01
01
MASTER OF PHARMACY
IN
PHARMACEUTICAL CHEMISTRY
SUBMITTED TO
SAVITRIBAI PHULEPUNE UNIVERSITY
Research Guide
Dr. P.D.Pawar
M. Pharm; Ph.D.
Research Student
Ms. Ravankole Pratiksha Rajkumar
B. Pharmacy
MASTER OF PHARMACY
IN
PHARMACEUTICAL CHEMISTRY
SUBMITTED TO
SAVITRIBAI PHULEPUNE UNIVERSITY
Research Co-Guide
Dr.P.D.Pawar
M. Pharm, PhD
Research Student
Ms. Ravankole Pratiksha Rajkumar
B. Pharmacy
Principal,
CERTIFICATE
This is to certify that the research work embodied in this thesis entitled
“Stability-indicating High performance liquid chromatography
Place: Principal
Seal of College
Dr. P. D. Pawar
Assistant Professor,
CERTIFICATE
This is to certify that the research work embodied in this thesis entitled
“Stability-indicating High performance liquid chromatography
Pune University. The research work has been carried out under my
supervision and to my satisfaction.
Seal of College
ACKNOWLEDGEMENT
I would like to express my sincere appreciation, deep sense of gratitude and cordial
thanks to my thesis Supervisor, Prof.Santosh A. Waghmare&
Prof.S.R.GhodakeAssistant Professor, Department of Pharmacy, for his valuable
instructions, continuous guidance, Encouraging, constructive criticisms and thoughtful
advice related to the experiments and theoretical
studiesduringpursuingthisresearchandpreparationofthis report.
5
ABBREVATIONS
i.e: That is
V: Volt
etc: Extra
%: Percentage
Std: Standard
Fig: Figure
No.: Number
°C: Degree Celsius
min: Minute
Sr.
Name of Chapter Page No.
No.
1. Introduction 1-55
9. References
APPENDIX
I. Presentation and Publications
II. Errata
LIST OF TABLES
1.1 UV Spectrophotometer 3
1. INTRODUCTION
CHAPTER I INTRODUCATION
The newly developed methods find their importance in various fields such as:
Research institutions
Quality control departments
Approved testing laboratories
Biopharmaceutics & bioequivalence studies
The combination dosage forms are complex in nature because of different additives and
individual drug components present in it, in the process of estimation it is important to
confirm that one component does not interfere with the estimation of the other. Hence the
analytical approach should be different.
1.2.4Mass Analysis:
CHAPTER I INTRODUCATION
Mass spectrometry measures the interaction of charged materials and electric and magnetic
fields.
1.2.5Thermal Analysis:
Calorimetric and thermo gravimetric analysis measure the interaction of material and heat. In
order to utilize the techniques available currently, complex material mixtures must be
separated into simpler samples for individual analysis.
Instrumentation of HPLC:
3) Column:
Straight lengths of stainless steel tubing make excellent columns. These come invarious
diameter and lengths depending upon the particular application. Usually the internal diameter
is 3.9 or 4.6mm. A well packed 4.6 mm column of 5 µm diameter (d p) should give a plate
count on the order of 60,000 to 90,000 plates/meter at flow rates of 1 ml/min.
A small 3 to 10cm guard column or precolumn is placed between the injector and the
analytical column.
4) Detectors:
The function of the detector in HPLC is to monitor the mobile phase as it merges from the
column. Detectors are usually of two types:
It compares overall changes in a physical property of the mobile phase with and without an
eluting solute. e.g. refractive index, dielectric constant or density.
Modes of HPLC:
1) Normal phase chromatography: In normal phase mode, the nature of stationary phase
is polar and the mobile phase is non-polar. In this technique, non-polar compounds travel
faster and are eluted first because of the lower affinity between the non-polar compounds
and stationary phase. Polar compounds are retained for longer time and take more time to
elute because if their higher affinity with the stationary phase. Normal phase mode of
separation is, therefore, not generally used pharmaceutical applications because most of the
drug molecules are polar in nature and hence take longer time toelute.
2) Reversed phase chromatography: Reversed phase mode is the most popular mode for
analytical and preparative separations of compounds of interest inchemical,biological,
pharmaceutical, food and biomedical sciences. In this mode, the stationary phase is non-
polar hydrophobic packing with octyl and octadecyl functional group bonded to silica gel
and the mobile phase is a polar solvent, often a partially or fully aqueous mobile phase.
Polar substances prefer the mobile phase and elute first. As the hydrophobic character of
the solutes increases, retention increases. Generally, the lower the polarity of the mobile
phase, the higher is its eluent strength. The elution order of the classes of compounds is
reversed (thus the name reverse-phase chromatography).
separation goal. Then estimate the best separation condition from trial runs. After
optimizing the separation condition, validate the method for release to routine laboratory.
1.4.1 The Mobile Phase: In reverse-phase chromatography, the mobile phase is more polar
than the stationary phase. Mobile phase in these systems is usually mixtures of two or more
individual solvents with or without additives or organic solvent modifiers. The usual
approach is to choose what appears to be the most appropriate column, and then to design a
mobile phase that will optimize the retention and selectivity of the system. Separations in
these systems are considered to be due to different degrees of hydrophobicity of the solutes.
The simple alteration of composition of the mobile phase or of the flow rate allows the rate
of the elution of the solutes to be adjusted to an optimum value and permits the separation
of a wide range of the chemical types. First isocratic run followed by gradient run
ispreferred.
1.4.2 The Detector: The next consideration should be the choice of detector. UV-visible
detectors are the most popular as they can detect a broad range of compounds and have a
fair degree of selectivity for someanalyses.
1.4.3 The Column Length: Many chromatographers make the mistake of simply using
what is available. Often this is a 250 cm × 4.6 mm C18 column. These columns are ableto
resolve a wide variety of compounds. While many reverse phase separations can be carried
out on such column. Method development can be streamlined by starting with shorter
columns; 150, 100 or even 50 cmlong.
1.4.4 The Stationary Phase: Selecting an appropriate stationary phase can also help to
improve the efficiency of method development. For example, a C8 phase (reversed phase)
can provide a further time saving over a C18, as it does not retain analytes as strongly as the
C18 phase. For normal phase applications, cyano (nitrile) phases are mostversatile.
1.4.5 The Internal Diameter: By selecting a shorter column with an appropriate phase, run
CHAPTER I INTRODUCATION
times can be minimized so that an elution order and an optimum mobile phase can be
quicklydetermined.
1.4.6 Gradient Programming: The fastest and easiest way to develop a method is to use a
mobile phase gradient. Always start with a weak solvent strength and move to a higher
solvent strength. To begin, use a very fast gradient (e.g.10 minutes) and then
modifythestartingand finishing mobilephasestoachieveasuitableseparation.
1.4.7 Retention: Analytes may be too strongly retained (producing long run times). If this
occurs, the solvent strength should be increased. In reverse phase analysis this means a
higher % of organic solvent in the mobile phase.
1.4.8 Poor Separation: Analytes often co-elute with each other or impurities. To overcome
this, the analysis should be run at both higher and lower solvent strengths so the best
separation conditions may bedetermined.
1.4.9 Peak Shape: This is often a problem, especially for basic compounds analyzed by
reversed phase HPLC. To minimize any potential problems always use a high purity silica
phase such as Wakosil II. These modern phases are very highly deactivated so secondary
interactions with the support are minimal. To maximize the reproducibility of a method, it is
best to use a column heater to control the temperature of the separation. A temperature of 35
– 40°C isrecommended.
1.4.10 Buffer selection: In reverse phase HPLC, the retention of analytes is related to their
hydrophobicity. The more hydrophobic the analyte, the longer it is retained. When an
analyte is ionized, it becomes less hydrophobic and, therefore, its retention decreases. When
separating mixtures containing acid and/or bases by reversed phase HPLC, it is necessary to
control the pH of mobile phase using appropriate buffer in order to achieve reproducible
results. Buffers play an additional role in the reproducibility of a separation. The buffer
salts reduce peak tailing for basic compounds by effectively masking silanols.
They also reduce potential ion-exchange interactions with unprotonated silanols (Figure 8).
