Download as pdf or txt
Download as pdf or txt
You are on page 1of 23

Analytica Chimica Acta 890 (2015) 21e43

Contents lists available at ScienceDirect

Analytica Chimica Acta


journal homepage: www.elsevier.com/locate/aca

Review

Challenges in the determination of aminoglycoside antibiotics,


a review
Faten Farouk a, b, Hassan M.E. Azzazy b, Wilfried M.A. Niessen c, d, *
a
Ahram Canadian University, Faculty of Pharmacy, Pharmaceutical Chemistry Department, 6th of October City, Giza, Egypt
b
The American University in Cairo, School of Sciences and Engineering, Department of Chemistry, P.O. Box 74, New Cairo 11835, Egypt
c
VU University Amsterdam, Faculty of Sciences, Department of Chemistry and Pharmaceutical Sciences, AIMMS Division of BioAnalytical Chemistry,
De Boelelaan 1083, 1081 HV Amsterdam, The Netherlands
d
hyphen MassSpec, Herenweg 95, 2361 EK Warmond, The Netherlands

h i g h l i g h t s g r a p h i c a l a b s t r a c t

 Residues of antibiotics in the envi-


ronment and food represent a major
concern.
 Aminoglycosides lack a chromophore
necessary for UV detection.
 Aminoglycosides show poor chro-
matographic retention on RPLC
column.
 A general workflow for the analysis
of aminoglycosides is presented.

a r t i c l e i n f o a b s t r a c t

Article history: Residues of antibiotics (ABs) in the aquatic environment and in food of animal origin represent a major
Received 12 March 2015 concern, as prolonged exposure to ABs is a serious health hazard, related to both the side effects of
Received in revised form prolonged use and the risk of developing bacterial resistance to various ABs. Given the low levels of the
14 June 2015
AB residues in complex matrices, the development of sensitive analytical methods represents a major
Accepted 18 June 2015
challenge. This is certainly true for the aminoglycoside ABs (AGs) which lack a chromophore and show
Available online 7 August 2015
poor chromatographic properties in reversed-phase liquid chromatography. This paper reviews the
current state of the art in the determination of AGs. Attention is paid to extraction, sample clean-up,
Keywords:
Antibiotic residue
chromatographic separation, and detection of AGs in both environmental and food samples and in
Residue analysis plasma and serum. A general workflow for the analysis of AGs is presented which takes into account the
Aminoglycosides matrix and required level of information.
Food contamination © 2015 Elsevier B.V. All rights reserved.
Water contamination
Therapeutic drug monitoring

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 22
2. Application, pharmacology and toxicity of aminoglycosides . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 24

Abbrevations: AB, antibiotic; AG, aminoglycoside.


* Corresponding author. hyphen MassSpec, Herenweg 95, 2361 EK Warmond, The Netherlands.
E-mail address: w.m.a.niessen@vu.nl (W.M.A. Niessen).

http://dx.doi.org/10.1016/j.aca.2015.06.038
0003-2670/© 2015 Elsevier B.V. All rights reserved.
22 F. Farouk et al. / Analytica Chimica Acta 890 (2015) 21e43

3. Analytical challenges in aminoglycoside determination . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 24


3.1. Residues in food substances . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 26
3.2. Residues in water compartments . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 26
3.3. Impurity profiling and identification . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 26
3.4. Clinical determination . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 26
4. Extraction and clean-up methods . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 29
5. Liquid chromatographic methods . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 29
5.1. Ion-pair liquid chromatography . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 30
5.2. Capillary electrophoresis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 31
5.3. Thin-layer chromatography . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 31
5.4. Reserved-phase liquid chromatography after derivatization . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 31
5.4.1. Pre-column derivatization . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 31
5.4.2. Post-column derivatization . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 32
5.4.3. High-pH mobile phase . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 32
6. Liquid chromatographyemass spectrometry . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 32
6.1. Fragmentation of aminoglycosides in MS/MS . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 32
6.2. IPLCeMS/MS . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 34
6.3. HILICeMS/MS . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 35
6.4. High-pH RPLCeMS . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 36
6.5. Comparison between IPLCeMS and HILICeMS . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 36
7. Non-chromatographic methods . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 39
8. Conclusions and perspectives . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 39
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 39

1. Introduction dibekacin [2]. Their general structure is characterized by two or


more aminosugars linked by glycosidic bonds to an aminocyclitol
Aminoglycosides (AGs) are broad-spectrum antibiotics (ABs) scaffold. Two types of aminocyclitol scaffolds are found: streptidine
that are used against Gram-positive and Gram-negative bacteria. or deoxystreptamine (Fig. 1). The latter is more abundant [3,4]. The
Their history goes back to 1943 with the discovery of streptomycin structures of some important AGs are given in Table 1.
[1]. AGs are produced by various species of Streptomyces and AGs (and its related substances) possess oto- and nephrotoxicity
Micromonospora bacteria. AGs derived from Streptomyces species which, due to its low cost, did not hinder the widespread use of AGs
are given the suffix “mycin”, while those derived from Micro- in therapeutic and veterinary applications. Because of this, AG
monospora are given the suffix “micin”. In addition, there are semi- residues are abundant in the environment [5]. The analysis of AGs is
synthetic products such as netilmicin, arbekacin, isepamicin, and challenging due to the lack of a chromophore, which prevents

Fig. 1. Chemical classification of aminoglycosides.


F. Farouk et al. / Analytica Chimica Acta 890 (2015) 21e43 23

Table 1
Structural classification of some aminoglycoside.

Streptidine derivatives Streptomycin

Dihydrostreptomycin

Deoxy-streptamine derivatives 4,5-Substituted Neomycin

Paromomycin

Ribostamycin

(continued on next page)


24 F. Farouk et al. / Analytica Chimica Acta 890 (2015) 21e43

Table 1 (continued )

4,6-Disubstituted Kanamycin

Tobramycin

Gentamicin

direct determination by UV absorption. Advancement in the recently announced it will sell antibiotics-free chicken within two
detection technologies for liquid chromatography (LC) such as the years in its US restaurants [7]). ABs may also be used either directly
use of pulsed amperometric detectors (PAD), evaporative light- or indirectly during the production, processing and storage of milk
scattering detection (ELSD) and tandem mass spectrometry (MS/ and milk products.
MS) made their determination possible to some extent. However, AGs have a narrow therapeutic window, e.g., 2e10 mg mL1 for
these advanced techniques are not applicable in poor economies tobramycin [8], and their pharmacokinetics may vary from one
and developing countries, as these techniques are too expensive person to another according to differences in renal function and
and require highly trained personnel. The total number of LCeMS/ tissue distribution. Therefore, close monitoring of the serum level is
MS instruments is limited in these countries and their use is mostly required to minimize potential toxic effects, especially in patients
restricted to cost-worthy bioequivalence studies. The LCePAD is with renal impairment [9] and in long-term treatment, as AGs
even rarer in these countries, but much less expensive than LCeMS/ accumulate in tissues [8].
MS. The toxicity of AGs includes (usually reversible) nephrotoxicity,
In this paper, we critically review the analytical methods in use (irreversible) ototoxicity in the cochlea and vestibular organs and,
for AGs. After briefly discussing the pharmacology and toxicity, the rarely, neuromuscular blockade and hypersensitivity reactions.
challenges in the analytic method development are addressed. Streptomycin and gentamicin are primarily vestibulotoxic, causing
dizziness, ataxia, and/or nystagmus, whereas amikacin, neomycin,
2. Application, pharmacology and toxicity of aminoglycosides dihydrosterptomycin, and kanamycin are primarily cochleotoxic,
causing permanent hearing loss [10]. The polycationic nature of the
AGs have a poor systemic absorption. Therefore, they are AGs causes poor oral absorption, poor penetration into cerebro-
administered by injection or topically in ointments, creams, ear spinal fluid, and a rapid renal clearance. However, the polycationic
drops, and nasal drops. charge may also contribute to their nephrotoxicity [8,10].
In veterinary applications, AGs as well as other ABs are not only
administered for their bactericidal or static properties, but also (at 3. Analytical challenges in aminoglycoside determination
sub-therapeutic doses) as a growth promoter, especially in young
growing animals and poultry [6], and to prevent possible infection. The analysis of AGs is important for a number of applications,
This type of administration has been strictly forbidden within the i.e., residue analysis in food of animal origin (food safety) and, given
European Union (EU) since 2006, mainly because of the risks of its frequent use in veterinary applications, in environmental sam-
allergic and physiological problems that might directly occur to ples such as different water compartments and soil. In addition,
consumers and indirectly in causing AB resistance (McDonalds has analysis of AGs and their related products is important in drug
F. Farouk et al. / Analytica Chimica Acta 890 (2015) 21e43 25

Table 2
MRL of some aminoglycosides in various food (defers for CODEX for vet drugs) [11].

Active ingredient Species or product Market commodity (mg kg1)

Kidney Fat, liver, muscle Milk Eggs

Dihydrostreptomycin Cattle 1000 600 200


Hogs 1000 600
Sheep 200
Neomycin Cattle 10,000 500 1500
sheep 10,000 500 1500
Pig 10,000 500
Turkey 10,000 500
Spectinomycin Cattle 1000 600
Chicken 5000 2000 (fat and liver)
500 (muscle)
Streptomycin Chicken 1000 600
Pig 1000 600
Gentamicin Cattle 5000 100 (muscle, fat) 200
2000 (liver)
Pig 5000 100 (muscle, fat)
2000 (liver)
Avilamycin Turkey 200 200 (muscle, fat)
300 (liver)
Chicken 200 200 (muscle, fat)
300 (liver)
Pig 200 200 (muscle, fat)
300 (liver)
Rabbit 200 200 (muscle, fat)
300 (liver)

formulations and in therapeutic drug monitoring (TDM) in body concentration are difficult to achieve. In conventional reversed-
fluids and tissues. phase LC (RPLC), the AGs show little interaction and thus little
Given these application areas, chromatographic separation retention. In addition, the AGs do not have a chromophore or flu-
methods play an important role in the analysis of AGs. Next to the orophore, thus ruling out the use of common UV and fluorescence
application-related problems, outlined below, there are some detectors.
general issues. AGs are highly polar compounds, present in poly-
ionic form in aqueous liquids. Thus, extraction and/or pre-

Household Hospitals Industry Farms


1
DomesƟc disposal

Sewage 2

Sewage
treatment

+ +
Soil Surface water 3 Soil

Ground water Drinking water Ground water

Consequences of anƟbioƟc contaminaƟon:


1- AnƟbioƟc contaminated Human food
2- Development of mulƟple resistant bacteria
3- DestrucƟon of aquaƟc environment

Fig. 2. Environmental contamination with antibiotics.


26 F. Farouk et al. / Analytica Chimica Acta 890 (2015) 21e43

3.1. Residues in food substances II). Neomycin LP-A (3-acetyl neamine) and LP-B (mono-N-acetyl
derivatives of neomycins A and B) can also be found. Of these, the
AB residues in food are a challenge because in many cases the AB neomycin B and neomycin C are the most active species. The Eu-
species is unknown and the expected levels are also unknown. ropean Pharmacopoeia limits the neomycin C content in neomycin
Thus, screening to determine the presence and identity of AB is to 3e15% [20]. Other AGs may also contain several associated
needed prior to its quantitative determination. This represents related substances, some of which may even be still undiscovered
challenges in both the extraction of AB residues as well as their and/or difficult to separate. Serious research has to be done in that
detection and quantitation. In this review, we also pay some area.
attention to AG residue analysis in developing countries, where Impurity profiling may be even more important for developing
strict documentation and efficient regulatory agencies in relation to countries, where local pharmaceutical industries preferably
food safety are less common than for instance in the EU. import the active ingredients, often from the least expensive
In the past, some countries have proposed a zero limit of AB source with minimal acceptable quality, and only formulate them
residue in food products. Currently, maximum residual limits into a final pharmaceutical product. Thus, the bulk material may
(MRLs) have been established, providing compromises between contain impurities ranging from synthetic starter materials, re-
producer and consumer benefits. Some typical MRL values, estab- agents, solvents, intermediate compounds to degradation prod-
lished by the EU and FDA, for the AG residues in milk, edible tissues, ucts. Many of these potentially harmful components will not be
honey and other food stuff are given in Table 2 [11]. To reinforce this detected by the available analytical methodologies, which in the
regulations and for its application in developing economies, cheap, case of AGs are even more demanding, as the separation of the
fast and to a great extent inclusive methods are needed that can isomeric impurities still is challenging, even when sophisticated
decrease the number of samples and simplify the analytical tech- technology is used.
nique for instant grading and prohibition of food containing ABs Analytical technologies for impurity profiling of AGs in practice
above the MRL. very much rely on advanced technologies, such as LCeMS, LCePAD
Analytical methods for monitoring AB residues in food stuff can and LCeELSD. Chromatographic and non-chromatographic
be classified as screening methods, in which its detection limit (immunoassay, spectroscopic and microbiological) methods are
must be lower than the MRL of the analyte, and quantitative reported for AG determination. Microbiological assays are generally
methods. excellent regarding their specificity. It also provides insight in the
effectiveness of the drug molecule [21]. A microbiological assay for
3.2. Residues in water compartments neomycin sulfate is recommended by the WHO [22].

