You are on page 1of 7

Journal of Chromatographic Science, 2016, Vol. 54, No.

10, 1813–1819
doi: 10.1093/chromsci/bmw162
Advance Access Publication Date: 7 October 2016
Article

Article

Downloaded from https://academic.oup.com/chromsci/article/54/10/1813/2527514 by National Science & Technology Library user on 23 May 2023
Development and Validation of a Normal Phase
Chiral HPLC Method for Analysis of Afoxolaner
Using a Chiralpak® AD-3 Column
Jinyou Zhuang*, Satish Kumar, and Abu Rustum
Merial Inc., A Sanofi Company, 631 Rt 1 South, North Brunswick, NJ 08902, USA
*
Author to whom correspondence should be addressed. Email: jinyou.zhuang@merial.com
Received 20 April 2016; Revised 2 August 2016

Abstract
Afoxolaner is a new antiparasitic molecule from the isoxazoline family that acts on the insect acar-
ine gamma-aminobutyric acid and glutamate receptors. Isoxazoline family of compounds has
been employed as active pharmaceutical ingredient in drug products prescribed for control of
fleas and ticks in dogs. Afoxolaner with a chiral center at isoxazoline ring exists as a racemic mix-
ture. A normal phase chiral high performance liquid chromatography analytical method has been
developed to verify that afoxolaner is a racemic mixture as demonstrated by specific rotation, as
well as to determine enantiomeric purity of single enantiomer samples. A Chiralpak® AD-3 column
(150 × 4.6 mm I.D.) maintained at 35°C was used in the method. Analytes were analyzed with an
isocratic elution using n-Hexane/IPA/MeOH (89:10:1, v/v/v) as the mobile phase with a detection
wavelength of 312 nm. Desired separation of the two enantiomers was achieved in <10 minutes
with resolution and selectivity factors of 5.0 and 1.54, respectively. The analytical method was
appropriately validated according to ICH guidelines for its intended use. ® All marks are the prop-
erty of their respective owners.

Introduction Use of chiral chromatography has proven to be immensely valu-


Single enantiomers of biologically active chiral compounds have been able for identification and quantitation of chiral compounds. high
used in drug products by pharmaceutical industry for many decades. performance liquid chromatography (HPLC) analysis for chiral
Individual enantiomer of a chiral compound can potentially exhibit compounds can be conducted in two ways—indirect or direct. An
different pharmacology, toxicology and metabolism activities in the indirect way of chiral analysis involves derivatization of the analyte,
living system (1). Potentially only one of the two enantiomers may be leading to two diastereomers followed by separation using tradi-
responsible for the desired therapeutic response. Whereas the other tional reversed-phase chromatography (3). Whereas, a direct method
enantiomer may be inactive or may even have deleterious effect. utilizes a chiral selector on chiral stationary phases (CSPs) (4–10) or
Therefore, it is of a paramount importance to develop analytical in the mobile phase to distinguish the enantiomers (11). Direct meth-
methods that can identify and determine enantiomeric purity of a chi- ods are most widely used for chiral separations due to (i) availability
ral active pharmaceutical ingredient (API) in bulk lots as well as in of a wide range of chiral selectors such as Pirkle type, cyclodextrins,
drug products. To provide pharmaceutical industry further guidance chirobiotic phases, protein and polysaccharide based (12, 13), and
on this key topic US Food and Drug Administration (FDA) issued a (ii) no pretreatment of the analyte is needed.
guidance entitled “Development of New Stereoisomeric Drugs” in Among the various types of CSP, modified cellulose and
1992 (2). As stated in the FDA guidance document, specifications for amylose-based polysaccharides that were originally developed by
the final product should assure identity, strength, quality and purity Okamoto and Yashima (14) have demonstrated very good chiral
from a stereochemical viewpoint. recognition ability in a wide range of applications (15).

© The Author 2016. Published by Oxford University Press. All rights reserved. For Permissions, please email: journals.permissions@oup.com 1813
1814 Zhuang et al.

