You are on page 1of 9

Chromatographia

DOI 10.1007/s10337-014-2699-4

Original

Multiple Response Optimization of a HPLC Method


for the Determination of Enantiomeric Purity of S‑Ofloxacin
Valliappan Kannappan · Sai Sandeep Mannemala 

Received: 28 February 2014 / Revised: 21 April 2014 / Accepted: 18 May 2014


© Springer-Verlag Berlin Heidelberg 2014

Abstract  The aim of the study was to develop a new Keywords  Column liquid chromatography · Ofloxacin
HPLC method for direct chiral separation of Ofloxacin enantiomers · Chiral HPLC separation · Central composite
enantiomers using polar non-aqueous mobile phase by design · Polysaccharide chiral stationary phase ·
application of response surface methodology. Rotatable Chromatographic optimization function
central composite design (CCD) with eight factorial points,
six axial points and six replications in central point was
used to evaluate the influence of three independent vari- Introduction
ables (concentration of methanol, diethylamine and flow
rate) on the output responses (capacity factor of first peak, Chiral drugs constitute one-third of the worldwide marketed
tailing factors of both the enantiomers, resolution between drugs. It is now well documented that the enantiomers of
the Ofloxacin enantiomers, retention time of the last peak chiral drugs exhibit different pharmacological and toxico-
and chromatographic optimization function). Further, CCD logical properties [1]. Therefore, the determination of enan-
data were combined with multiple response optimiza- tiomeric purity of chiral drugs is important from the point of
tion in order to obtain a set of optimal experimental con- view of safety and efficacy of drug therapy [2]. Ofloxacin
ditions (% methanol/hexane/acetonitrile-43.33/10/46.62 (Fig.  1) designated as [(±)-9-fluoro-2,3-dihydro-3-methyl-
(v/v), % acetic acid/diethylamine-0.4/0.2 and flow rate as 10-(4-methyl-1-piperazinyl)-7-oxo-7H-pyridol(1,2,3-de)-
1.4 mL min−1) leading to the most desirable compromise 1,4-benzoxazine-6-carboxylic acid] is a second-generation
between resolution and analysis time. The method demon- broad spectrum antibiotic. Ofloxacin possesses one stereo-
strated good correlation between observed and predicted genic center in the oxazidine ring and exists in two enan-
responses. The developed method was validated according tiomeric forms. Levofloxacin (S-OFL) is the S-enantiomer
to ICH guidelines and applied for quantitative analysis of of Ofloxacin, the eutomer, which shows 8-fold to 128-fold
two commercially available tablets Zenoflox (Ofloxacin) higher activity than its R-enantiomer (R-OFL) with differ-
and Glevo (Levofloxacin). Good agreement was found ent in vitro bacterial strains [3]. Levofloxacin products may
between the assay results and the label claim of the mar- contain traces of the R-OFL, the distomer, residual from
keted formulations by showing good %recovery and %CV. Levofloxacin synthesis. Hence, it is necessary to identify and
The study resulted in a better chromatographic system for determine both the Ofloxacin enantiomers for chiral quality
the determination of Ofloxacin enantiomers. control. The monograph of n is official in Indian Pharmaco-
poeia [4], British Pharmacopoeia [5] and USP NF 32 [6]. It
is available in various dosage forms, viz. tablets, eye drops,
infusion, suspension and capsules in Indian market.
The determination of n enantiomers in pharmaceutical dos-
V. Kannappan (*) · S. S. Mannemala  age form and biological matrices was performed by capillary
Department of Pharmacy, Faculty of Engineering
electrokinetic chromatography [7], capillary electrophoresis
and Technology, Annamalai University, Annamalai Nagar,
Chidambaram 608002, Tamil Nadu, India [8], ligand exchange methods [9–11]. Also a semiprepara-
e-mail: vallikonnect@gmail.com tive method by employing immobilized β-cyclodextrin chiral