CHAPTER I INTRODUCATION
1.4.11 Selection of pH: The pH range most often used for reversed-phase HPLC is 1 - 8
and can be divided into low pH (1 - 4) and intermediate pH (4 - 8) ranges. Each range has a
number of advantages. Low pH has the advantage of creating an environment in which peak
tailing is minimized and method ruggedness is, maximized. For this reason, operating at
low pH isrecommended.
At a mobile phase pH greater than 7, dissolution of silica can severely shorten the lifetime
of columns packed with silica-based stationary phases.
The pKa value (acid dissociation [ionization] constant) for a compound is the pH at which
equal concentrations of the acidic and basic forms of the molecule are present in aqueous
solutions. Analytes may sometimes appear as broad or tailing peaks when the mobile phase
pH is at, or near, their pKa values. A more rugged mobile phase pH will be at least 1 pH
unit different from the analyte pKa. This shifts the equilibrium so that 99% of the sample
will be in one form. The result is consistent chromatography.
Where, VA = Elution time of A and V0 = Elution volume of non retained compound. (Void
volume)
At a constant flow rate, retention times t A – t0 can be used instead of retention volumes.
Retention data is sometimes expressed, relative to a known internal standard (B). The ratio of
CHAPTER I INTRODUCATION
retention times tA/ tB can be used, but the ratios of adjusted retention times (t A- t0 / tB-t0) is
better when data need to be transferred between different chromatographs.
II) Resolution:
Resolution is a measure of the extent of separation of two components and the baseline
separation achieved. Resolution is equal to the distance between the peaks centers divided by
the average band width. The ideal value of R s for baseline separation is 1.5. It is calculated by
using the formula,
Where, t1 and t2 are the retention times of the first and second adjacent bandwidth.
K’ = V1 –V0/ V0
in the stationary phase to the number of molecules of the same in the mobile phase. The ideal
value of K’ ranges from 2-10. Capacity factor can be determined by testing the formula,
Where, V1 = retention volume at the apex of the system and V0 = Void volume of the system.
When conditions are changed so that K’ is made larger, resolution usually improves.
Typically an increase in percentage of the organic phase by 10 % by volume will decrease K’
of the bands by a factor of 2-3.
The selectivity (α) is a measure of relative retention of two components in a mixture. The
ideal value of selectivity is 2. It calculated by using the formula,
α = V2 –V0 / V1 -V0
Where, V2 &V1 = retention volume at the apex of the system and V 0 = Void volume of the
column.
CHAPTER I INTRODUCATION
N = 16 (TR) 2 / W
Ax =b/a The
equation for determining peak asymmetry is, Where, b = the distance between the
perpendicular connecting the baseline to peak maximum and the latest eluting portion of the
curve and a = the distance between the perpendicular connecting the baseline to peak
maximum and the earliest eluting portion of the curve.
determine limits of allowed variability for the conditions needed to run the method. [10]
1.5.1 Type of analytical procedures to be validated: Validation of analytical procedures
is directed to the four most common types of analyticalprocedures.
Identificationtest.
Quantitative test for impuritiescontent.
Limit test for the control ofimpurities.
Quantitative test of the active moiety in samples of drug substance on drug product
CHAPTER I INTRODUCATION
Safety precautions.
Critical parameters taken from robustness testing.
Listing of equipment and its functional and performance requirements, e.g., cell
dimensions baseline noise and column temperature range. For complex equipment, a
picture or schematic diagram may be useful.
Detailed conditions on how the experiments were conducted, including sample
preparation. The report must be detailed enough to ensure that it can be reproduced by
a competent technician with comparable equipment.
Statistical procedures and representative calculations.
Procedures for QC in routine analyses, e.g. system suitability tests.
Representative plots, e.g., chromatograms, spectra and calibration curves.
Method acceptance limits performance data. The expected uncertainty of
measurement results.
Criteria for revalidation.
The person(s) who developed and validated the method.
References (if any).
Summary and conclusions.
A direct consequence and most significant outcome from any method validation exercise is
“the development of meaningful specifications can be predicted upon the use of validated
analytical procedures that can assess changes in a drug substance or drug product during its
lifetime.”
Impurities (Quantitation)
Accuracy should be assessed on samples (drug substance/drug product) spiked with known
amounts of impurities. In cases where it is impossible to obtain samples of certain impurities
and/or degradation products, it is considered acceptable to compare results obtained by an
independent procedure.
Recommended Data
Accuracy should be assessed using a minimum of 9 determinations over a minimum of 3
concentration levels covering the specified range (e.g., 3 concentrations of 3 replicates each
of the total analytical procedure). Accuracy should be reported as percent recovery by the
assay of known added amount of analyte in the sample or as difference between the mean and
the accepted true value together with the confidence intervals.
b) Precision:
The precision of the analytical method is related to the degree of the agreement among
individual test results when the procedure or method is applied repeatedly to multiple
sampling of homogenous sample. It is expressed as standard deviation or as coefficient of
variation or relative standard deviation and it is possible to calculate statistically valid
estimates of standard deviations or relative standard deviation(S).
i) Repeatability
CHAPTER I INTRODUCATION
The result of the methods operating over a short time interval under the same conditions
(within a laboratory over a short period of time using the same analyst with the same
equipment). Repeatability is also termed as intra-assay precision.
a) A minimum of 9 determinations covering the specified range for the procedure (e.g. 3
concentrations/3 replicate each).
b) A minimum of 6 determinations at 100 % of the test concentration.
ii) Intermediate precision
Intermediate precision expresses within laboratory variation (as on different days, or with
different analyst, or equipment within the same laboratory).
iii) Reproducibility
It expresses the precision between laboratories and is often determined in collaborative
studies or method transfer experiments.
c) Linearity:
The linearity of an analytical procedure is its ability to obtain test results that are directly
proportional to the concentration of the analyte in the sample within a given range. Linearity
should be evaluated by visual inspections of a plot of signals as a function of analyte
concentration or content. Data from regression line itself may helpful to provide
mathematical estimates of the degree of linearity. For establishing the linearity, minimum of
5 concentrations are recommended.
d) Range:
The range of an analytical method is the interval between the upper and lower concentration
levels of analyte for which it has been demonstrated that the analytical range has a suitable
level of precision, accuracy and linearity.
e) Specificity:
It is the ability to asses unequivocally the analyte in the presence of the components which
may be expected to be present in the sample matrix. It is obtained by analysis of sample
containing added impurities, degradation products, related chemical compounds or placebo
ingredients when compared to test results from samples without added substance. The
CHAPTER I
INTRODUCATION
impurity should not interfere in detection. Specificity has been divided into two separate
categories by ICH:
Identification test
Assay and impurity tests
h) Robustness:
It is the measure of its capacity to remain unaffected by small, but deliberate variation in
method parameters and provides indication of its reliability during its normal usage.
i) Ruggedness:
This is reproducibility of the results when the method is performed under actual use
conditions. This includes different analyst, laboratories, column, instruments, sources of
reagents, chemicals, solvents and so on.
j) Stability:
In order to make reliable measurements, the intensity of solution must remain constant for
long-times.
In2002,inordertoestablishamoresystematicandriskbasedapproachtothedevelopmentofpharmac
euticalproducts,usingtheprogressesinscienceandtechnology,FoodandDrugAdministration(FDA
)announcedthe“cGMPforthe21stCentury:ARiskbasedApproach”Initiative.Thisinitiative,focus
edonQbD,andthepublicationoftheProcessAnalyticalTechnology(PAT)Guidancein2004bytheFD
Acontributeddecisivelyforthemodernizationofthepharmaceuticalindustryandchallengedthemto
lookbeyondthetraditionalapproachofQualitybyTesting(QbT).Inadditiontothesenewideas,threei
mportantguidancedocumentswerepublished
aspartofInternationalConferenceonHarmonization(ICH)guidelines:Q8PharmaceuticalDevelop
mentandQ9 Quality
RiskManagement,in2005,andICHQ10PharmaceuticalQualitySystem,in2008.Theseguidancedo
cumentsimplementedtogether,inaholisticmanner,providesaneffectivesystemthatemphasizesahar
monizedscienceandrisk-
basedapproachtoproductdevelopment,assuringanimprovinginQualityinpharmaceuticalindustry.
