offers no interference with the indicator end-point,

 

(i)          Ce3+ always solely results on reduction of Ce4+, whereas permanganate (MaO4 ) can be reduced to any of several oxidation states,

 

(ii)       ammonium ceric sulphate unlike potassium permanganate, may be conveniently employed as an oxidizing agent in the presence of high concentrations of HCl, thereby facilitating determinations of Fe2+ in the presence of Cl, and

 

(iii)     ferrous phenanthrolone ion (ferroin) has proved to be a very successful indicator in titrations with ceric salts. Thus, we have :


Orthophenanthroline (base) dissolves rapidly in aqueous solutions of ferrous salts, thereby three moles combine with one Fe2+ ion to give a complex termed as ‘ferroin’ having an, intense red colour. Now, any strong oxidizing agent converts the ferrous to a corresponding ferric complex having a slight blue colour.

 

1. Preparation of 0.1 N Ammonium Ceric Sulphate Solution

 

Materials Required : Ceric ammonium sulphate : 66 g ; sulphuric acid (conc.) : 30 ml.

 

Procedure : Dissolve 66 g of ceric ammonium sulphate, with the help of gentle heat, in a mixture of ml of sulphuric acid and 500 ml DW. Cool, filter the solution through a fine-porosity sintered-glass crucible, dilute to 1 litre mark in a volumetric flask and mix thoroughly.

 

Since the oxidation reaction is given by :


 

2. Stadardization of 0.1 N Ammonium Ceric Sulphate Solution

 

Materials Required : Arsenic trioxide : 0.2 g ; sodium hydroxide solution (8.0% w/v) : 25 ml ; diluted sulphuric acid (10% w/v) : 30 ml ; osmic acid solution (1.0% w/v in water) : 0.15 ml ; ferroin sulphate solution (dissolve 0.7 g of ferrous sulphate in 70 ml of DW and add 1.5 g of 1, 10-phenanthroline and sufficient water to produce 100 ml) : 0.1 ml.

 

Procedure : Weigh accurately about 0.2 g of arsenic trioxide previously dried at 105°C for 1 hour and transfer to a 500 ml conical flask. Wash down the inner walls of the flask with 25 ml of sodium hydroxide solution, swirl to dissolve, add 100 ml of water and mix. Add 30 ml of diluted sulphuric acid, 0.15 ml of osmic acid solution, 0.1 ml of ferroin sulphate solution and slowly titrate with ceric ammonium sulphate solution until the pink colour is changed to a very pale blue. Each 4.946 mg of arsenic trioxide is equivalent to 1 ml of 0.1 N ammonium ceric sulphate or 0.06326 g of Ce(SO4)2. 2(NH4)2SO4 . 2H2O.

 

Equations :


It is evident from the above equations that 4 equivalents of ceric sulphate is required to oxidise 1 mole of arsenic trioxide, hence, 1 equivalent weight of arsenic trioxide is 1/4 mole or 197.84/4 or 49.46 g and 1 milliequivalent shall contain 49.46 mg or 0.04946 g.

 

Calculations : Therefore, the normality of ammonium ceric sulphate solution may be expressed as follows :


 

2.1.  Ferrous Fumarate

 

Materials Required : Ferrous fumarate : 0.3 g ; diluted H2SO4 (10% w/v) : 15 ml ; ferroin sulphate solution ; 0.1 N ammonium ceric sulphate solution.

 

Procedure : Weigh accurately about 0.3 g of ferrous fumarate and dissolve in 15 ml of dilute sulphu-ric acid by the help of gentle heating. Cool, add 50 ml of water and titrate immediately with 0.1 N ammonium ceric sulphate, employing ferroin sulphate solution as indicator. Each ml of 0.1 N ammonium ceric sulphate is equivalent to 0.01699 g of C4H2FeO4.

 

Equations and Calculations :


 

2.2.  Acetomenaphthone

 

Materials Required : Acetomenaphthone : 0.2 g ; glacial acetic acid : 15 ml ; dilute hydrochloric acid (10% w/v) : 15 ml ; ammonium ceric sulphate 0.05 N ; ferroin sulphate solution.

 

Procedure : Weigh accurately about 0.2 g of acetomenaphthone and boil it with 15 ml of glacial acetic acid and 15 ml of dilute hydrochloric acid under a reflux condenser for 15 minutes. Cool the contents carefully and taking adequate precautions to avoid any atmospheric oxidation. Add 0.1 ml of ferroin sulphate solution as indicator and titrate with 0.05 N ammonium ceric sulphate. Repeat the assay without the substance being examined (blank determination) and incorporate the correction, if any. Each ml of 0.05 N ammonium ceric sulphate is equivalent to 0.006457 g of C15H14O4.

 

Equations :


First, acetamenaphthone (I) undergoes hydrolysis in acidic medium to yield the corresponding phenol and secondly, this phenol is oxidised quantitatively with ammonium ceric sulphate to give the resulting 1, 4-dione derivative (II).

Calculations :

Thus, we have :


 

2.3.  Cognate Assays

 

A number of pharmaceutical substances and dosage forms may be determined by the help of ceric sulphate titration methods as given in Table 6.1.


Chapter: Pharmaceutical Drug Analysis: Iodimetric and Iodometric Titrations

Iodimetric and Iodometric Titrations

Iodimetric and iodometric titrations constitute another class of oxidation-reduction titrations wherein either iodine solutions are employed directly for the assay or an equivalent amount of iodine is liberated indirectly from the reaction mixture and then assayed.

IODIMETRIC AND IODOMETRIC TITRATIONS

 

NTRODUCTION

Iodimetric and iodometric titrations constitute another class of oxidation-reduction titrations wherein either iodine solutions are employed directly for the assay or an equivalent amount of iodine is liberated indirectly from the reaction mixture and then assayed.

 

Iodimetry is a procedure based on the following reversible reaction :

 

2I–   < = =>   I2 + 2e

 

Hence, it can be utilized for the quantitative estimation of reducing agents like arsenites (H3AsO3) and thiosulphates (Na2S2O3) by employing a standard solution of iodine.

 

Iodometry is an indirect procedure based on the aforesaid reversible reaction whereby the assay of oxidizing agents, for instance : ‘available chlorine’ in bleaching powder, cupric and ferric salts may be carried out by reducing them with an excess potassium iodide thereby liberating an equivalent quantity of iodine which can be estimated using a standard solution of thiosulphate.

Chapter: Pharmaceutical Drug Analysis: Iodimetric and Iodometric Titrations

Theory - Iodimetric and Iodometric Titrations

In iodimetry, quantitative oxidation of reducing agents, such as arsenious acid (H2AsO3) may be carried out by employing standard solutions of iodine as shown under :

THEORY

In iodimetry, quantitative oxidation of reducing agents, such as arsenious acid (H2AsO3) may be carried out by employing standard solutions of iodine as shown under :


This type of assay is known as ‘direct method of iodimetry’.

 

In another situation, a known excess quantity of standard iodine solution is added in the substance (a reducing agent) to be assayed and then the excess iodine may be titrated with the help of standard sodium thiosulphate solution, such as : the estimation of sodium bisulphite :


This category of assay is termed as ‘residual method of iodimetry’.

 

In iodometry, an equivalent amount of iodine is liberated when the given sample of an oxidizing agent oxidizes potassium iodide in an acidic medium, for example : the determination of cupric sulphate (CuSO4) :

2CuSO4  +  4 KI                           →     2CuI   +  I2  +  2K2SO4

 

Consequently, the equivalent amount of iodine generated by the above reaction may be conveniently assayed by titration against a standard sodium thiosulphate solution. In this context a point of caution must be observed while KI is being oxidized under a strongly acidic medium so as to avoid simultaneous oxidation of the iodide by atmospheric oxygen that may result high erroneous titer values leading to false estimations.

 

It is, however, pertinent to mention here that iodometric assays are never performed in a strongly basic medium, because of the fact that the reaction between I2 and OH produces hypoiodide and iodate ions respectively as shown below :

 

I2  +  OH   → HI  +  IO

3IO    IO3 +  2I

 

The said two ions partially oxidize thiosulphate to a higher oxidation form, such as sulphate (SO42–) thereby the stoichiometry achieved is always false.

 

 

Chapter: Pharmaceutical Drug Analysis: Iodimetric and Iodometric Titrations

Assay Methods - Iodimetric and Iodometric Titrations

Assay methods involving iodine can be categorized under the following heads namely :

ASSAY METHODS

Assay methods involving iodine can be categorized under the following heads namely :

 

A. Iodimetric Assays :

 

(a) Direct titration with iodine,

 

(b) Residual titration method : i.e., excess of iodine is titrated with sodium thiosuphate,

 

B. Iodometric Assays : 

i.e., release of iodine and subsequent titration with sodium thiosulphate.

Chapter: Pharmaceutical Drug Analysis: Iodimetric and Iodometric Titrations

Iodimetric Assays

In such estimations, the pharmaceutical substances can be measured either directly or back titration of excess iodine with sodium thiosulphate solution.

IODIMETRIC ASSAYS

 

In such estimations, the pharmaceutical substances can be measured either directly or back titration of excess iodine with sodium thiosulphate solution.

 

1. Direct Titration with Iodine

 

(a) Preparation of 0.1 Iodine Solution

 

Theory : Iodine in aqueous solution acts as an oxidizing agent which forms the basis of assay methods involving direct titration with iodine. Thus, we have :


 

Materials Required : Iodine : 3.2 g ; potassium iodide : 7.5 g.

 

Procedure : Weigh accurately 3.2 g of crushed iodine crystals on a watch glass and transfer to a beaker containing potassium iodide (7.5 g) and water (10 ml). Dissolve the contents of the beaker with the help of a glass rod and frequent swirling. Transfer the contents of the beaker quantitatively to a 250 ml volumetric flask and make up the volume with DW.

 

Explanation : Iodine is sparingly soluble in water but undergoes rapid dissolution in the presence of potassium iodide due to the formation of the corresponding triiodide ion :

 

I2  +  I I3

 

Thus, potassium iodide plays dual role, viz., in iodimetry—to solubilize iodine in aqueous KI solution, and in iodometry—as reducing agent, the excess KI helps in retaining liberated I2 in solution through interaction with KI.

 

(b) Standardization of 0.1 Iodine Solution with the aid of Arsenic Trioxide (As2O3)

 

Theory : This particular standardization is solely governed by the following equations, namely :


Hydroiodic acid (HI) possesses strong reducing characteristics which renders the oxidation with iodine into a reversible reaction as follows :

 


In order to shift the equilibrium to the right-hand-side (i.e., towards As2O5) in the above reaction, sodium bicarbonate (NaHCO3) is employed to remove the HI generated. It is important to record here that neither sodium hydroxide nor sodium carbonate can be used as both of them produce sodium iodide (NaI) and sodium iodate (NaIO3) as designated below :


Materials Required : Arsenic trioxide : 0.5 g ; sodium hydroxide solution (20% w/v in water) : 2 ml ; dilute hydrochloric acid (2N) ; sodium bicarbonate : 4 g ; 0.1 N iodine solution.

 

Procedure : Weigh accurately 0.5 g arsenic trioxide into a beaker, add to it 2 ml of sodium hydroxide solution, and heat to dissolve. Cool and transfer the contents quantitatively to a 100 ml volumetric flask and make up the volume upto the mark with DW. Pipette 20 ml into an iodine-flask, acidify with dilute HCl carefully and confirm it by adding a little NaHCO3 to remove the free excess acid, followed by a further 2 g to get rid of HI formed in the reaction mixture. Now, titrate with 0.1 N iodine solution till the end-point is achieved by the appearance of the first permanent pale straw colour.

 

(c) Standardization of 0.1 Iodine Solution by the aid of Sodium Thiosulphate

 

Theory : Iodine solution may also be standardized by using sodium thiosulphate (AR-Grade) whereby the latter gets oxidized to sodium tetrathionate as expressed below :


Materials Required : Sodium thiosulphate (AR) : 6.025 g ; 0.1 N I2 solution.

 

Procedure : Weigh accurately 6.025 g of sodium thiosulphate (AR) to a 250 ml volumetric flask. Dissolve it in DW, shake well and make up the volume to the mark with DW. Pipette 25 ml of 0.1 iodine solution into an iodine flask and titrate with the standard sodium thiosulphate solution (as primary standard) until the solution becomes almost colourless.

 

Note : Stock solutions of sodium thiosulphate may be preserved by the addition of a few drops of sodium hydroxide solution (20% w/v) which serves as stabilizer as well as prevents decomposition.

 

(d) Preparation of Starch Solution

 

Material Required : Starch (arrowroot) : 1.0 g.

 

Procedure : Weigh 1.0 g starch in a glass in a glass pestle-mortar and triturate thoroughly with 10 ml of cold DW. Boil separately 200 ml of DW in a beaker and add the starch paste to it with vigorous stirring. The resulting mixture is boiled gently for a further period of 30 minutes till a transluscent and thin liquid having an uniform consistency is obtained.

 

Note : (1) The prepared solution of starch undergoes rapid deterioration, hence it is always desired to use freshly prepared solution every day,

 

(2) It is now more or less believed that the iodine is held as an ‘absorption complex’ within the helical chain of the macromolecule β-amylose i.e., a component of most starches. However, another component, α-amylose, is undesirable because it produces a red-colouration with io-dine which is not readily reversible, and

 

(3) ‘Soluble Starch’ comprises principally of β-amylose, with the α-fraction having been removed. Always, it is a practice to prepare indicator-solutions from this product exclusively.

 

1.1. Analgin

 

Materials Required : Analgin : 0.4 g ; alcohol (95%) : 40 ml ; 0.01 N hydrochloric acid : 10 ml ; 0.1 N iodine solution.

 

Theory : The estimation of analgin depends upon the oxidation of the enolic group with iodine. The reaction is not reversible :


Procedure : Weigh accurately about 0.4 g and dissolve in a mixture of 40 ml of alcohol and 10 ml of 0.01 N hydrochloric acid. Titrate the resulting mixture with 0.1 N iodine solution till a yellow colour that remains stable for 30 seconds is achieved. Each ml of 0.1 N iodine is equivalent to 0.016670 g of C13H16N3NaO4S.

 

1.2. Acetarsol

 

Materials Required : Acetarsol : 0.25 g ; sulphuric acid (conc.) : 7.5 ml ; nitric acid (fuming) : 2.5 ml ; ammonium sulphate : 5 g ; potassium iodide : 1.0 g ; sodium sulphite (0.1 N) : 1.0 ml ; phenolphthalein solution : 2 drops ; NaOH solution (0.1 N) ; dilute sulphuric acid (6 N) ; sodium bicarbonate : 8.0 g ; iodine solution (0.1 N).

 

Theory : Acetarsol is an organic arsenal, hence arsenic may be estimated by carrying out the oxidation As3+ to As5+ state with the help of 0.1 N iodine solution.

 

The organic entity present in acetarsol is destroyed primarily by boiling it with aqua-regia (a mixture of conc. H2SO4 and fuming nitric acid). The resulting mixture is heated in the presence of ammonium sulphate to  get rid of nitric acid finally in the form of nitrous oxide (N2O) as follows :


Previously added H2SO4 maintains an acidic medium which on adding KI liberates HI that reduces the As5+ to As3+ state. Reduction is completed by boiling the solution which also expels the liberated I

as shown below :


The resulting mixture is cooled to room temperature and the residual iodine is removed by titration with 0.1 N sodium sulphite solution. Now, the solution is treated with sodium hydroxide solution to make it alkaline and then acidified carefully with dilute H2SO4 to remove the free NaOH. Finally, the resulting solution is made alkaline with NaHCO3 so that the equilibrium is shifted to the right (i.e., AS3+ gets converted to As5+) quantitatively on carrying out the titration with 0.1 N iodine solution. Thus, we have :


Procedure : Weigh accurately about 0.25 g of acetarsol into a 500 ml iodine flask and add to it sulphu-ric acid (conc.) 7.5 ml, followed by nitric acid (fuming) 1.5 ml. Boil the contents of the flask gently for 45 minutes preferably in a fume-cupboard. Cool the solution, add 0.5 ml of fuming HNO3 and boil till brown vapours (N2O) stop coming. Again cool the contents and add carefully 5 g of ammonium sulphate in small lots at intervals and heat till there is no evolution of N2O thereby giving rise to a colourless liquid. Bring the solution to room temperature, dilute with 100 ml DW, add 1 g KI and heat gently till the volume becomes 50 ml. Cool and add a few drops of 0.1 N sodium sulphite to effect decolourisation. Add 60 ml DW to dilute the resulting contents and make it just alkaline with NaOH solution by adding phenolphthalein indicator. Finally, acidify with dilute H2SO4, neutralize with NaHCO3 and add 4 g of NaHCO3 in excess. Swirl the contents of the flask and titrate with 0.1 N iodine solution. Each ml of 0.1 N iodine solution is equivalent to 0.01375 g of C8H10AsNO5.

 

1.3. Cognate Assays

 

The following pharmaceutical substances can be assayed by direct titration with iodine as stated in Table 7.1.


 

2. Residual Titration Method (Excess of Iodine Titrated with Sodium Thiosulphate)

 

In this titration method an excess of iodine solution is added to the solution of the substance and thus, the latter gets oxidized quantitatively. The excess of iodine is subsequently back titrated with sodium thiosulphate using freshly prepared starch solution as indicator with an end-point from violet to colourless.

 

2.1. Benzylpenicillin

 

Theory : Benzylpenicillin can be assayed efficiently by adopting the following three steps sequentially, namely :

 

Step 1 : Benzylpenicillin is first converted to the corresponding penicilloic acid (a dicarboxylic acid) by carrying out the hydrolysis with sodium hydroxide solution, as follows :


Step 2 : Penicilloic acid on treatment with acid yields D-penicillamine and benzylpenilic acid, as shown under :


Step 3 : D-Penicillamine thus obtained is oxidised quantitatively by iodine to give rise to a disulphide, as expressed in the following equation ; whereas, the excess iodine is back titrated with 0.02 N sodium thiosulphate solution :


From the above reaction, we have :

 

C16Hl7N2NaO4S I e

 

In usual practice, however, benzylpenicillin sodium is standardised against a chemical reference sub-stance of pre-determined potency.

 

Materials Required : Benzylpenicillin : 0.1 g ; (N) sodium hydroxide solution : 5 ml ; buffer solution (5.44% w/v of CH3COONa and 2.40%  w/v  of  glacial  acetic  acid) : 20 ml ; (N)  hydrochloric acid : 5 ml ; 0.02 N iodine solution : 25 ml ; 0.02 N sodium thiosulphate solution ; starch solution.

 

Procedure : Weigh accurately about 0.1 g of benzylpenicillin in DW and dilute to 100 ml in a volumet-ric flask. Transfer 10.0 ml to an iodine flask, add 5 ml of N sodium hydroxide and allow to stand for 20 minutes. Now, add 20 ml of freshly prepared buffer solution, 5 ml of N HCl and 25.0 ml of 0.02 N iodine solution. Close the flask with a wet glass-stopper and allow to stand for 20 minutes in a dark place (i.e., protected from light). Titrate the excess of iodine with 0.02 N sodium thiosulphate, employing freshly pre-pared starch solution as an indicator added towards the end-point.

 

To another 10.0 ml of the initial solution add 20 ml of the buffer solution, allow to stand for 20 minutes in the dark and titrate with 0.02 N sodium thiosulphate, using starch solution, added towards the end of the titration as indicator.

 

The difference between the two titrations represents the volume of 0.02 N iodine equivalent to the total penicillins present in the given sample of benzylpenicillin. An assay may be carried out simultaneously by benzylpenicillin sodium (reference sample) so as to determine the exact equivalent of each ml of 0.02 N iodine.

 

Calculations : Calculate the potency in Units of penicillin from the declared number of Units of pencillin in benzylpenicillin sodium (reference sample).

 

2.2. Sodium Metabisulphite

 

Theory : Sodium metabisulphite in acidic medium (HCl) yields SO2 which reacts with water to pro-duce sulphurous acid. The generated sulphurous acid is quantitatively oxidized by iodine to sulphuric acid, and the excess iodine is subsequently back titrated with sodium thiosulphate. The various reactions can be expressed as shown below :


Materials       Required :        Sodium       metabisulphite : 0.2 g ; 0.1 N Iodine solution ; hydrochloric acid ( ~  11.5 N) : 1 ml ; 0.1 N sodium thiosulphate ; starch solution.

 

Procedure : Weigh accurately about 0.2 g of sodium metabisulphite and dissolve in 50.0 ml of 0.1 N iodine solution and add 1 ml hydrochloric acid. Titrate the excess of iodine with 0.1 N sodium thiosulphate employing freshly prepared starch solution, added towards the end of the titration, as indicator. Each ml of 0.1 N iodine is equivalent to 0.0047453 g of Na2S2O5.

 

2.3. Cognate Assays

 

A few other pharmaceutical substances may also be assayed by adopting the residual titration method as shown in Table 7.2.


 

Chapter: Pharmaceutical Drug Analysis: Iodimetric and Iodometric Titrations

Iodometric Assays

In iodometric determinations the pharmaceutical substance oxidizes KI in an acidic medium to produce an equivalent quantity of iodine that may be assayed by titration with a standard solution of sodium thiosulphate.

IODOMETRIC ASSAYS

 

In iodometric determinations the pharmaceutical substance oxidizes KI in an acidic medium to produce an equivalent quantity of iodine that may be assayed by titration with a standard solution of sodium thiosulphate.

 

1. Chlorinated Lime

 

Chlorinated lime or bleaching powder, CaOCl2, contains about 30% w/w of available chlorine.

 

Theory : Chlorinated lime reacts with acetic acid to produce a mole each of calcium acetate, hydro-chloric acid and hydrochlorous acid. The two acids interact to give water and chlorine, and the latter reacts with HI to liberate iodine that can be estimated by titrating with 0.1 N sodium thiosulphate solution. The various reactions involved may be expressed as given below :


 

Materials Required : Chlorinated lime : 4 g ; dilute acetic acid : 5 ml ; potassium iodide : 3 g ; acetic acid : 5 ml ; 0.1 N sodium thiosulphate solution.

 

Procedure : Weigh accurately 4.0 g of chlorinated lime and triturate it in a glass-pestle-mortar with a little DW. Transfer the paste quantitatively into a 1 litre volumetric flask and shake thoroughly. Take a 100 ml volumetric flask, rinse it with a small quantity of the suspension from the 1 litre flask and finally fill it up with the suspension. Rinse out a 250 ml iodine flask containing a little dilute acetic acid and a little of the suspension from the 1-litre flask in order to oxidise any inorganic substance present in the iodine flask. Finally, wash it thoroughly with DW. Now, transfer 100 ml of the suspension completely from the 100 ml volumetric flask to the iodine flask by washing the former repeatedly with DW. Add to it acetic acid 5 ml followed by KI 3.0 g and shake the contents of the flask thoroughly. Titrate the liberated iodine with 0.1 N sodium thiosulphate which is equivalent to 0.003546 g of chlorine.

 

From this value the percentage of chlorine present in the given sample of chlorinated lime can be calculated.

 

2. Ferric Ammonium Citrate

 

Theory : In ferric ammonium citrate it is taken for granted that the entire iron is oxidized to the Fe2+ state and practically little Fe2+ is present. Thus, the ferric ion present in a known amount of the sample liberates an equivalent amount of iodine from an acidified KI solution. Thus, we have :


Materials Required : Ferric ammonium citrate : 0.5 g ; sulphuric acid conc. : 1 ml ; 0.1 N KMnO4 solution : 50 ml ; hydrochloric acid : 15 ml ; potassium iodide : 2.0 g ; 0.1 N sodium thiosulphate.

 

Procedure : Weigh accurately about 0.5 g of ferric ammonium citrate and dissolve the sample in 15 ml DW. Add to it slowly 1 ml of sulphuric acid and warm gently to attain a yellow colouration so as to decompose the iron and ammonium citrate complex completely. Cool and add 0.1 N potassium permanganate solution dropwise from a burette to obtain a pink colour that persists for 5 seconds. To the resulting solution add hydrochloric acid 15 ml and potassium iodide 2.0 g, shake well and set aside for 3 minutes so that iodine may be liberated completely. Now, add 60 ml of water and titrate with 0.1 N sodium thiosulphate solution while shaking the contents continuously till a colourless end-point is achieved.

 

Precautions :

 

(i) Addition of excess of KMnO4 solution must be avoided, since pink colour developed shall disap-pear within a short span, which may ultimately give false high results,

 

(ii) Washing down during the course of titration must be checked rigidly in order to maintain the right proportion of various substances in the solution,

 

(iii) End-point is almost colourless, hence starch indicator can be skipped totally, and

 

(iv) KMnO4 oxidizes the traces of Fe2+ to Fe3+ in the sample, if any.

 

3. Thyroid

 

Thyroxine and diidotyrosine are the two iodine-substituted organic compounds which essentially con-stitute the active principles present in dried thyroid gland. The latter on being subjected to pyrolysis with anhydrous K2CO3, gives rise to an equivalent amount of KI present in the sample. Soon after the completion of carbonization, the crucible is cooled and the residue is extracted with water to dissolve KI, carbonates and other soluble compounds. The resulting solution is filtered and treated with Br2 in the presence of phosphoric acid (H3PO4) so that complete oxidation of iodide to iodate is caused. The following reaction takes place :


The excess of bromine is removed by warming the acidic solution gently till the vapours show a negative test with starch-iodide paper. However, the residual traces of Br2 are reduced by treatment of the resulting solution with phenol to yield the corresponding 2,4,6-tribromophenol as shown below :


Lastly, iodate (IO3) in a weak acidic medium quantitatively oxidizes KI to an equivalent amount of iodine, as expressed below :


It is evident from the above equation that each gram-atomic weight of iodine in thyroid is converted to 1 mol of iodate and finally to 3 mol or 6 equivalent of iodine. Therefore, the equivalent weight of the iodine present in the dried thyroid gland is 21.15 g (i.e., 1/6 × 127 At. wt. of I 2). Hence, each millilitre of 0.01 N sodium thiosulphate is equivalent to 0.0002115 g of iodine (i.e., 0.01 × 0.02115 g).

Materials Required : Thyroid gland dried 1.0 g ; anhydrous potassium carbonate : 17.0 g ; bromine solution (9.6 ml of Br2 and 30 g of KBr in 100 ml DW) : 7.0 ml ; dilute phosphoric acid (10% w/v) : 42.0 ml ; starch iodide paper ; phenol solution (saturated solution of phenol in water) : 5.0 ml ; potassium iodide solution (10% w/v in water) ; 0.01 N sodium thiosulphate solution ; starch solution.