13
V. Kannappan, S. S. Mannemala

Fig. 1  Structure of n enantiom-
ers

stationary phase (CSP) is reported [12]. However, an inten- coated on silica particles (5 µm) from Phenomenex (Phe-
sive literature survey revealed that there are three direct chi- nomenex, USA) was used. Absorbance spectra were recorded
ral analysis HPLC methods reported for determination of n using an UV–Visible spectrophotometer (Model UV-1601PC;
enantiomers. Lehr et al. [13] and Rabba et al. [14] developed Japan) using quartz cell of 1.00 cm of path length.
methods based on bovine serum albumin (BSA) CSP which
suffers from limitation of usage of delicate column. Sun et al. Software
[15] proposed a method based on Chiralcel OD-H column,
which has major weakness like excess analysis time (20 min), Experimental design, data analysis and desirability func-
low sensitivity (LOD = 10 µg/L) and a poor resolution tion calculations were performed by using JMP®, 9.0 trial
(Rs  = 1.3) between the enantiomers. Further, none of these version (SAS Institute Inc., Cary, NC) [16]. Perturbation
methods utilized a systematic optimization approach (viz. plot was generated using Design expert®, 8.0 version (Stat-
chemometric methods) for the separation and quantitation of Ease, MN, USA). The rest of the calculations for the anal-
the n enantiomers, but employed a time-consuming trial and ysis were performed by the use of Microsoft Excel 2010
error approach which will result only in an apparent optimum software (Microsoft, USA).
and information regarding the sensitivity of the factors on the
analytes separation and interaction between the factors is not Chemicals and Reagents
available. All the above indicated direct chiral HPLC meth-
ods suffer from one or the other limitation and may not be Working standards of Ofloxacin (±)-Ofloxacin (98.9 %)
suitable for routine quality control purpose. Hence, this study was donated by East India Pharmaceuticals (Kolkata, India),
attempts to develop an improved HPLC method for the deter- Levofloxacin (99.7 %) was a gift sample from M/S Phar-
mination of Ofloxacin enantiomers using chemometric proce- maceuticals (Puducherry, India) and probenecid, internal
dure and also demonstrate the utility of the developed method standard (IS) (99.8 %), was purchased from Sigma-Aldrich
by analyzing two commercially available tablets Zenoflox (Bangalore, India); acetonitrile (MeCN), methanol (MeOH)
(Ofloxacin) and Glevo (:Levofloxacin). of HPLC grade and acetic acid (AcOH) and diethyl amine
(DEA) of analytical reagent grade were procured from M/S
SD Fine Chemicals (Mumbai, India). The HPLC grade water
Experimental was prepared by using Milli-Q Academic, Millipore (Banga-
lore, India). The tablets Zenoflox (Ofloxacin, 200 mg) from
Apparatus Mankind Pharma Ltd (Mumbai, India) and Glevo (Levoflox-
acin 500 mg) from Glenmark Pharmaceuticals Ltd (Chennai,
The chromatographic system from Shimadzu (Shimadzu Cor- India) was procured from local pharmacy.
poration, Kyoto, Japan) was used for the analysis which con-
sisted of two LC 20 AD solvent delivery modules, a SPD-M Preparation of Solutions and Chromatographic Conditions
20A PDA detector and a rheodyne injector (model 7125,
USA) valve fitted with a 20-µL loop. The system was con- Stock and Working Standard Solutions
trolled through a system controller (SCL-10A) and a personal
computer using a Shimadzu chromatographic software (LC Stock standard solutions of (±)-Ofloxacin, S-OFL and
Solution, Release 1.11SP1) installed on it. The mobile phase probenecid (IS) 1,000 (µg mL−1) were prepared in mobile
was degassed using Branson sonicator (Branson Ultrasonics, phase. The prepared stock solutions were stored at 4 °C pro-
USA). A Lux Cellulose-4 column (250 × 4.6 mm, ID) packed tected from light. Working standard solutions were freshly
with cellulose tris (4-chloro-3-methylphenylcarbamate) prepared by diluting the stock standard solutions with mobile

