throbber
International Journal of Pharmaceutics 360 (2008) 77–82
`
`Contents lists available at ScienceDirect
`
`International Journal of Pharmaceutics
`
`j o u r n a l h o m e p a g e : w w w . e l s e v i e r . c o m / l o c a t e / i j p h a r m
`
`Stability indicating validated HPLC method for quantification of levothyroxine
`with eight degradation peaks in the presence of excipients
`R.B. Shah a, A. Bryant a, J. Collier a,b, M.J. Habib b, M.A. Khan a,∗
`
`a Division of Product Quality Research, Office of Testing and Research, Office of Pharmaceutical Sciences, Center for Drug Evaluation and Research,
`Food and Drug Administration, United States
`b Department of Pharmaceutical Sciences, School of Pharmacy, Howard University, United States
`
`a r t i c l e
`
`i n f o
`
`a b s t r a c t
`
`Article history:
`Received 12 December 2007
`Received in revised form 20 March 2008
`Accepted 12 April 2008
`Available online 20 April 2008
`
`Keywords:
`Levothyroxine
`Impurities
`Degradation products
`Validation
`Excipients
`
`A simple, sensitive, accurate, and robust stability indicating analytical method is presented for identi-
`fication, separation, and quantitation of l-thyroxine and eight degradation impurities with an internal
`standard. The method was used in the presence of commonly used formulation excipients such as
`butylated hydroxyanisole, povidone, crospovidone, croscarmellose sodium, mannitol, sucrose, acacia,
`lactose monohydrate, confectionary sugar, microcrystalline cellulose, sodium laurel sulfate, magnesium
`stearate, talc, and silicon dioxide. The two active thyroid hormones: 3,3(cid:3),5,5(cid:3)-tetra-iodo-l-thyronine (l-
`thyroxine-T4) and 3,3(cid:3),5-tri-iodo-l-thyronine (T3) and degradation products including di-iodothyronine
`(T2), thyronine (T0), tyrosine (Tyr), di-iodotyrosine (DIT), mono-iodotyrosine (MIT), 3,3(cid:3),5,5(cid:3)-tetra-
`iodothyroacetic acid (T4AA) and 3,3(cid:3),5-tri-iodothyroacetic acid (T3AA) were assayed by the current
`method. The separation of l-thyroxine and eight metabolites along with theophylline (internal stan-
`dard) was achieved using a C18 column (25 ◦C) with a mobile phase of trifluoroacetic acid (0.1%, v/v,
`pH 3)–acetonitrile in gradient elution at 0.8 ml/min at 223 nm. The sample diluent was 0.01 M methanolic
`NaOH. Method was validated according to FDA, USP, and ICH guidelines for inter-day accuracy, preci-
`sion, and robustness after checking performance with system suitability. Tyr (4.97 min), theophylline
`(9.09 min), MIT (9.55 min), DIT (11.37 min), T0 (11.63 min), T2 (14.47 min), T3 (16.29 min), T4 (17.60 min),
`T3AA (22.71 min), and T4AA (24.83 min) separated in a single chromatographic run. Linear relationship
`(r2 > 0.99) was observed between the peak area ratio and the concentrations for all of the compounds
`within the range of 2–20 ␮g/ml. The total time for analysis, equilibration and recovery was 40 min. The
`method was shown to separate well from commonly employed formulation excipients. Accuracy ranged
`from 95 to 105% for T4 and 90 to 110% for all other compounds. Precision was <2% for all the compounds.
`The method was found to be robust with minor changes in injection volume, flow rate, column tem-
`perature, and gradient ratio. Validation results indicated that the method shows satisfactory linearity,
`precision, accuracy, and ruggedness and also stress degradation studies indicated that the method can be
`used as stability indicating method for l-thyroxine in the presence of excipients.
`Published by Elsevier B.V.
`
`1. Introduction
`
`Levothyroxine sodium pentahydrate, the sodium salt of the
`levo-isomer of thyroxine is an active physiological substance
`secreted by thyroid gland. With three ionizable moieties: carboxyl
`group (pKa = 2.4), phenolic group (pKa = 6.87) and amino group
`(pKa = 9.96), its aqueous solubility reduces from pH 1 to 3 and
`increases above pH of 7 (Patel et al., 2003). Another thyroid hor-
`
`∗ Corresponding author at: 10903 New Hampshire Avenue, Life Sciences Building
`64, Silver Spring, MD 20993-0002, United States. Tel.: +1 301 796 0132;
`fax: +1 301 796 9816.
`E-mail address: Mansoor.khan@fda.hhs.gov (M.A. Khan).
`
`0378-5173/$ – see front matter. Published by Elsevier B.V.
`doi:10.1016/j.ijpharm.2008.04.018
`
`mone, 3,3(cid:3),5-tri-iodo-l-thyronine (T3), is also pharmacologically
`active. The precursors or metabolites include di-iodothyronine (T2),
`the parent compound of the iodinated series of thyroid-active
`hormones, thyronine (T0), tyrosine (Tyr), di-iodotyrosine (DIT),
`mono-iodotyrosine (MIT) (Gika et al., 2005), as well as 3,3(cid:3),5-tri-
`iodo-l-thyroacetic acid, and 3,3(cid:3),5,5(cid:3)-tetra-iodo-l-thyroacetic acid
`have no pharmacological activity.