 

Procedure : Weigh accurately about 1.0 g of dried thyroid gland in a porcelain crucible, add 7.0 g of anhydrous K2CO3, mix thoroughly and overlay with further 10 g more of anhydrous K2CO3, finally compact the mixture by tapping gently. Incenerate for 25 minutes at 675°—700°C in a preheated muffle furnace. Cool the contents, add 20 ml of DW, boil gently and decant through a filter paper into a flask. Repeat the extraction by boiling with 20 ml DW, wash the crucible and the residue on the filter with hot water until the filtrate is about 200 ml. To it add 7.0 ml of freshly prepared bromine solution followed by 40 ml of dilute phosphoric acid and continue boiling slowly till starch iodide paper is no longer coloured blue by the vapours. While boiling is in progress top up the volume to 200 ml by adding DW at intervals. Cool and add 5 ml of phenol solution and allow to stand for 5 minutes. Add 2 ml of dilute phosphoric acid and 5 ml of potassium iodide solution and titrate immediately with 0.01 N sodium thiosulphate solution employing starch solution as indi-cator towards the end-point. A blank estimation is also carried out simultaneously and necessary correction incorporated. Each ml 0.1 N sodium thiosulphate is equivalent to 0.0002115 g of I.

 

Precautions :

 

(i) Potassium carbonate should be perfectly anhydrous otherwise decrepitation would take place caus-ing loss of material during pyrolysis,

 

(ii) Both the temperature of the muffle furnace and the extent of heating should be monitored closely, because KI is significantly volatile at an elevated temperature and part of it may be lost due to extended heating, and

 

(iii) The solution from which excess Br2 is removed by heating must be acidic, otherwise a portion of Br2 shall be fixed in the form of potassium hypobromite (KBrO).

 

4. Cognate Assays

 

A few pharmaceutical substances can be assayed by titrating the liberated iodine from potassium iodide with sodium thiosulphate as stated in Table 7.3.

Chapter: Pharmaceutical Drug Analysis: Iodimetric and Iodometric Titrations

Argentometric Precipitation Methods

In general, titrations governed by precipitation reactions do not really constitute an appreciable number in volumetric determinations in comparison to either redox or acid-base reactions.

ARGENTOMETRIC PRECIPITATION METHODS

 

INTRODUCTION

 

In general, titrations governed by precipitation reactions do not really constitute an appreciable number in volumetric determinations in comparison to either redox or acid-base reactions. The interaction between silver-nitrate and sodium chloride in solutions result into the precipitation of silver chloride as shown below :

 

NaCl  +  AgNO3   → AgCl   +  NaNO3

 

In actual practice, however, such titrations are more or less restricted to those involving precipitation of Ag+ with anions, for instance : halogens (Cl, Br, I) and thiocyanate (SCN). Generally, it is quite difficult and tedious to locate the exact point at which further addition of reagent affords no more precipitation. There-fore, the choice and wisdom of a chemical reaction is preferably sought so as to result in either a coloured solution or a coloured precipitate at the end point. A typical instance may be cited by application of potassium chromate (K2CrO4) solution in the above case whereby any extra drop of silver nitrate, after all the chloride has been precipitated, immediately causes precipitation of red chromate showing that the end point has been duly achieved.

 

It is, however, interesting to observe here that such reactions do offer limited usage because of the following two facts, namely :

 

(i) Co-precipitation effects do not give a real composition of the precipitate, and

 

(ii) Choice of appropriate indicator is very much limited.

 

Besides, the foregoing facts another vital aspect to be taken into consideration is the solubility product that plays a major role in such titration. Hence, the equilibrium constant of the reaction giving the precipitate of AgCl may be expressed as :


From the above expression the solubility product constant Ksp may be designated as :


assuming the activity of solid AgCl being constant.

Following are the four cardinal parameters that may be considered for a feasible argentometric analysis, namely :

 

(i) Precipitate formed must be insoluble,

 

(ii) Precipitation process should be fast and rapid,

 

(iii) Co-precipitation effects must be minimal, and

 

(iv) Detection of equivalence point must be apparently visible.


Chapter: Pharmaceutical Drug Analysis: Iodimetric and Iodometric Titrations

Argentometric Precipitation Methods: Theory

In the precipitation reaction involving chloride and silver nitrate, the addition of even a small quantity of the latter shall effect precipitation of AgCl provided that Ksp has been exceeded significantly.

THEORY

In the precipitation reaction involving chloride and silver nitrate, the addition of even a small quantity of the latter shall effect precipitation of AgCl provided that Ksp has been exceeded significantly. At this juncture, the concentrations of both Ag+ and Cl are related by the solubility-product equilibrium constant  thus, we have :


Chromate ion concentration required to initiate the precipitation of Ag2CrO4 commences at the equivalence point and may be calculated with the solubility products for AgCl and Ag2CrO4 :


In actual practice, the concentration of chromate produces an intense yellow colour to such an extent that the end point is masked. Therefore, normally concentrations of 5 × 10 –3 M are employed in analytical procedures. It suggests that [Ag+] shall be > 1.3 × 10 –5 M at the end-point thereby introducing a positive determinate error. However, it has been proved experimentally that even with concentrations as low as 2 × 10 –3 M, the extent of error caused is negligibly small.

 

Adsorption-coprecipitation phenomenon using fluorescein, dichlorofluorescein and tetrabromofluo-rescein (eosin) essentially impart the fluoresceinate ion that is absorbed on the AgCl particles. At the equiva-lence point, the AgCl particles change from white to pink due to the coprecipitation of silver fluoresceinate. In short, the adsorption indicator method is quite rapid and capable of providing very accurate results for the estimation of Cl with AgNO3.

 

Furthermore, Br, I and SCN ions can also be titrated with AgNO3 employing eosin as an adsorption indicator.


Chapter: Pharmaceutical Drug Analysis: Iodimetric and Iodometric Titrations

Argentometric Precipitation Methods: Assay Methods

Argentometric titrations may be divided into two broad categories, namely : (i) Direct titration with silver-nitrate, and (ii) Ammonium thiocyanate-silver nitrate titrations (Volhard’s Method).

ASSAY METHODS

Argentometric titrations may be divided into two broad categories, namely :

 

(i) Direct titration with silver-nitrate, and

 

(ii) Ammonium thiocyanate-silver nitrate titrations (Volhard’s Method).

 

1. DIRECT TITRATION WITH SILVER NITRATE

 

Pharmaceutical substances essentially containing halides may be estimated by direct titration with silver nitrate solution as a titrant.

 

1.1 Preparation of 0.1 N Silver Nitrate Solution

 

Materials Required : Silver nitrate (AR) : 16.989 g.

 

Procedure : Weigh accurately 16.989 g of silver nitrate on a watch-glass and transfer quantitatively into a 1 litre volumetric flask. Add freshly prepared DW and make up the volume to 1000 ml. Thus, we have :


 

1.2. Standardization of 0.1 N Silver Nitrate Solution

 

Materials Required : Sodium chloride : 0.1 g ; acetic acid (33% w/v) : 5 ml ; methyl alcohol (95%) : 50 ml ; eosin solution (0.5% w/v in water) : 5 ml ; 0.1 N silver nitrate solution.

 

Procedure : Weigh accurately about 0.1 g of sodium chloride, previously dried at 110°C for 2 hours, and dissolve in 5 ml of water. Add 5 ml of acetic acid, 50 ml of methyl alcohol and three drops of eosin solution. Stir thoroughly on a magnetic stirrer and titrate with the silver nitrate solution till the white particles of AgCl change from white to pink. Each 0.005844 g of sodium chloride is equivalent to 1 ml of 0.1 N silver nitrate.

 

1.2.1.  Potassium Chlroride

 

Materials Required : Potassium chloride : 0.25 g ; potassium chromate solution (5% w/v in water) : 10 ml ; 0.1 N silver nitrate solution.

 

Procedure : Weigh accurately about 0.25 g of potassium chloride in a conical flask and dissolve it in 50 ml of DW and titrate with 0.1 N silver nitrate solution, using 2-3 drops of potassium chromate solution as indicator till precipitation of red chromate is indicated. Each ml of 0.1 N silver nitrate solution is equivalent to 0.007455 g of KCl.

Equations :


 

1.2.2. Chloral Hydrate

 

Materials Required : Chloral hydrate : 4.0 g ; sodium hydroxide (N) : 30 ml ; sulphuric acid (N) ; phenolphthalein solution (1.0% w/v in 50% v/v alcohol) ; 0.1 N silver nitrate solution ; potassium chromate solution (5% w/v in water).

 

Procedure : Weigh accurately about 4 g of chloral hydrate and dissolve in 10 ml of DW and add 30 ml of N sodium hydroxide solution. Allow the resulting mixture to stand for 2 minutes, and then titrate with N sulphuric acid, employing phenolphthalein solution as indicator till a colour change from pink to colourless is achieved. Titrate the neutralized liquid thus obtained with 0.1 N silver nitrate using potassium chromate solution as indicator till precipitation of red chromate is obtained, Add, now 2/15th of the amount of 0.1 N silver nitrate used to the amount of N sulphuric acid used in the first titration and deduct the figure so obtained from the amount of N sodium hydroxide added. Each ml of N sodium hydroxide, obtained as difference, is equivalent to 0.1654 g of C2H3Cl3O2.

 

Explanation :

 

(i) The estimation depends upon the interaction between chloral hydrate and sodium hydroxide as shown by the following equation :


(ii) As the chloroform generated in Eq. (a) undergoes chemical reaction with the alkali to a certain degree; therefore, addition of alkali followed by back titration does not afford the correct assay. Thus, we have :


(iii) The ionized chloride generated from the additional side reaction (b) may be estimated by titration with 0.1 N silver nitrate solution, and necessarily a correction has got to be made to the alkali-titration reading so as to adequately compensate for this side reaction. Thus, from equation (b) we have :


Therefore, it is evident that 2/15th of the volume of 0.1 N AgNO3 (i.e., 2/15th of 30,000 = 4,000) needed shall give the volume of N NaOH that reacted with chloroform as per Eq. (b).

 

1.2. Cognate Assays

 

The pharmaceutical substances in Table 8.1, can be assayed by direct titration with silver nitrate using a suitable indicator.


 

2. AMMONIUM THIOCYANATE-SILVER NITRATE TITRATIONS (VOLHARD’S METHOD)

 

Volhard’s method is based on two major aspects, namely :

 

(a) Complete precipitation of insoluble silver salts from nitric acid solution by adding an excess of silver nitrate solution to a corresponding soluble salt, and

 

(b) Estimation of excess of silver nitrate solution by carrying out residual titration with standard am-monium thiocyanate solution, employing ferric ammonium sulphate as an indicator.

 

Thus, ammonium thiocyanate reacts with silver nitrate in nitric acid solution as below :

 

NH4SCN + AgNO3 AgSCN + NH4NO3

 

However, in actual practice the thiocyanate solution is always taken in the burette and is run directly into the silver nitrate solution in the flask that has been duly acidified with nitric acid. Ferric ammonium sulphate is the choicest indicator since the end point is visibly detected by a deep red colour (ferric thiocyanate) due to the interaction of Fe2+ ions with a trace of SCN ion.

 

Precautions :

 

(i) Nitric acid must be free from nitrous acid, otherwise thiocyanic acid may give an instant red colouration, and

 

(ii) Temperature of the solution should be maintained below 25°C since at an elevated temperature the red colour of the ferric thiocyanate complex fades away rapidly. Therefore, we have :

 

NH4SCN AgNO3 H

 

2.1. Preparation of 0.1 N Ammonium Thiocyanate Solution

 

Materials Required : Ammonium thiocyanate : 8.0 g.

 

Procedure : Weigh about 8.0 g of ammonium thiocyanate and transfer it quantitatively in 1 litre volu-metric flask. Dissolve it in DW and make up the volume upto the mark.

Equation :


 

2.2. Standardization of 0.1 N Ammonium Thiocyanate Solution

 

Materials Required : 0.1 N Silver nitrate solution : 25 ml ; nitric acid (16 N) : 2 ml ; ferric ammonium sulphate (10% w/v in water) : 2 ml ; 0.1 N ammonium thiocyanate solution.

 

Procedure : Pipette 25 ml of a standard 0.1 N AgNO3 solution into a glass-stoppered flask (iodine-flask), dilute with 50 ml of DW, add to it 2 ml of nitric acid and 2 ml of ferric ammonium sulphate solution and titrate with ammonium solution to the first appearance of red-brown colour. Each ml of 0.1 N silver nitrate is equivalent to 0.007612 g of NH4SCN.

 

Note : Soon after the addition of ammonium thiocyanate a white precipitate of silver thiocyanate is formed first and then a reddish-brown colour appears that fades out completely upon shaking thereby leaving a white precipitate of silver thiocyanate. The end-point is indicated by the appearance of a permanent faint reddish brown colour that does not vanish upon shaking.

 

2.2.1.  Chlorobutol

 

Materials Required : Chlorobutol : 0.2 g ; alcohol (95%) : 5 ml ; sodium hydroxide solution (20% w/v in water) : 5 ml ; nitric acid (16 N) : 5 ml ; nitrobenzene : 1 ml ; 0.1 N silver nitrate solution : 50 ml ; ferric ammonium sulphate solution (10% w/v in water) ; 0.1 N ammonium thiocyanate solution.

 

Procedure : Weigh accurately about 0.2 g of chlorobutol in a flask and dissolve in 5 ml of alcohol. Add to it 5 ml of sodium hydroxide solution, and boil under a reflux condenser for 15 minutes. Cool, dilute with 20 ml of DW, add 5 ml of nitric acid, 1 ml of nitrobenzene and 50 ml of 0.1 N silver nitrate solution. Shake the contents vigorously for 1 minute, add 4 ml of ferric ammonium sulphate solution and titrate the excess of silver nitrate with 0.1 N ammonium thiocyanate solution. Each ml of 0.1 N silver nitrate is equivalent to 0.005917 g of C4H7Cl3O.

 

Explanation : Chlorine combined originally to chlorobutol is being converted by hydrolysis in the presence of sodium hydroxide to ionic chloride that may be estimated quantitatively by Volhard’s method in the presence of nitrobenzene.

Thus, we have :


 

2.2.2.  Ethionamide

 

Theory : Theoretically the cleavage of thioamide link in ethionamide takes place in an acidic medium. Subsequent neutralization with NH4OH yields ammonium sulphide which on addition of silver nitrate yields a precipitate of Ag2S. Thus we have :


 

Materials Required : Ethionamide : 0.3 g ; dilute sulphuric acid (10% w/w) : 10 ml ; dilute ammonia solution (4.25 ml of strong ammonia solution in 100 ml of water) ; 0.1 N silver nitrate : 50 ml ; dilute nitric acid (10.6 ml of nitric acid to 100 ml of water) : 60 ml ; ferric ammonium sulphate solution (10% w/v in water) : 5 ml ; and 0.1 N ammonium thiocyanate solution.

 

Procedure : Weigh accurately about 0.3 g of ethionamide in a flask and dissolve in 10 ml of dilute sulphuric acid. Add to it 100 ml of water, 20 ml of dilute ammonia solution and rapidly 50 ml of 0.1 N silver nitrate solution. Allow the resulting mixture to stand for a few minutes, filter and wash the filter paper with three successive quantities, each of 10 ml of DW. To the combined filtrate and washings, add 60 ml of dilute nitric acid, cool and titrate with 0.1 N ammonium thiocyanate employing 5 ml of ferric ammonium sulphate solution as an indicator. Each ml of 0.1 N silver nitrate is equivalent to 0.008312 g of C8H10N2S.

 

2.2.3. Cognate Assays

 

A good number of pharmaceutical substances can be assayed by Volhard’s method and are mentioned in Table 8.2.


Chapter: Pharmaceutical Drug Analysis: Complexometric Analysis

Complexometric Analysis

A host of inorganic pharmaceutical substances essentially containing polyvalent and bivalent metal ions, for instance :

COMPLEXOMETRIC ANALYSIS

INTRODUCTION

A host of inorganic pharmaceutical substances essentially containing polyvalent and bivalent metal ions, for instance : Al3+, Ca2+, Mg2+, Bi2+ and Zn2+ were initially analysed quantitatively by the aid of gravimetric methods. These historically important procedures have become more or less obsolete since they are extremely time consuming and tedious owing to several steps involved e.g., precipitation, filtration, washing, drying and finally ignition to a constant weight.

 

Thereafter, analysis was done by faster techniques based on oxalate-permanganate titrations which involved a precipitation procedure followed by quick titration of pre-heated solutions containing oxalate ions.

 

More recently the introduction of an analytical reagent disodium ethylene-diaminetetraacetate, invariably termed as EDTA, an altogether latest titrimetric method has been used exclusively for the estimation of metals using metal-ion indicators.


Chapter: Pharmaceutical Drug Analysis: Complexometric Analysis

Complexometric Analysis: Theory

1. Effect of pH on Complexation 2. Stability of Complexes 3. Colouration of Complexes 4. Titrability of Polyvalent Metal Ions Employing Disodium Edetate 5. Usage of pM Indicators in Complexometric Titrations

THEORY

Complex is a compound that is formed by the combination of a metal ion with a molecule that is capable of donating electrons, for example :


In the above two examples both Cu2+ and Co2+ form complexes with lone pair of electrons present in the neutral molecule ammonia e.g., N’’H3.

 

Chelate is a complex that is formed by the combination of a polyvalent metal ion with a molecule which essentially contains two or more groups that can donate electrons.

 

Specifically, disodium ethylenediaminetetraacetate (EDTA) reacts with polyvalent metal ions to result in the formation of a fairly stable water-soluble complex, or a chelate compound.

 

It is, however, pertinent to mention here that the predominant state of the dissociated forms of EDTA (viz . Y4–, HY3– , H2Y2– and H3Y) is solely dependent upon the pH of the medium at which complexation takes place :

 

where, H4Y = ethylenediaminetetraacetic acid, and

Y4– = tetracetate ion.

In general, all EDTA complexation reactions essentially have the ratio of EDTA to metal ion as 1 : 1 Thus, we have :


Ligand is a molecule that affords groups for attachment to metal ions such as EDTA.

 

Some examples of polyvalent metal ions are given below :

 

Bivalent Metal ions : Ca2+, Mg2+, Zn2+,

 

Trivalent Metal ions : Fe3+, Al3+, Cr3+,

 

Tetravalent Metal ions : Sn4+, Ce4+, Cr4+, Pt4+.

 

The structures of the complexes formed with di-, tri- and tetra-valent metal ions give rise to three, four and five rings respectively as depicted below :


There are various aspects in complex formation and detection, namely :

 

(i) Effect of pH on complexation,

 

(ii) Stability of complexes,

 

(iii) Colouration of complexes,

 

(iv) Titrability of polyvalent metal ions employing disodium edetate, and

 

(v) Usage of pM indicators in complexometric titrations.

 

 

1. EFFECT OF pH ON COMPLEXATION

 

Ethylenediamine tetracetic acid (H4Y) undergoes ionization at four, different stages, namely :


In reality, the actual complexing species is the tetracetate ion i.e., Y4– ; therefore, complexation will take effect more efficiently and be more stable in an aikaline medium. Hence, it is evident that EDTA com-plexes of many divalent metals are quite stable in ammoniacal solution.

 

As we have seen earlier that the trivalent metal complexes are normally bound still more firmly due to the formation of four rings (unlike three rings with divalent metal complexes) and stable in strongly acidic solutions, for instance : cobalt (Co2+) EDTA complex is fairly stable in concentrated hydrochloric acid ( ~ 11.5 N).

 

Though a good number of metal-EDTA complexes are found to be quite stable over a wide-spectrum of pH, yet in actual practice solutions are normally buffered for two specific reasons :

 

(a) to stabilize the complex formed, and

 

(b) to achieve the most distinct colour-change of the indicator.

 

2. STABILITY OF COMPLEXES

 

Generally, the formation of a 1 : 1 chelate complex (MX) may be designated by the following equation :


where, M = Metal ion, and

 

X = Chelating ion.

 

Hence, the stability constant, K, may be expressed as :


where, items within the ‘square brackets’ represent activities.

There are two cardinal factors which influence the stability constant (K), namely :

 

(a) Elevation in temperature affords a slight enhancement in the ionization of the complex and a slight lowering of K, and

 

(b) Stability constant is decreased on the addition of electrolytes with no common ion ; whereas, ethyl alcohol enhances K, perhaps on account of the suppression of ionization.

 

Table 9.1, provides the values of the logarithms of stability constants (K) of EDTA-complexes of certain metals normally occurring in pharmaceutical substances :


 

3. COLOURATION OF COMPLEXES

 

The formation of EDTA-metal ion complexes invariably attribute a change in the absorption spectrum pattern which ultimately forms the basis of a large number of colorimetric assays.

 

4. TITRABILITY OF POLYVALENT METAL IONS EMPLOYING DISODIUM EDETATE

 

Ethylenediamine tetracetic acid is found to be sparingly soluble in water ( ~ 0.2% w/v) whereas its corresponding disodium salt is almost 50 times more soluble than the parent compound (solubility ~ 10% w/v). Therefore, it is the disodium salt of EDTA which is normally used in complexometric titrations.

 

In actual practice, whenever the disodium EDTA solution is added to a solution of a metal ion previously buffered to augment complexation, it has been observed that initially the rate of change of concentration of metal ion is rather slow, but interestingly it picks up quite rapidly as further addition of sodium-EDTA approaches one equivalent.

 

5. USAGE OF pM INDICATORS IN COMPLEXOMETRIC TITRATIONS

 

The equivalence point in complexometric titrations is invariably observed by the help of pM indicators. The relationship amongst pM, concentrations of ligand, chelate complex and stability constant may be established by the following equations :

 

Assuming K as the stability constant, we have :


Now, considering Eq. (a), if a solution is made in such a manner that [X] = [MX], we have :

 

pM = – pK

 

or pM = pK ... (b)

where, K is the dissociation constant.

 

From Eq. (b) it may be concluded that a solution having equal activities of free chelating agent and the metal-complex formed, the concentration of metal ions shall remain almost constant and would be buffered exactly in a similar fashion as are H+ ions in a pH-buffer. As we know that the various chelating agents are mostly basic in character, therefore, the equilibrium attained in a metal-buffer solution is largely influenced by a change in pH. Hence, it may be concluded that the amino acid type chelating agents, such as : ethylenediamine tetracetic acid and ammoniatriacetic acid, when [X] = [MX], pM increases proportionately with pH until it reaches a value pH 10, thereby attaining a constant value. Hence, this particular pH is the ‘Ideal pH’ at which complexometric titrations of metals with chelating agents in buffered solution must be performed.

 

pM Indicator : It is a dye that serves as a chelating agent to yield a dye-metal complex, which apparently differs in colour from the original dye, besides possessing a lower stability constant than the corresponding chelate-metal complex. Hence, the colour imparted to the solution is mostly attributed due to the dye-complex formed until the end-point, when an equivalent amount of sodium-EDTA has been incorporated. The critical point at which the metal-dye complex decomposes to yield free-dye on addition of the slightest excess of sodium-EDTA, is distinctly shown by a visible change in colour.

 

Examples :

 

(i)          Mordant Black 2 : (Syn. : Eriochrome Black T ; Solochrome Black T)


(ii)       Murexide : (Syn. : Ammonium Purpurate)



Chapter: Pharmaceutical Drug Analysis: Complexometric Analysis

Complexometric Analysis: Assay Methods

The complexometric titrations involving various inorganic pharmaceutical substances may be catego-rized into three broad heads, namely : (i) Direct titration methods, (ii) Masking and demasking agents, and (iii) Residual titration methods.

ASSAY METHODS

The complexometric titrations involving various inorganic pharmaceutical substances may be catego-rized into three broad heads, namely :

(i) Direct titration methods,

 

(ii) Masking and demasking agents, and

 

(iii) Residual titration methods.

 

1. DIRECT TITRATION METHODS

 

In direct titration, usually an appropriate buffer solution and a suitable indicator are added to the M2+ (metal-ion) solution and subsequently the resulting solution is titrated with previously standardized disodium-EDTA until the indicator just changes colour. Sometimes, a simultaneous blank determination is also recommended to have a check for the presence of traces of metallic impurities in the reagents.

 

1.1. Preparation of 0.05 M Disodium Ethylenediamine Tetracetate Solution (Disodium Edetate 0.05 M)

 

Materials Required : Disodium ethylenediaminetetracetate : 18.6 g.

 

Procedure : Weigh accurately 18.6 g of disodium ethylenediaminetetracetae, dissolve in sufficient DW in a 1 litre volumetric flask and make up the volume upto the mark.

 

Calculations :


 

1.2. Standardization of 0.05 M Disodium Edetate Solution

 

Materials Required : Granulated zinc : 0.8 g ; dilute HCl (10% w/v of HCl) : 12.0 ml ; bromine water (3 ml Br2 in 100 ml H2O) : 5 ml ; sodium hydroxide (2 N) : 20.0 ml ; ammonia buffer (pH 10.0) (dissolve 5.4 g of NH4Cl in 70 ml of 5 N ammonia and dilute with water to 100 ml) : 100 ml ; Mordant Black II mixture (mixture of 0.2 part Mordant Black II with 100 parts of NaCl) : 50 mg ; disodium edetate : 0.05 M.

 

Procedure : Weigh accurately about 0.8 g of granulated zinc, dissolve by gentle warming in 12 ml of dilute hydrochloric acid and 5 drops of bromine water. Boil to remove excess bromine, cool and add sufficient DW to produce 200 ml in a volumetric flask. Pipette 20 ml of the resulting solution into a flask and neutralize carefully with 2 N sodium hydroxide. Dilute to about 150 ml with DW, add to it sufficient ammonia buffer (pH 10.0) to dissolve the precipitate and add a further 5 ml quantity in excess. Finally add 50 mg of Mordant Black II mixture and titrate with the disodium edetate solution until the solution turns green. Each 0.003269 g of granulated zinc is equivalent to 1 ml of 0.05 M disodium ethylenediaminetetracetate.

Calculations :


 

1.3. Calcium Chloride

 

Materials Required : Calcium chloride dihydrate : 0.15 g ; dilute hydrochloric acid (10% w/w of HCl) : 3.0 ml ; 0.05 M disodium edetate ; sodium hydroxide solution (20% w/v in water) ; calcon mixture (a mixture of 1 part of calcon with 99 parts of freshly ignited anhydrous Na2SO4) : 0.1 g.