13
Multiple Response Optimization of a HPLC Method

phase during the day of analysis. Calibration curves report- separation and to understand the interaction of selected fac-
ing peak area ratios of S-OFL and R-OFL to that of the IS tors on separation attributes. The selection of key factors
versus drug concentrations were established in the range for optimization was based on preliminary experiments and
of 2–10 µg mL−1 for all the analytes in the presence of prior knowledge from literature [18–21]. For instance, the
probenecid (10 µg mL−1) as IS. The standard solution pre- concentration of hexane was fixed at 10 % v/v and the con-
pared for the optimization procedure comprised Ofloxacin centration of AcOH was fixed at 0.4 % v/v in the mobile
(10 µg mL−1), S-OFL (10 µg mL−1) and IS (10 µg mL−1). phase as it aided in maintaining a better peak shape for
both enantiomers. So, the factors selected for optimization
Preparation of Sample Solution process were % MeOH concentration (A), % DEA concen-
tration (B) and flow rate (C). The factor space of this design
Twenty tablets were weighed and finely powdered. An was expanded with in the following range: MeOH concen-
amount of tablet powder equivalent to 20 mg of (±)-Ofloxa- tration was varied from 40 to 60 % v/v, DEA concentration
cin was accurately weighed and transferred in a 25-mL vol- from 0.2 to 0.3 % and flow rate from 1.0 to 1.4 mL min−1
umetric flask; a suitable quantity of IS (25 mg) was added by keeping concentration of hexane (10 % v/v), AcOH
followed by 12 mL of mobile phase. This mixture was sub- (0.4 % v/v) constant and % MeCN constituting the rest of
jected to sonication for 10 min for complete extraction of the volume of mobile phase. In order to judge the quality
drugs and the solution was made up to the mark with mobile of the method under different conditions, the following
phase and further dilutions were made to obtain a concen- responses of interest were defined (1) capacity factor of
tration of (±)-Ofloxacin and IS as 8.0 and 10.0 µg mL−1, the first eluted peak (k1), (2) tailing factor of the 2nd peak,
respectively. The solution was centrifuged at 4,000 rpm S-OFL (tF2), (3) tailing factor of the 3rd peak, R-OFL
for 10 min; the clear supernatant was collected and filtered (tF3), (4) resolution between the Ofloxacin enantiomers
through a 0.2-µm membrane filter (Gelman Science, India), (Rs23), (5) retention time of the last peak, R-OFL (tR3) and
and 20 µL of this solution was injected for HPLC analysis. (6) chromatographic optimization function (COF).
K
Chromatographic Conditions
 
 Rsi
COF = Ai ln + B(tM − tL ) (1)
Rsid
i=1
The chromatographic separation was carried out using a
mobile phase consisting of a mixture of MeOH, hexane, COF is calculated according to Eq. (1) [22]. Where
MeCN and small amounts of AcOH and DEA. The analytes Ai and B are weighted parameters (equal to unity in this
were detected at 285 nm based on isobestic point. Prior to use, study), Rsi is the resolution of the ith pair, Rsid is the
the mobile phase was degassed for 15 min in an ultrasonic desired resolution for the specific pair (equals to 4), tM rep-
bath and vacuum filtered through 0.45-µm membrane filter resents the desired maximum analysis time (here assumed
(Gelman Science, India). The injection volume was 20 µL. 10 min), and tL is the actual time of the last eluted peak. In
The mobile phase was prepared by first mixing the appropri- the present study, COF was used as it enabled to decrease
ate proportions of MeOH in hexane and then with ACN; sub- data from each chromatogram to a single number which can
sequently small amounts of AcOH and DEA were added to be used in the optimization procedure. The elution order
this mixture as per design. The HPLC system was used in an of analytes was confirmed by injecting the pure standards
air-conditioned laboratory atmosphere (25 ± 2 °C). individually. Probenecid (IS) was used as an internal stand-
ard as it presented acceptable resolution and retention time
Method Validation with both of the enantiomers.
Table  1 summarizes the conducted experiments,
The optimized method was validated according to ICH viz. (n  = 14 + 6) six replicates at center point and the
Q2(R1) guidelines [17]. The validation parameters like responses. All experiments were conducted in randomized
linearity, limit of detection and quantification, specificity, order to minimize the effects of uncontrolled variables that
accuracy, precision and robustness were addressed. may introduce a bias on the measurements. Two replicates
were performed for each experiment in order to know the
experimental error variance and to test the predictive valid-
Results and Discussion ity of the model.
A cross-effect between the three factors (MeOH, DEA
Optimization of Chromatographic Conditions and flow rate) and the selected responses were then ana-
lyzed using a “standard least squares” model. Calculated
Rotatable central composite design (RCCD) was employed coefficients of the response model and obtained P values
in this study for the optimization of chromatographic are given in Table 2. The insignificant terms (P > 0.05)

13
V. Kannappan, S. S. Mannemala

Table 1  Central composite Design points Coded factor levels Responses


rotatable design arrangement
and responses %MeOH %DEA Flow rate k1 Rs23 tF2 tF3 tR3 COF