InICHQ8guidance,theconceptofQbDwasmentioned,statingthat“qualitycannotbetestedintoprod
ucts,i.e.,qualityshouldbebuiltinbydesign”.In2009,theICHQ8guidancewasreviewed,clarifyingk
eyconceptsoftheoriginalguidance.Additionally,theprinciplesofQbDweredescribesandQbDdefi
ningas“asystematicapproachtodevelopmentthatbeginswithpredefinedobjectivesandemphasizes
productandprocessunderstanding and process control,based on sound science and quality risk
management”.
Infact,QbDisacomprehensiveapproachtargetingallphasesofdrugdiscovery,manufacture,anddeli
very.The aimistoimprovethequality and reduce thecostsofmedicinesfortheconsumer.
Thismaybeaninteractivesystematicapproachandthusthe circular designasshown
inFigure9.Thiscircle ofQbDcan bedividedintotwogeneral
areas,productknowledgeandprocessunderstanding.
Thesetwoareasmeetinthedesignspaceandtheinteractionofproductknowledgeandprocessundersta
ndingallowsforcontinuousimprovement.
Figure 1.5:QualitybyDesignconcept
QbDbeginsbydefiningthedesiredproductperformanceandalsobydefiningaproductthatmeetsthos
eperformancerequirements.Thecharacteristicsofthedesiredproductarethebasisfordesigningthem
anufacturingprocess,whichneedstobemonitoredintermsofperformance.Eachofthesestepsinflue
nceseachother,continuingthecycle.Theinnercircleinteractswithmanyotherspecificmeasuresofph
armaceuticalmanufacturing,suchasspecifications,criticalprocessparameters,ensuringtheproduc
tknowledgeandprocessunderstanding.
TheunderlyingprinciplesofQbDareexplainedinthequalityguidelinesofinternational
conferenceonharmonizationi.e.ICH
Q8PharmaceuticalDevelopment,ICHQ9QualityRiskManagement,andICHQ10Pharmaceutical
QualitySystem.Fig. No.10presentstheguidelines that explainQbD.
CHAPTER I INTRODUCATION
Quality
byDesig
n
ICHQ9
QualityRiskM
anagement
ICHQ8 ICHQ10
Pharmaceutica Pharmaceutical
lDevelopment QualitySystem
TheapplicationofQbDpresentsseveraladvantagesandcanbesummarizedas:
Patientsafetyandproductefficacyarefocused
Scientificunderstandingofpharmaceuticalprocessandmethodsisdone
Itinvolvesproductdesignandprocessdevelopment
Sciencebasedriskassessmentiscarried
Criticalqualityattributesareidentifiedandtheireffectonfinalqualityofproductisanalyzed
Itoffersrobustmethodorprocess
BusinessbenefitsarealsodrivingforcetoadoptQbD.
Target measurement
Select Technique
Risk Assessment
CHAPTER I INTRODUCATION
Method Development /
Validation
The approach provides an in-depth knowledge and enables the creation of a chromatographic
database than can be utilized to provide alternative method if required, in future
Development of a robustmethod.
Understand, reduce and control sources of variability.
Applicable throughout the life cycle of the method.
Regulatory flexibility Movements within “Analytical Design Space”.
b. Independent factors
i) Sample preparation variations
Solvents of stock solution
Solvents of sub-dilutions
deliver the intended performance of product. The information and knowledge gained from
pharmaceutical development studies and manufacturing experience provide scientific
understanding to support the establishment of the specifications, and
manufacturingcontrols.Duringpharmaceuticaldevelopment,QbD
suggeststhatitshouldincludethefollowingelements:
Definingthequalitytargetproductprofile(QTPP)
Identifyingpotentialcriticalqualityattributes(CQAs)
LinkrawmaterialattributesandprocessparameterstoCQAsandperformriskassessment
Developingadesignspace
Designingandimplementingcontrolstrategy
Continuousimprovement
1) DefiningProductDesignRequirementsandCriticalQualityAttributes
Theproductdesignrequirementsmustbewellunderstoodintheearlydesignphase,andtheycanbefo
undinaQualityTargetProductProfile(QTPP).TheQTPPisderivedfromthedesiredproductinform
ationandithasbeendefinedas“aprospectivesummaryofthequalitycharacteristicsofadrugproductt
hatideallywillbeachievedtoensurethedesiredquality,takingintoaccountsafetyandefficacyofthed
rugproduct”.
Therefore,pharmaceuticalcompaniesconstructatargetproductprofilethatdescribes:
Intendeduseinclinicalsetting,routeofadministration,dosageform,deliverySystems
Dosagestrength(s),Containerclosuresystem
Therapeutic moiety release or delivery and attributes
affecting,Pharmacokineticcharacteristics(e.g.,dissolution,aerodynamicperformance) Drug
product quality criteria likesterility,purity,stability and drug release
asappropriatefordosageformtheintendedformarketing.
TheQTPPguidesscientiststoestablishstrategiesandkeeptheproductdeveloping
effortfocusedandefficient.
Inadditiontodefiningtherequirementstodesigntheproduct,theQTPPwillhelpidentifycriticalqual
ityattributes(CQAs).ICHQ8definesCQAas“aphysical,chemical,biological,ormicrobiologicalp
ropertyorcharacteristicthatshouldbewithinanappropriatelimit,range,ordistributiontoensurethe
desiredproductquality”.CQAsaregenerallylinkedwiththedrugsubstance,excipients,intermediat
es(in-
processmaterials)anddrugproduct.Qualityriskmanagementtools,foundintheICHQ9guideline,
CHAPTER I INTRODUCATION
areoftenusedtoidentifyandprioritizethepotentialCQAs.RelevantCQAscanbeidentifiedbyadyna
micprocessqualityriskmanagementandexperimentationthatevaluatestheextenttowhichtheirvar
iationcanhaveanimpactontheultimatequalityproduct.Theaccumulatedexperience,theknowledg
e obtained from similarproductsandfrom
literaturereferencesareessentialtomaketheseriskassessments.Takentogether,thisdataprovidesara
tionalethatlinkstheCQAwiththesafetyandefficacyoftheproduct.Theoutcomeoftheriskassessme
ntwouldbealistofCQAsrankedinorderofimportance.ThepotentialCQAscanbemodifiedwhenth
eformulationandmanufacturingprocessesareselectedandasproductknowledgeandprocessunder
standingincrease.
Qualityriskmanagementsupportsascientificandpracticalapproachtodecision-
making,assessingtheprobability,severityandsometimesdetectabilityoftherisk.Inpharmaceutical
development,riskassessmentisimportantinidentifyingwhichmaterialattributesandprocessparam
eterspotentiallyhaveaneffectonproductCQAs–
CHAPTER I INTRODUCATION
CriticalMaterialAttributes(CMAs)andCriticalProcessParameters(CPPs).Riskassessmentistypi
callyperformedearlyinthepharmaceuticaldevelopmentprocessandisrepeatedasmoreinformation
becomesavailableandgreaterknowledgeisobtained.
Riskstoqualitycanbeassessedinavarietyofinformalways(empiricaland/orinternalprocedures)bas
edon,forexample,compilationofobservations,trendsandotherinformation.Suchapproachesconti
nuetoprovideusefulinformationthatmightsupporttopicssuchashandling of complaints,quality
defects,deviationsandallocation of
resources.Additionally,thepharmaceuticalindustrycanevaluatetheriskusingrecognizedriskmana
gementtools.Someofthesetoolsare:
FailureModeEffectsAnalysis(FMEA)
FailureMode,EffectsandCriticalityAnalysis(FMECA)
FaultTreeAnalysis(FTA)
HazardAnalysisandCriticalControlPoints(HACCP)
HazardOperabilityAnalysis(HAZOP)
PreliminaryHazardAnalysis(PHA)
Riskrankingandfiltering.
Thesetoolsmightbeadaptedforuseinspecificareastodrugsubstanceanddrugproductquality.Also,q
ualityriskmanagementmethodsandsomesupportingstatisticaltoolscanbeusedincombination.Co
mbineduseprovidesflexibilitythatcanfacilitatetheapplicationofqualityriskmanagementprinciple
s.