Equations :


Procedure : Weigh accurately about 0.15 g of calcium chloride dihydrate and dissolve it in 50 ml of DW. Titrate with 0.05 M disodium ethylenediamine tetracetate to within a few ml of the expected end point, add 8.0 ml of sodium hydroxide solution and 0.1 g of calcon mixture and continue the titration until the colour of the solution changes from pink to a full blue colour. Each ml of 0.05 M disodium ethylene disodium tetracetate is equivalent to 0.007351 g of CaCl2 . 2H2O.

 

1.4. Magnesium Sulphate

 

Materials Required : Magnesium sulphate heptahydrate : 0.3 g ; strong ammonia-ammonium chloride solution (6.75 g NH4Cl in 74.0 ml strong ammonia solution add water q.s. to produce to 100 ml) ; 0.05 M disodium edetate ; Mordant Black II mixture (mixture of 0.2 part mordant black II with 100 parts of NaCl) : 0.1 g.

 

Equations :

 

The assay of MgSO4.7H2O is based upon the reactions designated by the following equations :


Procedure : Weigh accurately about 0.3 g of magnesium sulphate heptahydrate and dissolve in 50 ml of DW. Add to it 10 ml of strong ammonia-ammonium chloride solution, and titrate with 0.05 M disodium ethylenediaminetetracetate employing 0.1 g of mordant black II mixture as indicator, until the pink colour is discharged from the blue. Each ml of 0.05 M disodium ethylenediaminetetracetate is equivalent to 0.00602 g of MgSO4.

 

1.5. Cognate Assays

 

A number of pharmaceutical inorganic substances may be assayed by the direct titration method using disodium ethylenediaminetetracetate. A few typical examples are cited in the following Table 9.2.



 

2. MASKING AND DEMASKING AGENTS

 

The disodium ethylenediaminetetracetate usually complexes with a wide spectrum of cations, which ultimately renders the selectivity of the titration procedure adversely, thereby providing enough scope for the accompanying metal impurities to be titrated along with the ion it is aimed at for actual estimation. Therefore, in a situation where one or two ions present in a mixture of cations is specifically required to be determined with a view to eliminate completely the possible effects of unwanted impurities that may enhance the titre value, a third substance is added, which is known as the Masking Agent. These agents must fulfil the follow-ing three requirements, namely :

 

(a) should act by precipitation,

 

(b) should form complexes that are definitely more stable than the interfering ion-edetate complex, and

 

(c) colour developed by either precipitates or auxiliary complexes should not obscure the end-point.

 

A few typical examples are cited below in Table 9.3 where masking has been accomplished by precipitation.



 

3. RESIDUAL TITRATION METHODS

 

Direct titration method offers a serious limitation for the assay of aluminium and bismuth containing pharmaceutical inorganic substances because of the precipitation of the metal as their corresponding hydroxides in alkaline media thereby introducing undesirable errors.

 

In actual practice, an excess of the standard solution of disodium edetate is added to the sample, pH is adequately adjusted for the residual titration with a metal-ion solution e.g., ZnSO4 and employing an appropriate indicator which is sensitive enough to the respective titrant. However, the metal ion under estimation remains firmly complexed with the EDTA and offers little interference with the Zn-EDTA complex formed. It has been established experimentally that bismuth readily yields a highly stable complex which may be titrated conveniently between pH 1 and 2. Bismuth forms a stable complex by reacting with EDTA quantitatively at pH 4.0 and, therefore, dithizone is employed as an indicator to detect the end-point for it has a transition state of colour at pH 4.6.

 

 

3.1. Potassium Alum, KAl(SO4)2, 12H2O

 

Materials Required : Potassium alum : 1.7 g ; 0.05 M disodium edetate : 30.0 ml ; hexamine : 1.0 g ; 0.05 M lead nitrate ; xylenol orange solution (0.1% w/v in water) : 0.4 ml.

 

Theory : The solution of potassium alum is heated with an excess of disodium edetate to ensure complete formation of aluminium-edetate complex. Hexamine serves as a buffer thereby stabilizing the pH between 5 and 6, the ideal pH for the titration of the disodium edetate not required by the Al with 0.05 M lead nitrate employing xylenol orange as indicator. The various reactions involved may be represented by the following equations :


Procedure : Weigh accurately 1.7 g of potassium alum and dissolve it in suffcient DW in a flask. Heat the contents of flask over a water-bath for 10 minutes to allow completion of complexation and cool to ambient temperature. Now, add 1 g hexamine to act as buffer and titrate with 0.05 M lead nitrate employing 0.4 ml of xylenol orange solution as an indicator. The colour shall change from that of the indicator (yellow at the pH of the titration) to the corresponding reddish purple, the colour of the lead complex of the indicator. Each ml of 0.05 M disodium edetate is equivalent to 0.02372 g of KAl(SO4)2, 12H2O.

 

3.2. Glycobiarsol [Bismethyl-N-glycolyl-arsanilate]

 

Materials Required : Glycobiarsol : 0.2 g ; 0.05 M disodium edetate : 10.0 ml ; acetic acid-ammo-nium acetate buffer (mix 13.6 g of sodium acetate and 7.7 g of ammonium acetate in water to make 100 ml. Add 25.0 ml of glacial acetic acid and mix) : 10.0 ml ; alcohol : 25.0 ml : dithizone solution (0.05% w/v in chloroform) : 2.0 ml ; 0.025 M ZnSO4 solution.

 

Procedure : Weigh accurately 0.20 g of glycobiarsol into a 250-ml conical flask and add 10.0 ml of 0.05 M disodium edetate. Warm the contents of the flask over a water-bath until glycobiarsol gets dissolved completely and then cool the contents to the room temperature (25°C). Add to it 10.0 ml of acetic acid-ammonium acetate buffer, 25.00 ml of alcohol and 2 ml of dithizone solution as an indicator. Titrate the excess of disodium edetate with 0.025 M zinc sulphate until the resulting solution turns rose pink in colour. Each millilitre of 0.05 M disodium edetate consumed is equivalent to 10.45 mg of Bi.

Note : The content of Bi, calculated on dried basis, lies between 38 to 42.5%.

 

3.3. Cognate Assays

 

A number of inorganic pharmaceutical substances may be assayed by adopting the residual titration method as depicted in Table 9.4.


Chapter: Pharmaceutical Drug Analysis: Gravimetric Analysis

Gravimetric Analysis

Gravimetric analysis is an unique technique by means of which either an element or a compound is obtained in its purest form through isolation and subsequent weighing.

GRAVIMETRIC ANALYSIS

 

INTRODUCTION

Gravimetric analysis is an unique technique by means of which either an element or a compound is obtained in its purest form through isolation and subsequent weighing. In order to achieve this, the element or compound is first and foremost separated from a specific portion of the pharmaceutical substance being determined and consequently the weight of the constituent in the given sample is calculated on the basis of the weight of the product.

 

However, in actual gravimetric analysis, the final weight of the product is usually accomplished by adopting anyone of the following standard methods, namely :

 

(a) Solvent extraction,

 

(b) Ignition or volatalization, and

 

(c) Precipitation from solution.

 

Gravimetric techniques are broadly based upon the quantitative precipitation of the respective cation or anion from a given solution in two different ways :

 

(i) as an insoluble compound that yields a residue having a specific composition after ignition, and

 

(ii) as an insoluble compound having a known composition.

 

There are four vital steps that are essentially required for a successful gravimetric method, namely :

 

(a) Identify an insoluble form with a definite composition,

 

(b) Separate the analyte exclusively from another constituents which may cause interference,

 

(c) Wash the precipitate free of coprecipitants and impurities as far as possible, and

 

(d) Convert the precipitate ultimately to a reasonably measurable form.


Chapter: Pharmaceutical Drug Analysis: Gravimetric Analysis

Gravimetric Analysis: Theory

The underlying principles and theories of gravimetric analysis are as stated below : (i) Law of mass action and reversible reactions, (ii) Principle of solubility product, and (iii) Common ion effect.

THEORY

The underlying principles and theories of gravimetric analysis are as stated below :

 

(i)          Law of mass action and reversible reactions,

 

(ii) Principle of solubility product, and

 

(iii) Common ion effect.

 

All the above three aspects shall be described briefly vis-a-vis their direct impact on the gravimetric analysis.

 

 

1. LAW OF MASS ACTION AND REVERSIBLE REACTIONS

 

A plethora of chemical reactions that are intimately associated with the quantitative analysis essen-tially belong to the class of reversible reactions. These reactions under certain prevailing experimental param-eters are made to proceed to completion, whereas in certain other conditions they may even attain equilibrium before completion. In the latter instance, erroneous results may creep in with regard to the pharmaceutical substance under estimation. Hence, it has become absolutely necessary first to establish the appropriate con-ditions whereby the reactions must move forward to attain completion so as to achieve the ultimate objective in all quantitative assays.

 

In general, there are three cardinal experimental parameters that must be observed rigidly in order to check the reversal processes and help the completion of a reaction, namely :

 

(a) formation of very slightly ionized molecules,

 

(b) formation of an insoluble gas, and

 

(c) formation of a sparingly soluble solid.

 

The ‘law of mass action’ advocates that the rate of a reaction is directly proportional to the product of the molecular concentrations of the reacting substances. For example :


In the above reaction the rate of reaction of barium chloride with sulphuric acid is designated by the following expression :

 

Forward reaction :

            .......................(a)

where, k = a constant that corrects for all factors which affect the rate other than concentration.

 

Likewise, in the opposing reaction, we have :

 

Opposing reaction :

            ...........................(b)

where, k1 = another constant.

 

At equilibrium the rates of the forward reaction (a) and opposing reaction (b) are equal. Hence, we have :


As k and kl are constants, their quotient K is also a constant known as the equilibrium constant. From Eq. (d), K, the equilibrium constant has a fixed value at a definite temperature, irrespective of concentrations of other components present.

 

Therefore, if the concentration of sulphuric acid is enhanced, consequently all other concentrations should change accordingly, the concentration of BaCl2 must become less and that of both BaSO4 and HCl be proportionately greater so as to maintain the equilibrium constant, thereby having the net impact of shifting the equilibrium towards the right hand side. Evidently, in most quantitative analysis one entity is added invariably to allow the reaction to proceed as closely to completion as possible.

2. PRINCIPLE OF SOLUBILITY PRODUCT

 

The principle of solubility product may be stated as follows :

 

The product of the concentration of the constituent ions in a saturated solution of a difficultly soluble salt for any given temperature is practically a constant, each concentration being raised to a power equal to the relative number of ions supplied by one molecule of the salt upon dissociating’.

 

The principle of solubility product is applicable to :

 

(i) difficultly soluble salts in their saturated solutions,

 

(ii) occurrence of precipitation,

 

(iii) prevention of precipitation, and

 

(iv) dissolution of a substance.

 

For instance, a difficultly soluble salt ApBq on dissociation provides a relative number of p cations and q anions. Thus, we have :


Hence, solubility product ApBq = [A+]p × [B ]q

 

where, [ ] are generally used to express the molar concentrations.

 

Table 10.1, contains the solubility products of certain difficultly soluble salts generally encountered in pharmaceutical analysis.



The interaction of AgNO3 and NaCl results into the formation of AgCl which is slightly soluble in water, the solubility being approximately 0.00001 ml litre–1 i.e., 1.5 mg litre–1. On exceeding this concentra-tion, the AgCl gets precipitated which remains in equilibrium with the dissolved AgCl. Therefore, at equilib-rium, the clear supernatant liquid is a saturated solution, and at this critical juncture the rate at which the dissolved salt gets precipitated is almost equal to the rate at which the solid undergoes dissolution. This establishes the following equilibria :


Hence, the ionization equilibrium may be expressed as follows :


Considering the following two assumptions :

 

(i) solution remains saturated with AgCl at a aiven temperature, and

 

(ii) concentration of unionized AgCl remains constant, it follows, that the product K × [AgCl] too becomes constant.

 

Therefore, it may be inferred that—‘in a saturated solution of a difficultly soluble salt, the product of the molecular concentration of its ions is constant’.


It is an usual practice to express the concentration of the solubility product in terms of moles per litre i.e., molar concentrations.

 

3. COMNION ION EFFECT

 

It has been observed that there is no change in the equilibrium constant even if :

 

(a) the concentrations of reacting components may change, and

 

(b) the relative concentration of the reacting substances may change.

 

When a solution of BaCl2 is added to a solution of sulphuric acid, the sulphate ion for a while is present in a concentration in such a manner that its ionic product with the barium ion exceeds the solubility product of barium sulphate, and the insoluble barium sulphate gets precipitated :


However, at equilibrium the concentration of Ba2+ ions shall be exactly equal to the concentration of sulphate ions.

 

Now, if to the resulting supernatant liquid, which is nothing but a saturated solution of barium sul-phate, an additional small quantity of either a soluble barium salt or a soluble sulphate is provided, a slight further precipitation may occur.

 

Hence, the equilibrium that represents the ionization constant may be expressed as :


From Eq. (a), it may be derived that if the concentration of Ba2+ ion is enhanced by the addition of a soluble barium salt, the concentration of sulphate ion should decrease simultaneously and conversely, that if the concentration of sulphate ion is enhanced by the addition of a soluble sulphate salt, the concentration of Ba2+ ion should decrease as their product almost remains constant. Evidently, this decrease in the concentration of the ions in either instance may be achieved by the combination of barium and sulphate ions to give rise to the insoluble barium sulphate thereby forcing the reaction towards completion.

 

In short, the common-ion effect is employed invariably in carrying out the gravimetric analysis of pharmaceutical substances so as to drive reactions toward completion.

 

Calculations : In gravimetric analysis the percentage of the desired constituent may be achieved by the following expression :


The term ‘gravimetric factor’ is generally employed which represents the number of grams of the desired constituent in 1 g of the substance weighed. It can be further expatiated with the help of the following examples :

 

(i)          One mole of BaSO4 (233.39 g) contains one mole of SO4 atoms (96.06 g).


(ii)       One mole of AgCl (143.323 g) contains one mole of Cl atoms (35.453 g).



Chapter: Pharmaceutical Drug Analysis: Gravimetric Analysis

Gravimetric Analysis: Assay Methods

A good number of pharmaceutical substances can be assayed gravimetrically.

ASSAY METHODS

A good number of pharmaceutical substances can be assayed gravimetrically. The gravimetric meth-ods adopted vary according to the nature of the substance under determination. However, most of the sub-stances being estimated gravimetrically fall into one or the other categories stated below, which would be discussed briefly with suitable examples :

 

(a) Substances assayed gravimetrically,

 

(b) Substances assayed after conversion :

 

(i) Substances assayed after conversion to Free Acid,

 

(ii) Substances assayed after conversion to Free Base,

 

(iii) Substances assayed after conversion to Free Compound, and

 

(iv) Substances assayed after conversion to Derivatives or Substitution Products.

 

Chapter: Pharmaceutical Drug Analysis: Gravimetric Analysis

Substances Assayed Gravimetrically

A good number of pharmaceutical substances may be determined gravimetrically by obtaining their respective difficultly soluble salts as precipitates, weighing to a constant weight and finding the percentage purity of the substance in question.

SUBSTANCES ASSAYED GRAVIMETRICALLY

 

A good number of pharmaceutical substances may be determined gravimetrically by obtaining their respective difficultly soluble salts as precipitates, weighing to a constant weight and finding the percentage purity of the substance in question.

 

A few typical examples are cited below so as to expatiate the procedure as well as the theoretical aspects.

 

 

1. Sodium Chloride

 

Materials Required : Sodium chloride : 0.25 g ; 5% w/v silver nitrate in DW (+ 2-3) drops of conc. HNO3 ; dilute nitric acid (6 N) ; asbestos fibre.

 

Theory : The following reaction forms the basis for the calculation of the theoretical amount of silver nitrate solution required as well as the purity of the given sample of NaCl. Thus, we have :


As 0.2570 g of NaCl has been used (from experimental data); therefore, the exact amount of AgNO3 required would be :


Hence, the amount of AgNO3 solution required theoretically would be 0.7470/0.05 = 14.94 ml.

From above, the percentage purity of the given sample of NaCl may be found as shown below :


The weight of AgCl is found to be 0.6288 g experimentally, or 0.4078 is the ‘gravimetric factor’.

 

Consequently, the percentage purity of the sample is determined by the formula :


where, W = Wt. of the product of a chemical reaction with the substance under determination,

 

E = Gravimetric Factor, and

 

S = Wt. of the sample.

 

By incorporating the data given above, the amount of sodium chloride present in 100 g of the sample i.e., the percentage purity of NaCl in the given sample may be calculated as follows :


Procedure : Weigh accurately between 0.20 to 0.30 g of sodium chloride and dissolve in 100 ml of DW. Add to it 1 ml of dilute nitric acid gradually with constant stirring. Check and confirm that the resulting solution is acidic with the help of blue litmus paper. Measure out 5.0 ml in excess of the amount of silver nitrate solution calculated on theoretical basis to precipitate all the available chlorine as silver chloride. The requisite quantity of silver nitrate solution must be added in small lots at intervals with constant stirring with a glass rod. Cover the beaker with a watch-glass and boil the contents very gently with occasional stirring (to avoid bumping of the liquid and loss of volume). Stop heating and digest the mixture for 10 minutes so as to agglomerate the precipitate and enhance settling thereby leaving a clear supernatant liquid. Add 2 drops of silver nitrate solution to the hot supernatant liquid in order to confirm whether precipitation is completed. Keep the beaker away from direct sunlight to allow the precipitate to settle.

 

Take a properly prepared Gooch crucible, heat to constant weight and fit it into the suction flask. Decant most of the supernatant liquid first into the Gooch crucible by applying gentle suction to hasten filtration. Wash the precipitate on the Gooch crucible at least thrice with 15 ml portions of 0.01 N nitric acid.

 

Test the above filtrate to be free of AgNO3. Finally wash the precipitate twice with 5 ml portion of DW to get rid of most of the HNO3 previously retained by the precipitate from the former wash solution. Now, apply vigorous suction to drain out the liquid from the precipitate to the maximum extent. Dry the crucible to a constant weight between 110-120°C in an electric oven until two concurrent weighings are achieved. Thus, the weight of the crucible (tare) must be deducted from the weight of the crucible plus the precipitate to arrive at the weight of silver chloride duly obtained from the sample.

Precautions :

 

1.           The solution of the substance is usually acidified with HNO3 to check the precipitation of other substances insoluble in water but soluble in HNO3 e.g., CO32–, O2– and PO43–. Besides HNO3 also helps to coagulate any colloidal AgCl,

2.           The excess of HNO3 must be avoided to cause solvolysis of silver halides,

3.           Heating should be affected only after the addition of AgNO3, otherwise Cl2 may be liberated and lost. Thus, we have :


4.           The precipitation should preferably be carried out in the absence of strong light because AgCl undergoes decomposition in sunlight with loss of Cl2,

5.           Washing of the precipitate (AgCl) with 0.01 N HNO3 is always recommended to prevent loss of AgCl by virtue of its return to colloidal condition (peptization) and to get rid of the soluble salts, namely : AgNO and NaNO3, and

6.           AgCl is significantly volatile on ignition, hence it must always be dried at a comparatively lower temperature.

 

2. Potassium Alum, KAl(SO4)2, 12H2O

 

Theory : The percentage of Al in potassium alum can be determined volumetrically by complexometric titration.

 

However, gravimetric procedure provides a fairly reliable and useful alternative method of analysis for

 

Al which may be accomplished by :

 

(a) precipitation from a solution of the aluminium salt by the addition of NH4OH in the presence of NH4Cl, and

 

(b) complexation from a solution of the aluminium salt with 8-hydroxyquinoline (oxine) either from an ammoniacal solution or from acetic acid-acetate buffer.

 

In the first method, the following reaction takes place :

 

Al3+  +  3OH  → Al(OH)3

 

The gelatinous white precipitate of Al(OH)3 is duly filtered, washed with dilute NH4NO3 solution, transformed to the corresponding oxide and finally weighed as Al2O3 .

 

Disadvantages : There are a number of serious disadvantages of this method, namely :

 

(i) excess of NH4OH may directly affect the solubility of Al(OH)3,

(ii) coprecipitation of metal hydroxides that are usually soluble in NH4OH,

(iii) heated oxide (Al2O3) is hygroscopic in nature, and

(iv) hydroxides may not undergo complete thermal decomposition.

 

Due to the above short-comings, the second method is usually preferred which shall be discussed below :

 

Equation :


The resulting precipitate of aluminium-oxine complex is crystalline in nature and hence can be filtered conveniently, washed with water and finally dried at 130-150°C to constant weight.

 

Disadvantages : There are two disadvantages of the metal-oxine-complex method, namely :

 

(i) aluminium-oxinate is prone to adsorb oxine, and

 

(ii) lack of selectivity of oxine such that all metals except the alkaline earths (Ba, Mg, Ca, Sr, Be) and alkali (Li, Na, K, Rb, Cs) should be totally absent.

 

Calculations :


Materials Required : Potassium alum : 0.3 g ; 0.1 N hydrochloric acid : 1.0 ml ; 8-hydroxyquinoline reagent (or oxine-reagent) (25 ml of a 2% w/v solution of oxine in 2 N acetic acid) ; 2 N ammonium acetate (dissolve 15.0 g of ammonium acetate in 20.0 ml of DW, add 0.3 ml of glacial acetic acid and dilute to 100 ml with DW) ; sintered glass crucible No : 3 or 4.

 

Procedure : Weigh accurately about 0.3 g of potassium alum in a 400-ml beaker, dissolve it in 150 ml of DW containing 1.0 ml of 0.1 N HCl and warm the contents of the beaker to about 60°C. Add the requisite quantity of the oxine reagent and then add a 2 N solution of ammonium acetate gradually from a pipette till precipitation just commences. Add a further portion (50 ml) of ammonium acetate solution with vigorous stirring. Allow the contents of the beaker to stand for 60 minutes with frequent stirring. Filter the precipitate through No : 3 or 4 sintered glass crucible that has been previously dried to a constant weight at 130—150°C. Wash the precipitate throughly with cold DW and dry at 130 to 150°C to constant weight. Each gram of aluminium oxinate is equivalent to 0.05873 g of Al.

 

3. Cognate Assays

 

A good deal of pharmaceutical substances are officially assayed gravimetrically as appears in Table 10.2.


Chapter: Pharmaceutical Drug Analysis: Gravimetric Analysis

Substances Assayed After Conversion - Gravimetric Analysis

1. Substances Assayed after Conversion to Free Acid 2. Substances Assayed after Conversion to Free Base 3. Substances Assayed After Conversion to Free Compound 4. Substances Assayed after Conversion to Derivatives or Substitution Products

SUBSTANCES ASSAYED AFTER CONVERSION

 

There are certain pharmaceutical substances that can be assayed gravimetrically after their suitable conversion to free acid, or free base, or free compound or corresponding derivatives (or substitution products). All these typical cases shall be discussed briefly with their appropriate examples in the following sections.

 

1. Substances Assayed after Conversion to Free Acid

 

A few official pharmaceutical substances may be assayed gravimetrically by affecting separation, purification, and weighing an organic medicinal compound without causing any permanent change in composition. It is an usual practice that before extraction of the organic medicinal compound, the sample of the crushed tablets is carefully washed with petroleum benzene to get rid of undesirable components, for instance : lubricants and binders that would be extracted along with the organic medicinal compound by such solvents as ether or chloroform which is employed subsequently.

 

In case, the organic medicinal compound is acidic in nature e.g., amobarbital in sodium amobarbital tablets, it is first and foremost extracted with an aqueous solution of an acid or base to cause separation from the neutral substance which might be present. The resulting aqueous solution of the salt of the respective organic medicinal compound is subsequently made acidic and the liberated organic acid (amobarbital) is finally extracted with ether or chloroform.

 

Interestingly, in a situation where either magnesium stearate or stearic acid forms a component in the formulation, the organic medicinal compound which is acidic (amobarbital) cannot be extracted with NaOH solution for obvious reason that sodium stearate shall also be extracted along with the salt of the organic acid. Therefore, instead a saturated solution of Ba(OH)2 is employed thereby the insoluble precipitate of barium stearate may be discarded by filtration.

 

1.1.  Phenobarbitone Sodium

 

Materials Required : Phenobarbitone sodium : 0.5 g ; hydrochloric acid (2 M) : (dissolve 17.0 ml (~  11.5 N) in 100 ml DW) : 5.0 ml ; ether : 13.5 ml ; absolute ethanol : 2.0 ml.

Procedure : Weigh accurately 0.5 g phenobarbitone sodium and dissolve in 15 ml of DW. Add to it 5 ml of 2 M hydrochloric acid and extract with 50 ml of ether and then with successive 25 ml quantities of ether until complete extraction is affected. Wash the combined extracts with two 5 ml quantities of DW and wash the combined aqueous extracts with 10 ml quantities of ether. Add the ether to the main ethereal extract, evaporate to low bulk, add 2 ml of absolute ethanol, evaporate to dryness and dry the residue to constant weight at 105°C. Each g of residue is equivalent to C12H11N2NaO3.

 

Calculations :


 

1.2.  Cognate Assays

 

There are certain pharmaceutical substances that may be assayed after their conversion to the respec-tive free acids as shown in Table 10.3.


 

2. Substances Assayed after Conversion to Free Base

 

In a specific instance where the organic medicinal substance is basic in nature e.g., papaverine in papaverine hydrochloride, it is primarily treated with an aqueous solution of a base and subsequently the liberated organic base is extracted with either chloroform or ether.

 

A typical example is described below :

 

2.1. Papaverine Hydrochloride Tablets

Materials Required : Sodium hydroxide (2 M) (dissolve 8.0 g of NaOH pellets in 100 ml of CO2 free DW : 50 ml ; chloroform : 100 ml ; absolute ethanol : 5 ml.

Calculations :


Procedure : Weigh 20 tablets and crush them in a pestle mortar and find out the average weight of a single tablet. Accurately weigh 0.5 g equivalent of papaverine hydrochloride and dissolve in 15 ml of DW. Add to it 15 ml of 2 M sodium hydroxide and extract with 50 ml of chloroform and then with successive 25 ml quantities of chloroform until complete extraction is affected. Wash the combined extracts with two 5 ml quantities of DW and wash the combined aqueous extract with two 10 ml quantities of chloroform. Add the chloroform to the main chloroform extract, evaporate to a small volume, add 2 ml of absolute ethanol, evaporate to dryness and dry the residue to constant weight at 105°C.