`Stability is considered one of the most important requirements
`of pharmaceutical product quality. Only stable preparations would
`promise precise delivery of the drug to the patients. Expiration dat-
`ing on any drug product is based upon scientific studies at normal
`and or stressed conditions of certain batches and strengths of prod-
`ucts that are developed in multiple strengths. Levothyroxine is one
`such example where products are available in multiple strengths.
`
`Mylan Ex 1016, Page 1
`
`

`
`78
`
`R.B. Shah et al. / International Journal of Pharmaceutics 360 (2008) 77–82
`
`Previous studies have shown that different dosage forms of levothy-
`roxine are susceptible to degradation under the influence of various
`environmental stress factors such as humidity and temperature.
`Won (1992) reported that levothyroxine degrades with high tem-
`perature and extremes of pH. Levothyroxine has been a subject
`of FDA Advisory Committee meetings where the clinical conse-
`quences of marketing product with approved specification limits
`of 90–110% has been reported as a problem. There were numerous
`recalls of levothyroxine due to stability issues (FDA, 2006). Further,
`lacks of potency and stability assurances has brought in concerns
`from physicians regarding their therapeutic substitutions and are
`believed not to deliver right doses to the patients (Thyroid, 2004).
`In order to understand the degradation mechanisms of levothy-
`roxine systematically, there is a need for a reliable and simple
`validated stability indicating method (ICH Q1A (R2), 2003). The
`stability indicating method should not only identify the degra-
`dation products of levothyroxine but also quantitate them. There
`are numerous reported methods to assay levothyroxine (Takahashi
`et al., 2002; Smith et al., 1981; Rapaka et al., 1981; Garnick et
`al., 1984; Richheimer and Amer, 1983). However, these methods
`either require derivatization of levothyroxine and liothyroxine for
`separation on HPLC (Takahashi et al., 2002; Smith et al., 1981)
`or lengthy and more tedious extraction–evaporation procedures
`before injecting into HPLC (Rapaka et al., 1981). There are meth-
`ods reported to assay content uniformity (Garnick et al., 1984)
`which cannot be used as stability indicating method. Richheimer
`and Amer (1983) reported a stability indicating assay method for
`levothyroxine. However, it is limited by the number of impurities.
`Quantification of impurities was not proposed in this method. Thin
`layer chromatography (TLC) has traditionally been used to identify
`degradation kinetics of levothyroxine (Won, 1992). However, it is
`not very accurate method to quantify the related compounds.
`Thus none of the previously reported methods satisfied the
`criteria of stability indicating methods. Very recently, a novel
`HPLC-based assay to quantify the impurities of levothyroxine was
`reported in the literature (Gika et al., 2005). The method included
`quantification of levothyroxine and six of its degradation products.
`However, in an attempt to reproduce the method in our laboratory,
`it was found to be erroneous in the order of mobile phase gradi-
`ent. The other limitation of the method was that the major acidic
`impurities of levothyroxine, namely, tri-iodo thyroacetic acid and
`tetra-iodo thyroacetic acid were not a part of impurity profile. The
`purpose of the current work was to modify this assay method to
`include these two impurities and also demonstrate that the assay
`was stability indicating as per FDA and ICH guidelines. Stress con-
`ditions used were high temperatures, acid and base hydrolysis,
`oxidation, and photolysis (Bakshi and Singh, 2002). Also some of the
`commonly used formulation excipients were mixed with levothy-
`roxine and eight impurities, and the chromatography was evaluated
`with a good resolution of all the peaks.
`
`2. Materials and methods
`
`l-Thyroxine sodium (l-T4) was obtained from KVPharmaceutical
`(St. Louis, MO). 3,3(cid:3),5-Tri-iodo-l-thyronine (l-T3) 3,5-di-iodo-l-
`thyronine (l-T2), 3,5-di-iodo-l-tyrosine (l-DIT), 3-iodo-l-tyrosine
`(l-MIT), l-thyronine (l-T0), l-tyrosine (l-Tyr), 3,3(cid:3),5-tri-iodo-l-
`thyroacetic acid, and 3,3(cid:3),5,5(cid:3)-tetra-iodo-l-thyroacetic acid, buty-
`lated hydroxyanisole, mannitol, sucrose, acacia, sodium laurel sul-
`fate, magnesium stearate, Inertsil 5 ␮m column, 250 mm× 4.6 mm,
`and security guard cartridge were purchased from Sigma (St.
`Louis, MO). Theophylline reagents, methanol, 0.01 M NaOH, 0.1%
`trifluoroacetic acid (TFA), Acetonitrile, and Fisherbrand low adhe-
`sion specialty tips (21-381-83) were purchased from Fisher Sci
`
`(Suwanee, GA). Povidone (BASF, Florham Park, NJ), crospovidone
`(ISP technologies Inc., Wayne, NJ), lactose monohydrate (Kerry
`BioScience, Chicago, IL), confectionary sugar (Domino’s sugar, Balti-
`more, MD), talc (Spectrum Chemicals, Gardena, CA), silicon dioxide
`(Aerosil, Evonik Degussa, Orange, CA) croscarmellose sodium, and
`microcrystalline cellulose (FMC Biopolymer, Philadelphia, PA),were
`used as received. For all studies, distilled and deionized water was
`used.
