43
CHAPTER I 1.1 GENERAL INTRODUCTION In absorptiometric method of analysis, simultaneous determination of the components of a binary mixture of drugs, whereby the two known absorbing substances exert mutual interferences at their individual measuring wavelengths, may be easily carried out by utilizing the theoretical principles inherent in the absorbance ratio technique. Determination of both absolute and relative concentration of individual component in such mixture by this technique is decidedly faster and has certain qualities that warrants its use for analysis of binary mixture. Determination of components in a binary mixture using simultaneous (1) equation technique was first applied by Vierordt almost 120 years ago. The technique was based on the assumption that if the two components do not react or interact in any manner with one another and thus neither affects the light absorbing properties of the other, the total absorbance of the two components in the solution is the sum of the absorbances which the two substances would have exhibited invidually if the substances were in separate solutions under similar conditions and had the same concentrations as in the mixture (2,3) . Despite simplicity of the Vierordt’s method, the method is far more sensitive to wavelength errors because some of the absorbance measurement will have to be made on the slopes of the absorbance curves (4) which require greater care specially with wavelength calibration of spectrophotometer visavis solutions of known concentrations are required for establishment of numerical coefficients. The absorbance ratio method was first used in Germany and Hufner (1) was the originator of the technique. The technique is based on the theoretical criterion

CHAPTER I 1.1 GENERAL INTRODUCTION - Shodhgangashodhganga.inflibnet.ac.in/bitstream/10603/25065/5/05...CHAPTER I 1.1 GENERAL INTRODUCTION In absorptiometric method of analysis, simultaneous

  • Upload
    others

  • View
    6

  • Download
    0

Embed Size (px)

Citation preview

  • CHAPTER I

    1.1 GENERAL INTRODUCTION

    In absorptiometric method of analysis, simultaneous determination of the

    components of a binary mixture of drugs, whereby the two known absorbing

    substances exert mutual interferences at their individual measuring wavelengths,

    may be easily carried out by utilizing the theoretical principles inherent in the

    absorbance ratio technique. Determination of both absolute and relative

    concentration of individual component in such mixture by this technique is

    decidedly faster and has certain qualities that warrants its use for analysis of binary

    mixture.

    Determination of components in a binary mixture using simultaneous(1)

    equation technique was first applied by Vierordt almost 120 years ago. The

    technique was based on the assumption that if the two components do not react or

    interact in any manner with one another and thus neither affects the light absorbing

    properties of the other, the total absorbance of the two components in the solution

    is the sum of the absorbances which the two substances would have exhibited

    invidually if the substances were in separate solutions under similar conditions and

    had the same concentrations as in the mixture(2,3). Despite simplicity of the

    Vierordt’s method, the method is far more sensitive to wavelength errors because

    some of the absorbance measurement will have to be made on the slopes of the

    absorbance curves(4) which require greater care specially with wavelength

    calibration of spectrophotometer visavis solutions of known concentrations are

    required for establishment of numerical coefficients.

    The absorbance ratio method was first used in Germany and Hufner(1) was

    the originator of the technique. The technique is based on the theoretical criterion

  • 2

    that the ratio of two absorbance values determined on the same solution at two

    different wavelengths is a constant. Schroeder and his coworkers(5) were perhaps

    the first to tabulate the absorbance ratio values for a relatively large number of

    substances of chemical importance. Kuratani(6) Hirt et.al.(7) and Bonnier et.al.(8)

    pointed out that such ratios can be used to assess the relative concentrations of the

    two components in a binary mixture. Moshe IshShalom et.al.(9) compared the

    method as described by Hirt et.al.(7) with the Vierordt’s method and concluded

    that the former method gave better results. Glenn(10) had developed a modified

    Vierordt equation in terms of absorbance ratios which could be determined from

    solutions of unknown concentrations and may require absorptivity value at the

    max of each component (where dA/d=0).

    Pernarowski et.al.(11) used absorbance ratios to determine both relative and

    absolute concentrations of the components of binary mixtures and derived an

    equation similar to Glenn’s equation(10). However, these author(11) assumed that an

    isoabsorptive point as one of the two wavelengths must be chosen to apply their

    equations. These authors(11) have also applied ‘Q Analysis’ based on the

    relationship between the absorbance ratio values of a binary mixture and the

    relative concentration of such a mixture. Relative analysis of binary mixture by

    this technique is comparatively faster than the simultaneous equation technique

    but, unlike the later method, results are in terms of fraction of total mixture.

    Cho and Parnarowski(12) used absorbance ratios to determine absolute

    concentration and derived an equation which do not require isoabsorptive point.

    The method is based on the use of two absorbance ratio values, one absorptivity

    value and the difference at two wavelengths between the absorbance values of a

    solution and the values of a reference solution containing one of the two

  • 3

    components in the mixture. However, if the difference in absorbance values at the

    two specified wavelengths is small, the error in the analysis is likely to be higher.

    Drug dissolution study is generally done in acidic media by the use of either

    0.1 N HCl or buffer solution. Direct spectrophotometric assay in application of

    such study is very much useful because of rapidity, accuracy and simplicity. The

    drugs which are acid labile in character, deteriorates quicly in acidic media and the

    spectral behaviour changes with time. If the spectra of such drug at different time

    intervals show an isoabsorptive point, then by measuring absorbance at the

    wavelength of isoabsorptive point, total drug content can be determined

    irrespective of acid degradation, which has valuable utilization in the drug

    dissolution rate study. Utilization of the procedure has been described by Fedynec

    et.al.(13) and the technique have been made official in USP[3] in the dissolution

    study of Aspirin Capsules.

    Literature reveals that applications of the absorbance ratio technique have

    been applied for the simultaneous assay of binary mixtures of Aminophylline

    Phenobarbital(14), AnatazolineNaphazoline(15), TrimethoprimSulphamethoxa

    zole(1618), Sulphathiazole in presence of Sulphadiazine and Sulphamerazine(19),

    SulphacetamideSulphanilamide(20), NifuroximeFurazolidone(21), Primaquine

    Amodiaquine(22), Diloxanide furoate in presence of degradation products(23),

    AnalginParacetamol and AnalginOxyphenbutazone(24), Morphine in presence of

    Pseudomorphine(25), StrychnineBrucine(26,27) and mixture of alkaloids.(28)

    In application of zeroorder simultaneous spectrophotometric methods, the

    presence of spectral interferences and/or spectral overlapping such as many

    originate from batchtobatch differences between the sample and reference

    standard or from the pharmaceutical formulation matrix, would certainly lead to

  • 4

    erroneous results. The high excipientdrug ratio and high sample weight require

    for these formulations result in backgroundirrelevant absorption of a sufficiently

    high intensity to possibly prohibit the application of simple spectrophotometric

    methods. Irrelevant absorption in spectra may originate from diluents, moistening

    agents, binder, lubricants, excipients, adjuvants and preservatives. Although

    simultaneous spectrophotometric methods are applicable by eliminating specific

    interference from degradation products and coformulated drugs, however, these

    methods require special attention in selecting assay parameters and application of

    several predetermined factors. Madsen et.al.(29) has made a linear least squares

    approach to analyse mixtures of drugs in the presence of known background

    absorption by simultaneous spectrophotometry.

    The methods aiming at to reduce or eliminate matrix interference in the

    assay of coformulated drugs include MortonSturbs(30) correction procedure,

    which requires that the irrelevant absorption is linear over the wavelength range of

    the absorption band of the drug; Glenn’s method of orthogonal polynomials for

    equally spaced intervals(3136); pj method(37) compensation spectrophotometry, in

    which the reference solution contains the matrix or sample at the same

    concentration present in the sample solution(38,39) difference spectrophotometry(40)

    derivative spectrophotometry(41,42); derivativedifference spectrophotometry(4345);

    Vidicon Spectrophotometry(46) and trigonometric functions(47).

    Difference spectrophotometry is a recently explored spectrophotometric

    technique applicable to acidic, basic, and amphoteric drug substances that undergo

    reproducible spectral changes due to pH changes or the effect of reagents. Doyle

    and Fazzari(48) stated that assay of mixtures of drugs by difference

    spectrophotometry, where more than one drug undergo spectral shifts, constitutes a

  • 5

    special challenge which can sometimes be met, but the method require meticulous

    technique and depend on the fortuitous juxtaposition of an isoabsorptive point of

    one compound with a maximum of another. By contrast the method for single

    component dosage forms are usually simple and rugged.

    Difference spectrophotometry provides an approximation of the ideal

    reference solution by employing an aliquot of the sample solution itself as

    reference, adjusted by change in pH or other parameters but containing both the

    substance being analysed and all extraneous substances at exactly the same

    concentrations as the sample. If the pH or other variation causes an alteration in

    the spectrum of the sample, the instrument records this as a characteristic

    difference spectrum. If other materials present are unaffected by the change in

    conditions, their contribution to the total absorbance of each solution will be

    identical and their effect will be exactly cancelled. It has been proved useful

    particularly in the assay of medicinal substances by eliminating specific

    interference from degradation products and coformulated drugs and also

    nonspecific irrelevant absorption from the formulation matrix. The technique is

    also applied to substances that exhibit a difference in absorbance between the

    equimolar solutions which has been induced by the addition of reagents to one or

    both of the solutions. The difference spectrophotometric assay of these substances

    in samples that also contain other absorbing components may be carried out

    provided the absorbances of the interfering substances remain unaltered by the

    reagents. Thus, pHinduced and reactioninduced simultaneous spectro

    photometric procedures are specific for certain coformulated drugs and

    formulation excipients.