This paper describes development of a normal phase chiral HPLC columns


HPLC method using amylose-based polysaccharides column for sep- Different polysaccharide based HPLC columns were used for
aration of a racemic mixture of afoxolaner (Scheme 1). Afoxolaner method development. Columns used were Chiralpak® AD-3 (150 ×
is a new insecticide–acaricide molecule from isoxazoline family. This 4.6 mm, 3 µm particle size), Chiralcel® OD-H (150 × 4.6 mm, 5 µm
compound is used as the API in veterinary drugs (e.g., NexGard®) particle size) and Chiralcel® OJ-3 columns (150 × 4.6 mm, 3 µm
for fleas and ticks control by acting on the insect’s gamma- particle size). Chiralpak® AD column has tris(3,5-dimethylphenyl-
aminobutyric acid and glutamate receptors. Afoxolaner is neutral carbamate) amylose derivative as the stationary phase which is

Downloaded from https://academic.oup.com/chromsci/article/54/10/1813/2527514 by National Science & Technology Library user on 23 May 2023
molecule without any ionizable groups. It has a logP of 5.15 indicat- coated on silica gel. While Chiralcel® OD and Chiralcel® OJ col-
ing its strong hydrophobic character. As shown in Scheme 1 afoxo- umns contain cellulose modified with tris(3,5-dimethylphenylcarba-
laner has one chiral center on the isoxazoline ring. For the purpose mate) and tris(4-methylbenzoate), respectively on silica gel. All
of the method development two enantiomers of the compound were columns were purchased from Chiral Technologies Co. (West
named “Enantiomer 1” and “Enantiomer 2”. Chester, PA).
As afoxolaner is a relatively new chemical entity, at present there
is no published report about the chiral separation of afoxolaner. For
the first time in this paper we report method development and vali- Chromatographic analysis
dation of a chiral HPLC method to analyze afoxolaner and its indi- Agilent 1100 or 1200 series HPLC system (Agilent Technologies,
vidual enantiomers. The major goals of the method development Santa Clara, CA) with UV or DAD detector equipped with
were to develop a sensitive, rugged and QC-friendly normal phase ChromSword® method development software (Merck KGaA,
chiral HPLC method that can verify afoxolaner sample is racemic Darmstadt, Germany) and LC Spiderling line HPLC Column
mixture as well as can determine enantiomeric purity of the single Selector (Chiralizer Services, L.L.C., Newtown, PA) were used for
enantiomers. In this paper, “Enantiomer 1” is treated as the API method development and/or method validation. The major parts of
and “Enantiomer 2” as impurity or undesired enantiomer. As both the mobile phase were n-Hexane and IPA. Several organic modifiers
enantiomers are expected to behave similarly in chromatography (various alcohols) were evaluated as well. Chromatographic perfor-
(except the retention time), method can be used as well when mance was evaluated by retention time (Rt), selectivity (α), resolu-
“Enantiomer 1” is treated as impurity. Desired separation of the tion (Rs) and peak efficiency (N) which were calculated by
two enantiomers was achieved in <10 minutes on Chiralpak® AD-3 ChemStation or Empower.
column (150 × 4.6 mm I.D.) at 35°C with an isocratic elution using Afoxolaner (racemic mixture) reference standard and individual
n-Hexane/IPA/MeOH (89:10:1, v/v/v). Analytes were monitored enantiomer solutions were prepared in IPA. Each mobile phase solu-
with a detection wavelength of 312 nm. The new analytical method tions were prepared by pre-mixing required amount of n-Hexane/
has been successfully validated for its intended purpose as per ICH IPA and organic modifier (as needed).
guidelines and demonstrated to be accurate, linear, precise, specific,
sensitive and robust.
Results
Optimization of chromatographic conditions
The main purpose of this work is to develop a fast Normal Phase
Experimental HPLC method to separate and quantitate the two enantiomers in
Materials Afoxolaner (racemic mixture). During method development work,
Reference standards and afoxolaner samples were obtained from several cellulose based chiral HPLC columns were selected on the
Merial, Inc. (North Brunswick, NJ). All solvents were HPLC grade. basis of the chemical structure and location of the chiral center of
Solvents/reagents were purchased from commercial sources and the analyte and also on the characteristics of the chiral phase of the
used as is. n-Hexane was purchased from VWR (Radnor, analytical columns. Three columns namely Chiralcel® OJ-3,
Pennsylvania). Isopropanol (IPA), n-propanol and 1-butanol were Chiralcel® OD-H and Chiralpak® AD-3 were selected and screened
purchased from EMD (Billerica, Massachusetts). Ethanol (200 by using low polarity mobile phase system. Figure 1 shows the over-
proof) was purchased from Sigma-Aldrich (St. Louis, Missouri). laid chromatograms using these columns with mobile phase of n-
Cyclohexanol, 2-butanol and 3-methyl-1-butanol were purchased Hexane/IPA (95:5, v/v). Best chiral selectivity and resolution was ob-
from Alfa Aesar (Haverhill, Massachusetts). Methanol was pur- tained on Chiralpak® AD-3 column. Therefore, Chiralpak® AD-3
chased from Honeywell (Morris Plain, NJ). column was selected for further optimization.
As indicated in Figure 2, the separation of the two enantiomers
was obtained using isocratic elution by use of n-Hexane and IPA
with the ratios of 90:10 and 95:5. Better and fast separation is
F O achieved with n-Hexane/IPA (90:10, v/v). For optimization of the
F H
N mobile phases, the effect of a variety of organic modifiers such as
F N methanol, ethanol, 1-butanol, hexanol, t-butanol and cyclohexanol
H F F
O F were investigated. The final mobile phase is n-Hexane/IPA/MeOH
F
* F (89:10:1, v/v/v). Table I summarized the final chromatographic con-
N O F ditions. The final method was then used for analysis of both
Afoxolaner (racemic mixture) and Enantiomer 1 samples. Typical
chromatograms of both afoxolaner (racemate) and “Enantiomer 1”
Cl that had a low level of “Enantiomer 2” as the impurity are shown in
Figures 3 and 4, respectively. Figure 3 is a chromatogram of
Scheme 1. Molecular structure of afoxolaner. Afoxolaner (racemic mixture) which indicates the peak area ratio of
Development and Validation of a Normal Phase Chiral HPLC Method 1815

