6. / BRIEF RESUME OF THE INTENDED WORK:
6.1 NEED OF THE STUDY:
The stability-indicating assay is a method is employed for the analysis of stability samples in pharmaceutical industry. With the advent of International Conference on Harmonization (ICH) guidelines, the requirement of establishment of stability-indicating assay method (SIAM) has become more clearly mandated.
The purpose of stability studies is to monitor possible changes to product or material over time. Stability is essential factor to determine quality, safety and efficacy of a drug product. The stability assay method developed as per ICH guidelines which reveal the stability, storage conditions, shelf-lives, and expiry date of drug product.ICH Q5C states that not just one method is stability- indicating but stability can be inferred by a combination of identity, purity, and density of the drug.
SIAM mainly relays on forced degradation samples, the ability of forced degradation studies is to forecast real-time degradation has been the object of several studies. SIAM supports the quality and efficacy claims of drug, by which it ensures the quality of production to support site or changes to the product.
Gastrointestinal (GI) Problems are becoming very common in present world; this is due to change in food style and food habits. There are different class of GI agents are available, Antacids like Magnesium Trisilicate, Aluminum hydroxide, etc., H2antagonists like famotidine, ranitidine, etc. and Proton Pump Inhibitors like omeprazole, Lansoprazole. These are used for various GI disorders. Ranitidine is H2 antihistaminic drug, as H2antagonists are sparing as compared to Proton pump inhibitors and this has been in use from long time. Ranitidine is classical drug, due concerns of yield change in regulations, and intellectual property issues, industry constantly develops newermanufacturing methods. To ensure drugs are stable in these newly developed manufacturing methods SIAMs are essential. This work is carried out with goal of developing stability indicative assay method of one of the APIs that is used in treatment of GI disorders.
Hence it is necessary to develop a method for assessing the stability drug product of API.
6.2 REVIEW OF LITERATURE:
The literature was found to be instinct with publications on development ofstability indicatingassayof specific drugs. A general review was published in 1971, it gave general principles. Ho and Chen reviewedstability indicating HPLCassay methods reported till 1996. A compilation ofstability indicatingassay (>500) for various drugs was published in 1999 by Xu and Trissel, The book ‘DrugStability Principles and Practices’ by Carstensen and Rhodesreview on the development of validatedstability indicatingassay methods(SIAMs) for drug substances and products. Critical issues related to development of SIAMs, such as separation of all degradation products, establishment of mass balance, stress testing of formulations, development of SIAMs for combination products, etc. are also addressed.1
According to literature survey states that Quantitative analysis of polymorphic mixtures ofranitidine hydrochloride by Raman spectroscopy and principal components analysisdrug exists as two polymorphs, forms I and II, both of which are used to manufacture commercial tablets quantitative analysis of one polymorph as an impurity in the other lack sensitivity by using this principal components analysis (PCA) of Raman spectra to binary mixtures of the two polymorphs and to binary mixtures prepared by adding one polymorph to powdered tablets of the other. Based on absorption measurements of seven spectral regions, it was found that >97% of the spectral variation was accounted for by three principal components. Quantitative calibration models generated by multiple linear regressions predicted a detection limit and quantization limit for either forms I or II in mixtures of the two of 0.6 and 1.8%, respectively.PCA of Raman spectroscopicbest tool for quantitative analysis of polymorphic impurities of drugs in commercial tablets with a quantization limit of less than 2%.2
International Conference for Harmonization (ICH) has outlined purpose of stability studies, criteria for selection of batches, storage conditions, study/test designs during stability studies along with description on evaluation of stability data under guidelines for stability testing of new drug substances and products.4,5,6
United states Food And Drug Administration (USFDA) have described the criteria for determining the content uniformity at powder blend stage, lubrication stage and at compression stage under guidance for industry on powder blends and finished products stratified In-Process Dosage Unit sampling and assessment. 3
.