  • 6

    Some of the formulations such as Cinnamic and Benzoic acids(49,50),

    Acetylsalicylic acid SalicylamideAcetaminophenCaffeine(51), Morphine(52,53),

    HydrochlorothiazideReserpine(54), Acetaminophen(55), Chlordiazepoxide and

    Demoxepam(56), AcetaminophenSalicylamide and Codeine phosphate(57),

    Caffeine(58), analgesics(59). Oxyphenbutazone(60), Phenylbutazone(61,62), Tetra

    cycline and Oxytetracycline(63) have been analysed by difference

    spectrophotometry based on pHinduced spectral changes. Reported methods

    based on reactioninduced spectral changes are for Corticosteriods(64,65),

    Phenothiazine drugs(66), and Dipyrone(67).

    Purely spectroscopic evidence for irrelevant absorption always involves

    observed distortion of the contaminated substances absorption curve, of the more

    obvious distortions, max diminishes in the presence of impurities whose

    absorption decreases with increasing wavelength in the region of max, and

    viceversa. On the other hand, irrelevant absorption possessing a constant

    contribution at all wavelengths has no effect on max.

    The accuracy and specificity of U.V. absorption methods may be

    considerably improved by conversion of the normal zeroorder spectrum into a

    higher order derivative spectrum. The improved resolution of overlapping

    absorption bands and the discrimination in the favour of narrow bands against

    broader bands, which are the principal characteristics of derivative

    spectrophotometry, can result in the complete elimination of both nonspecific

    matrix intereference(6870) and specific interference from coformulated

    compounds(71,72). Thus the technique is applicable for determination of single

    component drug dosage forms by eliminating irrelevant absorption due to

    excipients which interfere in direct spectrophotometric analysis and more

  • 7

    successfully for analysis of ingredients of binary component drug formulations for

    which absorbance ratio and difference spectrophotometric techniques are not

    applicable. The technique was introduced(73) as a useful means of resolving two

    overlapping spectral bands of almost coincident wavelengths(74) and eliminating

    marix interference in the assay of many drugs(75,76). The principal advantages of

    derivative measurements are the improvement in the detectability of minor spectral

    features and in quantitative analysis, a potential reduction in error caused by

    overalp of the analyte spectral band by interfering bands of unknown/known

    and/or variable intensity.

    Analytical applications of derivative spectrophotometry has been increasing

    in the past few years(7781) by the introduction of commercial spectrophotometers

    operating in derivative mode, the recently introduced commercial systems that can

    produce a graphic display of the derivative (dA/d) or (d2A/d 2) of the anlog

    signal given by the spectrophotometer provide a different approach to these

    problems. Methods to generate derivative spectra have included direct quantitation

    by digital(82,83) and analog computers(84) or by mechanical modulation of

    wavelength dispersion(8588). The basic difference between the mechanical

    modulation and computational methods is that the former produces a

    representation of derivative of intensity or absorbance with respect to wavelength,

    while the latter produces a derivative of intensity or absorbance with respect to

    time which is assumed to be directly related to wavelength. Hager(85) and Green

    and O’Haver(84) have given lucid description of the manner in which wavelength

    modulation methods coupled with tuned amplifiers generate first and second

    derivatives. Pardue et.al.(82), Cook et.al.(89,90), Milano et.al.(91) & McDowell

    et.al.(97) reported a brief description of vidicon based derivative spectrophotometer

  • 8

    in which derivative spectra are generated directly by the spectrophotometer used in

    conjugation with a phase sensitive lockin amplifier(90).

    Reported pharmaceutical applications of derivative spectroscopy are few

    and for the part have been limited to enhancing the spectral features of a drug to

    facilitate its identification or quantitization. Successful application of this

    technique has been reported for the determination of single component drug

    dosage forms containing excipients which interfere with the spectrometric analysis

    and simultaneous determination of more than one active ingredient in

    multicomponent drug dosage forms, for which both the absorbance ratio and

    difference spectrophotometric techniques are not applicable. Such study is of great

    help in content uniformity analysis and quantitization of the drugs.

    When the irrelevant absorption is not cancelled by difference

    spectrophotometery or derivative spectrophotometry, in such a case difference

    spectroscopy coupled with derivative spectroscopy known as derivative

    difference spectroscopy(D) is sometimes applicable for further compensation of

    irrelevant absorption. This will be the major advantage for the application of

    derivativedifference spectrophotometry over the use of each technique alone. The

    D method unlike the A method can be applied to the assay of drugs after their

    spectra have been changed via change in pH or any other chemical reaction. The

    main criteria for such application is that spectral change induction, which may be

    do not only to pH change but also due to any other chemical reaction such as

    complexation(93), condensation(94), and bromination(95), obviously the limitation(96),

    of the A method should be considered before using the D method.

    Derivative spectrophotometry has been applied for determination of single

    component pharmaceutical dosage forms containing excipients(97101); degradation

  • 9

    products such as Procaine in presence of 4Aminobenzoic acid(102); 1,4Benzo

    diazepins in presence of their acidinduced degradation products(103); Thiamine

    and Pyridoxine in aged pharmaceutical formulations(104); Cephalosporins in the

    presence of their degradation products(105); Acetaminophen and Phenacetin in

    presence of their degradation products(106); determination of respective degradation

    products in presence of intact drugs such as Salicylic acid in Aspirin(107,108) and

    sulfoxide in Chlorpromazine(109); estimation of drugs based on zero

    crossing(110113) technique and simultaneous determination of more than one active

    ingredient in multicomponent drug dosage(114118). Derivativedifference spectro

    photometry has been reported for the drug formulations such as, Corticosteriods,

    Oxytetracycline and Tetracycline(119), Oxazepam or Phenobarbitone and

    Dipyridamole(120).

    Spectral studies of chromophore in the visible region have been extensively

    used in numerous fields and will continue to remain an important field of study as

    it involves very simple instrumentation, resulting nevertheless in sensitive and

    accurate measurements with the advantages of speed and simplicity. The

    limitations of the procedures lies in the chemical reactions characteristic of various

    functional groups capable of giving rise to coloured species. In several instances

    chemical species do not possess suitable chromogenic properties and may be

    converted to an absorbing species or be made to react with an absorbing reagent,

    which forms the basis of their analysis. One of the most common studies in the

    field in recent time is the use of compounds having phenolic functional groups to

    yield spectrophotometrically useful chromogenes through diazocoupling and

    nitrosation(121125) reactions.

  • 10

    Aromatic hydroxyl compounds having free ortho or para position form

    nitroso derivatives in acidic medium. Inamdar and Kadji(126) have reported that

    the stability of the chromogen produced in an acidic medium was increased by the

    addition of alcohol. Chafetz et.al.(127) had claimed that nitro rather than nitroso

    derivative was actually produced. Belal et.al.(121) reported that alkalinization of the

    medium stabilizes the chromophore. Coordination of a transition metal with the

    polydentate ligand (orthonitroso derivative) results in the formation of a stable

    water soluble metal chelate(121). Thus the nitroso derivatives and their metal

    chelates offer a good scope for analysis of phenolic compounds. The compounds

    which are reported in the literature using the technique of nitrosation and chelation

    reaction are Acetaminophen and Salicylamide(123), Oxyphenbutazone,

    Ethinyloestradiol(128) and Amoxicillin(129).

    Chromatography first discovered by Michael Tswett in 1903(130) is

    extensively applied for separation, isolation purification, identification and

    quantitation of the compounds of pharmaceutical interest. The various

    chromatographic techniques are thinlayer chromatorgraphy (TLC), high

    performance thinlayer chromatography (HPTLC), column and paper

    chromatography, ionexchange chromatography, gas chromatography (GC) and

    highperformance liquid chromatography (HPLC). Compounds having similar

    chemical nature often defy separation by other analytical techniques, are identified

    and quantitised by the chromatographic techniques even in microgram quantities.

    All the chromatographic techniques are based on basic separation principles such

    as adsorption, partition and ionexchange.

    TLC was first introduced as a procedure for analytical adsorption

    chromatography by Stahl(131). The technique is now widely been used in official

  • 11

    compendias(13) for limit test of decomposition products and related foreign

    substances. Other wide applications, of TLC are isolation and quantitation of

    multicomponent mixture of drugs.(132,133)

    The real development of adsorption chromatography began in 1931 when

    Kuhn and Lederer(134) introduced the method in the preparative chemistry.

    Martin and Synge(135) introduced partition chromatography using columns of

    silica gel and Consdon et.al.(136) developed paper chromatography. Applications of

    column, paper and ionexchange chromatography are available in the official

    compendias(137143) and literature.