`
`2.1. Preparation of calibration standards
`
`In all cases the sample diluent used for preparing the standards
`and samples was the 0.01 M methanolic sodium hydroxide solution,
`which was prepared as described in USP monograph (USP, 2007).
`Two stock solutions (I and II) of each of nine components (T4, T3,
`T2, T0, MIT, DIT, T3AA, T4AA, and Tyr) prepared at 1000 ␮g/ml were
`prepared by dissolving them individuallly in the sample diluent.
`From the stock solution I, a working mix I was prepared by mix-
`ing 10 ml of each of these components and making the volume to
`100 ml. In a similar way working mix II was prepared from stock II.
`This was used on 3 different days but final dilutions were made on
`each day of validation. Working mix I was used for the calibration
`standards, and working mix II was used for quality control samples.
`Six different standard solutions were prepared from the working I
`to yield all the nine components in a concentration range from 2
`to 20 ␮g/ml. An internal standard, theophylline, was also added to
`all the above diluted calibration ranges. The standards were then
`transferred to an automatic injector for HPLC analysis.
`
`2.2. Preparation of quality control (QC) standards
`
`Three quality control standards were prepared from the working
`mix II to yield concentrations of 8, 10, and 12 ␮g/ml with 10 ␮g/ml
`as target concentration (100%). These were then transferred to an
`automatic injector for HPLC analysis.
`
`2.3. Preparation of resolution mixture and system suitability
`standard
`
`A combination solution containing all nine components at
`10 ␮g/ml each and theophylline was prepared from stock solution
`I and was used as system suitability standard.
`
`2.4. Chromatography
`
`HP 1100 HPLC equipment from Agilent (Wilmington, DE) con-
`sisted of quaternary pump, an automatic injector, a diode array
`wavelength detector, and a column oven. Various columns and
`mobile phases were tested. Finally, the method was validated
`with a reversed phase Inertsil ODS 2 column (250 mm× 4.6 mm,
`5 ␮m, 150 A) with a Inertsil ODS Security Guard cartridge
`(4.0 mm× 3.0 mm, 10 ␮m). It provided baseline separation with
`gradient conditions with 0.1% TFA (A) and acetonitrile (B) from 92
`to 8% A in 25 min, at 8% A from 25 to 30 min, from 8 to 92% A from
`30 to 35 min run time of 40 min for all the nine components and
`IS in a single chromatographic run. The flow rate was 0.8 ml/min,
`column temperature was 25 ◦C and the injection volume was 50 ␮l.
`The UV detection wavelength was set at 215, 223, 228, 232, and 240.
`However, all the calculations were performed at 223 nm.
`
`2.5. Validation
`
`Validation was carried out according to ICH and FDA guidelines
`for chromatographic methods (Bakshi and Singh, 2002). Speci-
`ficity, selectivity, linearity, accuracy, precision, and robustness were
`
`Mylan Ex 1016, Page 2
`
`

`
`R.B. Shah et al. / International Journal of Pharmaceutics 360 (2008) 77–82
`
`79
`
`established for the method. System suitability and resolution was
`performed utilizing related compound C and ranitidine HCl as the
`standards.
`
`2.6. Stress degradation studies
`
`Stress conditions applied for degradation of levothyroxine pow-
`der include refluxing it (1 mg) at room temperature under acidic
`(0.1N HCl, 24 h) and alkaline (0.1N NaOH, 24 h) conditions, oxida-
`tion (3% hydrogen peroxide, 24 h), and photolysis (exposure to UV-A
`and UV-B rays). Also the degradation was carried out at 40 ◦C for a
`period of 14 h. All these samples were appropriately diluted with
`sample diluent and injected into the HPLC.
`
`2.7. Excipient analysis
`
`Some of the commonly used formulation excipients were
`selected based on the commercial product inserts. They included
`butylated hydroxyanisole, povidone, crospovidone, croscarmellose
`sodium, mannitol, sucrose, acacia, lactose monohydrate, confec-
`tionary sugar, microcrystalline cellulose, sodium laurel sulfate,
`magnesium stearate, talc, and silicon dioxide. Initially, all the excip-
`ients were diluted in the sample diluent, filtered, and analyzed
`on HPLC for detection and evaluating their retention times. Fol-
`lowing that, l-thyroxine along with internal standard and all the
`eight degradation compounds were mixed with the excipient in
`1:1 ratios and were analyzed on HPLC as described earlier. The
`detector was set at multiple wavelengths of 215, 223, 228, 232,
`and 240 so as to ensure non-interference of excipients with either
`the active pharmaceutical ingredient (API), internal standard or
`impurities.