It has been revealed in literature survey about Development of an isocraticstabilityindicating HPLC separationRanitidine hydrochloride, the development of stability indicating
assay for Ranitidinehydrochlorideusing a mobile phase added ion-interaction reagent was achieved. Thisis be used for the determination of purity, identity and strength for the active ingredient and finished dosage forms. Placebo samples were analyzed for all of the dosage forms and did not interfere with the separation. The system was found to be linear over a range of 0.056 to 44.4 μg/g, with a limit of detection of 0.028 μg/g and a limit of quantification of 0.056 μg/g. The system precision was determined to be 0.7%. 7
Injectable solutionsofranitidineby dissolvingranitidine HCLin water for injections.Thebuffering system has been used was sodium phosphate (Anhydrous),Potassiumdihydrogenphosphate andphenolas a preservativeInert gas (nitrogen) was used to displaceOxygenfrom a solution and reduce the possibility of oxidative changes in the formulation. The solution was poured into 2-ml brown glass ampoules in aseptic condition. Ampoules samples have been stored at three different temperatures. They have been stored at 55 and 40°C for 6 months and at 25°C for 12 months. TLC technique has been used for monitoring related substances, and HPLC technique formonitoring phenolandranitidine content. It has been shown that only those samples that were stored at 25°C were actually stable.8
From literature survey, aselective and accurate high-performance liquid chromatographic method has been developed and validated for the simultaneous determination ofmethylParaben(MP) andpropylparaben(PP) in oral liquids. Samples were purified by solid-phase extraction (SPE) using a copolymeric [poly (divinylbenzene-co-N-inylpyrrolidone)] sorbent. The chromatographic separation was achieved by HPLC using a mixture ofammonium acetatesolution (0.5M),acetonitrileand methanol as the mobile phase with gradient elution, a Nucleosil C18 column and UV detection at 254nm. The Ranitidine was validated with respect to linearity, precision, accuracy, selectivity, and robustness. Themethodwas found to be applicable to routine analysis (Assay and stability tests) of active compound (Ranitidine and preservatives (MPandPP).9
A selective, sensitive and accurate high-performance liquid chromatographicmethodhas been developed, validated and applied for the determination ofranitidineandcimetidineinplasmasamples. The effects of mobile phase composition, buffer concentration, mobile phase pH and concentration of organic modifiers on retention of investigated drugs were investigated. Sample preparation was carried out by adding an internal standard, famotidine, and the clean-up procedure was accomplished using solid-phase extraction (SPE). Thismethoduses ultraviolet detection, the separation used a LichrocartLichrospher 60 RP-select B column and the mobile phase consisted of 0.2%triethylamine(TEA), 0.04 mol l−1KH2PO4at pH 6.8 and 14%acetonitrile. The recovery, selectivity, linearity, precision and accuracy of themethod were evaluated from spiked humanplasma. Themethod has been implemented to monitorranitidine levels in clinical samples.10
Literature view has revealedthat theKinetic spectrophotometric determination ofNizatidineandranitidinein pharmaceutical preparations involves the reaction of the drugs with alkalinepotassiumpermanganate, where by a green color peaking at 610 nm is produced. The reaction is monitored spectrophotometrically by measuring the rate of change of absorbance of the resulting manganate species at 610 nm. Calibration graphs are linear over the concentration range 0.8–4.0 μg/ml and the precision is quite acceptable. 11
Asimple and universal HPLC-UV method was determined and Validated, for the determination ofcimetidine,famotidine,nizatidineandranitidine inhumanurineis presented. This is the first single HPLC method reported for the analysis of all four H2antagonists inhumanbiological samples. Thismethod was also utilized for the analysis ofranitidineand its metabolites inhumanurine. All calibration curves showed good linear regression (r20.9960) within test ranges.Themethod showed good precision and accuracy with overall intra- and inter-day variations of 0.2–13.6% and 0.2–12.1%, respectively. Separation ofranitidine and its metabolites using thisassay provided significantly improved resolution, precision and accuracy compared to previously reportedmethod. It successfully applied to ahumanvolunteer study usingranitidine as the model compound.12
Literature has been revealed that on processcharacterization, optimization and validation by design of experiments approach. Powder filled sachets containing a 3:1 (W/W) powder mixture of diacetylmorphine base and caffeine anhydrate were developed as a dosage form. The powder mixture was filled into sachets using a micro dose auger filler machine. Five variables were tested: auger speed, agitator speed, hopper fill level, dose interval and dose. A 9-term regression modelwas constructedexplaining 94% of the observed variation in dose weight variation coefficient. The results of four test batches manufactured with these optimized setting showed accurate (accuracy: 99.0-101.0%) and precise (Cv:3.2-5.3%) filling of diacetylmorphine/caffeine sachets is possible using the micro dose auger filler machine.13
A Simple and sensitive capillary electrophoresismethodusing UV detection has been developed for the direct determination ofranitidine andfamotidineinserum,urineand pharmaceuticalformulations. A buffer consisting of 60 mMphosphate bufferadjusted to pH 6.5 was found to provide a very efficient and stable electrophoretic system for the analysis of both drugs. The detection limits obtained were 0.088 μgml−1forRANITIDINEand 0.16 μgml−1for
FAMOTIDINE.14
An analytical technique was developed for determining the composition of two solid forms of ranitidine hydrochloride using two peaks of Fourier transform infrared (FTIR) spectra without the need to grind the samples. Solubility studies of ranitidine hydrochloride showed that Form 2 has a higher solubility than Form 1. Solution-mediated transformation is very slowand occurs from Form 2 to Form 1 and not the reverse. No solid–solid transformation was observed due to grinding or compressing the pure samples of either forms and of a 50/50 wt. % mixture.Grinding was found to be a proper technique for increasing the bulk solid density of the ranitidine hydrochloride without therisk of solid–solid transformation. Dissolution rate found to be equally fast for both forms.15
6.3OBJECTIVE OF STUDY:
The Present Study “Development and Validation of Stability Indicating Assay Method” designed to meet the current regulatory requirements and to ensure that the products meet the predetermined specifications, quality and stability attributes.