    Gas chromatography (GC) was first applied by Ramsey in 1905 to separate

    gaseous mixtures(144). In 1952, James and Martin(145) introduced gasliquid

    chromatography based on the suggestion of Martin and Synge(146). Gassolid

    chromatography (GSC) and gasliquid chromatography (GLC) are the two

    common technqieus. GC has increasing application in the analysis of drugs and

    their metabolites. The speed, resolution and sensitivity makes this techniques very

    attractive for drug analysis problems dealing with bioavailability, raw material,

    and pharmaceutical formulations. In pharmaceutical analysis, GC has been applied

    for the assay of the raw material, drug substances, quantitation of drugs in

    formulations, and assay of impurities including minor components in the drug

    substance.(147148)

    Highperformance liquid chromatography (HPLC) or often called

    highpressure liquid chromatography is a technique in which separation is

    accomplished by partitioning between a mobile solvent and a stationary column

    packing material of small uniform particle size (10 µm or less). The combined

    advantage of HPLC have led to the very rapid growth in its technology and

  • 12

    popularity since it began to develop a separate discipline in the late 1960s. In

    recent years, due to the development of HPLC instrumentation the technique is

    preferred over gas chromatographic analysis which require derivatization.

    In surveying the literature involving HPLC pharmaceutical analysis, one

    finds that probably more than 90 per cent of the applications involve reverse phase

    liquid chromatography of a bonded phase packing material consisting of silica

    support with an organic moiety bonded to it through a siliconoxygen Silicon

    carbon covalent bonding system prepared with a functionalized chlorosilane. The

    technique of reverse phase chromatography was introduced in 1950 by Howard

    and Martin(149) and involves the use of nonpolar stationary phases and polar

    eluents. Reverse phase chromatography has developed mainly since the

    introduction of chemical bonded stationary phases in 1969 by Halasz and

    Sebestian(150). Now very efficient columns packed with chemically bonded phases

    on microparticles of silica have been prepared. The advantages of reversedphase

    technique are numerous, the most outstanding being the extremely simple

    operating conditions. The ease of sample preparation, speed of analysis,

    specificity, accuracy and precision associated with this method are the main

    advantages. In pharmaceutical analysis HPLC has many useful applications in

    isolation and quantitation of multicomponent mixture of drugs and metabolities.

    The Taxanes docetaxel (Taxotere) and Paclitaxel (Taxol) are used in the

    treatment of cancer. The naturally occurring paclitaxel was first isolated from the

    bark of the pacific yew tree (Taxus brevifolia) in the 1960s and gained commercial

    approval in December 1992. Docetaxel was first synthesized starting from

    10deacetyl baceatin III, a non toxic precursor found in the European yew (Texus

    baccate) in 1986(151). Today these drugs have contributed significantly to the

  • 13

    treatment of a variety of malignancies such as varian, breast and non small cell

    lung cancers, as well as head and neck cancer and some cancers of the digestive

    system.(152)

    Despite the major befits of these products, patients receiving

    chemotherapeutic treatment can experience severe to life threatening side effects

    primarily myelosuppression leading to neutropenia. On the other hand under

    dosage might result in sub optimal treatment of the cancer. In addition to their

    narrow therapeutic range these substances also display highly variable pharmaco

    kinetics. Traditionally the dosing of anticancer agents is calculated on the basis of

    the patient body surface area. It has been suggested that pharmaco kinetically

    guided chemotherapy and dose individualization might lead to a better treatment

    outcome.

    Although this subject is still under discussion, it is clear that further clinical

    studies are necessary to reveal the optimal treatment schedule(153154) for this

    purpose validated analytical methods for the quantification of these compounds in

    plasma are a necessity.

    In plasma paclitaxel and docetaxel highly bound to proteins, with free

    fractions generally lower than 10%. This free active, fraction is better related to

    the pharmacological and/or toxic effect and measuring free fractions could

    therefore be superior to total plasma concentration. The determination of free

    fractions is however complicated and time consuming, limiting its use in clinical

    practice. Oral fluid as a therapeutic drug monitoring matrix offers some interesting

    opportunities.

    This matrix can be viewed as a natural ultrafiltrate of plasma, and oral fluid

    concentrations often correlate to free drug levels in plasma. In addition

    administration of taxaues is based on short infusion duration and patients are often

  • 14

    not hospitalized. Under these conditions, monitoring plasma concentrations, which

    requires blood sampling by medical personnel is laborious and increases stress on

    patients and health care workers. The collection of oral fluid with a collection

    device could be performed by the patients, thus not requiring medical personnel or

    a hospital visit. To investigate the correlation between oral fluid and plasma

    concentrations a method for the quantification of paci taxel and doce taxel in oral

    fluid was developed. In the past, methods have already been developed to monitor

    decetaxel or paclitaxel concentrations in plasma or serum. Earlier methods using

    liquid chromatography with ultraviolet detection suffered from limited sensitivity

    and selectivity, due to their relatively low UVabsorbance and nonselective.

    UVmaximum (227 nm). As a consequence methods based on liquid

    chromatography coupled to mass spectrometry (LCMS) were developed.

    Most published methods report the quantification of either docetaxel(155159)

    or paclitaxel(160164). The simultaneous analysis was also reported(165). Most

    methods used isocratic elution, which minimizes the total run time but does not

    provide a column wash. In addition previous methods did not evaluate the ion

    suppression by the drug formulations vehicle. Recent paper reported(165) ion

    suppression by the drug formulations of docetaxel (Tween 80) and peclitaxel

    (Cremophor EL) due to carry over in subsequent runs with an isocratic LC

    elution(166). To monitor paclitaxel and decetaxel levels in patient samples, a new

    robust method needed to be developed, devoid of matrix effect.

    All these literary calls encouraged the author to study simultaneous

    spectrophotometric determination of some binary or ternary mixture of drugs using

    absorbance ratio, difference spectrophotometric, derivative spectrophotometric,

    and derivativedifference spectrophotometric techniques; visible spectrophoto

  • 15

    metric determination of some phenolic drugs through nitrosation and subsequent

    chelation; application of TLC, paper chromatography and gasliquid

    chromatography for identification and quantitation of active ingradients, added

    impurities or degradation products; simultaneous determination of some binary

    mixture of drugs by column chromatography and HPLC. Literature survey upto

    date revealed that no such study has so far been done on the selected formulations,

    hence it was considered worthwhile to undertake this project.

    1.2 PLANNING OF WORK:

    1.2.1 Category of the Drugs Slected:

    The drug is defined as any substance or product that is used to modify or

    explore physiological systems or pathological states for the benefit of the recipient.

    The drugs are first grouped according to their therapeutic action and then

    subdivided according to the chemical structure of drugs. Category of the drugs

    selected for the work are as enumerated below:

    Antineoplastic:

    (i) Alkylating Drugs

    Procarbazine (Matulane); Dacarbazine (DTIC) Altretamine (Hexalen).

    (ii) Purine Antagonists

    Mercaptopurine (6MP); Fludarabine Phosphate.

    (iii) Pyrimidine Antagonists

    Cytarabine (ARAC); Azacitidine

    (iv) Plant Alkaloids

    Vinblastine (Velban); Vincristine (Oncovin); Etoposide (VP16, Ve

    PeSid); Teniposide (Vumon); Paclitaxel (Taxol); Docetaxel (Taxotere);

  • 16

    Dicloxacillin Sodium; Epirubicin; Epirubicin HCl; Epirubicin Benzoate;

    Mitoxantrone.

    (v) Sulphonamides

    Sulphamethoxy Pyridazine (SMPZ)

    (vi) Diuretic and Antihypertensive

    Frusemide; 4chloro5sulphamoylanthranilic acid (CSAA) (Decom

    position Product of Frusemide)

    It is a potent diuretic, and is also used in the treatment of hypertension.

    (vii) Keratolytic

    4methyl benzoic acid; 4methyl salicylic acid. They have bacteriostatic

    and Fungicidal properties.

    (viii) Antimalarial

    Pyrimethamine; 5(4chlorophenyl)6ethyl pyrimidine2,4diamine.

    (ix) Methyclo thiazide and candesartan cilexetil

    (x) Cycloxacillin; (2S, 5R, 6R)6{[3(2chloro phenyl)5methyloxazole

    4carbonyl}amino}3,3Dimethyl7oxo4thia1azabicyclo [3.2.0]

    heptane2carboxylic acid (C19H18ClN3O5S) and oxacillin; (2S, 5R,

    6R)3,3Dimethyl6[(5methyl3phenyl1,2oxazole4carbonyl)

    amino]7oxo4thia1Azabicyclo [3.2.0] Heptane2carboxylic acid.

    (xi) Moxonidine;4chloroN(Imidazolidin2ylidene)6methoxy2 methyl

    pyrimidine5Amine (C9H12ClN5O) and Amlodipine.

    (xii) Gatifloxacin and propyphenazone.

    1.2.2 Categorisation of work:

    In the present work an attempt has been made to develop quick and reliable

    methodology for control analysis of the selected drugs, available in formulations

  • 17

    as single component, binary or ternary mixtures. Literature survey reveal no such

    work, so far has been done on these formulations using the applied techniques.