`
`3. Results and discussion
`
`3.1. Analytical method development
`
`Levothyroxine products have a history of stability failures which
`could result into sub-potency products with patients receiving less
`than optimal dose. The problem is aggravated while switching
`from one product to another although they are listed as ther-
`apeutic equivalents. To understand mechanism of levothyroxine
`degradation, an assay which will not only identify but quan-
`tify the impurities is essential. A reported method (Gika et al.,
`2005) was corrected and used with modifications to add two
`additional degradation products of levothyroxine, T3AA and T4AA.
`These two are considered to be significant degradation prod-
`ucts for levothyroxine. Five different wavelengths were used to
`observe the chromatograms, but only 223 nm was used for calcu-
`lation purpose. Although the peak areas were highest at 215 nm,
`the baseline showed very high negative drift. The wavelength
`of 223 also showed comparatively higher peak area for all the
`impurities as well as levothyroxine compared to 228, 232, and
`240 nm. Therefore, that wavelength was used. However, the detec-
`tion at all wavelengths was continued considering that stability
`samples might show some impurities which might be detected
`at one wavelength as opposed to other. Fig. 1 depicts the chro-
`matogram obtained with the current method. The peaks of all
`the impurities and levothyroxine were well resolved. This method
`was further validated as given in Section 2.7. The order of elu-
`tion was Tyr (4.97 min), theophylline (9.09 min), MIT (9.55 min),
`DIT (11.37 min), T0 (11.63 min), T2 (14.47 min), T3 (16.29 min), T4
`(17.60 min), T3AA (22.71 min), and T4AA (24.83 min). The current
`method can be used to assay levothyroxine and its major degrada-
`tion products.
`
`Fig. 1. Typical chromatogram of the iodothyronines and iodotyrosines separation
`using the HPLCs. All analytes are at 10 ␮g/ml. Peaks: Tyr (4.97 min), theophylline
`(9.09 min), MIT (9.55 min), DIT (11.37 min), T0 (11.63 min), T2 (14.47 min), T3
`(16.29 min), T4 (17.60 min), T3AA (22.71 min), and T4AA (24.83 min).
`
`3.2. Analytical method validation
`
`Specificity was established by determining that levothyroxine,
`internal std and degradation products have no co-eluting peaks
`in preparative solvents, mobile phase (blanks), or related matrices
`(Fig. 1).
`Selectivity was tested by running solutions containing the eight
`impurities and one internal standard in the same quantities and
`conditions as the samples to show that there was no peak at the
`retention times corresponding to the API.
`The detection limit (LOD) was 1 ␮g/ml which was evaluated by
`measuring the baseline noise and by calculating the analyte con-
`centration that gave S/N = 3, while the limit of quantification (LOQ)
`was 2 ␮g/ml which was established for the analyte concentration
`that gave S/N = 10.
`Linearity was established across the analytical calibration range.
`At least five non-zero calibration standards and a zero calibration
`standard and or blanks were utilized for each calibration curve.
`Table 1 shows the calibration curves of levothyroxine and all eight
`degradation compounds on 3 different days showing a linear corre-
`lation with R2 > 0.99 for all the components. Range was established
`by demonstrating a suitable level of accuracy, precision, and linear-
`ity.
`
`Accuracy and precision of the analytical method was established
`across its analytical range (Table 2). The accuracy was measured at
`each quality control (QC) standard level (n = 3) over the analyti-
`cal range as defined by the 80% of target concentration (8 ␮g/ml),
`l00% of target concentration (10 ␮g/ml), and 120% of target concen-
`tration (12 ␮g/ml), against the calibration curve. The levels were
`selected based on FDA and ICH guidelines (FDA, 1994; ICH Q2 (R1),
`1995). Nominal values are no greater than 15% at the LLOQ and 10%
`at the low, intermediate and high QC levels.
