You are on page 1of 14

Critical Reviews in Analytical Chemistry

ISSN: (Print) (Online) Journal homepage: https://www.tandfonline.com/loi/batc20

Stability Indicating Methods for Determination of


Third Generation Antiepileptic Drugs and Their
Related Substances

Jéssica Domingos da Silva, Lucio Mendes Cabral & Valéria Pereira de Sousa

To cite this article: Jéssica Domingos da Silva, Lucio Mendes Cabral & Valéria Pereira de
Sousa (2021): Stability Indicating Methods for Determination of Third Generation Antiepileptic
Drugs and Their Related Substances, Critical Reviews in Analytical Chemistry, DOI:
10.1080/10408347.2021.1890544

To link to this article: https://doi.org/10.1080/10408347.2021.1890544

Published online: 06 Mar 2021.

Submit your article to this journal

Article views: 46

View related articles

Full Terms & Conditions of access and use can be found at


https://www.tandfonline.com/action/journalInformation?journalCode=batc20
CRITICAL REVIEWS IN ANALYTICAL CHEMISTRY
https://doi.org/10.1080/10408347.2021.1890544

REVIEW ARTICLE

Stability Indicating Methods for Determination of Third Generation


Antiepileptic Drugs and Their Related Substances
J
essica Domingos da Silva , Lucio Mendes Cabral , and Valeria Pereira de Sousa
Department of Drugs and Pharmaceutics, Faculty of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil

ABSTRACT KEYWORDS
The third generation of antiepileptic drugs that have been approved by international regulatory Antiepileptic drugs; third
agencies between 2007 and 2018 include rufinamide, stiripentol, eslicarbazepine acetate, lacosa- generation; stability
mide, perampanel, brivaracetam and everolimus. As part of demonstrating their safety profile, sta- indicating methods; HPLC;
related substances
bility indicating methods are developed to monitor these drugs and their impurities. In this
context, this review describe some characteristics, impurities and the stability indicating methods
used for the determination of these drugs and the presence of their related substances. Through
a search in official compendia and scientific articles, fifty-six analytical methodologies were identi-
fied up to October 2020. The methodologies were developed using techniques of HPLC, UPLC,
HPTLC, GC and UV/Vis spectrophotometry. A majority of the methods (70%) employed HPLC-UV.
A number of these antiepileptic drugs were found to have had a small number of studies related
to their stability and for the detection of impurities. The presentation of the current level of
research on third generation antiepileptic drugs highlights the need for new stability and safety
studies that are necessary to develop new pharmaceutical products containing these drugs.

1. Introduction some advantages such as more favorable pharmacokinetics,


improved tolerability, lower teratogenic potential and/or
Epilepsy is one of the most common chronic neurological
reduced probability to cause enzyme inductions and drug
disorders in the world with recurrent crises that affects
interactions.[9,10]
approximately 50 million people of all ages.[1] An epileptic
The monitoring for impurities and degradation products
seizure is defined as a transient occurrence of abnormally
is essential for establishing a safety profile for these seven
excessive or synchronous neuronal activity in the brain that
most recently approved AEDs, since these related substances
is visibly apparent.[2] Antiepileptic drugs (AEDs) used to
may have genotoxic potential and induce adverse effects.[11]
treat epilepsy aim to decrease the probability of seizures by
acting on the main mechanisms contributing to brain Thresholds of impurities and degradation products are
excitability.[3] expressed as total daily intake (TDI) of these related sub-
Pharmacological treatment of epilepsy began in the 20th stances or as a percentage of active pharmaceutical ingredi-
century with discovery of phenobarbital, which represents ent (API) determined by maximum daily dose of API. It is
the first generation of AEDs. The second generation com- recommended that the exposure be zero or very close to
prise vigabatrin, oxcarbazepine, lamotrigine, felbamate gaba- zero. To meet this recommendation, monitoring must be
pentin, topiramate, tiagabine, levetiracetam and zonisamide, performed through highly sensitive analytical methods with
these drugs were licensed during the period from 1980 to an adequate capacity to detect these related substances.[12–14]
2000.[4,5] Currently, there are at least 27 molecules approved Considering this objective, the purpose of this review is
for use as an AED by the Food and Drug Administration to describe the most current analytical methodologies that
(FDA) and European Medicines Agency (EMA).[6] The are being applied to determine the third generation AEDs in
third, and most recent, generation of AEDs is formed by the presence of their related substances. In addition, we pre-
rufinamide (RUF), stiripentol (STP), lacosamide (LAC), esli- sent an overview of the chemical characteristics of these
carbazebine acetate (ESA), perampanel (PER), brivaracetam drugs and their organic impurities reported in the literature.
(BRV) and everolimus (EVR). These drugs were approved In addition, we identify several of the products that can be
for treatment of epilepsy between 2007 and 2018 by the formed in forced degradation studies. The analytical meth-
FDA and EMA.[4,5,7] The search for new molecules and for- ods described here were compiled from official compendia
mulations that aim to improve efficacy continues, especially and published scientific articles, which were found by data-
in cases of resistance to therapy, also known as refractory base searches as recently as October 2020 in Google Scholar,
epilepsy.[8] In addition, third generation AEDs can offer Science Direct and PubMed. The search terms include the

CONTACT Valeria Pereira de Sousa valeria@pharma.ufrj.br Department of Drugs and Pharmaceutics, Faculty of Pharmacy, Federal University of Rio de
Janeiro, Av. Carlos Chagas Filho, 373, CCS, Bss, sl15, Rio de Janeiro, RJ 21941-902, Brazil.
ß 2021 Taylor & Francis Group, LLC
2 J. D. DASILVA ET AL.

Table 1. Therapeutics indications, mechanisms of action and dosage forms of third generation AEDs.
AED Approval date Indication Mechanism of Action Dosage forms Reference
RUF 2007 (EMA) Lennox-Gastaut syndrome in patients aged 1 year Modulation of VGSCs, Tablet [15,16]

2008 (FDA) particularly the Oral suspension


Nav1.1 isoform
STP 2007 (EMA) Dravet’s syndrome in a patient aged 2 years in Positive allosteric modulator Capsule Powder for [17,18]

2018 (FDA) association with clobazam and valproate at GABAA receptors oral suspension
[19,20]
LAC 2008 (FDA and EMA) Seizures with partial onset with or without secondary Blocking VGSCs through slow Tablet
generalization in patients aged 4 years inactivation Oral solution
Intravenous injection
[21,22]
ESA 2009 (EMA) Refractory partial-onset seizures, with or without Blocking VGSCs through slow Tablet
2013 (FDA) secondary generalization in adult patients (FDA) inactivation Oral suspension
and children aged  6 years (EMA)
[23,24]
PER 2012 (FDA and EMA) Primary generalized tonic-clonic seizures and focal Non-competitive negative Tablet
epilepsies with or without generalized seizures in allosteric antagonist of Oral suspension
patients aged  12 years AMPA receptors
[25,26]
BRV 2016 (FDA and EMA) Partial onset seizures with or without secondary Binding to SV2A Tablet
generalization in patients aged 4 years Oral solution
Intravenous injection
[27,28]
EVR 2017 (EMA) Tuberous sclerosis complex associated partial-onset Inhibition of the Tablet
2018 (FDA) seizures in patients aged 2 years mTOR pathway Dispersible tablet
AMPA - a-amino-3-hydroxyl-5-methyl-4- isoxazole-propionic acid; GABA- gamma-amino butyric acid; mTOR - mammalian target of rapamycin; SV2A- synaptic ves-
icle protein 2 A; VGSCs – voltage-gated sodium channels.

individual names of each AED combined with “stability RUF shows that it is a triazole derivative and PER is a
indicating methods”, “forced degradation” and “impurities”. bipyridine derivative. Neither have a chiral carbon in their
The intention of the data compiled in this review is to sup- structures.[33,34] STP is a derivative of a-ethylene alcohol
port researchers and manufacturers in the development of whereby the dosage forms are composed by racemic equi-
stability studies and analytical methodologies for determin- molar mixture of the (R) - (þ) - and (S) - (-) - stiripentol
ing the third generation AEDs and their related substances. enantiomers.[35,36] LAC is a functionalized amino acid with
a substitution on carbon 2 that originates a chiral center.
The (R) -enantiomer is the pharmacologically active form
2. Mechanisms of action and therapeutic indications with antiepileptic activity.[37,38]
The third generation of AEDs described in this review act in ESA is a prodrug derived from the same family as carba-
the reduction of abnormal cerebral hyperexcitability through mazepine and oxcarbazepine that has in its chemical struc-
five different mechanisms of action as summarized in ture a dibenzazepine nucleus with the 5-carboxamide
Table 1. RUF, LAC and ESA act in the modulation of volt- substituent but structurally different in positions 10 and
age-controlled ion channels; STP increases inhibition of 11.[32] The hydrolysis of this prodrug generates oxcarbaze-
gamma-amino butyric acid (GABA); BRV modulates synap- pine and the active metabolite (S) -licarbazepine (eslicarba-
tic release by binding to protein 2 A on synaptic vesicle zepine). Oxcarbazepine is metabolized to (R)-licarbazepine
(SV2A); PER inhibits synaptic excitation mediated by ino- and these two enantiomers are biologically active with the S
tropic glutamate receptors by binding to a-amino-3- form displaying less toxicity and is more efficient at crossing
hydroxy-5-methyl-4-isoxazol-propionate (AMPA) receptors; the blood brain barrier.[39]
and EVR is considered an agent with a targeted mechanism BRV is a 2-pyrrolidone derivative of levetiracetam with
to inhibit mTOR protein pathway, which is the mammalian two chiral centers, one between the bonding of the butana-
target of rapamycin.[3,29] mide portion to the pyrrolidone ring, and the other at pos-
These AEDs also differ in terms of their therapeutic indi- ition 4 of pyrrolidone ring, which permits formation of four
cation, also summarized in Table 1. RUF, STP and EVR are different diastereoisomers. (S, R) -brivaracetam is the form
considered orphan drugs and indicated for the treatment of present in pharmaceutical forms, as it has been rationally
seizures associated with rare diseases, such as Lennox- designed to have a greater affinity for binding to SV2A
Gastaut and Dravet syndromes as well as the tuberous scler- protein.[40,41]
osis complex.[15–18,27,28] LAC, BRV and ESA are indicated EVR is a hydroxyethyl derived from rapamycin (siroli-
for the treatment of partial seizures with or without second- mus), a macrolide isolated from Streptomyces hygroscopicus.
ary generalization in pediatric and adult patients.[19–22,25,26] This drug contains 15 asymmetric carbons and 4 substituted
PER is indicated for the treatment of generalized and partial double bonds that maintain the original rapamycin configur-
seizures in adult patients only.[23,24] ation. EVR is unstable at temperatures above 25  C and is
extremely sensitive to oxidation, which is stabilized with the
antioxidant butylhydroxytoluene (BHT).[42,43]
3. Chemical characteristics
In addition to their different mechanisms of action and 4. Stability indicating analytical methods
therapeutic indications, third generation AEDs also have dif-
ferent chemical structures, solubility and physical chemical Stability indicating methods are essential for detecting deg-
characteristics, as shown in Table 2. The chemical structure radation products formed during forced degradation studies
Table 2. Chemical structure and physicochemical characteristics of third generation AEDs.
Molecular Molecular Melting
AED Chemical Structure IUPAC name formula Weight (g/mol) pka Log P range ( C) Solubility References
[15,30,31]
RUF 1- (2,6-difluorobenzyl) -1H-1,2,3-triazole-4-carboxamide C10H8F2N4O 238.19 14.37 ± 0.50 0.65 233  238 Insoluble in water, slightly
soluble in gastric and
intestinal fluids, ethanol
and methanol
[30,31]
STP rac-(E)-1-(benzo [d] [1,3]dioxol-5-yl)-4,4-dimethylpent- C14H18O3 234.29 14.45 ± 0.20 2.95 64 Insoluble in water, soluble in
1-en-3-ol acetone and alcohol,
moderately soluble
in chloroform
[30,31]
LAC (R) -2-acetamido-N-benzyl-3-methoxypropionamide C13H18N2O3 250.29 14.19 ± 0.46 0.728 140–146 Soluble in aqueous medium