 

Each g of the residue is equivalent to 1.105 g of C20H21NO4 . HCl.

 

2.2.  Amodiaquine Hydrochloride

 

Materials Required : Amodiaquine hydrochloride : 0.3 g ; dilute ammonia solution (42.5 ml of strong ammonia solution to 100 ml in water) ; NO. 4 sintered glass crucible.

 

Theory : Amodiaquine hydrochloride possesses two moles of inherent water of crystallization, and hence the precentage base is calculated with reference to the substance dried over P2O5 at a pressure not exceeding 5 mm of Hg. Usually, the assay is performed on one portion of the sample and the drying on a separate portion altogether.

 

The underlying principle of the method is based upon the precipitation of amodiaquine base that is generated as a precipitate when the salt is decomposed in aqueous medium with dilute ammonia.


Procedure : Weigh accurately 0.3 g of previously dried amodiaquine hydrochloride into a 100 ml beaker provided with a stirring rod and watch glass cover. Dissolve it in 50 ml of DW and dilute ammonia solution with constant gentle stirring until the solution is just alkaline (to litmus paper). Allow the contents of the flask to stand for 30 minutes and then quantitatively filter through a NO. 4 sintered glass-crucible previously dried to a constant weight at 105°C. Wash the precipitate several times with DW, until the washings do not give a positive test for chloride (test with standard AgNO3 Solution). Dry the residue to a constant weight at 105°C. Each gram of residue is equivalent to 1.306 g of C20H22ON3Cl, 2HCl, 2H2O.

 

2.2.  Cognate Assays

 

A few other pharmaceutical substances are also determined after conversion to free bases as recorded in Table : 10.4.


 

3. Substances Assayed After Conversion to Free Compound

 

In certain specific cases either the pure pharmaceutical substance or dosage forms are quantitatively converted to free compound. This conversion to free compound is quantitative and hence forms the basis of gravimetric analysis. A few typical examples belonging to this category are, namely : progesterone suspension sterile, progesterone tablets, sodium lauryl sulphate, mephobarbital tablets and sorbitan monooleate.

 

 

3.1. Mephobarbital Tablets

 

Materials Required : Mephobarbital : 300 mg ; hexane : 100 ml ; chloroform : 150 ml ; alcohol (95% v/v) : l0 ml.

 

Procedure : Weigh and finely powder not less than 20 mephobarbital tablets. Transfer an accurately weighed portion of the powder equivalent to about 300 mg of mephobarbital to an extraction thimble. Extract with 15 ml of solvent hexane, allow the thimble to drain, transfer to a continuous extraction apparatus pro-vided with a tared flask, and extract the mephobarbital with chloroform for 2 hours. Evaporate the chloroform on a steam bath with the aid of a current of air, cool, dissolve the residue in about 10 ml of alcohol, evaporate, dry the residue at 105°C for 1 hour, cool and weigh.

 

The weight of the residue represents the weight Cl3H14N2O3 in the portion of the tablets taken.

 

4. Substances Assayed after Conversion to Derivatives or Substitution Products

 

In pharmaceutical drug analysis a host of organic pharmaceutical substances are invariably converted quantitatively to their corresponding derivatives by virtue of interactions with certain functional entities, namely : aldehyde, ketone, amino, carboxyl, phenolic, hydroxyl etc. However, in some cases it may be feasible to obtain uniform substitution products of organic pharmaceutical substances quantitatively, for instance : tetraido derivative of phenolphthalein is obtained from the phenolphthalein tablets. It is important to mention here that the number of organic pharmaceutical substances which may be analysed by this method is limited because of two vital reasons, they are :

 

(a) the reversible nature of reactions, and

 

(b) the formation of products of side reactions simultaneously.

 

 

4.1. Benzylpenicillin(Syn : Benzylpenicillin Sodium or Potassium Salt)

 

Materials Required : Benzylpenicillin sodium (say) : 0.12 g ; amyl acetate (previously saturated with 1-ethylpiperidinium benzylpencillin at room temperature, cooled in ice and filtered) : 5.0 ml ; phosphoric acid (20% v/v) : 0.5 ml ; anhydrous sodium sulphate (freshly ignited and powdered) : 0.5 g ; dry acetone (previously saturated with 1-ethylpiperidinium benzylpenicillin at room temperature cooled in ice and filtered) : 3.0 ml ; 1-ethylpiperidine amyl acetate solution (prepared from l-ethyl piperidine, 1 .0 ml, and amyl acetate, 8.0 ml, saturated at room temperature with 1-ethylpiperidinium benzylpenicillin, cooled in ice and filtered) : 1.5 ml ; dry acetone in amyl acetate (1 : 1) previously saturated with 1-ethylpiperidinium benzylpenicillin : 2.0 ml ; solvent ether : 4.0 ml.

 

Theory : Benzylpenicillin (sodium or potassium salt) may be assayed gravimetrically by quantitative conversion to the 1-ethylpiperidinium benzylpenicillin derivative. The ultimate precipitation is caused by l-ethyl piperidine after the respective sodium or potassium salt of benzylpencillin has been duly converted with phosphoric acid to the corresponding penicillanic acid (i.e. parent acid) and the latter finally extracted with amyl alcohol. The reactions may be expressed as follows :


Procedure : Weigh accurately 0.12 g of benzyl penicillin sodium, dissolve in 5 ml of ice-cold DW in a flask and cool in an ice-bath. Add to it 5.0 ml of amyl acetate followed by 0.5 ml of ice-cold H3PO4, stopper, shake the contents immediately for 15 seconds, and centrifuge for 30 seconds. Remove the aqueous layer as completely as possible with the help of a pipette. Add 0.5 g anhydrous Na2SO4, stir the contents vigorously and cool in an ice-bath for 5 minutes. Centrifuge for about 30 seconds and again cool in ice-bath for 5 minutes. Pipette 3.0 ml of the supernatant liquid into a tared centrifuge tube. Add to it 3.0 ml of ice-cold acetone and 1.5 ml of 1-ethylpiperidine amyl acetate solution, stir, stopper the tube and cool in ice-bath for 2 hours. Now, centrifuge for 1 minute, break the surface with the help of a pointed glass rod, so that all crystalline particles are covered by liquid, and again centrifuge for 1 minute. Decant off the supernatant liquid, wash the precipitate with 2 ml of ice-cold dry acetone in amyl acetate (1 : 1) and again centrifuge for 1.5 minutes. Decant the supernatant liquid, wash twice with 2.0 ml portion of solvent ether, centrifuging for 1.5 minutes and decanting each time. Dry to constant weight under vacuum at room temperature. Each gram of residue is equivalent to 0.8300 g of C16H17N2NaO4S.

 

4.2.  Cholesterol

 

Materials Required : Cholesterol : 0.1 g ; ethanol (90% v/v) : 12.0 ml ; digitonin solution (0.5% w/v in 90% v/v ethanol) : 40.0 ml ; ethanol (90% v/v) : 100 ml ; acetone ; carbon tetrachloride.

 

Theory : The assay of cholesterol is solely based on the fact that practically all 3 β-hydroxysterols e.g., cholesterol, readily produces an insoluble molecular addition complex with pure digitonin (1 : 1)—a steroidal saponin isolated from either Digitalis purpurea or Digitalis lanata. The complex thus obtained is crystalline in nature, fairly stable and possesses very low solubilities.


The complexation of cholesterol and digitonin may be expressed as follows :


Procedure : Weigh accurately about 0.1 g of cholesterol into a 100 ml flask and dissolve it in 12.0 ml ethanol. Insert the stopper and allow to stand at room temperature (25 ± 2°C) for 12 hours, filter through a Gooch crucible, and wash with 5.0 ml of ethanol. Mix the washings to the filtrate and add to it 40.0 ml solution of digitonin and make it warm to 60°C to ensure that the complexation is almost complete. Filter the precipitate of the resulting complex through a prepared Gooch crucible, previously dried to constant weight at 105°C. Wash the precipitate with ethanol followed by acetone and carbon tetrachloride, allow to drain as completely as possible, and dry to a constant weight at 105°C. Each g of the residue is equivalent to 0.2390 g of cholesterol.

 

Note : All solutions must be ice-cold.

 

4.3. Thiamine Hydrochloride

 

Materials Required : Thiamine hydrochloride : 0.5 g ; hydrochloric acid ( ~  11.5 N) : 2.0 ml ; silicotungstic acid solution (10% w/v in water) : 4.0 ml ; NO : 4-sintered glass-crucible ; dilute hydrochloric acid (1 part HCl + 19 parts H2O) : 50 ml.

 

Theory : The gravimetric assay of thiamine hydrochloride is based upon the precipitation of it as thiamine silicotungstate with silicotungstic acid in a slightly acidic medium. It has been observed that the precipitating reagent is a complex silicate SiO2, 12 WO2, n H2O having somewhat variable composition with regard to the degree of hydration. For a reasonably precise and accurate determination the precipitating reagent must contain <| 1.85% SiO2 and <| 85% WO3. Interestingly, the thiamine silicotungstate complex possesses more or less a constant composition.

The precipitation of insoluble thiamine silicotungstate may be designated by the following reaction :


Procedure : Weigh accurately 0.05 g of thiamine hydrochloride, previously dried at 105°C, and dissolve it in 50 ml DW in a 250 ml beaker having a stirring rod and watch glass cover. Add to it 2.0 ml of hydrochloric acid, heat to boiling and then add 4.0 ml of silicotungstic acid solution as rapidly as possible. Now, boil the solution gently for 2 minutes and quickly filter through a NO. 4 sintered-glass crucible, previously dried to a constant weight at 105°C. Wash the residue with a boiling mixture of HCl and H2O (1 : 19) about 40 ml, then with DW 10.0 ml and ultimately with two portions of 5 ml each of acetone. Finally dry the residue to constant weight at 105°C. Each g of thiamine silicotungstate residue is equivalent to 0.1938 g of C12Hl7ON4SCl, HCl.

 

Precautions :

 

(a) An excess of HCl is a must so as to produce a readily filterable precipitate,

 

(b) In case the sample is pure enough, the rate of addition of silicotungstic acid has little influence on the result, but on the contrary if the sample has significant impurity it may afford poor results,

 

(c) To achieve complete complexation boiling must be done for more than 2 minutes, otherwise it would yield low results, and

 

(d) A 50-ml wash-liquid is quite ideal, further washings (volume) may offer poor results.

 

4.4. Histamine Acid Phosphate (C5H9N3, 2H3PO4)

 

Materials Required : Histamine : 0.15 g ; nitranilic acid solution (3.5% w/v in 95% ethanol) : 10.0 ml ; ethanol (95%) : 30.0 ml ; sintered-glass crucible (NO : 3) ; ether : 10.0 ml.

 

Theory : The gravimetric assay of histamine acid phosphate is based upon the formation of insoluble histamine-nitranilic acid complex as depicted in the following equation :


Procedure : Weigh accurately about 0.15 g of histamine acid phosphate into a 250 ml beaker provided with a stirring rod and watch glass cover. Add to it 10.0 ml of DW to dissolve the sample. Now, add 10.0 ml of nitranilic acid solution, stir and allow to stand for 15 ininutes. Pour in 10.0 ml of ethanol, keep it in an ice-bath for 3 hours and filter through a No. 3 sintered-glass crucible, previously dried to a constant weight at 130°C. Transfer the precipitate quantitatively and wash it thoroughly with four quantities each of 5.0 ml of ethanol and ultimately with 10.0 ml of ether. Dry to constant weight at 130°C. Simultaneously, determine the loss in weight on drying a separate portion of the sample at 105°C. Each gram of the histamine-nitranilic acid complex is equivalent to 0.8998 g of C5H9N3, 2 H3PO4.

 

4.5.  Proguanil Hydrochloride

 

Materials Required : Proguanil hydrochloride : 0.6 g ; ammoniacal cupric chloride solution (dissolve 22.5 g of copper (II) chloride in 200 ml of DW and mix with 100 ml of 13.5 M ammonia) ; NO. 4 sintered-glass crucible ; mixture of dilute solution of ammonia and DW (1 : 5).

 

Theory : Gravimetric analysis of proguanil hydrochloride involves the precipitation of the proguanil-cupric complex that results on the addition of ammoniacal cupric chloride solution to a solution of proguanil hydrochloride. The reaction can be expressed by the following equation :


Procedure : Weigh accurately 0.6 g of proguanil hydrochloride into a 250 ml beaker fitted with a stirring rod and watch-glass cover. Add to it 50.0 ml of DW and heat gently to dissolve the sample. Chill the solution below 10°C in an ice-bath and then add ammoniacal-cupric-chloride solution with continuous stirring till the resulting solution attains a permanent deep-colour. Allow the solution to stand for 90 minutes to complete the complexation and then filter through a No. 4 sintered glass crucible previously dried to constant weight at 130°C. Transfer the precipitate quantitatively into the crucible, wash first with a mixture of dilute solution of ammonia and DW (1 : 5) adequately followed by cold water until the washings are practically colourless thereby showing the complete absence of soluble copper salts. Dry the precipitate to a constant weight at 130°C. Simultaneously, find out the loss in weight on drying with a separate portion of the sample at 105°C and incorporate this in the calculation. Each gram of proguanil-cupric-complex is equivalent to 1.0199 g of C11Hl6N5Cl, HCl.

 

4.6. Benzethonium Chloride

 

Theory : In general, quaternary nitrogen containing compounds like—choline chloride, acetylpyridinium chloride, benzethonium chloride, and bethanechol chloride readily form insoluble salts quantitatively with tetraphenyl boron and this puts forward the basis for the gravimetric assay of the above cited pharmaceutical substances.

 

The various reactions involved may be summarized and expressed as follows :


Eq. (a) shows that the quaternary salt gets quantitatively precipitated by sodium tetraphenyl boron as the complexing agent. Eq. (b) depicts that quaternary compounds shall readily react with certain anionic dye, such as bromophenol blue, to yield a blue, chloroform-soluble complex.

 

Eq. (c) finally illustrates that the blue-coloured complex shall react quantitatively with sodium tetraphenyl boron to give an insoluble compound.


Therefore, we have :


Materials Required : Benzethonium chloride : 0.15 g ; Chloroform : 50 ml ; bromophenol blue solution (Dissolve with heating 0.2 g of bromophenol blue in 3 ml of 0.1 M NaOH and 10 ml of ethanol (96%). Allow to cool and dilute to 100 ml with ethanol 96%] : 50 ml ; sodium tetraphenyl borate solution (1% w/v in chloroform) : 50 ml ; sintered-glass crucible No : 4.

 

Procedure : Weigh accurately about 0.15 g of benzethonium chloride sample into a 250-ml beaker placed on a magnetic-stirrer and watch-glass cover. Add to it 25 ml of chloroform and warm gently to dissolve. Cool to ambient temperature and add suffcient bromophenol blue solution gradually till the solution yields a blue Chloroform-soluble complex. Now, add sodium tetraphenyl borate solution in small lots at intervals with constant stirring until the complete precipitation of insoluble benzethonium tetraphenyl borate complex takes place. Allow the solution to stand for 60 minutes to complete the complexation and subsequently filter through a No. 4 sintered-glass crucible previously dried to constant weight at 130°C. Transfer the precipitate quantitatively into the crucible and wash the precipitate with cold chloroform. Dry the precipitate to a constant weight at 110°C. Each gram of benzethonium tetraphenyl borate complex is equivalent to 0.6117 g of C27H4lO2NCl.

 

4.7.  Cognate Assays

 

Quite a few official pharmaceutical substances and their respective dosage forms can be assayed gravimetrically after conversion to their corresponding derivatives or substitution products. Table 10.5 records some examples from official compendia.


Chapter: Pharmaceutical Drug Analysis: Thermoanalytical Analysis

Thermoanalytical Analysis

Thermoanalytical methods essentially encompass such techniques that are based entirely on the concept of heating a sample followed by well-defned modified procedures, such as : gravimetric analysis, differential analysis and titrimetric analysis.

THERMOANALYTICAL ANALYSIS

 

INTRODUCTION

Thermoanalytical methods essentially encompass such techniques that are based entirely on the con-cept of heating a sample followed by well-defned modified procedures, such as : gravimetric analysis, differ-ential analysis and titrimetric analysis. In usual practice, data are generated as a result of continuously re-corded curves that may be considered as ‘thermal spectra’. These thermal spectra also termed as ‘thermograms, often characterize a single or multicomponent system in terms of :

 

(a) temperature dependencies of its thermodynamic properties, and

 

(b) physicochemical reaction kinetics.

 

Broadly speaking the thermoanalytical methods are normally classified into the following three categories, namely :

 

(i) Thermogravimetric Analysis (TGA),

 

(ii) Differential Thermal Analysis (DTA), and

 

(iii) Thermometric Titrations.

 

All the above mentioned techniques shall be discussed briefly with specific reference to their theory, instrumentation, methodology and applications wherever necessary.


Chapter: Pharmaceutical Drug Analysis: Thermoanalytical Analysis

Thermogravimetric Analysis (TGA)

Thermogravimetric Analysis (TGA)
A large number of chemical substances invariably decompose upon heating, and this idea of heating a sample to observe weight changes is the underlying principle of thermogravimetric analysis (TGA).

THERMOGRAVIMETRIC ANALYSIS (TGA)

 

1. THEORY

 

A large number of chemical substances invariably decompose upon heating, and this idea of heating a sample to observe weight changes is the underlying principle of thermogravimetric analysis (TGA). However, TGA may be sub-divided into two heads, namely :

 

(a) Static (or Isothermal) Thermogravimetric Analysis, and

 

(b) Dynamic Thermogravimetric Analysis.

 

1.1. Static Thermogravimetric Analysis

 

In this particular instance the sample under analysis is maintained at a constant temperature for a period of time during which any changes in weight are observed carefully.

 

1.2. Dynamic Thermogravimetric Analysis

 

In dynamic thermogravimetric analysis a sample is subjected to conditions of predetermined, carefully controlled continuous increase in temperature that is invariably found to be linear with time.

 

 

2. INSTRUMENTATION

 

The essential requirements for an instrument (Figure 11.1) meant for thermogravimetric analysis are, namely :

 

(a) A high-precision balance,

 

(b) A furnace adequately programmed for a linear rise of temperature with time, and

 

(a)       A sensitive recorder.


 

2.1. Balances

 

They are usually of two types :

 

(a) Null-point Type : It makes use of an appropriate sensing-element which aptly detects any slightest deviation of the balance beam and provides the application of a restoring force, directly propor-tional to the change in weight, thereby returning the beam to its original null-point. The restoring-force is subsequently recorded either directly or with the aid of a transducer.

 

(b) Deflection Type : It is essentially based on either a conventional analytical balance consisting of helical spring, cantilever beam and strain gauze or a torsion analytical balance involving the conversion of deviations directly into a record of the weight change.

 

 

2.2. Furnace

 

The furnace must be designed in such a fashion so as to incorporate an appropriate smooth input thereby maintaining either a fixed temperature or a predetermined linear-heating programme (e.g., 10°C-600°C per hour).

 

The temperature control of the furnace is satisfactorily achieved via a thermocouple mounted very close to the furnace-winding. The maximum operational temperature may be obtained by using different thermocouples as indicated below :


 

2.3. Recorder

 

The recording device must be such so as to :

 

(i) record both temperature and weight continuously, and

 

(ii) make a definite periodic record of the time.

 

 

3. METHODOLOGY

 

The ‘thermogram’ for calcium oxalate monohydrate (CaC2O4.H2O) is presented in Figure 11.2. The successive plateaus correspond to the anhydrous oxalate (100-250°C), calcium carbonate (400-500°C), and finally calcium oxide (700-850°C). In other words, these plateaus on the decomposition curve designate two vital aspects, namely :

 

(a) clear indication of constant weight, and

 

(b) stable phases within a specified temperature interval. The chemical reactions involved may be summarized as follows :


 

3.1. Interpretation of Thermogram

 

In  the  thermogram  (Figure  11.2),  which  vividly  illustrates  the  thermogravimetric  evaluation  of CaC2O4.H2O, it is ensured that the weight of this product decreases in several stages, namely :


Stage 1 : The water of hydration (or crystallization) from calcium oxalate monohydrate is lost which corresponds to 2.46 mg (12.3%) equivalent to 1 mole of H2O in the temperature range 100-250°C.

 

Actually, the 12.3% weight loss that took place between 100-250°C should correspond to 12.3% of the original formula weight for CaCO3 H2O (FW = 146). Hence, the product being lost has a formula weight of 0.123 × 146 = 17.958 ( ~ 18.0), and it corresponds to H2O.

 

 

Stage 2 : One mole of carbon monoxide is evolved subsequently from calcium oxalate, corresponding to 3.84 mg (19.2%) in the temperature range 400-500°C.

 

The 19.2% weight loss that occurred between 400-500°C should correspond to 19.2% of the original formula weight of 146. Therefore, the product being given out has a formula weight of 1.92 × 146 = 28.0, that corresponds to CO.

 

Stage 3 : Finally, a mole of CO2 is evolved from calcium carbonate that corresponds to 6.02 mg (3.01%) in the temperature range 700-850°C.

 

The weight loss amounting to 3.01% which took place in the range 700-850°C must, in fact, corresponds to 3.01% of the original formula weight of 146. Therefore, the product being evolved has a formula weight of 0.301 × 146 = 43.946 ( ~ 44), and it corresponds to CO2.

It is quite evident that in a multicomponent system wherein more than one component exhibits weight variations and that too at different temperature regions, the composition of the original compound may be estimated as depicted in Figure 11.2.

 

In a situation whereby an inert material is present along with a pure substance, from the generated thermogram the composition of the mixture may be derived from the percentage weight variation which takes place relative to the percentage weight variation observed with the pure compound (A), by employing the following expression :


 

4. APPLICATIONS

 

The most broadly based application of the thermogravimetric analysis (TGA) has been visualized and exploited in the investigation of analytical methods, such as :

 

(i) Determining appropriate forms for many elements,

 

(ii) Screening and testing of substances which may be used as potential analytical standards (primary standard), and

 

(iii) Direct application of the technique in analytical assays.

 

A few typical applications of TGA are, namely :

 

(a) Plateaus for hydrates are sometimes based on the initial water content (i.e., water of crystalliza-tion). It has been observed that in humidified air at low heating rates, hydrates usually give rise to good plateaus.

 

Example : Dehydration of sodium tungstate 28-hydrate [Na2WO4.28 H2O (5 : 12)]

 

Experimental parameters* :

 

A. Humidified air, 300°C/hour,

 

B. Humidified air, 150°C/hour,

 

C. Humidified air 10°C/hour,

 

D. Room air, 10°C/hour,

 

Sample weight : 0.5000 g ;

 

n = Moles water per 5Na2O, 12 WO3

 

(b) Analysis of flue-gas scrubber system in environmental analysis.

 

The flue-gas emitted from a coal-fired-power-plant is subjected to scrubbing by the aid of wet limestone to get rid of sulphur dioxide (SO2) as completely as possible. TGA helps in monitoring the system by carrying out the analysis of the products resulting from the scrubbing process, that mainly consist of (i) CaCO3 ; (ii) CaSO3 . CaSO3 . 1/4 H2O, and (iii) CaSO4 . 2H2O.

 

The thermogram obtained from TGA provides the following valuable informations which suggests the decomposition occurring at three distinct stages thereby causing the loss due to two moles of water, half-a-mole of water and one mole of CO2.


 

(c) The stepwise degradation of organic polymers has received adequate attention which has broadened the in-depth knowledge of polymer chemistry. In this specific instance the sample is either heated under vacuum or in an inert atmosphere (of N2).

 

(d) The thermogravimetric data may be employed to evaluate the kinetic parameters of weight varia-tions in reactions.


Chapter: Pharmaceutical Drug Analysis: Thermoanalytical Analysis

Differential Thermal Analysis (DTA)

Differential Thermal Analysis (DTA)
The difference of temperature between the sample under estimation and a thermally-inert reference material is continuously recorded as a function of furnace temperature in differential thermal analysis (DTA).

DIFFERENTIAL THERMAL ANALYSIS (DTA)

 

1. THEORY

 

The difference of temperature between the sample under estimation and a thermally-inert reference material is continuously recorded as a function of furnace temperature in differential thermal analysis (DTA). In actual practice both TGA and DTA are regarded as complementary techniques whereby information gathered by the usage of one approach is invariably supplemented and enhanced by the application of the other method. The range of phenomena measurable during a DTA-run is found to be much larger than in a TGA-run.

 

It is pertinent to mention here that in the course of TGA many vital processes, for instance : crystalli-zation, crystalline transitions, pure fusion reactions, glass transitions, and solid-state reactions devoid of vola-tile components might not be indicated as they happen to cause little change in weight of the sample. TGA invariably describes with ample precision the stoichiometry related to chemical changes that are indi-cated during DTA by an endothermal or exothermal duration from the base-line.

 

 

2. INSTRUMENTATION

 

A differential thermal analyzer is composed of five basic components, namely :

 

(i) Sample holder with built-in thermocouple assembly,

 

(ii) Flow-control system,

 

(iii) Furnace assembly,

 

(iv) Preamplifier and Recorder, and

 

(v) Furnace Power Programmer and Controller.

 

A typical commercial differential thermal analyzer is schematically illustrated in Figure 11.3.


(a) Thermocouples employed are normally unsheathed Platinum Vs Platinum and Sodium Vs 10% Rhodium. The said two thermocouples help in measuring the difference in temperature between a sample S and an absolutely inert reference substance R, as both are subjected to heating in a ceramic or metal block inside a furnace being operated by a temperature programmer and controller.

(b) The output of the differential thermocouple is amplified adequately through a high gain, low-noise preamplifier and subsequently hooked to the recorder, one axis of which is driven by the block temperature signal and is measured by a third thermocouple.

(c) Heating/Cooling Device : A sufficient versatility is achieved by the aid of a pressure-vacuum, high-temperature electric furnace. An almost constant heating rate is usually achieved by using a motor-driven variable auto transformer.

 

Both heating rates and cooling rates may be conveniently adjusted continuously :

 

(i) From 0°-30°C/minute by some instruments, and

 

(ii) From a choice of several commonly employed heating rates viz., 2°, 4°, 8° and 16°C/minute.

 

Usual workable sample temperatures are upto : 500°C. Exceptional maximum temperatures are upto : 1000°C.

 

(d) Relatively small sample volumes help in two ways : first, they make evacuation easy ; and secondly, they minimize thermal gradients.