`
`Table 1
`Linearity and sensitivity data
`
`Analyte
`
`Calibration range (␮g/ml)
`
`Tyr
`MIT
`DIT
`T0
`T2
`T3
`T4
`T3AA
`T4AA
`
`2–20
`2–20
`2–20
`2–20
`2–20
`2–20
`2–20
`2–20
`2–20
`
`Slope
`0.051 ± 0.004
`0.124 ± 0.008
`0.192 ± 0.014
`0.149 ± 0.009
`0.183 ± 0.013
`0.187 ± 0.012
`0.157 ± 0.011
`0.208 ± 0.014
`0.192 ± 0.014
`
`Intercept
`0.016 ± 0.008
`0.015 ± 0.002
`0.009 ± 0.012
`0.002 ± 0.003
`0.008 ± 0.009
`0.010 ± 0.003
`0.014 ± 0.006
`0.007 ± 0.003
`0.012 ± 0.010
`
`R2
`
`0.9992
`0.9999
`1.0000
`0.9998
`0.9999
`0.9998
`0.9999
`1.0000
`1.0000
`
`Mylan Ex 1016, Page 3
`
`

`
`80
`
`R.B. Shah et al. / International Journal of Pharmaceutics 360 (2008) 77–82
`
`Table 2
`Accuracy and precision data
`
`Inter-day (n = 9)
`
`80
`
`100.09 ± 3.04
`102.18 ± 1.34
`100.19 ± 2.85
`98.19 ± 3.18
`99.46 ± 3.32
`99.26 ± 3.28
`100.25 ± 2.96
`97.30 ± 2.91
`103.61 ± 3.02
`
`Intra-day (n = 3)
`
`80
`
`96.38 ± 0.24
`100.52 ± 0.13
`96.40 ± 0.07
`94.08 ± 0.05
`95.04 ± 0.03
`94.94 ± 0.34
`96.31 ± 0.00
`93.55 ± 0.04
`99.59 ± 0.03
`
`Accuracy (%)
`Tyr
`MIT
`DIT
`T0
`T2
`T3
`T4
`T3AA
`T4AA
`
`Precision (%)
`Tyr
`MIT
`DIT
`T0
`T2
`T3
`T4
`T3AA
`T4AA
`
`100
`
`120
`
`100.72 ± 0.56
`100.57 ± 0.12
`99.82 ± 0.11
`97.36 ± 0.09
`98.39 ± 0.05
`98.20 ± 0.28
`99.91 ± 0.10
`96.86 ± 0.20
`103.22 ± 0.15
`
`101.62 ± 1.33
`100.57 ± 0.02
`100.53 ± 0.07
`98.25 ± 0.10
`99.21 ± 0.10
`100.01 ± 0.41
`100.71 ± 0.08
`97.58 ± 0.06
`103.90 ± 0.05
`
`Intra-day (n = 6)
`
`0.94
`0.12
`0.08
`0.09
`0.08
`0.29
`0.10
`0.16
`0.12
`
`100
`
`120
`
`100.63 ± 1.47
`100.62 ± 0.72
`100.04 ± 0.45
`98.19 ± 1.35
`99.43 ± 0.98
`99.31 ± 1.27
`100.38 ± 0.60
`97.29 ± 1.07
`103.55 ± 0.51
`
`102.92 ± 2.21
`101.28 ± 0.54
`101.00 ± 0.46
`99.28 ± 0.98
`100.47 ± 0.95
`100.59 ± 0.60
`101.41 ± 0.56
`98.29 ± 0.68
`104.52 ± 0.53
`
`Inter-day (n = 18)
`
`1.48
`0.70
`0.43
`1.33
`0.95
`1.22
`0.58
`1.07
`0.47
`
`The system suitability standard contains levothyroxine, inter-
`nal standard, and all eight degradation products. Table 3 depicts
`the system suitability and resolution factors. The specifications are
`also given in Table 3. It was observed that all the parameters passed
`the USP specifications. An important system suitability parameter
`is resolution, a measure of how well two peaks are separated. For a
`reliable quantification, well-separated peaks are essential. This is a
`
`very useful parameter if potential interference peak may be of con-
`cern. The degradation impurities of levothyroxine were selected to
`measure the resolution parameter. It is desirable to have resolu-
`tion of >1 between the two peaks. With the current method, we
`obtained satisfactory resolution of >1 in all cases. The tailing fac-
`tor was also considered as the accuracy of quantification decreases
`with increase in peak tailing because of the difficulties encountered
`
`Table 3
`System suitability parameters
`
`RT %R.S.D.
`
`Peak area %R.S.D.
`
`USP tailing
`1.00 ± 0.00
`1.01 ± 0.01
`1.00 ± 0.00
`1.07 ± 0.03
`1.10 ± 0.01
`1.07 ± 0.01
`1.23 ± 0.00
`1.17 ± 0.00
`1.17 ± 0.00
`1.22 ± 0.00
`1.16 ± 0.00
`1.15 ± 0.00
`1.32 ± 0.00
`1.25 ± 0.00
`1.25 ± 0.01
`1.35 ± 0.01
`1.29 ± 0.00
`1.31 ± 0.01
`1.38 ± 0.01
`1.34 ± 0.00
`1.38 ± 0.01
`1.30 ± 0.01
`1.24 ± 0.00
`1.26 ± 0.00
`1.29 ± 0.01
`1.23 ± 0.08
`1.26 ± 0.00
`
`Theoretical plates (X05)
`3.03 ± 0.04
`2.54 ± 0.05
`3.07 ± 0.05
`1.64 ± 0.02
`1.79 ± 0.02
`1.34 ± 0.05
`2.54 ± 0.01