[30–32]
ESA (10S) -5-carbamoyl-10,11-dihydro-5H-dibenzo [b, C17H16N2O3 296.32 13.97 ± 0.40 8.8 184–187 Soluble in organic solvents
f]azepin-10-yl acetate and aqueous buffers (pH
1.2  7.4 at 37  C)

[30,31,33]
PER 2- (2-oxo-1-phenyl-5- (pyridin-2-yl) -1,2-dihydropyridin- C23H15N3O 349.38 4.73 ± 0.19 1.36 180 Insoluble in water, with
3-yl) benzonitrile improvement in pH acidic

[25,30]
BRV (2S) -2- [ (40 R) -2-oxo-4-propylpyrrolidin-1- C11H20N2O2 212.29 15.74 ± 0.50 1.04 76–78.7 Soluble in aqueous medium
yl] butanamide and organic solvents

[30,31]
EVR (1R,9S,12S,15R,16E,18R,19R,21R,23S,24E,26E,28E,30S,32S, C53H83NO14 958.22 10.40 ± 0.70 4.18 97–101 Slightly soluble in water,
35R) 1,18-Dihydroxy-12-[(2R)-1-[(1S,3R,4R) 4- (2- soluble in organic solvents
hydroxy-ethoxy)-3-methoxycyclohexyl]propan-2-yl]-
19,30-di-methoxy-15,17,21,23,29,35-hexamethyl-
11,36-dioxa-4-azatricyclo [30.3.1.04,9]hexatriaconta-
16,24,26,28-tetraene-2,3,10,14,20-pentone
CRITICAL REVIEWS IN ANALYTICAL CHEMISTRY
3
4 J. D. DASILVA ET AL.

4.1. Rufinamide
Several methodologies have been found for the determin-
ation of RUF in the presence of its three related substances
(compounds 1-3; Appendix 1). USP 43 describes two meth-
ods, one for the API and the other for tablets, for the deter-
mination of RUF and two of its related substances
(compounds 1 and 2; Appendix 1). The two methodologies
Figure 1. Frequency of use for the techniques used in the stability indicating employ reverse phase HPLC (column C18), the minimum
methods (n ¼ 56) applied to determination of third generation AEDs in the differences between these methods is the composition of
presence of their related substances. Data based on the review of the scien-
tific literature. mobile phase (80% of potassium phosphate buffer in tablet
method instead of 83% of water in API method) and detec-
or that may be formed over the shelf life of the product. tion (210 nm for tablet method and 220 nm for API
These methods must be able to quantify the drug in the method). The resolutions recommend by USP 43 between
presence of related substances, excipients and matrix com- RUF and compound 1 (Appendix 1) for tablet and API
ponents.[13,44,45] Analytical methods reported in this review methods cannot be less than 1.5 and 2.5, respectively.[48]
were developed for the determination of third generation Most of the methods for RUF found in the literature also
AEDs in presence of organic impurities present with the used HPLC and the most applied parameters were C18 and
API, in the dosage forms or identified in forced degrad- C8 stationary phases (lenghts with a UV detection in the
ation studies. range of 210 to 243 nm. The mobile phases consisted of
Organic impurities can originate from starting materials, aqueous buffers and solvents such as acetonitrile, methanol
intermediate by-products or degradation, any of which can and tetrahydrofuran. Shorter RUF retention times of 2.4
arise during the manufacturing and/or storage of the API from 4.0 min were observed in methods that used aceto-
and finished product.[13] Seventy-nine organic impurities for nitrile in the mobile phase.[50–53,93] In contrast, the use of
third generation AEDs have been reported in the literature: methanol as an organic modifier increases RUF retention in
3 for RUF, 1 for STP, 26 for LAC, 21 for ESA, 13 for PER, stationary phase with an elution between 5 to 8 min, since
4 for BRV and 11 for EVR. Chemical structures, nomencla- this solvent has lower eluotropic strength than aceto-
tures, formulas and molecular weights of these related sub- nitrile[54–56]. Hassib et al.,[54] method shower greater sensi-
stances are detailed in Supplementary data (Appendix 1). tivity when compared to other methodologies with isocratic
Liquid chromatography, either HPLC (High Performance elution, however it was applied only for a separation of RUF
Liquid Chromatography) or UPLC (Ultra Performance and compound 1. The method of Mahamudi et al.[51],
Liquid Chromatography), were the most used techniques for obtained adequate separation between RUF and compounds
the development of the indicative stability methods for third 1 and 2, but the authors did not present the LOD values,
generation AEDs as shown in Figure 1. These techniques not allowing to assess the sensitivity of the method.
allow for the separation of several components quickly at Parashar et al.[94] and Ranjith et al.[58] were the only works
high resolution and sensitivity.[46] Other techniques in which separated RUF from all three related substances
employed were HPTLC (High performance thin layer chro- using gradient mode. Although these methods showed good
matography), GC (Gas chromatography) and UV/Vis spec- sensitivity, the disadvantage was a an increase in the reten-
trophotometry. In most methodologies, detection was tion time of RUF and compounds 1,2 and 3 (19.98 to
performed by UV due to the presence of chromophoric 35.31 min) as well as the total run time of 90 min.
groups in the structures of the AEDs (Figure 1). Two methods used UPLC[59] and HTPLC[60] as the chroma-
Employment of photodiode array detectors (DAD) delivers tographic separation technique for determination of RUF in the
sensitivity, compatibility with most solvents and coverage presence of its related substances in the API and when formu-
over a wide linear range of concentrations with an absorp- lated into tablets. The method developed for UPLC was advan-
tion profile proportional to concentration that follows Beer’s tageous, because the separation of RUF and all three related
law.[47] The stationary phases most often used were the substances has been achieved with an analysis time of 17 min
reverse phase (columns C18 and C8), but the separation of and a resolution greater than 2. Composition of the mobile
enantiomeric impurities employed direct methods that phase for this methodology was similar to that recommended
included chiral separation columns (CSP) derived from pol- by USP 43 for tablets (Table 3) and only differed by the pH of
ysaccharides. Among the AEDs reported in this review, the buffer.[59] HPTLC has proven to be an alternative technique
RUF, LAC and EVR have official methods described in US for the development of a stability indicating method. Using a
Pharmacopeia (USP 43) for determining these drugs and mobile phase composed of chloroform, methanol and glacial
their related substances in API and pharmaceutical forms[48] acetic acid (9:1:0.1, v/v/v), it was shown that RUF could be sep-
and European Pharmacopeia (Eur. Ph. 10.0th)[49]. These arated from its degradation product (compound 3; Appendix
methodologies are detailed in Table 3 as well as non- 1)[62]. Another alternative for determining RUF in the presence
compendial methods for the determination of these third of degradation product was through the use of first derivative
generation AEDs in presence of impurities ratio spectrophotometry. In this methodology, RUF was deter-
and degradation products. mined by measuring the peak amplitude at 269.5 nm.[54]
Table 3. Stability indicating methods by chromatography for the determination of third generation AEDs in presence of their related substances.
Method/ Retention
AED Matrix Detection Stationary phase/ Temperature (  C) Mobile phase/Flow rate/Injection volume time Run Time LOD/LOQ References
[48]
RUF API HPLC-UV C18 (125 x 4.6 mm, 5 lm) at RT MeOH: tetrahydrofuran: water (12:5:83, v/v). FR: – – –
220 nm 1.0 ml.min-1; IV: 20 mL
[48]
Tablet HPLC-UV C18 (125 x 4.6 mm, 5 lm) at RT MeOH: tetrahydrofuran: 2.7g/L of potassium dihydrogen – – –
210 nm phosphate in water (15:5:80, v/v). FR: 1.0 ml.min-1; IV:
25 mL
[50]
API, tablet HPLC-UV C18 (250 x 4.6 mm, 5 mm) at 25  C 10 mM tetra butyl ammonium hydrogen sulfate buffer 3.53 10.0 0.29 /0.87 mg.mL-1
215 nm pH 3.37: ACN (50: 50, v/v). FR: 1.0 ml.min-1; IV: 20 mL
[51]
API HPLC-UV Xbrigde C18 (250 mm  4.6 mm, 5 lm) Water: ACN (50: 50, v/v). FR: 1.0 ml.min-1 – – –
215 nm at RT
[52]
API, tablet HPLC-UV C18 (250 x 4.6 mm, 5 mm) at 25  C ACN: water (60:40, v/v). FR: 0.8 ml.min-1; IV: 20 mL 3.89 10.0 0.24/0.73 mg.mL-1
215 nm
[53]
API, tablet HPLC-UV Phenomenex Luna C18 (250 [ 4.6 mm, 5 ACN: water (60: 40, v/v). FR: 1.0 ml.min-1; IV: 20 mL 3.047 7.0 0.89/2.69 mg.mL-1
210 nm mm) at 25  C
[54]
API, tablet HPLC-UV Kromasil C8 (250mm x 4.6 mm,5 mm) ACN: water (50: 50, v/v). FR: 1.0 ml.min-1; IV: 20 mL 4.008 – 0.778/2.356 mg.mL-1
210 nm
[55]
API, tablet HPLC-UV C18 (250 x 4.6 mm, 5 mm) at 25  C MeOH: water (52:48, v/v). FR: 1.0 ml.min-1;IV: 20 mL 5.55 10.0 0.0028/0.0086 mg.mL-1
210 nm
R [56]
API, tablet HPLC-UV PhenomenexV Hyperclone BDS C18 (250 x MeOH: 0.1 M octane sulfonic acid, 0.1 M, potassium – – 0.156/0.311 mg.mL-1
210 nm 4.6 mm, 5 mm) at 35  C dihydrogen phosphate buffer: water pH 6.5
(30:10:5:55, v/v/v/v). FR: 1.0 ml.min-1;IV: 20 mL
[57]
API HPLC-UV Inertsil ODS 3V C18 (250 mm  4.6 mm, 5 0.1% ortophosphoric acid in water and MeOH: ACN: 20.03 90.0 0.003% / 0.01%
220 nm mm) at 30  C tetrahydrofuran (90:70:30, v/v/v); gradient elution. FR:
1.0 ml.min-1; IV: 50 mL
[58]
API HPLC-UV Inertsil ODS-3V (250  4.6 mm, 5 mm) 0.1% triethylamine in water pH 2.2 and MeOH: 19.89 90.0 0.0046 / 0.014 mg.mL-1
215 nm at 35  C tetrahydrofuran (98:2, v/v); gradient elution. FR:
1.0 ml.min-1; IV: 50 mL
[59]
API, tablet HPLC-UV Acquity TM UPLC BEH C18 (150 mm  0.02 M potassium dihydrogen orthophosphate buffer pH 4.16 17.0 0.01% /0.03%
210 nm 2.1 mm, 1.7 mm) at 40  C 4.5: MeOH: tetrahydrofuran (80:15:5, v/v). FR:
0.3 ml.min-1; IV: 2 mL
[60]
API, tablet HPTLC Precoated silica gel aluminum plate Chloroform: MeOH : glacial acetic acid (9:1:0.1, v/v/v) – – 196.59/595.74 ng.spot-1
210 nm 60 F254
[61]
STP API, capsule HPLC-UV Symmetry C18 (75  4.6 mm, 3.5 mm) ACN: 50 mM potassium dihydrogen phosphate buffer pH 1.80 6.0 0.081/0.242 mg.mL-1
262.5 nm at 25  C 4.1 (60 : 40, v/v).FR: 1.0 ml.min-1; IV: 20 mL
[49]
LAC API HPLC-UV Amylose derivative (250  4.6 mm, 10 mm) Water: 2-propanol: heptane (3:100:900, v/v/v). FR: 25.0 42.5 –
215 nm at RT 1.0 ml.min-1; IV: 20 mL
[49]
API HPLC-UV C8 (150  4.6 mm, 3.5 mm) at RT 0.1% solution of trifluoroacetic acid and trifluoroacetic 12.0 21.0 –
258 nm acid: ACN: MeOH (0.3:500:500, v/v/v); gradient elution.
FR: 1.2 ml.min-1; IV: 20 mL
[49]
Tablet, infusion HPLC-UV C8 (150  4.6 mm, 5 mm) at 35  C Methanesulfonic acid: ACN: water (0.75:130:870, v/v/v). 6.0 15.0 –
215 nm FR: 2.0 ml.min-1; IV: 5 mL
[49]
Oral solution HPLC-UV C18 (250  4.6 mm, 5 mm) at 30  C Trifluoroacetic acid: ACN: water (0.56:100:900, v/v/v) and 27.0 33.0 –
215 nm trifluoroacetic acid: ACN (0.5:1000, v/v); gradient
elution. FR: 1.5 ml.min-1; IV: 5 mL
[62]
API HPLC-UV Lichrosphere C18 (250  4.6 mm, 5 mm) 0.1 % trimethylamine in water pH 3.0: MeOH (70:30, v/ 10.7 15.0 0.036/ 0.108 mg.mL-1
215 nm at 25  C v). FR: 1.0 ml.min-1; IV: 25 mL
[63]
API HPLC-UV Hypersil BDS C18 (250  4.6 mm, 5 mm) 0.01M monobasic potassium phosphate pH 4.0: ACN 3.6 10.0 2.8/8.9 mg.mL-1
215 nm at 25  C (30:70, v/v). FR: 1.0 ml.min-1; IV: 20 mL
[64]
API, tablet HPLC-UV Hypersil C18 (150  4.6 mm, 5 mm) at RT ACN: water (20:80, v/v). FR: 1.0 ml.min-1;IV: 20 mL 8.9 – 2/ 5 mg.mL-1
258 nm
[65]
API HPLC-UV Develosil ODS HG-5 (150  4.6 mm, 5 mm) 0.01M sodium dihydrogen phosphate monohydrate 16.58 60.0 0.011/ 0.036 mg.mL-1
CRITICAL REVIEWS IN ANALYTICAL CHEMISTRY