In industry there may be scale up of batch or may alter some process or modify a process, for changes made to the processes stability testing is to be done . This stability method developed and validated ensures how stable the drug due to changes.
Following is plan of work that is designed. It mainly consists of two sections, first method development followed by validation of method.
STEP WISE DESCRIPTION OF METHOD DEVELOPMENT:
Step I: Critical study of the drug structure to assess the likely decomposition route(s).
Step II: Collection of information on physicochemical properties.
Step III: Stress ( forced decomposition)Studies
Step IV: Preliminary separation studies on stressed samples
Step V: Final method development and optimization
Step VI: Identification and characterization of degradation products, and preparation ofStandards.
VALIDATION OF METHOD:
Following parameters are to be evaluated to ensure that is method validated.
  1. Linearity
  2. Accuracy
3. Precision
a) Repeatability
b) Intermediate Precision:
c)Reproducibility
4. Range:
5. Specificity
6. Quantitation limit
7. Detection limit
8. Robustness.
7. / MATERIALS AND METHOD:
7.1 SOURCE OF DATA:
Data will be from the journals, USFDA, ICH Guidelines, WHO Guidelines, Internet, Science Direct, Articles, e-library as well as from Krupanidhi College of Pharmacy.
7.2 METHOD OF COLLECTION OF DATA:
The data will be collected on stability development and validation indicating from standard books and literature survey. The method developed is to be validated to check the variation in method conditions for robustness. The data will also be collected from methodologies which are outlined briefly as follows.
Following data to be collected during development of method:
PRELIMINARY REQUIREMENTS:
( A )SAMPLES REQUIRED FOR METHOD DEVELOPMENT:
Synthetic process with enriched impurities and individual intermediates, if available, arerequired to start the development of a selective method. A cocktail of impurities and a cocktail of the key degradants willenable the start of method development
( B) PHYSICO-CHEMICAL PROPERTIES OF THE DRUGS:
Information on the compound and the (potential) formulations is essential in helpingto frame the development of the method, primarily to determine whether HPLC/UVis appropriate (this chapter assumes that to be the case), to select the diluents and thechromatographic mode.
( C ) FUNCTIONAL GROUP EFFECTS:
Structural understanding of the compound, especially functionalgroups present that may undergo chemical transformation, is important. Guide the selection of Chromatographicconditions, including pH of the mobile phase and choice of organic modifiers.
(D) RELATED STRUCTURES
Small changes in chemical structures, whetherbackbones or functional groups, can have a profound effect on the reactivity so anyinformation on related compounds should always be used as a guide only and not asextrapolation.
METHOD DEVELOPMENT APPROACHES :
( A ) STABILITY-INDICATING CHROMATOGRAPHY CONDITIONS.
In selecting initial chromatographic conditions for a SIM of a new entity, most important is to make sure that degradants are in solution, separated, and detected. Most pharmaceuticals have a usable chromophore, allowing for UV detection. UV spectra may be different between the API and the impurities/degradants. Consideration of likely impurities and degradants as to whether they have a chromophore is important for both mass balance reasons as well as experimental setup (choice of detector(s)). At the column scouting phase, the use of a photo-diode array (PDA) detector will increase the likelihood of detecting degradants with different UV spectrum to that of the API. Alternatively, a wavelength in the lower UV range 210–254 nm may be appropriate.
( B) PEAK PURITY
Peak purity (or peak homogeneity) analysis of the main peak, to assess for thepresence of impurities under the main peak, is an essential part of the validationof a SIM. PDA only works well for degradants that have a different UV spectrum from that of the drug. Indirect evaluation of peak purity can be accomplished by changing one or morechromatographic parameters (column, mobile phase, gradient composition, etc.)that will significantly impact the separation selectivity.