    Hence it was considered worth while to undertake this project. The methodology

    applicable to the selected formulations are summarized in table1 and are

    categorized in nine different chapters as enumerated below:

    ChapterII, SectionA: Simultaneous spectrophotometric analysis, using

    absorbance absorbitivity ratio techniques of the following binary mixture of drugs:

    (A) Dicloxacillin Sodium Docetaxel, EpirubicinEpirubicin salt Mitoxantrone;

    Mercaptopurine; Fludarabine phosphate; cytarabine; Azacitidine; Viblastine; Vin

    cristine, Etoposide; Teniposide, Procarbazine; Dacarbazine; Paclitaxel;

    Altretamine; Sulphamethoxy Pyridazine; Frusemide; 4chloro5sulphamoyl

    anthranilic acid; 4methyl benzoic acid; 4methyl salicylic acid; pyrimethamine;

    methyclo, Thiazide, candesartan; cilexetil; cycloxacillin; oxacillin; Moxonidine;

    Amlodipine; Gatigloxacin and propyphenazone and thiabenzazone.

    ChapterII, SectionB: Spectrophotometric estimation of total sulphonamides,

    using isoabsorptive point in a ternary mixture of trisulphadrugs.

    ChapterII, SectionC: Determination of isoabsorptive point, of an intact

    molecule and its decomposition product of an acid labile drug thia benzazone.

    ChapterIII, SectionA: Simultaneous spectrophotometric analysis using

    difference absorbance/difference absorbance ratio technique based on pHinduced

    spectral changes of the following binary component drug formulations:

    Epirubicin/Epirubicin benzoateMitoxantsrone; Mercaptopurine; Fudarabine

    phosphate; viblastine; vincristine; Paceitaxel; Altretamine; Sulphamethoxy

    Pyridazine in presence of pyrimethamine.

  • 18

    ChapterIII, SectionB: Simultaneous spectrophotometric analysis using

    difference absorbance/difference absorbance ratio technique based on pHinduced

    spectral changes, of a ternary mixture of salicylamide, propyphenazone and

    pyrithyldione in presence of caffeine.

    ChapterIII, SectionC: Difference spectrophotometric analysis, based on

    reactioninduced spectral changes of Frusemide in presence of its degradation

    product.

    ChapterIV, SectionA: First derivative spectrophotometric analysis of the

    following drugs. Dicloxacillin sodiumDocetaxel, Mercaptopuyrine; Epirubicin/

    Fudarabinephosphate; DacarbazineEpirubicin; Epirubicinbenzoate; Dacarbazine

    Mitoxantrone.

    ChapterIV, SectionB: Second derivative spectrophotometric analysis of the

    following drugs. ProcarbazineEpirubicin/Epirubicin benzoate; Frusemide in

    presence of its degradation products; 4methyl Benzoic acid and 4methyl

    salicyclic acid.

    ChapterV: Validated spectrophotometric method for simultaneous estimation of

    methylothiazide and candesartan, cilexetil in Tablet Dosage form.

    ChapterVI: Determination of Degradation product for combination containing

    cycloxacillin and oxacillin in capsule dosage form by LCmass spectroscopy.

    ChapterVII: Quantitative determination of paclitaxel first order derivative

    UVspectrophotometry using area under curve.

    ChapterVIII: Simultaneous spectrophotometric estimation of moxonidine and

    Amlodipine in tablet dosage form.

    ChapterIX: Simultaneous spectrophotometric estimation of gatifloxacin and

    proyphenazone in Bulk Drug and in ophthalmic Dosage form.

  • 19

    1.2.3 Further Scope of the work:

    The present investigations embodied in this thesis still point towards a

    further scope for a fertile investigations to develop the fields in pharmaceutical

    analysis with the outcome of new formulations.

    TABLE 1

    THE METHODOLOGY DEVELOPED PERTAINING TO EACH

    FORMULATIONS

    No. Formulation Method developed

    M1 Dicloxacillin sodium

    Docetaxel

    Absorbance Ratio Spectrophotometry;

    Derivative Spectrophotometry

    M2 Epirubicin/Epirubicin

    saltMitoxantrone

    Absorbance Ratio Spectrophotometry;

    Difference Spectrophotometry.

    M3 Mercapto Purine Epirubincin

    Fludarabine phosphate

    Absorbance Ratio Spectrophotometry;

    Difference Spectrophotometry; Derivative

    Spectrophotometry.

    M4 CyatarabineAzacitidine Absorbance Ratio Spectrophotometry.

    M5 VinblastineVincristine Absorbance Ratio Spectrophotometry;

    Difference Spectrophotometry; HPLC.

    M6 EtoposideTeniposide Absorbance Ratio Spectrophotometry.

    M7 ProcarbazineAltretamine;

    Epirubicin and its salt

    Absorbance Ratio Spectrophotometry;

    Derivative Spectrophotometry

    M8 PaclitaxelAltretamine Difference Spectrophotometry; HPLC

    M9 Sulphamethoxy pyridazine Difference Spectrophotometry; Derivative

    Spectrophotometry

    M10 Sulpha Drugs Absorbance Ratio Spectrophotometry.

  • 20

    TABLE 1 Contd… No. Formulation Method developed

    M11 SalicylamidePropyphenazone

    Pyrithyldione

    Difference Spectrophotometry; Derivative

    Difference Spectrophotometry; HPLC.

    M12 Frusemide in presence of

    degradation product.

    Difference spectrophotmetry; Derivative

    spectrophotometry.

    M13 DacarbazineMitoxantrone Derivative Spectrophotometry.

    M14 4Methyl Benzoic Acid4

    methyl Salicyclic Acid

    Derivative Spectrophotometry

    1.3 APPROACH TO WORK:

    In development of the methods detailed under ChapterIIIX, the

    investigations were carried out in the following manner:

    1.3.1 Study of Physical Parameters:

    Some essential physical characteristics of the compounds like solubility,

    pKa values, melting and boiling point, polarity, spectral characteristics, etc. were

    compiled from literatures.

    1.3.2 Choice of Solvents:

    Selection of suitable solvents for stock solutions, standard preparations and

    sample preparations have been done taking into consideration that all the active

    ingredients are easily extractable and stable in the solvent for prolonged time. The

    solvent selected are suitable for repeated dilutions under experimental conditions.

    1.3.3 Selection of Formulatiions:

    In application of simulataneous spectrophotometric analysis, the proportion

    of active ingredients to be determined, is more important for accuracy of the

  • 21

    results. Thus a 5050 drug mixture would yield for more accurate results than a

    94:6 mixtures of the minor component. However, if the absorption intensity of the

    minor component is much greater than that of the major component, then the

    accuracy and precision can be attained to a satisfactory limit. The formulations

    were selected on these considerations.

    1.3.4 Possible Interaction Between the Components:

    In application of the simultaneous spectrophotometric methods it was taken

    into consideration that the components did not form complexes with each other

    under the experimental conditions so that the simple sum of the individual

    absorbances of the components at a particular wavelength are equal to the total

    absorbance of the mixed components of identical concentrations.[34]

    1.3.5 For the Absorbance/Absorptivity Ratio Method:

    Absorbance media: For selection of suitable absorbance media, spectral

    characteristics of all the components present in a particular formulation were

    studied in different acidic, basic, netural (water) and organic media. Finally the

    solvent was selected which gave well separated position of maximum absorption

    peaks or isoabsorptive point with optimum absorption intensity.

    Locatioin of Isoabsorptive point (Isobestic point): An isoabsorptive point is

    defined as the wavelength at which two dissimilar substances have identical

    absorptivity values, the solvent being the same for both substances. For location of

    isoabsorptive point, spectrum of the equiconcentration solutions of the two

    substances were recorded against the solvent blank. The wavelength at which both

    the curve intersected was the isoabsorptive point. Another alternative method

    adopted was that the spectrum of a solution of one substance was recorded relative

    to equiconcentration solution of another substance, the wavelength at which zero

    absorbance was observed represented the isoabsorptive point.

  • 22

    Choice of wavelength: When the maximum of both the components are well

    separated and have optimum absorptivity values, the wavelengths selected are the

    max. of both the components. In applicatioin of isoabsorptive point method and

    ‘Q curve’ analysis these are usually be the wavelengths at which one of the two

    components exhibit maximum absorption and the isoabsorptive point. In selection

    of the wavelengths it was taken into consideration that at the selected wavelengths

    both the components had optimum absorptivity values, the wavelengths of

    measurement were unaffected by irrelevant spectral interferences, and the

    combined spectra of the two components did not have steep slope of the curve at

    the wavelengths of determination.

    ‘Q Curve’ plot: In application of simulataneous spectrophotometry using

    isoabsorptive point as one of the selected wavelength, ‘Q curve’ was plotted to

    determine the applicability and accuracy of the method. Details of such plot are

    discussed under Chapter II (2.2.5).

    1.3.6 For the Difference Absorbance Method:

    Choice of pH/solvent: For application of pHinduced difference

    spectrophotometry for the compounds showing bathochromic or hyperchromic

    shift together with hypochromic or hypsochromic effect, spectral interference of

    each other were eliminated by measuring interalia absorbance of acidic, basic or

    neutral solution of identical concentrations.

    Plot of absorptivity versus different pH media were studied at a particular

    wavelength. For acidic and basic pH media, HCl and NaOH solutions of different

    strengths were used and for neutral pH, glass distilled water (pH~7) was used.