 

 

3. METHODOLOGY

 

(i) Insert a very thin thermocouple into a disposable sample tube 2 mm in diameter and containing 0.1-10 mg of sample,

 

(ii) Another identical tube is either kept empty or filled with a reference substance, such as quartz, sand, alumina or alundum powder,

 

(iii) The two tubes are simultaneously inserted into the sample block and subsequently heated (or cooled) at a uniform predetermined programmed rate, and

 

(iv) DTA—being a dynamic process, it is extremely important that all aspects of the technique must be thoroughly standardised so as to obtain reproducible results. A few of these aspects vital aspects are :

 

·              Pretreatment of the specimen,

 

·              Particle size and packing of the specimen,

 

·              Dilution of the specimen,

 

·              Nature of the inert diluent,

 

·              Crystalline substances must be powdered, and sieved through 100-mesh sieve,

 

·              For colloidal particles (e.g., clays), micelle-size is very critical, and

 

·              Either to supress an unwanted reaction (e.g., oxidation), or to explore the study of a reaction (e.g., gaseous reaction product)—the atmosphere should be controlled adequately.

 

Figure 11.4, depicts the differential thermal analysis investigation of calcium acetate monohydrate at a uniform programmed heating rate of 12°C/minute..


The chemical reactions involved in the differentiated thermal analysis of calcium acetate monohydrate may be expressed as follows :


 

Stage I : The endothermal dehydration of calcium acetate monohydrate occurs giving rise to the anhydrous salt. It is easily noticed by an endothermal band on DTA curve between 200°C and 250°C.

 

Stage II : The anhydrous salt undergoes endothermal decomposition reaction at 350-400°C resulting into the formation of CaCO3. It has been observed that this decomposition reaction seems to be almost alike in the presence of either CO2 or Ar.

 

Stage III : The decomposition of calcium carbonate to calcium oxide, which is a function of the partial pressure of the CO2 in contact with the sample. The endothermal band for the carbonate decomposition is sharply peaked spread over a relatively narrower temperature range in an atmosphere of CO2.

 

 

4. APPLICATIONS

 

The various important applications of DTA are :

 

(i) Rapid identification of the compositions of mixed clays,

 

(ii) Studying the thermal stabilities of inorganic compounds,

 

(iii) Critically examining in a specific reaction whether a new compound is actually formed or the product is nothing but an unreacted original substance, and

 

(iv) DTA offers a wide spectrum of useful investigations related to reaction kinetics, polymerization, solvent retention, phase-transformations, solid-phase reactions and curing or drying properties of a product.

Chapter: Pharmaceutical Drug Analysis: Thermoanalytical Analysis

Thermometric Titrations (TT)

Thermometric Titrations (TT)
The thermometric titrations (TT) make use of ‘heats of reaction’ to obtain titration curves. In usual practice, the temperature of solution is plotted against the volume of titrant.

THERMOMETRIC TITRATIONS (T T)

 

1. THEORY

 

The thermometric titrations (TT) make use of ‘heats of reaction’ to obtain titration curves. In usual practice, the temperature of solution is plotted against the volume of titrant. TT is performed by allowing the titrant to flow from a thermostated-burette directly into a solution contained in a thermally-insulated vessel, and subsequently the observed change in temperature of the solution is recorded precisely either during con-tinuous addition of titrant or after every successive incremental addition. The end-point is aptly indicated by a sharp break in the curve.

 

As the dielectric constant of a solvent exerts little effect on the thermometric titrations, the latter may be employed effectively in most non-aqueous media.

 

Hence, in a broader-sense TT may be utilized in a number of reactions with greater efficacy, for in-stance : complexation, precipitation, redox, neutralization. Further, TT can be used to titrate gases against other gases devoid of a liquid-phase ; and to titrate liquid solutions with gaseous reagents.

 

 

2. INSTRUMENTATION

 

A standard thermometric titration assembly essentially consists of the following four vital components, namely :

 

(i) Motor-driven Burette,

 

(ii) Adiabatic Titration Chamber

 

(iii) Thermister Bridge Assembly, and

 

(iv) Recorder.

 

Fiaure 11.5, represents the schematic thermometric titration assembly complete with a bridge-circuit. To minimise heat transfer losses from the solution by its immediate surroundings, the thermometric titrations are usually carried out in an isolated-beaker tightly closed with a stopper having provision for a burette-tip, a motorized-glass stirrer, and a temperature-monitoring arrangement.


 

Procedure :

 

(a) Introduce the titrant from a burette that is duly mounted in a thermostated-water-jacket to maintain the temperature of the titrant within ± 0.05°C,

 

(b) Experimental parameters are predetermined in such a fashion such that the volume of titrant needed for each titration must lie between 1-3 ml,

 

(c) Automated device delivering reagent at a steady and constant rate of 600 µl per minute usually permits recording,

 

(d) Constant-speed motorized stirrer at 600 rpm is employed to effect uniform mixing of solution,

 

(e) Variations in temperature are measured with the help of a sensitive thermister-sensing-element with fast response, that is sealed completely in glass and immersed in solution,

 

(f) In the course of a thermometric titration, the thermister attached to the insulated-beaker is connected to one arm of the Wheatstone Bridge as displayed in Figure 11.5. The values of the circuit component listed are for a thermister having an approximate resistance of 2 K and a sensitivity of 0.04 //°C in the 25°C temperature range. Hence, an observed change of 1°C an unbalanced potential of 15.7 mV, and

 

(g) The heat of reaction is either absorbed or generated upon addition of the titrant to the beaker, thereby unbalancing the Wheatstone Bridge caused by simultaneous variations in the resistance (temperature) in the insulated-beaker thermister. Thus, the bridge unbalance potential is promptly plotted by the recorder.

 

Note : (i) To minimise the temperature variations between the titrant and the solution and also to obviate volume corrections, the concentration of the titrant is invariably maintained 10–100 times higher than that of the reactant, and

(ii) To obtain optimum results, temperatures of the titrant and the solution must be always within 0.2°C of each other before a titration is commenced.

 

 

 

3. METHODOLOGY

 

Thermometric titration curves usually represent both the entropy and the free energy involved. The titrant is added to the solution at a constant rate in order that the voltage output of the thermister-temperature-transducer changes linearly with time upto the equivalence point.

 

TT-method affords exact end-point due to :

 

(a) Coloured solutions, and

 

(b) Poisoning of Electrodes.

 

In usual practice it has been observed that thermometric titrations are mostly feasible with such sys-tems that provide rates of temperature change more than 0.01°C/second.

 

A few typical examples are cited below :


Precautions :

 

(i) Lower limit of concentrations which can be titrated effectively is 0.002 M,

 

(ii) No transfer of heat between the titration vessel and its immediate surroundings is allowed, and

 

(iii) During titration temperature fluctuation must not exceed 0.001°C.

 

 

4. APPLICATIONS

 

Various important applications of thermometric titrations are enumerated below :

 

(i) Precipitation Reactions : e.g., Chloride ions (Cl) with Ag+ ions. Besides, phase relations have been studied extensively in precipitation reactions.

 

(ii) Ion-combination Reactions : e.g., divalent cations like Ca2+, Mg2+ with EDTA (complexometric estimation),

 

(i)          Conversion of Amides to Amines : e.g.,


(iv) Estimation of H2O and (CH3CO)2O concentrations in a mixture : The concentration of either of these reactions in the presence of the other may be determined successfully by measuring the rise in temperature taking place during the exothermic reactions of water and acetic anhydride in gla-cial acid solution along with a trace of perchloric acid (HClO4) acting as a catalyst, and

 

(v) Benzene in Cyclohexane : Benzene may be estimated rapidly with fairly good accuracy in cyclohexane by measuring the heat of nitration, whereby a previously prepared standard nitrating acid mixture (benzene and cyclohexane) and the subsequent temperature rise is noted which is a direct function of the quantity of benzene present.

 

Details involving various experimental parameters for the above estimation are enumerated below :

 

4.1. Estimation of Benzene in Cyclohexane

 

Materials Required : Thermometric titration assembly as per Figure 11.5, minus the burette; a stop-watch or timer ; standard nitrating acid mixture [mix 2 volumes of 70% HNO3 (d = 1.41) with 1 volume of 95% H2SO4 (d = 1.82)] ; Bakelite screw-cap bottle (4 oz. capacity) : 2.

 

Procedure :

 

·              Weigh 50 g of sample in a Bakelite screw-cap bottle and in a similar bottle put the standard nitrating mixture. Place these two bottles in a thermostat maintained at 20°C until the contents have attained an equilibrium temperature,

 

·              Transfer 50 ml of the standard nitrating-acid to the insulated vessel and insert the motorised stirrer. Just wait for about 3-5 minutes and then start the motorized stirrer. After exactly 1 minute record the initial temperature,

 

·              Stop the motor. Insert the sample into the reaction vessel and start the stirrer. Now, start taking readings of the rise in temperature after each interval 1, 2, 3 and 5 minutes respectively, and

 

·              Plot a ‘calibration curve’ between the observed temperature-rise in a 3 minute interval Vs percent benzene present in cyclohexane. Run pure cyclohexane and standards containing 0.5-5.0 percent benzene by weight.


Chapter: Pharmaceutical Drug Analysis: Diazotization (Sodium Nitrite Titration)

Diazotization (Sodium Nitrite Titration)

In general, aromatic primary amino moiety (i.e., Ar-NH2), as present in a host of sulphadrugs viz., succinyl sulphathiazole, sulphamethoxazole, sulphaphenazole and other potent pharmaceutical substances.

DIAZOTIZATION (SODIUM NITRITE TITRATION)

 

INTRODUCTION

In general, aromatic primary amino moiety (i.e., Ar-NH2), as present in a host of sulphadrugs viz., succinyl sulphathiazole, sulphamethoxazole, sulphaphenazole and other potent pharmaceutical substances, for instance sodium or calcium aminosalicylate, isocarboxazid, primaquine phosphate, procainamide hydro-chloride, procaine hydrochloride and dapsone react with sodium nitrite in an acidic medium to yield the corresponding diazonium salts as expressed below :


It is interesting to observe here that the above reaction is absolutely quantitative under experimental parameters. Therefore, it forms the basis for the estimation of pharmaceutical substances essentially contain-ing a free primary amino function as already illustrated earlier.

 

THEORY

Nitrous acid is formed by the interaction of sodium nitrite and hydrochloric acid as follows :

NaNO2 + HCl  ------à NaCl + HNO2

 

The end-point in the sodium nitrite titration is determined by the liberation of iodine from iodide which may be expressed by the following equations :

KI  +  HCl   → HI  +  KCl

 

2HI  +  2HNO2   → I2  +  2NO  +  2H2O

In other words, the small excess of HNO2 present at the end-point can be detected visually by employ-ing either starch-iodide paper or paste as an external indicator. Thus, the liberated iodine reacts with starch to form a blue green colour which is a very sensitive reaction. Besides, the end-point may also be accomplished electrometrically by adopting the dead-stop end-point technique, using a pair of platinum electrodes immersed in the titration liquid.

Chapter: Pharmaceutical Drug Analysis: Diazotization (Sodium Nitrite Titration)

Diazotization (Sodium Nitrite Titration): Assay Methods

1. Preparation of 0.1 M Sodium Nitrite Solution 2. Standardization of 0.1 M Sodium Nitrite Solution with Sulphanilamide 3. Calcium Aminosalicylate 4. Isocarboxazid 5. Phthalylsulphathiazole 6. Cognate Assays

ASSAY METHODS

A number of pharmaceutical substances can be assayed by official methods employing sodium nitrite titrations. A few typical examples are described below to get an indepth knowledge about sodium nitrite titrations.

 

1. PREPARATION OF 0.1 M SODIUM NITRITE SOLUTION

 

Materials Required : Sodium nitrite : 7.5 g.

 

Procedure : Weigh accurately 7.5 g of sodium nitrite and add sufficient DW to produce 1 litre in a 1000 ml volumetric flask.

 

2. STANDARDIZATION OF 0.1 M SODIUM NITRITE SOLUTIOlN WITH SULPHANILAMIDE

 

Materials Required : Sulphanilamide (previously dried at 105°C for 3 hours) : 0.5 g ; hydrochloric acid ( ~ 11.5 N) : 20 ml ; 0.1 M sodium nitrite.

 

Theory : The nitrous acid, generated on the introduction of sodium nitrite solution into the acidic reaction mixture, reacts with the primary amino group of sulphanilamide quantitatively, resulting into the formation of an unstable nitrite that decomposes ultimately with the formation of a diazonium salt. The diazonium salt thus produced is also unstable, and if the reaction mixture is not maintained between 5-10°C, it shall undergo decomposition thereby forming phenol products which may react further with nitrous acid. The reactions involving the formation of the diazonium salt may be expressed in the following manner :

 

NaNO2 + HCl HNO2 + NaCl


At the equivalence point a slight excess of HNO2 is present which must persist for at least 1 minute. This excess HNO2 may be detected by employing either starch iodide strip or paste and designated by the following equation :


Procedure : Weigh accurately 0.5 g of suphanilamide and transfer to a beaker. Add to it 20 ml of hydrochloric acid and 50 ml of DW, stir until dissolved and cool to 15°C in an ice-bath. Add to it 25 g of crushed ice, and titrate slowly with sodium nitrite solution, stirring vigorously, until the tip of the glass rod dipped into the titrated solution immediately produces a distinct blue ring on being touched to starch-iodide paper. The titration is supposed to be complete when the end-point is deducible after the resulting mixture has been allowed to stand for 1 minute. Each 0.01722 g of sulphanilamide is equivalent to 1 ml of 0.1 N sodium nitrite.

 

3. CALCIUM AMINOSALICYLATE

Materials Required : Calcium aminosalicylate : 0.5 g ; hydrochloric acid ( ~  11.5 N) : 10.0 ml ; potassium bromide : 1.0 g ; 0.1 M sodium nitrite ; starch-iodide paper.

 

Theory : The assay of calcium aminosalicylate is based upon the reaction designated by the following equation :


 

Procedure : Weigh accurately about 0.5 g of calcium aminosalicylate, into a funnel placed in the mouth of a 250 ml volumetric flask. Wash through with 10 ml of hydrochloric acid and enough DW to dissolve, add 1.0 g potassium bromide and make up the volume upto 250 ml mark. Pipette 50 ml into a conical flask, cool to below 15°C (in ice-bath) and titrate gradually with 0.1 M sodium nitrite solution while shaking the contents of the flask vigorously and continuously until a distinct blue colour is achieved when a drop of the titrated solution is placed on a starch-iodide paper 5 minutes after the last addition of the 0.1 M NaNO2 solution. Care must be taken to add NaNO2 solution at the rate of 0.1 ml near the end of the titration. Each ml of 0.1 M sodium nitrite is equivalent to 0.01722 g of C14Hl2CaN2O6.

 

4. ISOCARBOXAZID

 

Materials Required : Isocarboxazid : 0.5 g ; glacial acetic acid (99% w/w or 17.5 N) : 20.0 ml ; hydrochloric acid ( ~  11.5 N) : 20.0 ml ; 0.1 M sodium nitrite ; starch-iodide paper.

 

Theory : The estimation is based on the fact that isocarboxazid undergoes rapid cleavage in acidic medium to produce benzylhydrazine. The latter reacts quantitatively with nitrous acid (NaNO2 and HCl) to give rise to benzylazide. The various reactions involved are expressed as follows :


Procedure : Weigh accurately about 0.5 g of isocarboxazid and dissolve it in 20 ml of glacial acetic acid. Add to it 20 ml of hydrochloric acid and 50 ml of DW. Cool to about 15°C in an ice-bath and titrate slowly with 0.1 M NaNO2 while shaking vigorously and continuously until a distinct blue colour is obtained on a starch-iodide paper that lasts for 5 minutes after the final addition of the 0.1 M NaNO2 solution to the titrated solution. Add NaNO2 solution very carefully at the rate of 0.1 ml at a time as the end-point is approached. Each mole of 0.1 M sodium nitrite is equivalent to 0.02313 g of C12H13N3O2 .

 

5. PHTHALYLSULPHATHIAZOLE

 

Materials Required : Phthalylsulphathiazole : 0.5 g ; sodium hydroxide solution (20% w/v in water) : 10.0 ml ; hydrochloric acid ( ~  11.5 N) : 20.0 ml ; 0.1 M sodium nitrite ; starch-iodide paper.

 

Theory : The assay is based upon the reactions designated by the following equations :


Phthalylsulphathiazole undergoes hydrolysis to give phthalic acid and sulphathizole. The latter reacts with nitrous acid to yield the corresponding diazonium salt quantitatively.

 

Procedure : Weigh accurately about 0.5 g of phthalylsulphathiazole and heat on a water-bath for 2 hours after the addition of 10.0 ml of sodium hydroxide solution. Cool the contents of the flask to 15°C in an ice-bath, add to it 10.0 ml of water and 20.0 ml of hydrochloric acid and carry out the titration slowly with 0.1 M sodium nitrite solution. The contents of the flask are shaken thoroughly and continuously until a distinctly visible blue colour is obtained when a drop of the titrated solution is placed on a starch-iodide paper 5 minutes after the last addition of the 0.1 M NaNO2 solution. Towards the approach of the end-point the addition of NaNO2 solution must be at the rate of 0.1 ml. Each ml of 0.1 M sodium nitrite is equivalent to 0.04034 g of C17H13N3O5S2.

 

6. COGNATE ASSAYS

 

A plethora of pharmaceutical substances that can be assayed by the help of sodium nitrite titrations are mentioned in Table 12.1.


Chapter: Pharmaceutical Drug Analysis: Estimation of Phenols and Related Compounds

Estimation of Phenols and Related Compounds

In oxidation-reduction assays the use of bromine is judiciously carried out as an oxidizing agent effectively for such specific compounds which ultimately results into the formation of both bromine substitution and bromine additive compounds.

ESTIMATION OF PHENOLS AND RELATED COMPOUNDS

 

INTRODUCTION

In oxidation-reduction methods bromine is employed as an oxidizing agent in place of iodine, because it is reduced quantitatively be the readily oxidized pharmaceutical organic substances in a reaction which results in either water-insoluble bromine substitution products, for instance :


or corresponding water-insoluble bromine-addition products, such as :



However, the standard solution used does not have bromine (Br2) as such but it does contain an equivalent amount of potassium bromate and an excess of potassium bromide and the resulting mixture on subsequent acidification liberates bromine. The reaction may be expressed as follows :


The liberated bromine helps in oxidizing iodide to an equivalent amount of iodine as shown below :


The free iodine thus produced is titrated with previously standardized sodium thiosulphate solution as depicted below :


 

THEORY

In oxidation-reduction assays the use of bromine is judiciously carried out as an oxidizing agent effectively for such specific compounds which ultimately results into the formation of both bromine substitution and bromine additive compounds. These products of reaction are produced quantitatively and are mostly water-insoluble in characteristics ; and more interestingly they take place in an acidic medium.

 

As it has been discussed earlier, iodine cannot be used directly as an oxidizing agent in such type of assays, whereas the liberated iodine quantitatively produced by the oxidation of iodide with bromine (excess) may be assayed by titrating against sodium thiosulphate solution.

Chapter: Pharmaceutical Drug Analysis: Estimation of Phenols and Related Compounds

Estimation of Phenols and Related Compounds: Assay Methods

Assay methods based on bromine may be classified under the following three heads, namely : (i) Titrations with 0.1 N Bromine, (ii) Titrations with Potassium Bromate, and (iii) Titrations with Potassium Iodate.

ASSAY METHODS

Assay methods based on bromine may be classified under the following three heads, namely :

 

(i) Titrations with 0.1 N Bromine,

 

(ii) Titrations with Potassium Bromate, and

 

(iii) Titrations with Potassium Iodate.

 

 

1. TITRATIONS WITH 0.1 N BROMINE

 

This involves the preparation of 0.1 N bromine solution and subsequent standardization with 0.1 N sodium thiosulphate solution. Bromine solution is also known as Koppeschaar’s Solution in some literature.

 

1.1. Preparation of 0.1 N Bromine Solution

 

Materials Required : Potassium bromate : 3.0 g ; potassium bromide : 15 g.

 

Procedure : Weigh 3 g of potassium bromate and 15 g of potassium bromide in a beaker and dissolve with water. Transfer it quantitatively into a 1 litre volumetric flask and make up the volume with DW.

 

 

1.2. Standardization of 0.1 N Bromine with 0.1 N Sodium Thiosulphate Solution

 

Materials Required : 0.1 N Bromine solution ; hydrochloric acid ( ~ 11.5 N) : 5 ml ; potassium iodide solution (10% w/v in water) : 5.0 ml ; 0.1 N sodium thiosulphate ; starch solution.

 

Procedure : Transfer 25 ml of 0.1 N bromine solution with the help of a pipette into a 500 ml iodine flask and dilute it with 120 ml of DW. Add to it 5 ml of hydrochloric acid, moisten the glass-stopper with water and insert the stopper in the flask. Shake the contents gently. Now, add 5 ml of potassium iodide solution, again lace the stopper and allow the resulting mixture to stand for 5 minutes in the dark. Titrate the liberated iodine with previously standardized 0.1 N sodium thiosulphate solution, adding 3 ml of freshly prepared starch solution towards the end-point. Each ml of 0.1 N sodium thiosulphate is equivalent to 0.01598 g of Br2.

 

1.3. Thymol

 

Materials Required : Thymol : 0.1 g ; N sodium hydroxide : 25.0 ml ; dilute hydrochloric acid (10% v/v of HCl) : 20.0 ml ; 0.1 N bromine ; methyl orange solution (0.1% w/v soln. in 20% alcohol).

 

Procedure : Weigh accurately about 0.1 g of thymol, transfer to a 250-ml iodine flask and dissolve in 25.0 ml of N sodium hydroxide. Add to it 20.0 ml of dilute hydrochloric acid and immediatelv titrate with 0.1 N bromine to within 1 to 2 ml of the calculated end-point. Warm the solution to about 75°C, add 2 drops of methyl orange solution and continue the titration gradually while swirling the contents of the flask thor-oughly after each addition. When the colour of the methyl orange is discharged, add 2 drops of 0.1 N bromine, shake well, add 1 drop of methyl orange solution and shake vigorously. If the colour of the solution is still red, continue the titration dropwise and with constant stirring until the red colour of the indicator is discharged completely. Repeat the alternate addition of 0.1 N bromine and methyl orange solution until the red colour is discharged after the addition of the methyl orange solution. Each ml of 0.1 N bromine is equivalent to 0.003755 g of C10H14O.

 

Calculations :         C10H14O + 2Br2   → C10Hl2Br2O + 2HBr 150.22

 

Since, 1 mole of thymol reacts with 2 mol, 4 equivalent of bromine under the conditions of the assay, the equivalent weight of thymol is 37.55 g, 1/4 gramme molecular weight (i.e., 150.22/4 = 37.55). Therefore, each milliliter of 0.1 N bromine consumed in the reaction with thymol is equivalent to 0.1 × 0.03755 = 0.003755 g or 0.1 meq. of thymol (C10H14O).

 

1.4. Ethacrynic Acid

 

Theory : Active bromine is liberated from the standard solution of bromine in an acidic medium (HCl) that subsequently attacks the double bond present in the side chain of the ethacrynic acid molecule thereby resulting into the formation of the corresponding dibromo derivative. This particular reaction takes place quantitatively. Hence, the reactions involved in this assay may be expressed as follows :


A blank determination is always performed simultaneously to account for the losses caused by the bromine as well as iodine vapours due to the interaction of excess bromine on potassium iodide.

 

Materials Required : Ethacrynic acid : 0.2 g ; glacial acetic acid : 40.0 ml ; 0.1 N bromine : 20.0 ml ; hydrochloric acid ( ~ 11.5 N) : 3.0 ml ; potassium iodide solution ; (10% w/v in water) : 20 ml ; 0.1 N sodium thiosulphate ; starch solution.

 

Procedure : Weigh accurately about 0.2 g of ethacrynic acid, dissolve in 40 ml of glacial acetic acid in a 250 ml iodine flask. Add to it 20 ml of 0.1 N bromine and 30.0 ml of hydrochloric acid, immediately place in position the moistened stopper to the ffask, mix the contents vigorously and allow it to stand in a dark place for 60 minutes (to complete the reaction with bromine). Add to it 100 ml of water and 20 ml of KI Solution and titrate immediately with 0.1 sodium thiosulphate, employing freshly prepared starch solution as an indicator towards the end of the titration. Repeat an operation without the pharmaceutical substance (blank titration) ; thus the difference between the titrations represents the amount of bromine required by the ethacrynic acid. Each ml of 0.1 N bromine is equivalent to 0.01516 g of C13H12Cl2O4.

 

Calculations : From the above equations, we have :


 

1.5. Cognate Assays

 

A number of pharmaceutical substances may be determined quantitatively by titration with bromine as given in Table 13.1.


 

 

 

2. TITRATIONS WITH POTASSIUM BROMATE

 

Potassium bromate can also be employed as an oxidizing agent in the assay of a number of pharmaceutical substances, namely : mephenesin, phenol, and sodium salicylate. This particular method solely depends upon the formation of iodine monobromide (IBr) in relatively higher concentration of hydrochloric acid solution.

 

2.1. Preparation of 0.1 N Potassium Bromate

 

Theory : Potassium bromate can be estimated by the addition of potassium iodide and dilute hydrochloric acid. Thus, we have :


Materials Required : Potassium bromate : 2.784 g.

 

Procedure : Weigh accurately 2.784 g of potassium iodide into a beaker and dissolve it in suffcient DW. Transfer the solution quantitatively into a 1 litre volumetric flask and make up the volume to the mark.

 

2.2. Standardization of 0.1 N Potassium Bromate Solution with the help of 0.1 N Sodium Thiosulphate

 

Materials Required : 0.1 N Potassium bromate ; potassium iodide : 3.0 g ; hydrochloric acid (~ 11.5 N) : 3.0 ml ; 0.1 N sodium thiosulphate ; starch solution : 3.0 ml.

 

Procedure : Transfer an accurately measured volume of about 30.0 ml of 0.1 N potassium bromate solution into a 250 ml iodine flask. Add to it 3.0 g potassium iodide, followed by 3.0 ml of potassium iodide, followed by 3.0 ml of hydrochloric acid. Mix the contents thoroughly and allow it to stand for 5 minutes with its stopper in position. Titrate the liberated iodine with previously standardized 0.1 N sodium thiosulphate, using 3.0 ml of freshly prepared starch solution as an indicator at the end-point. Carry out a blank run using the same quantities of the reagents and incorporate the necessary corrections, if any. Each ml of 0.1 N sodium thiosulphate is equivalent to 0.002784 g of KBrO3.