In validating some critical parameters Following Conditions are to be Investigated:
( A ) HPLC CONDITIONS
  • HPLC column (lot, age, brand)
  • Mobile-phase composition (pH ± 0.05 unit, percent organic ± 2%)
  • HPLC instrument (dwell volume, detection wavelength ± 2 nm, column
( B ) SAMPLE PREPARATION
  • Sample solvent (pH ± 0.05 unit, percent organic ± 2%)
  • Sample preparation procedure (shaking time, different membrane filters
  • HPLC solution stability
7.3 DOES THE STUDY REQUIRE ANY INVESIGATIONS OF INTERVENTIONS TO BE CONDUCTED ON PATIENTS OR OTHER HUMAN OR ANIMALS? IF SO PLEASE DESCRIBE BRIEFLY?
No.
7.4 HAS ETHICAL CLEARANCE BEEN OBTAINED FROM YOUR INSTITUTE IN CASE OF AS ABOVE?
Not applicable
8. LIST OF REFERENCES:
  1. Bakshi M, Singh S. Development of validated stability-indicating assay methods- Critical review. Pharma Biomed Anal 2002;28:1011-40.
  1. DestariPratiwia, Paul Fawcetta J, Keith C. Gordonb, ThomasRades. Quantitative analysis of polymorphic mixtures of ranitidine hydrochloride by Raman spectroscopy and principal components analysis. Journal of Pharmaceutical and Biomedical Analysis 2002;54;337–341.
  1. FDA Guidance for Industry; Analytical Procedures and Methods Validation (draft), 2000.
  1. ICH Harmonized Tripartite Guideline, ICH Q2A, Text on Validation of Analytical Procedures, Mar. 1995.
  1. ICH Harmonized Tripartite Guideline ICH Q2B, Validation of Analytical Procedures: Methodology, May 1997.
  1. ICH Harmonized Tripartite Guideline ICH Q3B(R), Impurities in New Drug Products, Oct. 1999.
  1. James S. Munro, Thomas A. Walker. Ranitidine hydrochloride development of an isocratic stability indicating high-performance liquid chromatographic separation.J Chrom B. 2001;914:13–21.
  1. Vehabovic M, SabiraHadzovic, Fatima Stambolic, AminaHadzic, Elvedina Vranjes, Ediba Haracic. Stability of ranitidine in Injectable solution. Journal of Pharmaceutical and Biomedical Analysis 2003;256:109–11.
  1. XenouKokoletsiM, Kafkala.S, TsiaganisM. A novel gradient HPLC method for simultaneous determination of ranitidine, methyl paraben and propylparaben in oral liquid pharmaceutical formulation.Journal of Pharmaceutical and Biomedical Analysis 2005;38 ;763–767.
  1. DragicaZ and Stafilov.T. Development of an HPLC method for the determination of ranitidine and cimetidine in human plasma following SPE.Journal of Pharmaceutical and Biomedical Analysis 2003;33:165-173.
  1. E.M. Hassan, F. Belal. Kinetic spectrophotometric determination of nizatidine and ranitidine in pharmaceutical preparations. Journal of Pharmaceutical and Biomedical Analysis2002; 27:31–38.
  1. Diane AI,AshiruRajesh,PatelAbdul, Basit W.Simple and universal HPLC-UV method to determine cimetidine, famotidine, nizatidine, ranitidine inurine. J. Chrom B 2007;860;235-240.
  1. KlousMG, Nuijen B, Van den Brink W, Van Ree JM, Beijnen JH. Process characterization, optimization and validation of production of diacetylmorphine/caffeine sachets: a design of experiments approach. Int J Pharm. 2004 Nov 5;285(1-2);65-75
  1. Toma´sPe´rez-Ruiz, Martı´nez-Lozano V, Toma V, Bravo V, R. Galera. Direct determination of ranitidine and famotidine by CE in serum, urine and pharmaceutical formulations. Journal of Pharmaceutical and Biomedical Analysis 2002;30:1055-1061.
  1. Mirmehrabi M, Rohani S, Murthy KSK, Radatus B. Solubility, dissolution rate and phase transition studies of ranitidine hydrochloride tautomeric forms. International Journal of Pharmaceutics 2004; 282:73–85.
  1. Jens T.Carstensen, C.T. Rhodes. DrugStability Principles and Practices. 3rded. New York;Marcel Dekker, Inc; p. 329-84.

Page 1 of 12