    Selection of solvent were done on the basis of maximum absorbance

    difference (A), isoabsorptive point of zero absorbance and prolonged stability of

  • 23

    the components in the medium. The acidic or basic solutions were so selected, that

    both are at least two pH units removed from the pKa on opposite sides of this

    value and 10% variation of the strength of the acids or bases did not alter the pH

    of the medium.

    The influence of pH changes on unbuffered sample/standard solutions were

    also studied. The final sample solution was found to have a pH~7 in all the cases,

    i.e. equal to the ph of the water used for dilution. Therefore, the use of buffered

    solution was not considered to have any advantage over the use of water. Thus the

    solutions were prepared in water, suitable strength of acid (HCl) and alkali

    (NaOH) because of suitability and simplicity.

    Isoabsorptive Point: Isoabsorptive point of zero difference absorbance (A=O)

    find many uses in the development and selection of optimum analytical conditions.

    The conditions are so selected, that at the wavelength of maximum A of one

    component, the other components have isoabsorptive point of zeroA so that the

    component is determined directly without interference of the other.

    Choice of wavelengths: The wavelength was selected, preferably at or near the

    maximum A of the components to be determined, at which other component(s)

    had isoabsorptive point of ZeroA. In some cases where isoabsorptive point not

    lie at or near A maximum, the wavelength of maximum A was selected and the

    result was calculated with the vector sum of the A of the other component at that

    wavelength.

    Irrelevant absorption: Graphs of log A versus were plotted for sample

    solution and for the authentic mixture of identical ratio. The graphs were

    completely superimposable, indicating that the irrelevant absorbance was

    unaffected by pHchange and so the irrelevant absorption were totally nullified.

  • 24

    1.3.7. For the Derivative Spectrophotometric Method:

    Zerocrossing deterimination: While developing the derivative

    spectrophotometric methods for binary component mixtures, solvent systems were

    so selected that would have placed the derivativeabsorbance maximum of the

    component to be determined at or near the zerocrossing wavelength of the other

    i.e. at the isoabsoptive point, where derivativeabsorbance is equal to zero. Thus at

    the zerocrossing wavelength the measuringcomponent could be determined by

    measuring absolute value of derivative absorbance at that wavelength. For

    zerocrossing determination, recorded the derivative absorption spectra of various

    concentrations of the components to be determined on the same chart paper using

    identical parameters, and suitable zerocrossing wavelengths at zeroderivative

    absorbance were selected for the measurements.

    Derivative amplitude measurement: The derivative amplitudes were measured

    with respect to the derivative zero i.e. from zerocrossing point at derivative zero

    to the measuring derivative curve, with positive or negative value.

    1.3.8 For the DerivativeDifference Spectrophotometric Method:

    Since in the derivativedifference spectrophotometry, the difference spectra

    (A) are differentiated with respect to wavelength, thus the zerocrossing

    determination and peak amplitude measurement were done as under 1.3.7.

    1.3.9 For the visible spectrophotometric method:

    Spectral characteristics of the chromophore: Absorption spectra of the

    chromophore were recorded in the visible and near UltraViolet range

    (350700nm). The wavelength was selected at which sharp maxima with

    maximum intensity was obtained.

  • 25

    Reagent concentration: To determine appropriate concentration of each reagents,

    a fixed volume of the reagent of different concentrations were added to the

    reaction mixture having fixed volume and concentration of the other reagent and

    the corresponding drug. The colour intensity of the reaction mixtures for each

    concentration after complete reaction under a fixed assay parameters was

    measured. The concentration of the reagent which produced maximum colour

    intensity was taken for analysis.

    Effect of temperature: Variation of colour intensity with temperature was studied

    at room temperature, as well as by heating on waterbath for different time

    intervals. The heating parameters which produced the maximum colour intensity

    within shortest time interval, was used for analysis.

    Order of addition of reagents and colour development time: Order of addition of

    reagents were varied taking a fixed concentration of the reagents and the

    corresponding drug. The colour development time to get the maximum colour

    intensity for each set of the order of addition was observed. The order, which

    produced maximum colour intensity in minimum time interval, was selected for

    assay parameters.

    Stability of colour: Stability of the coloured species was studied by measuring the

    developed colour intensity at different time intervals.

    Effect of solvent: The studies on the influence of other water miscible polar

    solvents such as methanol, ethanol, isopropanol and tbutanol instead of water

    revealed that aqueous medium was the best for maximum colour development.

    Cobalt chelates of nitrosoderivative were extractable in chloroform.

    Calibration curve and optimum concentration range: Calibration curves were

    constructed by measurement of the absorbance developed by known concentration

  • 26

    of the constituents under optimum conditions against reagent blank. Straight lines

    were obtained conforming following of Beer’s law in the concentration under use.

    Sensitivity: Sensitivity of the colour reaction has been expressed as molar

    absorptivity (), which is calculated from the equation

    = bCA

    where, A = absorbance; C = concentration of coloured species

    (mol. 11); b = light path length (cm); is expressed

    as 1 mol1. cm1.

    1.3.10 For the Chromatographic Methods:

    Selection of solvents: Selection of solvent for preparation of standard and sample

    solutions are based on compatibility with the techniques adopted. Selection of

    solvents for mobile phase are based on polarity of the solute and solvents.

    Study of impurities/adultration: The methodology concerned with the detection of

    impurities and adultration were developed by considering possible existence of

    such substances.

    Sensitivity of detection: Minimum detectability limit under each experimental

    condition were studied.

    System suitabiulity test: To ascertion the suitability and effectiveness of the final

    operating system for the HPLC determinations, it was subjected to a suitability test

    prior to use. Specific data were collected from the replicate injections of the

    standard preparations and efficiency, precision, tailing factor, resolution, retention

    time, nature of the calibration curves and recovery were studied.

    1.3.11 Interference Studies:

    In the estimations of therapeutically important drug substances, the effect of

    wide range of excipients, diluents, adjuvants, lubricants, binders, preservatives and

  • 27

    other coformulated potent compounds usually present in dosage forms, were

    studied according to the nature of formulations as follows:

    Excipients: Aerosil 400, Dibasic Calcium Phosphate.

    Adjuvants: Activated Charcoal, Light Kaolin, Heavy Kaolin and Talc.

    Lubricants: Stearic Acid and Magnesium Stearate.

    Diluents: Lactose, Starch and Sucrose.

    Ointment base: Anhydrous Lanolin, Soft Paraffin, Bee’s Wax.

    Moistening agents/binders: Anacia Mucilage, Gelatin, Liquid Glucose.

    Preservatives: Methylparaben, Propylparaben, Phenol, Cresol and Sodium

    Benzoate.

    In the initial interference studies, a fixed concentration of the drug substance

    was determined several times by the optimum procedure in the presence of a

    suitable (1100 fold) molar excess of the foreign compounds under investigation,

    and its effect on the applicability of the method was observed. The foreign

    compound was considered to be not interfering, if at those concentrations it

    consistently produced an error less than 1 per cent.

    1.3.12 Application to Commerical Formulations and Stnadard Mixtures:

    The developed methodology were applied to commercial formulations as

    well as to synthetic standard mixtures to determine validity and applicability of the

    methods.

    1.4. STATISTICAL EVALUATION OF RESULTS:

    1.4.1 Precision:

    The precision or reproducibility of the analytical results were calculated in

    terms of Standard Deviation ‘s’.

    s = V

    NNXX

    1

    /)(22

  • 28

    where, ‘N’ is number of repeated measurements of a quantity

    ‘X’, and ‘V’ is the Variance about the Mean ‘ X ’

    X = (X)/N

    For practical interpretation sometimes it is more convenient to express ‘s’ in

    terms of per cent of the average of the repeated measure ‘ X ’ used in the

    calculation of ‘s’. This is called the per cent Coefficient of Variation (% C.V.) or

    per cent Relative Standard Deviation (% RSD)

    % C.V. or % RSD = 100Xs

    Standard Error is the quantity used to calculate the Limits of Error of a

    Mean. For most of the assays a probability level ‘P’ of 0.95 (P’ = 0.05) is taken,

    meaning that in 19 cases out of 20, the true result will lie within the limits. When

    the standard errors are small, a probability level of 0.99 (P|=0.01) i.e. 99 cases out

    of 100 is often used, Thus,

    Standard error = s/N

    and Limits of error = t s/N

    where ‘t’ is the factor available from Students Tbale.

    1.4.2 Tests of Significance:

    The ttest: The test decides whether the difference between the mean results of a

    sample by two different methods are significant or insignificant. If ‘mA’ and ‘mB’

    are means of two results of ‘NA’ and ‘NB’ values respectively, then in order to

    decide whether the difference (mAmB) is significant

    t = (mAmB)

    22

    )2(

    BABA

    BABAddNN

    NNNN

    where dA2 = sA2 (NA1) and dB2 = sB2 (NB1)

  • 29

    The value is compared with the critical limiting values of ‘t’ for different

    probability levels.

    The Ftest (Variance ratio test): This test is used to compare variance values of

    two results of a sample by two different methods, and compared with the

    theoretical limiting values of ‘F’ for given probability levels.

    F = VA/VB = sA2 /sB2

    where ‘sA2’ and ‘sB2’ are the standard deviations of two results.