`2.61 ± 0.02
`2.55 ± 0.02
`2.78 ± 0.02
`2.84 ± 0.03
`2.78 ± 0.00
`3.73 ± 0.03
`3.72 ± 0.06
`3.56 ± 0.02
`4.26 ± 0.07
`4.16 ± 0.04
`3.93 ± 0.03
`4.37 ± 0.06
`4.18 ± 0.07
`3.90 ± 0.03
`5.76 ± 0.09
`5.64 ± 0.06
`5.40 ± 0.05
`6.38 ± 0.09
`6.18 ± 0.08
`5.93 ± 0.02
`
`Resolution
`
`Selectivity
`
`1.82 ± 0.04
`1.05 ± 0.00
`1.03 ± 0.00
`1.20 ± 0.00
`1.19 ± 0.00
`1.20 ± 0.00
`1.02 ± 0.00
`1.02 ± 0.00
`1.02 ± 0.00
`1.25 ± 0.00
`1.24 ± 0.00
`1.25 ± 0.00
`1.13 ± 0.00
`1.12 ± 0.00
`1.13 ± 0.00
`1.08 ± 0.00
`1.08 ± 0.00
`1.08 ± 0.00
`1.30 ± 0.00
`1.30 ± 0.00
`1.29 ± 0.00
`1.09 ± 0.00
`1.09 ± 0.00
`1.09 ± 0.00
`
`1.03 ± 0.00
`2.49 ± 0.05
`1.55 ± 0.02
`10.3 ± 0.05
`10.1 ± 0.07
`10.3 ± 0.04
`1.54 ± 0.02
`1.48 ± 0.01
`1.53 ± 0.01
`15.9 ± 0.08
`15.6 ± 0.14
`15.6 ± 0.06
`9.52 ± 0.06
`9.30 ± 0.10
`9.18 ± 0.04
`6.43 ± 0.04
`6.21 ± 0.07
`6.14 ± 0.03
`23.6 ± 0.29
`22.8 ± 0.10
`22.2 ± 0.14
`8.97 ± 0.06
`8.69 ± 0.05
`8.58 ± 0.03
`
`Day-1
`Day-2
`Day-3
`
`Day-1
`Day-2
`Day-3
`
`Day-1
`Day-2
`Day-3
`
`Day-1
`Day-2
`Day-3
`
`Day-1
`Day-2
`Day-3
`
`Day-1
`Day-2
`Day-3
`
`Day-1
`Day-2
`Day-3
`
`Day-1
`Day-2
`Day-3
`
`Day-1
`Day-2
`Day-3
`
`Tyr
`
`MIT
`
`DIT
`
`T0
`
`T2
`
`T3
`
`T4
`
`T3AA
`
`T4AA
`
`Spec
`
`0.05
`0.05
`0.08
`
`0.15
`0.23
`0.13
`
`0.09
`0.17
`0.05
`
`0.21
`0.16
`0.05
`
`0.26
`0.09
`0.03
`
`0.03
`0.04
`0.04
`
`0.02
`0.02
`0.04
`
`0.05
`0.07
`0.04
`
`0.04
`0.06
`0.04
`
`1.38
`1.97
`1.02
`
`0.24
`0.07
`0.18
`
`0.23
`0.09
`0.13
`
`0.24
`0.09
`0.11
`
`0.11
`0.14
`0.10
`
`0.16
`0.15
`0.27
`
`0.08
`0.12
`0.09
`
`0.09
`0.06
`0.12
`
`0.05
`0.10
`0.10
`
`<2
`
`<2
`
`<2
`
`>1
`
`>1
`
`>1
`
`Mylan Ex 1016, Page 4
`
`

`
`R.B. Shah et al. / International Journal of Pharmaceutics 360 (2008) 77–82
`
`81
`
`Table 4
`Robustness with flow rate variation (nominal was 0.8 ml/min)
`
`Flow rate (ml/min)
`
`RT %R.S.D.
`
`Peak area %R.S.D.
`
`0.7
`0.9
`
`0.7
`0.9
`
`0.7
`0.9
`
`0.7
`0.9
`
`0.7
`0.9
`
`0.7
`0.9
`
`0.7
`0.9
`
`0.7
`0.9
`
`0.7
`0.9
`
`Tyr
`
`MIT
`
`DIT
`
`T0
`
`T2
`
`T3
`
`T4
`
`T3AA
`
`T4AA
`
`Spec
`
`0.03
`0.05
`
`0.04
`0.06
`
`0.02
`0.07
`
`0.01
`0.07
`
`0.02
`0.04
`
`0.01
`0.03
`
`0.01
`0.03
`
`0.01
`0.02
`
`0.01
`0.02
`
`<2
`
`0.83
`1.51
`
`0.19
`0.16
`
`0.07
`0.19
`
`0.04
`0.12
`
`0.05
`0.09
`
`0.03
`0.05
`
`0.11
`0.20
`
`0.06
`0.05
`
`0.10
`0.08
`
`<2
`
`in calculating the area under the peak. A desirable USP tailing factor
`is <2 which is consistent with the factor obtained with the current
`method (Table 3).
`Robustness was established by analyzing the system suitability
`standard (n = 6) at 20 and 30 ◦C (nominal = 25 ◦C), at flow rates of 90
`and 110% of the nominal flow (i.e., 1 ml/min.) and injector volumes
`at 50 and 150% of system suitability standard injection volume. Also
`since gradient method was used, a slight variation in gradient was
`also evaluated which included 93% (A) −7% (B) and 91% (A) −9%
`(B). Table 4 shows the results obtained for robustness of the sys-
`tem. A low CV (%) indicates that the system was robust and could
`be used without any problem if a minor change is to occur. The
`determination of robustness or ruggedness is especially important
`for gradient elution systems which might be impacted significantly
`by minor variations due to gradient ratio, temperature or other fac-
`tors. However the current method was found to be robust for such
`minor changes as demonstrated in Table 4 for flow rate variation.