AGs are excreted unchanged from human and animal bodies. In 3.4. Clinical determination
addition, the intentional and unintentional disposal of AGs from the
pharmaceutical industry and hospital sewage increases the level of AGs are an old group of ABs with known ototoxicity, neph-
contamination of sewage and surface water [12]. Generally, only rotoxicity and some neuromuscular toxicity issues. However, to
half of the prescribed drugs are removed by sewage water treat- date they are clinically used against serious infections by gram
ment plants according to the report by the International Joint negative bacteria due to the relatively low resistance developed
Commission [13] (Fig. 2). Appropriate dealing with sewage water against it.
and hospital waste water is a critical problem in developing econ- Given the narrow therapeutic window, TDM is important in AG
omies due to technical, health and environmental shortcomings therapy in order to adequately maintain an active plasma concen-
among many of the sector stake holders. AB residues in sewage tration of AG against bacteria and meanwhile avoid its associated
water represent a great risk in the development of AB resistant toxicity. The ratio between the therapeutic and toxic plasma con-
species. AB residues may destroy the aquatic environment [12]. centration of AG is critical [8]. Furthermore, clearance of AG mainly
In the analysis of sewage and surface waters, pre-concentrating depends on the kidney function, thus individual variation in the
sample preparation procedures are required to reach detectable pharmacokinetics of AG exists due to difference in renal function
levels of AG residues. As an example, solid-phase extraction of and tissue distribution, e.g., in patients with kidney disease or
gentamicin on a weak cation exchanger had been applied for elderly patients with impaired kidney functions [8]. Thus, a suitable
determination of gentamicin in hospital water [14]. method for the plasma monitoring of AGs must be readily available
Analytical technologies for residue analysis of AGs, both in food at hospitals and points of care [8].
and environmental samples, often involve immunoassays for Relevant plasma concentrations in AG therapy are the peak
screening and LCeMS for quantification and confirmation of iden- plasma concentration (Cmax), which is the maximum plasma level
tity. Immunoassay (ELISA) was reported for the determination of reached after dose administration, and the trough concentration,
gentamicin, neomycin, streptomycin, and dihydrostreptomycin in which is the minimum plasma concentration before the next dose
milk and kidney tissue [15]. For AGs, lacking a chromophore, is administered. The trough level is used to ensure adequate
spectrophotometric methods can only be applied after chemical clearance of AG from the kidney. If optimum trough level is not
derivatization of the analytes [16e19]. This type of fast and rela- reached, the treatment regimen should be revised to avoid AG
tively simple tests, requiring little advanced instrumentation, can toxicity.
only be applied to not-too-complex matrices. They provide a AG therapy generally involves either a preventive one-dose
limited dynamic range, usually not down to residue level (ng kg1). administration for surgical prophylaxis, an empirical therapy in
Therefore, methods including LC are more appropriate. which the illness is severe and pathogen is unknown (2 days
therapy), or a directed therapy in which the pathogen is known
3.3. Impurity profiling and identification (long duration therapy of >3 days). The latter may involve different
dosing regimens (extended, traditional and synergistic regimens).
Many of the aminoglycosides are actually composed of mixtures. Generally, the plasma level should be monitored if the AG program
Neomycin, for instance, generally is a mixture of neomycin A is prolonged over 3 days (direct therapy). Sampling time largely
(neamine), neomycin B (framycetin), neomycin C, as well as neo- depends on the dosing regimen [23e30].
mycins D (paromamine), E (paromomycin I) and F (paromomycin In an extended dose regimen, one sample is recommended
F. Farouk et al. / Analytica Chimica Acta 890 (2015) 21e43 27

Aminoglycoside Therapy
Type

PrevenƟve Empirical Direct


therapy therapy therapy
Eligibility

Serious illness
Surgical
pathogen Pathogen known
prophylaxis
unknown
DuraƟon

Long therapy > 3


One dose 2 Days therapy
days
Monitoring
in plasma

Monitoring
Monitoring unrequired
required

ClassificaƟon of direct therapy and general guidelines for its monitoring


Type

Extended dosing TradiƟonal SynergisƟc


“Once Daily” dosing dosing
Sampling

One sample One sample Peak level measured 30 min aŌer 3rd dose
(6 – 14) h of 24 h aŌer Trough level measured 30 min prior 4th
first dose first dose dose

Meet Reached
Plasma

values in desired Within reference interval


level

nomogram trough

ConƟnue Therapy

Fig. 3. A summary of the AG therapy regiments and their therapeutic monitoring requirements [23e31].

6e14 h after the dose. Additional sampling may be done prior to the trough levels are monitored. Sampling for peak level determination
second dose (trough) to assure adequate clearance of the first dose. should be 30 min after the end of the infusion while for trough
For traditional as well as synergistic regimens, both peak levels and levels sampling should be “within 30 min” prior the next dose.
28 F. Farouk et al. / Analytica Chimica Acta 890 (2015) 21e43

Preferred pathway Pure Aminoglycoside


Crude Sample Substance
Un Preferred pathway
XXXXX

Sample prepara on
Forbidden pathway

Contains protein No protein

To remove buffer if any

No extrac on needed
Organic solvent Acid
precipita on precipita on

No Cl- (e.g. TFA) Cl- acid( e.g. TCA)

To remove buffer
SPE
if any

Another method of sample clean up (de-fa ng


by hexane)

Chromatography

Chromatographic separa on
High pH mobile phase
Quan ta ve/residue Determina on of
determina on impuri es

Sensi vity Separa on Cost

Advanced Aminoglycoside
HILIC IPC column
technology deriva za on
XXXXXXX

Nega ve Posi ve Vola le Nonvola le


Pre-column Post-column
charged column charged column IPR IPR
XXXXXXXX XXXXXX

Zwi er ion
column
Detec on

MS detector
PAD

Other type of universal detector

UV/ fluorometry detector

Fig. 4. Suggested workflow for analysis of AG in different matrices. TCA; Trifluoroacetic acid (TFA); Solid phase extraction (SPE); Hydrophilic interaction liquid chromatography
(HILIC); Ion pairing chromatography (IPLC); Ion pairing reagent (IPR); Mass spectrometry (MS).

Sampling of extended dose regimen should be done after the first Pharmacists, the desired plasma/serum levels of gentamicin and
dose while that of the traditional and synergistic regimens are tobramycin in a traditional regimen should be 8e10 mg mL1
better after the third dose [23,25,29,31]. Fig. 3 summarizes the AG (10e12 times the minimum inhibitory concentration (MIC) of the
therapy regimens and their corresponding TDM recommendations. infecting organism) and <2 mg mL1 for peak and trough concen-
According to the American Society of Health System trations, respectively, while in a synergistic regimen they should be
F. Farouk et al. / Analytica Chimica Acta 890 (2015) 21e43 29

3e5 mg mL1 and <1 mg mL1, respectively. In the extended dose extraction [41].
regimen, the peak concentration should be 10e12 times MIC of the Gentamicin and kanamycin were extracted from hospital
infectious organism while the trough level should not exceed sewage water by passing 20e50 mL of water (pH 7e8) through a
1 mg mL1. For amikacin, the desired peak concentration is pre-conditioned widepore Ansys CBX SPE cartridge at 50 mL h1.
30e40 mg mL1 (10e12 times MIC of infecting organism), while the The cartridge is eluted with 1 mL of methanoleacetic acid (10:1, v/
trough level should not exceed 7 mg mL1 in both traditional and v) into polypropylene vials. The extracts were evaporated to dry-
one-dose regimens [28]. ness in a gentle nitrogen stream and reconstituted prior to injection
More recently, two sample measurements have been recom- and analysis [14].
mended to calculate the pharmacokinetic parameters of AG using A molecular imprinted polymer (MIP) was synthesized by
computerized models, such as Aladdin®, TCIWorks®, or SeBagen®, polymerization of methacrylic acid and ethylene glycol dimetha-
to enable quick calculation of future dose based on the obtained crylate in the presence of streptomycin as template molecule.
information rather than dose adjustment based on measured peak Model AGs studied were streptomycin, gentamicin, spectinomycin
and trough level which includes a trial and error period until and dihydrostreptomycin. The MIP exhibited a recognizable ca-
adequate levels are reached [25,29]. pacity and specificity to the AG. The MIP was grinded and packed as
an alternative packing material in SPE. The performance was tested
4. Extraction and clean-up methods in combination with hydrophilic interaction chromatography
(HILIC). Recoveries for the four model AGs in honey samples ranged
General steps for the extraction of AGs from the relevant from 90% to 110% [42].
matrices involve sample homogenization, protein precipitation In clinical applications, extraction of AG from plasma is typically
using acid or an organic solvent, mechanical shaking or sonication performed by two methods, i.e., protein precipitation and SPE.
for release of adsorbed AGs into solution, separation of the pre- Plasma protein precipitation involves the use of a precipitating
cipitate and the liquid phase, clean-up of the extracts using solid- agent (organic solvent or acid). After ultracentrifugation, AGs are
phase extraction (SPE) to remove the acid or salt ruminants, analyzed in the supernatant, after evaporation and reconstitution
defatting using n-hexane, and possibly pre-concentration steps. in a suitable solvent. As precipitating agents, either acetonitrile
Fig. 4 shows a general workflow of AG analysis in different matrices [43e52], methanol [53], TCA [54e57], or heptafluorobutyric acid
and for different application areas, highlighting the different (HFBA) [58] are used.
technologies that are discussed in more detail in this paper. This SPE (preceded with plasma protein precipitation) involves
section discusses general extraction and clean-up procedures conditioning of the SPE, loading of the sample, washing the car-
applied in the relevant application areas. tridge, and then eluting the analyte. Selection of the solvent
A typical method for the extraction of AGs from muscle, milk composition for each step depends on the type of SPE cartridge
and kidney (poultry, swine, equine, and bovine) involves extraction used. The eluate is evaporated to dryness and reconstituted
using an extraction solvent containing trichloroacetic acid (TCA) (mostly) in the mobile phase.
and EDTA, sample clean-up by SPE, pre-concentration using evap- A typical example is in the use of Isolute® CBA carboxylate weak
oration to dryness of the eluate from the SPE cartridge, and cation exchange SPE cartridges (100 mg, 3 mL) (Mid Glamorgan,
reconstituted in a smaller solvent volume [32]. A typical protocol UK) for the extraction of AGs from plasma. The cartridge was first
starts by adding 10 mL of the extraction solution (2% TCA and 0.1% conditioned with 2 mL methanol, followed by 2 mL H2OeNH3 (25%)
Na2EDTA$2H2O in water) to the sample, vortexing, agitating and (10:0.1, v/v). Plasma sample diluted 1:1 in water was loaded onto
subjecting it to an ultrasonic bath for 5 min prior to centrifugation. the cartridge. The cartridge was washed with methanol and dried.
The supernatant is collected in a 15-mL vial. For clean-up of the AG was then eluted from the cartridge using 1.0 mL of H2OeTFA
extract, SPE is applied, for instance a Chromabond HR-X cartridge, (10:0.1, v/v) [59].
which is first conditioned with 5 mL of methanol, 5 mL of water and Alternatively, Oasis MCX cartridges (30 mg, 1 mL) (Waters,
5 mL of extraction solution. Then, the collected extract (12 mL) is Milford, MA, USA) were used for the extraction of 6 AGs from
applied. After washing with 1 mL of water, the cartridge is dried for plasma. The cartridge was first conditioned with 1 mL methanol
30 min in an air stream. Analytes are eluted with 6 mL of methanol. and 1 mL water. After mixing with 9% formic acid (50:50, v/v), the
The eluate is evaporated to dryness under nitrogen in a TurboVap sample was loaded. Then, the cartridge was washed using 1 mL of
evaporator at 40  C. The residue is re-dissolved in 0.5 mL (muscle, 50% methanol in 9% formic acid and 0.7 mL water and finally eluted
milk) or 1.0 mL (kidney) of water, centrifuged and transferred into a with 0.8 mL 50% methanol in 25% ammonia. The eluate was then
polypropylene LC vial [32]. The use of polypropylene instead of evaporated to dryness at 70  C and reconstituted into mobile phase
glass vials is important to avoid losses due to adsorption at the glass [60].
surface. The typical recovery of AGs ranged from 94 to 103% [32].
Described modifications of this method involve somewhat different 5. Liquid chromatographic methods
concentrations and amounts of extraction solutions. Also, different
SPE materials have been applied, such as a weak cation-exchange Determination of aminoglycosides can be performed either
carboxypropyl SPE cartridge [33], a cation-exchange cartridge directly, e.g., by spectrophotometric, immunochemical, or micro-
(BakerBond SPE Wide Pore CBX [34] or DSC-WCX cartridge from biological methods (see Section 7), or after LC separation. With
Supelco [35]), a turbulent-flow pre-column (TurboFlow Cyclone P respect to the LC-based methods, there are two general challenges:
(50  0.5 mm)) [36], and a polymeric phase (Bond Elute Plexa the lack of a chromophore and the poor retention in RPLC.
cartridge from Agilent [37]). Very similar procedures were applied Because of the lack of a chromophore, UV or fluorescence
for the extraction of AGs from honey [38] and apple [39], both detection cannot be used, unless at a wavelength of 195 nm, which
involving a Waters Oasis HLB SPE cartridge, and from fish, using a is not applicable in complex (biological) samples. Instead, detection
Strata X-CW cartridge [40]. should be achieved using electrochemical detection, especially
Streptomycin, gentamicin C1, neomycin B, amikacin, and kana- PAD, electrospray ionization mass spectrometry (ESI-MS), ELSD, or
mycin together with spectinomycin and ABs from seven other charge aerosol detection (CAD). Detection by ESI-MS, ELSD or CAD
classes were extracted from veal muscle using CH3CN/H2O (86:14, requires the use of volatile mobile-phase additives, including vol-
v/v) followed by a defatting step using hexane liquideliquid atile ion-pairing agents (IPRs). Detection using PAD requires post-
30 F. Farouk et al. / Analytica Chimica Acta 890 (2015) 21e43

Fig. 5. Typical chromatograms of AG obtained by IPLC. A: Ion-pair LC separation of neomycin B, framycetin (neomycin C) and its related substances using 170 mM TFA as IPR and
ELSD. Reprinted from Ref. [20] with permission. B: Chromatogram of a commercial amikacin sample under the LC conditions mentioned. Column: 250  4.6 mm Discovery C18,
5 mm. Elution order: (1) l-HABA; (2) unknown 1; (3) unknown 2; (4) unknown 3; (5) unknown 4; (6) unknown 5; (7) unknown 6; (8) 1,60 -di-HABAekanamycin A; (9) 1,3-di-
HABAekanamycin A; (10) amikacin; (11) BB-K9; (12) BB-K11; (13) BB-K6; (14) kanamycin A; (15) BB-K29; (16) unknown 7; (17) BB-K26. Reprinted from Ref. [68] with permission.