    1.4.3 Linear Regression:

    To determine best fitting line or curve, regression analysis is performed by

    application of method of least squares. The equations of the regression, or least

    squares, line is

    Y Y = XX

    b =

    NXXNYXXY

    /

    /22

    where Y & X are the arithmetic mean and ‘b’ is the slope of the line.

    Correlation coefficient: Before carrying out a regression calculation, a test is

    made to determine whether there is a significant linear correlation between the sets

    of figures. This can be done by calculating the correlation coefficient ‘r’.

    r =

    }]/}{/[{

    /2222 NYYNXX

    NYXXY

    1.4.4 Accuracy:

    The accuracy of the recommended procedure was evaluated by comparing

    the results of the proposed method with the well established reported method,

    using either synthetic mixtures or commercial formulations. In absence of well

  • 30

    established methods the recovery results of standard mixtures were evaluated to

    determine accuracy.

    1.4.5 Percent Recovery Studies:

    Recovery studies were performed by adding known quantities of the drug

    substance to the preanalysed formulations, using the proposed procedure. To

    study per cent recovery, fixed amount of the sample was taken and different levels

    of standard solutions were added. Each level were subjected to the desired

    repeated measure and total amount of the drug substance was then determined.

    Recovery (%) =

    100.

    ..22

    xXXN

    YXXYN

    where X = amount of the drug added in mg per g or mg per ml of

    the sample.

    Y = amount of the drug found in mg per g or mg per ml of

    the sample.

    N = total number of observations.

    1.4.6 Adherence to Beer’s Law

    To study linearity of the methods, Beer’s law curves were plotted and the

    concentrations range within which the method followed linearity were determined.

    1.5. INSTRUMENTS AND EQUIPMENTS USED:

    The following instruments and equipments (R&D Division, Central Indian

    Pharmacopoeia Laboratory, Ghaziabad) were used for conducting the

    experimental work and SGSIndia Private Ltd., Gurgoan.

    i) Beckman 24 UV/Vis, double beam spectrophotometer with recorder.

    ii) Hitachi 15020 double beam recording Spectrophotometer

    (microprocessor controlled).

  • 31

    iii) PerkinElmer Gas Chromatograph, Model 8500, with GP100 Graphics

    Printer. (microprocessor controlled).

    iv) Waters Liquid Chromatograph, equipped with Data Module M 730, M45

    and 6000 A solvent delivery system; Series 440 Absorbance Detecter;

    Model U 6 K Universal Injector.

    v) Ultrasonic bath (Brasonic 220).

    vi) Corning pH meter, Model 7.

    vii) Camag TLC Scanner II, with Camag PC Evaluation system (MBC 990)

    and Printer Star NL10.

    viii) Camag Linomat III, TLC applicator.

    ix) Camag Shortware (254 nm) U.V. lamp (Portable).

    x) Mettler DL 40 RC Memo Titrator; with GA 40 Printer.

    xi) Millipore Filtration Unit.

    xii) Remi Centrifuge,Model R8C.

    xiii) Mettler H 54 AR Balance.

    xiv) Hot Air Oven

    xv) Dessicating Cabinet

    xvi) Hemilton Syringe (1 µl; 10 µl; 25 µl).

    1.6 GENERAL NOTICES:

    Reference Standards: The reference standards used for preparation of the standard

    solutions were either C.D.L. reference standards or authentic specimens that have

    been verified with the official compendia. (obtained from Central Indian

    Pharmacopoeia Laboratory, Ghaziabad).

    Commercial formulations: All the usually available marketed samples or received

    from C.I.P.L. Ghaziabad were used for verification of the methodology.

  • 32

    Chemicals and reagents: Unless otherwise specified, all laboratory grade

    chemicals and the reagents as spedcified in I.P. [1] were used.

    Water: Unless otherwise specified singleglass distilled water was used, (pH~7).

    Filtrations: Where it is directed to filter, without further qualification, it is

    employed that the solution was filtered through medium porosity sintered glass

    funnel using suction.

    Centrifugation: Where it is directed to centrifuge, it is implied that the solution

    was centrifuged at 3000 r.p.m. at room temperature.

    Waterbath: The term ‘water bath’ means a bath of boiling water, unless water at

    some specific temperature is indicated.

    Temperature: All the temperature measurements are expressed in Celsius

    (Centigrade) scale.

    Working temperature: Unless otherwise specified all the experiments were carried

    out at 25 5C.

    Warm: Any temperature between 30 and 40C.

    Cool: Any temperature between 8 and 25C.

    1.7 ABBREVIATIONS:

    g = Gram; mg = Milligram; mcg = Microgram; ng = Nanogram;

    l = Litre; ml – Millilitre; µl = Microlitre;

    mol = Grammolecular weight (mole)

    cm = Centimeter; mm = Millimeter;

    psi = Pounds per square inch.

    min = Minutes; hr(s) = hour(s)

    mp = Melting point; bp = Boiling point

    AUFS = Absorbance unit full scale.

  • 33

    RT = Retention time; Rf = Chromatographic retardation factor;

    hRf = Rf 100

    A = Absorbance; A = Difference absorbance

    D = Derivative absorbance; D = Derivative difference absorbance

    D1 = First derivative absorbance;

    D2 = Secondderivative absorbance

    max = Wavelength of maximum absorption

    DMF = Dimethyl formamide

    MeOH = Methanol

    EtOH = Ethanol

  • 34

    CHAPTERI

    REFERENCES

    1. L.Heilmeyer, Spectrophotometry in Medicine, Adam Holger Ltd., London

    1943, P7, 65.

    2. C.N.Reilley and D.T.Sawyer, Experiments for Instrumental Methods,

    McGraw Hill Book Company, New York and London.

    3. L.G.Chatten, Pharmaceutical Chemistry, Vol. II, Marcel Dekker, New York

    and London 1969.

    4. M.Ismail & A.L.Glenn, J.Pharm. Pharmacol, 16 (1964) Suppl. 150 T 155T.

    5. W.A.Schroeder, P.E.Wilcox, K.N.True Blood and A.O.Dekker, Anal.

    Chem. 23 (1951) 1740.

    6. K.Kuratani, J. Chem. Soc. Japan Pure Chem. Sect. 72 (1951) 632.

    7. R.C.Hirt, F.T.King and R.G.Schmitt, Anal. Chem. 26 (1954) 1270.

    8. J.M.Bonnier, and G.De Gaumaris, Bull. Soc. Chim. France 67 (1955).

    9. M.Ish,Shalom, J.D.Fitzpatrik and M.Orchin, J. Chem. Educ. 34 (1957) 496.

    10. A.L.Glenn, J. Pharm. Pharmacol. 12 (1960) 595608.

    11. M.Pernarowski, A.M.Knevel and J.E.Christian, J. Pharm. Sci. 50(11) (1961)

    943953.

    12. M.J.Cho and M.Pernarowski, J.Pharm. Sci. 59(9) (1970) 13331335.

    13. R.C.Fedynec, G.A.Lewis and H.N.E., Stevens, J. Pharm. Pharmacol, 35

    (1983) (Supplement) 104P.

    14. F.Yokoyama and M.Pernarowski, J.Pharm. Sci. 50(11) (1961) 953, 2957.

    15. A.M.Wahbi, H.Abdine, M.A.Korany, and F.A. ElYazb, J.Pharm. Sci.

    67(1) (1978) 140141.

    16. A.Ghanem, M.Meshali, J.Pharm. Pharmacol. 31 (1979) 122123.

  • 35

    17. H.N.Joshi,G.N.Chaudhari, H.P.Tipnis, Indian Drugs 19 (1982) (8) 446450.

    18. M.A.Korany, A.M.Wahbi, M.A.Elsayed and S.Mandour, Anal. Lett; 17

    (1984) 13731389; Throu. Anal. Abst; 47 (5) (1985) E30.

    19. L.J.Dombrowski, R.S.Browing and E.L.Pratt, J.Pharm. Sci. 66 (10(1977)

    14131415.

    20. A.A.Wahbi, H.Abdine M.Korany and F.ELYazbi, J. Assoc. Off. Anal.

    Chem. 62 (1) (1979) 6770.

    21. L.Elsayed, S.M.Hassan, K.M.Kelani and H.M.ElFatatry, J.Assoc. Off.

    Anal. Chem; 63(5) (1980) 992995.

    22. S.M.Hassan, M.E.S.Metwally and A.M.A.Ouf, J. Assoc. Off. Anal. Chem;

    66(6) (1983) 14331435.

    23. N.M.Sanghavi and S.P.Kulkarni, Indian Drugs. 17(7) (1980) 299303.

    24. C.Bagavant and V.Roshani, Indian Drugs 23(6)(1986) 373376.

    25. R.R.A.Pride and E.s.Stern, J. Pharm. Pharmacol. 6 (1954) 590.

    26. S.El Masry & A.A.M. Wahbi, J.Assoc. off. Anal. Chem. 61(1)(1978)6567.

    27. M.Scott, A.Taub and C.Dosi Pianta, J.Am. Pharm. Assoc. Sci. Ed. 45(1956)

    232236.

    28. R.N.Bhattacharya & A.K.Ganguly, J.Pharm. Pharmacol. 4 (1952) 190196.

    29. B.W.Madsen, D.Herbinson, Evans. And J.S.Robertson, J. Pharm. Pharmacol

    26(1974) 629636.

    30. R.A.Mortan and A.L.Sturbs, Analyst. 71 (1946) 348356.

    31. A.L.Glenn, J.Pharm. Pharmacol 15 (Suppl) (1963) 123T130T.

    32. H.Abdine, A.M.Wahbi and M.A.Korany, J. Pharm. Pharmacol 23 (1971)

    444447.

  • 36

    33. H.Abdine, A.M.Wahbi and M.A.Korany, J. Pharm. Pharmacol 24 (1972)

    518524.

    34. M.A.Korany, M.Bedair and F.A.ElYazbi, Analyst 111 (1986) 4144.

    35. A.M.Wahbi and H.Abdine, J. Pharm. Pharmacol. 25 (1973) 6972.

    36. I.Agwu and A.L.Glenn, J.Pharm. Pharmacol 19 (Suppl.) (1967) 765.

    37. M.A.Korany and R.Haller, J. Assoc. Off. Anal. Chem. 65 (1982) 144.

    38. J.H.Jones,G.R.Clark & L.S.Harrow, J.Assoc.Off.Anal. Chem. 34 (1951)135.

    39. C.F.Hiskey, Anal. Chem; 33 (1961) 927.

    40. T.D.Doyle and F.R.Fazzari, J. Pharm. Sci. 63 (12) 1974) 19211926.

    41. T.C.O. Haver and G.L.Green, Anal. Chem. 48 (2) (1976) 312318.

    42. J.Traveset, V.Such, R.Gonzalo and E.Gelpi, J. Pharm. Sci. 69(6) (1980)

    629633.

    43. A.M.Wahbi, M.Barary, H.Mahgoub and A.H.Elsayed; J. Assoc. off. Anal.

    Chem; 68(5) (1985) 10461048.

    44. F.A.ElYazbi, M.A.Korany, O.A.Razak and M.A.Elsayed, J. Assoc. Off.

    Anal. Chem. 69(4) (1986) 614618.

    45. F.A.ElYazbi, M.A.Korany and M.Bedair, J. Pharm. Belg. 40(4) (1985)

    244248; Throu. Anal. Abst. 48 (7) (1986) E29.

    46. A.E.McDowell and H.L.Pardue, J. Pharm. Sci; 67 (1978) 822.

    47. A.M.Wahbi, H.Abdine and M.A. Korany, Pharmazie, 33 (1978) 278.

    48. T.D. Doyle and F.R. Fazzari, J. Pharm. Sci. 63 (1974) (12), 19211926.

    49. M.R.I. Saleh, A.A.M.Habib and N.E.Shaer, J. Assoc. Off. Anal. Chem.

    63(6) (1980) 11951199.

    50. A.A.M. Habib and M.R.I. Slach, J. Associ off. Anal. Chem. 63 (3) (1980)

    11951199.

  • 37

    51. N.Shane and M.Kowblansky, J. Pharm. Sci. 57(7) (1968) 12181223.

    52. A.M.Wahbi and A.M. Farghaly, J. Pharm. Pharmacol. 22 (1970) 848850.

    53. R.V.D. Ronadia, A.L.Bondani & J.D. Coussio, J. Pharm. Sci. 62 (1973)502.

    54. H.Abdine, A.M.Elsayed and Y.M.Elsayed, Analyst 103 (1978).

    55. M.A.H. Elsayed, S.F.Belal, A.F.M. Elwality and H.Abdine, J. Assoc. Off.

    Anal. Chem. 62(3) (1979) 549551.

    56. A.G.Davidson, J. Pharm. Sci; 73 (1) (1984) 5558.

    57. M.A.H.Elsayed, S.F.Belal, A.F.M.Elwality and H.Abdine, Analyst 104

    (1979) 620625.

    58. W.P.Ferren and N.A.Shane, J.Assoc. off. Anal. Chem. 57 (1968) 573.

    59. N.A.Shane and J.I.Routh, Anal. Chem. 39 (1967) 414.

    60. S.M.Amer, S.M.Hassan and M.F.E.Tarro Anal. Lett. Part (B) 13 (18) (1980)

    16251634 Throu Anal. Abst. 41 (3) (1981) E44.

    61. V.G.Belikov, E.N.Vergeichik, S.Mutsueva, Kh 33 (2), (1984) 4446;

    Throu; Anal. Abstr; 47(2) (1985) E31.

    62. Z.Berakova, M.Bachrata, M.Blesova and L.Knazko, Farm. Obz; 49 (1980)

    157167; Through; Anal. Abst; 39(5) (1980) E40.

    63. A.A.M. Wahbi M.B.H. Mahgoub, M.A.H. Elsayed, J. Assoc. Off. Anal.

    Chem. 68 (5) (1985) 10451050.

    64. S.Gorog, J. Pharm. Sci; 57 (1968) 1737.

    65. L.Chafetz, D.C.Tsilifonis, and J.M.Riedt, J. Pharm. Sci; 61 (1972) 148.

    66. A.G.Davidson, J. Pharm. Pharmacol; 28 (1976) 795800.

    67. M.A.H. Elsayed, H.Abdine and M.E.A. Hamid, Analyst. 104 (1979)

    568572.

  • 38

    68. J.Traneset, V.Such, R.Gonzalo and E.Gelpi, J.Pharm. Sci; 69(6) (1980)

    629633.

    69. A.F.Fell, Proc. Anal. Div. Chem. Soc; 15 (1978) 260.

    70. T.C.O.Haver, Anal. Chem; 51 (1979) 91A94A.

    71. A.F.Fell and G.Smith, Anal. Proc. 19 (1982) 28.

    72. A.G.Davidson, H.Elaheik, Analyst 107 (1982) 879.

    73. A.T.Giese and C.S.French, Appl. Spectrosc 9 (1955) 78.

    74. T.C.O.Haever and G.L.Green, Anal. Chem; 48(2) (1978) 312318.

    75. A.F.Fell, Proc. Anal. Div. Chem. Soc; 15 (1978) 260.

    76. A.M.Wahbi, M.A.Abounassif, and H.M.G.AlKahtani, Analyst. 111 (1986)

    777780.

    77. T.C.O.Haver and G.L.Green, Am. Lab; 7 (1975) 15.

    78. J.W.Strojek, D.Yates and T.Kuwana, Anal. Chem; 47 (1975) 1050.

    79. G.L.Green and T.C.O. Haver, Anal. Chem; 46 (1974) 2191.

    80. R.N.Hager, Anal. Chem. 45 (1973) 1131A.

    81. E.F.McFarren, R.J.Lisaka and J.H.Parker, Anal. Chem. 42 (1970) 358.

    82. H.L.Pardue, A.E.McDowell, D.M.Fast and M.J.Milan, Clin Chem. 21

    (1975) 11921200.

    83. A.Savitzky and M.J.E.Golay, Anal. Chem; 36 (1964) 1627.

    84. G.L.Green, T.C.O. Haver, Anal. Chem; 46 (1974) 2191.

    85. R.N.Hager, Anal. Chem. 45 (1973) 1131A.

    86. T.C.O. Haver and G.L.Green, Anal. Chem; 48(2) (1976), 312318.

    87. W.Snellman, T.C.Rains, K.W.Yee, H.D.Cook and O.Menis, Anal. Chem. 42

    (1970) 399.

    88. R.C.Elser and J.D.Winefordner, Anal. Chem. 44 (1972) 698.

  • 39

    89. T.E.Cook, H.L.Pardue and R.E.Santini, Anal. Chem. 48 (1976) 451.

    90. T.E.Cook, R.E.Santini and H.L.Pardue, Anal. Chem. 49 (1977) 871877.

    91. M.J.Milani, H.L. Pardue, T.E.Cook, R.E. Santini, D.W.Margerum and

    J.M.T.Raycheba, Anal. Chem. 46 (1974) 374.

    92. A.E.McDowell and H.L.Pardue, J. Pharm. Sci; 67 (1978) 822.

    93. N.A.Abdel Salam, M.A.Hadi and Y.A.Hamid, Acta Pharm. Jugosl, 27

    (1977) 8187.

    94. N.A.Abdel Salam, M.A.Hadi and S.M.K. Khafagy, Pharmazie, 30 (1975)

    402403.

    95. Y.A.Mohamed, N.A.Abdel Salam, N.A.Elsayed and M.A.Abdel Salam, Ind.

    J. Pharm. 28 (1976) 117119.

    96. G.M.Junego and A.L.Glenn, Chemy. Ind. 75 (1956) 813814.

    97. A.M.Wahbi and S.Ebel, Anal. Chim. Acta 70 (1974) 5763.

    98. M.A.Korany, A.M.Wahbi, M.A.Elsayed and S.Mandour, Farmaco Ed. Pr.

    39 (1984) 243252; Throu; Chem. Abst. 101 (1984) 116795w.

    99. A.GF.Davidson and S.M.Hassan, J.Pharm. Sci. 73(3) (1984), 413416.

    100. M.A.H.Elsayed, S.Belal, A.M.ElWality and H.Abdine, J. Pharm. Sci; 68

    (1979) 739741.

    101. M.S.Mahrous, M.M.Abdel, Khalek and M.E.AbdelHamid, J. Assoc. Off.

    Anal. Chem. 68 (1985) (31), 535539.

    102. M.A.Korany, A.M.Wahbi and I.I.Hewala, Arch. Pharm. Chem. Sci. Ed; 12

    (1984) 2630; Throu; Chem. Abstr. 101 (1984) 137106n.

    103. M.Abdel Hamid, M.A.Korany and M.Bedair, Acta Pharm. Jugosl, 34 (1984)

    183190.

    104. V.Such, J.Travest and R.Gonzalo, Anal. Chem. 52 (1980) 412419.

  • 40

    105. F.A.ElYazbi and M.Barary, Anal. Lett. 52 (1980) 412419.

    106. M.A.Korany, M.Bedair, H.Mahgoub and M.A.Elsayed, J.Assoc. Off. Anal.

    Chem. 69(4) (1986) 608611.

    107. K.Kitamura and R.Majima, Anal. Chem. 55 (1983) (1), 5456.

    108. K.Keisuke and M.Ryo, Anal. Chem; 55 (1983) 5456.

    109. A.F.Fell and A.G.Davidson, J. Pharm. Pharmacol. 32 (1980) (Suppl.) 97.

    110. D.Y.Tobias, J.Assoc. Anal. Chem. 66 (6) (1983) 14501454.

    111. M.A.Korany and A.A.ElDin Seif, J.Assoc. off Anal. Chem. 67 (1984)

    138141.

    112. S.Shibata, M.Furukowa and K.Goto, Anal. Chim. Acta 65 (1973) 4958.

    113. S.Shibata, K.Goto and Y.Ishiguro, Anal. Chim. Acta 62 (1972) 305310.

    114. M.A.Korany, A.M.Wahbi, S.Mandour and M.A.Elsayed, Anal. Lett; 18

    (1B) (1985) 2134.

    115. M.E.Mohamed, Anal. Lett; 19 (1986) (1112) 13231339.

    116. F.A.ElYazbi, M.A.Korany and M.Bedair, J. Pharm. Belg. 40(4) (1985)

    244248; Throu; Anal. Abstr. 48(7) (1986), E29.

    117. F.A.ElYazbi, M.A.Korany, O.Abdel Rajak and M.A.Elsayed, Anal. Lett;

    18 (1985) (B17); 21272142; Throu, Anal. Abstr. 48(9) (1986) E55.

    118. N.A.ElSebakly, A.A.ElDin Seif, and M.A.Korany, J.Pharm. Belg. 41(4)

    (1986) 222235. Throu. Anal. Abst. 49 (6) (1987) E11.

    119. A.M.Wahbi, M.Barary, H.Mahgoub, and A.H.Elsayed, J. Assoc. Off. Anal.

    Chem. 68(5) (1985) 10461048.

    120. F.A.ElYazbi, M.A.Korany and M.Bedair, J. Pharm. Belg. 40(4) (1985)

    244248 Throu; Anal. Abstr. 48(7) (1986), E29.

  • 41

    121. S.F.Belal, M.ElSayed, A.AbdelHady Elwality and H.Abdine, Analyst;

    104 (1970) 919927.

    122. S.M.Amer, M.M.Ellalitly and M.F.El Tarasse, Pharmazie 37(3) (1982)

    182184; Throu; Anal. Abst. 43(4) (1982) E58.

    123. A.A.El Kheir, S.Belal and E.L.Shanwani, Pharmazie 40 (1985) (1) 62;

    Throu; Anal. Abst. 47(9) (1985) E41.

    124. A.Abou Out, M.I.Walash, S.M.Hassan and S.M.ElSayed, Analyst, 105

    (1980) (1247), 169173.

    125. M.M.Sanghavi, N.G.Jivani and P.D.Mluye, Ind. J. Pharm. Sci; 39 (1977)87.

    126. M.C.Inamdar and N.M.Kadji, Ind. J. Pharm. 31 (1969) 79.

    127. L.Chafetz,R.E.Daly,H.Schriftman & J.J.Lomner,J.Pharm.Sci.60 (1971) 463.

    128. M.M.Amer, M.I.Walash, I.A.Haroun, and F.M.Ashour, J. Pharm. Belg.

    33(5) (1978) 297302; Throu; Anal. Abstr. 38(6) (1980) E23.

    129. M.G.Rana, H.K.Dhyani, M.R.Patel and B.J.Shah, Ind. Drugs 17 (5) (1980)

    142144.

    130. M.T.Swett, Proc.Warsaw, Soc.Nat.Sci; 1903, Biol. Section 14 Minute No.6.

    131. E.Stahl, Pharmazie 11 (1956) 633.

    132. Clarkes, Isolation and Identification Drugs 2nd Ed. The Pharmaceutical

    Press, London (1986).

    133. Kurt Randerath, Thin layer chromatography, 2nd Ed. Academic Press, New

    York (1968).

    134. R.Kuhn and E.Lederer, Berdtsch. Chem. Ges. 64 (1931) 1349.

    135. A.J.P.Martin and R.L.M.Synge, Biochem. J. 35 (1941) 1358.

    136. R.Consden, A.H.Gordon and A.J.P. Martin, Biochem. J. 38 (1944) 224.

    137. Pharmacopoeia of India, 3rd Ed. Controller of Publications, Delhi (1985).

  • 42

    138. British Pharmacopoeia, University Press, Cambridge 1980.

    139. The United States Pharmacopoeia/National Formulary, United States

    Pharmacopoeial Convention, Rock Ville 1985.

    140. Strain, Chromatographjic Adsorption Analysis, Inter Science Publishers,

    Inc. New York (1945).

    141. Ion exchange chromatography, F.Harold Walton, Dowden, Hutchinson &

    Ross Inc. Pennsylvania.

    142. Chromatographic and Electrophoretic Techniques, Ivor Smith Vol.I Inter

    Science Publishers, Inc. U.S.A. (1960).

    143. Official Methods of Analysis of A.O.A.C. 13th Ed. Association of Official

    Analytical Chemists, Benzamin Franklin Station, Washington, 1980 p704.

    144. W.Ramsey, Proc. Roy. Soc. A 76 (1905) 111.

    145. A.J.James and A.J.P.Martin, Analyst, 77 (1952) 915.

    146. A.J.P.Martin and R.L.M.Synge, Biochem. J; 35 (1941) 1358.

    147. S.Roger Schirmer, Modern methods of Pharmaceutical Analysis, Vol. III

    CRC Press, Inc. Florida.

    148. Gas Chromatography, A.B.Little Wood; The Institute of Petroleum, London

    (1966).

    149. G.A.Haward and A.J.P. Martin, Biochem. J. 56 (1950) 539.

    150. I.Halasz and I.Sebastian, Angew Chem. 8 (1969) 453.

    151. J.E.Cortes, R.Pazdur, J. Clin Oncol 13 (1995) 26432655.

    152. J.Dubois, D.Guenard, F.Gueritte, Expert Opin Ther. Pat. 13 (2003)

    18091823.

    153. C.H.Smorenburg, A.Sparreboom, M.Bontenbal, G.Stoter, K.Nooter, J.

    Verweij, J. Clin Oncol 21 (2003) 197202.

  • 43

    154. M.J.Egorin, J. Clin Oncol 21 (2003) 182183.

    155. S.Mao, X.Shuai, F.Unger, M.Simona, D.Bu T.Kissel, Int. J. Pharm. 281

    (2004) 4554.

    156. P.Dharmani, W.Singh Chauhan, U.Palit, Eur. J. Pharmacology, 519 (2005)

    27784.

    157. C.Immanuel, A.K.Kumar, J.Chromatogr. (B). Biomed. Sci. Appl. 760

    (2001) 9195.

    158. M.B.Patel, K.M.Patel, O.S.Patel, B.N.Suhagia, A.M.Prajapati, J.Liq.

    Chromatgr. Rel. Technol. 30(2007) 245971.

    159. J.Turek, M.Reinbers, D.Dreyer, T.K.Kiffmeyer, K.G.Schmidt, H.M.Kwens,

    J. Chromatogr. (B) 83 (2006) 7280.

    160. S.Sharma, A.Bhandari, U.R.Choudhary, H.Rayapurohit and P.Khandelwal,

    Ind. J. Pharm. Sci. 73(1) (2011) 8488.

    161. P.Pattanayak, U.P.Chaudhary, P.Sharma, P.Mahapatr, D.K.Setty, Res. J.

    Pharm. And Tech. 2 (2009) 2913.

    162. S.Mohammed, G.Mohsin, S.Anwar, J.Chromatogr. Sci. 40 (2002) 42931.

    163. R.Yanamandra, C.S.Vadla, U.M.Puppala, B.Patro, Y.L.N.Murty and

    B.R.Parimi, Jour. of Pharm. Sci.; 74(2) (2012) 116121.

    164. S.R.Desai, U.V.Laddi, R.S.Bennur and S.C.Bennur, Ind. Jour. of Pharm.

    Sciences 73(4) (2011) 478482.

    165. H.B.Lea,T.F.Perata,M.L.Svobodale, J.Chromatogr.(A) 1055 (2004) 22328.

    166. P.R.Kalra,R.Sharma,S.C.Chaturvedi,Ind.Jour.of Pharm.Sci.70(2008) 4914.