210 nm at 40  C buffer pH 3.0 and ACN; gradient elution. FR:


1.0 ml.min-1; IV: 30 mL
(continued)
5
Table 3. Continued. 6
Method/ Retention
AED Matrix Detection Stationary phase/ Temperature (  C) Mobile phase/Flow rate/Injection volume time Run Time LOD/LOQ References
[66]
API, tablet HPLC-UV Apollo Grace C18 (250  4.6 mm, 5 mm) 0.01M sodium dihydrogen ortho phosphate buffer pH 5.6 10.0 –
215 nm at RT 3.5: ACN (70:30, v/v). FR: 1.0 ml.min-1; IV: 20 mL
[67]
API HPLC-UV Kromasil C18 (250  4.6 mm, 5 mm) at RT 0.01 M potassium dihydrogen orthophosphate buffer pH 14.64 22.0 –
220 nm 3.0 and ACN; gradient elution. FR: 1.0 ml.min-1;
IV: 20 mL
[68]
API HPLC-UV Kinetex C18 (150  4.6 mm, 5 mm) at 25  C 0.02 M potassium dihydrogen phosphate buffer pH 4.5 13.3 26.0 0.08/ 0.2 mg.mL-1
J. D. DASILVA ET AL.

210 nm and MeOH; gradient elution. FR: 1.0 ml.min-1; IV:10 mL


[69]
Oral solution HPLC-UV Waters Symmetry C8, (250  4.6 mm, 5 mm) 0.012 M sodium dihydrogen phosphate anhydrous buffer 25.67 85.0 –/0.05%
210 nm at 30  C pH 2.0 and water: ACN (20:80); gradient elution. FR:
1.2 ml.min-1; IV: 10 mL
[70]
API, tablet HPLC-UV Regis Pack (250  4.6 mm, 5 mm) at 25  C 100 % Ethanol. FR: 1.0 ml.min-1; IV: 10 mL 7.06 30.0 0.0429/ 0.1286 mg.mL-1
210 nm
[71]
API HPLC-UV Chiralcel OD-H, (250  4.6 mm, 5 mm) Mixture of n- hexane–ethanol (74:6, v/v) in isopropyl 12.93 25.0 –
210 nm at 25  C alcohol–trifluoroacetic acid (72:6:0.08, v/v). FR:
0.5 ml.min-1; IV: 10 mL
[72]
API, tablet HPLC-UV Chiralpak-IC (250  4.6 mm, 5 mm) at 27  C N-hexane: ethanol (85:15, v/v). FR: 1.0 ml.min-1; IV: 10 mL 11.5 25.0 –
210 nm
[67]
API HPLC-MS Kromasil C18 (250  4.6 mm, 5 mm) at RT 0.01 M ammonium acetate buffer pH 3.0 and ACN; – – –
gradient elution. FR: 1.0 ml.min-1; IV: 10 mL
[68]
API HPLC-MS Kinetex C18 (150  4.6 mm, 5 mm) at 25  C 0.02 M ammonium formate buffer pH 4.5 and MeOH; – – –
gradient elution
[73]
API HPLC-MS Agilent Zorbax SB C18 (150  4.6 mm, 0.02 M ammonium acetate buffer with 0.1% formic acid 10.0 25.0 –
5 lm) at 30  C and ACN; gradient elution. IV: 5 mL
[74]
API UPLC-UV Acquity HSS C18 (2.1  100 mm, 1.8 mm) 0.01 M sodium dihydrogen phosphate monohydrate 2.1 5.0 –
210 nm at 45  C buffer pH 2.0: ACN (85:15, v/v). FR: 0.7 ml.min-1;
IV: 1 mL
[75]
API, tablet HPTLC Precoated silica gel aluminum Toluene: ethyl acetate: MeOH: triethylamine (7:2:1:0.1 v/ – – 893.65/2708.03 ng.spot-1
210 nm plate 60 F254 v/v/v)
[76]
API, tablet GC-FID Capilar column HP-5 (30 m x 0.320 mm) Helio gas. FR: 2.0 ml.min-1. IV: 1 mL (split mode, 5.7 7.0 5.87/19.57 mg.mL-1
260  C 100  C for 1 min to 250  C (40  C/min) ratio 50:1)
[77]
ESA API HPLC-UV Chiralpak IC-3 (150  4.6 mm, 3 mm) Dichloromethane: ethanol (90:10, v/v). FR: 1.0 ml.min-1; 6.51 – Enantiomer: 0.3/
240 nm at 25  C IV: 20 mL 0.9 mg.mL-1
[77]
API HPLC-UV Chiralpak IC-3 (150  4.6 mm, 3 mm) 100% ACN. FR: 0.5 ml.min-1; IV: 10 mL 11.84 – Enantiomer: 0.3/
220 nm at 25  C 1.0 mg.mL-1
[78,79]
API HPLC-UV C8 (250  4.6 mm, 5 mm) at 35  C 0.01 M potassium dihydrogen phosphate buffer pH 5.0: 14.35 60.0 0.020 / 0.060 mg.mL-1
215 nm ACN (95:5 v/v) and ACN: water (80:20 v/v); gradient
elution. FR: 1.0 ml.min-1; IV: 10 mL
[80]
API HPLC-UV Zorbax SB C18 (150  4.6 mm, 3.5 mm)a at 0.01 M potassium dihydrogen phosphate buffer: MeOH 7.0 20.0 0.05/0.15 mg.mL-1
210 nm 27  C at room temperature (80:20, v/v) and ACN: MeOH: water (75:5:20, v/v);
gradient elution. FR: 1.0 ml.min-1; IV: 10 mL
[81]
API HPLC-UV Inertsil ODS 3 V (150  4.6 mm, 5 mm) at 0.1% orthophosphoric acid in water: MeOH: ACN 4.2 10.0 2.40/7.29 mg.mL-1
210 nm room temperature (500:250:250, v/v). FR: 1.5 ml.min-1; IV: 20 mL
[78,79]
API HPLC-MS RP8 (250  4.6 mm, 5 mm) at 35  C 0.01 M ammonium bicarbonate buffer and ACN (95:5, v/ – – –
v) and ACN: water (80:20, v/v); gradient elution. FR:
1.0 ml.min-1; IV: 10 mL
[82]
API UPLC-UV Acquity BEH C18 (150  2.1 mm, 1.7 mm) Mobile phase A and phase B (50:50) 2.6 3.5 0.38/ 1.15 mg.mL-1
215 nm at 30  C (A) 0.01 M potassium dihydrogen orthophosphate and
ACN (90:10, v/v), (B) ACN: water: MeOH (75:5:25, v/v/v).
FR: 0.5 ml.min-1; IV: 2 mL
[82]
API HPTLC Precoated silica gel aluminum Toluene: MeOH: acetone (60:20:20, v/v/v) – – 893.65/2708.03 ng.spot-1
254 nm plate 60 F254
[83]
PER API, tablet HPLC-UV ODS partisil (250  4.6 mm, 5 lm) at room ACN: 0.1% trimethylamine in water pH 2.5 (60:40 v/v). 3.0 7.0 0.32/ 0.98 mg.mL-1
227 nm temperature FR: 1.5 ml.min-1; IV: 20 mL
[84]
API HPLC-UV Shimadzu Wondasil C18 (250 mm x 0.01M potassium dihydrogen phosphate buffer and ACN; 24.88 40.0 0.19/4.78 mg.mL-1
220 nm 4.6 mm, 5 mm) at room temperature gradient elution. FR: 1.0 ml.min-1; IV: 20 mL
[85]
API HPLC- MS Dikma Diamonsil 0.01 M ammonium acetate buffer and ACN; – 40.0 –
C18 (250  4.6 mm, 5 mm) at 30  C gradient elution
[85]
API, tablet UPLC-UV Phenomenex C8 (150  4.6 mm, 3.5 mm) Acetic acid: ACN: water (0.1:50: 50, v/v/v). FR: 1.0 ml.min- 10.41 15.0 –
1
240 nm at 25  C ; IV: 20 mL
[86]
API UPLC-UV- MS Acquity BEH C18 (100  2.1 mm, 1.7 mm) 0.1 % formic acid in water: ACN (60:40, v/v). FR: 3.1 6.0 0.03/ 0.06 mg.mL-1
290 nm at 40  C 0.4 ml.min-1
[87]
BRV API HPLC-UV C18 (250  4.6 mm, 5 mm) at room MeOH: water: ACN (30:10:60 v/v). FR: 1.0 ml.min-1; 7.9 – 0.1/ 0.4 mg.mL-1
242 nm temperature IV: 20 mL
[88]
API, tablet HPLC-UV ODS 3 V (150  4.6 mm, 5 mm) at 30  C 0.1% trifluoroacetic acid in water: ACN (60:40 v/v). FR: 3.1 – 0.047/ 0.132 mg.mL-1
210 nm 1.0 ml.min-1; IV: 10 mL
[89]
API UPLC- UV Acquity BEH C18 (100  2.1 mm, 1.7 mm) 0.1 % trifluroroacetic acid in water and water: ACN (20: 3.1 10.0 0.05/0.018 mg.mL-1
230 nm at 35  C 80 v/v); gradient elution. FR: 0.3 ml.min-1; IV: 1 mL
[90]
API UPLC-UV -MS Chiral PAK IG-U (100  3.0 mm, 1.6 lm) 0.01 M ammonium bicarbonate:ACN (65:35, v/v). FR: 9.4 15.0 0.3/0.8 mg.mL-1
215 nm at 40  C 0.3 ml.min-1; IV: 5 mL
[49]
EVR API HPLC-UV C18 (150  2.1 mm, 2.7 mm) at 60  C Formic acid: MeOH: 0.7 g/L solution of ammonium 16.0 28.0 –
278 nm format: ACN (0.05:100:450:450 v/v/v/v) and formic
acid: MeOH: 0.96 g/L solution of ammonium formate:
ACN; gradient elution. FR: 0.9 ml.min-1; IV: 3.5 ml;
autosampler at 6  C
[49]
API HPLC-UV Base – deactivated C18 (150  2.1 mm, ACN: 0.27 g/L solution of potassium dihydrogen 15.0 43.0 –
210 nm 2.7 mm) at 50  C phosphate (40:60 v/v) and ACN; gradient elution. FR:
1.1 ml.min-1; IV: 10 ml
[91]
API, tablet HPLC-UV Hypersil BDS C18 (100  4.6 mm, 5 lm) Ammonium acetate buffer: ACN (50:50 v/v) pH 6.5. FR: 3.1 6.0 0.036/ 0.109 mg.mL-1
280 nm at 30  C 1.0 ml.min-1; IV: 10 mL
[92]
Tablet HPLC-UV Zorbax SB C18 (250  4.6 mm, 5 mm) Ammonia solution and formic acid and MeOH: ACN 38.3 100.0 –
280 nm at 50  C (30:70, v/v); gradient elution. FR: 1.0 ml.min-1;
IV: 50 mL
ACN – acetonitrile; API- active pharmaceutical ingredient; FID – Flame ionization detector; FR- flow rate; GC-Gas chromatography; HPLC – High performance liquid chromatography; HPTLC- High performance thin layer
chromatography; IV – injection volume; LOD- limit of detection; LOQ – limit of quantification; MeOH – methanol; MS- mass spectrometry; RT – room temperature.
CRITICAL REVIEWS IN ANALYTICAL CHEMISTRY
7
8 J. D. DASILVA ET AL.

In forced degradation studies, RUF proved to be more pharmacopeial methods. The chromatographic conditions
susceptible to oxidation and hydrolysis by both acid and most often used in non-compendial HPLC methods
alkaline conditions. No degradation was observed under described in these reviews and published after 2016 were:
thermal and photolytic conditions. One product (compound reverse stationary phase (C18 and C8) with a temperature
3; Appendix 1) was formed in acidic and alkaline hydrolysis range of 25 to 45  C, a mobile phase composed of either
(1 N HCl and 1 N NaOH).[58] Another study demonstrated sodium or potassium phosphate buffers, an acid pH and
that this product could be methylated by methanol (co-solv- organic modifiers as acetonitrile or methanol with a detec-
ent) present in acid hydrolysis, forming a pseudo degrad- tion at 210, 215, 220 and 258 nm.[62–69,95,96] The method of
ation product (compound 2; Appendix 1)[52]. Ramisetti et al.[68] demonstrated to be sensitive, the LOD
value (0.08 mg.mL1) was not the lowest among all the
methods described in the present review, but it proved to be
4.2. Stiripentol
the most comprehensive for separating LAC and six degrad-
Only one HPLC-UV method was reported in the literature ation products (compounds 8.14, 15,19, 25-27, Appendix 1),
to determine STP in the presence of its degradation product with run time of 26 min, selectivity and precision.
(compound 4; Appendix 1)[63]. The chromatographic condi- The chiral separation of LAC from its (S)-enantiomer
tions employed a column C18 along with a mobile phase (compound 7; Appendix 1) was performed by HPLC using
composed of phosphate buffer and acetonitrile in the iso- chiral separation columns. In the chiral method that used
cratic mode, which enabled the elution of STP at 1.8 min RegisPack column, based on tris-(3,5-dimethylphenyl carba-
and the degradation product at 4.25 min that gave a com- moyl) of amylose and a mobile phase composed of 100%
plete analysis time of 6 min. This methodology showed good ethanol, retention time of LAC and its enantiomer was
sensitivity (0.081 mg.mL1) and advantageous for separated 5.36 min and 7.02 min, respectively.[70] Shorter retention times
STP and its degradation product. In this work, the authors were observed with this methodology when compared to
observed that STP was highly susceptible to acid hydrolysis methods that used the cellulose-derived columns of Chiracel
with an almost total decomposition in 0.5 N HCl. In con- OD-H, based on tris- (3, 5-dimethylphenyl carbamate) of cel-
trast, it proved to be stable under alkaline, oxidative, photo- lulose and Chiralpak-IC, based on tris- (3, 5-dichlorophenyl
lytic and thermal conditions.[61] carbamate) of cellulose. Even with their longer retention
times, these methods proved to be enantioselective.[71,72]
Mass spectrometry (MS) coupled to HPLC was another
4.3. Lacosamide technique used both for determination of LAC and to support
Different methodologies were found in the literature for the the identification of degradation products (compounds 8, 14-
determination of LAC in the presence of its 26 related sub- 19, 25-30; Appendix 1). The methodologies developed used
stances (compounds 5-30; Appendix 1). Four HPLC-UV volatile buffers such as ammonium acetate and formate with
methods described in Eur. Ph 10.0th are presented in either acetonitrile or methanol as an organic solvent in combin-
Table 3 for the determination of LAC and 11 impurities ation with a C18 column. Ionization was performed by electro-
(compounds 5-15; Appendix 1) in the API, infusion, oral spray (ESI) in both positive and negative modes. High
solution and tablet.[49] One of these methodologies is for the resolution spectrometers (HRMS), such as Ion trap and Time-
determination of an enantiomeric purity in the API that of-flight (TOF) were used for determination of mass and frag-
specifies the use of a chiral separation column derived from mentation pattern of LAC and degradation products.[67,68,73]
amylose. This determination is important as the R enantio- Other non-HPLC techniques, such as UPLC, HPTLC and
meric form is responsible for the pharmacological action of GC, were also reported to separate LAC from its related
LAC (see Section 3). Methodologies for determining LAC substances. The parameters used in the UPLC-UV technique
and impurities in the API, tablets and infusion recommend were similar to some of those described for HPLC methods
the use of a C8 column, whereas for oral solution, the rec- such as using a C18 column, potassium phosphate buffer
ommendation is to employ a C18 column. All three method- and methanol as a mobile phase in an isocratic mode. This
ologies incorporate acetonitrile as an organic modifier. The methodology show to be an alternative to HPLC methods,
chromatographic conditions employed in these methods dir- with total analysis time of 6 min and resolution greater than
ectly impact in the retention time of LAC and its related 2 between peaks of LAC and 4 impurities (compounds
substances, to guarantee the resolution recommended by 11,16,19 and 20; Appendix 1)[78]. The HPTLC methodology
official monograph. For the tablet and infusion method, the proved to be selective for the determination of LAC using a
resolution proposed between compounds 8 and 9 (Appendix mobile phase composed of toluene, ethyl acetate, methanol
1) is 1.5. For these same related substances in the oral solu- and triethylamine (7:2:1:0.1, v/v/v/v) that allowed for the
tion method, the resolution recommended is at least 3.0. In separation of LAC from 3 of its degradation products (com-
the API methodology the suggested resolutions between pounds 8, 11 and 24; Appendix 1)[79]. Another technique
LAC and compounds 11 and 7 (Appendix 1) are at least 2.0 used to determine LAC in forced degradation studies was
and 4.5, respectively.[49] gas chromatography with flame ionization detection (GC-
Two reviews[95,96] describe some stability indicating FID). In this method, the mobile phase was composed of
methods for determination of LAC and its related substan- helium gas and a temperature gradient was applied to the
ces published through 2016, but did not include any column that showed an elution of LAC at approximately
CRITICAL REVIEWS IN ANALYTICAL CHEMISTRY 9

5.7 min without any interference from peaks formed in deg- (compound 51; Appendix 1). This methodology enabled
radation conditions.[76] rapid analyzes with an elution of ESA and product in 2.6
Degradation products (compounds 8,11,14-17,24-30; and 1.8 min, respectively.[82] An HPTLC methodology, using
Appendix 1) of LAC were formed by oxidation (30% H2O2 toluene, methanol and acetone (60: 20:20, v/v/v) as the
at 100  C for 0.5 h), acidic and alkaline hydrolysis in differ- mobile phase, also proved to be selective for the analysis of
ent concentrations of stressors (0.1 M and 2 M HCl; 0.05 M, samples from forced degradation studies without interfer-
1 M and 2 M NaOH), reaction times and temperature. ence in the elution of ESA.[97]
Under thermal and photolytic conditions, LAC showed Two degradation products (compound 33 and 51;
insignificant degradation.[62,67,68,76] Appendix 1) of ESA described in the literature were formed
by oxidation (reflux with 30% H2O2 for 24 h), neutral
hydrolysis (60  C for 48 h) and, most notably, by acidic
4.4. Eslicarbazepine acetate (0.5 N HCl for 1 h and 2 N HCl for 15 min, both in room
Analytical methods described in the literature for determin- temperature) and alkaline hydrolysis (0.001 N NaOH for
ing ESA in the presence of its 21 related substances (com- 15 min and 0.015 N NaOH for 2 min, both in room
pounds 31-51; Appendix 1) widely employed HPLC[77–81]. temperature).[78,82]
Two reported on the use of UPLC[82] and HPTLC.[97] Two
different HPLC methodologies were developed in same 4.5. Perampanel
study for the separation of ESA and its (R)-enantiomer
(compound 31; Appendix 1) in API.[77] In these methods, Analytical methodologies for the separation of PER and 13
chiral separation was performed through a Chiralpak-IC-3 of its related substances (compounds 52-64; Appendix 1)
column that is based on tris-(3,5-dichlorophenyl carbamate) were performed by HPLC-UV and UPLC-UV.[83–86] The
of cellulose. One used a normal phase containing dichloro- chromatographic conditions employed columns of C18 and
methane and ethanol, and the other used an organic polar C8, a mobile phase composed by phosphate buffer, acidified
solvent, 100% acetonitrile. In the normal phase method, the water and acetonitrile in isocratic and gradient mode, and a
retention times were 4.24 min for the (R)-enantiomer and detection in the UV range of 220 to 290 nm. Saida et al.[86]
6.77 min for ESA with a resolution of 6. In the polar organic UPLC – UV methodology is the most sensitive, with shorter
method, the resolution was 3 with longer retention times of retention times for PER and its degradation products (com-
10.18 and 11.77 min for (R)-enantiomer and ESA, respect- pounds 52 and 53, Appendix 1). However, Xia et al.[83]
ively.[77] Control of the enantiomeric purity ensures that methodology even with longer retention time and run time,
during hydrolysis there is a greater conversion of the pro- it is the most advantageous for separating PER from thirteen
drug into the active metabolite (S)-licarbazepine and not the related substances (compounds 52-64, Appendix 1).
formation of carbamazepine-10, 11-epoxide, which is the HPLC-MS was used again to support in the identification
metabolite responsible for adverse effects.[39] of degradation products. In one study, the determination of
In the Mohamed et al (2016) review are described three the mass of degradation products (compounds 52, 53, 62-64;
stability indicating methods.[78,80,81] The first method Appendix 1) was performed using an HPLC-MS (triple
described uses column C8 and was developed for the separ- quadrupole) methodology with ionization by ESI in positive
ation of ESA in face of potential bulk impurities and deg- mode. The coupling to MS differed extensively from the
radation products (compounds 32-46, Appendix 1)[82]. This method for HPLC with a UV detection in relation to the
method proved to be more sensitive (LOD 0.02 mg.mL1) composition of mobile phase, time and gradient proportion,
than the other two methods described in the review, how- as well as column temperature.[84]
ever it presented longer run time (60 min) to obtain PER was shown to be stable under thermal and photo-
adequate resolution between the chromatographic peaks. lytic conditions although degradation products were formed
The other two methods used column C18 as a stationary under oxidation as well as acid and alkaline hydrolysis with
phase, with shorter retention times for ESA, when compared different concentrations of stressors (0.5 M and 1 M HCl;
to the first methodology. In addition, the methods proved to 1 M NaOH, H2O2 at 3% and 30%), temperatures (60 and
be selective in determining ESA in the presence of its related 100  C) and reaction times[84,86]
substances (compounds 33-35, 39, 41, 48-50, Appendix 1).
Thomas et al. (2012, 2014)[78,79] used the same HRMS
4.6. Brivaracetam
(HPLC-Ion trap) methodology at two works as support tool
for the identification of unknown impurities and degrad- BRV has four related substances with structures and nomen-
ation products. In this methodology, unlike the HPLC-UV clatures described in the literature, three diasteroisomers
method, ammonium bicarbonate buffer was used as the (compounds 65-67; Appendix 1) and one degradation prod-
mobile phase since it is volatile and compatible with ioniza- uct (compound 68; Appendix 1). Analytical methods devel-
tion by ESI and APCI (atmospheric pressure oped for BRV determination in presence of these substances
ionization).[78,79,98] were performed by HPLC and UPLC coupled to DAD/UV
An analytical method developed for UPLC used chroma- detectors.[87–90] In the HPLC-UV methods, a longer reten-
tographic conditions similar to the methodologies for HPLC tion time of 7.9 min was observed for BRV when methanol
in the separation of ESA from degradation product was used in mobile phase.[79] BRV retention time was
10 J. D. DASILVA ET AL.

reduced to 3.1 min when the mobile phase was composed of 2.6. However, the overall analysis time was greater than
acetonitrile and acidified water together with a column tem- 100 min.[92] EVR showed little degradation under the stress
perature that was maintained at 30  C.[88] In addition, sensi- conditions used in these two studies and without the formation
tivity was improved with value of LOD 2-fold lower than of degradation products.[91,92]
previous method.
In the UPLC-UV method, a C18 column was also used
with a gradient elution of mobile phase that resulted in a 5. Conclusion
BRV retention time of 3.1 min and that provided an This review covered the stability indicating methods for the
adequate separation from its impurities with resolution separation of third generation AEDs from their impurities
greater than 2.[89] Chiral separation of BRV and its three and degradation products. The frequency of the techniques
diastereomers was achieved by UPLC-UV using a Chiral used in the methods for obtaining fast and sensitive analyses
PAK IG-U column (tris (3, 5-chloromethylphenyl carba- were: HPLC (79%), UPLC (12%), HPTLC (5%), GC (2%)
mate) of amylose), ammonium bicarbonate buffer and aceto- and UV/Vis spectrophotometry (2%). Mass spectrometry
nitrile as the mobile phase in the isocratic mode. With these was employed mainly for the identification of impurities
chromatographic conditions, a complete analysis was and degradation products that provided support information
obtained in 15 min with an elution of BRV at 9.4 min and in the structural elucidation of these compounds. USP 43
separation of the stereoisomers with a resolution greater and Ph. Eur. 10th describe methods for separating RUF,
than 1.5. This chiral methodology was also used with an LAC and EVR from their related substances in dosage forms
UPLC-MS (Ion trap) with ionization by ESI to support in and API. The official methods, as well as most of the meth-
the identification of diasteroisomer (R,R)-brivaracetam odologies found in scientific articles, suggest the use of
(compound 66; Appendix 1) in alkaline hydrolysis.[90] liquid chromatography, UV detection and reverse phase col-
In forced degradation studies of BRV, the formation of dia- umns (C18 and C8). Chiral separation of LAC, ESA and
stereomer (R,R)-brivaracetam (compound 66; Appendix 1) was BRV enantiomers was performed using direct methods with
observed after alkaline hydrolysis in 1 N NaOH for 4 h. chiral separation columns. In general, the AEDs reported in
Another degradation product (compound 68; Appendix 1) was this review showed susceptibility to degradation under oxi-
also formed in alkaline hydrolysis in 1 N NaOH, but with 24 h dative condition as well as to acid and alkaline hydrolysis.
of reaction. In both studies, BRV was directly exposed to deg- Forced degradation studies along with the use of known
radation conditions without a co-solvent.[88,90] These studies impurities, allowed for the demonstration of analytical
indicated that BRV is stable under conditions of acidic, ther- methodologies selectivity. Some of these AEDs, having been
mal, photolytic and oxidative degradation.[90] only approved during the last decade, have few studies
focused on their stability and the identification of their
4.7. Everolimus impurities/degradation products. Importantly, this review
shows that further studies are clearly needed to contribute
EVR has 14 related substances described in the literature, of to the establishment of more complete safety profiles of
which, 11 have known chemical structures (compounds 69- these third-generation antiepileptic drugs.
79; Appendix 1). Among the analytical methods used for
determining EVR in presence of these impurities, two are
found in Eur. Ph. 10.0th and are applied for the analysis of Acknowledgments
the API.[49] The first methodology is for separation of EVR The authors acknowledge CAPES (Brasılia, Brazil), CNPq (Brasılia,
and sirolimus (compound 69; Appendix 1), that as discuss Brazil) and FAPERJ (Rio de Janeiro, Brazil). We are grateful to
in Section 3 is the compound from which EVR is derived. William Provence Jr for English review.
The second method was developed to determine EVR in the
presence of 7 impurities, 4 with known structures (com-
Disclosure statement
pounds 71-74; Appendix 1) and 3 unknown structures. The
two methodologies are reverse phase (column C18) with an The authors declare that there are no conflicts of interest.
elution in a gradient mode. Only for a second method, Eur.
Ph. 10.0th recommend resolutions at least 1.5 between EVR
Funding
and compound 70 and 72 (Appendix 1) peaks.
Another two methods found in the literature for the deter- This article was funded by Conselho Nacional de Desenvolvimento
mination of EVR in the presence of its related substances are Cientıfico e Tecnol
ogico;Coordenaç~ao de Aperfeiçoamento de Pessoal
de Nıvel Superior;Fundaç~ao Carlos Chagas Filho de Amparo a Pesquisa
also by reverse phase HPLC-UV.[91,92] The method with an iso- do Estado do Rio de Janeiro.
cratic elution, showed good sensitivity (LOD 0.036 mg.mL1)
and the EVR retention time was 3.1 min.[90] In other method, a
Quality by Design (QbD) approach by Design of Experiments
(DOE) was applied to determine EVR in presence of 8 impur- ORCID
ities (compounds 69-70, 75-79; Appendix 1). One of criteria for Jessica Domingos da Silva http://orcid.org/0000-0001-8728-1906
optimizing the method was the resolution between EVR and Lucio Mendes Cabral http://orcid.org/0000-0002-4550-5729
sirolimus (compound 69; Appendix 1) to achieve a value of Valeria Pereira de Sousa http://orcid.org/0000-0003-1589-0846
CRITICAL REVIEWS IN ANALYTICAL CHEMISTRY 11

References scripts/cder/daf/index.cfm?event=overview.process&ApplNo=
211199. (accessed Aug 15, 2020).
[1] World Health Organization. Epilepsy. https://www.who.int/ [23] EMA. Fycompa, Perampanel. https://www.ema.europa.eu/en/
news-room/fact-sheets/detail/epilepsy. (accessed Sept 13, 2020). medicines/human/EPAR/fycompa. (accessed Aug 15, 2020).
[2] Moshe, S. L.; Perucca, E.; Ryvlin, P.; Tomson, T. Epilepsy: New [24] FDA. Drugs@FDA: FDA-Approved Drugs. New Drug
Advances. Lancet 2015, 385, 884–898. DOI: 10.1016/S0140- Application (NDA): 208277. https://www.accessdata.fda.gov/
6736(14)60456-6. scripts/cder/daf/index.cfm?event=overview.process&applno=
[3] Sills, G. J.; Rogawski, M. A. Mechanisms of Action of Currently 208277. (accessed Aug 15, 2020).
Used Antiseizure Drugs. Neuropharmacology 2020, 168, [25] EMA. Briviact, Brivaracetam. https://www.ema.europa.eu/en/
107913–107966. DOI: 10.1016/j.neuropharm.2020.107966. medicines/human/EPAR/briviact-italy-nubriveo#authorisation-
[4] Landmark, C. J.; Johannessen, S. I.; Patsalos, P. N. Therapeutic details-section. (accessed Aug 15, 2020).
Drug Monitoring of Antiepileptic Drugs: Current Status and [26] FDA. FDA. Drugs@FDA: FDA-Approved Drugs. New Drug
Future Prospects. Expert Opin. Drug Metab. Toxicol. 2020, 16, Application (NDA): 205836. https://www.accessdata.fda.gov/
227–238. DOI: 10.1080/17425255.2020.1724956. scripts/cder/daf/index.cfm?event=overview.process&varApplNo=
[5] Singh, K. P.; Verma, N. Teratogenic Potential of Third- 205836. (accessed Aug 15, 2020).
Generation Antiepileptic Drugs: Current Status and Research [27] EMA. Votubia, Everolimus. https://www.ema.europa.eu/en/med-
Needs. Pharmacol. Rep. 2019, 71, 491–502. DOI: 10.1016/j. icines/human/EPAR/votubia. (accessed Aug 15, 2020).
pharep.2019.01.011. [28] FDA. Drugs@FDA: FDA-Approved Drugs. New Drug
[6] Patsalos, P.; Spencer, E.; Berry, D. Therapeutic Drug Application (NDA): 022334. H https://www.accessdata.fda.gov/
Monitoring of Antiepileptic Drugs in Epilepsy: A 2018 Update. scripts/cder/daf/index.cfm?event=overview.process&ApplNo=
Ther. Drug Monit. 2018, 40, 526–548. DOI: 10.1097/FTD. 022334. (accessed Aug 17, 2020).
0000000000000546. [29] Rogawski, M. A.; L€oscher, W.; Rho, J. M. Mechanisms of
[7] Maggio, R. M.; Vignaduzzo, S. E.; Kaufman, T. S. Practical and Action of Antiseizure Drugs and the Ketogenic Diet. Cold
Regulatory Considerations for Stability-Indicating Methods for Spring Harb. Perspect. Med. 2016, 6, a022780–28. DOI: 10.1101/
the Assay of Bulk Drugs and Drug Formulations. TrAC - Trends cshperspect.a022780.
Anal. Chem. 2013, 49, 57–70. DOI: 10.1016/j.trac.2013.05.008. [30] Scifinder. https://scifinder.cas.org (acessed May, 2020).
[8] Santulli, L.; Coppola, A.; Balestrini, S.; Striano, S. The Challenges [31] Pubchem. hppts://pubchem.ncbi.nlm.nih.gov/ (acessed May, 2020).
of Treating Epilepsy with 25 Antiepileptic Drugs. Pharmacol. Res. [32] Almeida, L.; Soares-da-Silva, P. Eslicarbazepine Acetate (BIA
2016, 107, 211–219. DOI: 10.1016/j.phrs.2016.03.016. 2-093). Neurotherapeutics 2007, 4, 88–96. DOI: 10.1016/j.nurt.
[9] de Biase, S.; Nilo, A.; Bernardini, A.; Gigli, G. L.; Valente, M.; 2006.10.005.
Merlino, G. Timing Use of Novel anti-Epileptic Drugs: Is [33] Hibi, S.; Ueno, K.; Nagato, S.; Kawano, K.; Ito, K.; Norimine,
Earlier Better? Expert Rev. Neurother. 2019, 19, 945–954. DOI: Y.; Takenaka, O.; Hanada, T.; Yonaga, M. Discovery of 2-(2-
10.1080/14737175.2019.1636649. oxo-1-phenyl-5-pyridin-2-yl-1,2-dihydropyridin-3-yl)benzoni-
[10] Perucca, E.; Brodie, M. J.; Kwan, P. Tomson, T. 30 Years of trile (perampanel): A Novel, Noncompetitive a-Amino-3-
Second-Generation Antiseizure Medications: Impact and Future hydroxy-5-methyl-4-isoxazolepropanoic acid (AMPA) Receptor
Perspectives. Lancet Neurol. 2020, 4422, 1–12. DOI: 10.1016/ Antagonist. J. Med. Chem. 2012, 55, 10584–10600. DOI: 10.
s1474-4422(20)30035-1. 1021/jm301268u.
[11] Liu, D. Q.; Sun, M.; Kord, A. S. Recent Advances in Trace [34] Perucca, E.; Cloyd, J.; Critchley, D.; Fuseau, E. Rufinamide:
Analysis of Pharmaceutical Genotoxic Impurities. J. Pharm. Clinical Pharmacokinetics and Concentration-Response
Biomed. Anal. 2010, 51, 999–1014. DOI: 10.1016/j.jpba.2009.11.009. Relationships in Patients with Epilepsy. Epilepsia 2008, 49,
[12] International Conference Harmonisation Q3A (R2): Impurities 1123–1141. DOI: 10.1111/j.1528-1167.2008.01665.x.
in New Drug Substances, ICH Harmon. Tripart. Guidel, 2006. [35] Trojnar, M. K.; Wojtal, K.; Trojnar, M. P.; Czuczwar, S. J.
[13] International Conference on Harmonisation. Q3B (R2): Stiripentol. A Novel Antiepileptic Drug. Pharmacol. Rep. 2005,
Impurities in New Drug Products. ICH Harmon. Tripart. 57, 154–160.
Guidel, 2006. [36] Czuczwar, S. J.; Trojnar, M. K.; Gergont, A.; Kroczka, S.;
[14] Jain, D.; Basniwal, P. K. Forced Degradation and Impurity Kacinski, M. Stiripentol - Characteristic of a New Antiepileptic
Profiling: Recent Trends in Analytical Perspectives. J. Pharm. Drug. Expert Opin. Drug Discov. 2008, 3, 453–460. DOI: 10.
Biomed. Anal. 2013, 86, 11–35. DOI: 10.1016/j.jpba.2013.07.013. 1517/17460441.3.4.453.
[15] EMA. Inovelon, Rufinamide. https://www.ema.europa.eu/en/ [37] Beyreuther, B. K.; Freitag, J.; Heers, C.; Krebsf€anger, N.;
medicines/human/EPAR/inovelon. (accessed Aug 17, 2020). Scharfenecker, U.; St€ ohr, T. Lacosamide: A Review of
[16] FDA. Drugs@FDA: FDA-Approved Drugs. New Drug Preclinical Properties. CNS Drug Rev. 2007, 13, 21–42. DOI: 10.
Application (NDA): 021911. https://www.accessdata.fda.gov/ 1111/j.1527-3458.2007.00001.x.
scripts/cder/daf/index.cfm?event=overview.process&applno= [38] Rogawski, M. A.; Tofighy, A.; White, H. S.; Matagne, A.; Wolff,
021911. (accessed Aug 17, 2020). C. Current Understanding of the Mechanism of Action of the
[17] EMA. Diacomit, Stiripentol. https://www.ema.europa.eu/en/ Antiepileptic Drug Lacosamide. Epilepsy Res. 2015, 110,
medicines/human/EPAR/diacomit. (accessed Aug 17, 2020). 189–205. DOI: 10.1016/j.eplepsyres.2014.11.021.
[18] FDA. Drugs@FDA: FDA-Approved Drugs. New Drug [39] Rocamora, R. A Review of the Efficacy and Safety of
Application (NDA): 206709. https://www.accessdata.fda.gov/ Eslicarbazepine Acetate in the Management of Partial-Onset
scripts/cder/daf/index.cfm?event=overview.process&ApplNo= Seizures. Ther. Adv. Neurol. Disord. 2015, 8, 178–186. DOI: 10.
206709. (accessed Aug 17, 2020). 1177/1756285615589711.
[19] EMA. Vimpat, Lacosamide. https://www.ema.europa.eu/en/med- [40] Malawska, B.; Kulig, K. Brivaracetam: A New Drug in
icines/human/EPAR/vimpat. (accessed Aug 15, 2020). Development for Epilepsy and Neuropathic Pain. Expert Opin.
[20] FDA. FDA. Drugs@FDA: FDA-Approved Drugs. New Drug Investig. Drugs 2008, 17, 361–369. DOI: 10.1517/13543784.17.3.361.
Application (NDA): 022253. https://www.accessdata.fda.gov/ [41] Qiu, S.; Tehrani, K. A.; Sergeyev, S.; Bultinck, P.; Herrebout, W.;
scripts/cder/daf/index.cfm?event=overview.process&ApplNo= Mathieu, B. Stereochemistry of the Brivaracetam Diastereoisomers.
022253. (accessed Aug 15, 2020). Chirality 2016, 28, 215–225. DOI: 10.1002/chir.22558.
[21] EMA. Zebinix, Esliscarbazepine Acetate. https://www.ema.europa. [42] European Medicines Agency, E. Votubia, Everolimus. Comm.
eu/en/medicines/human/EPAR/zebinix. (accessed Aug 15, 2020). Med. Prod. Hum. Use. 2011, 44, 0–88.
[22] FDA. FDA. Drugs@FDA: FDA-Approved Drugs. New Drug [43] Jang, S. W.; Kang, M. J. Improved Oral Absorption and
Application (NDA): 211199. https://www.accessdata.fda.gov/ Chemical Stability of Everolimus via Preparation of Solid
12 J. D. DASILVA ET AL.

Dispersion Using Solvent Wetting Technique. Int. J. Pharm. Development, Validation, Kinetics, Structure Elucidation and
2014, 473, 187–193. DOI: 10.1016/j.ijpharm.2014.06.006. Application to Commercial Dosage Form. J Anal Methods
[44] Moraes do Carmo, A. C.; Pereira, R. S.; Gratieri, T. Brazilian Chem. 2014, 2014, 638910–638951. DOI: 10.1155/2014/638951.
Requirements for Stability Indicating Methods. TrAC - Trends [62] Shah, S.; Vasantharaju, S. G.; Karthik, A.; Muddukrishna, B. S.
Anal. Chem. 2018, 98, 58–63. DOI: 10.1016/j.trac.2017.10.017. Development and Validation of Stability-Indicating Assay
[45] Sengupta, P.; Chatterjee, B.; Tekade, R. K. Current Regulatory Method for Lacosamide by RP- HPLC. Elixir Int. J. 2011, 38,
Requirements and Practical Approaches for Stability Analysis of 4174–4177.
Pharmaceutical Products: A Comprehensive Review. Int. J. [63] Sreenivasulu, V.; Rao, D. R.; Maheswari, B. N. U.; Das, S. K.;
Pharm. 2018, 543, 328–344. DOI: 10.1016/j.ijpharm.2018.04.007. Krishnaiah, A. Development and Validation of a Stabiliting-
[46] Ramachandra, B. Critical Reviews in Analytical Chemistry Indicating RP-HPLC Method for Determination of Lacosamide.
Development of Impurity Profiling Methods Using Modern Res. J. Pharm. Biol. Chem. Sci. 2011, 2, 1–11.
Analytical Techniques. Crit. Rev. Anal. Chem. 2017, 47, 24–36. [64] Chhalotiya, U. K.; Bhatt, K. K.; Shah, D. A.; Baldania, S. L.;
DOI: 10.1080/10408347.2016.1169913. Patel, J. R. Stability-Indicating Liquid Chromatographic Method
[47] Zhang, K.; Kurita, K. L.; Venkatramani, C.; Russell, D. Seeking for Quantification of New anti-Epileptic Drug Lacosamide in
Universal Detectors for Analytical Characterizations. J. Pharm. Bulk and Pharmaceutical Formulation. CI&CEQ. 2012, 18,
Biomed. Anal. 2019, 162, 192–204. DOI: 10.1016/j.jpba.2018.09.029. 35–42. DOI: 10.2298/CICEQ110821044C.
[48] USP. United States Pharmacopeia, 43th ed.; United States [65] Chakravarthy, V. K.; Sankar, D. G. Stability Indicating Hplc
Convention: Rockville, 2020. Method for Determination of Lacosamide and Its Degradants/
[49] Eur. Ph. European Pharmacopoeia, 10th Ed.; Council of Europe: Impurities in Bulk and Pharmaceutical Formulation. Rasayan J.
Strasbourg, 2019. Chem. 2012, 5, 293–310.
[50] Annapurna, M. M.; Goutam, S.; Pavani, S. New Stability [66] Patil, S. N.; Agrawal, P. N.; Kadam, A.; Manish, M.; Askshay, S.
Indicating Liquid Chromatographic Method for the Development and Validation of Stability Indicating RP-HPLC
Determination of Rufinamide in Presence of Degradant Method for Estimation of Lacosamide in Bulk and Its
Products. Drug Invent. Today 2012, 4, 501–506. DOI: 10.5958/ Pharmaceutical Formulations. Int. J. Pharm. Sci. Rev. Res. 2014,
0974-360X.2018.00705.9. 28, 164–168.
[51] Mahamuni, B. S.; Srinivas, R.; Talluri, M. V K. Characterization [67] Tiwari, R. N.; Bonde, C. G. Identification and Characterization of
of Forced Degradation Products of Rufinamide by LC/QTOF/ Degradation Products of Lacosamide by Liquid-Chromatography/
MS/MS, NMR and IR Studies. Anal. Chem. Lett. 2018, 8, Time-of-Flight Mass Spectrometric and Multi-Stage Mass
405–415. DOI: 10.1080/22297928.2018.1438315. Spectrometric Analysis. J. Liq. Chromatogr. Relat. Technol. 2014,
[52] Kumar, B. S. P.; Annapurna, M. M.; Pavani, S. Development 37, 2046–2061. DOI: 10.1080/10826076.2013.825860.
and Validation of a Stability Indicating RP-HPLC Method for [68] Ramisetti, N. R.; Kuntamukkala, R.; Lakshetti, S.; Sripadi, P.
the Determination of Rufinamide. J. Pharm. Anal. 2013, 3, Identification and Characterization of Stress Degradants of
66–70. DOI: 10.1016/j.jpha.2012.08.003. Lacosamide by LC-MS and ESI-Q-TOF-MS/MS: Development
[53] Patel, A.; Suhagia, B. N.; Patwari, A. Development and and Validation of a Stability Indicating RP-HPLC Method. J.
Validation of Stability Indicating HPLC Method for Estimation Pharm. Biomed. Anal. 2014, 95, 256–264. DOI: 10.1016/j.jpba.
of Rufinamide in Bulk and Its Pharmaceutical Dosage Form. 2014.03.010.
World J. Pharm. Res. 2014, 3, 1798–1810. [69] Mahesh, H. R. K.; Babu, S. K. Quantitative Estimation of
[54] Hassib, S. T.; Hashem, H. M. A.; Mahrouse, M. A.; Mostafa, Related Compounds of Lacosamide in Oral Solution by Using
E. A. Determination of Rufinamide in the Presence of 1-[(2,6- Reverse Phase HPLC. Der Pharm. Lett. 2015, 7, 285–291.
Difluorophenyl)Methyl]-1H-1,2,3-Triazole-4 Carboxylic Acid [70] Kalyan Chakravarthy, V.; Gowri Shankar, D. HPLC Method for
Using RP-HPLC and Derivative Ratio Methods as Stability Determination of Lacosamide S(-)Enantiomer in Bulk and
Indicating Assays to Be Applied on Dosage Form. J. AOAC Int. Pharmaceutical Formulation. Rasayan J. Chem. 2011, 4, 744–752.
2020, 103, 1–8. DOI: 10.1093/jaoacint/qsaa020. [71] Parmar, M.; Nimavat, K.; Vyas, K.; Rao, D.; Pande, R. A
[55] Annapurna, M. M.; Kumar, B. S. P.; Goutam, S.; Srinivas, L. Stability-Indicating Liquid Chromatographic Method for the
Stability Indicating Liquid Chromatographic Method for the Quantification of New anti-Epileptic Drug Lacosamide and Its
Quantitative Determination of Rufinamide in Pharmaceutical Intermediates. Int. J. Pharm. Res. Sch. 2012, 1, 40–47.
Dosage Forms. J. Drug Deliv. Ther. 2012, 2, 167–174. DOI: 10. [72] Charagondla, K. A Validated Chiral Liquid Chromatographic
22270/jddt.v3i3.498. Method for the Enantiomeric Separation of Lacosamide Drug
[56] Ngumo, P.; Abuga, K. O.; Njogu, P. M.; Ongarora, D. S. B. A Product and Its Dosage Forms. J. Chromatogr. Sep. Tech 2015,
Stability Indicating Liquid Chromatography Method for the 06, 1–9. DOI: 10.4172/2157-7064.1000280.
Assay of Rufinamide Bulk Material and Tablets. East Cent. [73] Zhou, N.; Li, T.; Ai, L.; Guo, C.; Zhang, J.; Fu, S.; Wang, Q.
African J. Pharm. Sci. 2016, 19, 16–21. DOI: 10.1017/ Identification of Forced Degradation Products of Lacosamide
CBO9781107415324.004. by LC-QQLIT-MS and LC-IT/TOF-MS. Acta Chromatogr.
[57] Snyder, J. K. Introduction to Modern Liquid Chromatography, 2019, 31, 12–18. DOI: 10.1556/1326.2018.00279.
John Wiley and Sons, New York, 2011. [74] Molleti, S.; Rao, V.; Jayaveera, K. N. Stability Indicating RP-
[58] Ranjith, K.; Rao, M.; Murthy, T. Development and Validation UPLC Method for the Determination of Lacosamide and Its
of Related Substances Method for Rufinamide Tablets by RP- Impurities in Bulk Drugs and Its Pharmaceutical Dosage
HPLC. J. Chem. Pharm. Res. 2017, 4, 306–314. DOI: 10.26420/ Forms. Der. Pharma. Chem. 2013, 5, 81–89.
austinjanalpharmchemg.2017.1087. [75] Patel, A.; Suhagia, B. N.; Patwari, A. Stability Indicating Assay
[59] Geetha, M.; Sait, S.; Sripal Reddy, P. A Stability Indicating Method for Quantification of Lacosamide in Bulk and Its
UPLC Method for the Estimation of Related Substances, Assay Pharmaceutical Dosage Form and Characterization of Major
and Dissolution of Rufinamide. Asian J. Chem. 2013, 25, Degradation Products. Int. J. Pharm. Pharm. Sci. 2013, 6, 593–599.
9775–9778. DOI: 10.14233/ajchem.2013.15314. [76] Korany, M. A.; Mahgoub, H.; Haggag, R. S.; Ragab, M. A. A.;
[60] Patel, A.; Patwari, A.; Suhagia, B. Development of a Validated Elmallah, O. A. Green Gas Chromatographic Stability-
Stability-Indicating HPTLC Method for Rufinamide in Bulk Indicating Method for the Determination of Lacosamide in
and Its Pharmaceutical Dosage Form. J. Chromatogr. Sci. 2014, Tablets. Application to in-Vivo Human Urine Profiling. J.
52, 1294–1301. DOI: 10.1093/chromsci/bmt171. Chromatogr. B Analyt. Technol. Biomed. Life Sci. 2018, 1083,
[61] Darwish, H. W.; Abdelhameed, A. S.; Attia, M. I.; Bakheit, 75–85. DOI: 10.1016/j.jchromb.2018.02.033.
A. H.; Khalil, N. Y.; Al-Majed, A. A. A. A Stability-Indicating [77] Mone, M. K.; Chandrasekhar, K. B. Development of Liquid
HPLC-DAD Method for Determination of Stiripentol: Chromatographic Enantiomer Separation Methods and
CRITICAL REVIEWS IN ANALYTICAL CHEMISTRY 13

Validation for the Estimation of (R)-Enantiomer in Stability – Indicating HPLC Assay Method. IJSRM Human
Eslicarbazepine Acetate. J. Pharm. Biomed. Anal. 2011, 54, 2016, 4, 43–57.
248–251. DOI: 10.1016/j.jpba.2010.08.015. [88] Bhamare, P.; Umadoss, P.; Upmanyu, N.; Dubey, R.
[78] Thomas, S.; Bharti, A.; Maddhesia, P. K.; Shandilya, S.; Identification, Isolation, Structural Characterisation, Synthesis
Agarwal, A.; Dharamvir; Biswas, S.; Bhansal, V.; Gupta, A. K.; and in-Silico Toxicity Prediction of Alkaline Hydrolytic
Tewari, P. K.; Mathela, C. S. Highly Efficient, Selective, Degradation Product of Brivaracetam by Using LC-PDA,
Sensitive and Stability Indicating RP-HPLC-UV Method for the Preparative HPLC, LC/HESI/LTQ, FTIR, 1H NMR. Anal.
Quantitative Determination of Potential Impurities and Methods 2020, 12, 1868–1881. DOI: 10.1039/C9AY02582K.
Characterization of Four Novel Impurities in Eslicarbazepine [89] Vishweshwar, V.; Babu, J. M.; Muralikrishna, R.;
Acetate Active Pharmaceutical Ingredient by LC/ES. J. Pharm. Pharmaceutical, C.; Pradesh, A. Development and Validation of
Biomed. Anal. 2012, 61, 165–175. DOI: 10.1016/j.jpba.2011.11. Stability-Indicating Uplc Method for the Determination of
024. Brivaracetam, Its Related Impurities and Degradation Products.
[79] Thomas, S.; Paul, S. K.; Joshi, S. C.; Kumar, V.; Agarwal, A.; Int. J. Pharm. Sci. Res. 2018, 9, 2315–2327. DOI: 10.13040/
Vir, D. Vir, D. Identification, Synthesis and Characterization of IJPSR.0975-8232.9(6).2315-27.
an Unknown Process Related Impurity in Eslicarbazepine [90] Baksam, V.; Saritha, N.; Pocha, V. R.; Chakka, V. B.; Ummadi,
Acetate Active Pharmaceutical Ingredient by LC/ESI-IT/MS, R. R.; Kumar, P. Development of an Effective Novel Validated
1H, 13C and 1H-1H COSY NMR. J. Pharm. Anal. 2014, 4, Stability-Indicating HPLC Method for the Resolution of
339–344. DOI: 10.1016/j.jpha.2013.08.004. Brivaracetam Stereoisomeric Impurities. Chirality 2020, 32,
[80] Srinivas, M.; Avupati, N. R.; Sait, S.; Mukkanti, K. Stability 1208–1212. DOI: 10.1002/chir.23269.
Indicating HPLC Method for the Determination of [91] Sharmila, D.; Rao Lakshmana, A.; Kalyani, L. Development and
Eslicarbazepine Acetate and Its Impurities in Bulk Drugs and Validation of Stability-Indicating High Performance Liquid
Pharmaceutical Dosage Forms. J. Liq. Chromatogr. Relat. Chromatographic Method for the Estimation of Everolimus in
Technol. 2012, 35, 1550–1564. DOI: 10.1080/10826076.2011. Tablets. Indian J. Pharm. Sci. 2015, 77, 599–604. DOI: 10.4103/
619043.
0250-474x.169044.
[81] Kallam, S. R.; Srikanth, J.; Prakash, K. V. Development and
[92] Prasad, S. S.; Krishna Mohan, G. V.; Naga Babu, A.
Validation of an RP-HPLC Method for Related Substance and
Development of Simple and Robust RP-HPLC Method for
Quantitative Estimation of Eslicarbazepine Acetate in Bulk
Determination of Everolimus and Its Impurities in Oral Solid
Drug and Pharmaceutical Dosage Form. J. Pharm. Biol. Sci.
Dosage Form. Asian J. Chem. 2019, 31, 1002–1008. DOI: 10.
2015, 10, 155–162. DOI: 10.9790/3008-1062155162.
14233/ajchem.2019.21723.
[82] Iram, F.; Alam, P.; Siddiqui, N. A.; Alqasoumi, S. I.; Siddiqui,
[93] Kumaraswamy, G.; Lalitha, R.; Vijaypraksh, K. Method
A. A.; Khan, S. A.; Husain, A. Development of a Stress Induced
Development and Validation for Estimation of Rufinamide in
Validated UPLC-PDA Method for the Analysis of
Eslicarbazepine Acetate. Saudi Pharm J 2018, 26, 286–291. Tablet Dosage Forms by RP-HPLC. Int. J. Pharma. Chem. Anal.
DOI: 10.1016/j.jsps.2017.11.009. 2016, 3, 99. DOI: 10.5958/2394-2797.2016.00015.0.
[83] Patel, C. J.; Patel, S. S.; Patel, M. M. Method Development and [94] Parashar, V.; Todkar, M.; Shaikh, F.; Reddy, S.; Mehra, V.;
Stability Study by Chromatographic Method for Perampanel in Sinha, S. Validated Liquid Chromatographic Method for
API and Tablet Dosage Form. Int. J. Pharm. Drug Anal. 2017, Quantitative Determination of Rufinamide Active
5, 229–240. Pharmaceutical Ingredient Form and Its Impurities. Pharm.
[84] Xia, Y.-Y.; Zou, Q.-G.; Yang, Y.-F.; Sun, Q.; Han, C.-Q. Methods 2013, 4, 6–10. DOI: 10.1016/j.phme.2013.08.001.
Determination of Impurities in Perampanel Bulk Drugs by [95] Mohamed, F. A.; Bakr, M. F.; Rageh, A. H.; Mostafa, A. M. The
High-Performance Liquid Chromatography and Gas Use of Separation Techniques in the Analysis of Some
Chromatography. Curr. Pharm. Anal. 2020, 16, 1–12. DOI: 10. Antiepileptic Drugs: A Critical Review. J. Liq. Chromatogr.
2174/1573412916999200513105657. Relat. Technol. 2016, 39, 783–798. DOI: 10.1080/10826076.2016.
[85] Annapurna, M. M.; Kaibada, R.; Srinivas, M. New Stability 1266654.
Indicating Rp-Ultra Fast Liquid Chromatographic Method for [96] Valarmathi, R.; Banu, S. F.; Akilandeswari, S.; Senthamarai, R.;
the Determination of Perampanel–an Antiepileptic Drug. Rese. Dhivya, C. S. A Review on New Antiepileptic Drug –
J. Pharm. Technol. 2019, 12, 2657–2663. DOI: 10.5958/0974- Lacosamide and Its Analytical Methods. Int. J. Pharm. Chem.
360X.2019.00444.X. Sci. 2013, 2, 181–186.
[86] Saida, S. J.; Muthuchamy, M.; Kaliyaperumal, M.; Rumalla, S.; [97] Mali, N. V.; Bansode, D. A. Stability Indicating Thin-Layer
Yanaka, R.; Rao, S. Isolation and Spectral Characterization of Chromatographic Determination of Eslicarbazepine Acetate as
Degradation Impurity in Perampanel Drug Substance Using Bulk Drug: Application to Forced Degradation Study. Der
UPLC-MS and NMR Spectroscopy: Validation of Assay Method Pharm. Lett. 2016, 8, 38–47.
by UPLC. Asian J. Chem. 2018, 30, 2215–2219. DOI: 10.14233/ [98] Narayanam, M.; Sahu, A.; Singh, S. Use of LC-MS/TOF, LC-
ajchem.2018.21405. MS(n), NMR and LC-NMR in characterization of stress degrad-
[87] Mali, N. V.; Mhaske, D. V. HPLC Studies on Degradation ation products: Application to cilazapril. J. Pharm. Biomed.
Behavior of Brivaracetam and Development of Validated Anal. 2015, 111, 190–203. DOI: 10.1016/j.jpba.2015.03.038.

You might also like