1 0 0 0 0.40 4.7 2.1 1.8 6.4 7.89


2 0 0 0 0.40 4.7 2.1 1.9 6.4 7.89
3 0 0 0 0.41 4.8 2.0 1.7 6.5 7.71
4 0 0 0 0.43 4.7 1.9 1.8 6.5 7.69
5 0 0 0 0.41 4.7 1.9 1.7 6.5 7.69
6 0 0 0 0.40 4.7 1.9 1.8 6.5 7.70
7 0 0 −1.682 0.42 5.2 2.3 1.9 9.2 2.45
8 0 0 +1.682 0.45 4.2 1.6 1.5 5 10.38
9 0 −1.682 0 0.61 4.3 1.9 1.8 6.3 7.47
10 −1.682 0 0 0.57 7.5 1.8 1.5 6.9 7.53
11 +1.682 0 0 0.52 3.6 2.1 1.9 6.8 6.83
12 0 +1.682 0 0.51 4.9 2.1 1.7 6.6 7.68
13 +1 +1 −1 0.45 4.1 2.6 2.1 7.9 4.92
14 +1 −1 −1 0.60 4.1 2.4 2.1 7.6 4.99
15 −1 −1 −1 0.52 5.9 1.9 1.68 8.2 4.39
16 −1 +1 −1 0.43 5.6 2.0 1.6 8.7 3.41
17 +1 +1 +1 0.39 3.9 2.0 1.8 5.7 9.20
18 +1 −1 +1 0.55 3.8 2.0 1.8 5.6 9.14
Central composite rotatable 19 −1 +1 +1 0.48 5.8 1.8 1.5 6 8.97
design arrangements are in 20 −1 −1 +1 0.69 5.7 1.6 1.5 6.9 6.87
random

Table 2  Reduced response models and statistical parameters obtained from ANOVA (after backward elimination)
Responses Reduced response modelsa Adjusted R2 Model P value %CV Adequate precision

k1 k1 = +0.42 − 0.015 A − 0.057 B + 0.012 C − 0.042 AC 0.8664 0.000 6.50 14.088


+ 0.046 A2 + 0.051 B2
Rs23 Rs23 = +4.65 − 1.00 A − 0.16 C + 0.28 A2 0.9406 0.000 4.61 33.672
tF2 tF2 = + 2.00 + 0.16 A + 0.061 B − 0.20 C 0.8186 0.001 5.12 18.270
tF3 tF3 = +1.75 + 0.16 A − 0.11 C 0.8476 0.000 4.00 20.209
tR3 tR3 = +6.81 − 1.12 C 0.8144 0.000 6.60 26.440
COF COF = +7.04 + 2.18 C 0.8249 0.000 12.05 27.381
a
  Only significant coefficients with P < 0.05 are included. Factors are in coded levels

were eliminated from the model through a backward Since the assumptions of normality and constant variance
elimination process to obtain a simple and realistic model. of the residuals were found to be satisfied, the fitted model
The adjusted R2 were well within the acceptable lim- for the response COF was accepted.
its of R2  ≥ 0.80 [23], which revealed that the experimen- Response models (Table 2) suggest that among the fitted
tal data show a good fit with the second-order polynomial models, k1 has the interaction term (AC) with largest abso-
equations. For all the reduced models, P value of >0.05 is lute coefficient (+0.042). The positive interaction between
obtained, implying that these models are significant. The A and C is statistically significant (P < 0.0001) for the k1
adequate precision values were found to be in the range of model. Perturbation plot was examined for response k1 to
14.08–33.67, which indicates an adequate signal, and there- understand the effect of an independent factor on a specific
fore, the model is significant for the separation process. The response, with all other factors held constant at a reference
%CV for all the models was found to be <10 %, except for point [25]. It is evident from Fig. 2 that the response k1 was
COF (12.05 %). Therefore, the diagnostic plots (a) normal highly influenced by the level of factor B followed by fac-
probability plot of the residuals and (b) plot of residual ver- tor A. As expected, capacity factor k1 is least sensitive to
sus predicted values [24] were analyzed for response COF. factor C.

13
Multiple Response Optimization of a HPLC Method

performing the experiment under the optimal condition,


and the corresponding chromatogram is shown in Fig. 4.
The average errors for k1, Rs23, tF2, tF3, tR3 and COF were
1.63, 0.05, −1.60, −3.95, −2.61 and 0.73 %, respectively,
within a difference of <4 %, indicating a good correlation
between the experimental and the predicted responses.