column addition of NaOH to increase pH to 12. Alternatively, AGs aminoglycosides at 195 nm, but are not compatible with MS and
can be subjected to pre- or post-column derivatization to introduce ELSD detection. The volatile ammonium acetate buffer is compat-
a chromophore or fluorophore, thus enabling UV/VIS or fluores- ible with MS, but not applicable in 195-nm UV detection. Some
cence detection. examples of IPLC are discussed below.
Because of their highly polar character, i.e., in-solution proton- The IPLC separation of neomycin and framycetin and their
ated species in the applicable pH range, AGs show poor retention related substances using 170 mM TFA as IPR and ELSD detection is
characteristics in RPLC. The use of ion-pair liquid chromatography shown in Fig. 5A [20]. The method enabled the detection of im-
(IPLC) or HILIC seems to be the most straightforward ways to solve purities down to 0.20% (m/m) in pharmaceutical preparations [20].
this problem. HILIC is more readily combined with ESI-MS detec- Neomycin could be determined down to 0.6 mg mL1 (LOD in ELSD)
tion than IPLC. Alternatively, capillary electrophoresis (CE) can be in pharmaceutical formulations with IPLC on a C18 column using a
applied. Pre-column derivatization to introduce a chromophore mobile phase of watereacetone (50:50) containing 11.6 mM HFBA
results in derivatives that can be analyzed using RPLC. Recently, as the IPR [64]. Other examples of IPLC coupled to ELSD are the
some studies utilized a mobile phase with high pH to render AG an determination of tobramycin impurities [65] and kanamycin B in
un-ionized molecule which shows retention in RPLC [61]. The presence of kanamycin A [66] using HFBA as the IPR. Similarly,
choice between these general approaches, summarized in Fig. 4, neamine was determined in plasma using pentafluoropropionic
largely depends on the specific application area. If the aim of the acid as the IPR [67]. Streptomycin and dihydrostreptomycin were
analysis is the determination of degradation products or impurities, separated and determined on a C18 column using TFA as IPR.
derivatization may cause unwanted or unexpected modifications to Separation of streptomycin B in streptomycin and determination of
the unknown impurities or degradation products. Other applica- dihydrostreptomycin in the presence of penicillin G procaine were
tions may be directed at the sensitive analysis of particular well- also performed with ELSD detection [59].
defined target compounds. In this section, different chromato- Impurity profiling of amikacin was achieved using a sodium
graphic approaches are discussed, whereas in the next section the sulfate salt gradient (0e70% B in 38 min). The mobile phase con-
additional dimension of having MS detection is addressed (see sisted of 8.3 mM sodium 1-octanesulfonate, 14 mL L1 tetrahy-
Section 6). drofuran, 50 mL L1 0.2 M phosphate buffer (pH 3.0). In solvent A,
20 g L1 sodium sulfate was added; in solvent B, 28 g L1. The LOD
5.1. Ion-pair liquid chromatography was 0.2 mg mL1 using PAD. A typical chromatogram is shown in
Fig. 5B [68]. For the analysis of neomycin B and C in ear drops, IPLC
In IPLC, an IPR is added to the mobile phase. An IPR is a molecule was applied using an PLRP-S column. An aqueous phosphate-
containing a large hydrophobic moiety, which interacts with the buffered mobile phase (pH 3) with 0.5 M sodium sulfate and
RPLC stationary phase (C18 or PLRP-S), and an ionic site with a 6.5 mM of 1-octanesulfonate as IPR was used. Post-column addition
charge opposite to that of the analyte of interest, i.e., a negatively of 0.3 mL min1 of 0.5 M NaOH was needed in order to achieve
charged group in the case of AGs. Compounds such as alkyl sulfo- adequate PAD at pH >12. The LOD on PAD was 0.05% (m/m) (5 ng).
nates can be used as IPR, whereas volatile alternatives like per- Other components in the ear drops (polymyxin B and dexametha-
fluoroalkylacetic acids (trifluoroacetic acid (TFA) up to sone) were analyzed separately using RPLC with UV detection
heptafluorobutyric acid (HFBA)) have to be applied when ESI-MS is [69,70]. Coupling of IPLC to PAD was also applied to the in vitro
used in detection. The effect of alkyl chain length of alkyl (C5, C6, or release and ex vivo permeation studies of paromomycin [71].
C7) sulfonates as IPR on the retention was tested for the separation Analysis of micronomicin and its related substances using IPLC
of streptomycin and dihydrostreptomycin. The retention increased coupled to pulsed electrochemical detector (PED) was reported.
with the length of the alkyl chain. Hexane sulfonate was chosen to The stationary phase was a Hypersil BDS C18 column and the
give optimal separation in a relatively short time [62]. As high mobile phase consisted of 1% acetonitrile: 99% aqueous solution
concentrations of IPR (>20 mM) may damage the RPLC column containing 1.25% (v/v) TFA, 0.025% (v/v) pentafluoropropionic acid
material [63], the IPR concentration should be kept as low as and 0.85% (v/v) 50% NaOH, pH was 2.6 (adjusted by 0.5 N NaOH).
possible while achieving appropriate retention and peak shape. Post-column, the pH of the effluent was adjusted to 12 by adding
In most cases, an additional buffer system is applied in IPLC in 0.5 N NaOH to enhance PED detection. LOD was 0.08 mg mL1. The
order to control the pH of the mobile phase, and thus the level of method was linear in range 0.25e60 mg mL1. Nineteen impurities
protolysis of the analytes. Acetate and phosphate buffers are most were identified and the method was applied to commercial sam-
commonly used. Phosphate buffers allow UV detection of ples [72].
F. Farouk et al. / Analytica Chimica Acta 890 (2015) 21e43 31

Spectinomycin and its impurities were determined by IPLC was amino F254S plates while the mobile phase was methanol-
coupled to CAD detector. The stationary phase was a RP C18 column acetonitrile (40:60, v/v). Detection was by a bioluminescence
and the mobile phase was composed of 0.3% TFA and acetonitrile assay [83].
(98:2). The LOD was 0.5 mg mL1. Two linearity ranges were vali-
dated for the assay: 2e45 and 80e200 mg mL1. The method was 5.4. Reserved-phase liquid chromatography after derivatization
applied to pharmaceutical preparations [73]. Neomycin and its
impurities were separated using HFBA as an IPR and CAD for In a targeted analysis directed at particular AGs, pre-column or
detection [74]. post-column derivatization can be applied to introduce a chromo-
phore or fluorophore. The choice between the pre- and post-
5.2. Capillary electrophoresis column derivatization depends on several factors such as: the
required sensitivity, the amount of sample present and the eco-
Given the ionic character of the aminoglycosides over a wide pH nomic and experience level of the laboratory. Post-column deriv-
range, CE can also be applied for the separation. Several examples atization requires more complex instrumentation. Derivatization of
have been reported [75e79]. Neomycin was determined in oint- AGs can also be applied for direct spectrophotometric analysis,
ments using an aqueous background electrolyte containing 35 mM without chromatography.
phosphate and 15 mM acetate buffer (pH 4.7), a polyacrylamide- Derivatization of AGs is directed at modifying the primary amine
coated fused-silica capillary, and UV detection at 200 nm [75]. functions. Therefore, a number of derivatization reagents can be
Netilmicin, tobramycin, lincomycin, kanamycin, and amikacin were applied, which have been described for other purposes as well.
determined simultaneously in pharmaceutical preparations using Some reagents such as 9-fluorenylmethyl chloroformate (FMOC-
of 125 mM NaOH as a background electrolyte, an uncoated fused- Cl), dansyl chloride, and 1-fluoro-2,4-dinitrobenzene (Sanger re-
silica capillary, and electrochemical detection involving a copper agent) can only be applied in pre-column derivatization, while
microparticle-modified carbon fiber microdisk array electrode. some others in principle can be used either in pre-column or in
LODs were ranging from 1.2 mM for amikacin to 6.7 mM for linco- post-column derivatization, as is the case for o-phthaldialdehyde
mycin [76]. CE coupled to contactless conductivity detection was (OPA) and 1,2-naphthoquinone-4-sulfonate (Folin's reagent).
used for the determination of amikacin [77] and tobramycin [78]
with their related impurities. 5.4.1. Pre-column derivatization
Pre-column derivatization of AGs alters their chromatographic
behavior, enabling the use of conventional RPLC rather than the
5.3. Thin-layer chromatography more complicated IPLC. Pre-column derivatization is easy to
perform, also in high-throughput applications, as the sample pre-
Thin-layer or planar chromatography (TLC) has been advocated treatment step is decoupled from the separation and detection
in the WHO International Pharmacopoeia [22] for the identification step. It is more flexible than post-column derivatization in terms of
of neomycin and its impurity neamine; detection is performed by reaction time, reaction solvent, and eventual elimination of excess
spraying triketohydrindene/butanol for visualization in daylight. In reagent. However, the additional treatment steps may result in
other studies, visualization of neomycin has been done by densi- analyte losses. A complete derivatization reaction into stable
tometry [80,81] or fluorimetry [82]. Although TLC can be relatively products is necessary, and excess reagent or derived products
cheap, as no extensive instrumentation is required, the repeat- thereof may interfere in the analysis.
ability of the assay for quantitative determination is poor and the As an example, the pre-column derivatization of amikacin with
method is difficult to validate. HPTLC was performed for the FMOC-Cl was performed for preclinical pharmacokinetic studies
determination of AG in food of animal origin. The stationary phase and in parenteral formulations. The derivatization reaction pro-
ceeds in aqueous borate buffer (pH 7.3) at room temperature with
the instantaneous formation of the corresponding derivative of
amikacin which is stable for at least 48 h. Isepamicin was used as
the internal standard (IS). After derivatization, the resulting de-
rivatives can be analyzed using a C8 RPLC column, a water-
eacetonitrile (35:65, v/v) mobile phase without IPR. The LOD was
90 ng mL1 with fluorescence detection (excitation 265 nm and
emission 315 nm). A typical example is shown in Fig. 6 [84]. Simi-
larly, pre-column FMOC-Cl derivatization and fluorescence detec-
tion of isepamicin in rat plasma was performed using amikacin as
an IS [45]. For the analysis of neomycin, netilmicin and sisomicin in
cell culture media or plasma, FMOC-Cl derivatization was per-
formed. Separation was performed on an ODS Hypersil column
using a mobile phase of acetonitrileewater (90:10, v/v) at a flow
rate of 1 mL min1 with fluorescence detection. The LOQ was
0.1 mg mL1 [85]. Similar FMOC-Cl derivatization and detection was
performed for the determination etimicin in rat plasma using an RP
C8 column [52].
For the determination of tobramycin in human plasma, pre-
column derivatization was developed using 1-naphthyl isothiocy-
anate (NITC) in pyridine at 70  C. The stationary phase was a Pur-
ospher STAR RP-18e column and the mobile phase
Fig. 6. Separation of (1) amikacin and (2) isepamicin (I.S.) after pre-column FMOC-Cl-
watereacetonitrile (50:50, v/v). UV detection was performed at
derivatization in a rat plasma sample 2 h after intravenous administration of 230 nm. Linearity range was 0.93e9.34 mg L1; the LOD was
10 mg kg1 amikacin. Reprinted from Ref. [84] with permission. 0.23 mg L1 [51]. The same method was applied in the analysis of
32 F. Farouk et al. / Analytica Chimica Acta 890 (2015) 21e43