Thestatisticaltoolscansupportandfacilitatequalityriskmanagement.Theycanenableeffectivedata
assessment,aidindeterminingthesignificanceofthedataset(s),andfacilitatemore reliable decision
making.Exampleof statisticaltool are Designof Experiments,ControlCharts,Histograms,etc.
gprocess.TheoutputofDoEconfirmsCQAsandCPPsthatneedtobecontrolledinthemanufacturing
process.
Inanexperiment,oneormorefactorsaredeliberatelychangedinordertoobservetheeffectononeormo
reresponsevariables.Thismayleadtoanextendnumberofexperiments.InDoE,itisensuredthatthese
lectedexperimentsproducethemaximumamountofrelevantinformation,keepingcostslowbycond
uctingfewexperiments.
CreatedbySirRonalA.Fisherinthe1920sand1930s,DoEisdefinedasastructuredandefficientstatist
icalmethodforplanningexperiments,sothatthedataobtainedcanbeanalyzedto yield valid and
objective conclusions andfordetermining therelationships
amongthefactorsaffectingaprocessanditsoutput.
DoEinitiateswithdefiningtheobjectivesofanexperimentandselectingtheprocessfactorsforthestud
y.Anexperimentaldesignisthelayingoutofadetailedexperimentalplaninadvanceofdoingtheexper
iment.
ThestatisticaltheoryunderlyingDoEgenerallybeginswiththeconceptofprocessmodels,andthemo
stcommonitistheprocessmodelofthe“blackbox”type,withseveraldiscreteorcontinuousinputfacto
rsthatcanbecontrolledandoneormoremeasuredoutputresponses,asshowninFigure12.Themeasur
edresponsesdescribethepropertiesoftheinvestigatedsystem.Bychangingthemostinfluentialfacto
rs(e.g.amountofdisintegrant,timeofmixture,force
ofcompression)thefeaturesofthesystemmightbealtered
accordingtoaresponse(e.g.disintegrationtime,contentuniformity,hardness).Frequently,theexperi
mentsareaffectedbyanumberofuncontrolledfactorsthatmaybediscrete,suchasdifferentmachines
oroperators,and/orcontinuoussuchasambienttemperatureor humidity.
Oncefactorshavebeenchosenandresponsesmeasured,itisdesirabletogetanunderstandingoftherel
ationshipbetweenthem,thatis,linkingthechangesinthefactorstothechangesintheresponseswitha
mathematicalmodel.Infact,thebaseforDoEisanapproximationofrealitywiththehelpofamathemat
icalmodel.Thismodelisnever100%right,butsimplyhelpstotransportthecomplexityoftherealityint
oanequationwhichiseasytohandle.Themostcommonempiricalmathematicalmodelsfittotheexper
imentaldatatakearepolynomialfunctions,usuallyinalinearformorquadraticform.
ThechoiceofanexperimentaldesignisanimportantpartofaDoEprocess,
beingcriticalforthesuccessofthestudy.Thischoicedependsonanumberofaspects,includingthenatu
reoftheproblemandstudy(e.g.,ascreening,optimization,or robustness
CHAPTER I INTRODUCATION
study),thefactorsandinteractionstobestudied(e.g.,four,six,orninefactors,andmaineffectsortwo-
wayinteractions),andavailableresources(e.g.,time,labour,cost,andmaterials).Numerousstatistica
lexperimentaldesigns areknown.Thefollowinglist gives thecommonlyuseddesigntypes:
1. Fullfactorialdesign
2. Box-Behnkendesign
3. Centralcompositedesign
4. Doehlert design
2. Box–Behnken designs
Box and Behnken suggested how to select points from the three-level factorial arrangement,
which allows the efficient estimation of the first and second-order coefficients of the
mathematical model. These designs are, in this way, more efficient and economical then their
corresponding 3k designs, mainly for a large number of variables. Its principal characteristics
are: (1) Requires an experiment number according to N = 2k (k − 1) + cp, where k is the
number of factors and (cp) is the number of the central points; (2) All factor levels have to be
adjusted only at three levels (−1, 0, +1) with equally spaced intervals between these levels.
This experimental design has been applied for the optimization of several chemical and
physical processes; however, its application in analytical chemistry is still much smaller in
comparison with central composite design.
CHAPTER I INTRODUCATION
4)Doehlert design
Developed by Doehlert, the design is a practical and economical alternative in relation to
other second-order experimental matrices. This design describes a circular domain for two
variables, spherical for three variables, and hyper spherical for more than three variables,
which accents the uniformity of the studied variables in the experimental domain. Although
its matrices are not routable as previous designs, it presents some advantages, such as
requiring few experimental points for its application and high efficiency. Other characteristics
are presented below:
Applications of the Doehlert design in analytical chemistry are increasing in recent years,
mainly because of its advantageous characteristics in relation to other designs. Application of
response surface methodology in the optimization of analytical procedures is today largely
diffused and consolidated principally because of its advantages to classical one-variable-a-
time optimization, such as the generation of large amounts of information from a small
number of experiments and the possibility of evaluating the interaction effect between the
variables on the response. The central composite design is still the symmetrical second- order
experimental design most utilized for the development of analytical procedures. The
application of three-level factorial designs is not frequent, and the use of this design has been
limited to the optimization of two variables because its efficiency is very low for higher
numbers of variables. However, the Box–Behnken and Doehlert designs present more
efficient matrices and have increased the number of published works in recent years. Multiple
response optimization using desirability functions have until now had its utilization limited to
the chromatographic field, its related techniques, and to electrochemical methods. However,
its principles can be applied to the development of procedures using various analytical
techniques, which demand a search for optimal conditions for a set of responses
simultaneously.
ADesignSpaceisawaytorepresenttheproductandprocessunderstandingwhichwillbeestablish.Th
eproductandprocessunderstandingandDesignSpacehelpsto
identifyandexplaintheallsourcesofvariabilityandthuswayoutfromthisvariabilitybymeasuringan
dcontrollingtheCPPsandCMAsresponsibleforvariability.Finally,thisassignmentpredictstheaccu
rateandreliableproductqualityattributeswithinspecificationsintermsofquality.
Once a sufficient level of product and process understanding is achieved, through Design
Space, a Control Strategy should be developed that assures that the process will remain
incontrolwithin the normal variation in material attributes and process operating ranges.
CHAPTER I INTRODUCATION
Figure 8 shows how Control Strategy are connected and interact with Design Space and
Knowledge Space.
ControlStrategyisdefinedas“aplannedsetofcontrols,derivedfromcurrentproductandprocessunde
rstandingthatensuresprocessperformanceandproductquality.Thecontrolscanincludeparametersa
ndattributesrelatedtodrugsubstanceanddrug productmaterialsand components, facility and
equipment operating conditions, in-process controls, finished product specifications, and the
associated methods and frequency of monitoring and control.”
AControlStrategyisdesignedtoensurethataproductofrequiredqualitywillbeproducedconsistently
.Theelementsofthecontrolstrategyshoulddescribeandjustifyhowin-
processcontrolsandthecontrolsofinputmaterials(drugsubstanceandexcipients),intermediates(in
-
processmaterials),containerclosuresystem,anddrugproductscontributetothefinalproductquality.
Thesecontrolsshouldbebasedonproduct,formulationandprocessunderstandingandshould
include,at a minimum,control oftheCPPsandCMAs.In
aQbDapproach,pharmaceuticaldevelopmentwillgenerateprocessandproductunderstandingandi
dentifysourcesofvariability.Thissourcesofvariabilitymayimpactonproductqualityandtherefores
houldbeidentified,understood,andsubsequentlycontrolled.Productandprocessunderstanding,inc
ombinationwithqualityriskmanagement,willsupportthecontroloftheprocesssuchthatthevariabili
tycanbecompensatedforinanadaptablemannertodeliverconsistentproductquality
Scale-
up,technologytransferandmanufacturingexperiencecanleadtorefinementsofthecontrolstrategy.
ICHQ10describesamodelfortheestablishmentofaneffectivePharmaceuticalQualitySystem(PQS
) that can be used by manufacturers implementing QbD systems and can evaluate and
improve product quality throughout the product lifecycle. In fact, PQS facilitate continual
improvement,helpingthe identificationandimplementationof
appropriateproductandprocessqualityimprovements,reducingthevariability,andidentifyingandp
rioritizingareasforcontinualimprovement.Itisimportanttosharetheknowledgegainedduringdevel
CHAPTER I INTRODUCATION
opmentandimplementationthatisrelevantforutilizationofthatDesignSpaceonthemanufacturingfl
oorandunderthePQS.Thisknowledgecanincluderesultsofriskassessments,assumptionsbasedonp
riorknowledge,andstatisticaldesignconsiderations.LinkagesamongtheDesignSpace,ControlStr
ategy,CQAandQTPPareanimportantpartofthissharedknowledge.
Inthecaseofchangestoanapproved designspace,appropriatefilingsshouldbemade
tomeetregulatoryrequirements.Movementwithintheapproveddesignspace,asdefinedintheICHQ
8(R2)glossary,doesnotcallforaregulatoryfiling.Formovementoutsidethedesignspace,
theuseofriskassessmentcouldbehelpfulindeterminingtheimpactofthechange on
quality,safetyand efficacy and theappropriate regulatory filing strategy.
CHAPTER II DRUG PROFILE
CHAPTER 2
DRUG PROFILE
CHAPTER II DRUG PROFILE
Name Trabectedin
Structure
Structure of Trabectedin
CAS NO 114899-77-3
(1'R,6R,6aR,7R,13S,14S,16R)-6',8,14-trihydroxy-7',9-
dimethoxy-4,10,23-trimethyl-19-oxo-
IUPAC Name 3',4',6,7,12,13,14,16-octahydrospiro[6,16-
(epithiopropano-oxymethano)-7,13-imino-6aH-1,3-
dioxolo[7,8]isoquino[3,2-b][3]benzazocine-20,1'(2'H)-
isoquinolin]-5-yl acetate
DNA Blocker in Myxoid liposarcoma
Metastatic leiomyosarcoma
Category & Medical use Metastatic liposarcoma
Relapsed platinum-sensitive ovarian cancer
Unresectable leiomyosarcoma
pKa 9.26
CHAPTER 3
LITERATURE
REVIEW
CHAPTER III LITERATURE REVIEW
Abstract
A rapid, sensitive and specific HPLC–MS/MS method was developed and validated for the
quantification of trabectedin in human plasma after using deuterated trabectedin as Internal
Standard (IS). After the addition of ammonium sulphate, the analyte was extracted from
human plasma with acidified methanol (0.1 M HCl). Chromatographic separation was done
on an Accucore XL C18 column (4 μm; 50 mm × 2.1 mm) using a Mobile Phase (MP)
consisting of CH3COONH4 10 mM, pH 6.8 (MP A) and CH3OH (MP B). The mass
spectrometer worked with electrospray ionization in positive ion mode and Selected Reaction
Monitoring (SRM), using target ions at [M−H2O+H]+ m/z 744.4 for trabectedin and
[M−H2O+H]+ m/z 747.5 for the IS. The standard curve was linear (R2 ≥ 0.9955) over the
concentration range 0.025–1.0 ng/ml and had good back-calculated accuracy and precision.
The intra- and inter-day precision and accuracy determined on three quality control samples
(0.04, 0.08 and 0.80 ng/ml) were <9.9% and between 98.3% and 105.3%, respectively. The
extraction recovery was >81% and the lower limit of quantification 0.025 ng/ml. The method
was successfully applied to study trabectedin pharmacokinetics in a patient with a
liposarcoma who received 1.3 mg/m2 as a 24 h continuous i.v. infusion.
performed according to the most recent FDA guidelines for bioanalytical method validation.
The time needed for off-line sample preparation has been reduced 10-fold compared with an
existing LC/MS/MS method for ET-743 in human plasma, employing a labor-intensive solid-
phase extraction procedure for sample pretreatment. The proposed column switching method
was successfully applied in phase II clinical trials with Yondelis and pharmacokinetic
monitoring. Copyright © 2004 John Wiley & Sons, Ltd.
G Sirisha et al, Analytical Method Development And Validation For The Estimation Of
Trabectedin In Bulk And Parenteral Dosage Form By RP-HPLC, 2018
Abstract: A new RP-HPLC method for the quantitative determination of Trabectedin was
developed and validated as per ICH guidelines. The drugs were injected into Zorbax SB,
C18,(150x4.6mm); 3.5µm column maintained at ambient temperature and effluent
monitored at 215nm. The mobile phase consisted of phosphate buffer (pH 3.0) and
Acetonitrile in the ratio of 70:30 v/v. The flow rate was maintained at 0.8 ml/min. The
calibration curve for Trabectedin was linear from 50-175µg/ml (r2 for Trabectedin = 1). The
proposed method was adequate, sensitive, reproducible, accurate and precise for the
determination of Trabectedin in bulk and pharmaceutical dosage forms.
degradation compounds in forced degradation study and excipients was noticed. The
Robustness study and percentage of the assay of the formulations was established within the
limit of ICH guidelines
2.
(b) National status
1. Blessy M et al, (2013) Forced degradation is a degradation of new drug substance and
drug product at conditions more severe than accelerated conditions. It is required to
demonstrate specificity of stability indicating methods and also provides an insight into
degradation pathways and degradation products of the drug substance and helps in
elucidation of the structure of the degradation products. Forced degradation studies show the
chemical behavior of the molecule which in turn helps in the development of formulation and
package. In addition, the regulatory guidance is very general and does not explain about the
performance of forced degradation studies. Thus, this review discusses the current trends in
performance of forced degradation studies by providing a strategy for conducting studies on
degradation mechanisms and also describes the analytical methods helpful for development
of stability indicating method.
2. S. V. Mulgund et al, A simple, specific, accurate and stability-indicating reversed phase high
performance liquid chromatographic method was developed for the simultaneous
determination of mephenesin and diclofenac diethylamine, using a Spheri-5-RP-18 column
and a mobile phase composed of methanol: water (70:30, v/v), pH 3.0 adjusted with o-
phosphoric acid. The retention times of mephenesin and diclofenac diethylamine were found
to be 3.9 min and 14.5 min, respectively. Linearity was established for mephenesin and
diclofenac diethylamine in the range of 50-300 μg/ml and 10-60 μg/ml, respectively. The
percentage recoveries of mephenesin and diclofenac diethylamine were found to be in the
range of 99.06-100.60% and 98.95-99.98%, respectively. Both the drugs were subjected to
acid, alkali and neutral hydrolysis, oxidation, dry heat, photolytic and UV degradation. The
degradation studies indicated, mephenesin to be susceptible to neutral hydrolysis, while
diclofenac diethylamine showed degradation in acid, H2O2, photolytic and in presence of UV
radiation. The degradation products of diclofenac diethylamine in acidic and photolytic
conditions were well resolved from the pure drug with significant differences in their
retention time values. This method can be successfully employed for simultaneous
quantitative analysis of mephenesin and diclofenac diethylamine in bulk drugs and
CHAPTER III LITERATURE REVIEW
formulations
3.M. Vamsi Krishna et al, Quality by Design (QbD) approach to develop HPLC method for
berconazole nitrate:Application to hydrolytic, thermal, oxidative and photolytic degradation
kinetics, Stability of eberconazole nitrate (EBZ) was investigated using a stability indicating
HPLC method. Quality by Design (QbD) approach was used to facilitate method
development. EBZ was exposed to different stress conditions, including hydrolytic (acid,
base, neutral), oxidative, thermal and photolytic. Relevant degradation was found to take
place in all the conditions. The degradation of EBZ followed (pseudo) first-order kinetics
under experimental conditions. The kinetic parameters (rate constant,t1/2, andt90) of the
degradation of EBZ were calculated
generic approach can be used as a starting point to set up a stress testing study, but a case-by-
case approach for stress testing is essential to allow flexibility. This is also recognized by the
regulatory agencies because very detailed instructions about how to perform stress testing are
not given in the available guidance documents. In first instance, a SIM should be applied to
quantification of the main active in a precise and accurately, even in the presence of
interferences (degradation products, impurities, and placebo). In this manner, some
evaluation of mass balance can be done, but the applicant must have in mind, the limitation
about the obtained values, since %area is a useful but not precise/accurate tool to determine
concentrations. If during long-term stability studies or quality control determinations, the
main active concentration decreases, mass balance investigations may start, evaluating
%peak, DAD spectra and inparallel, different/contributing analytical techniques.