`The specifications were met for all of the peaks under minor flow
`rate condition. Similar data was obtained for minor variations in
`injection volume, gradient, as well as column temperature (data
`not shown).
`
`3.3. Stress degradation studies
`
`Stress studies were carried out following an ICH guideline which
`establishes the requirements of stability indicating methods. A vari-
`ety of conditions, such as pH, light, oxidation, dry heat, etc. were
`applied and separation of drug from the degradation products was
`observed in the chromatograms. Similar studies are carried out in
`the literature for many drugs (Bakshi et al., 2004; Ojha et al., 2003).
`However all the literature methods fall short in meeting the current
`regulatory requirements for levothyroxine assay methods (Garnick
`et al., 1984; Graham et al., 1974; Rapaka et al., 1981). Therefore, the
`current work comprised of performing forced degradation studies
`to establish suitability of the method as stability indicating. Another
`ICH guideline on stability of testing of new drug substances and
`products (ICH Q1A (R2), 2003) advocates the use of stability testing
`assay methods for highly susceptible drugs such as levothyroxine.
`
`USP tailing
`0.94 ± 0.02
`0.95 ± 0.03
`1.06 ± 0.01
`1.00 ± 0.00
`1.10 ± 0.00
`1.15 ± 0.01
`1.21 ± 0.01
`1.14 ± 0.00
`1.27 ± 0.00
`1.23 ± 0.00
`1.31 ± 0.00
`1.28 ± 0.01
`1.40 ± 0.00
`1.35 ± 0.01
`1.29 ± 0.01
`1.24 ± 0.01
`1.27 ± 0.01
`1.23 ± 0.01
`
`Theoretical plates (X05)
`3.58 ± 0.02
`2.18 ± 0.06
`1.87 ± 0.00
`1.46 ± 0.01
`2.97 ± 0.01
`2.44 ± 0.02
`2.73 ± 0.02
`2.71 ± 0.03
`3.63 ± 0.05
`3.61 ± 0.04
`3.99 ± 0.05
`4.07 ± 0.05
`3.89 ± 0.04
`4.07 ± 0.06
`5.27 ± 0.03
`5.67 ± 0.07
`5.74 ± 0.05
`6.21 ± 0.06
`
`Resolution
`
`Selectivity
`
`1.03 ± 0.00
`1.04 ± 0.00
`1.02 ± 0.00
`1.22 ± 0.00
`1.18 ± 0.00
`1.03 ± 0.00
`1.23 ± 0.00
`1.27 ± 0.00
`1.12 ± 0.00
`1.13 ± 0.00
`1.08 ± 0.00
`1.08 ± 0.00
`1.29 ± 0.00
`1.30 ± 0.00
`1.09 ± 0.00
`1.10 ± 0.00
`
`1.81 ± 0.01
`1.76 ± 0.01
`1.31 ± 0.00
`10.69 ± 0.07
`9.80 ± 0.02
`1.70 ± 0.01
`14.55 ± 0.12
`16.56 ± 0.10
`8.66 ± 0.07
`9.79 ± 0.08
`5.88 ± 0.03
`6.48 ± 0.06
`21.61 ± 0.10
`23.06 ± 0.19
`8.31 ± 0.03
`8.96 ± 0.05
`
`<2
`
`>1
`
`>1
`
`>1
`
`In the current study, stress decomposition studies at temperatures
`in 40 ◦C increments above the accelerated temperature, extremes
`of pH and under oxidative and photolytic conditions were carried
`out on the drug substance, levothyroxine. The suitability of the
`proposed analytical method as a stability indicating method was
`supported by these stress degradation studies. Fig. 2 represents
`the stress degradation of the drug substance and the drug product
`in acidic, alkaline, oxidative, and photolytic (UVA and UVB) con-
`ditions, respectively. It was observed that the degraded products
`eluted far from the drug peak in case of oxidized sample where the
`degradation peaks were well separated from l-thyroxine peak. The
`main degradation compound formed by oxidation was observed at
`a retention time of 22.9 min which corresponds to T3AA. Thus it was
`even possible to identify the degradation compounds of l-thyroxine
`under the stress condition. There was no degradation observed with
`acidic, alkaline or with UV exposure under the conditions specified.
`Basic pH condition was found to enhance l-thyroxine pentahydrate
`stability in one of the studies (Patel et al., 2003).
`
`Fig. 2. Stres-degradation samples of levothyroxine with acid, alkali, oxidation, UV-
`A, and UV-b conditions (expanded view). The degradation compounds were seen
`under oxidation condition which were well separated from l-thyroxine peak. The
`primary degradation peak with retention time of 22.9 min corresponded to T3AA
`impurity. There was no degradation observed under acidic, alkaline, or photolysis
`conditions.
`
`Mylan Ex 1016, Page 5
`
`

`
`82
`
`R.B. Shah et al. / International Journal of Pharmaceutics 360 (2008) 77–82
`
`iodo-l-tyrosine (l-MIT), l-thyronine (l-T0), and l-tyrosine (l-Tyr).