Validation of the Test Procedure

Linearity

The linearity of the proposed method was estimated by car-


rying out regression analysis at five concentration levels in
the range of 2–10 µg mL−1 for Ofloxacin (approximately
20–200 % of the nominal range of the analyte). The cali-
bration curve was obtained using the linear least squares
regression procedure. The representative linear equation
was y = 0.2375x − 0.0413 and y = 0.2356x − 0.0462 for
S-OFL and R-OFL, respectively. Correlation coefficients
were found to be more than 0.998 for both enantiomers.
Fig. 2  Perturbation plot showing the effect of each of the independ-
ent factors on response k1, where A is %MeOH, B is %DEA and C is The homoscedasticity for the calibration curves was veri-
mobile phase flow rate fied by using Bartlett’s test and in that no statistical differ-
ence (P > 0.05) was found between the variances [26].

Global Optimization of Multiple Responses Limits of Detection and Quantitation

The targeted criteria for the optimization was to minimize Calibration curves were plotted at five levels ranging from
the resolution between the Ofloxacin enantiomeric pair, 0.05 to 1.0 % of the nominal analyte concentration [27].
reduce the tailing factors of enantiomers, minimize the The residual standard deviation of the response (σ) and
analysis time, increase the COF value as well as the reten- slope (s) of the calibration curve was used to calculate
tion factor k1. Global optimization of multiple responses the LOD as 3.3 σ/s and LOQ as 10 σ/s. Using the above
was achieved by employing desirability function computed equations, the LOD and LOQ were estimated as 4.95 and
using JMP prediction profiler (Fig. 3). Desirability function 14.99 ng mL−1 for S-OFL, and 4.26 and 12.92 ng mL−1 for
transfers the response variable to a 0–1 scale. A response R-enantiomer, respectively.
of 0 represents a completely undesirable response and 1
represents the most desirable response. Figure 3 depicts Specificity
that a smaller % of MeOH has positive impact on all the
factors except for the resolution Rs23 in which it leads to Specificity of the method was evaluated by comparing the
excess resolution between the enantiomer pair, small % chromatograms of placebo sample containing a mixture of
of DEA showed a good impact on k1 value. However, its the commonly used excipients (starch, lactose monohy-
effect on other factors was not significant as it showed drate, methyl cellulose, titanium dioxide and magnesium
less curvature and the higher flow rate values showed sig- stearate) with that of the standard drugs. There were no
nificant effect on k1, COF and tR3. The operating condi- excipient peaks co-eluted with the analyte as well as with
tions were chosen to achieve the maximum overall desir- IS, which shows that the developed method is selective and
ability; all the responses were optimized simultaneously. specific to the excipients used in this study (Fig. 4).
The optimum conditions were chosen by total desirability
as near as 1. The highest desirability value of 0.823 was Accuracy
achieved at MeOH/hexane/MeCN-43.33/10/46.62 (v/v)
%, %AcOH/DEA-0.4/0.2 and flow rate as 1.4 mL min−1. Accuracy of the method was demonstrated by analyzing
The predicted response values corresponding to the high- quality control (QC) standards prepared at three levels of
est desirability value (0.823) were k1 = 0.61, Rs23 = 5.193, 80, 100 and 120 % of the expected assay value in the mar-
tF2 = 1.68, tF3 = 1.57, tR3 = 6.14 min and COF = 8.25. keted formulation. QC samples were prepared as three rep-
The prediction efficiency of the model was confirmed by licates at each concentration level by spiking the standard

13
V. Kannappan, S. S. Mannemala

Fig. 3  Prediction profiler
obtained for the responses Rs23,
tF2, tF3, tR3, COF and k1

drugs with the placebo excipients, which were left over- hypothesis being that the recovery is unity or 100 %. Since
night to allow matrix–analyte interactions to occur, and then the calculated t-value (tCalc) for S-OFL (2.73) and R-OFL
analyzed as described in “Preparation of Sample Solution” (1.64) is less than the theoretical t-value (tCrit  = 4.30), at
section. The % recovery of the enantiomers at each level 5 % significance level, the null hypothesis was accepted.
(n  = 3) and mean % recovery (n  = 9) were determined, These results indicate that the method is accurate, and there
and data are presented in Table 3 where accuracy (%) was was no interference from placebo in this study.
expressed as [(calculated amount/predicted amount) × 100].
The recoveries of both enantiomers at each level were found Precision
to lie within the acceptable criteria of the bias ±2 % [28].
The mean % recovery (n = 9) for each enantiomer was also The precision was established by injecting three concen-
tested for significance by using Student’s t test, the null tration levels (2, 6 and 10 µg mL−1) for Ofloxacin with