0.180

0.135 AD-3
AU

0.090

Downloaded from https://academic.oup.com/chromsci/article/54/10/1813/2527514 by National Science & Technology Library user on 23 May 2023
OD-H

0.045

OJ-3
0.000

0.00 12.00 24.00 36.00 48.00 60.00


Minutes

Figure 1. Chromatograms of afoxolaner (0.1 mg/mL) on various chiral HPLC columns with mobile phase n-Hexane/IPA (95:5, v/v); flow rate: 0.8 mL/min; wave-
length: 312 nm; column temperature: 35°C; injection volume: 10 μL.

0.340

0.255

0.170
AU

Hexane/IPA (90:10, v/v)


0.085

Hexane/IPA (95:5, v/v)


0.000

0.00 12.00 24.00 36.00 48.00 60.00


Minutes

Figure 2. Chromatograms of afoxolaner (0.8 mg/mL) on AD-3 column with different mobile phases; flow rate: 0.8 mL/min; wavelength: 312 nm; column tempera-
ture: 35°C; injection volume: 10 μL.

Table I. Final Chromatographic Conditions of the Method Results of these evaluations are summarized in sections below.
®
System suitability requirements [blank baseline: no interfering peaks
HPLC column Chiralpak AD-3 150 × 4.6 mm, 3 µm
above 0.1% level at the retention time of two afoxolaner enantio-
Mobile phase n-Hexane/IPA/MeOH (89:10:1, v/v/v)
mers, 0.2% standard solution have signal-to-noise ratio (S/N) ≥10
Temperature 35°C
Flow rate 0.8 mL/min for each enantiomer; standard solution preparation agreement
Wavelength 312 nm (≤2.0%), resolution factor (≥1.2), injector agreement (difference
Injection volume 10 μl between two injections ≤2.0%)] were met prior to performing the
Afoxolaner 0.8 mg/mL method validation experiments.