 

2.3. Mephenesin

 

Theory : Mephenesin undergoes oxidation with bromine to yield a dibromo derivative as expressed in the following equation :


In this instance an excess of potassium bromate is employed. Therefore, any bromide formed [Eq. (a)] is oxidized to bromine, and the excess bromate and the bromine are assayed bromometrically. The reduction of bromate to bromine may be designated as in [Eq. (b)].



Materials Required : Mephenesin : 0.15 g ; 0.1 N potassium bromate : 25.0 ml ; potassium bromide powder : 10.0 g ; hydrochloric acid (25% w/v) : 10.0 ml ; potassium iodide solution (10% w/v in water) : 10.0 ml ; 0.1 N sodium thiosulphate solution ; starch solution.

 

Procedure : Weigh accurately 0.15 g of mephenesin and dissolve in 50 ml of DW into a 250 ml iodine-flask. Add to it 25.0 ml of 0.1 N potassium bromate solution and 10.0 g of powdered potassium bromide. After the dissolution of KBr, add 10 ml of hydrochloric acid, insert the moistened stopper, and after 10 seconds add 10 ml of potassium iodide solution. Titrate with 0.1 N sodium thiosulphate using starch solution as indicator. Each ml of 0.1 N potassium bromate is equivalent to 0.00911 g of C10Hl4O3.

 

2.4. Phenol

 

Theory : Phenol interacts with bromine whereby the former undergoes bromination to yield a water-insoluble 2, 4, 6-tribromophenol. This reaction takes place quantitatively as shown below :


Materials Required : Phenol : 0.5 g ; 0.1 N potassium bromate : 25.0 ml ; potassium iodide (powdered) : 1.0 g ; dilute hydrochloric acid (10% w/w of HCl) : 10.0 ml ; potassium iodide (10% w/v in water) : 10 ml ; chloroform : 10.0 ml ; 0.1 N sodium thiosulphate ; starch solution.

 

Procedure : Weigh accurately 0.5 g of phenol and dissolve in sufficient water to produce 500 ml in a volumetric flask. Mix 25.0 ml of this solution with 25.0 ml of 0.1 N potassium bromate in a 250 ml iodine flask and add to it 1 g of powdered KI and 10.0 ml of dilute hydrochloric acid. Moisten the glass stopper with a few drops of KI solution and place it in position. Set it aside in a dark place for 20 minutes while shaking the contents frequently in between. Add to it 10 ml of KI solution, shake the contents thoroughly and allow it to stand in the dark for a further duration of 5 minutes. Wash the stopper and neck of the flask carefully with DW, add 10 ml chloroform and titrate with the liberated iodine with 0.1 N sodium thiosulphate using freshly prepared starch as an indicator. Carry out a blank titration simultaneously and incorporate any necessary correction, if required. Each ml of 0.1 N potassium bromate is equivalent to 0.001569 g of C6H6O.

 

2.5. Cognate Assays

 

A few other pharmaceutical substances may also be assayed by titrating with 0.1 N potassium bromate as indicated in Table 13.2.


 

3. TITRATIONS WITH POTASSIUM IODATE

 

Potassium iodate is a fairly strong oxidizing agent that may be used in the assay of a number of pharmaceutical substances, for instance : benzalkonium chloride, cetrimide, hydralazine hydrochloride, potassium iodide, phenylhydrazine hydrochloride, semicarbazide hydrochloride and the like. Under appropriate experimental parameters the iodate reacts quantitatively with both iodides and iodine. It is, however, interesting to observe here that the iodate titrations may be carried out effectively in the presence of saturated organic acids, alcohol and a host of other organic substances.

 

The oxidation-reduction methods with potassium iodate invariably based on the formation of iodine monochloride (ICl) in a medium of strong hydrochloric acid solution.

 

 

3.1. Preparation of 0.05 M Potassium Iodate

 

Theory : First of all the potassium iodate is dried to a constant weight at 110°C to make it completely free from moisture and then brought to room temperature in a desiccator. It is pertinent to mention here that KIO3 is a very stable salt and may be obtained in a very pure form. Therefore, it is possible to prepare the standard solutions of KIO3 by dissolving the calculated weight of the salt in water and diluting the same to an approximate volume.

 

Since, the normality of iodate solution varies significantly depending on the nature of the reaction, therefore, in usual practice standard iodate solutions of known molarity are used.

 

The reduction of potassium iodate to iodide is usually not feasible in a direct titrimetric method (unlike the reduction of potassium bromate to bromide) and hence, has no viable application in the official procedures

           ...........................(a)

In this type of reaction, 1 mol of KIO3 is 6 equivalent and a 0.05 M solution would be 0.3 N.

 

In a situation, whereby excess of potassium iodate is employed, any I formed [Eq. (a)] is readily oxidized to iodine, and subsequently the excess iodate and the iodine are estimated by the iodometric proce-dure. Thus, the reduction of the iodate to iodine may be expressed as shown below :

          ............................(b)

In such a reaction, 1 mol of iodate is 5 equivalent and a 0.05 M solution would be 0.25 N. This reaction of iodate is never used in the offcial assay methods.

 

Interestingly, at higher concentrations of hydrochloric acid, both the iodide and iodine obtained as reduction products of iodate [Eqs. (a) and (b)] are quantitatively converted to I+. It forms the basis of official procedures for iodate titrations.

 

The iodine produced intially by the reduction of iodate [Eq. (b)] undergoes solvolysis in a polar solvent as expressed in the following reaction :


The iodine cation forms iodine monochloride (ICl) in a medium having sufficiently high concentration of HCl and the latter is subsequently stabilized by complex ion formation. Thus, we have :


Adding Equations (c) and (d), we may have :


In actual practice, either carbon tetrachloride or chloroform is usually added so as to make the end-point distinctly visible. Iodine is liberated at the initial stages of the titration which renders the chloroform layer coloured. At that material point when all the reducing agent under estimation has been duly oxidized, the iodate completes the oxidation of iodine and iodide to I+, and hence the colour from the chloroform layer disappears.

 

In official methods of analysis i.e., the iodine monochloride method, the reduction of KIO3 can be expressed as follows :


In Eq. (e), 1 mol of KIO3 is 4 equivalent, and a 0.05 solution would be 0.2 N.

 

Materials Required : Potassium iodate : 10.7 g.

 

Procedure : Weigh accurately 10.7 g of pure potassium iodate, previously dried at 110°C to constant weight, in sufficient DW to produce 1 litre in a volumetric flask.

 

 

3.2. Benzalkonium Chloride

 

Materials Required : Benzalkonium chloride : 4.0 g ; chloroform : 60.0 ml ; 0.1 N sodium hydroxide : 10.0 ml ; potassium iodide (5% w/v in water) : 10.0 ml ; hydrochloric acid ( ~ 11.5 N) : 40.0 ml ; 0.05 M potassium iodate.

 

Procedure : Weigh accurately benzalkonium chloride 4.0 g and dissolve it in sufficient DW to make 100 ml. Pipette 25.0 ml into a separating funnel, add 25 ml of chloroform, 10 ml of 0.1 N NaOH and 10 ml of potassium iodide solution. Shake the contents thoroughly, allow to separate and collect the chloroform layer in another separating funnel. Treat the aqueous layer with 3 further quantities each of 10 ml of chloroform and discard the chloroform layer. To the aqueous layer add 40 ml of hydrochloric acid, cool and titrate with 0.05 M potassium iodate till the solution becomes pale brown in colour. Add 2 ml of chloroform and continue the titration until the chlorofonn layer becomes colourless. Titrate a mixture of 29 ml of water, 10 ml of KI solution and 40 ml of hydrochloric acid with 0.05 M potassium iodate under identical conditions (Blank Titration). The differences between the titrations represent the amount of 0.05 M potassium iodate required. Each ml of 0.05 M potassium iodate is equivalent to 0.0354 g of C22H40ClN.


 

3.3. Potassium Iodide

 

Theory : The iodine monochloride method described earlier employing standard potassium iodate is the basis for the official assay of potassium iodide. Vigorous shaking is a prime requirement, as the end-point is approached in this assay, because of the fact that both iodine and iodate in different phases attribute a heterogeneous medium. However, the reaction involving the oxidation of KI by iodate may be designated as shown below :


The reduction of KIO3 may be expressed as :


Hence, from the above equation we have, 1 mol of KIO3 is 4 equivalent and a 0.05 M solution would be 0.2 N.

Thus, we have :


Materials Required : Potassium iodide : 0.5 g ; hydrochloric acid ( ~ 11.5 N) : 35 ml ; chloroform : 5 ml ; 0.05 M potassium iodate.

 

Procedure : Weigh accurately 0.5 g of potassium iodide and dissolve it in about 10 ml of DW. Add to it 35 ml of hydrochloric acid and 5 ml of chloroform. Titrate with 0.05 M potassium iodate till the purple colour of iodine disappears from the chloroform layer. Add the last portion of the iodate solution carefully and dropwise while shaking the contents of the flask vigorously and continuously. Allow to stand for 5 minutes. In case any colour still develops in the chloroform layer continue the titration. Each ml of 0.05 M potassium iodate is equivalent to 0.0166 g of potassium iodide.

 

3.4. Cognate Assays

 

A host of other pharmaceutical substances, namely : cetrimide, hydralazine hydrochloride, phenylhydrazine hydrochloride may be assayed by titration with potassium iodate as mentioned in Table : 13.3.


 

Chapter: Pharmaceutical Drug Analysis: Karl Fischer Method for Determination of Water

Karl Fischer Method for Determination of Water

A plethora of chemical compounds for the determination of small amounts of water present in organic solids, pharmaceutical substances and organic solvents have been devised over a length of time.

KARL FISCHER METHOD FOR DETERMINATION OF WATER

 

INTRODUCTION

A plethora of chemical compounds for the determination of small amounts of water present in organic solids, pharmaceutical substances and organic solvents have been devised over a length of time. But unquestionably the most important of these is the one proposed by Karl Fischer (1935), which is considered to be relatively specific for water*. It essentially makes use of the Karl Fischer reagent which is composed of iodine, sulphur dioxide, pyridine and methanol.

 

Note : Both pyridine and methanol should be anhydrous.

 

THEORY

Water present in the analyte reacts with the Karl Fischer reagent in a two-stage process as shown below :


From Eq. (a) step l, it is obvious that the oxidation of sulphur dioxide takes place by iodine to yield sulphur trioxide and hydrogen iodide thereby consuming one mole of water. In other words, each one molecule of iodine disappears against each molecule of water present in the given sample. It is pertinent to mention here that in the presence of a large excess of pyridine (C5H5N), all reactants as well as the resulting products of reaction mostly exist as complexes as evident from Eqs. (a) and (b).

 

Stability of the Reagent : The stability of the original Karl Fischer reagent initially prepared with an excess of methanol was found to be fairly poor and hence, evidently needed frequent standardization. However, it was estabtished subsequently that the stability could be improved significantly by replacing the methanol by 2-methoxyethanol.

 

It has been observed that the titer of the Karl Fischer reagent, which stands at 3.5 mg of water per milliliter of reagent, falls rapidly upon standing with the passage of time. Hence, the following precautions must be observed rigidly using the Karl Fischer reagent, namely :

 

(a) Always prepare the reagent a day or two before it is to be used,

 

(b) Great care must be taken to prevent and check any possible contamination either of the reagent or the sample by atmospheric moisture,

 

(c) All glassware(s) must be thoroughly dried before use,

 

(d) Standard solution should be stored out of contact with air, and

 

(e) Essential to minimise contact between the atmosphere and the solution during the course of titration.

 

End-point Detection : The end-point of the Karl Fischer titration may be determined quite easily by adopting the electrometric technique employing the dead-stop end-point method. When a small quantum of e.m.f. is applied across two platinum electrodes immersed in the reaction mixture, a current shall tend to flow till free iodine exists, to remove hydrogen and ultimately depolarize the cathode. A situation will soon arise when practically all the traces of iodine have reacted completely thereby setting the current to almost zero or very close to zero or attain the end-point.

 

Limitations of Karl Fischer Titration : The Karl Fischer titration has a number of serious limitations due to possible interferences tantamount to erroneous results, namely :

 

(i) Oxidizing agents, for instance : chromates, Cu(II), Fe(III), Cr2O72–, peroxides, salts, higher oxides,

 

Example :

 

MnO2 + 4C5H5NH+ + 2I   → Mn2+ + 4C5H5N + I2 + H2O

 

(ii) Reducing agents, such as : Sn(II) salts, sulphides, and S2O32–, and

 

(iii) Compounds that have a tendency to form water with the ingredients of the Karl Fischer reagent, for instance :

 

(a) basic oxides : e.g., ZnO ;

 

Example : ZnO  +  2C5H5NH+              →      Zn2+  +  C5H5N  +  H2O

(b) salts of weak oxy-acids e.g., NaHCO3 ;

 

Example :  NaHCO3  +  C5H5NH+        →      Na+  +  H2O  +  CO2  +  C5H5N

 

Note : As H2CO3, carbonic acid, is very unstable ; hence it splits up to yield a mole each of water and CO2.

Chapter: Pharmaceutical Drug Analysis: Karl Fischer Method for Determination of Water

Instrumentation - Karl Fischer Method for Determination of Water

Instrumentation - Karl Fischer Method for Determination of Water
Automated Electrochemical Karl Fischer Analysis

INSTRUMENTATION

Figure 14.1 illustrates a simple dead-stop end-point assembly or a Karl Fischer titration apparatus. The titration vessel is fitted with a pair of identical platinum electrodes, a mechanical stirrer with adjustable speed, and a burette. It will be observed that absolutely little or no current may flow unless and until the solution is totally free from any polarizing substances ; this could perhaps be due to the absorbed layers of oxygen and hydrogen on the anode and cathode respectively. However, the current shall flow only when the two electrodes get depolarized. The Karl Fischer reagent is pumped into the burette by means of hand bellows, the eccess of moisture is usually prevented by employing an appropriate arrangement of desiccant tubes. Alternatively, the stirring may also be accomplished either by using a magnetic stirrer or by means of a suitably dried nitrogen passed gently through the solution during the course of titration.


The end-point is achieved by employing an eiectrical circuit comprising of a microammeter (A), platinum electrodes, together with a 1.5 V to 2.0 V battery connected across a variable resistance of about 2.5 k. First of all the resistance is adjusted in such a manner that an initial current passes through the platinum electrodes in series with a microammeter (A). After each addition of reagent, the pointer of the microammeter gets deflected but quickly returns to its original position. At the end of the reaction a deflection is obtained which persists for 10-15 seconds.

 

1. AUTOMATED ELECTROCHEMICAL KARL FISCHER ANALYSIS

 

Commercially available Modern KF-Titrators are usually equipped with specifically designed titration vessels that are exclusively meant to check and prevent the contact with atmospheric moisture. Quite a few such devices are armed with microprocessors that will perform the requisite operations sequentially in a programmed manner automatically ; and may also dish out a print-out of the desired results including the percentage moisture content. In fact, these Modern KF-Titrators not only afford greater accuracy and precision in results but also offer much ease and convenience in routine analysis as compared to the classical techniques based on either caulometry or controlled current potentiometry using two indicator electrodes.

 

In this procedure the iodide needed for the reaction with water is normally generated within the titration vessel caulometrically as shown below :

 

H2O  +  I2  +  SO2  +  3C5H5N  +  CH3OH    →     2C5H5N.HI  +  C5H5NH.SO4.CH3

 

Thus, the basis of the analysis rests upon the quantitative relationship existing between charge passed and iodine produced by the reagent according to the above reaction. Therefore, the generation of iodine is automatically stopped when an excess of it is detected by the indicator electrode. It essentially consists of two platinum electrodes across which an AC is applied and subsequently a marked drop in voltage between the electrodes takes place as soon as an excess of iodine is present. Normally such automated instruments make use of proprietory reagents exclusively.

 

The major advantage of this approach to KF-analysis being that no calibration is required as the method is absolute and is entirely based on the stoichiometry of the aforesaid equation. It is noteworthy that one may determine the amounts of water ranging between 10 mcg and 10 mg in solid as well as liquid samples.

Chapter: Pharmaceutical Drug Analysis: Karl Fischer Method for Determination of Water

Applications of Karl Fischer Method for Determination of Water in Pharmaceutical Analysis

The Karl Fischer method for the determination of water is used for prednisolone sodium phosphate as described below.

APPLICATIONS OF KARL FISCHER METHOD FOR DETERMINATION OF WATER IN PHARMACEUTICAL ANALYSIS

The Karl Fischer method for the determination of water is used for prednisolone sodium phosphate as described below.

 

1. PREDNISOLONE SODIUM PHOSPHATE

 

Materials Required : Karl Fischer Reagent* : 100 ml ; prednisolone sodium phosphate : 0.2 g ; anhydrous methanol : 20.0 ml.

 

Procedure : Add about 20 ml of anhydrous methanol to the titration vessel and titrate to the amperometric end-point with the Karl Fischer reagent. Quickly add 0.2 g of prednisolone sodium phosphate sample, stir for 1 minute and again titrate to the amperometric end-point with the Karl Fischer reagent. The difference between the two titrations gives the volume (v) of Karl Fischer reagent consumed by the sample.

 

The minimum water equivalent is 3.5 mg of water per ml of Karl Fischer reagent. Hence, the percentage of water w/w in the given sample may be calculated by the following expression :


Precautions :

 

·              The reagents and solutions used must be kept anhydrous and necessary care should be taken through-out to prevent exposure to atmospheric moisture,

 

·              The Karl Fischer reagent should be protected from light and preferably stored in a bottle fitted with an automatic burette, and

 

·              The water equivalent of Karl Fischer reagent should always be determined before use.

 

2. COGNATE ASSAYS

 

A number of other official pharmaceutical substances may be assayed for their water content by the Karl Fischer method as summarized in the following Table 14.1.


 

Chapter: Pharmaceutical Drug Analysis: Tetrazolium Assay of Steroids

Tetrazolium Assay of Steroids

A number of steroids essentially having a α-ketol (21-hydroxy-20 keto) side-chain group, for instance :

TETRAZOLIUM ASSAY OF STEROIDS

 

INTRODUCTION

A number of steroids essentially having a α-ketol (21-hydroxy-20 keto) side-chain group, for instance : hydrocortisone, hvdrocortisone acetate, prednisolone, methylprednisolone, methylprednisolone acetate, flucocinolone acetonide, triamcionolone acetonide and the like—are quantitatively reduced by tetrazolium salts to their respective coloured formazan derivatives. Thus, it is possible to carry out the assay of a number of formulations that contain corticosteroids by using triphenyltetrazolium chloride. The said reaction is usually performed in an alkaline medium (tetramethylammonium hydroxide) between a temperature ranging between 30° to 35°C for a duration of 1 to 2 hours. The absorbance of the resulting formazan derivative producing a red product is usually measured around 484 nm.

Chapter: Pharmaceutical Drug Analysis: Tetrazolium Assay of Steroids

Tetrazolium Assay of Steroids: Theory

Precautions : All these assays are to be carried out strictly in the absence of light and atmospheric oxygen to get optimum results.

THEORY

The oxidation of the α-ketol moiety present in the steroid under examination and the subsequent reduction of triphenyltetrazolium chloride to the corresponding triphenylformazan are depicted in the following reaction :



The triphenyltetrazolium chloride ring undergoes cleavage, as shown by the dotted line, and 2H-atoms are given out by the steroid prednisolone in being converted from C-21, —CH2OH to C-21, —CHO function ; one of the H-atoms from above is utilized in the formation of the open-chain compound i.e., triphenylformazan derivative ; whereas, the second H-atom abstracts the Cl ion as a mole of HCl. The above interaction is of a quantitative nature.

 

However, it is pertinent to mention here that certain steroids esterified at C-21 position, such as : hydrocortisone acetate, methylprednisolone acetate are duly hydrolyzed in the alkaline medium to give rise to the corresponding free C-21 hydroxy steroids and hence, may also be assayed by adopting the same procedure.

 

Precautions : 

All these assays are to be carried out strictly in the absence of light and atmospheric oxygen to get optimum results.

Chapter: Pharmaceutical Drug Analysis: Tetrazolium Assay of Steroids

Assay of Pharmaceutical Substances - Tetrazolium Assay of Steroids

A number of steroidal pharmaceutical substances listed in the official compendia may be assayed by the tetrazolium method of analysis. A few typical examples are described below :

ASSAY OF PHARMACEUTICAL SUBSTANCES

A number of steroidal pharmaceutical substances listed in the official compendia may be assayed by the tetrazolium method of analysis. A few typical examples are described below :

 

 

1. HYDROCORTISONE ACETATE

 

Theory : 

Hydrocortisone acetate is first hydrolysed by strong trimethylammonium hydroxide solu-tion to yield the free 21-hydroxysteroid i.e., hydrocortisone as shown below :


The resulting hydrolysed product is then treated with triphenyltetrazolium chloride and the coloured triphenylformazan is measured at 525 nm.

 

Materials Required : 

Hydrocortisone acetate : 0.350 g ; aldehyde-free absolute ethanol : 100 ml ; triphenyltetrazolium chloride solution [a 0.5% w/v solution of 2,3,5,-triphenyltetrazolium chloride in aldehyde-free ethanol (96%)] : 10 ml ; dilute tetramethylammonium hydroxide solution [Dilute 10 ml of tetramethylammonium hydroxide solution (10%) to 100 ml with aldehyde-free ethanol (96%). It contains about 1% w/v of C4H13NO. To be prepared immediately before use] : 10 ml.

 

Procedure : 

The following steps are to be followed sequentially strictly protected from light :

 

·              Dissolve accurately weighed hydrocortisone acetate 300 to 350 mg in 10 ml aldehyde-free absolute ethanol,

 

·              Transfer 10 ml to a 25 ml graduated flask, add 2 ml of triphenyltetrazolium chloride solution, displace the air in the flask with oxygen-free nitrogen,

 

·              Immediately add 2 ml of dilute tetramethylammonium hydroxide solution and again displace the air with oxygen-free nitrogen,

 

·              Stopper the flask, mix the contents by gently swirling and allow to stand in a water-bath maintained at 30°C for 1 hour,

 

·              Cool rapidly, add sufficient aldehyde-free absolute ethanol to produce 25 ml,

 

·              Mix well and immediately determine the absorbance of the resulting solution in a stoppered cell at the maximum at 485 nm, using in the reference cell a solution prepared at the same time and in the same manner using 10 ml of aidehyde-free absolute ethanol, and

 

·              Repeat the operation using the hydrocortisone acetate EPCRS* in place of the substance being examined under the same experimental parameters.

 

2. COGNATE ASSAYS

 

The following pharmaceutical substances may also be assayed by the above method, namely :

 

(a) Methylprednisolone,

 

(b) Hydrocortisone,

 

(c) Prednisolone, and

      

(a)       Prednisone.

Chapter: Pharmaceutical Drug Analysis: Potentiometric Methods

Potentiometric Methods

Generally speaking the actual concentration of a broad spectrum of solutes may be measured conveniently by forming an appropriate electrochemical cell.

POTENTIOMETRIC METHODS

 

INTRODUCTION

Generally speaking the actual concentration of a broad spectrum of solutes may be measured conveniently by forming an appropriate electrochemical cell. Thus, most electrochemical cells invariably comprise of two electrodes, namely : (a) an Indicator Electrode—the voltage of which solely depends on the thermodynamic activity (i.e., concentration) of one specific component in the solution ; and (b) a Reference Electrode—the voltage of which must be absolutely independent of the nature and composition of the solutions wherein it is immersed. Placing together of these two electrodes in a solution obviously gives rise to an electrochemical cell ; and consequently the voltage thus generated across the electrodes may be determined by connecting it either to a potentiometer or a millivoltmeter that has a sensitivity to measure ± 0.2 mV, besides possessing a high impedence-input of minimum 1012 ohms ().

 

Under these experimental parameters when an extremely feeble current, of the order of less than 5 pA, is drawn from the electrodes, the e.m.f. of the cell may be expressed as below :

 

Ecell = E+ – E + Ej                      ........(a)

where, Ej = e.m.f. at the liquid junction.

In Eq. (a), Ej may be eliminated completely by emp1oying a saltbridge integral with the reference electrode. In usual practice, the loss of electrons or reduction occurs from the prevailing chemical system at the cathode ; whereas the gain of electrons or oxidation takes place at the anode.

Chapter: Pharmaceutical Drug Analysis: Potentiometric Methods

Potentiometric Methods: Theory

In a situation, where a metal M is placed in a solution containing its own ions Mn+, an electrode potential is established across the two electrodes.

THEORY

In a situation, where a metal M is placed in a solution containing its own ions Mn+, an electrode potential is established across the two electrodes, whose actual value is provided by the Nernst equation as shown below :

E = E + (RT/nF) 1n a Mn+                   ..(b)

 

From Eq. (b) the relationship to a cationic electrode, i.e., sensitive only to a cation concentration, may be expressed as :

E = E Yn+, Y + (RT/nF) 1n aYn+.........................(c)

to an anionic electrode :

E = E Xn, X – (RT/nF) 1n a Xn.........................(d)

or to a redox electrode :

       ..........................(e)

where,  

E– = Standard electrode potential (SEP)    (or reduction potential of the half-cell involved),      

a = Thermodynamic activity of the ion to which the electrode is sensitive,         

R = Gas constant (8.314 JK–1 mol–1),        

T = Absolute temperature (K),         

F = Faraday (96500 C/mole of electrons), and    

n = Number of electrons involved in the electrode reaction.   

Direct Potentiometry : The procedure adopted of employing a single measurement of electrode potential to determine the concentration of an ionic species in a solution is usually termed as direct potentiometry.

 

Disadvantages : Direct potentiometry has the following two serious disadvantages namely :

 

(a) From the Nernst Eq. (b) : Considering n = 1, temperature 25°C, RT/nF being a constant, and introducing the factor for the conversion of natural logarithms to logarithms to base 10, the term RT/nF shows a value of 0.0591 V. Therefore, for an ion M+ (monovalent) a ten-time change in the electrode potential E by approximately 60 millivolts (mV) ; whereas for an ion M2+ (bivalent) a change in identical magnitude of activity shall bring forth alternation of E by about 30 mV. Hence, it is evident that to attain a desired accuracy and precision to the extent of 1% in the estimated value for the direct concentration using the technique of direct potentiometry, for M+ ion—the E should be measurable correctly within 0.26 mV ; and for M2+ ion-within 0.1 mV.