kanamycin A in human plasma and oral dosage [46]. special column materials as well as a detection system that are
Pre-column derivatization with OPA in the presence of b-mer- stable, durable and applicable at highly alkaline conditions. The
captopropionic acid or another mercaptol is also widely used. The analysis of tobramycin in pharmaceutical formulations using high-
resulting derivatives can be detected using fluorescence detection pH RPLC was described. A Purospher RP-8e column was used as a
(excitation 340 nm and emission 450 nm). A typical example is the stationary phase using mobile phase of 0.05 M diammonium
determination of neomycin in milk [86]. Neomycin was extracted hydrogen phosphate (pH adjusted to 10.0 ± 0.05 using 25% aqueous
from milk using an Amberlite CG-50 ion-exchange resin column tetramethyl ammonium hydroxide) at 1 mL min1 and UV detec-
and derivatized on that column with OPA. After elution with po- tion at 210 nm [94]. A Waters XBridge C18 column with a high-pH
tassium borate buffer/methanol, the derivatives are analyzed on a mobile phase (pH > 11) was applied for determination of plazo-
HISEP LC column with fluorescence detection. The LOD is 50 mg L1 micin and its impurities [61]. The mobile phase was a gradient of
in milk; typical recoveries range from 94 to 102% [86]. 0.25 M ammonium hydroxide in water and acetonitrile. UV detec-
Despite the fact that pre-column derivatization could avoid the tion at 210 nm was performed. The linearity range was
use of IPLC, in some applications IPLC of derivatized AGs is still 0.0025e3.0 mg mL1. The high detection range can be attributed to
used. The determination of sisomicin and its N-ethyl derivative, the poor UV absorbance of AGs [61].
netilmicin, in whole blood is an example of this [87]. The com- Applications of high-pH RPLC with MS detection are discussed
pounds were extracted from dried blood spots using a phosphate below. The high pH mobile phase for AG separation is readily
buffer by ultrasonication. After OPA derivatization, the derivatives applicable with PAD systems.
are analyzed using IPLC on a C18 column and with 1-
heptanesulfonate as IPR. The method was used for TDM in geri- 6. Liquid chromatographyemass spectrometry
atric and pediatric patients [87].
Pre-capillary derivatization of paromomycin and its impurities LCeMS is widely applied in the determination of AGs. The
with OPA prior to CE analysis was performed with UV detection at challenge in the LCeMS analysis of AGs is their structural similarity
330 nm [88]. Pre-capillary derivatization into sulfindocyanine which results in similar product ions upon fragmentation. This is to
succinimidyl esters was used for the determination of AGs in milk be added to the challenges in LC separation, already addressed in
by micellar electrokinetic chromatography (MEKC) using laser- Section 5. Thus, the selection of precursor-ion/product-ion combi-
induced fluorescence detection [89]. nation for selected-reaction monitoring (SRM) should be done with
great care to avoid interferences. After briefly discussing the frag-
5.4.2. Post-column derivatization mentation of protonated AGs in MS/MS to derive characteristic ions
Post-column derivatization is instrumentally more complex, for SRM transitions, applications of LCeMS with HILIC, IPLC or high-
because a post-column reactor and one or more additional pumps pH RPLC are discussed.
are necessary. The possibilities of post-column derivatization is
generally limited by the reaction time, as only relatively fast re- 6.1. Fragmentation of aminoglycosides in MS/MS
actions can be applied, and by the solvent system, as the reaction
conditions should be compatible with the mobile-phase composi- The general fragmentation of AGs in positive-ion MS/MS can be
tion. However, as long as it is repeatable, the reaction does not have readily understood using early data on gentamicins [95] (cf.
to be complete, and long-term stability of the derivative generally is Fig. 7A). The three major fragment ions observed were an ion with
less of an issue. Obviously, for the AGs, the difficulties in chro- m/z 160 due to the garosamine unit (ring A in Fig. 7A), an ion with
matographic separation, demanding for the use of IPLC, still exist m/z 163 due to the 2-deoxystreptamine unit (ring B), and an ion due
when performing post-column derivatization. to the purpurosamine unit (ring C), being m/z 129 for gentamicin
An example of post-column derivatization is the quenching of C1a, m/z 143 for the gentamicins C2, C2a and C2b, and m/z 157 for
Cu2þ-luminol chemiluminescence by complex formation between gentamicin C1. Additionally, an ion with m/z 322 due to the loss of
Cu2þ and AGs [90]. This approach is applied in the determination of either the purpurosamine unit (resulting in the AB fragment) or the
amikacin in plasma and urine, using RPLC on a C18 column and a garosamine unit (resulting in the BC fragment) can be observed
mobile phase consisting of 102 mol L1 aqueous potassium (see Fig. 7A). Subsequent losses of H2O, NH3, or CH3NH2 from these
hydrogen phthalate (pH 3.35) and acetonitrile (90:10, v/v). The LOD fragment ions may result in secondary fragment ions. This frag-
was 50 mg L1 in plasma or urine [90]. mentation pattern is in agreement with schemes reported by
Post-column derivatization of paromomycin in bulk with OPA others [96e102].
and fluorescence detection is achieved after IPLC using a C8 column Under positive-ion ESI-MS conditions, depending on the
and sodium 1-heptanesulfonate as IPR. The LOD was 10 ng [91]. mobile-phase composition, most AGs may show both double-
The potential of IPLC coupled to resonance Rayleigh scattering charge ions [M þ 2H]2þ and single-charge ions [M þ H]þ [103]. In
(RRS) detection was explored for the determination of AGs ami- the absence of IPR, e.g., in HILIC, the double-charge ion is most
kacin, netilmicin, and etimicin in plasma and urine. After IPLC abundant, whereas the single-charge ion is most abundant in IPLC.
separation on a Synergi Hydro-RP column (Phenomenex) using a In negative-ion ESI-MS, applied under high-pH RPLC conditions,
0.1% TFA (pH 2.2) as IPR at a flow rate of 0.4 mL min1, binary deprotonated molecules [M  H] are observed; fragmentation
compounds are formed after a post-column reaction with Congo under negative-ion conditions was not investigated [104]. Upon
red. From the RRS spectrum, it was established that detection is fragmentation in positive-ion MS/MS, the double-charge ions
best performed at 370 nm. LODs were 0.018, 0.021 and generate mostly single-charge fragment ions, similar to gentami-
0.055 mg mL1 for amikacin, netilmicin, and etimicin, respectively cins (cf. Fig. 7A). Characteristic fragments for various AGs are
[49]. Other examples of post-column derivatization of AGs involve summarized in Table 3 [96,103,105e107]. The ions frequently
the determination of neomycins B and C in animal feed [92] and of applied in SRM transitions are underlined in Table 3. Identification
streptomycin residues in honey [93]. of fragment ions of AGs, relevant to SRM, has been reported [96].
The relative abundance of fragment ions may differ, depending on
5.4.3. High-pH mobile phase whether single-charge or double-charge precursor ions are frag-
The use of a mobile phase with a pH 11 is believed to render mented, e.g., the most abundant fragment ions from single-charge
the AGs in an unionized form, thus enabling RPLC. This requires streptomycin and dihydrostreptomycin are the ions with m/z 263
F. Farouk et al. / Analytica Chimica Acta 890 (2015) 21e43 33

Fig. 7. (A) General fragmentation of aminoglycosides, illustrated for gentamicin C1. (B) Proposed structures for important fragment ions of spectinomycin.

Table 3
Fragmentation of aminoglycosides in MSeMS (for annotation, cf. Fig. 7a). The underlined m/z values are used as product ions in SRM [96,105e107].

Aminoglycoside [M þ H]þ m/z [M þ 2H]2þ m/z A m/z B m/z C m/z AB m/z BC m/z Other m/z

Amikacin 586 293.7 162 264 162, 180 425 425 102, 163, 247
Apramycin 540 270.6 162 235 145, 163 396, 378 379 199, 217
Arbekacin 553 277.2 129 264 162, 180 392 425 102, 163
Dihydro-streptomycin 584 292.6 176 165 245, 263 340 409 246
Streptomycin 582 291.6 176 167 245, 263 342 411 246
Gentamicin C1a 450 225.7 160 163 129, 147 322 291 112
Gentamicin C2, C2a, C2b 464 232.7 160 163 143, 161 322 305
Gentamycin C1 478 239.7 160 163 157, 175 322 319 139
Hygromycin B 528 264.6 n.a. n.a. 159, 177 370 n.a. 352, 257
Kanamycin A 484 242.6 161 163 162, 180 323 324 205, 102
Kanamycin B 485 243.1 162 163 162, 180 324 324
Neomycin B 615 308.2 293 163 161, 179 455 323
Paromomycin 616 308.7 293 163 162, 180 455 323 203, 161
Netilmicin 476 238.2 127 191 160, 180 317 350 458, 281
Ribostamycin 455 228.2 133 163 161, 179 295 323
Sisomycin 448 224.6 160 163 127, 145 322 289 271, 254
Tobramycin 468 234.6 145 163 162, 180 307 324
34 F. Farouk et al. / Analytica Chimica Acta 890 (2015) 21e43

and 245, whereas from double-charge ions the most abundant ions
are m/z 176 and 409 for dihydrostreptomycin and m/z 176 and 263
for streptomycin [108]. As can be seen from Table 3, several frag-
ment ions show the same m/z-values for different AGs. This is due
to the high structural similarity. If insufficient LC separation is
achieved in a multi-residue analysis, this may result in possible
interferences. This is an important point of attention during sample
pretreatment and analysis.
In order to identify impurities in tobramycin and kanamycin B,
the fragmentation of these two compounds was studied in some
detail [97]. The fragmentation pattern is greatly in agreement with
Fig. 7A and Table 3, although two additional fragments are dis-
cussed. The ions with m/z 349 (tobramycin) and m/z 365 (kana-
mycin B) are due to cross-ring cleavages in the C-ring. Subsequent
loss of the A-ring results in the fragment ion with m/z 205 [97].
Similarly, related substances of dihydrostreptomycin [98], genta-
Fig. 8. HPLC chromatogram of streptomycin sulfate with detection by charged aerosol
micins [99,100], vertilmicin [101], and micronomicin [102] were detector (CAD); 250  4.6 mm YMC-Pack Pro column (particle size 3 mm); column
identified. Again, detailed fragmentation schemes were developed. temperature: 40  C; mobile phase: 20 mmoL L1 of PFPA in a mixture of water/acetone
Spectinomycin is an aminocyclitol antimicrobial, closely related 99/1 (v/v); flow rate: 0.8 mL min1; injection of 10 mL of a 5 mg ml1 solution of
to aminoglycosides, produced by the bacterium Streptomyces streptomycin sulfate in water. Reprinted from Ref. [114] with permission.
spectabilis. In LCeMS, spectinomycin forms strong solvent adducts
with water or methanol [105,109]. Fragmentation schemes for injection system between the runs was found to be necessary.
spectinomycin ([M þ H]þ with m/z 333) have been proposed In another study, the determination of 34 antibacterial drugs
[110,111]. Fragment ions are observed with m/z 315 due to the loss from the same compound classes, among which were strepto-
of water, m/z 305 due to CO loss, m/z 302 due to CH3NH2 loss, and mycin, dihydrostrepromycin, neomycin, gentamicin as well as
m/z 289 due to CO2 loss. Subsequent fragmentation of the ion with spectinomycin, in fish samples was reported [40]. In this case, a
m/z 289 results in fragment ions with m/z 271 due to water loss and mobile phase of (A) 0.025% HFBA in water and (B) acetonitrile was
m/z 245 due to CO2 loss. The loss of CO and CO2 from the ion with m/ used. The gradient started at 5% B, increased to 50% B in 2 min, and
z 271 results in the fragment ions with m/z 243 and 227, respec- to 90% B in another 7 min. With column wash and re-equilibration,
tively. Another fragmentation route involves cleavage of the the total run time was 18 min. Positive-ion ESI-MS detection was
dioxane ring to a fragment ion with m/z 207, which in turn shows performed in SRM mode (characteristic SRM transitions in Table 3).
subsequent losses of water to m/z 189, of CH3NH2 to m/z 158, and of CCa and CCb were 589.8 and 529.4 mg kg1 and 714.9 and
water to an ion with m/z 140. The ion with m/z 140 can show the 571.0 mg kg1 for streptomycin and dihydrostreptomycin, respec-
loss of water to an ion with m/z 122, of CO to m/z 112, or of H2C]C] tively [40]. Because of poor extraction recovery (<60%), the method
O to m/z 98. As the fragment ions with m/z 207 and 189 are was not applicable for neomycin and gentamicin.
frequently used in SRM transitions for the analysis of spectino- Ten AGs were determined in poultry, swine, equine, and bovine
mycin, these are explained in Fig. 7B. kidney [34]. The procedure was an adaptation of the US Depart-
ment of Agriculture, Food Safety and Inspection Service (USDA-
6.2. IPLCeMS/MS FSIS) qualitative method [115] by inclusion of additional clean-up
and quantification at lower levels. The mobile phase consisted of
IPLCeMS is primarily used in multiresidue analysis of AGs in (A) water and (B) methanol, and (C) 0.1 M HFBA. The gradient
food of animal origin [34,36,37,40] and in impurity profiling program was 5e50% B in 0.75 min, 50e60% B in 9 min; C was kept
[112e114]. Some recent examples are discussed below. at 20%. With column wash and re-equilibration, the total run time
The determination of the AGs hygromycin B, dihydros- was 18 min. Detection was done in ESI-MS/MS in SRM mode. CCa
treptomycin, amikacin, kanamycin, apramycin, tobramycin, genta- and CCb varied between 1036 and 12,293 mg kg1, and between
micin and neomycin together with spectinomycin in bovine 1073 and 14,588 mg kg1, respectively. LOQs ranged from 27 to
muscles using IPLCeMS was described [37]. After extraction and 688 mg kg1 [34].
cleanup (see Section 4), providing recoveries between 71 and 98%, Characterization of tobramycin impurities using IPLCeMS on an
IPLC separation was achieved using an acetonitrile/water gradient, ion-trap MS was reported as well. The mobile phase consisting of an
containing 20 mM HFBA, on a C18 column. Compounds were aqueous solution containing 1.2 mM HFBA and 50 mM ammonium
detected in SRM with linearity ranging from 10 to 500 ng g1. LODs formate, adjusted to pH 3.0 by formic acid before bringing to vol-
ranged from 0.1 ng g1 for tobramycin to 5 ng g1 for streptomycin ume/methanol (68:32, v/v) at a flow rate of 0.2 mL min1 was used
[37]. with a C18 column. The total run time for separation was 20 min
In a multiresidue study, kanamycin and neomycin, together with [113].
34 other ABs from different classes like tetracyclines, sulfonamides, Turbulent flow chromatography (TFC) coupled to IPLCeMS/MS
(fluoro) quinolones and macrolides, were determined in chicken was used for the quantitation of different AB classes including AGs.
meat using a 50  2.1 mm Betasil phenyl hexyl column and a TFC is used to remove macromolecules and to pre-concentrate the
mobile phase of (A) 1 mM HFBA with 0.5% formic acid in water and ABs prior to analysis. The mobile phases were A (1 mM HFBA with
(B) 0.5% formic acid in acetonitrile/methanol (1:1, v/v) and a 0.5% formic acid in water) and B (0.5% FA in acetonitrileemethanol
gradient of 0e95% B in 12 min. With column wash and re- (1:1, v/v)). Detection was performed by MS/MS. The LODs were 10
equilibration, the total run time was 19 min. LOQs were 25 and and 40 mg kg1 for kanamycin and neomycin [116]. HFPA as an IPR
120 mg kg1 for kanamycin and neomycin, respectively. The deci- was also used in clinical studies to determine tobramycin [56],
sion limit CCa and detection capability CCb were 121 and amikacin [58], and etimicin [117] in serum or plasma.
143 mg kg1 for kanamycin and 602 and 704 mg kg1 for neomycin Rapid qualitative determination of 9 AGs, i.e., neomycin,
[36]. In order to avoid the risk of carryover, rigorous washing of the streptomycin, dihydrosptreptomycin, spectinomycin, kanamycin,
F. Farouk et al. / Analytica Chimica Acta 890 (2015) 21e43 35