`The method employed is under gradient condition in 40 min of
`total run. The method has been validated and it has been shown
`that it is reliable, linear, and precise both in upper and lower
`concentration range as well as robust with minor variations in
`chromatographic parameters. Therefore, it can be applied for quan-
`tification of the active compound and all of the eight degradation
`compounds. Excipient analysis study indicated that the method
`was found to separate l-thyroxine from the commonly used for-
`mulation excipient, butylated hydroxyanisole. The other excipients
`did not interfere with the analysis as they did not elute in the chro-
`matographic run.
`
`Acknowledgements
`
`Authors wish to express their sincere thanks to Everett Jefferson
`and Anthony Ciavarella from DPQR for their invaluable help.
`Disclaimer. The opinions expressed in this work are only of
`authors, and do not necessarily reflect the policy and statements
`of the FDA.
`
`References
`
`Bakshi, M., Singh, S., 2002. Development of validated stability-indicating assay
`methods—critical review. J. Pharm. Biomed. Anal. 28, 1011–1040.
`Bakshi, M., Ojha, T., Singh, S., 2004. Validated specific HPLC methods for deter-
`mination of prazosin, terazosin and doxazosin in the presence of degradation
`products formed under ICH-recommended stress conditions. J. Pharm. Biomed.
`Anal. 34, 19–26.
`FDA CDER (1994). Reviewer Guidance, Validation of chromatographic methods. FDA.
`FDA, 2006. Regulatory History and Current Issues. Dept. Health Human Ser.
`Garnick, R.L., Burt, G.F., Long, D.A., Bastian, J.W., Aldred, J.P., 1984. High-performance
`liquid chromatographic assay for sodium levothyroxine in tablet formulations:
`content uniformity applications. J. Pharm. Sci. 73, 75–77.
`Gika, H.G., Samanidou, V.F., Papadoyannis, I.N., 2005. Development of a validated
`HPLC method for the determination of iodotyrosines and iodothyronines in phar-
`maceuticals and biological samples using solid phase extraction. J. Chromatogr.
`B: Anal. Technol. Biomed. Life Sci. 814, 163–172.
`Graham, J.H., Banes, D., Biesemeyer, M.E., Nadkarni, A., 1974. Analysis of sodium
`levothyroxine or sodium liothyronine in tablets. J. Pharm. Sci. 63, 763–766.
`ICH Q2 (R1), 1995. Validation of analytical procedures: text and methodology. In:
`Proceedings of the International Conference on Harmonization.
`ICH Q1A (R2), 2003. Stability testing of new drug substances and products. In: Pro-
`ceedings of the International Conference on Harmonization.
`Ojha, T., Bakshi, M., Chakraborti, A.K., Singh, S., 2003. The ICH guidance in prac-
`tice: stress decomposition studies on three piperazinyl quinazoline adrenergic
`receptor-blocking agents and comparison of their degradation behaviour. J.
`Pharm. Biomed. Anal. 31, 775–783.
`Patel, H., Stalcup, A., Dansereau, R., Sakr, A., 2003. The effect of excipients on the
`stability of levothyroxine sodium pentahydrate tablets. Int. J. Pharm. 264, 35–43.
`Rapaka, R.S., Knight, P.W., Prasad, V.K., 1981. Reversed-phase high-performance liq-
`uid chromatographic analysis of liothyronine sodium and levothyroxine sodium
`in tablet formulations: preliminary studies on dissolution and content unifor-
`mity. J. Pharm. Sci. 70, 131–134.
`Richheimer, S.L., Amer, T.M., 1983. Stability-indicating assay, dissolution, and content
`uniformity of sodium levothyroxine in tablets. J. Pharm. Sci. 72, 1349–1351.
`Smith, D.J., Biesemeyer, M., Yaciw, C., 1981. The separation and determination of
`liothyronine and levothyroxine in tablets by reversed-phase high performance
`liquid chromatography. J. Chromatogr. Sci. 19, 72–78.
`Takahashi, M., Nagashima, M., Shigeoka, S., Kamimura, H., Kamata, K., 2002. Determi-
`nation of thyroid hormones in pharmaceutical preparations, after derivatization
`with 9-anthroylnitrile, by high-performance liquid chromatography with fluo-
`rescence detection. J. Chromatogr. A 958, 299–303.
`Thyroid Foundation of America, 2004. Thyroid Patient Advocacy Group Warns Thy-
`roid Patients of Potential Problems with Thyroid Drug Resulting From FDA
`Approval of Generic Versions. All Thyroid Org.: Thyroid Issues in the News.
`USP monograph, 2007 Levothyroxine Sodium. United States Pharmacopeia.
`Won, C.M., 1992. Kinetics of degradation of levothyroxine in aqueous solution and
`in solid state. Pharm. Res. 9, 131–137.
`
`Fig. 3. Expanded view of chromatograph of l-thyroxine degradation compounds
`(T3AA and T4AA) with formulation excipient, BHA. The retention times are T3AA
`(22.71 min), BHA (23.36 min) and T4AA (24.83 min).
`
`3.4. Excipient analysis
`
`The current method was used to assay various excipients which
`are commonly used in l-thyroxine formulations. The information
`was obtained from package insert or label of currently marketed
`lev

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket