Sei sulla pagina 1di 16

J. Sep. Sci.

2015, 00, 1–16 1

Ambavaram Vijaya Bhaskar Review


Reddy1
Jafariah Jaafar1
Khalid Umar2 Identification, control strategies, and
Zaiton Abdul Majid1
Azmi Bin Aris2 analytical approaches for the determination
Juhaizah Talib2
Gajulapalle Madhavi3
of potential genotoxic impurities in
1 Department of Chemistry,
pharmaceuticals: A comprehensive review
Faculty of Science, Universiti
Teknologi Malaysia, Johor, Potential genotoxic impurities in pharmaceuticals at trace levels are of increasing concern to
Malaysia
2 Department of Environmental both pharmaceutical industries and regulatory agencies due to their possibility for human
Engineering, Faculty of Civil carcinogenesis. Molecular functional groups that render starting materials and synthetic
Engineering, Universiti intermediates as reactive building blocks for small molecules may also be responsible for
Teknologi Malaysia, Johor, their genotoxicity. Determination of these genotoxic impurities at trace levels requires highly
Malaysia
3 Department of Chemistry, Sri sensitive and selective analytical methodologies, which poses tremendous challenges on an-
Venkateswara University, alytical communities in pharmaceutical research and development. Experimental guidance
Tirupati, India for the analytical determination of some important classes of genotoxic impurities is still
unavailable in the literature. Therefore, the present review explores the structural alerts of
Received October 24, 2014 commonly encountered potential genotoxic impurities, draft guidance of various regulatory
Revised December 12, 2014 authorities in order to control the level of impurities in drug substances and to assess their
Accepted December 16, 2014 toxicity. This review also describes the analytical considerations for the determination of
potential genotoxic impurities at trace levels and finally few case studies are also discussed
for the determination of some important classes of potential genotoxic impurities. It is
the authors’ intention to provide a complete strategy that helps analytical scientists for the
analysis of such potential genotoxic impurities in pharmaceuticals.

Keywords: Alkyl halides / Control strategies / Regulatory guidance / Toxicology /


Trace analysis
DOI 10.1002/jssc.201401143

1 Introduction

During the manufacturing of active pharmaceutical ingredi-


ents (APIs) some starting materials, intermediates, reagents,
Correspondence: Dr. Jafariah Jaafar, Department of Chemistry,
Faculty of Science, Universiti Teknologi Malaysia, Johor Bahru, and reaction byproducts inevitably end up in the final prod-
Johor 81310, Malaysia ucts as impurities [1]. The safety of a drug product is de-
E-mail: jafariah@kimia.fs.utm.my pendent not only on the toxicological properties of the active
drug substance itself, but on the impurities it contains. Or-
Abbreviations: ALARP, as low as reasonably practicable;
ganic impurities that have the potential to induce genetic
APIs, active pharmaceutical ingredients; APP, (2S)-2-amino-
3-(4-aminophenyl)propan-1-ol; CCP, 1-(3-chloropropyl)-4- mutations, chromosomal breaks, and/or chromosomal rear-
(3-chlorophenyl) piperazine HCl; CDP, 2-chloromethyl-3, rangements are considered as potential genotoxic impurities
4-dimethoxy pyridine hydrochloride; DEREK, deductive (PGIs), which may cause cancer in humans [2, 3]. Therefore,
estimation of risk from existing knowledge; EMA, Eu- there is an ever-increasing interest in controlling and mon-
ropean Medicines Agency; FID, flame ionization detector; itoring of PGIs in APIs [4–6]. The process chemists always
HS-GC, headspace gas chromatography; ICH, Interna- try to explore the possible opportunities to avoid the use and
tional Conference on Harmonization; MAP, (4-methyl-
generation of these genotoxic substances in the manufactur-
acetophenone)-p-sulfonamide phenylhydrazine hydrochlo-
ride; MCASE, multicomputer-automated structure evalua- ing process. However, the complete elimination/prevention
tion; MNA, (S)-methyl-4-nitrophenylalanate hydrochloride; of such impurities is always not achievable [7], and if not ad-
NPA, (S)-4-nitrophenylalanine hydrate; NPP, (S)-2-amino-3- dressed correctly these PGIs could lead to clinical holds or
(4-nitrophenyl)propanol; PCV, N-phenoxycarbonyl-L-valine; delays the approval from regulatory agencies. This poses an
PhRMA, pharmaceutical research and manufacturers of imperative challenge on analytical scientists to develop ap-
America; PGI, potential genotoxic impurity; QL, quanti-
propriate analytical methodologies to accurately measure as
tation limit; R&D, research and development; SHH, (4-
sulfamoylphenyl)hydrazine hydrochloride; TTC, threshold
well as to control the levels of PGIs in pharmaceuticals. In
of toxicological concern; USFDA, U.S. Food and Drug addition to the process impurities, certain drugs may also
Administration generate PGIs via degradation during the synthetic process,


C 2014 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.jss-journal.com
2 A. V. B. Reddy et al. J. Sep. Sci. 2015, 00, 1–16

Table 1. PhRMA, USFDA, and EMA recommended acceptable generation of potential genotoxic chemicals in any new manu-
qualification thresholds for genotoxic impurities in phar- facturing processes should be minimized whenever possible,
maceuticals in clinical studies
but the complete removal of PGIs from the drug substances
or drug products is often impractical. This necessitates that
Duration of exposure Limit (␮g/day)
the PGIs should be tested in the final products to ensure
PhRMA USFDA EMA the product quality and patient safety. Moreover, from the
process understanding point of view, the analytical methods
Single dose 120 120 120 that are capable of measuring the PGIs at trace levels are
<14 days 120 60 120 needed to monitor the fate of PGIs during chemical synthe-
14 days to 1 month 120 60 60 sis. This in turn allows for the establishment of effective PGI
1–3 months 40 20 20 control strategies. In response to the USFDA recent quality-
3–6 months 20 10 10 by-design initiative, the pharmaceutical industry has been
6–12 months 10 5 5
struggling to gain process understanding and to implement
>12 months 1.5 1.5 1.5
the process controls. This requires analytical chemists to de-
liver sensitive and robust analytical methods to support pro-
cess development, thereby an effective control strategy can
storage, formulation of dosage forms, and aging [8, 9]. For be drafted and implemented to ensure the API quality with
instance, penicillins and cephalosporins are classic examples respect to PGIs concentration [20]. Furthermore, in cases
for the formation of degradation products [10]. Another exam- where multiple structurally similar PGIs are identified, they
ple is the formation of oxidative degradation products such are assumed to have same mode of action and similar mech-
as hydroperoxides/epoxides and hydrolytic products such as anisms for toxicity, and might exert effects in an additive
anilines are of potential genotoxicity concern. In addition, manner. Therefore EMA recommended that the appropriate
the components in excipients may react with APIs or its individual limit should be applied to the sum of structurally
counterions and forms a new class of impurities, which similar PGIs. Hence the TTC becomes the total exposure to
have the genotoxic concern (e.g., halogenated furanones) [11], all of the related compounds and the sum of the genotoxic
this burdens the drug development process with additional impurities should not exceed 1.5 ␮g/day [21]. These consid-
roadblocks. erations pose tremendous a challenge on analytical method
Regulatory guidance acknowledge that the identified sensitivity, and lower the analytical testing limit by several
PGIs are unusually toxic and require lower reporting, iden- folds beyond an already very low limit, which brings addi-
tification, and qualification limits than outlined in the Inter- tional challenges to the analytical scientists in pharmaceutical
national Conference on Harmonization (ICH) guidelines for R&D.
typical impurities in drug substances or drug products [12,13]. Ideally, conventional analytical instrumentation in the
A guidance on the genotoxic impurities from the European pharmaceutical analysis such as HPLC with UV detection
Medicines Agency (EMA) was issued in 2006 followed by (for typical nonvolatile analytes) or GC with flame ioniza-
three rounds of questions and answers, providing a clarifi- tion detection (FID; for volatile small molecules) should be
cation on the aspects associated with this guidance [14, 15]. employed as the choice of techniques for PGIs analysis, but
The U.S. Food and Drug Administration (USFDA) draft guid- these are often inadequate for accurate determination of ana-
ance was subsequently issued in 2008, providing the direction lytes at low ppm levels depending on the properties of an-
greatly consistent with the concepts as outlined in the EMA alytes and sample matrices. In the past few years, much
guidance. Therefore the EMA and USFDA have strengthened progress has been attained in the development of highly
their guidelines to control the limit of genotoxic impurities in sensitive analytical methods for the analysis of various PGIs
new commercial drugs. Both the agencies have set a thresh- in pharmaceuticals [22]. Although conventional HPLC–UV
old of toxicological concern (TTC) of 1.5 ␮g/person/day for methods are sometimes viable options [23], recently hyphen-
each impurity, which can be considered as an acceptable qual- ated MS has gained popularity in this field [24–28] due to
ification threshold for supporting a marketing authorization its superior sensitivity and specificity. Because of the ad-
by EMA and USFDA [16, 17]. As such, pharmaceutical re- vantage of mass-selective detection, MS methods are gen-
search and development (R&D) has devoted great efforts to erally less prone to interference compared to the nonspe-
the development of manufacturing processes that can effec- cific detectors such as UV detector. Therefore, the efforts
tively control the PGIs below TTC levels. During the clinical required for method development regarding the PGIs anal-
development stages, a staged TTC applies where greater daily ysis was greatly reduced. With regard to the ionization tech-
intake can be allowed for shorter dosing durations as pre- niques in LC–MS, API, which includes ESI and APCI are the
sented in Table 1 [18, 19]. The methods capable for such a most popular tools because of their simplicity and reliabil-
low level of detection are uncommon in the context of tra- ity. Consequently, in the past few years analytical scientists
ditional pharmaceutical analysis, where the typical level of in the pharmaceutical industry have strived to develop an-
interest is above 0.05% (equivalent to 500 ppm), which poses alytical strategies to meet the regulatory guidance [29]. As
significant challenge on analytical method development a result, various types of sample introduction methodolo-
for controlling these impurities. Therefore the usage and gies, different chromatographic separation tools, and various


C 2014 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.jss-journal.com
J. Sep. Sci. 2015, 00, 1–16 Liquid Chromatography 3

kinds of detectors have been explored and demonstrated as synthesis may only require testing in the intermediate (and
useful approaches [30–35]. An increasing number of publica- not the final product) that immediately follows the introduc-
tions with regard to individual genotoxic impurity or a spe- tion of the impurity. The synthetic process may significantly
cific class of impurities has appeared in the recent literature purge the impurity during various reaction steps such as pro-
[36–38]. cessing and purification. Further testing is required in later
However, practical guidance for the analytical determi- intermediates, only if the amount of impurity in the first
nation of some paramount classes of PGIs is still lacking. intermediate tested exceeds a predetermined level. This pre-
Therefore, this review is focused to explore recent advances determined level can be found by spiking studies that relate
in the analysis of some important classes of PGIs that are impurity survival throughout the synthesis. Obviously, the
commonly encountered during the process development. It testing point and impurity level specifications will vary on
also encompasses the trends in recent literature, together a case-by-case basis. The additional work involved in con-
with several practical approaches developed in the authors’ trolling the genotoxic impurities level is the development
laboratory in recent years. As such, this article is organized and validation of analytical methods, which can significantly
into the following main sections: (i) genotoxic impurities impact the drug development timeline. Because of this, a
identification, assessment, and control strategies, (ii) struc- strong risk assessment approach is needed to be applied for
tural alerts of commonly encountered PGIs and various reg- the developmental process that balances patient safety with
ulatory guidance issued to control their limit in final APIs, the control of PGIs. Otherwise, it needs more time and re-
(iii) toxicology assessment and analytical strategies includ- sources and there is a chance to delay the drug to reach the
ing the selection of analytical technique for the analysis of market.
PGIs, and (iv) case studies for the determination of some im- It is relatively easy to perform a risk assessment on the
portant classes of PGIs including hydrazines, alkyl halides, synthetic route and identification of all theoretical PGIs that
phenols, and benzene derivatives. It is the authors’ inten- could be presented in the final API. Such approaches produce
tion to provide an industrial perspective with respect to the a significant number of theoretical PGIs and additional chem-
identification, assessment, and control strategies for the PGIs ical reasoning should be used to evaluate their significance,
that guides the analytical communities in the pharmaceutical particularly if they arise at an early stage in a long synthetic
industry. route [41]. This kind of exercise can be applied based on the
known reactivity of a particular PGI in conjunction with the
known downstream chemistry. Some industry experts have
raised their concerns toward risk assessment of the synthetic
2 Genotoxic impurities assessment processes, namely, Snodin et al. argued that highly reactive
and control reagents (possessing a structural alert for genotoxicity) that
are utilized in the early stages of complex synthetic pathways
Usually genotoxic impurities can be identified by any of the are unlikely to be carried over to the final API [42]. Similarly,
following methods. (i) Already known genotoxins, (ii) pos- Pierson et al. reasoned that most PGIs raised from reagents
sessing similar functional groups with known genotoxins, or synthetic intermediates are likely to be eliminated or sig-
(iii) testing positive by genotoxicity assays, (iv) marked as a po- nificantly reduced by the downstream chemistries and pro-
tential genotoxin by one of many computer-based structure– cesses [41]. In the recent EMA questions and answer update,
activity software programs. the EMA does describe different scenarios based on where
The main focus of the risk assessment is to monitor the the impurity is introduced into the synthetic route. If the
impurities that arise particularly in the penultimate and final impurity is introduced before the final stage of the synthe-
synthetic stages. If the genotoxic impurity is formed during sis then it can be omitted from the final API specification.
the API synthesis, elimination may be achieved by changing In contrast, if the impurity is introduced into the final stage
the synthetic route and reagents used. However, this may not of the synthesis then it should be included in the final API
be feasible as the API synthesis is quite complicated and lim- specification.
ited based on the chemistry and available reagents making
these approaches impractical. Another means of genotoxic
impurity control in this situation is the implementation of
additional purification steps to the synthetic route to get rid of 3 Structural alerts of commonly
the impurities. However, this is not effective if the genotoxic encountered PGIs
impurity forms after the API synthesis (e.g., API degradation
or reactions with excipients/containers). Hence the impu- Knowledge about the chemical functional groups that cause
rity levels are still needed to be measured and monitored in DNA mutations have been used to develop computer-based
pharmaceuticals [39]. programs such as deductive estimation of risk from exist-
The genotoxic impurity testing point can vary and needs ing knowledge (DEREK), multicomputer-automated struc-
to be determined with scientific basis and supporting data ture evaluation (MCASE), and TOPKAT for the prediction
[40]. The testing strategy for PGIs analysis is shown in Fig. 1. of potential genotoxicity [43–45]. A positive in silico result
For instance, an impurity that has formed during the API from a chemical structure infers potential genotoxicity. This


C 2014 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.jss-journal.com
4 A. V. B. Reddy et al. J. Sep. Sci. 2015, 00, 1–16

Figure 1. Decision tree for PGIs testing strategy


and assessment.

potential alert is usually investigated by a bacterial reverse Some of the commonly encountered potentially geno-
mutation test such as the “Ames test” by which DNA re- toxic structural moieties are illustrated in Fig. 3 [51]. One
active genotoxins can be identified [46]. Conversely, a clear important group among them is alkylating agents, such
negative result in an appropriate genotoxicity test usually in- as alkyl halides [52], alkyl sulfonates [53], and other re-
dicates the absence of genotoxicity [47]. For the purpose of lated structures. These molecules are generally used as
deciding whether a given impurity possesses a genotoxicity reagents or might be generated during the chemical synthe-
risk as well as to control the PGIs below TTC level, Mueller sis. These result in cytotoxicity with the effect of inhibition
et al. [48] has classified the impurities into groups based on of the cell growth, initiation of programmed cell death, or
their respective genotoxicity potentials: Class 1 impurities are apoptosis, examples include: ␤-propiolactone, dimethyl sul-
known to be both genotoxic and carcinogenic; Class 2 impu- fate, diepoxybutane, anticancer drugs. In addition, aromatic
rities are genotoxic but with unknown carcinogenic potential; amines and nitro compounds do not directly form cova-
Class 3 impurities are structural alerts with unknown geno- lent bonds with DNA bases, but these molecules undergo
toxic potential (namely PGIs) and for which the structures metabolic activation to arylnitrenium ions, which will then
are unrelated to the API structure; Class 4 impurities are react with DNA bases and become genotoxic, the example
structural alerts but share the alerting structure with the API; includes: 2-naphthylamine [54, 55]. Acrylates are Michael
Class 5 impurities are not structural alerts, they are controlled acceptors that are susceptible to nucleophilic additions,
as ordinary impurities covered by ICH Q3 guidelines [49, 50] although recent data demonstrated that ethyl acrylate is
(Fig. 2). nongenotoxic in humans [56]. Epoxides and aziridines can

Figure 2. Genotoxic impurities control and clas-


sification strategy.


C 2014 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.jss-journal.com
J. Sep. Sci. 2015, 00, 1–16 Liquid Chromatography 5

Figure 3. Structural alerts of com-


monly encountered PGIs.

alkylate DNA via ring-opening reactions [57, 58]. Some hy- Table 2. Drug substance impurity thresholds
droperoxides can result in oxidative damage to DNA, and
their degradation products may react with DNA. Hydrazines Maximum Reporting Identification Qualification
are also known genotoxic impurities and potential human daily dosea) thresholdb),c) thresholdc) thresholdc)
carcinogens [59]. Figure 3 is by no means an exhaustive list
ࣘ2 g/day 0.05% 0.10% or 1.0 0.15% or 1.0
of PGIs, but is presented to show the representative structural
mg/day mg/day
features of some crucial PGIs, those frequently required for
(whichever (whichever
the analysis during the chemical synthesis of drugs. is lower) is lower)
>2 g/day 0.03% 0.05% 0.05%

4 Regulatory aspects a) The amount of drug substance administered per day.


b) Higher reporting threshold should be scientifically justified.
c) Lower threshold can be appropriate if the impurities are un-
There are several regulatory guidelines and position papers
usually toxic.
focused on controlling the amount of PGIs in pharmaceuti-
cals using specified limits. Different organizations from in-
dustry and regulatory authorities have developed guidelines benefits to the patients. For instance, ICH Q3A15 regulates
specifically addressing genotoxic impurities. Pharmaceutical the impurities in new drug substances with thresholds for
regulatory agencies such as USFDA, ICH, and EMA have reporting, identification, and qualification of impurities [60].
raised concerns on the presence of PGIs in APIs and issued ICH Q3B16 and ICH Q3C17 are equivalent guidelines for
the guidelines to control their limits in APIs. To address impurities in new drugs, which control residual solvents. De-
their concerns, R&D scientists have to identify the PGIs in pending on the potential risk to human health, residual sol-
advance during the process development, develop the proper vents are categorized into three classes [61]. Class I solvents
analytical methods, and also need to demonstrate the control should be avoided, class II solvents have permitted daily ex-
strategies of synthetic processes leading to PGIs. The accep- posure limits, and class III solvents have no health-based
tance criterion for impurities in the drug substances should exposure limits if the daily exposure is ࣘ50 mg/day. ICH
be set lower than the qualified level to protect the patients Q3D is currently under development and will include ele-
(Table 2). ments and limits for heavy metal impurities [62]. Currently
released ICH guidelines for impurity limits are not suitable
for most genotoxic impurities. The main issue related to geno-
4.1 ICH guidelines toxic impurities is that the synthesis of drug substances often
requires the use of reactive materials that have the capacity
According to ICH guidance, the impurities in drug sub- to interact with human DNA and cause mutations, as well as
stances and drug products are potentially toxic and give no cancer even at extremely lowest levels. Therefore, genotoxic


C 2014 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.jss-journal.com
6 A. V. B. Reddy et al. J. Sep. Sci. 2015, 00, 1–16

impurities should be avoided, and if this is not possible they and safety evaluations. This guideline has summarized its
must be reduced below a defined threshold. recommendations in the form of a decision tree in which
the preferred option is to eliminate PGIs, second preference
is to apply ALARP principle, and the final alternative is the
4.2 USFDA draft guidance TTC approach. EMA also released “Question and Answer”
document with clarifications for the questions raised in its
In December 2008, the USFDA published a draft guidance original guidance [66].
entitled “Genotoxic and Carcinogenic Impurities in Drug
Substances and Products-Recommended Approaches” [63].
Recently, it has been replaced by the ICH M7 guideline 4.4 PhRMA approach
on “Assessment and Control of DNA Reactive Impurities
(PGIs) in Pharmaceuticals to Limit Potential Carcinogenic The Pharmaceutical Research and Manufacturing Associa-
Risk.” The USFDA guidelines have provided the specific rec- tion (PhRMA) formed a task force in 2004 to discuss about
ommendations regarding the safety qualification of impu- testing, classification, qualification, and toxicological risk as-
rities with known/suspected genotoxicity. These guidelines sessment of PGIs in pharmaceuticals [48]. It stipulates that,
describe a variety of ways to characterize and reduce the po- some structurally alerting functional groups are known to
tential cancer risk associated with the patient exposure to be involved in reactions with DNA. These DNA-affecting
genotoxic and carcinogenic impurities. The recommended functional groups were categorized into three groups viz.,
approaches include (i) prevention of genotoxic and carcino- Group 1: aromatic groups, for example, N-hydroxyaryls, N-
genic impurities formation, (ii) reduction of genotoxic and acylated aminoaryls, aza-aryl N-oxides, aminoaryls and alky-
carcinogenic impurities level (maximum daily allowable tar- lated aminoaryls, purines or pyrimidines, intercalators, etc.,
get of 1.5 ␮g/day), (iii) additional characterization of geno- Group 2: alkyl and aryl groups, for example, aldehydes, N-
toxic and carcinogenic risk, and (iv) considerations for flex- methylols, N-nitrosamines, nitro compounds, carbamates,
ibility in approaches to better support appropriate impurity epoxides, aziridines, propiolactones, propiosultones, beta
specifications. haloethyl, hydrazines, and azo compounds, Group 3: het-
eroaromatic groups includes Michael-reactive acceptors, alkyl
esters of phosphonates or sulfonates, haloalkenes, primary
4.3 EMA guidelines halides (alkyl and aryl-CH2 ). PhRMA categorized the impu-
rities into five classes. Class 1: impurities known to be both
The EMA is one of the pioneering regulatory bodies to im- genotoxic (mutagenic) and carcinogenic. These impurities
pose detailed guidelines to handle genotoxic impurities. In represent the most serious risk and the default preference
June 2006, the EMA’s CHMP published the final guideline is to eliminate them by modifying the process. If this is not
on the limits of genotoxic impurities. The document applies possible, the TTC concept must be employed as a last option.
to the genotoxic impurities in new drug substances. It also ap- Class 2: impurities known to be genotoxic (mutagenic) but
plies to new applications for existing active substances, where with unknown carcinogenic potential. These impurities are to
assessment of the route of synthesis, process control, and im- be controlled using TTC principles. Class 3: impurities con-
purity profile does not provide reasonable assurance. EMA taining alerting structures, unrelated to the structure of the
guidelines on the limit of PGIs are classified into two cate- API, and of unknown genotoxic (mutagenic) potential. This
gories. (i) PGIs with sufficient (experimental) evidence for a group includes impurities with functional moieties that can
threshold-related mechanism, these are to be regulated using be linked to genotoxicity based on the structure. Class 4: im-
methods outlined in ICH Q3C (R4) for class 2 solvents [64] purities containing alerting structures related to the API. This
and (ii) PGIs without sufficient (experimental) evidence for group includes impurities that contain an alerting functional
a threshold-related mechanism, these are to be controlled in moiety that is shared with the parent structure. Class 5: im-
accordance with “as low as reasonably practicable” (ALARP) purities with no alerting structures shall be treated as normal
principle. Although the second approach is acceptable in impurities and controlled according to the ICH guidelines.
most instances, sufficient mechanistic data that allows the If class 3 or 4 compounds are genotoxic or not tested, they
assessment of threshold mechanism is still lacking. Hence, are moved into class 2, and if they are nongenotoxic, they are
the EMA guidelines proposed the use of “TTC” concept, considered as class 5.
which refers a threshold exposure level to compounds that
do not pose a significant risk of carcinogenicity or other toxic
effects. 4.5 European pharmacopoeia guidelines
A TTC value of 1.5 ␮g/day intake of PGIs is considered to
be associated with an acceptable risk [65]. The concentration European pharmacopoeia required a practical approach to
limit in ppm of a PGI permitted in drug substance is the elaborate or revise monographs on PGIs control. It says
ratio of TTC in microgram per day and daily dose in gram that, the products that received the marketing authorization
per day. The TTC approach benefits the consumers, indus- after the issuance of the EMA guidelines [67] have to be
try, and regulators by avoiding unnecessary toxicity testing evaluated for the presence of PGIs and this should be the


C 2014 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.jss-journal.com
J. Sep. Sci. 2015, 00, 1–16 Liquid Chromatography 7

basis for a new monograph [68]. The ICH S9 guideline on 6 Considerations for analytical testing
nonclinical evaluation for anticancer pharmaceuticals [65]
also states that, “for genotoxic impurities, several approaches 6.1 Analytical strategies
have been used to set limits based on the increase in life-
time risk of cancer.” Such limits are not appropriate for Analysis of PGIs in pharmaceuticals is the challenging task
pharmaceuticals intended to treat patients with advanced because they must be controlled at levels significantly lower
cancer and justification should be considered to set higher than 0.01–0.03%. Ideally, the analytical procedure should al-
limits. low the detection limits in the range of 1.0–5.0 ppm (0.0001–
0.0005% w/w). The analysis at such lower levels require not
only sensitive analytical instruments, but also place higher
5 Toxicology assessments demands on selectivity because more number of other or-
ganic impurities may be present at lower concentrations, for
Toxicology assessment should be done by pharmaceutical example, from excipients. The relatively large amount of API
scientists and toxicologists to identify PGIs and their entry may also interfere with low-level impurities. Furthermore,
into the synthetic process [69–71]. In addition, the synthetic it is very difficult to analyze all PGIs with a single ana-
route must be reviewed by a team of process chemists, an- lytical procedure because they contain different functional
alytical chemists, and toxicologists to identify likely reaction groups and come from different sources. In addition, due
byproducts and their potential for carry through to the APIs. to their highly reactive nature most of the PGIs can eas-
Utilizing the experts opinion, PGI alert structures can be ily react during extraction, sample preparation, or analysis
identified among the compounds for which no data is avail- and may cause inaccurate results. Also, some of the geno-
able using either of the commercial software applications toxic impurities are low-molecular-weight compounds that
including MDL-QSAR [72], MC4PC [73], and DEREK for are quite volatile and therefore they might disappear during
Windows [74]. Articles containing several structural alerts the sample preparation step. Depending on the nature and
were published [75–79]. However, due to the uncertain rel- quantity of the genotoxic impurity being investigated, care-
evance of the structural alerts, regulatory action cannot be ful investigation must be needed for the selection of appro-
solely based on the presence of a particular functional group. priate analytical techniques (or a combination of analytical
The accuracy of the predicted genotoxicity should be evaluated techniques).
case by case based on the available literature and genotoxicity Numerous articles have appeared in the scientific litera-
test results. ture mainly for the analysis and testing of PGIs in APIs. But,
If PGIs are introduced as starting materials and/or the general API determination methods are not suitable for
reagents or synthesized in the form of an intermediate or reac- the determination of PGIs due to their lower quantitation lim-
tion byproduct, different approaches must be considered for its (QLs). In the analytical point of view, the actual QL should
the identification of structural alerts. Compounds that yield be much more sensitive than the PGIs concentration limit. In
negative results in the alert assessment are placed in Class addition to this, certain drug substances may generate PGIs
5 and no additional action beyond normal impurity monitor- via degradation or storage and are to be separated from pro-
ing is needed. For any positive result (Classes 3 and 4), the cess impurities to achieve specificity in the analysis [83–86].
samples must be submitted for in vitro mutagenicity testing, Besides the sensitivity and specificity, the other challenges
usually with the Ames or mini-Ames test [80]. If the muta- in PGIs determination includes (i) diverse structural types of
genicity test is negative, this overrides the alert assessment PGIs that require the application of various analytical tech-
and the compound is placed in Class 5. Compounds giving a niques, (ii) PGIs without structural features are not favorable
positive mutagenicity results are placed in Class 2 [81]. The to common analytical detectors, (iii) chemically reactive or
need for toxicology assessment depends on the PGIs entry (as unstable PGIs lead to low recovery and poor sensitivity, and
starting materials or as intermediates or as reagents) in the require special handling techniques, (iv) interference of sam-
process and opportunities for their removal. For byproducts ple matrix resulted by enhanced test concentration of API to
that are known to be genotoxic, a toxicology limit is obtained achieve lower detection limits [87]. Sample solubility is also
regarding acceptable levels in the API. For byproducts that an important issue, because high concentrations of samples
have alerting structures, isolation or preparation of material are often required to enable the analysis of low-level geno-
may be required for mini-Ames testing. In general, theoret- toxic impurities in the sample. Achieving high solubilities
ical byproducts that are not anticipated to be formed are not can often be a significant challenge, and there is also a risk
assessed. This is consistent with EMA guidance recommend- of the sample precipitating out of the solution especially if
ing that the assessment of genotoxicity could be limited to the sample solution needs to be cooled to slow the degra-
those impurities that might be reasonably expected on the dation. In addition to the solubility requirements, param-
basis of the chemical reactions and conditions involved [82]. eters such as detection limit, quantification limit, linearity
Scientific judgment is required to balance the potential for and range, accuracy, and solution stability are to be estab-
impurity formation and carry through with consideration of lished as per ICH Q2(R1) validation guidelines [87]. How-
safety risk that would be caused by the presence of the impu- ever, the extent of validation depends on the purpose of the
rity in the API. study.


C 2014 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.jss-journal.com
8 A. V. B. Reddy et al. J. Sep. Sci. 2015, 00, 1–16

6.2 Selection of analytical techniques

The selection of an analytical technique for the determination


of PGIs can be divided into two groups based on their volatil-
ity. HPLC with UV detection should be considered as first
choice for the analysis of nonvolatile PGIs due its simplic-
ity and availability [88]. However, HPLC–UV may not offer
sufficient sensitivity for certain PGIs in trace level analysis.
If PGIs offer insufficient UV response, UHPLC or ultrafast
LC can be used because they have enhanced UV-detector
sensitivity. UHPLC methods are more sensitive than HPLC
because of the holistic design with advanced technology in the
pump, detector, and autosampler, which has evolved from the
HPLC [89,90]. As a brief description, UHPLC is just a special
variant of HPLC, but two properties makes UHPLC more
practical and in use worldwide, that is, having high-quality Figure 4. Decision tree for the selection of analytical technique in
small porous packing material and the capability of very high PGIs analysis.
pressure. The speed, resolution, and sensitivity of the detec-
tor cannot be compromised as compared to the traditional
HPLC. Now it is possible to take the full advantage of chro-
matographic principles to run separations using speed, supe- from the sample matrix. Headspace injection is desirable be-
rior resolution, and sensitivity afforded by smaller particles. cause it minimizes potential contamination of the injector or
When PGIs lack chromophores, evaporative light-scattering column by avoiding the introduction of a large quantity of
detection is the alternative choice. But, this detector is limited API. More recently, GC coupled with MS (GC–MS or GC–
in sensitivity and dynamic range. Refractive index detector MS/MS) has gained attention for the confirmation and iden-
and fluorescence detector are the other alternative detectors tification purposes, highlighting the flexibility of this tech-
used in HPLC. However, lower QL establishment is always a nology [97–100]. If PGIs are labile, possess no chromophores,
challenging task in the PGIs analysis. and have reactive functional groups, they can be derivatized to
As the trend toward smaller and smaller analyte con- form detectable species (e.g., hydrazine derivatizes benzalde-
centrations progresses, conventional analytical methods are hyde to form 1,2-dibenzylidenehydrazine) [101]. Derivatiza-
becoming less effective to address the PGIs at trace levels. tion reagent can be selected based on the functional groups
Hence, attention in the last few years has shifted to the appli- in the analyte. Derivatization helps in stabilization, incorpo-
cation of hyphenated techniques as preferred choice for the ration of a unique structural moiety, enhancing fluorescence,
identification and characterization of PGIs at trace level. Hy- ionization for mass detection, and volatization for GC. The
phenation of GC and HPLC/UHPLC with MS significantly proposed decision tree for the selection of analytical tech-
improves the method sensitivity and makes the methods nique is shown in Fig. 4.
more rapid [91, 92]. Broadly, the available hyphenated instru- MS-based methods generally provide additional robust-
ments are GC, LC, or CE on the front end and MS, NMR, or IR ness and ruggedness compared to the techniques such as UV
on the detection side, for example, GC–MS, LC–MS, CE–MS, alone, due to their high specificity and sensitivity. Highly sen-
LC–NMR, LC–NMR/MS, CE–NMR, LC–IR, etc. In particular, sitive Q-TOF mass spectrometers provide higher resolution
LC–MS instruments are commercialized in multiple variants. and mass accuracy that enables the unambiguous identifica-
These hyphenated instruments are slowly penetrating into tion of unknown trace impurities, making them very useful
the analytical laboratories worldwide for the characterization for PGIs analysis. Usually MS-based methods are often se-
of new emerging impurities [93–95]. lected for the impurity profiling of APIs during process de-
MS detectors are highly selective and sensitive, which are velopment, while UV-based methods are generally selected
capable of minimizing the issues caused by the interferences for the testing of PGIs in QC laboratories at manufacturing
in the sample matrices. However, hyphenated instruments sites.
are expensive, differ from vendor to vendor, and therefore
they need optimization of instrumental parameters while
transferring the methods between the development stages
to receiving laboratories [96]. In combination with FID, GC is
the standard choice for the analysis of small molecule PGIs. 7 Case studies for the determination of
Headspace gas chromatography (HS-GC) methods allow the some crucial PGIs
determination and identification of volatile low-molecular-
weight organic compounds, volatile degradation products, The following sections provide the analytical strategies for
and other volatile compounds. HS-GC–FID can be used to im- the analysis of some important classes of PGIs that have
prove sensitivity of the analytes and minimize interferences been carried out recently in our research laboratory.


C 2014 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.jss-journal.com
J. Sep. Sci. 2015, 00, 1–16 Liquid Chromatography 9

7.1 Analysis of hydrazines and related analogues 1,1-dimethylhydrazine in a pharmaceutical intermediate at


LOQ concentration of 200 ppm. In addition, several research
Hydrazines are commonly used as starting materials in the groups used nitrogen-selective detection for the characteriza-
synthesis of some pharmaceuticals. Some N-alkyl, N-aryl, tion of hydrazine analogues [99, 100], but Kirtley et al. [101]
and N-acyl analogues have been subjected to extensive tox- commented on the poor sensitivity of nitrogen phospho-
icological evaluations [88]. Hydrazine adducts with DNA and rous detection and they found inadequate LOD to address
the mechanism of adduction could include the formation of the analytical issues. Static headspace techniques combined
methyldiazonium ions or methyl free radicals. In addition, it with quadrupole electron ionization (EI) detection with se-
seems that the hydrazines react with endogenous formalde- lected ion monitoring (SIM) have been routinely used by
hyde to produce formaldehyde hydrazones [91]. In a study, Sun et al. [102]. These authors reported a generic in situ
20 hydrazine derivatives were found to induce a direct DNA derivatization HS-GC–MS method for the determination
damage in Escherichia coli and 16 of them (80%) were Ames of hydrazine at low concentrations. Unlike alkyl halides,
positive as well [88, 92, 96]. Other methods have reported that hydrazine analogues commonly needed derivatization as one
hydrazine, methyl hydrazine, 1,1-dimethylhydrazine, and 1,2- of the main analytical strategy toward enhancing the sensi-
dimethylhydrazine and other analogues are carcinogenic in tivity and addressing volatility.
rodents and possibly in human. Besides this, it was also found To overcome the difficulties in the analysis of hydrazine
that the hydrazine derivatives like hydralazine and its hy- analogues, a simple and sensitive LC–MS/MS method
drochloride salts were tumorigenic in rodents [92, 96]. was developed for the simultaneous determination of
A study was reported by Hmelnickis et al. [97] for two hydrazine PGIs namely (4-sulfamoylphenyl)hydrazine
the separation of six polar impurities including the hy- hydrochloride (SHH) and (4-methyl-acetophenone)-
drazine and 1,1,1-trimethylhydrazinium bromide impurities p-sulfonamide phenylhydrazine hydrochloride (MAP) in
in mildronate API using HILIC method. Another sensi- celecoxib without any derivatization [103]. Better separation
tive analytical method was developed by Liu et al. [98] us- was achieved with symmetry C18 (150 mm × 4.6 mm,
ing HILIC for the direct determination of hydrazine and 3.5 ␮m) column in positive ESI using multiple-reaction

Figure 5. Specificity chromatogram of SHH, MAP, and celecoxib. Chromatographic separation was achieved on Symmetry C18 column
(150 mm × 4.6 mm, 3.5 ␮m) kept at 40⬚C, using the mobile phase of 5.0 mM ammonium acetate-acetonitrile in the ratio of 30:70 (v/v), with
a flow rate of 0.7 mL/min, and the injection volume of 10 ␮L [103].


C 2014 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.jss-journal.com
10 A. V. B. Reddy et al. J. Sep. Sci. 2015, 00, 1–16

Figure 6. Specificity chromatogram of ritonavir and its three phenol impurities viz., phenol, 4-nitrophenol, and NPV. Chromatographic
conditions: Acquity UPLC BEH C18 column (100 mm × 2.1 mm, 1.7 ␮m) kept at 35⬚C, mobile phase consisted of 5.0 mm ammonium
acetate–0.05% ammonia in methanol 30:70 (v/v), mobile phase flow was 0.2 mL/min, and the injection volume was 8 ␮L [106].

monitoring mode. To achieve the proper separation of SHH moter. ATSDR (2008) noted that “under certain conditions,
and MAP from celecoxib, different mobile phase solvents at especially at high doses” phenols have the potential to exhibit
various compositions were evaluated, 5.0 mM ammonium genotoxicity [104, 105].
acetate-acetonitrile (30:70, v/v) was found to be the most In the present study, we have developed an UHPLC–
suitable composition at a flow rate of 0.7 mL/min. The MS/MS method for the simultaneous determination of phe-
method showed good linearity over the concentration range nol, 4-nitrophenol, and N-phenoxycarbonyl-L-valine (PCV) in
of 0.1–2.5 ppm for SHH and 0.06–2.5 ppm for MAP PGIs. ritonavir. Initially the method development was started with
The LOD and LOQ of SHH are 0.03 and 0.1 ppm, for MAP LC–MS/MS for the determination of three closely related
they are 0.02 and 0.06 ppm, respectively. The method has phenol impurities namely phenol, 4-nitrophenol, and PCV.
good recovery in the range of 95.0–104.0% for both the But the separation and peak shapes are not satisfactory with
analytes. The developed method was also applied for the LC–MS/MS method. Therefore as the next option, UHPLC–
determination of SHH and MAP PGIs in three formulation MS/MS was chosen for the simultaneous determination of
batches of celecoxib and the specificity chromatogram is three phenol impurities in ritonavir. Good separation was
shown in Fig. 5. achieved on Acquity UPLC BEH C18 column (2.1 mm ×
100 mm, 1.7 ␮m) using gradient elution of 0.05% ammo-
nia in methanol (30:70, v/v) and 5.0 mM ammonium acetate
buffer at a flow rate of 0.2 mL/min. Several attempts were
7.2 Analysis of phenol impurities in ritonavir made with different C18 UHPLC columns (Inertsil ODS-3,
50 mm × 2.1 mm, 2.0 ␮m and Zorbax XDB C-18, 50 mm × 4.6
The available data regarding the genotoxicity and car- mm, 1.8 ␮m) using the gradient elution, but phenol and PCV
cinogenicity of phenols and its analogue impurities are were merged together, 4-nitrophenol was coeluted with riton-
inconclusive. But the reported data (RIVM 2001) on animal avir using the above columns excluding the UPLC BEH C18
testing suggested that the phenol can act as a tumor pro- column. The calibration curves showed good linearity over


C 2014 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.jss-journal.com
J. Sep. Sci. 2015, 00, 1–16 Liquid Chromatography 11

Figure 7. Specificity chromatogram and mass spectra of 2-chloromethyl-3,4-dimethoxy pyridine hydrochloride and pantoprazole. Chro-
matographic conditions: Hypersil BDS C18 column (50 mm × 4.6 mm, 3.0 ␮m) kept at 40⬚C, mobile phase consisted of 10 mm ammonium
acetate/acetonitrile 79:21 v/v at a flow rate of 1.0 mL/min, and the injection volume of 20 ␮L [117].

the concentration range of 0.3–1.5 ppm for phenol and 0.1– that are insufficiently volatile and/or too thermally labile for
1.5 ppm for both 4-nitrophenol and PCV. The method has reliable GC analysis. While many historical references em-
very low LOD and LOQ and the accuracy lies between 97.8 and ployed HPLC with single-wavelength UV detection, the more
103.2% for all the three impurities. The developed method recent literature employs a variety of MS techniques [110].
was successfully applied for the formulation batches of riton- Somewhat surprisingly, there are few reports in the literature
avir to determine its phenol impurities [106]. The specificity using SFC for the analysis of alkylating agents. Similarly, few
chromatogram is shown in Fig. 6. methods of CZE have been reported for the analysis of alkyl
halides.
Ellison et al. [111] reported a HS-GC–ECD method for
the determination of 23 genotoxic alkyl/aryl halides as po-
7.3 Analysis of alkyl halides
tential impurities in API. The method was generically vali-
dated with two different APIs, thereafter it was validated for
Owing to the electrophilic nature of alkylating agents, they
every additional API using spiked batches. The detection lim-
can introduce lesions at nucleophilic centers of DNA. Strong
its for the 23 analytes ranged from 2.5 to 290 ng/mL, and
acid–base interactions during the process of drug salt forma-
the results were greatly correlated with the results of Skett
tion produce alkylating agents such as alkyl halides and alkyl
et al. [112]. Another HS-GC method for the determination
esters of alkyl sulfonic acids. As the salt formation is a com-
of volatile alkylating impurities in the antiemetic, azasetron
mon method in drug formulation processes, alkyl halides
hydrochloride was reported by Wang [113]. Further, Frost
exist as impurities in several drug substances and leads to
et al. [114] reported a GC–FID method for the determination
mutagenicity [107,108]. The nucleophilic attack mechanisms
of trace levels of pharmaceutical process impurities of toxi-
of alkylating compounds determine their reactivity against
cological concern, including volatile alkylating agents, benzyl
DNA [109].
chloride and chloroethyl methyl ether. The authors have uti-
Historically, analysts have relied on the volatility of alkyl
lized a powerful combination of automated headspace and
halides and developed the GC methodologies with FID for
solid-phase microextraction to concentrate the analytes. Lee
their separation and quantification. GC still remained as the
et al. [115] demonstrated that the HPLC–ESI/MS gave bet-
preferred technique in many cases, but is now more routinely
ter responses than APCI and was used to measure N,N-
used in combination with more sensitive and selective detec-
dimethylaminoethyl chloride in diltiazem hydrochloride us-
tion techniques, such as GC–MS or GC–ECD. HPLC also
ing SIM mode. Clarke [116] used HPLC–MS to determine
remained as a key separation technique for those analytes


C 2014 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.jss-journal.com
12 A. V. B. Reddy et al. J. Sep. Sci. 2015, 00, 1–16

Figure 8. Specificity chromatogram


and mass spectra of 1-(3-choropropyl)-
4-(3-chlorophenyl)piperazine and tra-
zodone. Chromatographic conditions:
C18 symmetry column (100 mm ×
4.6 mm, 3.5 ␮m) kept at 40⬚C, mobile
phase consisted of 5.0 mM ammonium
acetate-acetonitrile in the ratio of 30:70
(v/v at pH 5.0 ± 0.05) at a flow rate of
0.8 mL/min (split down to 0.2 mL/min
into the MS source), and the injection
volume of 10 ␮L [118].

three residual alkyl bromides (A, B, and C) in a secondary and robustness. The specificity chromatogram of the CCP
amine API. The method gave acceptable LODs for all the impurity and trazodone is shown in Fig. 8.
three impurities with LODs of 10 ng/g and the method was
validated as well as used to analyze and release API for clinical
use.
In our study, a sensitive and selective LC–MS/MS 7.4 Analysis of benzene derivatives in zolmitriptan
method was developed for the trace level determination of
2-chloromethyl-3,4-dimethoxy pyridine hydrochloride (CDP), Benzene and its derivatives have the probability to increase
a PGI in pantoprazole sodium drug substance [117]. The the likelihood of DNA damage [119]. Several compounds of
separation was achieved on Hypersil BDS C18 (50 mm × this group are capable of reacting with DNA either directly
4.6 mm, 3.0 ␮m) column using 10 mM ammonium acetate in or after metabolic transformation and become potent mu-
1000 mL of water as buffer. The mobile phase was optimized tagens and carcinogens [120, 121]. Therefore, in the present
in the ratio of buffer/acetonitrile (79:21, v/v) to achieve bet- study we have developed a selective and sensitive UHPLC–
ter separation and high sensitivity. Trace level of ammonium MS/MS method for the simultaneous determination of
acetate is added to the mobile phase to enhance the ioniza- four PGIs, namely, (2S)-2-amino-3-(4-aminophenyl)propan-
tion and detection. The method was validated according to 1-ol, (S)-4-nitrophenylalanine hydrate, (S)-2-amino-3-(4-
ICH guidelines and was able to quantify CDP impurity at nitrophenyl)propanol, and (S)-methyl-4-nitrophenylalanate
0.3 ppm. The specificity chromatogram of CDP impurity and hydrochloride (APP, NPA, NPP, and MNA respectively). The
pantoprazole is shown in Fig. 7. method development was started with LC–MS/MS, but no
Another simple and sensitive LC–MS/MS method was proper separation was achieved using LC–MS/MS as well as
developed for the determination of 1-(3-chloropropyl)-4-(3- the peaks were merged with each other due to their structural
chlorophenyl) piperazine HCl (CCP), a process-related impu- similarity. Therefore, further method development was car-
rity in trazodone [118]. The separation was achieved on C18 ried out using UHPLC–MS/MS, in which all the four PGIs
Symmetry (100 mm × 4.6 mm, 3.5 ␮m) column interfaced viz., APP, NPA, NPP, and MNA were separated with better
with a triple quadruple tandem mass spectrometer operated resolution. The method utilized Hypersil BDS C8 column
in the multiple-reaction monitoring mode. Positive ESI was (50 mm × 4.6 mm, 3.0 ␮m) with ESI in selected ion record-
employed as the ionization source and the mobile phase used ing mode for the quantitation of four PGIs. The method
was 5.0 mM ammonium acetate/acetonitrile (30:70, v/v). The was able to quantitate APP at 0.1 ppm and NPA, NPP, and
LOD and LOQ were found to be 0.01 and 0.03 ppm, re- MNA at 0.15 ppm with respect to 5.0 mg/mL of zolmitrip-
spectively, with linearity ranging from 0.03 to 2.5 ppm. The tan. The proposed method was validated for its specificity,
method was fully validated by following ICH guidelines to linearity, accuracy, and precision by following the ICH guide-
assess LOD, LOQ, linearity, precision, accuracy, specificity, lines. The method was linear in the range of 0.1–2.0 ppm


C 2014 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.jss-journal.com
J. Sep. Sci. 2015, 00, 1–16 Liquid Chromatography 13

Figure 9. Specificity chromatogram of zolmitriptan and its four PGIs, that is, APP, NPA, NPP, and MNA. Chromatographic conditions:
Hypersil BDS C8 column (50 mm × 4.6 mm, 3.0 ␮m) kept at 35⬚C, gradient elution using 5.0 mM ammonium acetate as solution A and
acetonitrile/methanol (90:10, v/v) as solution B at time/%solution A; 0/95, 6/95, 9.0/80, 10/80, 13/70, 14/70, 20/95 using the flow rate of
0.5 mL/min, and the injection volume of 8 ␮L [122].

for APP and 0.15–2.0 ppm for NPA, NPP, and MNA and the in the trace analysis of PGIs in various stages of preclini-
LOD, LOQ values were found very low with >0.999 correla- cal and clinical drug development. It is not surprising that
tion coefficient [122]. The accuracy of the method was ranged the vast majority of the methods end up using MS detec-
between 98.1 and 102.8% for four PGIs. The impurities were tion because of the unparallel specificity and sensitivity of
not found in the three studied pure and formulation batches this technique. Because of their highly reactive nature, the
of zolmitriptan. The specificity chromatogram of impurity direct analysis of most PGIs is difficult and results in poor
and pantoprazole is shown in Fig. 9. recovery and sensitivity. Therefore, chemical derivatization
and coordination ion spray MS have been found as alterna-
tive analytical strategies for stabilizing and enhancing ana-
lyte detectability. Therefore, the trend clearly shows a def-
8 Conclusions inite shift of the PGIs analysis from conventional way of
structure elucidation to the use of modern hyphenated tech-
The identification and control of PGIs in a synthetic pro- niques. The strategies including regulatory guidance, flow
cess is always challenging, owing to their evolving nature charts, decision trees, and toxicology assessment approaches
and variable points of entry. Therefore, synthetic routes must are developed for the better understanding and to explain
be screened for the identification of structural alerts, which the risks and liabilities associated with PGIs throughout
causes genotoxicity. If PGIs are found, alternative synthetic the development process. By coupling the above practical
routes should be adopted to control them in the final APIs. If strategies with hyphenated MS instrumentation, PGIs anal-
this is not technically feasible, the safety limits must be fixed ysis will experience a major leap forward over the next few
based on the TTC concept. These limits generally come into years.
lower levels and need analytical determinations with adequate This review has presented the recent analytical meth-
selectivity and sensitivity. Rapid developments of extremely ods carried out in our laboratory for the quantitative analysis
sensitive and selective analytical methodologies are crucial of selected PGIs in pharmaceuticals. The case studies dis-
to monitor PGIs at very low levels. Synthetic and analytical cussed in this review for the determination of some impor-
chemistry is required to balance the risk and cost during tant classes of PGIs including hydrazines and its analogues in
the development of drug substances. In recent years, MS celecoxib, phenol impurities in ritonavir, benzene derivatives
detection coupled with GC or HPLC played a critical role in zolmitriptan, and finally alkyl halides in pantoprazole and


C 2014 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.jss-journal.com
14 A. V. B. Reddy et al. J. Sep. Sci. 2015, 00, 1–16

trazodone provided a better understanding of PGIs-related [10] Ahuja, S., Isolation and Characterization of Pharmaceu-
risk and assessment, such an attention is ultimately focused tical Impurities Evaluation, Marcel Dekker, New York
on analytical control of those specific PGIs that pose a the- 2000.
oretical risk of presence in the final drug substance. All the [11] Froetschl, R., Presentation at the 2009 PhRMA
developed methods were fully validated for their linearity, API Workshop, New Brunswick, NJ 2009, May 18–
21.
specificity, accuracy, precision, and robustness. The LOD and
LOQ values of all the developed methods were achieved be- [12] Committee for Proprietary Medicinal Products (CPMP),
Position Paper on the Limits of Genotoxic Impurities.
low the TTC threshold concentration. Some of the developed
CPMP, London, UK 2002.
methods were successfully applied for the determination of
[13] ICH, Genotoxicity: A standard battery for genotoxicity
corresponding PGIs in formulation batches of APIs. The de-
testing of pharmaceuticals S2B, 1997.
veloped methods were proved to be capable for monitoring
[14] Snyder, R. D., Green, J. W., Mutat. Res. 2001, 488, 151–
of the mentioned PGIs in corresponding drug substances
169.
during their synthesis/manufacturing.
[15] European Medicines Agency (EMA), Questions and an-
To offer a product to society that is devoid of PGIs
swers on the “guideline on the limits of genotoxic im-
or acceptable to regulatory agencies, avoiding the usage of purities,” 2010.
raw materials, reagents, solvents having potential genotoxic
[16] Guidance for industry genotoxic and carcinogenic im-
alerts is the most idealistic approach. But, in real life sce- purities in drug substances and products: recom-
nario, it would be a difficult and complex activity to any mended approaches draft, CDER, December 2008.
chemist. Therefore, it is worth to conclude the review with [17] U.S. FDA, Guidance for industry: M3(R2) nonclinical
the USFDA statement “Although marketed medicinal prod- safety studies for the conduct of human clinical trials
ucts are required to be safe, safety does not mean zero and marketing authorization for pharmaceuticals, 2010.
risk. A safe product is one that has reasonable risks, given [18] Kenyon, M. O., Cheung, J. R., Dobo, K. L., Ku, W. W.,
the magnitude of the benefits expected and the alternatives Reg. Toxic. Pharm. 2007, 48, 75–86.
available.” [19] Liu, D. Q., Chen, T. K., McGuire, M. A., Kord, A. S., J.
Pharm. Biomed. Anal. 2009, 50, 144–150.
The authors gratefully acknowledged the postdoctoral fi- [20] Yu, L. X., Pharm. Res. 2008, 25, 781–791.
nancial support by the Research University Grant, Universiti
[21] ICH Q3C, Guideline for Residual Solvents, ICH Tripartite
Teknologi Malaysia, and the Ministry of Education Malaysia Guideline, February 4, 2011.
for providing LRGS Grant on Water Security entitled “Pro-
[22] David, F., Vanhoenacker, G., Sandra, P., Jacq, K., Baker,
tection of Drinking Water: Source Abstraction and Treatment A., LCGC Europe, 2009.
(203/PKT/6720006).”
[23] Soman, A., Jacob, S., Swanek, F., J. Chromatogr. Sci.
2009, 47, 315–319.
The authors have declared no conflicts of interest. [24] An, J., Sun, M., Bai, L., Chen, T., Liu, D. Q., Kord, A., J.
Pharm. Biomed. Anal. 2008, 48, 1006–1010.
[25] Alzaga, R., Ryan, R. W., Taylor-Worth, K., Lipczynski, A.
M., Szucs, R., Sandra, P., J. Pharm. Biomed. Anal. 2007,
45, 472–479.
9 References [26] Sun, M., Bai, L., Liu, D. Q., J. Pharm. Biomed. Anal.
2009, 49, 529–533.
[1] Bolt, H. M., Foth, H., Hengstler, J. G., Degen, G. H., [27] Vanhoenacker, G., Dumont, E., David, F., Baker, A., San-
Toxicol. Lett. 2004, 151, 29–41. dra, P., J. Chromatogr. A 2009, 126, 3563–3570.
[2] Bouder, F., Expert Rev. Clin. Pharmacol. 2008, 1, 241– [28] Yuabova, Z. Y., Holschlag, D. R., Rodriguez, S. A., Qin,
250. C., Papov, V. V., Qiu, F., McCaffrey, J. F., Norwood, D.
[3] Cunningham, A. R., Rosenkranz, H. S., Zhang, Y. P., Kl- L., J. Liq. Chromatogr. Relat. Technol. 2008, 31, 2318–
opman, G., Mutat. Res. 1998, 398, 1–17. 2330.
[4] Kondo, K., Watanabe, A., Iwanaga, Y., Abe, I., Tanaka, [29] Gorog, S., Determination of Impurities in Drugs, Else-
H., Nagaoka, M. H., Akiyama, H., Maitani, T., Food Addit. vier Sciences, Amsterdam 1999.
Contam. 2006, 23, 1179–1186. [30] Alegre, M. R., Biochem. Pharmacol. 2012 1, e123.
[5] Leblanc, B., Charuel, C., Ku, W., Ogilvie, R., Int. J. [31] Ramakrishna, K., Raman, N. V. V. S. S., Narayana Rao,
Pharm. Med. 2004, 18, 215–220. K. M. V., Prasad, A. V. S. S., Subhaschander Reddy, K.,
[6] Jacobson Kram, D., McGovern, T., Adv. Drug Deliv. Rev. J. Pharm. Biomed. Anal. 2008, 46, 780–783.
2007, 59, 38–42. [32] Friscia, O., Pulci, R., Fassio, F., Comelli, R., J. Environ.
[7] Zhang, K., Li, Y., Tsang, M., Chetwyn, N. P., J. Sep. Sci. Pathol. Toxicol. Oncol. 1994, 13, 89–110.
2013, 36, 2986–2992. [33] Qixin, D., Jiajun, Z., Qiang, S., Chao, T., Xiaomei, W.,
[8] Kushwaha, P., Pharma. Info. Net. 2008, 6. Yunqiu, Y., J. Sep. Sci. 2013, 36, 1200–1208.
[9] Zhou, Y. X., Zhou, W. T., Sun, L. L., Zou, Q. G., Wei, P., [34] Rao, R. N., Narasa Raju, A., Ramaram, N., Raveendra
Ou Yang, P. K., J. Sep. Sci. 2014, 37, 1248–1255. Babu, G., J. Sep. Sci. 2008, 31, 107–118.


C 2014 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.jss-journal.com
J. Sep. Sci. 2015, 00, 1–16 Liquid Chromatography 15

[35] Wang, X., Sun, H., Zhang, A., Wang, P., Han, Y., J. Sep. [59] Elder, D. P., Snodin, D., Teasdale, A., J. Pharm. Biomed.
Sci. 2011, 34, 3451–3459. Anal. 2011, 54, 900–910.
[36] Zhang, X., Zheng, B., Zhang, H., Chen, X., Mei, G., J. [60] ICH Q3A (R2), Impurities in New Drug Substances,
Sep. Sci. 2011, 34, 469–474. 2006.
[37] Elder, D. P., Delaney, E. D., Teasdale, A., Eyley, S., Reif, [61] ICH Q3B (R2), Impurities in New Drug Substances, 2006.
V. D., Jacq, K., Facchine, K. L., Oestrich, R. S., Sandra, [62] ICH Q3D, Guideline for elemental Impurities, 2013.
P., David, F., J. Pharm. Sci. 2010, 99, 2948–2961.
[63] FDA Draft guidance, Genotoxic and carcinogenic impu-
[38] Elder, D. P., Snodin, D., Teasdale, A., J. Pharm. Biomed. rities in drug substances and products: Recommended
Anal. 2010, 51, 1015–1023. approaches, 2008.
[39] Deepti, J., Pawan Kumar B., J. Pharm. Biomed. Anal. [64] International Conference on Harmonization Guideline
2013, 86, 11–35. for Residual Solvents, Q3C (R4), 2009.
[40] Pierson, D. A., Olsen, B. A., Robbins, D. K., DeVries, K. [65] International Conference on Harmonization Guideline
M., Varie, D. L., Org. Process Res. Dev. 2009, 13, 285– on Nonclinical Evaluation for Anticancer Pharmaceuti-
291. cals, S9, 2009.
[41] Teasdale, A., Fenner, S., Ray, A., Ford, A., Philips, A., [66] Question & Answers on the CHMP Guideline on the
Org. Process Res. Dev. 2010, 14, 943–945. Limits of Genotoxic Impurities, 2008.
[42] Snodin, D. J., Org. Process Res. Dev. 2010, 14, 960–976. [67] European Medicines Agency, Guideline on the Limits
[43] Snyder, R. D., Pearl, G. S., Mandakas, G., Choy, W. N., of Genotoxic Impurities, 2007.
Goodsaid, F., Rosenblum, I. Y., Environ. Mol. Mutagen. [68] Potentially Genotoxic Impurities and European Phar-
2004, 43, 143–158. macopoeia Monographs on substances for human use,
[44] Snyder, R. D., Smith, M. D., Drug Discov. Today, 2005, Pharmeuropa 2008, 20, 426–427.
10, 1119–1124. [69] Humfrey, C. D. N., Toxicol. Sci. 2007, 100, 24–28.
[45] Matthews, E. J., Kruhlak, N. L., Benz, R. D., Contrera, [70] Bercu, J. P., Morton, S. M., Deahl, J. T., Gombar, V. K.,
J. F., Merchant, C. A., Yang, C., Toxicol. Mech. Methods Callis, C. M., Van Lier, R. B. L., Regul. Toxicol. Pharma-
2008, 18, 189–206. col. 2010, 57, 300–306.
[46] Bercu, J. P., Dobo, K. L., Gocke, E., McGovern, T. J., Int. [71] Fiori, J. M., Meyerhoff, R. D., Regul. Toxicol. Pharmacol.
J. Toxicol. 2009, 28, 468–478. 2002, 35, 209–216.
[47] Kruhlak, N. L., Contrera, J. F., Benz, R. D., Matthews, E. [72] Votano, J. R., Parham, M., Hall, L. H., Kier, L. B., Oloff,
J., Adv. Drug Deliv. Rev. 2007, 59, 43–55. S., Tropsha, A., Xie, Q., Tong, W., Mutagenesis 2004,
[48] Mueller, L., Mauthe, R. J., Riley, C. M., Andino, M. M., 19, 365–377.
De Antonis, D., Beels, C., De George, J., De Knaep, A. [73] Fellows, M.D., Boyer, S., O’Donovan, M.R., Mutagene-
K. M., Ellison, D., Fagerland, J. A., Frank, R., Fritschel, sis 2011, 26, 529–532.
B., Galloway, S., Harpur, E., Humfrey, C. D. N., Jacks,
[74] Deductive Estimation of Risk from Existing Knowledge,
A. S., Jagota, N., MacKinnon, J., Mohan, G., Ness, D.
marketed by LHASA Ltd., Leeds, UK.
K., Michael, R. O., Smith, M. D., Gopi, V., Larry, Y., Reg.
Toxicol. Pharm. 2006, 44, 198–211. [75] Ashby, J., Tennant, R. W., Mutat. Res. 1988, 204, 17–115.
[49] International Conference on Harmonization (ICH) [76] Ashby, J., Tennant, R. W., Mutat. Res. 1991, 257, 229–
Guidelines, Q3A (R): Impurities in New Drug Sub- 308.
stances (Revised Guideline), 2002. [77] Fetterman, B. A., Kim, B. S., Margolin, B. H., Schildcrout,
[50] International Conference on Harmonization (ICH) J. S., Smith, M. G., Wagner, S. M., Zeiger, E., Environ.
Guidelines, Q3B(R): Impurities in New Drug Products Mol. Mutagen. 1997, 29, 312–322.
(Revised Guideline), 2003. [78] Dobo, K. L., Greene, N., Cyr, M. O., Caron, S., Ku, W. W.,
[51] Derek, I., Org. Process Res. Dev. 2010, 14, 946–959. Regul. Toxicol. Pharmacol. 2006, 44, 282–293.
[52] Elder, D. P., Lipczynski, A. M., Teasdale, A., J. Pharm. [79] Snyder, R. D., Environ. Mol. Mutagen. 2009, 50, 435–
Biomed. Anal. 2008, 48, 497–507. 450.
[53] Elder, D. P., Teasdale, A., Lipczynski, A. M., J. Pharm. [80] Flamand, N., Meunier, J. R., Meunier, P. A., Agapakis
Biomed. Anal. 2008, 46, 1–8. Causse, C., Toxicol. In Vitro 2001, 15, 105–114.
[54] Ohnishi, S., Murata, M., Kawanishi, S., Jpn. J. Cancer [81] European Medicines Agency (EMA), Guideline on the
Res. 2002, 93, 736–743. Limits of Genotoxic Impurities, London, UK 2006.
[55] Parks, J. M., Ford, G. P., Cramer, C. J., J. Org. Chem. [82] European Medicines Agency, Question and answers on
2001, 66, 8997–9004. the CHMP, Guideline on the Limits of Genotoxic Impu-
rities, 2009.
[56] Waegemaekers, T. H., Bensink, M. P., Mutat. Res. 1984,
137, 95–102. [83] Raillard, S. P., Bercu, J., Baertschi, S. W., Riley, C. M.,
Org. Process Res. Dev. 2010, 14, 1015–1020.
[57] Zang, H., Kent, S. G., Biochemistry 2000, 39, 14968–
14975. [84] Patterson, D. E., Powers, J. D., LeBlanc, M., Sharkey, T.,
Boehler, E., Irdam, E., Osterhout, M. H., Org. Process
[58] Liu, D. Q., Sun, M., Kord, A. S., J. Pharm. Biomed. Anal.
Res. Dev. 2009, 13, 900–906.
2010, 51, 999–1014.


C 2014 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.jss-journal.com
16 A. V. B. Reddy et al. J. Sep. Sci. 2015, 00, 1–16

[85] Cimarosti, Z., Bravo, F., Stonestreet, P., Tinazzi, F., Vec- Registry, US Department of Health and Human Ser-
chi, O., Camurri, G., Org. Process Res. Dev. 2010, 14, vices, Atlanta, GA 2008.
993–998. [106] Venugopal, N., Vijaya Bhaskar Reddy, A., Madhavi, G.,
[86] Kovarikova, P., Vavrova, K., Tomalova, K., Schongut, M., J. Pharm. Biomed. Anal. 2014, 90, 127–133.
Hruskova, K., Haskova, P., Klimes, J., J. Pharm. Biomed. [107] Lee, C. R., Hubert, M., Van Dau, C. N., Peter,
Anal. 2008, 48, 295–302. D., Krstulovic, A. M., Analyst 2000, 125, 1255–
[87] International Conference on Harmonization, Guideline 1259.
on Validation of Analytical Procedures, Q2(R1), 2005.
[108] Sobol, Z., Engel, M. E., Rubitski, E., Ku, W. W., Aubrecht,
[88] Raman, N. V. V. S. S., Ratnakar Reddy, K., Prasad, A. V. J., Schiestl, R. H., Mutat. Res. Genet. Toxicol. Environ.
S. S., Ramakrishna, K., J. Pharm. Biomed. Anal. 2008, Mutagen. 2007, 633, 80–94.
48, 227–230.
[109] Teasdale, A., Elder, D., Chang, S. J., Wang, S., Thomp-
[89] Zhou, Y., Fung Kei Choi, F., He, Z. Z., Song, J. Z., Qiao, son, R., Benz, N., Ignacio, H., Flores, S., Org. Process
C. F., Liu, X., Ding, L. S., Gesang, S. L., Xu, H. X., J. Sep. Res. Dev. 2013, 17, 221–230.
Sci. 2010, 33, 3675–3682.
[110] Borman, P. J., Chatfield, M. J., Crowley, E. L., Eckers, C.,
[90] Li, Y., Zhang, T., Zhang, X., Xu, H., Liu, C., J. Sep. Sci. Elder, D. P., Francey, S. W., Laures, A. M. F., Wolff, J. C.,
2010, 33, 3347–3353. J. Pharm. Biomed. Anal. 2008, 48, 1082–1089.
[91] Raman, N. V. V. S. S., Ratnakar Reddy, K., Prasad, A. V.
[111] Ellison, G. K., Pharmaceutical Analysis Science Group
S. S., Ramakrishna, K., Talanta 2009, 77, 1869–1872.
(PASG) Autumn Meeting, Milton Keynes, UK, October
[92] Bai, L., Sun, M., An, J., Liu, D. Q., Chen, T. K., Kord, A. 02, 2006.
S., J. Chromatogr. A 2010, 1217, 302–306.
[112] Skett, P. W., Smith, R. J., Webb, M. L. (Eds.). Low Level
[93] Hao, Z., Xiao, B., Weng, N., J. Sep. Sci. 2008, 31, 1449– Measurement of Potent Toxins. In: Analysis of Drug
1464. Impurities, Blackwell Publishing, Oxford 2007, pp. 82–
[94] Dejaegher, B., Mangelings, D., Vander Heyden, Y., J. 123.
Sep. Sci. 2008, 31, 1438–1448. [113] Wang, Y., Chin. J. Pharm. Anal. 2004, 24, 293–295.
[95] Regalado, E. L., Helmy, R., Green, M. D., Welch, C. J., J. [114] Frost, R. P., Hussain, M. S., Raghani, A. R., J. Sep. Sci.
Sep. Sci. 2014, 37, 1094–1102. 2013, 26, 1097–1103.
[96] Sun, M., Liu, D. Q., Kord, A. S., Org. Process Res. Dev. [115] Lee, K. C., Cheuk, M. W., Chan, W., Lee, A. W., Zhao, Z.
2010, 14, 977–985. Z., Jiang, Z. H., Cai, Z., Anal. Bioanal. Chem. 2006, 386,
[97] Hmelnickis, J., Pugovics, O., Kazoka, H., Viksna, A., 2225–2232.
Susinskis, I., Kokums, K., J. Pharm. Biomed. Anal. 2008, [116] Clarke, C., Proceedings of International September
48, 649–656. Conference, Spain 2004.
[98] Liu, J., Zhou, W., You, T., Li, F., Wang, E., Dong, S., Anal. [117] Venugopal, N., Vijaya Bhaskar Reddy, A., Madhavi, V.,
Chem. 1996, 68, 3350–3353. Gangadhara Reddy, K., Madhavi, G., J. Pharm. Biomed.
[99] Glyenhaal, O., Gronberg, L., Vessman, J., J. Chro- Anal. 2012, 70, 592–597.
matogr. A 1990, 511, 303–315. [118] Venugopal, N., Vijaya Bhaskar Reddy, A., Madhavi, G.,
[100] Matsui, F., Robertson, D. L., Lovering, E. G., J. Pharm. Jaafar, J., Madhavi, V., Gangadhara Reddy, K., Arab. J.
Sci. 1983, 72, 948–951. Chem. doi:10.1016/j.arabjc.2014.10.006
[101] Kirtley, A., Strickfuss, S., Yehl, P. M., PASG Annual Meet- [119] Carere, A., Antoccia, A., Crebelli, R., Degrassi, F., Fiore,
ing, Bedfordshire, UK 2006. M., Iavarone, I., Isacchi, G., Lagorio, S., Leopardi, P.,
[102] Sun, M., Bai, L., Terfloth, G. J., Liu, D. Q., Kord, A. S., J. Marcon, F., Palitti, F., Tanzarella, C., Zijno, A., Mutat. Res.
Pharm. Biomed. Anal. 2010, 52, 30–36. 1995, 332, 17–25.
[120] Whysner, J., Reddy, M. V., Ross, P. M., Mohan, M., Lax,
[103] Vijaya Bhaskar Reddy, A., Venugopal, N., Madhavi, G.,
E. A., Mutat. Res. 2004, 566, 99–130.
J. Anal. Sci. Technol. 2014, 5, 1–8.
[121] Srinivasa Rao, M., Vinay Rao, S., Ramesh Babu, K. P.,
[104] Baars, A. J., Theelen, R. M. C., Janssen, P. J. C. M.,
Satish Kumar, P., Kumar Sharma, H., Der Pharm. Lett.
Hesse, J. M., Van Apeldoorn, M. E., Meijerink, M. C. M.,
2014, 6, 47–55.
Verdam, L., Zeilmaker, M. J., RIVM Report 711701 025,
The Netherlands 2001. [122] Vijaya Bhaskar Reddy, A., Venugopal, N., Madhavi, G.,
Gangadhara Reddy, K., Madhavi, V., J. Pharm. Biomed.
[105] Frumkin, H., Gerberding, J.L., Toxicological Profile for
Anal. 2013, 84, 84–89.
Phenol, Agency for Toxic Substances and Disease


C 2014 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.jss-journal.com

Potrebbero piacerti anche