Enantiomer 1 and Enantiomer 2 is 50:50. Shown in Figure 4 is a Specificity


chromatogram of Enantiomer 1 obtained using the final chro- No significant interfering peaks (peak area >0.1%) was observed at
matographic conditions. Enantiomer 1 and Enantiomer 2 were well the retention time of the two enantiomers in blank solution. In addi-
separated with Rs of 5.4. The %peak area of Enantiomer 1 and tion, no evidence of co-elution was noted using peak purity analysis
Enantiomer 2 are 99.63% and 0.37%, respectively. (purity angle was less than the purity threshold using Empower
Software) for the two enantiomers.

Validation of method DL/QL evaluation


To ensure that method is suitable for the intended purpose key The DL and QL of method was evaluated using diluted afoxolaner
method characteristics, i.e., specificity, accuracy, linearity, precision, (racemate) standard solution. The estimated DL (S/N~3) was
DL (detection limit) and QL (quantitation limit) were evaluated. 0.06 μg/mL (0.02%) for each enantiomer. The QL for the method
1816 Zhuang et al.

0.260

0.195

AU 0.130

0.065

Downloaded from https://academic.oup.com/chromsci/article/54/10/1813/2527514 by National Science & Technology Library user on 23 May 2023
0.000

0.00 3.00 6.00 9.00 15.00 12.00


Minutes

Figure 3. A typical chromatogram of 0.8 mg/mL afoxolaner racemate analyzed using the final chromatographic conditions (Chiralpak® AD-3 150 × 4.6 mm, 3 µm;
mobile phase = n-Hexane/IPA/MeOH (89:10:1, v/v/v); flow rate: 0.8 mL/min; wavelength: 312 nm; column temperature: 35°C; injection volume: 10 μL).

0.012

0.008 Impurity
Enantiomer 1 (Enantiomer 2)
AU

0.004

0.000

–0.004
0.00 3.00 6.00 9.00 12.00 15.00
Minutes

Figure 4. A typical chromatogram of 0.4 mg/mL “Enantiomer 1” lot analyzed using the final chromatographic conditions (Chiralpak® AD-3 150 × 4.6 mm, 3 µm;
mobile phase = n-Hexane/IPA/MeOH (89:10:1, v/v/v); flow rate: 0.8 mL/min; wavelength: 312 nm; column temperature: 35°C; injection volume: 10 μL).

was set to (0.2 μg/mL; 0.05%) as S/N for “Enantiomer 1” and “Enantiomer 2”. The method is proven to be linear within the inves-
“Enantiomer 2” was 21 and 15, respectively. These results demon- tigational range.
strate the method is sensitive for detection and estimation of both The recoveries of “Enantiomer 2” at each level were also calcu-
afoxolaner enantiomers. lated and the results ranged from 98% to 104% meeting the accep-
tance criteria of 70–130%. The %RSD of the recovery from five
levels in the linearity/recovery study was calculated to be 2% meet-
Linearity/accuracy ing the acceptance criteria of ≤10%. These results demonstrate the
In this study, “Enantiomer 1” is treated as API and “Enantiomer 2” method is linear and accurate for “Enantiomer 1” (as API) and
is treated as impurity. The linearity of both enantiomers was evalu- “Enantiomer 2” (as an impurity).
ated. Since “Enantiomer 1” was treated as API, its linearity was
evaluated from 0.2% to 125% of the nominal concentration of
Precision
0.4 mg/mL. A linear least square analysis of the data gave correla-
The method precision was evaluated by analyzing six solutions of
tion coefficient (r) of 1.000. The y-intercept obtained was ≤0.5% of
Enantiomer 1 at 100% level (0.4 mg/mL). The enantiomer purity
the 100% level indicating that there is no significant bias for quanti-
value was calculated for each preparation. The average enantiomer
tation for “Enantiomer 1”. Method is considered accurate for
purity was 99.6% and %RSD = 0.0% (n = 6) thus met the criteria
“Enantiomer 1” because as per ICH Q2B method’s precision (see
of %RSD ≤2%. These results demonstrate that the method is pre-
section precision), linearity and specificity have been established.
cise for determination enantiomer purity of the API samples.
“Enantiomer 2” is treated as impurity and the linearity and
recovery were studied together by preparing a series of “Enantiomer
2” solution with concentrations corresponded to 0.2%, 0.5%,
Discussion
1.0%, 2.5% and 5.0% in “Enantiomer 1” (0.4 mg/mL). The solu-
tions were prepared by spiking Enantiomer 2 into the “Enantiomer Column and mobile phase screening
1” (0.4 mg/mL) solution at calculated ratios. At each level, duplicate Polysaccharide-based HPLC columns were selected for the method
injections were performed. development. As polysaccharide-based CSPs lack ionic sites, they
A linear least square analysis of the data gave correlation coeffi- are suitable for chromatography of a neutral compound such as
cient (r) of 0.999. The y-intercept obtained was ≤3.0% of the 100% afoxolaner (16, 17). In addition, polysaccharide-based columns
level indicating that there is no significant bias for quantitation for have been demonstrated to be very useful in separation of a wide
Development and Validation of a Normal Phase Chiral HPLC Method 1817

spectrum of racemate in pharmaceutical industry due to their versa- peaks eluted around 22 and 30 minutes, respectively with Rs of 1.6
tility and durability (18). and selectivity of 1.4. Both peaks showed peak tailing factor great
Normal phase chromatographic conditions were selected for than 1.5. No separation was achieved on Chiralcel® OD-H column
the method. This is because hydrogen bonding, dipole–dipole under this mobile phase condition.
and π–π interactions which are considered critical for chiral As it has been reported in literature (19) changing from one alco-
recognition are known to be more effective under normal pha- hol to another can affect chiral separation (including elution order),
se conditions (14). Among the large number of available alcohols other than IPA were studied as well in combination with

Downloaded from https://academic.oup.com/chromsci/article/54/10/1813/2527514 by National Science & Technology Library user on 23 May 2023
polysaccharide-based columns, Chiralcel® OD, Chiralpak ® AD IPA. The other alcohols evaluated were methanol, ethanol, n-propa-
and Chiralcel® OJ columns are most commonly used for pharma- nol, 1-butanol, 2-butanol, t-butanol, 3-Methyl-1-butanol, Hexanol
ceutical analysis; therefore, these were selected for the method and cyclohexanol. These alcohols are different in terms of their
development. A detection wavelength of 312 nm was selected for polarity, chain length and steric effects. The different butanols stud-
analysis since afoxolaner has λmax around this wavelength ied have varied steric effect. All analyses were performed on
(Figure 5). In addition, selection of higher wavelength minimizes Chiralpak® AD-3 column with afoxolaner standard solution
potential interference from common solvent/reagents contami- (0.8 mg/mL) with n-Hexane/alcohol (95:5, v/v) as mobile phase.
nants. At the initial stage of method development 0.1 mg/mL Partial or no separation was obtained for all alcohols except 3-
afoxolaner reference standard (a racemic mixture) solution was Methyl-1-Butanol. However, selectivity factor for 3-Methyl-1-
used. To increase sensitivity of the method, the concentration Butanol (α = 1.23) was much lower than IPA (α = 1.60). Based on
was increased to 0.8 mg/mL later during method development. these experiments only IPA was pursed further as the polar compo-
The HPLC columns utilized were Chiralpak® AD-3 (150 × nent in the mobile phase.
4.6 mm, 3 µm particle size), Chiralcel® OD-H (150 × 4.6 mm,
5 µm particle size) and Chiralcel® OJ-3 columns (150 × 4.6 mm,
3 µm particle size) columns. Effect of organic modifiers
For the method isocratic mobile phase was preferred over a gra- In normal phase chiral chromatography, it is common to use organic
dient mobile phase due to slow equilibrium of mobile phase in nor- modifiers such as acids or alcohol to improve HPLC column’s selec-
mal phase chromatography. N-Hexane and IPA were selected as tivity. The addition of organic acid in the mobile phase is to mini-
non-polar and polar components, respectively as they are the most mize interactions with residuals silanols for better peak shape and
commonly used solvent combination in normal phase chromatogra- resolution (20). Also, as reported in literature (5, 21) addition of
phy and were found to suitable for afoxolaner separation. small amounts of alcohols does affect column selectivity as well.
Chromatographic analysis with n-Hexane/IPA (95:5, v/v) mobile Various alcohols were evaluated as organic modifier to the mobile
phase and Chiralcel® OJ-3 column showed two broad peaks around phase. First, the n-Hexane/IPA (90:10, v/v) was modified to include
6 and 37 minutes. With same mobile phase there was no separation 1% methanol. Addition of 1% methanol improved both resolution
of two enantiomers on Chiralcel® OD-H column. Using this mobile and selectivity compared to n-Hexane/IPA (90:10, v/v). It appears
phase combination, the best separation was achieved on Chiralpak® that small amount of methanol alters the steric environment around
AD-3 column with a resolution factor (Rs) of 6.0 and selectivity (α) the chiral cavities thus favoring further differentiation of the two en-
of 1.8 between the two enantiomers (Figure 1). However, the reten- antiomers. However, addition of more methanol (up to 3%) did not
tion times of “Enantiomer 1” and “Enantiomer 2” peaks were ~17 improve resolution or selectivity (Table II).
and 28 minutes, respectively. Besides methanol several other alcohols were also evaluated as the
To reduce analysis time, the mobile phase polarity was changed organic modifiers at 1% level. These were longer linear alcohols such
to n-Hexane/IPA (90:10, v/v). On Chiralpak® AD-3 column, the as ethanol, 1-butanol and hexanol, branched alcohols such as t-buta-
two enantiomer peaks eluted at 6 and 8 minutes, respectively with nol and cyclohexanol and diol (propylene glycol). Results of this eval-
resolution of 5.6 (see Figure 2). On Chiralcel® OJ-3 column, two uation are summarized in Table III. As shown in Table III, methanol

0.80

0.60

0.40
AU

312 nm

0.20

0.00

228.00 266.00 304.00 342.00 380.00


nm

Figure 5. Typical UV spectrum of afoxolaner in n-Hexane/IPA/MeOH (89:10:/1, v/v/v).


1818 Zhuang et al.

Table II. Effect of MeOH as Organic Modifier in Mobile Phase using AD-3 Column

n-Hexane (%) IPA (%) MeOH (%) RT (min) Resolution (Rs) Selectivity (α)

Enantiomer 1 Enantiomer 2

90 10 6.1 8.3 5.9 1.36


89 10 1 6.3 9.0 8.2 1.45
87 10 3 5.6 7.4 7.0 1.3

Downloaded from https://academic.oup.com/chromsci/article/54/10/1813/2527514 by National Science & Technology Library user on 23 May 2023
(Afoxolaner concentration: 0.1 mg/mL; flow rate: 0.8 mL/min; Wavelength: 312 nm; Column temperature: 35°C; Injection volume: 10 μL).

Table III. Effect of Various Alcohols as Organic Modifiers in Mobile Phase with AD-3 Column

n-Hexane (%) IPA (%) Organic modifier RT (min) Resolution (Rs) Selectivity (α)

Enantiomer 1 Enantiomer 2

89 10 1% MeOH 5.9 8.5 5.0 1.54


89 10 1% Ethanol 6.2 8.7 4.5 1.48
89 10 1% 1-Butanol 6.0 8.2 4.2 1.46
89 10 1% 2-Butanol 6.1 8.6 4.5 1.50
89 10 1% t-Butanol 6.3 9.0 4.5 1.50
89 10 1% Hexanol 6.0 8.3 4.1 1.45
89 10 1% Cyclohexanol 5.8 7.8 4.1 1.42
89 10 1% Propylene Glycol 5.8 8.0 4.3 1.47

(Afoxolaner concentration: 0.8 mg/mL; flow rate: 0.8 mL/min; Wavelength: 312 nm; Column temperature: 35°C; Injection volume: 10 μL).

Table IV. Results Summary for Method Robustness Study

Parameters Variation Retention time (min) Resolution (Rs)

Enantiomer 1 Enantiomer 2

Normal None 6.2 8.6 4.4


Column Temperature −2°C 6.3 8.8 4.7
+2°C 6.1 8.3 4.2
Flow rate −0.1 (mL/min) 7.1 9.8 4.5
+0.1 (mL/min) 5.5 7.6 4.4
Injection volume −2 µl 6.2 8.6 5.0
+2 µl 6.2 8.6 4.0
Wavelength −2 nm 6.3 8.8 4.7
+2 nm 6.3 8.8 4.7
%IPA n-Hexane/IPA/MeOH (91:8:1, v/v/v) 6.8 9.3 4.5
n-Hexane/IPA/MeOH (87:12:1, v/v/v) 4.6 6.0 3.5

(Afoxolaner concentration: 0.8 mg/mL; Chiralpak® AD-3 150 × 4.6 mm, 3 µm).

gave the best selectivity. So based on the data n-Hexane/IPA/MeOH Robustness study
(89:10:1, v/v/v) was selected as the mobile phase for the final method. The robustness of the method was demonstrated by showing the
capacity of the method remained unaffected while deliberately
changing HPLC parameters. Several key parameters, including %
Column temperature IPA in mobile phase, flow rate, detector wavelength, temperature
The column temperature effect on the separation of the two enantio- and injection volume were varied around the procedural values.
mers was studied at 25, 30, 35 and 40°C. At lower temperature (25°C) Resolution of the two enantiomers under each HPLC parameter var-
better separation was achieved (Rs: 6.6) compared to higher tempera- iation was assessed against the procedural parameters. Except varia-
tures (35°C) (Rs: 5.0) and 40°C (Rs: 4.2). For rugged HPLC method, tion of %IPA in mobile phase, all other method robustness
a well-controlled column temperature is crucial. Ideally, column tem- parameters were evaluated using ChromSword AutoRobust, a com-
perature should be at least 10°C above or below the ambient temper- puter software that tests method’s robustness with minimal analyst
ature (~25°C) for better temperature control. Column temperature set intervention. Method robustness results are summarized in
around ambient is difficult to tightly control due to environmental Table IV. In all varied conditions, the two enantiomers were well
fluctuations. Furthermore, 40°C is the highest recommended tempera- separated with a minimum resolution factor of 4 and runtime of
ture for this type of column. Based on these rationales the column <10 minutes. This shows method is robust as well as efficient and
temperature of 35°C was selected for the final method. suitable for high throughput analysis in a QC laboratory.
Development and Validation of a Normal Phase Chiral HPLC Method 1819

Conclusions chromatography: application to chiral inversion studies; Journal of


Pharmaceutical and Biomedical Analysis, (1998); 17: 1439–1447.
A new chiral HPLC method was successfully developed and vali- 9. Rustum, A.M., Estrada, V.; Separation and quantitation of the
dated for afoxolaner API. Method has been shown to be linear, S-(+)-enantiomer in the bulk drug tiagabine·HCl by chiral high-
accurate, precise, robust and sensitive. The method can be used to performance-liquid chromatography using a Chiralcel-OD column;
verify that afoxolaner is a racemic mixture as demonstrated by Journal of Chromatography B: Biomedical Sciences and Applications,
specific rotation, as well as to determine enantiomeric purity of (1998); 705: 111–117.
single enantiomer samples. The method is also considered QC 10. Rustum, A.M.; Determination of chiral purity of ethyl nipecotate using a