Conclusion:
As per literature survey, No one has done the force degradation study for Trabectedin by
HPLC method also nobody did the quality by design approach any method development and
validation.
It was best opportunity to develop the stability indicating method for the estimation of
Trabectedin by Quality by Design Approach.
CHAPTER III LITERATURE REVIEW
CHAPTER IV AIM AND OBJECTIVES
CHAPTER 4
AIM AND
oBJECTIVES
CHAPTER IV AIM AND OBJECTIVES
CHAPTER 5
PLAN OF WORK
CHAPTER IV PLAN OF WORK
CHAPTER IV PLAN OF WORK
1.4Experimental
1.4.1 Materials
Sr.
Name ofChemicals Grade Manufacturer
No
1 Methanol HPLC Grade Merck Lie Sciences Pvt. Ltd, Mumbai.
2 Acetonitrile HPLC Grade Merck Lie Sciences Pvt. Ltd, Mumbai.
3 Ethanol HPLC Grade Merck Lie Sciences Pvt. Ltd, Mumbai.
4 Water HPLC Grade Merck Lie Sciences Pvt. Ltd, Mumbai.
Potassium dihydrogen
5 LR Grade Research Fine Chem. Indu.
phosphate
6 Sodium hydroxide LR Grade Research Fine Chem. Indu.
7 Triethylamine LR Grade Research Fine Chem. Indu.
CHAPTER IV PLAN OF WORK
Method
Method Development
The best peak parameter plays a significant role to determine a suitable solvent system and
column. (ACN: Buffer, ACN: Water, Methanol: Water) these three mobile phases were
selected for the factorial design. We performed trails on C18 columns at pH 4.0 and 6.0.
These mobile phases were screened by varying the organic phase composition from 70 to 90
% v/v and flow rate from 1.00 mL/ min flow rate consider standard. After performing the
trials, we optimized chromatographic conditions according to Peak properties by scaling it
with satisfactory, partially satisfactory, extremely satisfactory, dissatisfactory
RESULT AND
DISCUSSION
CHAPTER V RESULT AND DISCUSSION
Hydrolysis
Acid Degradation Studies
Approximately 1 mL of solution of stock Trabectedin was added, followed by 1 mL of 1 N
hydrochloric acid, which was then refluxed for 30 minutes at 60 0C/400C for 1 to 5 sampling
Days. A 400 ppm and a 50 ppm solution were prepared from the resulting solution, and 0.30
μL solutions were injected into the system, and the chromatograms were recorded in order to
determine the stability of the sample.
Table 1. Software has given Conditions for forced degradation studies.
Storage Sampling
Condition in time Retention Retention
Sr. Peak Are of Peak Are of Percentage
time of time of
No Trabectedin Degradent Degradation
Temp in Trabectedin Degradent
0 Days
C
1 60 1 5.689 5708920.514 3.23 608353.4862 9.63
2 40 5 5.687 5909809.827 3.231 407464.173 6.45
3 60 5 5.689 5251549.876 3.231 1065724.124 16.87
4 40 1 5.689 6077217.588 3.23 240056.412 3.80
Standard Peak Area of Trabectedin 30ug/mL: 6317274
Day 01
Results of Hydrolysis by 1 N HCl at 400C
The percentage degradation of Trabectedin was found to be 3.80 % in acidic condition. The
percentage degradation was within acceptable criteria (NMT 10%). The extra peaks were
eluted at retention times of 3.23min and the chromatogram was shown in Figure
CHAPTER V
RESULT AND DISCUSSION
Day 05
Results of Hydrolysis by 1 N HCl at 400C
The percentage degradation of Trabectedin was found to be 6.45% in acidic condition. The
percentage degradation was within acceptable criteria (NMT 10%). The extra peaks were
eluted at retention times of 3.231min and the chromatogram was shown in Figure
Day 01
Results of Hydrolysis by 1 N HCl at 600C
The percentage degradation of Trabectedin was found to be 9.63% in acidic condition. The
percentage degradation was within acceptable criteria (NMT 10%). The extra peaks were
eluted at retention times of 3.23min and the chromatogram was shown in Figure
CHAPTER V RESULT AND DISCUSSION
Day 05
Results of Hydrolysis by 1 N HCl at 600C
The percentage degradation of Trabectedin was found to be 16.87% in acidic condition. The
extra peaks were eluted at retention times of 3.23min and the chromatogram was shown in
Figure
Conclusion: Less degradation will exist 400C and Day 1st also all runs shown percentage
degradationis less than 10%. The percentage degradation was within acceptable criteria
(NMT 10%) and for 60 degree it shows more degradation.
Storage Sampling
Retention Retention
Sr. Condition in time Peak Are of Peak Are of Percentage
time of time of
No Trabectedin Degradent Degradation
Temp in Trabectedin Degradent
0 Days
C
1 60 1 5.689 4909153.625 3.231 1408120.375 22.29
2 40 5 5.688 5081615.206 3.231 1235658.794 19.56
3 60 5 5.689 4239522.581 3.231 2077751.419 32.89
4 40 1 5.689 5482762.105 3.231 834511.8954 13.21
Standard Peak Area of Trabectidin 30ug/mL: 6317274
CHAPTER V RESULT AND DISCUSSION
Day 01
Results of Hydrolysis by 1 N NaOH at 400C
The percentage degradation of Trabectedin was found to be 13.21 % in basic condition. The
extra peaks were eluted at retention times of 3.231 min and the chromatogram was shown in
Figure
Day 05
Results of Hydrolysis by 1 N NaOH at 400C
The percentage degradation of Trabectedin was found to be 19.56 % in basic condition. The
extra peaks were eluted at retention times of 3.231 min and the chromatogram was shown in
Figure
CHAPTER V
RESULT AND DISCUSSION
Day 01
Results of Hydrolysis by 1 N NaOH at 600C
The percentage degradation of Trabectedin was found to be 22.29 % in basic condition. The
extra peaks were eluted at retention times of 3.231 min and the chromatogram was shown in
Figure
Day 05
Results of Hydrolysis by 1 N NaOH at 600C
The percentage degradation of Trabectedin was found to be 32.89% in basic condition. The
extra peaks were eluted at retention times of 3.231 min and the chromatogram was shown in
Figure
CHAPTER V
RESULT AND DISCUSSION
Conclusion: Less degradation will exist 400C and Day 1stand its percentage is 13.21% . The
percentage degradation was not within acceptable criteria (NMT 10%) and for 60 degree it
shows more degradation so that method says that drug is basic degradable.
Storage Sampling
Condition in time Retention Peak Area Peak Area
r. Peak Are of Rt of 1st Rt of 2nd Percentag
time of of 1st of 2nd
o Trabectedin Degradent Degradent Degradatio
Temp in Trabectedin Degradent Degradent
0 Days
C
60 1 5.689 4279953 7.214 2037321 9.31 679107 32.25
25 5 5.687 4360183 7.214 1957091 9.31 652364 30.98
60 5 5.689 3655806 7.214 2661468 9.31 887156 42.13
25 1 5.689 4891465 7.214 1425809 9.31 475270 22.57
Standard Peak Area of Trabectedin 30ug/mL: 6317274
Day 01
Results of Oxidation by 20 PercentH2O2 at 600C
The percentage degradation of Trabectedin was found to be 32.25% in Oxidative condition.
The extra peaks were eluted at retention times of 7.214min and9.31 min, the chromatogram
was shown in Figure
Day 05
Results of Oxidation by 20 PercentH2O2 at 600C
The percentage degradation of Trabectedin was found to be 42.13% in Oxidative condition.
The extra peaks were eluted at retention times of 7.214min and9.31 min, the chromatogram
was shown in Figure
CHAPTER V RESULT AND DISCUSSION
Day 01
Results of Oxidised by 20 PercentH2O2 at 250C
The percentage degradation of Trabectedin was found to be 22.57% in Oxidative condition.
The extra peaks were eluted at retention times of 7.214min and9.31 min, the chromatogram
was shown in Figure
Day 05
Results of Oxidised by 20 PercentH2O2 at 250C
The percentage degradation of Trabectedin was found to be 30.98% in Oxidative condition.
The extra peaks were eluted at retention times of 7.214min and9.31 min, the chromatogram
was shown in Figure
CHAPTER V RESULT AND DISCUSSION
Conclusion: Less degradation will exist on 250C and 1st Day with 22.57% but all runs shown
percentage degradationis more. Method concluded that the drug is oxidative.
diluting the resulting solution to 400 parts per million (ppm) and 50 parts per million (ppm)
solutions, injecting 0.30 μL of the solution into the system and recording the chromatograms
to determine the stability of the sample.No major degradation wasobserved.
Storage Sampling
Condition in time Retention Retention Peak Are
Peak Are of Percentage
Sr. No time of time of of
Trabectedin Degradation
Temp in Trabectedin Degradent Degradent
0 Days
C
1 105 1 5.689 1756833.899 8.324 4560440.1 72.19%
2 30 5 5.687 2991229.239 8.324 3326044.76 52.65%
3 105 5 5.689 765653.6088 8.324 5551620.39 87.88%
4 30 1 5.689 3643803.643 8.324 2673470.36 42.32%
Standard Peak Area of Trabectedin 30ug/mL: 6317274
Day 01
Results of Thermolysis at 300C
The percentage degradation of Trabectedin was found to be 42.32% in thermolytic condition.
The extra peaks were eluted at retention times of 8.324min; the chromatogram was shown in
Figure
Day 05
Results of Thermolysis at 300C
CHAPTER V RESULT AND DISCUSSION
Day 01
Results of Thermolysis at 1050C
The percentage degradation of Trabectedin was found to be 72.19%in thermolytic condition.
The extra peaks were eluted at retention times of 8.324min; the chromatogram was shown in
Figure
Day 05
CHAPTER V
RESULT AND DISCUSSION
Conclusion: The drug is thermo labile because it shown the more degradation in all
conditions. It is not within prescribed limits.
Results of various trials performed on C1 Column at different Mobile phase with different pH
of aqueous phase.
Table 1.5: Trials performed at mobile phase (70:30 v/v) with aqueous phase pH 4
CHAPTER V
RESULT AND DISCUSSION
Sr.
Composition Observation Remarks
no.
Peak asymmetry is good but Partly
1 ACN: Water
less theoretical plates satisfactory
Greater peak asymmetry and
2 Methanol: Water Not satisfactory
lower theoretical plates
Good Retention Time, Less
Extremely
3 ACN: buffer Asymmetry & Remarkable
Satisfactory
theoretical plates
Table 1.6: Trials performed at mobile phase (90:10 v/v) with aqueous phase pH 6
Sr.
Composition Observation Remarks
no.
Less peak asymmetry with More than
1 ACN: Water
more theoretical plates Satisfied
Less peak asymmetry but less
2 Methanol: Water Satisfied
theoretical plates
Very
3 ACN: buffer No peak appearence
Dissatisfied
Table 1.7: Trials performed at mobile phase (70:30 v/v) with aqueous phase pH 6
Sr.
Composition Observation Remarks
no.
More retention time Not
1 ACN: Water
and less peak height satisfactory
Not at all
2 Methanol: Water More retention time
satisfactory
Very
3 ACN: buffer No peak appearence
Dissatisfied
Table 1.8: Trials performed at mobile phase (90:10 v/v) with aqueous phase pH 4
Sr.
Composition Observation Remarks
no.
1 ACN: Water Good peak properties Satisfactory
Not
2 Methanol: Water More retention time
satisfactory
Greater peak Not
3 ACN: buffer
asymmetry satisfactory
time: 10 min.
Calibration Curve
7000000
6000000 y = 210862x - 13333
R² = 0.9999
5000000
Peak Area
4000000
3000000
2000000
1000000
0
0 5 10 15 20 25 30 35
Concentration
Linearity
Selectivity
Brand name of Trabectedin Formulation is Yondelis1mg.Vial Injectable of Trabectedin
formulation in volumetric flask and 1ml (1 mg of pure drug) including other excipients was
shifted in 10 ml volumetric flask and volume were adjusted with Acetonitrile. Retention time
and another peak parameter of the formulation were compared with the API of the drug. A
chromatogram is shown in figure 5.11. Retention time, Area, Asymmetric factor, and
Theoretical Plates were found to be 5.689min, 1052879, 1.212 and 9895 respectively. These
results concluded that the excipient of the capsule does not interfere with the separation of the
drug in the selected mobile phase.
Figure
1.11: A typical chromatogram of Trabectedin [Concentration 25ug/ml]
Sensitivity
The sensitivity of the method can be determined by the limit of detection and the limit of
quantitation. Signal to noise ratio is of paramount importance in the calculation. Six
replicates of the blank sample were run and calculate noise level which is three times of
LOD and ten times of LOQ respectively.
Found
Concentration
Sr. No Peak area Concentration % Recovery
(μg/mL)
(μg/mL)
1 16 3394816.8 15.99 99.92
2 20 4243521 19.99 99.95
3 24 5092225.2 24.09 100.36
Precision
Precision is the series of measurements in different environments of all day. We performed
intraday precision and Interday precision. Six replicates of 20 µg/ml solution were prepared
and took 6 readings in a day right from morning to evening. Intraday precision proved that the
method remains stable in different climatic conditions of the day. Relative standard deviation
% was found to be 1.5652likewise Interday precision is the sample analysis for three days
and relative standard deviation % was found to be 1.730. Analytical precision yields reliable
results and complies with the ICH guideline. Repeatability was performed by preparing the
six replicates and RSD% was 0.6940. The results obtained are shown in table 5.30.
Table 1.18:SystemPrecisionresultsforTrabectedinbyRP-HPLC
Sr. Concentration
Intraday Interday
No (μg/mL)
1 20 4235992.5 4224355
2 20 4261017 4265265.333
3 20 4087630 4350678
4 20 4242997 4130308
5 20 4274752 4211218
6 20 4248203.25 4176939
Average 4225098.6 4226460.6
Standard Deviation 62750.91 69366.48
RSD% 1.4852 1.641
CHAPTER V RESULT AND DISCUSSION
RSD 1.641
Specificity
Chromatogram of Trabectedin Esilate in capsule formulation showed a peak at a retention time
of 4.217min. The mobile phase designed for the method resolved the drug very efficiently and
The Retention time of Trabectedin was 4.217± 0.0018 min. The detection of wavelength was
265 nm. The peak properties of a formulation containing the drug were compared with
standard and it was observed that the peak was adequately resolved without the interference of
the excipient. Recovery was achieved at 99.94%. The result was shown in table 5.31.
Table 1.15: Results of Specificity
Label Claim
Sample Amount Found Recovery Retention Time
(mg)
Vial 1 0.98 98% 5.689
CHAPTER V RESULT AND DISCUSSION
Repeatability
Demonstrationofprecisionwas done undertwocategories.Theinjectionrepeatabilit y
(SystemPrecision)wasassessedbyusingsixinjectionsofthestandardsolutiono fTrabectedinandthe
%RSDofthereplicateinjectionswascalculated. Results obtained are shown in table 100.
Table 1.16:Repeatability resultsforTrabectedinbyRP-HPLC
Concentration
Sr. No Peak Area
(μg/mL)
1 20 4216734
2 20 4196734
3 20 4216747
4 20 4209860
5 20 4266734
6 20 4214522
Average 4220221.8
Standard Deviation 21905.7
RSD% 0.519
Robustness
Robustnessisameasureofcapacityofamethodtoremainunaffectedbysmall,but
deliberatevariationsinthemethodconditions,andisindicationsofthereliabilityofthemethod.Ameth
odisrobust,ifitisunaffectedbysmallchangesinoperating
conditions.Todeterminetherobustnessofthismethod,theexperimentalconditionsweredeliberately
alteredatthreedifferentlevelsandretentiontimeand chromatographicresponsewereevaluated.
Onefactoratatimewaschangedtostudy theeffect.Variationofmobilephasecompositio n
(70:30v/v), flowrateby1ml/min(0.9 and1.1ml/min), Variation of wavelength by 265 nm (263
nm and 267 nm), Variations in pH by 5 (4.8 and 5.2)hadnosignificanteffectonthe retention
time and chromatographic response of 10 μg/ml solution, indicating that the method was
robust, The result was shown in table
Detection
Acetonitrile : Buffer Retention Wavelength Peak Area
Time (min)
(V/V) (nm)
CHAPTER V RESULT AND DISCUSSION
Standard
Standard Deviation 0.1001 43733.13
Deviation
Recovery
The The recovery of the method was evaluated by the addition of standard at three levels 80,
100, and 120 % to the mixture. Six replicates were run and calculated the mean amount of
drug recovered and percentage recovery. It found that results were observed within the
prescribed limit. Results were shown in table 8.
Amount Amount of
Amount of
of Drug Drug
Sample
Sr. No Added Recovered Recovery %
1 20 10 9.989 99.89
2 20 20 19.99 99.96
3 20 30 30.01 100.03
CHAPTER V RESULT AND DISCUSSION
Part-II
According to the above prescribed, the RP-HPLC method was developed and validated as per
ICH guideline. Optimized mobile phase by design expert software was used to estimate drug
in human plasma. Details are as follows.
Introduction of research:
Stability requirements during development of a drug
Chemical stability of pharmaceutical molecules is a matter of great concern as it affects the
safety and efficacy of the drug product. The FDA and ICH guidance’s state the requirement
of stability testing data to understand how the quality of a drug substance and drug product
changes with time under the influence of various environmental factors. Knowledge of the
stability of molecule helps in selecting proper formulation and package as well as providing
proper storage conditions and shelf life, which is essential for regulatory documentation.
Forced degradation is a process that involves degradation of drug products and drug
substances at conditions more severe than accelerated conditions and thus generates
degradation products that can be studied to determine the stability of the molecule. The ICH
guideline states that stress testing is intended to identify the likely degradation products
which further helps in determination of the intrinsic stability of the molecule and establishing
degradation pathways, and to validate the stability indicating procedures used. But these
guidelines are very general in conduct of forced degradation and do not provide details about
the practical approach towards stress testing. Although forced degradation studies are a
regulatory requirement and scientific necessity during drug development, it is not considered
as a requirement for formal stability program.
It has become mandatory to perform stability studies of new drug moiety before filing in
registration dossier. The stability studies include long term studies (12 months) and
CHAPTER V RESULT AND DISCUSSION
also storage produced. The accelerated testing on drug substance and drug product provides
the in sequence to the inherent stability of the bulk drug molecule formation and may
establish the likely degradation pathways. The formulation group also has the responsibility
for recommending to the toxicology group about stability of drug substance in the medium
Objectives
To Apply Stability indicating method to the Trabectedin
To establish degradation pathways of drug substances and drug products.
To determine the intrinsic stability of a drug substance in formulation.
To reveal the degradation mechanisms such as hydrolysis, oxidation, thermolysis or
photolysis of the drug substance and drug product
To establish stability indicating nature of a developed method.
To understand the chemical properties of drug molecules.
To produce a degradation profile similar to that of what would be observed in a formal
stability study under ICH conditions.
CHAPTER VI CONCLUSION
CONCLUSION
CHAPTER VI CONCLUSION
.
❖ Method I
REFERENCE
CHAPTER VII REFERENCE
Chapter
References
1. ChatwalGR,AnandSK.InstrumentalMethodsofChemicalAnalysis:Himalaya Publishing
House; 1979.
2. VA.ATextbookofQuantitativeAnalysis:ForeignLanguagesPublishingHouse.
3. Skoog DA, Holler FJ, Crouch SR. Principles of Instrumental Analysis:
ThomsonBrooks/Cole; 2007.
4. WillardHH,MerrittJrLL,DeanJA,SettleJrFA.Instrumentalmethodsofanalysis. 1988.
5. Lawrence XY. Pharmaceutical quality by design: product and process
development,understanding, and control. Pharmaceutical research. 2008;25(4):781-91.
6. MartensH,MartensM.Multivariateanalysisofquality.Anintroduction.IOPPublishing; 2001.
7. Guidance for industry [electronic resource] : Q8(R2) pharmaceutical
development.CenterforDrugE,Research,CenterforBiologicsE,Research,InternationalConf
erenceonH,editors.Rockville,MD:U.S.Dept.ofHealthandHumanServices,FoodandDrugA
dministration,CenterforDrugEvaluationandResearch : Center for Biologics Evaluation
and Research; 2009.
8. Guidance for industry [electronic resource] : Q9 quality risk management. Centerfor
Drug E, Research, Center for Biologics E, Research, International Conferenceon H,
editors. Rockville, MD: U.S. Dept. of Health and Human Services, Food
andDrugAdministration,CenterforDrugEvaluationandResearch:CenterforBiologics
Evaluation and Research; 2006.
9. Dudhe, P.; Shelke, P., Chavare, P. Determination of Apixaban From Bulk and Tablet
Dosage Form by Area Under Curve and First Order Derivative Spectrophotometric
Methods. Int J PharmTech Res., 2017,10(5), 703-711.
10. Katta, R.; Nagaraju, C.; Ramasrinivas.; Rao G. Two Novel Validated RP-HPLC and Uv
Spectrophotometric Methods for Estimation of Apixaban In Bulk and Pharmaceutical
Dosage Form. Am. j. PharmTech Res., 2015, 5(4), 451-460.
11. Kashid, A.; Vidhate, M.; Ingale, M. Analytical Method Development and Validation for
Estimation of Apixaban by RP-HPLC. Indian Drugs., 2017, 54(4), 76-79.
12. Landge, S.; Jadhav, S.; Dahale, S.; Solanki, P.; Bembalkar, S.; Mathad, V. Development
and Validation of Stability Indicating RP-HPLC Method on Core Shell Column for
Determination of Degradation and Process Related Impurities of Apixaban—An
CHAPTER VII REFERENCE
65. Bhinge, Somnath D., Sharangouda M. Malipatil, and Lalit V. Sonawane. "Bioanalytical
method development and validation for simultaneous estimation of cefixime and
dicloxacillin by RP-HPLC in human plasma." Acta Chimica Slovenica 61, no. 3 (2014):
580-586.
66. Jayaseelan, S., S. Suresh, G. Sathishkumar, V. Sekar, and P. Perumal. "Bioanalytical
method development and validation of Lamivudine by RP-HPLC method." Int J Chem
Tech Res 2 (2010): 163-7.
67. Lin, Dan, Li-man Qiao, Yu-niao Zhang, Yuan Liu, and Xin-she Liu. "Simultaneous
determination of nintedanib and its metabolite by UPLC–MS/MS in rat plasma and its
application to a pharmacokinetic study." Journal of pharmaceutical and biomedical
analysis 117 (2016): 173-177.
68. Ameeduzzafar, Javed and Asgar A, "Development and validation of UPLC/ESI-Q-TOF-
MS for carteolol in aqueous humor: Stability, stress degradation, and application in the
pharmacokinetics of nanoformulation"Arabian Journal of Chemistry 10, no. 3 (2017):
S2969-S2978.
69. Ameeduzzafar, Ibrahim E, Nabil K., Imam S, Ahmed F, Mohammed H, Ahmad N, and
Elmowafy M, Quality by design (QbD) based development and validation of
bioanalytical RP-HPLC method for dapagliflozin: Forced degradation and preclinical
pharmacokinetic stud, Journal of Liquid Chromatography & Related Technologies 43, no
1-2, (2020): 01-12.
CHAPTER VIII PRESENTATION AND PUBLICATION
PRESENTATION
&
PUBLICATION
CHAPTER VIII PRESENTATION AND PUBLICATION