 

(b) Uncertainty due to liquid-junction potential (Ej) : It has been observed that the liquid-junction potential (Ej) occurring between the two solutions, one related to the reference-electrode and the other to the indicator-electrode gives rise to a certain quantum of uncertainty with regard to e.m.f. measurement.

 

Remedial Measures : There are two ways to eliminate the above anomaly, namely :

 

(i)          to replace the reference electrode with a concentration-cell i.e., with an electrode comprised of a rod of the same metal as that employed in the indicator electrode plus a solution having the same cation as present in the test-solution, but with a known concentration. Thus, the ionic activity of the metal ion present in the test-solution may be represented by the following expression :

            ....................(f)

(ii) by using one solution which contains a high concentration of KCl or NH4NO3 i.e., such electro-lytes that offer almost identical values for ionic conductivities for both cation as well as anion.

 

Keeping in view the above serious anomalies commonly encountered with direct potentiometry, such as : an element of uncertainty triggered by liquid junction potential (Ej) and high degree of sensitivity required to measure electrode potential (E), it promptly gave birth to the phenomenon of potentiometric titrations,

which subsequently received a high level of sophistication and ultimately turned into a versatile analytical method. As the name suggests, it is indeed a titrimetric method whereby a series of potentiometric measurements are recorded so as to locate the end-point as correctly as possible. In this procedure, it is particularly of more interest to know the exact changes in the observed electrode potential after each addition of the titrant, rather than a precise and accurate electrode potential often brought about by a given solution. Thus, in a way the impact due to liquid-junction-potential (Ej) has been eliminated completely. It is pertinent to mention here that in a potentiometric titration procedure the apparent change in cell e.m.f. takes place not only most rapidly but also most distinctly in the vicinity of the end-point.

 

1. GENERAL CONSIDERATIONS

 

The potentiometric titrations invariably cover a broad-spectrum of chemical reactions that may be classified as follows :

 

(i) Neutralization reactions,

 

(ii) Redox reactions,

 

(iii) Precipitation reactions,

 

(iv) Complexation reactions, and

 

(v) Potentiometric titrations in non-aqueous solvents.

 

The general principles which govern the above different types of reactions will be discussed briefly in the sections that follow :

 

1.1. Neutralization Reactions

 

The accuracy and precision with which the end-point can be determined potentiometrically solely depends upon the quantum of change in the observed e.m.f. in the vicinity of the equivalence point, which in turn entirely depends upon the strength and the concentration of acid and base employed.

 

Merits of the Method : It is found to be useful to titrate a mixture of acids having a significant difference in their strengths, for instance : HCl and CH3COOH (alcoholic). In this case, the first-break in the titration curve signifies that the stronger of the two acids i.e., HCl, gets neutralized ; whereas, the second-break represents the entire completion (i.e., HCl + CH3COOH).

 

In order to get fruitful and reproducible results it is quite necessary that the strengths between either the two acids or bases in question must vary by at least 105 to 1.

 

Demerits of the Method : The neutralization reactions often found to be giving unsatisfactory results in the following two instances. They are :

 

(a) when both the acid and the base are appreciably weak, and

 

(b) when either the acid or the base is very weak (i.e., K < 10–8) and also the prevailing solutions are dilute.

 

Note : In (a) above, an accuracy upto 1% is achievable in 0.1 M solution.

 

Choice of Electrodes :

 

Indicator Electrodes : Hydrogen, Glass or Antimony electrodes ;

 

Reference Electrode : Calomel electrode.

 

1.2. Redox Reactions

 

In this particular case the ratio of the concentrations of the oxidized and reduced forms of ionic species establishes the determining factor. Considering the following reaction,


The electrode potential E is given by the following expression :

               .....................(g)

where,  E = Standard potential of the system.

 

In other words, the potential of the immersed indicator electrode is solely controlled and monitored by the ratio of the ionic concentrations in Eq. (g). Furthermore, in the course of either reduction of an oxidizing agent or vice-versa i.e. the said ratio, and hence the observed potential, undergoes an instant rapid change in the proximity of the end-point of the redox reaction.

 

Example : A typical example is that of titrations of Fe2+ with potassium permanganate or potassium dichromate or cerium (IV) sulphate.

 

Choice of Electrode : Indicator Electrode : Pt wire or foil.

 

The oxidizing agent is usually taken in the burette.

 

1.3. Precipitation Reactions

 

In this the determining factor mainly rests on the solubility product of the resulting nearly insoluble material generated in the course of a precipitation reaction and its ionic concentration at the equivalence point. It is, however, pertinent to mention here that the indicator electrode must readily come into equilib-rium with one of the ions.

 

Example : Titration of Ag+ with a halide (Cl, Br or I) or with SCN (thiocyanate ion).

 

Choice of Electrodes :

 

Reference Electrodes :  Saturated Calomel Electrode (SCE) :

 

Silver-silver chloride Electrode ;

 

Indicator Electrodes :      Silver wire or Platinum wire or gauze plated with silver and sealed into a glass-tube.

 

(It should readily come into equilibrium with one of the ions of the precipitate).

 

Salt-Bridge :                     For the determination of a halide the salt-bridge should be a saturated solution of potassium nitrate.

 

Note : Ion-selective electrode can also be employed.

 

1.4. Complexation Reaction

 

Complexation invariably occurs by the interaction of a sparingly soluble precipitate with an excess amount of the reagent, for instance : the classical example of titration between KCN and AgNO3 as expressed by the following reactions :


In Eq. (h) the precipitate of AgCN is produced at first instance ; consequently, the precipitate of AgCN initially produced gets dissolved by further addition of KCN to afford the complex ion [Ag(CN)2] Eq. (i) and only a negligible quantum of Ag+ ions remain in the solution. Thus, the entire process from ab initio to the final stage of titration may be divided into three distinct portions, namely :

 

(i) Upto end-point : Here, all the available CN ion has been virtually converted to the complex ion. At this stage the ever increasing concentration reflects a gradually increasing concentration of Ag+ ions, thereby slowly enhancing the potential of the Ag-electrode dipping in the solution,

 

(ii) At the end-point : It is usually visualized by a distinct and marked rise in potential, and (iii) Beyond end-point : Further addition of AgNO3 brings about only a gradual change in e.m.f. and AgCN gets precipitated. Ultimately, a second sudden change in potential may be visualized at this juncture when practically most of the CN ion gets precipitated as AgCN.

 

Choice of Electrodes :

 

Indicator Electrode : Silver electrode ;

 

Reference Electrodes : Colomel electrode ; Mercury-mercury (I) sulphate electrode.

 

Salt-Bridge : A saturated solution of KNO3 or K2SO4 isolated from the reference electrode.

 

1.5. Potentiometric Titration in Non-Aqueous Solvents

 

The potentiometric technique has proved to be of great significance and utility for determining end-points of titrations in a non-aqueous media. The mV scale rather than the pH scale of the potentiometer must be used for obvious reasons, namely :

 

(i) pH scale based upon buffers has no logical significance in a non-aqueous media, and

 

(ii) the potentials in non-aqueous media may exceed the pH scale.

 

The resulting titration curves are more or less emperical and afford a reasonably dependable and reproducible means of end-point detection.

Choice of Electrodes :

Indicator Electrodes : Glass electrode ;

Reference Electrode: Calomel electrode ;

Salt-Bridge: A saturated solution of KCl.

 

 

2. END-POINT DETERMINATION

In fact, there are several acceptable means to graph the potentiometric titration data generated from an actual titration in order to locate the exact (or nearest) end-point. These may be illustrated exclusively by employing the titration data provided in Table 16.1, between 25 ml of 0.01 M NaF and 0.01 M La (NO3)3.


The simplest and the most commonly used method is to plot the cell voltage E, millivolts (mV), versus the volume (ml) of titrant added. Ultimately, the end-point is determined from the point of maximum slope of the curve i.e., the point of inflexion, as depicted in Figure 16.1 (a). However, the degree of accuracy and precision with which this point of inflexion can be located from the plotted graph largely depends on the individual number of data points observed in the close proximities of the end-point.

 

Figure 16.1 (a) gives rise to a sigmoid-curve (or S-shaped curve) obtained either by using an appropriate equipment (automatic titrators) that plots the graph automatically* as the titration proceeds, or manually by plotting the raw experimental data. The central portion of the sigmoid curve, in fact is the critical zone where the point of inflexion resides and this may be located by adopting any one of the follow-ing three procedures, namely :

 

(i)          Method of parallel tangents,

(ii)       Method of bisection, and

(iii)     Method of circle fitting


Figure 16.1 (b) is obtained by plotting E/V against V which is termed as the first derivative curve. It gives a maximum at the point of inflexion of the titration curve i.e., at the end-point.

 

Figure 16.1 (c) is achieved by plotting the slope of the frst derivative curve against the volume of titrant added i.e., by plotting 2E/V2 Vs V and is known as the second derivative curve. Thus, the second derivative becomes zero at the point of inflexion and hence, affords a more exact measurement of the equiva-lence point.

 

The titration error (i.e., difference between end-point and equivalence point) is found to be small when the potential change at the equivalence point is large. Invariably, in most of the reactions employed in potentiometric analysis, the titration error is normally quite small and hence may be neglected.


Chapter: Pharmaceutical Drug Analysis: Potentiometric Methods

Potentiometric Methods Instrumentation: Electrodes, Automatic Titrator

Potentiometric Methods Instrumentation: Electrodes, Automatic Titrator
Figure 16.2 illustrates a typical assembly for carrying out a potentiometric titration. Broadly speak-ing, the titration essentially comprises of measuring and subsequently recording a cell potential in terms of either mV or pH, after each sequentially known addition of reagents.

INSTRUMENTATION

Figure 16.2 illustrates a typical assembly for carrying out a potentiometric titration. Broadly speak-ing, the titration essentially comprises of measuring and subsequently recording a cell potential in terms of either mV or pH, after each sequentially known addition of reagents.

 

In usual practice, the titrant (e.g., Lanthanum Nitrate) is added in large amounts at the initial stage ; as the end-point is approached, which is marked by distinct larger potential changes per addition, the subse-quent increments are made smaller to the tune of 0.1 ml for each addition.

 

It is always advisable to allow sufficient time lapse after each addition of titrant so as to attain equilibrium. A gentle and uniform stirring by means of a magnetic stirrer also helps in hastening the ultimate achievement of equilibrium :


The various components shown in Figure 16.2, are as follows :

 

A = Saturated Calomel Electrode (SCE),

 

B = Indicator Electrode,

 

C = Burette to discharge titrant in the reacting vessel,

 

D = pH Meter with a mV scale,

 

E = Magnetic stirrer with variable speed, and

 

F = Magnetic Guide.

 

1.  ELECTRODES

 

The accurate, precise and effective potentiometric measurements are evidently made with the aid of the following two types of electrodes namely :

 

(i) Reference Electrodes, such as :

 

(a) Standard Hydrogen Electrode,

 

(b) Saturated Calomel Electrode, and

 

(c) Silver-silver Chloride Electrode.

 

(ii) Indicator Electrodes, such as :

 

(a) Metal Indicator Electrode, and

 

(b) Membrane Indicator Electrode.

 

These various kinds of electrodes will be discussed briefly, along with a diagrammatic representation wherever possible, in the sections that follow :

 

1.1. Reference Electrodes

 

In general, reference electrodes exhibit a potential which is absolutely independent of the solution wherein it is used. Besides, it must not display any significant change even when a small quantum of current is passed through it.

1.1.1. Standard Hydrogen Electrode (SHE)

The standard hydrogen electrode (SHE), as shown       in Figure 16.3, is considered to be the universally accepted reference electrode. The metal electrode comprises of a small piece of platinum foil with a finely divided platinum,usually termed as platinum black because of its dark look.The coated foil is immersed in an acidic medium having a hydrogen ion activity of 0.1, and through which H2 gas is bubbled at a partial pressure of 1.0 atm (unit activity). The Pt-black-foil possesses a relatively large-surface-area thereby enabling it to absorb an appreciable amount of  H2+H (a = 1.0 M) gas, ultimately bringing it into direct contact with the surrounding H+ ions at the electrode surface. Consequently,the Pt-electrode attains a potential which is finally estimated   Figure 16.3 : A Standard Hydrogen by the relative tendencies of H+ ions to undergo reduction Electrode. and H2  (g) to undergo oxidation simultaneously. It is an usual convention to assign the potential of SHE a value exactly equal to zero at all temperatures.


1.1.2. Saturated Calomel Electrode

 

The schematic diagram of a commercial saturated calomel elec-trode (SCE) is depicted in Figure 16.4. It essentially consists of a plati-num wire immersed in a slurry made up of pure mercury, solid mercurous chloride Hg2Cl2 (commonly known as calomel), and aqueous saturated solution of KCl, packed in the inner-tube (c) having a small hole (B). The outer-tube contains a saturated solution of KCl (D) having a porous ce-ramic fiber (A) at its lower end. It serves as a salt-bridge which allows the entire set-up immersed directly into the solution to be measured. The po-rous ceramic fiber permits establishment of electrical contact between one side of the salt-bridge and the solution under the examination and serves as a barrier between the said two solutions. The small opening at the top end of the salt-bridge tube serves as a fill-hole (E) through which either KCl solution may be filled or replaced as and when required. 


The differ-ent parts of the saturated calomel electrode are as follows :

 

A = Porous ceramic fiber,

 

B = Small-hole,

 

C = Slurry of Hg, Hg2Cl2 and saturated KCl, D = Saturated KCl solution,

 

E = Fill-hole, and

 

F = Electrical lead.

 

The half-cell of SCE may be expressed as :


According to the Nernst equation, the potential of the electrode is represented by :


assuming the activities of Hg and Hg2Cl2 solid are both unity.

 

Advantages : The two major advantages of SCE are, namely :

 

(a) Concentration of Cl does not alter appreciably even if some of the solvent gets evaporated, and

 

(b) Generates a comparatively small junction potential (Ej) at the two salt-bridge solution interfaces.

1.1.3. Silver-silver Chloride Electrode

 

Figure 16.5 shows a silver-silver chloride electrode which com-prises of a silver wire coated with silver chloride (B) and is duly placed in a 1 M KCl solution saturated with AgCl (C).


 

The half-cell of silver-silver chloride electrode may be represented as :


for which half-reaction would be :


According to the Nernst equation, the potential of the electrode is expressed as :


considering that the potential of the electrode is solely dependent on the concentration of Cl.

 

The various components of a silver-silver chloride electrode are, namely :

 

A = Porous ceramic fiber,

 

B = Ag wire coated with AgCl,

 

C = 1 M KCl saturated with AgCl,

 

D = Fill-hole, and

 

E = Electrical lead.

 

1.2. Indicator Electrodes

An indicator electrode is invariably used exclusively in conjunction with a reference electrode the response of which solely depends upon the concentration of the analyte.

 

1.2.1. Metal Indicator Electrode

 

Metal indicator electrodes develop a potential which is usually determined by the equilibrium posi-tion of a redox half-reaction at the electrode surface. These are further classified into the following three types, namely :

 

(i) First order electrodes,

 

(ii) Second order electrodes, and

 

(iii) Inert electrodes.

 

which shall be discussed briefly below.

 

1.2.1.1.  First-order electrodes

 

They are essentially comprised of a metal placed in a solution of its respective ions, for instance : a silver wire immersed into a AgNO3 solution. Hence, the reversible half reaction may be represented as:


and the corresponding Nernst equation would be as follows :


The metals that display reversible half reactions with their respective ions and are found to be suitable for employing as first-order electrodes are Ag, Hg, Cu, Cd, Zn, Bi, Pb and Sn. However, several other metals like : Fe, Co, Cr and W are not useful due to the following reasons :

 

(i) Non-reproducible potentials largely influenced by impurities,

 

(ii) Irregular crystal structures in the solid-state, and

 

(iii) Formation of oxide layers on their surfaces.

 

1.2.1.2.  Second-order electrodes

 

Sometimes a metal electrode may be directly responsible to the concentration of an anion which either gives rise to a complex or a precipitate with the respective cations of the metal. Therefore, they are termed as second-order electrodes as they respond to an ion not directly involved in the electron transfer process. The silver-silver chloride electrode, as already described, is a typical example of a second-order electrode. In this particular instance, the coated Ag wire when dipped in a solution, sufficient AgCl dissolves to saturate the layer of solution just in contact with the respective electrode surface. Thus, the Ag+ ion concentration in the said layer of solution may be determined by the status of the solubility product (Ksp) equilibrium :


Disadvantages : The four serious disadvantages are, namely :

 

(a) May be used effectively over a certain range of anion concentration only so that the solution must remain saturated with the substance coating the metal,

 

(b) In the case of Ag-AgCl electrode, a very low Cl ion concentration would dissolve the AgCl-coating to a great extent,

 

(c) Likewise, a very high concentration of Cl ion would result into the formation of soluble complex ions as shown below :


(d) Ions like Br, I SCN, CN and S2– cause interference while using a Ag-AgCl electrode to esti-mate Cl ion concentrations because of the facts that these ions usually form salts with Ag+ ion which are significantly less soluble than AgCl.

 

1.2.1.3. Inert electrodes

 

Inert electrodes comprise of chemically inert conductors, for instance : Au, Pt and C which do not necessarily take part either directly or indirectly in the various redox processes. However, the potential developed at an inert electrode solely depends upon both the nature as well as the prevailing concentration of the different redox-reagents present in the solution.

Example : A Pt-electrode placed in a solution consisting of both Fe3+ and Fe2+ ions develops a poten-tial which is duly represented by the Nernst equation for ions as given below :


Advantages : The two main advantages of inert-electrodes are, namely :

 

(a) Exhibit no chemical selectivity, and

 

(b) Respond to any reversible redox-system.

 

1.2.2. Membrane Indicator Electrodes (or Ion-Selective Electrodes)

 

The underlying principle of this type of electrode is that the potential developed due to an unequal charge generated at the opposing surfaces of a ‘special’ membrane. The resulting charge at each surface of the membrane is exclusively controlled and monitored by the exact position of an equilibrium involving analyte ions, which in turn, solely depends upon the concentration of those ions present in the solution. Ion-selective electrodes occupy a very important place in the analytical chemistry by virtue of the fact that one may use the acquired skill, expertise and wisdom to design and commercially prepare membranes that are practically selective towards a specific ion besides producing potentials according to the Nernst-type equation. These are classified further into the following four kinds, namely :

 

(i) Glass membrane electrodes,

 

(ii) Polymer (liquid) membrane electrodes,

 

(iii) Crystalline membrane electrodes, and

 

(iv) Gas-sensing electrodes,

 

which will be described below briefly :

 

1.2.2.1. Glass Membrane Electrodes

 

The diagram of a typical glass-membrane electrode is depicted in Figure 16.6. The internal element essentially comprises of a Ag-AgCl electrode (B) dipped in a pH 7 buffer saturated with AgCl (A). The thin, ion-selective glass membrane (I) is carefully fused to the bottom of a high resistance non-responsive glass tube (H) so that the entire membrane may be immersed while taking measurements.

 

The half-cell of glass-membrane electrode may be expressed as :

 

Ag (s) | AgCl [saturated], Cl (inside), H+ (inside) | glass membrane | H+ (outside)

 

According to the Nernst equation, the potential of the electrode is represented by :

              .......................(i)

Now, separating the ratio of H+ ion concentrations into two log terms we may have :


As the activities (i.e., concentrations) of H+ and Cl in the internal electrolyte solution are constant, the first three components on the right hand side of Eq. (ii) may be confined into a single constant, K, and the equation could be rewritten as :

E = K + 0.0592 log [H+]outside

The various components of Figure 16.6 are as follows :                                                               

A = pH 7.0 buffer solution saturated with AgCl,         

 

B = Ag-AgCl internal reference electrode,           

                                                         

C = Mercury connection,                                                                 

                                                         

D = Connecting wire,                                          

                                                         

E = Shield,                                                          

                                                         

F = Cap,                                                              

         

G = Shielded and insulated connecting wire,                                                                

H = High resistance non-responsive glass, and                                                   

I = H+–selective glass membrane.                                                             

 

1.2.2.2. Polymer (Liquid) Membrane Electrode

Figure 16.7. illustrates a calcium-ion polymer (liquid) membrane electrode. It has a close similarity to the glass pH electrode, and it essentially comprises of an internal Ag-AgCl electrode (B) and an internal reference solution having a fixed composition e.g., aqueous CaCl2 saturated with AgCl (C). The liquid calcium di (n-decyl) phosphate, [{CH3(CH2)8CH2O}2PO2]2 Ca serves as the membrane, positioned at the lower end of the electrode, and strategically immobilized by a thin disk of PVC (polyvinyl chloride) (A) which is not penetrable with water.


 

Thus, the calcium di (n-decyl) phosphate forms an equilibrium with its ions at every membrane surface :


where, R = CH3(CH2)8 –CH2i.e., n-decyl hydrocarbon chain.

 

Interestingly, the didecylphosphate anion represents a fixed component of the non-aqueous liquid membrane. As the concentration of Ca+ ions present in the solutions on either side of the membrane varies ; hence, the concentration of didecylphosphate anion at every membrane surface would also vary accordinly, thereby causing a potential that may be expressed by the following equation :

           ..........................(i)

  • Classifieds
  • Chapter
  • Activiti
  • Hanex solid surface
  • The Case
  • Activities
  • 1 Ml
  • Advantages of credit cards
  • Anna University
  • Aqueous
  • Classifieds
  • Chapter
  • Activiti

Again, separating the ratio of Ca2+ ion concentration into two log terms we have :


Since the activities of Ca2+ and Cl in the internal electrolyte solution are more or less constant, the first three terms on the right hand side of Eq. (ii) may be combined to a single constant, K, and the same equation may be rewritten as follows :

      .....................(iii)

The different essential components of Figure 16.7 are as stated below :

 

A = Calcium di (n-decyl)-phosphate immobilizes in PVC,

B = Silver-silver chloride electrode, and

C = Aqueous CaCl2 saturated with AgCl.


 

1.2.2.3.  Crystalline Membrane Electrodes

 

The crystalline membrane electrodes have a very close similarity to those of glass-membrane electrodes except that glass has been replaced with crystalline membrane. In fact, these electrodes offer a means to devise responsive to anions by making use of a membrane containing specific anionic sites.

 

Example : Fluoride-ion Electrode : In this particular instance the mem-brane essentially comprises of a single crystal of lanthanum fluoride (LaF3), usually doped with a slight trace of europium (II), Eu2+, so as to initiate the crystal defects required for establishing its electrical conductivity. Therefore, the potential developed at each surface of the membrane is finally determined by the exact status of the equilibrium :


and is represented by the following equation :


Salient features of Fluoride-Ion Electrode are, namely :

 

(a) At low pH, F ion gets readily converted to the weak acid HF (pKa = 3.17) thereby rendering the electrode insensitive,

(b) It is almost 103 times more specific and selective for F ion as compared to other common anions, of course with the exception of OH ion, and

(c) This  electrode can tolerate conveniently the maximum concentration of OH ion to the extent of 1/10th as compared to the F ion concentration.

 

Table 16.2 records the characteristics of certain selected crystalline-membrane electrodes.



1.2.2.4.  Gas-Sensing Electrode

The schematic diagram of a gas-sensing electrode is illustrated in Figure 16.8, that comprises of essentially a reference electrode (E), a specific-ion electrode (B), and   internal electrolyte solution (F) contained in a cylindrical plastic tube (G). One end of the plastic tubing is provided with a thin, replaceable, gas-permeable membrane that separates the internal electrolyte solution from the external solution containing gaseous analyte. However, the exact composition and specifications of this gas-permeable membrane is  usually  described by  its respective manufacturers. It is normally made up of a thin microporous film fabricated from a hydrophobic plastic material.


The various components of Figure 16.8 are as follows :

A = Gas permeable membrane,

B = Specific ion electrode (a glass electrode),

C = ‘O’-Ring to hold the membrane,

D = External solution containing dissolved gaseous analyte,

E = Reference electrode (a Ag/AgCl electrode),

F = Internal electrolyte solution, and

G = Plastic tube.

 

In general, it must fulfil the following requirements, namely :

 

(a) It should act as a 100% barrier for both water and electrolytes i.e., they must not pass through this membrane,

 

(b) Pores of the film contain exclusively air or other gases to which it is exposed, and

 

(c) A solution containing a particular gaseous-analyte, for instance CO2, when comes in contact with the membrane the former migrates swiftly into the pores of the latter, as expressed by the follow-ing reaction :

        .......................(a)

As the number of pores in the gas-permeable membrane are plenty, therefore, an equilibrium is established. Evidently, the carbon-dioxide present in the pores is in direct contact with the internal-electrolyte solution (F), thereby giving rise to a second equilibrium reaction that may be represented as follows :

              ............................(b)

As a result of the above two reactions, Eq. (a) and Eq. (b), the external solution containing dissolved gaseous analyte (D) immediately attains an equilibrium with the film of internal electrolyte solution (F) present very close to the gas-permeable membrane (A). Thus, another equilibrium gets established that affords the pH of the internal-surface film to alter according to the following expression :

          .........................(c)

The above change in pH is instantly detected by means of a Ag/AgCl reference electrode pair (E) dipped in the film of internal solution as shown in Figure 16.8.

Therefore, the net overall reaction caused by the entire aforesaid process may be achieved by simply summing up the three chemical reactions (a), (b), and (c) to give :

              .............................(d)

The equilibrium constant, K, for Eq. (d) may be represented by :

                ..............................(e)

Assuming that the concentration of HCO3–present in the internal-electrolyte solution (F) is made comparatively high such that its concentrations do not undergo any appreciable change due to the migrating CO2, we may have :

                .............................(f)

Thus, Eq. (f) may be rewritten as follows :

   ...........................(g)

where, a1 = Internal hydrogen ion activity

 

It is given that :

 

Ecell = L + 0.0592 log a1            ...(h)

Consequently, the potential of the electrode system present in the internal-electrolyte solution (F) is solely dependent on a1 according to Eq. (h). Hence, substituting Eq. (g) into Eq. (h), we may have :


 

In short, therefore, the potential of the cell comprising of the Ag/AgCI reference electrode (E) i.e., the internal reference and the specific ion electrode (B) i.e., the indicator electrode is normally determined by the CO2 concentration of the external solution containing dissolved gaseous analyte.