13
Multiple Response Optimization of a HPLC Method

Fig. 4  Chromatograms cor-
responding to (a) placebo solu-
tion; b Levofloxacin (enantio-
pure); c a real sample of Glevo®
tablets containing Levofloxacin;
d a real sample of Zenoflox®
tablets containing Ofloxacin
enantiomers; e a synthetic
mixture of probenecid (IS) and
Ofloxacin enantiomers, under
optimum conditions

Table 3  Validation of method Validation parameters S-OFL R-OFL


for the determination of S-OFL
and R-OFL Concentration Results Concentration Results

Linearity (n = 6) 2–10 µg mL−1 y = 0.2375x − 0.0413 2–10 µg mL−1 y = 0.2356x − 0.0462


2
R  = 0.9988 R2 = 0.9981
LOD 4.95 ng mL−1 4.26 ng mL−1
−1
LOQ 14.99 ng mL 12.92 ng mL−1
Specificity The method is specific with respect to tablets ingredients (starch, lactose
monohydrate, methyl cellulose, titanium dioxide and magnesium stearate)
Accuracy (mean %recovery) (n = 3)
80 % W/W 98.78 80 % W/W 99.28
100 % W/W 99.63 100 % W/W 99.21
120 % W/W 99.47 120 % W/W 99.50
(mean %recovery, %CV) (n = 9) 99.29, 0.45 99.53, 0.49
Precision (%CV) (n = 6)
(a) Intraday precision 2.0 2.73 2.0 2.93
6.0 1.24 6.0 0.89
10.0 0.88 10.0 1.04
(b) Interday precision 2.0 1.84 2.0 2.73
6.0 1.39 6.0 0.87
10.0 0.70 10.0 0.91
Robustness (%assay, %CV)
%MeOH conc. (43.33 ± 0.5 %) 100.22, 0.53 99.84, 0.81
%DEA conc. (0.2 ± 2 %) 100.38, 0.25 100.05, 0.73

10 µg mL−1 of IS (probenecid) each in six replicates, for Robustness


intraday precision (repeatability) and on three consecutive
days for the intermediate precision (reproducibility). Pre- To demonstrate the reliability of the developed method
cision was expressed by the RSD (%) of the analyte peak for routine use, robustness of the proposed method was
area. Results for all studied compounds (Table 3) met the evaluated with respect to small, but deliberate variations
proposed requirement %RSD ≤ 3 % [28]. in experimental parameters such as variations in %MeOH

13
V. Kannappan, S. S. Mannemala

concentration (43.38 % ± 0.5) and % DEA (0.2 ± 0.02) References


did not alter the retention times, tailing factor (tF2) and res-
olution values more than 2 % in the proposed method. So, 1. Burke D, Henderson DJ (2002) Chirality: a blueprint for the
future. Br J Anaesth 88(4):563–576. doi:10.1093/bja/88.4.563
it could be concluded that the developed method is robust. 2. Francotte ER (2001) Enantioselective chromatography as a power-
ful alternative for the preparation of drug enantiomers. J Chroma-
Application of the Method togr A 906(1–2):379–397. doi:10.1016/S0021-9673(00)00951-1
3. Drew RH, Gallis HA (1988) Ofloxacin: its pharmacol-
ogy, pharmacokinetics, and potential for clinical applica-
The proposed RP-HPLC method was applied to the quan- tion. Pharmacother J Hum Pharmacol Drug Ther 8(1):35–46.
titative analysis of the Ofloxacin and Levofloxacin tab- doi:10.1002/j.1875-9114.1988.tb0406x
lets. Ofloxacin enantiomers in real samples (Zenoflox 4. Indian Pharmacopoeia (2010) vol v. 1. Indian Pharmacopoeia
tablets) containing racemic Ofloxacin were analyzed by Commission, Ghaziabad, India
5. British Pharmacopoeia (2011) vol v. 1. Her Majesty’s stationery
the proposed method. Representative chromatograms are Office, London, UK
presented in Fig. 4. The results achieved when analyzing 6. United States Pharmacopoeia (2009) vol v. 1. United States Phar-
Zenoflox tablets were 100.07 (0.11) mg of Levofloxacin macopeial Convention, Rockville, Maryland, USA
and 99.95 (0.06) mg of R-OFL, respectively, with the val- 7. de Boer T, Mol R, de Zeeuw RA, de Jong GJ, Ensing K (2001)
Enantioseparation of ofloxacin in urine by capillary electrokinetic
ues within parentheses being the %CV of the six replicates. chromatography using charged cyclodextrins as chiral selectors and
Assay of Levofloxacin was performed on Glevo-500 tab- assessment of enantioconversion. Electrophoresis 22(7):1413–1418.
lets in which the presence of R-enantiomer is considered to doi:10.1002/1522-2683(200105)22:7<1413::AID-ELPS1413>3.0.
be an impurity. The result obtained for the assay of Levo- CO;2-U
8. Awadallah B, Schmidt PC, Wahl MA (2003) Quantitation of
floxacin was 499.6 (0.1) mg with the values within paren- the enantiomers of ofloxacin by capillary electrophoresis in the
theses being the %CV of the six replicates. The content of parts per billion concentration range for in vitro drug absorp-
the R-OFL was not more than 0.5 %. Good agreement was tion studies. J Chromatogr A 988(1):135–143. doi:10.1016/
found between the assay results and the label claim of the S0021-9673(02)02015-0
9. Bi W, Tian M, Row KH (2011) Chiral separation and determi-
marketed formulations. The quantities found for Ofloxacin nation of ofloxacin enantiomers by ionic liquid-assisted ligand-
as well as for Levofloxacin in tablets were in conformity exchange chromatography. Analyst 136(2):379–387. doi:10.1039/
with the values claimed by the manufacturer. C0AN00657B
10. Yan H, Qiao F (2013) Rapid screening of ofloxacin enantiomers
in human urine by molecularly imprinted solid-phase extraction
coupled with ligand exchange chromatography. J Liq Chromatogr
Conclusions Relat Technol 37(9):1237–1248. doi:10.1080/10826076.2013.78
9795
11. Zeng S, Zhong J, Pan L, Li Y (1999) High-performance liquid
In this paper, an efficient chiral HPLC method was devel-
chromatography separation and quantitation of ofloxacin enan-
oped, optimized and validated for the simultaneous esti- tiomers in rat microsomes. J Chromatogr B Biomed Sci Appl
mation of the Ofloxacin enantiomers in pharmaceutical 728(1):151–155. doi:10.1016/S0378-4347(99)00085-7
formulations and in bulk drugs using response surface 12. Fang Z, Guo Z, Qin Q, Fan J, Yin Y, Zhang W (2013) Semi-pre-
parative enantiomeric separation of ofloxacin by HPLC. J Chro-
methodology. This method reduces overall analysis time
matogr Sci 51(2):133–137. doi:10.1093/chromsci/bms117
and provides essential information regarding the sensitiv- 13. Lehr K-H, Damm P (1988) Quantification of the enantiomers of
ity of various chromatographic factors and their interaction ofloxacin in biological fluids by high-performance liquid chro-
effects on separation attributes. Higher sensitivity, shorter matography. J Chromatogr B Biomed Sci Appl 425:153–161.
doi:10.1016/0378-4347(88)80015-X
analysis time, use of polar non-aqueous organic solvents
14. Rabbaa L, Dautrey S, Colas-Linhart N, Carbon C, Farinotti R
and adequate resolution of the developed method dem- (1997) Absorption of ofloxacin isomers in the rat small intestine.
onstrates that it can be extrapolated for semi-preparative Antimicrob Agents Chemother 41(10):2274–2277
purpose and for LC–MS analysis of Ofloxacin enantiom- 15. Sun X, Wu D, Shao B, Zhang J (2009) High-performance liquid-
chromatographic separation of ofloxacin using a chiral stationary
ers. The proposed method was found to be linear, sensitive,
phase. Anal Sci 25(7):931–933. doi:10.2116/analsci.25.931
selective, precise and accurate. Therefore, it could be suc- 16. Institute. SAS (2010) JMP statistical software version 9.0. SAS
cessfully adopted for routine analysis of Ofloxacin enanti- Institute Inc, Cary, North Carolina, USA
omers and in chiral impurity profiling of Levofloxacin in 17. Secretariat ICH (2005) Validation of analytical procedures: text
and methodology, Q2 (R1), current step 4 version, parent guide-
bulk drugs and pharmaceutical formulations.
lines on methodology dated November 6 1996, incorporated in
November 2005, vol 60. Fed.Register, Geneva, Switzerland
Acknowledgments  Sai Sandeep M is grateful to University Grants 18. Dossou KSS, Chiap P, Chankvetadze B, Servais A-C, Fillet M,
Commission (UGC), New Delhi, India, for providing UGC BSR fel- Crommen J (2010) Optimization of the LC enantioseparation of
lowship and to UGC SAP- DRS Phase-I sponsored Department of chiral pharmaceuticals using cellulose tris(4-chloro-3-methylphe-
Pharmacy, Annamalai University, Tamil Nadu, India, for providing nylcarbamate) as chiral selector and polar non-aqueous mobile
the facilities to carry this research work. phases. J Sep Sci 33(12):1699–1707. doi:10.1002/jssc.201000049