Downloaded from https://academic.oup.com/chromsci/article/54/10/1813/2527514 by National Science & Technology Library user on 23 May 2023
friendly as it is robust, uses isocratic mobile phase with both enan- Chiralcel-OG column; Journal of Chromatography A, (1995); 696:
75–81.
tiomers eluting in <10 minutes, and employs commonly used sol-
11. Zheng, C., Huang, W.X., Chen, Z., Rustum, A.M.; Separation of chiral
vents as mobile phase.
primary amino compounds by forming a sandwiched complex in
reversed-phase high performance liquid chromatography; Journal of
Chromatography A, (2010); 1217: 4965–4970.
Acknowledgments 12. Beesley, T.E., Scott, R.P.W.; Chiral chromatography. John Wiley and
The authors would like to thank all analytical scientists in Analytical R&D Sons Ltd, Chichester, West Sussex, (1998).
Merial for their support during this study. 13. Gübitz, G., Schmid, M.G.; Chiral separation by chromatographic and
electromigration techniques. A review; Biopharmaceutics and Drug
Disposition, (2001); 22: 291–336.
14. Okamoto, Y., Yashima, E.; Polysaccharide derivatives for chro-
References matographic separation of enantiomers; Angewandte Chemie
1. Nguyen, L.A., He, H., Chuong, P.H.; Chiral drugs: an overview; International Edition, (1998); 37: 1020–1043.
International Journal of Biomedical Science, (2006); 2: 85–100. 15. Bargann-Leyer, N., Tambute, A., Caude, M.; A comparison of LC and
2. FDA. (1992) Development of New Stereoisomeric Drugs. http://www.fda. SFC for cellulose- and amylose-derived chiral stationary phases; Chirality,
gov/Drugs/GuidanceComplianceRegulatoryInformation/Guidances/ucm1 (1995); 7: 311–325.
22883.htm (accessed April, 2016). 16. Yashima, E.; Polysaccharide-based chiral stationary phases for high-
3. Toyo’oka, T.; Resolution of chiral drugs by liquid chromatography based performance liquid chromatographic enantioseparation; Journal of
upon diastereomer formation with chiral derivatization reagents; Journal Chromatography A, (2001); 906: 105–125.
of Biochemical and Biophysical Methods, (2002); 54: 25–56. 17. Perrin, C., Vu, V.A., Matthijs, N., Maftouh, M., Massart, D.L., Heyden,
4. Neue, U.D.; HPLC columns: theory, technology and practices. Wiley- Y.V.; Screening approach for chiral separation of pharmaceuticals: Part I.
VCH, New York, NY, (1997). Normal-phase liquid chromatography; Journal of Chromatography A,
5. Lu, J., Rustum, A.M.; Separation of the two enantiomers of T-3811ME (2002); 947: 69–83.
by normal-phase HPLC using modified amylose as chiral stationary 18. Tachibana, K., Ohnishi, A.; Reversed-phase liquid chromatographic sepa-
phase; Journal of Chromatographic Science, (2008); 46: 466–471. ration of enantiomers on polysaccharide type chiral stationary phases;
6. Zhou, L., Welch, C., Lee, C., Gong, X., Antonucci, V., Ge, Z.; Journal of Chromatography A, (2001); 906: 127–154.
Development of LC chiral methods for neutral pharmaceutical related 19. Persson, B., Andersson, S.; Unusual effects of separation conditions on
compounds using reversed phase and normal phase liquid chromatogra- chiral separations; Journal of Chromatography A, (2001); 906: 195–203.
phy with different types of polysaccharide stationary phases; Journal of 20. Stringham, R.W., Ye, Y.K.; Chiral separation of amines by high-
Pharmaceutical and Biomedical Analysis, (2009); 49: 964–969. performance liquid chromatography using polysaccharide stationary
7. Rustum, A.M.; Separation of the R(−)- and S(+)-enantiomers of the ethyl phases and acidic additives; Journal of Chromatography A, (2006); 1101:
ester of tiagabine HCl using a Chiralcel-OG column; Journal of 86–93.
Chromatography. A, (1994); 684: 29–36. 21. Kunath, A., Theil, F., Jähnisch, K.; Influence of the kind of the alcoholic
8. Rustum, A.M., Menon, G., Patel, S.B.; Separation of the S(+) and R(−)- modifier on chiral separation on a Chiralpak AD column; Journal of
enantiomers of tiagabine HCl and its two chiral precursors by chiral Chromatography A, (1996); 728: 249–257.

You might also like