 

Notes : (i) None of the electrodes (reference & indicator) ever gets in contact directly with the analyte solution, and

 

(ii) The only substances which may cause interference with the measurement of potential are dissolved gases which may have a free-access through the membrane, and in turn may affect the pH of the internal solution accordingly.

 

Selectivity of Gas-sensing Electrode : The selectivity of the gas-sensing electrode may be enhanced by making use of such an internal electrode which is particularly sensitive enough to certain species other than the H+ ion.

 

Example : Nitrate-sensing electrode is employed to cater for a cell which will be sensitive exclusively to nitrogen dioxide (NO2). The equilibrium of such a reaction may be represented as follows :


The nitrate-sensing electrode allows the determination of NO2 in the presence of certain specific gases only, for instance, NH3, SO2 and CO2, that will also affect the change in pH of the internal electrolyte solution significantly.

 

2. AUTOMATIC TITRATOR (PRESET END-POINT TITRATOR)

 

The schematic diagram of an automatic titrator* is shown in Figure 16.9.


The various components of Figure 16.9 are, namely :

 

A = Magnetic stirrer with a Regulator,

 

B = Solenoid valve, and

 

C = Error Signal,

 

D = Amplifier,

 

E = Electronic Switch,

 

F and G = AC-Source,

 

H = End-point Potential,

 

I = Calibrated Potentiometer, and

 

J = Accurately calibrated Burette.

 

In this case a preset equivalence point potentiometer is applied at the two electrodes with the aid of a calibrated potentiometer (I). It will give rise to an “error” signal (C) provided a difference is caused between this potential and that of the electrodes. The feeble signal thus generated is duly amplified (D) and closes an electronic switch (E) which allows the electricity to flow through the solenoid operated value (B) of the burette (J). As the titration proceeds, the error signal (C) starts approaching a zero value, subsequently the current to the solenoid valve (B) is instantly switched off, and finally the flow of titrant from the burette (J) comes to a halt. The solution of the sample is constantly and uniformly stirred with the help of a magnetic stirrer (A).

 

Chapter: Pharmaceutical Drug Analysis: Potentiometric Methods

Applications of Potentiometric Titrations in Pharmaceutical Analysis

Potentiometric titrations have been used extensively for assay of a number of official compounds. A few typical examples would be described here, namely : Nitrazepam ; Allopurinol ; and Chloridine hydrochloride.

APPLICATIONS OF POTENTIOMETRIC TITRATIONS IN PHARMACEUTICAL ANALYSIS

Potentiometric titrations have been used extensively for assay of a number of official compounds. A few typical examples would be described here, namely : Nitrazepam ; Allopurinol ; and Chloridine hydrochloride.

 

A. Assay of Nitrazepam :

 

Materials Required : Nitrazepam : 0.25 g ; acetic anhydride : 25.0 ml ; perchloric acid (0.1 M) : 250 ml ; a Potentiometer ; a Magnetic Stirrer ; Burette (50 ml) ;

 

Theory : Nitrazepam is a weakly basic compound and hence, it may be titrated conveniently by means of a non-aqueous titration technique and determining the end-point potentiometrically.


Procedure : Weigh accurately 0.25 g of nitrazepam and dissolve in 25.0 ml of acetic anhydride. Titrate with 0.1 M perchloric acid placed in a burette and adding it carefully into the beaker kept on a magnetic stirrer potentiometrically. Each ml of 0.1 M perchloric acid is equivalent to 28.13 mg of C15H11N3O3.

 

B. Assay of Allopurinol :

 

Materials Required : Allopurinol : 0.12 g ; dimethylformamide : 100.0 ml ; tetrabutylammonium hydroxide (0.1 M) : 1 L ;

 

Preparation of 0.1 M Tetrabutylammonium hydroxide (1 Litre) : Dissolve 40 g of tetrabutylammonium iodide in 90 ml of anhydrous methanol, add 20 g of finely powdered silver oxide and shake vigorously for 1 hour. Centrifuge a few ml of the mixture and test the supernatant liquid for iodides. If a positive reaction is obtained add a further 2 g of silver oxide and shake for 30 minutes. Repeat this procedure until the mixture is free from iodides, filter through a fine sintered-glass filter and wash the reaction vessel and filter with three 50-ml quantities of toluene. Add the washings to the filtrate and add sufficient toluene to produce 1000 ml. Pass dry carbon-dioxide free N2 through the solution for 5 minutes.

 

Standardization of 0.1 M Tetrabutylammonium Hydroxide : To 10 ml of dimethylformamide add 0.05 ml of a 0.3 % w/v solution of thymol blue in methanol and titrate with the tetrabutylammonium hydroxide solution until a pure blue colour is produced. Immediately add 0.2 g of benzoic acid, stir to effect solution and titrate with the tetrabutylammonium hydroxide solution until the pure blue colour is restored. Protect the solution from atmospheric CO2 throughout the titration. The volume of titrant used in the second titration represents the amount of tetrabutylammonium hydroxide required. Each ml of 0.1 M tetrabutylammonium hydroxide Vs is equivalent to 12.21 mg of C7H6O2.

 

Procedure : Dissolve 0.12 g of accurately weighed allopurinol in 50 ml of dimethylformamide, with gentle heating, if necessary. Titrate to the colour change of the indicator that corresponds to the maximum absolute value of dE/dV in a potentiometric titration (where E is the electromnotive force and V is the volume of the titrant). Each ml of 0.1 M tetrabutylammonium hydroxide Vs is equivalent to 13.61 mg of C5H4N4O.

 

C. Clonidine Hydrochloride :

 

Materials Required : Clonidine hydrochloride : 0.2 g ; ethanol (96%) : 100 ml ; 0.1 M ethanolic sodium hydroxide Vs : 1 L (Add 3.3 g of 10 M sodium hydroxide solution to 250 ml of absolute ethanol).

 

Standardization of 0.1 M Ethanolic Sodium Hydroxide Solution Vs : Dissolve 0.2 g of benzoic acid in a mixture of 10 ml of ethanol (96%) and 2 ml of water and titrate with the ethanolic sodium hydroxide solution using 0.2 ml of thymolphthalein solution (a 0.1 % w/v solution of thymolphthalein in ethanol (96%) as indicator. Each ml of 0.1 M ethanolic sodium hydroxide Vs is equivalent to 12.21 mg of C7H6O2.

 

Procedure : Dissolve 0.2 g of clonidine hydrochloride in 70 ml of ethanol (96%) and titrate with 0.1M ethanolic sodium hydroxide Vs determining the end-point potentiometrically. Each ml of 0.1 M ethanolic sodium hydroxide Vs is equivalent to 26.66 mg of C9H9Cl2,N3, HCl.

 

1. COGNATE ASSAYS

 

A plethora of official drugs are assayed by the potentiometric method in various official compendia, and a few selected examples are given in Table 16.3, which may be assayed potentiometrically :


Chapter: Pharmaceutical Drug Analysis: Amperometric Methods

Amperometric Methods

An amperometric method or amperometry is concerned with the measurement of current under a constant applied voltage ; and under such experimental parameters the concentration of the ‘analyte’ exclusively determines the quantum and magnitude of the current.

AMPEROMETRIC METHODS

 

INTRODUCTION

An amperometric method or amperometry is concerned with the measurement of current under a constant applied voltage ; and under such experimental parameters the concentration of the ‘analyte’ exclusively determines the quantum and magnitude of the current. Hence, these measurements may be employed effectively to record the alteration in concentration of an ion in question in the course of a titration, and ultimately the end-point is established. This specific process is commonly referred to as amperometric method or amperometry.

 

In this particular case, the total current flowing shall remain almost equal to the current carried by the ions that undergoes equal electrolytic migration together with the current caused on account of the diffusion of the ions. Thus, we have :

 

I = Id + Im

 

where   I = Total current,

 

Id = Diffusion current, and

 

Im = Migration current.

 

An awkward situation arises when dealing with a dilute solution where it has been observed that the depletion of the electrode layer ultimately leads to an enhancement of the resistance of the solution and thereby affecting subsequently an alteration in the Ohm’s Law potential drop (I × R) in the cell. This ulti-mately gives rise to a doubtful observed potential operative at the electrode. In order to overcome this serious anomaly, it is a normal practice to add an excess of an indifferent electrolyte to the system, such as : 0.1 M KCl, which renders the solution to remain stable at a low and constant resistance, whereas the migration current (Im) of the species under examination almost vanishes i.e., I = Id.

The ion under investigation, whose rate of diffusion at the electrode surface is governed by Fick’s Law represented as under :


where, D = Diffusion coefficient,

 

C = Concentration,

 

t = Time, and

 

x = Distance from the electrode surface.

 

Thus, the potential of the electrode is controlled and monitored by the Nernst Equation as shown below:


Salient Features of Amperometric Methods : 

The various salient features of amperometric titrations are enumerated below :

 

(a) It is less dependent upon the characteristics of the electrode,

 

(b) It is quite independent of the nature and type of the supporting electrolyte,

 

(c) It does not require a constant temperature in the course of a titration but it should not necessarily be fixed accurately,

 

(d) The substance under investigation may not essentially be reactive at the electrode ; whereas either a reactive reagent or a product is just sufficient for a successful amperometric titration, and

 

(e) The amperometric method is inherently more accurate and precise, and therefore, has an edge as compared to the polarographic method.


Chapter: Pharmaceutical Drug Analysis: Amperometric Methods

Amperometric Methods: Theory

Amperometric Methods: Theory
1 Titration Curves 2. Corrections for the Volume Change 3. Advantages of Amperometric Titrations

THEORY

Assuming that the migration current (Im) is virtually eliminated by the addition of a reasonably enough supporting electrolyte then the only cardinal factor which would affect the limiting current would be the rate of diffusion of the electro-active substance from the main body of the solution to the surface of the electrode.

 

Thus, we may have :

 

Diffusion current = Limiting current – Residual current

 

It follows from above that the diffusion current is directly proportional to the concentration of the electro-active substance present in the solution. Now, if a situation is created whereby a portion of the electro-active substance is eliminated by interaction with a specific reagent, the diffusion current shall decrease significantly. It represents the fundamental underlying principle of amperometric method or amperometry. Hence, at an appropriate applied voltage the apparent diffusion current is measured as a function of the volume of the titrating solution added. Now, if a graph is plotted between the ‘current’ against the ‘volume of reagent added’, the end-point will be represented by the point of intersection of two lines indicating the change of current both before and after the equivalence is achieved.

 

 

1 TITRATION CURVES

 

The most commonly obtained various kinds of curves encountered in amperometric methods are illustrated in Fig. 17.1 (a) through (d) ; and each of them shall be discussed briefly as follows :

 

Fig. 17.1 (a) : It represents a titration wherein the analyte reacts at the electrode whereas the reagent does not. In other words, only the substance under titration gives rise to a diffusion current ; whereby the electro-active substance is removed from the solution by means of precipitation with an inactive substance.

Example : The titration of Pb2+ with SO4 2– or C2O42– ions. An appreciably high potential is usually applied to yield a diffusion current for lead. From Fig. : 1(A), one may evidently observe a linear decrease in current because Pb2+ ions are removed from the solution by precipitation. The small curvature just prior to the end-point (or equivalence point) shows the incompleteness of the analytical reaction in this particular region. However, the end-point may be achieved by extrapolation of the linear portions, as shown in the said figure.


Figure 17.1 (b) : It designates typical of an amperometric titration curve wherein the reagent exclu-sively reacts at the microelectrode surface and the analyte does not. In other words, the reagent gives rise to a diffusion current, whereas the solute does not ; it means an electro-active precipitating reagent is being added to an inactive substance.

 

Examples : (a) Titration of Mg2+ with 8-hydroxyquinoline. In this particular instance, a diffusion current for 8-hydroxyquinoline is normally achieved at – 1.6 V Vs Standard Calomel Electrode (SCE), whereas Mg2+ ion is more or less inert at this potential.

(b) Titration of Ba2+ or Pb2+ ions with SO4– ions.

 

Figure 17.1 (c) : It represents an amperometric method wherein the solute as well as the titrating reagent afford diffusion currents ; and give rise to a sharp V-shaped curve. The end-point may be obtained by extrapolation of the lower-end of the V-shaped portion of the curve as depicted in the above Figure.

 

Examples : (a) Titration of Ph2+ ion with Cr2O72– ion. The Figure : 17.1 (c) corresponds to the amperometric titrations of Pb2+ and Cr2O72– ion at an applied potential more than – 1.0 V ; when both these ions afford diffusion currents at this very potential and the end-point is duly signalled corresponding to a minimum in the curve.


Figure 17.1 (d) : In this particular instance the current undergoes a change from cathodic to anodic or vice-versa. Thus, the final end-point of the potentiometric titration is indicated by a zero-current as depicted in Figure 17.1 (d). Since the resulting diffusion coefficient of the reagent is found to be slightly different from the corresponding substance under titration, therefore, the slope of the line just before the end-point actually differs very slightly from that after the end-point. However, in actual practice it is rather convenient to add the reagent unless and until the current attains a zero value.

 

Examples: (a) Titration of I ion with Hg2+ ion (as nitrate),

(b) Titration of Ti3+ ion in an acidified tartaric acid,

[CH(OH)COOH]2, medium with Fe3+  ion.

 

In addition to the above four types of amperometric methods cited, there also exist a plethora of titrations involving neutralization and complex ion formation that have been accomplished successfully, for instance :

 

(i) Amperometric method for the study of precipitation reactions, e.g., salicylaldoxime (or salicylaldehyde oxime), dimethylglyoxime, have been used for such type of studies.

 

(ii) Halides, such as : I, Br and Cl have been titrated at a less negative potential by virtue of the fact that in these titrations the main indicator reaction is the deposition of silver from aquo-silver ions.

 

(iii) Micromolecular solutions of Cd2+ ions against ethylene diaminetetra-acetic acid (EDTA) have been carried out amperometrically.

 

2. CORRECTIONS FOR THE VOLUME CHANGE

 

The corrections for the volume change may be affected by adopting either of the two methods de-scribed below namely :

 

Method I : In order to obtain plots between current (μ A) and volume of reagent (ml) specifically with linear regions both before and after the end-point (or equivalence point), it is absolutely necessary to apply the corrections for the volume change which results from the added titrant. This correction is applied by multiplying the measured corresponding diffusion current (Id) by the following factor :

[V + v] / V

 

where, V = Initial volume of the solution, and

 

v = Volume of the titrating reagent added.

 

Method II : The above correction caused due to the volume change may be eliminated to a great extent by making use of the reagent at a concentration of 10 to 20 times higher than that of the corresponding solute, and subsequently adding the same from a semimicro-burette very carefully. The use of concentrated reagents have the following advantages, namely :

 

(a) Relatively very small amount of dissolved O2 is incorporated into the system, which eliminates completely the prolonged bubbling of inert gas (e.g., N2) through the medium after each addition of the reagent, and

 

(b) Elimination of ‘migration current’ by simple addition of enough supporting electrolyte. If need be, an appropriate maximum suppressor can also be incorporated judiciously.

 

3. ADVANTAGES OF AMPEROMETRIC TITRATIONS

 

A few cardinal advantages of amperometric titrations are described below, namely :

 

·              The amperometric titration may normally be performed very quickly, because the equivalence point (or end-point) is determined graphically. A series of measurements at constant applied volt-age just prior and latter to the end-point are more than enough.

 

·              The titrations can be carried out both satisfactorily and effectively in such situations where the solubility relations offer erroneous and unsatisfactory results given by visual indicator and potentiometric methods. For instance :

 

(a) A reaction product which is hydrolysed significantly e.g., acid base titrations, and

 

(b) A reaction product that is appreciably insoluble e.g., precipitation reaction.

 

It is quite evident that the readings in the vicinity of end-point offer practically no specific value and importance in amperometric titrations. Because the readings are mostly taken in particular zones where there exists either an excess of reagent or of titrant, and which specific points the hydrolysis or solubility is entirely suppressed by the effect of Mass Action. The point of intersection of these lines ultimately gives rise to the desired end-point.

 

·              A good number of amperometric titrations may be performed on considerably dilute solutions (say, 10–4 M) at which neither potentiometric nor visual indicator methods ever can give precise and accurate results, and

 

·              In order to eliminate the migration current (Im) completely either the ‘foreign salts’ already present cause little interference or invariably added so as to serve as the ‘supporting electrolyte’.


Chapter: Pharmaceutical Drug Analysis: Amperometric Methods

Amperometric Methods: Instrumentation

Amperometric Methods: Instrumentation
The amperometric titrations can be accomplished by any one of the three methods, namely : (i) Amperometric titrations with the dropping mercury electrode, (ii) Amperometric titrations with a rotating platinum microelectrode, and (iii) Amperometric titrations with twin-polarized microelectrodes (or Biamperometric Titrations or Dead-stop-end-point method).

INSTRUMENTATION

The amperometric titrations can be accomplished by any one of the three methods, namely :

 

(i) Amperometric titrations with the dropping mercury electrode,

 

(ii) Amperometric titrations with a rotating platinum microelectrode, and

 

(iii) Amperometric titrations with twin-polarized microelectrodes (or Biamperometric Titrations or Dead-stop-end-point method).

 

These three techniques will be discussed in the sections that follow.

 

1. AMPEROMETRIC TITRATIONS WITH THE DROPPING MERCURY ELECTRODE

 

Figure 17.2 (a) and (b) illustrates the schematic diagram of amperometric titrations with the dropping mercury electrode having a titration-cell and an electric circuit respectively.

 

The titration-cell Figure 17.2 (a) essentially comprises of a pyrex 100-ml, four-necked, flat-bottomed flask. A semimicro burette (B) (graduated in 0.01 ml), a 2-way gas-inlet tube (A) to enable N2 to pass either through the solution or simply over its surface, a dropping mercury electrode (C) and an agar-potassium salt-bridge* are duly fitted into the four necks with the help of air-tight rubber stoppers.

 

The electrical circuit, Figure 17.2 (b), consists of two 1.5 V dry cells that provides a voltage applied to the above titration cell. It is duly controlled and monitored by the potential divider (R) and is conveniently measured with the help of a digital voltmeter (V). Finally, the current flowing through the circuit may be read out on the micro-ammeter (M) installed.



The following steps may be carried out in a sequential manner for an amperometric titration, namely :

 

·              A known volume of the solution under investigation is introduced in the titration cell,

 

·              The apparatus is assembled and electrical connections are duly completed with dropping mercury electrode (C) as cathode and saturated calomel half-cell as anode,

 

·              A slow stream of pure analytical grade N2 – gas is bubbled through the solution for 15 minutes to get rid of dissolved O2 completely,

 

·              Applied voltage is adjusted to the desired value, and the initial diffusion current (Id) is noted carefully,

 

·              A known volume of the reagent is introduced from the semimicro burette (B), while N2 is again bubbled through the solution for about 2 minutes to ensure thorough mixing as well as complete elimination of traces of O2 from the added liquid,

 

·              The flow of N2 gas through the solution is stopped, but is continued to be passed over the surface of the solution gently so as to maintain an O2 free inert atmosphere in the reaction vessel,

 

·              The current (µA) and microburette readings are recorded simultaneously, and

 

·              Finally, the said procedure is repeated until sufficient readings have been obtained to allow the equivalence point to be determined as the intersection of the two linear portions of the graph thus achieved.

 

2. AMPEROMETRIC TITRATIONS WITH A ROTATING PLATINUM MICROELECTRODE

 

The rotating platinum microelectrode was first introduced by Laitinen and Kolthoff in 1941. Figure 17.3 (a) depicts a simple rotating platinum microelectrode which is made out from an usual standard ‘mer-cury seal’. A platinum wire (length : 5.0 mm ; diameter : 0.5 mm) protrudes from the lower end wall of a 21 cm long 6 mm glass tubing, which is bent at an angle of 90°. There are holes (H) in the stem of the mercury reservoir for making electrical contact with it. The mercury reservoir is provided with a flange fitted inward to prevent Hg from being thrown out.

 

Figure 17.3 (b) illustrates the electric circuit. The electrical connection is duly done to the electrode by means of a strong amalgamated Cu-wire passing through the glass tubing to the lower end of the Hg covering the sealed-in platinum wire ; the upper end of which passes through a small hole made in the stem of the stirrer and dips well into the Hg present in the Hg seal. Subsequently, a wire from the Hg seal is connected to the source of applied voltage. The glass tubing serves as the stem of the electrode that is rotated at a constant speed of 600 rpm.



 

3. AMPEROMETRIC TITRATIONS WITH TWIN-POLARIZED MICROELECTRODES (BIAMPEROMETRIC TITRATIONS OR DEAD-STOP-END-POINT METHOD)

 

Dead-stop-end-point method was first introduced by Foulk and Bawden* in 1926. Evidently, this particular technique is a modification of the classical amperometric titration. This technique is specifically applicable to only such systems where the phenomenon of oxidation-reduction exists both before as well as after the equivalence point has been duly accomplished.

 

It essentially makes use of two identical, stationary microelectrodes immersed in a well stirred solu-tion of the sample. A small potential ranging between these electrodes ; and the resulting current is measured subsequently as a function of the volume of reagent added. The end-point is distinctly characterized by a sudden current rise from zero or a decrease in the current to zero or a minimum at zero in a V-shaped curve.

 

Though this technique was first used in 1926, but it received its due recognition only around 1950**. F

igure 17.4 represents a simple diagram of an amperometric titration assembly with twin-polarized microelectrodes.


The various components are as follows :

 

A, A = Twin-polarized Platinum microelectrodes,

 

B= Micro-ammeter (µA),

 

C= 500 , 0.5 watt potentiometer,

 

D = 3-Volt dry torch cell or a 2-volt accumulator

 

E = Reaction vessel.

 

The potentiometer is adjusted in such a fashion that there is a distinct potential drop of about 80 to 100 millivolts between the two platinum electrodes.


 

Chapter: Pharmaceutical Drug Analysis: Amperometric Methods

Applications of Amperometric Titrations in Pharmaceutical Substances

Some pharmaceutical substances are assayed by amperometric titrations, namely : procainamide hydrochloride ;

APPLICATIONS OF AMPEROMETRIC TITRATIONS IN PHARMACEUTICAL SUBSTANCES

Some pharmaceutical substances are assayed by amperometric titrations, namely : procainamide hydrochloride ;

 

1. PROCAINAMIDE HYDROCHLORIDE

 

Materials Required : Procainamide hydrochloride : 0.25 g ; 2M hydrochloric acid : 100 ml ; potassium bromide : 3 g ; 0.1 M sodium nitrite Vs (dissolve sodium nitrite in sufficient water to produce 1000 ml) Standardization of 0.1 M Sodium Nitrite Vs : Dissolve 0.3 g of sulphanillic acid in 50 ml of 2M hydrochloric acid, add 3 g of KBr, cool in ice and titrate with 0.1 M sodium nitrite Vs determining the end-point amperometrically. Each ml of 0.1 N sodium nitrite Vs is equivalent to 17.32 mg of C6H7NO3S.

 

Procedure : Dissolve 0.25 g of procainamide hydrochloride in 50 ml of 2 M hydrochloric acid, add 3 g of potassium bromide, cool in ice and titrate slowly with 0.1 M sodium nitrite Vs, stirring constantly and determining the end-point amperometrically. Each ml of 0.1 M sodium nitrite Vs is equivalent of 27.18 mg of C13H21N3O . HCl.

 

2. COGNATE ASSAY

 

Procaine hydrochloride can be assayed exactly in a similar manner by using 0.4 g of the substance.

Each ml of 0.1 M sodium nitrite Vs is equivalent to 27.28 mg of C13H20N2O2, HCl.

 

3. ASSAY OF NICKEL WITH DIMETHYLGLYOXIME

 

Materials Required : 0.001 M Nickel solution ; supporting electrolyte [a mixture of NH4OH (1.0 M) and NH4Cl (0.2 M)] ; gelatin solution (0.2%) : 2 ml ;

 

Procedure : The following steps may be followed in a sequential manner :

 

·              Weigh accurately a sample of Ni-salt to yield a 0.001 M Ni-solution. To 25 ml of this solution placed in a titration cell add an equal volume (25.0 ml) of a supporting electrolyte and 2 ml of gelatin solution,

 

·              The solution must be deoxygenated. Set the applied e.m.f. to 01.85 V Vs SCE (standard-calomel electrode),

 

·              The diffusion current is measured, and

 

·              Finally, titrate with dimethylglyoxime solution (0.02 M) using the standard general method and obtain a V-shaped graph.

 

·              Each ml of dimethylglyoxime solution is equivalent to 0.5869 mg of Nickel.

 

4. ASSAY OF LEAD WITH POTASSIUM DICHROMATE SOLUTION

 

Materials Required :

 

(i) Buffered supporting electrolyte : Dissolve 10 g of KNO3 and 8.2 g of sodium acetate in 500 ml of DW. Add glacial acetic acid carefully until a pH of 4.2 is achieved (pH Meter) (approximately 10 ml of the acid will be required),

 

(ii) Standard 0.01 M K2Cr2O7 Solution : Weigh accurately ‘ANALAR’-grade 1.47 g K2Cr2O7 into a 500-ml volumetric flask. Dissolve in DW and make up the volume upto the mark, and

 

(iii) 0.1% w/v Gelatin Solution :      Dissolve 0.1 g gelatin in 100 ml of boiling DW.

 

Procedure : The amperometric titration may be carried out in a 100 ml beaker. A saturated KNO3 salt bridge is employed to provide contact between the saturated calomel electrode and the analyte solution. The various steps involved are as follows :

 

1.           Weigh accurately a sample of Pb-salt to give a 0.01 to 0.02 M lead solution,

 

2.           Transfer 10.0 ml aliquot to the titration vessel,

 

3.           Add to it 25 ml of the buffered supporting electrolyte, and 5 ml of the gelatin solution,

 

4.           Determine the current at zero applied potential,

 

5.           Add K2Cr2O7 (0.01 M) solution, in 1 ml increments, and measuring the resulting current after each addition,

 

6.           Continue the addition to at least 5.0 ml beyond the equivalence point,

 

7.           Correct the currents for the volume change, and plot the graph. Determine the end-point and calculate the number of milligrams of Pb in the given sample, and

 

8.           Repeat the titrations at – 1.0 V. It is essential to bubble N2 through the solution for 10–15 minutes before the titration and while addition of reagents are made. However, the flow of N2 must be stopped at the time of measuring the current. Again, correct the currents for dilution, plot the graph, determine the end-point, and report the number of milligrams of Pb present in the given sample.












Comments