13
Multiple Response Optimization of a HPLC Method

19. Chankvetadze B (2012) Recent developments on polysaccha-


of domperidone and pantoprazole. J Pharm Biomed Anal
ride-based chiral stationary phases for liquid-phase separation 43(5):1842–1848. doi:10.1016/j.jpba.2006.12.007
of enantiomers. J Chromatogr A 1269:26–51. doi:10.1016/j. 24. Choisnard L, Bigan M, Blondeau D, Dhulster P, Leman B, Guil-
chroma.2012.10.033 lochon D (2003) Application of the method of the experimental
20. Dossou KSS, Chiap P, Servais AC, Fillet M, Crommen J (2011) design to the study of a processing of unshrinkableness of wool
Development and validation of a LC method for the enantiomeric fibers. J Appl Polym Sci 89(2):535–547. doi:10.1002/app.12173
purity determination of S-ropivacaine in a pharmaceutical for- 25. Sivakumar T, Manavalan R, Muralidharan C, Valliappan K (2007)
mulation using a recently commercialized cellulose-based chiral An improved HPLC method with the aid of a chemometric proto-
stationary phase and polar non-aqueous mobile phase. J Pharm col: simultaneous analysis of amlodipine and atorvastatin in phar-
Biomed Anal 54(4):687–693. doi:10.1016/j.jpba.2010.10.020 maceutical formulations. J Sep Sci 30(18):3143–3153. doi:10.10
21. Valliappan K, Vaithiyanathan S, Palanivel V (2013) Direct chiral 02/jssc.200700148
HPLC method for the simultaneous determination of warfarin 26. Danzer K, Currie L (1998) Guidelines for calibration in ana-
enantiomers and its impurities in raw material and pharmaceuti- lytical chemistry-Part I. Fundamentals and single component
cal formulation: application of chemometric protocol. Chroma- calibration (IUPAC Recommendations 1998). Pure Appl Chem
tographia 76(5–6):287–292. doi:10.1007/s10337-012-2373-7 70(4):993–1014
22. Glajch JL, Kirkland JJ, Squire KM, Minor JM (1980) Opti-
27. Crowther JB (2001) Validation of pharmaceutical test meth-

mization of solvent strength and selectivity for reversed-phase ods. In: Satinder A, Stephen S (eds) Separation science and
liquid chromatography using an interactive mixture-design sta- technology, vol 3. Academic Press, pp 415–443. doi:10.1016/
tistical technique. J Chromatogr A 199:57–79. doi:10.1016/ S0149-6395(01)80014-3
S0021-9673(01)91361-5 28. Ermer J, Burgess C, Kleinschmidt G, McB. Miller JH (2005) Per-
23. Sivakumar T, Manavalan R, Muralidharan C, Valliappan K
formance parameters, calculations and tests. In: Method valida-
(2007) Multi-criteria decision making approach and experimen- tion in pharmaceutical analysis. Wiley-VCH Verlag GmbH & Co.
tal design as chemometric tools to optimize HPLC separation KGaA, pp 21–194. doi:10.1002/3527604685.ch2

13

You might also like