gentamicin, apramycin, amikacin, and hygromycin, in bovine tis- A HILICeMS/MS method was reported for the determination
sue matrices was performed by IPLCeMS/MS using tobramycin as of streptomycin and dihydrostreptomycin in honey. Two HILIC
IS [96]. Due to the use of column material with 1.7 mm ID particles, columns (150  2.1 mm; 3 mm particle size) were tested: a Wa-
analysis time was only 2.4 min. Initially, paromomycin was ters Atlantis silica column and a Varian Monochrom Si column.
included, but it was found that paromomycin could be a degra- Gradient elution was performed using solvent A (acetonitrile
dation production of neomycin by conversion of an NH2 into an with 0.05% formic acid) and solvent B (water with 0.05% formic
OH group [96]. This phenomenon has been previously reported acid): 10% B for 1 min; 10e90% B in 5 min. With column wash
[118] and stresses the importance of careful and critical data and a 9-min re-equilibration, the total run time was 20 min.
evaluation. Under these conditions, streptomycin and dihydrostreptomycin
The analysis of streptomycin sulfate and 21 of its impurities was co-eluted at 8.0 min. Because the m/z values of the precursor ions
achieved using a 250  4.6 mm YMC-Pack Pro column and a mobile differ by only 2 units and the same product ions are used for both
phase of 20 mM of PFPA in watereacetone 99/1 (v/v). The run time compounds (cf. Table 3), a selectivity check should be performed.
was 70 min. Monitoring was performed by time-of-flight mass The method can be applied in the range of 5e80 mg kg1. CCa
spectrometry (TOF-MS) and confirmed by tandem-quadrupole MS/ were 11.8 and 11.5 mg kg1 and CCb were 18.9 and 19.9 mg kg1
MS. The LOQs were tested for streptomycin sulfate and streptidine for streptomycin and dihydrostreptomycin, respectively [38].
sulfate to be 4.5 mg mL1 and 0.4 mg mL1, respectively [114]. A Streptomycin, dihydrostreptomycin, apramycin, kanamycin,
typical chromatogram is shown in Fig. 8. PFPA was used as IPR for paromomycin, gentamicin, and neomycin together with specti-
the determination of amikacin, gentamicin, and vancomycin levels nomycin were determined in bovine milk and in bovine and
in human newborn plasma [57]. porcine muscle and kidney tissues. A 2-min water/acetonitrile
Gentamicin, kanamycin, and apramycin were determined in gradient with 200 mM ammonium acetate and 1% formic acid was
rat plasma in ranges 1e5,000, 20e10,000, 10e10,000 ng mL1, applied on a ZIC-HILIC column. LCeMS analysis was achieved in
respectively. In this study, TCA functioned as both a precipi- SRM mode, using characteristic SRM transitions listed in Table 3.
tating agent for the plasma proteins and an IPR. Despite the The linearity of the procedure was tested in the range from 0.25 to
fact that TCA was only present in the sample and not in the 2*MRL [32].
mobile phase; it improved the retention of AGs on the C12 The AGs streptomycin, gentamicin C1, neomycin B, amikacin,
column applied. The elution was performed in a gradient pro- and kanamycin together with spectinomycin and ABs from seven
gram using water and acetonitrile, both with 0.1% formic acid. other classes were determined in veal muscle. For the AGs, a ZIC
The flow rate was 0.25 mL min1. The gradient started as 5% B column was applied, whereas the other compounds were analyzed
for 0.1 min and B was raised to 70% at 1.5 min and then to 90% in RPLC using a C18 column [41]. For HILIC, the mobile phase
at 2.5 min and reduced again to 5% at 3.50 min and gradient consisted of solvent A (acetonitrile) and solvent B (0.4% formic acid
stopped at 3.6 min. 1 min equilibration was performed between in water). The gradient started at 3% B for 1.0 min, then ramped to
runs [54]. 85% B in 1.0 min, and was held at 85% B for 3.0 min. Column wash
Gentamicin was determined in different bovine matrices and re-equilibration resulted in a total run time of 15.0 min. MS/MS
(plasma, urine, milk, kidney tissue). The separation was performed detection was performed in SRM mode, using characteristic SRM
on a Waters ODS-AM (3.5 mm particles) column. The mobile phase transitions (Table 3). The accuracies achieved in this method ranged
was a mixture of 95% solvent A (0.11 M aqueous TFA-methanol, 1:1) from 45% to 106% [41].
and 5% solvent B (acetonitrile). Flow rate was 0.22 mL min1 and A HILIC-based multiresidue method for the analysis of 24 ABs
detection was performed by MS/MS. The applicable linearity ranges from different classes (the aminoglycosides streptomycin and
were 1e5000 ng mL1 for plasma, 2.5e2500 ng mL1 for milk, dihydrostreptomycin, together with various b-lactams, lincosa-
1e100 ng mL1 for urine, and 10e50,000 ng g1 for kidney tissues mides, macrolides, quinolones, sulfonamides, tetracyclines, and
[55]. TFA was also used as the IPR for the determination of arbe- amprolium) in chicken muscle was reported as well [121].
kacin in serum [119]. HILICeMS/MS was achieved using a ZIC column in SRM mode. The
mobile phase was composed of solvent A (acetonitrile) and solvent B
6.3. HILICeMS/MS (50 mM ammonium formate buffer at pH 2.5). The gradient elution
started with 10% B for 10 min and then ramped from 10 to 90% B in
HILIC has been recognized as a powerful alternative to RPLC 5 min, followed by equilibration. The total runtime was 15 min. The
for especially highly polar compounds. In the analysis of AGs by LOD of the method was 20 mg kg1 for aminoglycosides. The linearity
HILIC, no IPR is needed. For HILIC, different column materials was 1e100 mg kg1 for all analytes [121]. A somewhat similar
are available. A comparative study on the analysis of AGs on six method was reported for the determination of AGs in bovine and
different HILIC stationary phases, i.e., (anionic) bare silica, porcine kidney and muscle tissue using HILICeMS/MS in SRM mode
(cationic) amino phenol, and (neutral) amide and zwitter ionic on a ZIC column. The LOQs were 25 ng g1 for gentamicin, 50 ng g1
(ZIC) materials, showed that the ZIC column showed best per- for spectinomycin, dihydrostreptomycin, kanamycin, and apramy-
formance [120]. This is probably due to the “mixed-mode” cin, and 100 ng g1 for streptomycin and neomycin M [33].
retention mechanism. The ZIC column provides interaction to Neomycin was determined in human serum using a ZIC-HILIC col-
both positively-charged amino groups and some hydroxyl umn using a mobile phase composed of solvent A (5/95/0.2, v/v/v,
groups. The optimal mobile phase was composed of A (0.2% acetonitrile, 2 mM ammonium acetate and formic acid) and solvent
formic acid in acetonitrile) and B (175 mM ammonium formate) B (95/5/0.2, v/v/v, acetonitrile, 2 mM ammonium acetate and formic
at pH 4.5. Detection was performed by MS/MS in SRM mode. acid). The gradient elution started with 80% B until 0.6 min and was
Absence of the IPR reduces mobile-phase related ionization decreased to 20% B until 5.8 min then elevated again to 80% until
suppression and results in improved sensitivity compared to 8.8 min the LOQ was 100 ng ml1 [122]. A similar approach was
IPLC. In the comparative study on HILIC material [120], the ZIC applied in the simultaneous determination of amikacin, gentamicin,
provided LOQs that ranged from 6 mg L1 for spectinomycin to kanamycin, neomycin, paromomycin, and tobramycin in human
154 mg L1 for neomycin. Therefore, HILICeMS is an attractive serum [60]. The use of a ZIC column was also reported for the
tool in both (veterinary) residue analysis and impurity determination of AGs in food of animal origin [123].
profiling. Some typical applications are discussed below. Impurity profiling of streptomycin and dihydrostreptomycin
36 F. Farouk et al. / Analytica Chimica Acta 890 (2015) 21e43

Fig. 9. A. Chromatogram of a spiked milk sample spiked with streptomycin and dihydrostreptomycin at half MRL (100 mg kg1) and analyzed by IPLC method. Reprinted from Ref.
[35] with permission. B: Chromatogram of a spiked milk sample fortified with streptomycin and dihydrostreptomycin at half MRL (100 mg kg1) and analyzed by HILIC method.
Reprinted from Ref. [35] with permission.

samples was performed using HILICeMS (single quadrupole) and a acid in the ratio 96:3.6:0.2 (v/v/v)) and solvent B (methanol) was
Halo unbound anionic silica HILIC column. The mobile phase was applied impurity profiling of vertilmicin [101] and of micronomicin
75% acetonitrile and 100e200 mM aqueous ammonium formate [102]. Using ESI-MS detection in positive-ion mode, 18 impurities
buffer (pH 3.0e4.5) in acetonitrile. According to the authors, better were detected for vertilmicin [101] and 36 impurities for micro-
peak shape was achieved at higher ammonium formate concen- nomicin [102]. The detectability of the impurities was enhanced by
trations. The pH was adjusted to pH 4.5 to reduce decomposition of injecting a high sample concentration (5 mg mL1 [102]) into the
silica column [124]. system.
An Inertsil HILIC column (3.5 mm) was used for the determina-
tion of tobramycin in human plasma. The mobile phase was
6.5. Comparison between IPLCeMS and HILICeMS
acetonitrilee5 mM ammonium acetate and 0.1% formic acid (60:40,
v/v) at 0.30 mL min1. Sisomicin was used as the IS. The method
Using streptomycin and dihydrostreptomycin in milk as
was linear in range of 0.51e1051 ng mL1 [53].
example, the performance of IPLC and HILIC was compared [35].
Both techniques were used with their typical experimental condi-
6.4. High-pH RPLCeMS tions. Thus, a water/acetonitrile gradient with 10 mM HFBA in both
solvents and a 50  2.1 mm C18 column (5 mm particles) was
High-pH RPLC in combination with ESI-MS in negative-ion applied in IPLC (gradient run time 11.1 min; total run time 15 min),
mode was applied for the analysis of tobramycin in pharmaceu- whereas isocratic elution with 65% acetonitrile in 150 mM aqueous
tical formulations [104]. A mobile phase gradient of solvent A ammonium acetate (pH 4.5) mobile phase and a 100  3 mm
(0.7 mL of 28e30% ammonia solution added to water; pH 11; anionic silica column (3 mm particles) was used in HILIC (Fig. 9).
adjusted with 2.5 N sodium hydroxide) and solvent B (100% Quantification was performed in selected-ion monitoring (SIM)
acetonitrile) at a flow rate of 0.2 mL min1. A Waters Xterra RP18 mode with m/z of [M þ H]þ on a single-quadrupole instrument. The
was used as a stationary phase. The LOD was 0.063 mg mL1. ESI capillary voltage was 3.0 kV in IPLC and 3.5 kV in HILIC. For
Similarly, a Gemini NX C18 column with a mobile phase gradient qualitative purposes, the occurrence of characteristic fragment
of solvent A (water, ammonia solution (25%, m/m) and glacial acetic ions, i.e., ions with m/z 320, 263, and 176 for streptomycin and with
Table 4
Selected chromatographic methods for AG determination.

Method Recommendation AG Column Mobile phase Detection Matrix LOD/LOQ Reference

Ion pairing In separation of Neomycin and RP-Polaris C18 170 mM of aqueous ELSD Pharmaceutical 0.2% [20]
aminoglycosides and framycetin and related (150  4.6 mm i.d., 3 mm) TFA preparation
their known impurities impurities
MS/MS can be Tobramycin impurities Zorbax SB-C18 H2Oeacetonitrile ELSD Pharmaceutical LOD: 0.75 mg mL1 [65]
hyphenated for (250  4.6 mm, 5 mm) eHFBA (96:4:1, v/v/v) preparations
identification and and B: H2O
characterization of eacetonitrileeHFBA
separated unknown (60:40:1, v/v/v).
impurities Spectinomycin and its C18 Hypersil® Gold 0.3% TFA in water CAD Pharmaceutical LOD: 0.5 mg mL1 [73]
impurities (150 mm  4.6 mm; 3 mm) eacetonitrile in ration preparation LOQ: 2.0 mg mL1
98:2
Neomycin and its A Hypersil® GOLD aQ C18 Wateremethanol CAD Pharmaceutical LOD: 3 mg/mL1 [74]
impurities column (150  4.0 mm; (40:60, v/v) with preparations
3 mm) 1.7 mL. L1 HFBA
RPLC Pre-column In absence of other Amikacin RP-C8 Watereacetonitrile Flourimetric Plasma LOD: 90 ng mL1 [84]

F. Farouk et al. / Analytica Chimica Acta 890 (2015) 21e43


derivatization types of universal (35:65, v/v) Reagent: FMOCL
detector Isepamicin Agilent XDB-C8 (particle Water and acetonitrile Flourimetric Plasma 0.10 mg mL1 [45]
size 5 mm, (68:32, v/v) Reagent: FMOCL
150 mm  4.6 mm)
Tobramycin A Purospher® STAR RP- Watereacetonitrile UV Plasma 0.23 mg L1 [51]
18e column (55  4 mm; (50:50, v/v) NITC
3 mm, Merck)
Post column Amikacin, netilmicin, RP Synergi Hydro-RP 0.1% trifluoroacetic Spectrophotometric: Plasma and urine LOD: [49]
derivatization and etimicin column acid (pH 2.2) resonance Rayleigh 0.018, 0.021 0.055
(150 mm  4.6 mm, 4 mm scattering (mg mL1)
Congo red
Amikacin Phenomenex C18 Synergi 102 mol L1 aqueous luminescence Plasma and urine LOD: 50 mg L1 [90]
RP 80A 250 mm  4.6 mm potassium hydrogen detector:
phthalate (pH 3.35)- quenching of Cu2þ-
acetonitrile (90:10, v/ luminol
v). chemiluminescence
High pH In coupling with Tobramycin Xterra® RP18 A: 0.7 mL of 28e30% MS detector in Pharmaceutical LOD: 0.063 mg mL1 [104]
detector system 2.1 mm  250 mm, 5 mm ammonia solution negative ionization preparation
require alkalization of added to water (pH 11; mode
column effluent e.g. adjusted with 2.5 N
PAD sodium hydroxide)
Solvent B was 100%
acetonitrile.
In gradient
Tobramycin Purospher RP-8e column 0.05 M diammonium UV at 210 Pharmaceutical LOQ: 0.47 mg mL1 [94]
(250 mm  4.6 mm, 5 mm) hydrogen phosphate, preparation
pH (10.0; adjusted
with using tetramethyl
ammonium
hydroxide)
Impurity profiling of Gemini NX C18 column A: water, ammonia MS detector Bulk e [101]
vertilmicin. (250 mm  4.6 mm, 5 mm) solution (25%, m/m)
and glacial acetic acid
in the ratio 96:3.6:0.2
(v/v/v). Mobile phase
B: methanol
pH ¼ 11
(continued on next page)

37
Table 4 (continued )

38
Method Recommendation AG Column Mobile phase Detection Matrix LOD/LOQ Reference

Plazomicin and its XBridge C18, A: Water-28 -30% MS detection Bulk e [61]
impurities 4.6 mm  150 mm, 3.5 mm Ammonia (98.33:1.67,
v/v)
B: Acetonitrile e 28
e30% Ammonia (98.33
e1.67, v/v)
LCeMS IPC-MS For separation of AG as Kanamycin and Betasil phenyl hexyl A: 1 mM HFBA þ 0.5% MS/MS Multi-residual analysis LOQ: [36]
well as separation neomycin, 50 mm  2.1 mm, 3 mm formic acid in water of 32 AB in chicken 25;
identification of B: 0.5% FA in meat 120 mg kg1
known and unknown acetonitrileemethanol
impurities of AG (1:1, v/v).
10 AG X-Terra MS C18 A: water; B: methanol, MS/MS Multiresidual analysis LOQ: 27 [34]
2.1  100 mm, 3.5 mm both containing 0.1 M in poultry, swine, e688 mg kg1
HFBA equine, and bovine
kidney
Streptomycin and its YMC-Pack Pro column 20 mmol L1 of PFPA in TOF-MS Bulk LOQ: 4.5 mg mL1 [114]
impurities (250 mm  4.6 mm; water/acetone (99/1, v/ CAD for streptomycin
particle size 3 mm) v) sulfate and

F. Farouk et al. / Analytica Chimica Acta 890 (2015) 21e43


0.4 mg mL1 for
streptidine sulfate
Streptomycin Luna C18 (2) 100A column A: acetonitrile B: MS/MS Multiresidual analysis LOQ: [138]
Dihydrostreptomycin (50  3.0 mm, 3 mm 0.025% HFBA in water 1
neomycin 2
10 ug.L1
HILICeMS In assay that require 10 aminoglycosides Zwitter ionic (ZIC®eHILIC, A: 0.2% formic acid in MS/MS Honey samples LOD: 3e46 mg mL1 [120]
high sensitivity e.g. 2.1  150 mm, 3.5 mm, acetonitrile
water analysis, B: ammonium formate
therapeutic drug (175 mM) at pH 4.5.
monitoring Tobramycin Inertsil HILIC column Acetonitrilee5 mM MS/MS Plasma LOQ: 0.51 ng mL1 [53]
(150 mm  2.1 mm, ammonium acetate
3.5 mm and 0.1% formic acid
(60:40, v/v)
Neomycin ZIC-HILIC column Acetonitrile, 2 mM MS/MS Plasma LOQ: 100 ng ml1 [122]
(100 mm  2.1 mm) ammonium acetate
and formic acid.
Solvent A was (5/95/
0.2, v/v/v) and solvent
B was (95/5/0.2, v/v/v)
Amikacin Atlantis HILIC column A ¼ 0.1% formic acid MS/MS Plasma LOQ: 1 mg mL1 [139]
Kanamycin (150  2.1 mm, 3 mm; solution
Sterptomycin Waters, Milford, MA, USA) B: acetonitrile
containing 0.1% formic
acid (B).
F. Farouk et al. / Analytica Chimica Acta 890 (2015) 21e43 39

m/z 524, 163, and 175 for dihydrostreptomycin, was checked using Magnetic iron nanoparticles (MNP) were used for the detection
in-source CID. The overall process efficiencies were 69.3% and 56.5% of kanamycin A in milk samples. To this end, the MNP or gold-core
in IPLC and 85.5% and 72.3% in HILIC, for streptomycin and dihy- silver shell (AuAg) nanoparticles were functionalized with anti-
drostreptomycin, respectively. The intra-day and inter-day preci- kanamycin antibodies. Upon reaction with kanamycin A, a sand-
sion, studied at 100, 200 and 300 mg kg1 levels in milk, gave %RSD wich kanamycin complex with both types of nanoparticles is
13 for both methods. LOQs were 109 and 31 mg kg1 in IPLC, for formed, which can be detected by surface-enhanced Raman scat-
streptomycin and dihydrostreptomycin, respectively, and tering. The method showed linearity over the range
14 mg kg1 for both analytes in HILIC. The sensitivity of the HILIC 2 pg mL1e80 ng mL1 for the determination of kanamycin in milk
method is greater than that of the IPLC method, for streptomycin and the LOD was 2 pg mL1 [136].
and dihydrostreptomycin, respectively [35]. AuNPs were also used for sensing AG presence in milk. The
Fig. 4 shows different chromatographic techniques that are model AGs used were kanamycin monosulfate, neomycin sulfate,
available for the analysis of AGs depending on the required analytic streptomycin sulfate and bleomycin sulfate. The method depended
information. AGs can be separated by IPLC, HILIC or RPLC after pre- on synthesis of gold nanoparticles stabilized by pyrocatechol violet.
column or post-column derivatization or at high pH. In absence of AGs, these particles are monodisperse and have a red
HILIC is better compatible to MS detectors than IPLC. This can be color. However, in presence of AGs, the hydroxy and amine groups
attributed to the ionization suppressing effect of the IPR which is of the AGs form hydrogen bonding with pyrocatechol violet,
used in IPLC. In applying HILIC, ZIC columns were generally resulting in cross linking and aggregation of the AuNPs. The
preferred over anionic and cationic HILIC columns [125]. aggregated AuNPs show a blue color. At very high AG concentration
Although the IPR is chosen to be volatile to suit the MS detector, of AGs, neither red nor blue color will be observed due to inter-
their charge may suppress the ionization of the analyte and thus ference in the synthesis of AuNP [137].
reduce the detection efficiency.
IPLC is superior over HILIC in its separation power and its wider 8. Conclusions and perspectives
column availability. Thus, if sensitivity is the concern, the HILIC is
the method of choice while in impurity detection or elucidation We propose a workflow for the analysis of AGs in different
IPLC is preferred. matrices according to the type of matrix, required level of infor-
High-pH RPLCeMS/MS so far has only be described for impurity mation and facilities (Fig. 4) aided by some examples in Table 4. In
profiling, thus no comparison in terms of quantitative performance the sample preparation step, the methodology of extraction will be
for residue analysis and TDM in plasma or serum can yet be made. largely affected by the suspected amount of AGs in the sample and
Recently, the use of a CarboPac® PA1 anion-exchange column was the available detection techniques. In case of a sample containing
reported for the determination of streptomycin and dihydros- protein, e.g., food, serum and plasma samples, protein precipitation
treptomycin in veterinary dosage forms. The mobile phase was is an initial step. After protein precipitation, sample clean-up to
0.07 N NaOH at a flow rate of 0.8 mL min1. The detection was per- remove fats (if any) by for example hexane liquideliquid extraction
formed by PAD with a multi-step potential waveforms to cope with should be performed. SPE can be used to remove salts from the
the tendency of AG to adsorb on the solid electrodes. The linearity sample as they might affect ionization in the MS detector. SPE can
range of the assay was 1e50 mg mL1 using glucose as IS [126]. also be used for other cleanup and pre-concentration purposes,
Another recent development involves the use of contactless depending to the nature of the sample.
conductivity detection (CD) in combination with IPLC for the The choice of the chromatographic method, IPLC, HILIC, or RPLC
determination of AGs. At present, the method seems to be limited after derivatization, depends on the level of information required
in the choice of the IPR and especially the concentration of the IPR. along with the availability of equipment and resources.
As a result, the IPLC is compromised and no adequate LODs could be HILIC provides retention of AGs without IPR to the mobile phase.
achieved [127]. In MS detection, HILIC can be used to achieve lower detection limits
(compared to IPLC or RPLC). However, IPLC provides better sepa-
7. Non-chromatographic methods ration efficiency. The IPR required must be volatile in case of MS
detection and may still compromise sensitivity due to ionization
Several non-chromatographic methods were recently described suppression. Thus, in food safety analysis, HILIC is our proposed
for the determination of AGs, including immunoassays [128e131], choice. For the detection of AG related substances, IPLC is recom-
microbiological assays [132], and fluorometric assays [43,48]. These mended. Alternatively, RPLC with pre-column analyte derivatiza-
methods exhibit comparable sensitivity to the LC methods and may tion can be applied, especially if no MS facilities are available. For
thus be further investigated as alternative to the mostly costly LC impurity profiling, good results have also been achieved using
and LCeMS methods. high-pH RPLC.
In addition to this, more advanced non-chromatographic The detector choice is largely affected by the availability and
methods have been reported in which nanoparticles are applied. financial aspects, especially in developing countries. As AGs lack a
A method using gold nanoparticles (AuNPs) carrying a dye-labeled chromophore, the widespread UV detection system is not the
fluorescent aptamer specially designed for kanamycin A was method of choice, unless analyte derivatization is performed. MS
recently developed. In the absence of kanamycin A, the aptamer is detection is the state of the art technology and is our recommen-
adsorbed on the AuNPs and the fluorescence of the aptamer is dation for AG determination. It can be coupled to all three types of
quenched. The aptamer shows higher affinity to kanamycin A and is LC mentioned, as long as volatile mobile-phase additives are
released from the AuNP, resulting in enhanced fluorescence. This applied. PAD and ELSD can be considered as alternative approaches.
process is concentration dependent. The method was applied for
the determination of kanamycin A in milk samples [133]. Another References
group reported a similar AuNP-based colorimetric method for the
detection of streptomycin in milk [134]. A recently reported [1] D. Greenwood, Antimicrobial Chemotherapy, fifth ed., Oxford University
biosensor for the detection of streptomycin and other classes of ABs Press, New York, 2007.
[2] E.M. Scholar, W.B. Pratt, The Antimicrobial Drugs, second ed., Oxford Uni-
is based on a multicolor quantum dot based immunofluorescence versity press, New York, 2000.
assay [135]. [3] W. Piepersberg, K. Aboushanab, H. Schmidt-Biebner, U.F. Wehmeier, The
40 F. Farouk et al. / Analytica Chimica Acta 890 (2015) 21e43

biochemistry and genetics of aminoglycosides producers, in: D.P. Arya (Ed.), [33] R. Ishii, M. Horie, W. Chan, J. MacNeil, Multi-residue quantitation of ami-
Aminoglycoside Antibiotics from Chemical Biology to Drug Discovery, Wiley, noglycoside antibiotics in kidney and meat by liquid chromatography with
New Jersey, 2007, pp. 15e119. tandem mass spectrometry, Food Addit. Contam. Part A Chem. Anal. Control
[4] G.F. Busscher, F.P.J.T. Rutjes, F.L. van Delft, 2-Deoxystreptamine: central Expo. Risk Assess. 25 (2008) 1509e1519.
scaffold of aminoglycoside antibiotics, Chem. Rev. 105 (2005) 775e779. [34] M.P. Almeida, C.P. Rezend, L.F. Souza, R.B. Brito, Validation of a quantitative
[5] Y. Zhu, T.A. Johnson, J. Su, M. Qiao, G. Guo, R.D. Stedtfeld, S.A. Hashsham, and confirmatory method for residue analysis of aminoglycoside antibiotics
J.M. Tiedje, Diverse and abundant antibiotic resistance genes in Chinese in poultry, bovine, equine and swine kidney through liquid chromatography-
swine farms, Proc. Natl. Acad. Sci. U. S. A. 110 (2013) 3435e3440. tandem mass spectrometry, Food Addit. Contam. Part A Chem. Anal. Control
[6] P. Hughes, J. Heritage, Antibiotic Growth-Promoters in Food Animals, http:// Expo. Risk Assess. 29 (2012) 517e525.
www.fao.org/DOCREP/ARTICLE/AGRIPPA/555_EN.HTM, (accessed 01.03.15). [35] A.M. Gremilogianni, N.C. Megoulas, M.A. Koupparis, Hydrophilic interaction
[7] http://www.nytimes.com/2015/03/05/business/mcdonalds-moving-to- vs ion pair liquid chromatography for the determination of streptomycin and
antibiotic-free-chicken.html, (accessed 31.05.15). dihydrostreptomycin residues in milk based on mass spectrometric detec-
[8] J.K. Aronson, D.J. Reynolds, ABC of monitoring drug therapy, aminoglycoside tion, J. Chromatogr. A 1217 (2010) 6646e6651.
antibiotics, BMJ 305 (1992) 1421e1424. [36] K. Bousova, H.Z. Senyuva, K. Mittendorf, Quantitative multi-residue method
[9] T.E. Young, Aminoglycoside therapy in neonates with particular reference to for determination antibiotics in chicken meat using turbulent flow chro-
gentamicin, NeoReviews 3 (2002) 243e248. matography coupled to liquid chromatography-tandem mass spectrometry,
[10] E. Selimoglu, Aminoglycoside-induced ototoxicity, Curr. Pharm. Des. 3 (2007) J. Chromatogr. A 1274 (2013) 19e27.
119e126. [37] Aminoglycosides in Bovine Muscle Using Agilent Bond Elut Plexa
[11] http://www.codexalimentarius.org/standards/list-of-standards/, (accessed SPE and Agilent Poroshell 120 Column and LC/Tandem MS, Application
01.03.15). Note: 5991-1321EN (2012). http://www.chem.agilent.com/search/?
[12] G.A. Osman, H.M. Hassan, M.M. Kamel, Resistance and sensitivity of some Ntt¼Aminoglycosides%20in%20bovine%20muscle%20using%20agilent%
bacterial strains isolated from hospital wastewater and Nile water using 20bond%20elut%20plexa%20SPE%20and%20agilent%20poroshell%20120%
chlorination and some antibiotics in Cairo (Egypt), J. Am. Sci. 7( (2011) 20column%20and%20LC%2Ftendam%20MS, (accessed 01.03.15).
1033e1041. [38] D.A. Bohm, C.S. Stachel, P. Gowik, Confirmatory method for the determination
[13] Report, Only Half of Prescription Drugs Removed from Sewage by of streptomycin and dihydrostreptomycin in honey by LC-MS/MS, Food Addit.
Treatment Plants, http://www.drugfree.org/join-together/report-only-half- Contam. Part A Chem. Anal. Control Expo. Risk Assess. 29 (2012) 189e196.
of-prescription-drugs-removed-from-sewage-by-treatment-plants/, [39] D.A. Bohm, C.S. Stachel, P. Gowik, Confirmatory method for the determination of
(accessed 01.03.15). streptomycin in apples by LCeMS/MS, Anal. Chim. Acta 672 (2010) 103e106.
[14] D. Lo€ ffler, T.A. Ternes, Analytical method for the determination of the ami- [40] M. Gbylik, A. Posyniak, K. Mitrowska, T. Bladeka, J. Zmudzki, Multi-residue
noglycoside gentamicin in hospital wastewater via liquid chromatography- determination of antibiotics in fish by liquid chromatography-tandem mass
eelectrospray-tandem mass spectrometry, J. Chromatogr. A 1000 (2003) spectrometry, Food Addit. Contam. Part A Chem. Anal. Control Expo. Risk
583e588. Assess. 30 (2013) 940e948.
[15] W. Haasnoot, P. Stouten, G. Cazemier, A. Lommen, J.F.M. Nouws, H.J. Keukens, [41] P.A. Martos, F. Jayasundara, J. Dolbeer, W. Jin, L. Spilsbury, M. Mitchell,
Immunochemical detection of aminoglycosides in milk and kidney, Analyst C. Varilla, B. Shurmer, Multiclass, multiresidue drug analysis including ami-
124 (1999) 301e305. noglycosides in animal tissue using liquid chromatography coupled to tan-
[16] S.M. Sunaric, S.S. Mitic, G.Z. Miletic, D.A. Kostic, N.V. Trutic, Spectrophoto- dem mass spectrometry, J. Agric. Food Chem. 58 (2010) 5932e5944.
metric initial-rate method for determination of neomycin in ophthalmic [42] S. Ji, F. Zhang, X. Luo, B. Yang, G. Jin, J. Yan, X. Liang, Synthesis of molecularly
drops, Chem. Anal. 52 (2007) 810e821. imprinted polymer sorbents and application for the determination of ami-
[17] M.R.C. Marques, E.R.M. Hackmann, T. Saito, Extraction and spectrophoto- noglycosides antibiotics in honey, J. Chromatogr. A 1313 (2013) 113e118.
metric determination of neomycin sulphate in pharmaceutical oral suspen- [43] M. Omar, D. Nagy, M. Hammad, A. Aly, Highly sensitive spectrofluorimetric
sions, Anal. Lett. 23 (1990) 1005e1016. method for determination of certain aminoglycosides in pharmaceutical
[18] M.R.C. Marques, E.R.M. Hackmann, T. Saito, Spectrophotometric determina- formulations and human plasma, AAPS Pharm. Sci. Tech. 14 (2013) 828e837.
tion of neomycin with 2, 4-dinitrofluorobenzene, Anal. Lett. 22 (1989) [44] A. Shen, L. Morgan, M. Barroso, X. Zhang, Method Development of LC-MS/MS
621e633. Analysis of Aminoglycoside Drugs: Challenges and Solutions, 2008. http://
[19] M. Confino, P. Bontchev, Spectrophotometric determination of amikacin, www.tandemlabs.com/documents/ASMS08-Angela-Web.pdf (accessed
kanamycin, neomycin and tobramycin, Mikrochim. Acta 102 (1990) 15.05.15).
305e309. [45] X. Chang, J. Peng, LC analysis of isepamicin in plasma samples post-
[20] I. Clarot, A. Regazzeti, N. Auzeil, F. Laadani, M. Citton, P. Netter, A. Nicolas, inhalation with fluorescence detection and its application to a pharmaco-
Analysis of neomycin sulfate and framycetin sulfate by high-performance kinetic study, Chromatographia 70 (2009) 1429e1433.
liquid chromatography using evaporative light scattering detection, [46] S. Chen, Y. Liang, Y. Chou, Analysis of kanamycin A in human plasma and in
J. Chromatogr. A 1087 (2005) 236e244. oral dosage form by derivatization with 1-naphthyl isothiocyanate and high-
[21] C.H. Yamamoto, T.J.A. Pinto, Rapid determination of neomycin by a micro- performance liquid chromatography, J. Sep. Sci. 29 (2006) 607e612.
biological agar diffusion assay using triphenyltetrazolium chloride, J. AOAC [47] M. Jonge, J. Bekkers, H.M. Straaten, R.W. Sparidans, E. Franssen, Simple and
Int. 79 (1996) 434e440. sensitive method for quantification of low tobramycin concentrations in
[22] Monographs, Pharmaceutical Substances: Neomycini Sulfas e Neomycin human plasma using HPLCeMS/MS, J. Chromatogr. B 862 (2008) 257e262.
Sulfate, International Pharmacopoeia, http://apps.who.int/phint/en/p/docf/, €
[48] S. Tekkeli, A. Onal, A. Sagırlı, Spectrofluorimetric determination of tobra-
(accessed 01.03.15). mycin in human serum and pharmaceutical preparations by derivatization
[23] http://idmp.ucsf.edu/aminoglycoside-dosing-and-monitoring- with fluorescamin, Luminescence 29 (2014) 87e91.
recommendations, (accessed 15.05). [49] L. Zhang, J. Peng, J. Tang, B. Yuan, R. He, Y. Xiao, Description and validation of
[24] http://www.uptodate.com/contents/dosing-and-administration-of- coupling high performance liquid chromatography with resonance Rayleigh
parenteral-aminoglycosides, (accessed 15.05). scattering in aminoglycosides determination, Anal. Chim. Acta 706 (2011)
[25] M. Avent, B. Rogers, A. Cheng, D. Paterson, Current use of aminoglycosides: 199e204.
indications, pharmacokinetics and monitoring for toxicity, Intern. Med. J. 41 [50] M. Omar, M. Hammad, D. Nagy, A. Aly, Development of spectrofluorimetric
(2011) 441e449. method for determination of certain aminoglycoside drugs in dosage forms
[26] R. Hatala, T. Dinh, D.J. Cook, Once-daily aminoglycoside dosing in immuno- and human plasma through condensation with ninhydrin and phenyl acet-
competent adults: a meta-analysis, Ann. Intern. Med. 124 (1996) 717e725. aldehyde, Spectrochim. Acta A Mol. Biomol. Spectrosc. 136 (2014)
[27] E. van Maarseveen, W. Man, J. Proost, C. Neef, D. Touw, Chro- 1760e1766.
nopharmacokinetics of once daily dosed aminoglycosides in hospitalized [51] C. Feng, S. Lin, H. Wu, S. Chen, Trace analysis of tobramycin in human plasma
infectious patients, Int. J. Clin. Pharm. 37 (2015) 342e347. by derivatization and high-performance liquid chromatography with ultra-
[28] http://www.ashp.org/DocLibrary/MemberCenter/NPF/2011Pearls/Antibiotic- violet detection, J. Chromatogr. B 780 (2002) 349e354.
Pharmacokinetic-Monitoring.aspx, (accessed 15.05). [52] X. Chang, Z. Yu, Determination of etimicin in rat plasma using 9-
[29] http://www.sahealth.sa.gov.au/wps/wcm/connect/ fluorenylmethyl chloroformate precolumn derivatization by HPLC with
e4c8cb004877c5c3a295f67675638bd8/Guideline_Aminoglycosides% fluorescence detection, J. Braz. Chem. Soc. 20 (2011) 1246e1252.
2Brecommendations%2Bfor%2Buse_Sept11.pdf?MOD¼AJPERES, (accessed [53] L. Chen, H. Chen, M. Shen, Hydrophilic interaction chromatography com-
15.05). bined with tandem mass spectrometry method for the quantification of
[30] http://www.ruh.nhs.uk/For_Clinicians/departments_ruh/Pathology/ tobramycin in human plasma and its application in a pharmacokinetic study,
documents/haematology/Dosing_of_Gentamicin_Vancomycin_and_ J. Chromatogr. B 973 (2014) 39e44.
Teicoplanin.pdf, (accessed 15.05). [54] C. Cheng, S. Liu, D. Xiao, S. Hansel, The application of trichloroacetic acid as
[31] G. Wong, F.B. Sime, J. Lipman, J.A. Robert, How do we use therapeutic drug an ion pairing reagent in LCeMSeMS method development for highly polar
monitoring to improve outcomes from severe infections in critically ill pa- aminoglycoside compounds, Chromatographia 72 (2010) 133e139.
tients? BMC Infect. Dis. 14 (2014) 288. [55] D. Heller, J. Peggins, C. Nochetto, M. Smith, O. Chiesa, K. Moulton, LC/MS/MS
[32] D.A. Bohm, C.S. Stachel, P. Gowik, Validation of a method for the determi- measurement of gentamicin in bovine plasma, urine, milk, and biopsy samples
nation of aminoglycosides in different matrices and species based on an in- taken from kidneys of standing animals, J. Chromatogr. B 821 (2005) 22e30.
house concept, Food Addit. Contam. Part A Chem. Anal. Control Expo. Risk [56] B. Keevil, S. Lockhart, D. Cooper, Determination of tobramycin in serum using
Assess. 30 (2013) 1037e1043. liquid chromatographyetandem mass spectrometry and comparison with a
F. Farouk et al. / Analytica Chimica Acta 890 (2015) 21e43 41

fluorescence polarisation assay, J. Chromatogr. B 794 (2003) 29e35. electrophoresis method with contactless conductivity detection for the
[57] Y. Bijleveld, T. de Haan, J. Toersche, S. Jorjani, J. van der Lee, F. Groenendaal, analysis of tobramycin and its related substances, J. Pharm. Biomed. Anal. 58
P. Dijk, A. van Heijst, A. Gavilanes, R. de Jonge, K. Dijkman, H. van Straaten, (2012) 49e57.
M. Rijken, I. Zonnenberg, F. Cools, D. Nuytemans, R. Matho ^ t, A simple [79] S. Ge, W. Tang, R. Hana, Y. Zhu, Q. Wang, P. He, Y. Fang, Sensitive analysis of
quantitative method analysing amikacin, gentamicin, and vancomycin levels aminoglycoside antibiotics via hyphenation of transient moving substitution
in human newborn plasma using ion-pair liquid chromatography/tandem boundary with field-enhanced sample injection in capillary electrophoresis,
mass spectrometry and its applicability to a clinical study, J. Chromatogr. B J. Chromatogr. A 1295 (2013) 128e135.
951e952 (2014) 110e118. [80] J. Krzek, M. Starek, A. Kwiecien  , W. Rzeszutko, Simultaneous identifica-
[58] C.-Y. Lu, C.-H. Feng, Micro-scale analysis of aminoglycoside antibiotics in tion and quantitative determination of neomycin sulfate, polymixin B
human plasma by capillary liquid chromatography and nanospray tandem sulfate, zinc bacytracin and methyl and propyl hydroxybenzoate in
mass spectrometry with column switching, J. Chromatogr. A 1156 (2007) ophthalmic ointment by TLC, J. Pharm. Biomed. Anal. 24 (2001)
249e253. 629e636.
[59] A.K. Sarri, N.C. Megoulas, M.A. Koupparis, Development of a novel method [81] U. Hubicka, B. Zuromska-Witek, J. Piotrowska, J. Krzek, Determination of
based on liquid chromatographyeevaporative light scattering detection for neomycin in the form of neomycin derivative with dabsyl chloride by thin
the direct determination of streptomycin and dihydrostreptomycin in raw layer chromatography and densitometry, Acta. Pol. Pharm. 72 (2015)
materials, pharmaceutical formulations, culture media and plasma, 31e37.
J. Chromatogr. A 1122 (2006) 275e278. [82] E. Roets, E. Adams, I.G. Muriithi, J. Hoogmartens, Determination of the rela-
[60] R. Oertel, V. Neumeister, W. Kirch, Hydrophilic interaction chromatography tive amounts of the B and C components of neomycin by thin-layer chro-
combined with tandem-mass spectrometry to determine six aminoglyco- matography using fluorescence detection, J. Chromatogr. A 696 (1995)
sides in serum, J. Chromatogr. A 1058 (2004) 197e201. 131e138.
[61] L. Tan, K. Wlasichuk, D. Schmidt Jr., R. Campbell, P. Hirtzer, L. Cheng, D. Karr, [83] Y. Chen, W. Schwack, High-performance thin-layer chromatography
A high pH based reversed-phase high performance liquid chromatographic screening of multi class antibiotics in animal food by bioluminescent bio-
method for the analysis of aminoglycoside plazomicin and its impurities, autography and electrospray ionization mass spectrometry, J. Chromatogr. A
J. Pharm. Biomed. Anal. 66 (2012) 75e84. 1356 (2014) 249e257.
[62] T.J. Whall, Determination of streptomycin sulfate and dihydrostreptomycin [84] X. Chang, J. Peng, S. Liu, A simple and rapid high performance liquid chro-
sulfate by high performance liquid chromatography, J. Chromatogr. A 219 matographic method with fluorescence detection for the estimation of
(1981) 89e100. amikacin in plasma e application to preclinical pharmacokinetics, J. Chin.
[63] J. Wang, S.B. Turnipseed, Chemical analysis: quantitative and confirmatory Chem. Soc. 57 (2010) 34e39.
methods, in: J. Wang, J. D. MacNeil and J. F. Kay (Eds.), Chemical Analysis of [85] D. Stead, R. Richards, Sensitive high-performance liquid chromatographic
Antibiotic Residues in Food, Wiley, New Jersey, 196e197. assay for aminoglycosides in biological matrices enables the direct estima-
[64] N.C. Megoulas, M.A. Koupparis, Enhancement of evaporative light scattering tion of bacterial drug uptake, J. Chromatogr. B 693 (1997) 415e421.
detection in high-performance liquid chromatographic determination of [86] V.K. Agarwal, High performance liquid chromatographic determination of
neomycin based on highly volatile mobile phase, high-molecular-mass ion- neomycin in milk using a Hisep column, J. Liq. Chromatogr. 13 (1990)
pairing reagents and controlled peak shape, J. Chromatogr. A 1057 (2004) 2475e2487.
125e131. [87] R. Tawa, S. Hirose, T. Fujimoto, Determination of the aminoglycoside anti-
[65] C. Pfeifer, G. Fassauer, H. Gerecke, T. Jira, Y. Remane, R. Frontini, biotics sisomicin and netilmicin in dried blood spots on filter discs, by high-
Jonathan Byrne, Robert Reinhardt, Purity determination of amphotericin B, performance liquid chromatography with pre-column derivatization and
colistin sulfate and tobramycin sulfate in a hydrophilic suspension by HPLC, fluorimetric detection, J. Chromatogr. B 490 (1989) 125e132.
J. Chromatogr. B 990 (2015) 7e14. [88] F. Wienen, U. Holzgrabe, Characterization of paromomycin sulfate by capil-
[66] Y. Zhang, H. He, J. Zhang, F. Liu, C. Li, B. Wang, R. Qiao, HPLC-ELSD deter- lary electrophoresis with UV detection after pre-capillary derivatization,
mination of kanamycin B in the presence of kanamycin A in fermentation Chromatographia 55 (2001) 327e331.
broth, Biomed. Chromatogr. 29 (2015) 396e401. [89] J.M. Serrano, M. Silva, Trace analysis of aminoglycoside antibiotics in bovine
[67] Y. Liu, X. Zhang, S. An, Y. Wu, G. Hu, Y. Wu, Pharmacokinetics of neamine in milk by MEKC with LIF detection, Electrophoresis 27 (2006) 4703e4710.
rats and anti-cervical cancer activity in vitro and in vivo, Cancer. Chemother. [90] J.M. Serrano, M. Silva, Determination of amikacin in body fluid by high-
Pharmacol. 75 (2015) 465e474. performance liquid-chromatography with chemiluminescence detection,
[68] N.H. Zawilla, B. Lia, J. Hoogmartens, E. Adams, Improved reversed-phase J. Chromatogr. B 843 (2006) 20e24.
liquid chromatographic method combined with pulsed electrochemical [91] L.L. Olson, J. Pick, W.Y. Ellis, P. Lim, A chemical assessment and HPLC assay
detection for the analysis of amikacin, J. Pharm. Biomed. Anal. 43 (2007) validation of bulk paromomycin sulfate, J. Pharm. Biomed. Anal. 15 (1997)
68e173. 783e793.
[69] E. Adams, R. Schepers, E. Roets, J. Hoogmartens, Determination of neomycin [92] J. Driver, N. Thiex, D. Raynie, M. Ofitserova, M. Pickering, Single-laboratory
sulfate by liquid chromatography with pulsed electrochemical detection, validation for the quantification of neomycin B and neomycin C in animal
J. Chromatogr. A 741 (1996) 233e240. feed by liquid chromatography, fluorescence detection with post column
[70] M. Pendela, E. Adams, J. Hoogmartens, Development of a liquid chromato- derivatization, J. AOAC Int. 92 (2009) 34e41.
graphic method for ear drops containing neomycin sulphate, polymyxin B [93] G. Pang, J. Zhang, Y. Cao, C. Fan, X. Lin, Z. Li, G. Jia, Evaluation of analyte
sulphate and dexamethasone sodium phosphate, J. Pharm. Biomed. Anal. 36 stability and method ruggedness in the determination of streptomycin res-
(2004) 751e757. idue in honey by liquid chromatography with post-column derivatization,
[71] A. Brugue s, A. Campmany, C. Lizandra, L. Bellowa, B. Naveros, Development J. AOAC Int. 87 (2004) 39e44.
of a liquid chromatographic method for the quantification of paromomycin. [94] K. Ruckmani, S. Shaikh, P. Khalil, M.S. Muneera, A simple and rapid high-
Application to in vitro release and ex vivo permeation studies, Spectrochim. performance liquid chromatographic method for determining tobramycin
Acta A Mol. Biomol. Spectrosc. 133 (2014) 657e662. in pharmaceutical formulations by direct UV detection, Pharm. Methods 2
[72] Y. Yuan, S. Chopra, X. Deng, M. Zhang, X. Fan, C. Hu, S. Jin, A. Van Schepdael, (2011) 117e123.
E. Adams, Analysis of micronomicin by liquid chromatography with pulsed [95] T.A. Getek, M.L. Vestal, T.G. Alexander, Analysis of gentamicin sulfate by
electrochemical detection, J. Chromatogr. A 1295 (2013) 90e98. high-performance liquid chromatography combined with thermospray mass
[73] K. Stypulkowska, A. Blazewicz, A. Brudzikowska, M. Waronwa-Grzeskiewicz, spectrometry, J. Chromatogr. A 554 (1991) 191e203.
K. Sarna, Z. Fijalek, Development of high performance liquid chromatography [96] S. Lehotay, K. Mastovska, A. Lightfield, A. Nun ~ ez, T. Dutko, C. Ng, L. Bluhm,
methods with charged aerosol detection for the determination of linco- Rapid analysis of aminoglycoside antibiotics in bovine tissues using dispos-
mycin, spectinomycin and its impurities in pharmaceutical products, able pipette extraction and ultrahigh performance liquid chromatogra-
J. Pharm. Biomed. Anal. 112 (2015) 8e14. phyetandem mass spectrometry, J. Chromatogr. A 1313 (2013) 103e112.
[74] K. Stypulkowska, A. Blazewicz, Z. Fijalek, M. Waronwa-Grzeskiewicz, [97] B. Li, A. Van Schepdael, J. Hoogmartens, E. Adams, Characterization of im-
K. Srebrzynska, Determination of neomycin and related substances in purities in tobramycin by liquid chromatography-mass spectrometry,
pharmaceutical preparations by reversed-phase high performance liquid J. Chromatogr. A 1216 (2009) 3941e3945.
chromatography with mass spectrometry and charged aerosol detection, [98] M. Pendela, J. Hoogmartens, A. Van Schepdael, E. Adams, Characterization of
J. Pharm. Biomed. Anal. 76 (2013) 207e214. dihydrostreptomycin-related substances by liquid chromatography coupled
[75] A.L. Huidobro, A. García, C. Barbas, Rapid analytical procedure for neomycin to ion trap mass spectrometry, Rapid Commun. Mass Spectrom. 23 (2009)
determination in ointments by CE with direct UV detection, J. Pharm. Bio- 1856e1862.
med. Anal. 50 (2009) 1303e1307. [99] B. Li, A. Van Schepdael, J. Hoogmartens, E. Adams, Mass spectrometric
[76] W. Yang, A. Mi, Applications of a copper microparticle-modified carbon fiber characterization of gentamicin components separated by the new European
microdisk array electrode for the simultaneous determination of amino- pharmacopoeia method, J. Pharm. Biomed. Anal. 55 (2011) 78e84.
glycoside antibiotics by capillary electrophoresis, J. Chromatogr. A 905 [100] R. Grahek, L. Zupancic-Kralj, Identification of gentamicin impurities by liquid
(2001) 309e318. chromatography tandem mass spectrometry, J. Pharm. Biomed. Anal. 50
[77] M. El-Attug, E. Adams, A. Van Schepdael, Development and validation of a (2009) 1037e1043.
capillary electrophoresis method with capacitively coupled contactless [101] Y. Yuan, M. Zhang, X. Fan, C. Hu, S. Jin, A. Van Schepdael, J. Hoogmartens,
conductivity detection (CE-C(4) D) for the analysis of amikacin and its related E. Adams, Analysis of impurities in vertilmicin sulfate by liquid chroma-
substances, Electrophoresis 33 (2012) 2777e2782. tography ion-trap mass spectrometry, J. Pharm. Biomed. Anal. 80 (2013)
[78] M. El-Attug, E. Adams, A. Van Schepdael, Optimization of capillary 1e8.
42 F. Farouk et al. / Analytica Chimica Acta 890 (2015) 21e43

[102] Y. Yuan, X. Zhao, M. Zhang, X. Fan, C. Hu, S. Jin, A. Van Schepdael, stationary phase and liquid chromatographyetandem mass spectrometry,
J. Hoogmartens, E. Adams, Impurity profiling of micronomicin sulfate in- Anal. Chim. Acta 882 (2015) 127e139.
jection by liquid chromatographyeion trap mass spectrometry, J. Pharm. [124] S. Kawano, Analysis of impurities in streptomycin and dihydrostreptomycin
Biomed. Anal. 75 (2013) 94e104. by hydrophilic interaction chromatography/electrospray ionization quadru-
[103] L.G. McLaughlin, J.D. Henion, P.J. Kijak, Multi-residue confirmation of ami- pole ion trap/time-of-flight mass spectrometry, Rapid Commun. Mass
noglycoside antibiotics and bovine kidney by ion spray high-performance Spectrom. 23 (2009) 907e914.
liquid chromatography/tandem mass spectrometry, Biol. Mass Spectrom. [125] G. Kahsay, H. Song, A. Van Schepdael, D. Cabooter, E. Adams, Hydrophilic
23 (1994) 417e429. interaction chromatography (HILIC) in the analysis of antibiotics, J. Pharm.
[104] M. Guo, L. Wrisley, E. Maygoo, Measurement of tobramycin by reversed- Biomed. Anal. 87 (2014) 142e154.
phase high-performance liquid chromatography with mass spectrometry [126] M. Martínez-Mejía, S. Rath, Use of experimental design to optimize a
detection, Anal. Chim. Acta 571 (2006) 12e16. triple-potential waveform to develop a method for the determination
[105] W.X. Zhu, J.Z. Yang, W. Wei, Y.F. Liu, S.S. Zhang, Simultaneous determination of streptomycin and dihydrostreptomycin in pharmaceutical veterinary
of 13 aminoglycoside residues in foods of animal origin by liquid dosage forms by HPLC-PAD, J. Pharm. Biomed. Anal. 104 (2015)
chromatography-electrospray ionization tandem mass spectrometry with 81e89.
two consecutive solid-phase extraction steps, J. Chromatogr. A 1207 (2008) [127] P. Jankovics, S. Chopra, M. El-Attug, D. Cabooter, K. Wolfs, B. Nosza l,
29e37. A. Van Schepdael, E. Adams, Exploring the possibilities of capacitively
[106] Y. Tao, D. Chen, H. Yu, L. Huang, Z. Liu, X. Cao, C. Yan, Y. Pan, Z. Liu, Z. Yuan, coupled contactless conductivity detection in combination with liquid
Simultaneous determination of 15 aminoglycoside(s) residues in animal derived chromatography for the analysis of polar compounds using amino-
foods by automated solid-phase extraction and liquid chromatography-tandem glycosides as test case, J. Pharm. Biomed. Anal. 112 (2015) 155e168.
mass spectrometry, Food Chem. 135 (2012) 676e683. [128] A. Losoya-Leal, M. Estevez, S. Martínez-Chapa, L. Lechug, Design of a surface
[107] F.L. van Holthoon, M.L. Essers, P.J. Mulder, S.L. Stead, M. Caldow, plasmon resonance immunoassay for therapeutic drug monitoring of ami-
H.M. Ashwin, M.A. Sharman, Generic method for the quantitative analysis of kacin, Talanta 141 (2015) 253e258.
aminoglycosides (and spectinomycin) in animal tissue using methylated [129] I. Darwish, A. Mahmoud, A. Al-Majed, Generic simple enzyme immunoassay
internal standards and liquid chromatography tandem mass spectrometry, approach to avert small molecule immobilization problems on solid phases:
Anal. Chim. Acta 637 (2009) 135e143. application to the determination of tobramycin in serum, Talanta 72 (2007)
[108] S. Bogialli, R. Curini, A. Di Corcia, A. Lagan a, M. Mele, M. Nazzari, Simple 1322e1328.
confirmatory assay for analyzing residues of aminoglycoside antibiotics in [130] I. Darwish, Development of generic continuous-flow enzyme immunoassay
bovine milk: hot water extraction followed by liquid chromatography- system for analysis of aminoglycosides in serum, J. Pharm. Biomed. Anal. 30
tandem mass spectrometry, J. Chromatogr. A 1067 (2005) 93e100. (2003) 1539e1548.
[109] K.M. Peru, S.L. Kuchta, J.V. Headley, A.J. Cessna, Development of a [131] https://www.beckmancoulter.com/wsrportal/techdocs?docname¼/cis/
hydrophilic interaction chromatographyemass spectrometry assay for 4T052/%25%25/EN_GENTAMICIN.pdf, (accessed 05.15).
spectinomycin and lincomycin in liquid hog manure supernatant and [132] A. Zuluaga, M. Agudelo, C. Rodriguez, O. Vesga, Application of microbiological
run-off from cropland, J. Chromatogr. A 1107 (2006) 152e158. assay to determine pharmaceutical equivalence of generic intravenous an-
[110] M.C. Carson, D.N. Heller, Confirmation of spectinomycin in milk using ion- tibiotics, BMC Clin. Pharm. 9 (2009) 1e11.
pair solid-phase extraction and liquid chromatographyeelectrospray ion [133] J. Chen, Z. Li, J. Ge, R. Yang, L. Zhang, L. Qu, H. Wang, L. Zhang, An aptamer-
trap mass spectrometry, J. Chromatogr. B 718 (1992) 95e102. based signal-on bio-assay for sensitive and selective detection of Kanamycin
[111] R.E. Hornish, J.R. Wiest, Quantitation of spectinomycin residues in bovine A by using gold nanoparticles, Talanta 139 (2015) 226e232.
tissues by ion-exchange high-performance liquid chromatography with [134] A.S. Emrani, N.M. Danesh, P. Lavaee, M. Ramezani, K. Abnous,
post-column derivatization and confirmation by reversed-phase high-per- S.M. Taghdisi, Colorimetric and fluorescence quenching aptasensors for
formance liquid chromatography-atmospheric pressure chemical ionization detection of streptomycin in blood serum and milk based on double-
tandem mass spectrometry, J. Chromatogr. A 812 (1998) 123e133. stranded DNA and gold nanoparticles, Food Chem. 190 (2016)
[112] Y. Wang, M. Wang, J. Li, S. Yao, J. Xue, W. Zou, C. Hu, Characterization of 115e121.
impurities in commercial spectinomycin by liquid chromatography with [135] E. Song, M. Yu, Y. Wang, W. Hu, D. Cheng, M.T. Swihart, Y. Songa, Multi-color
electrospray ionization tandem mass spectrometry, J. Antibiot. 67 (2014) quantum DoT-based fluorescence immunoassay array for simultaneous vi-
511e518. sual detection of multiple antibiotic residues in milk, Biosens. Bioelectron. 72
[113] B. Li, A. Van Schepdael, J. Hoogmartens, E. Adams, Characterization of im- (2015) 320e325.
purities in tobramycin by liquid chromatographyemass spectrometry, [136] A. Zengin, U. Tamer, T. Caykar, Extremely sensitive sandwich assay of
J. Chromatogr. A 1216 (2009) 3941e3945. kanamycin using surface-enhanced Raman scattering of 2-mercaptobenzo-
[114] U. Holzgrabe, C. Nap, N. Kunz, S. Almeling, Identification and control of thiazole labeled gold@silver nanoparticles, Anal. Chim. Acta 817 (2014)
impurities in streptomycin sulfate by high-performance liquid chromatog- 33e41.
raphy coupled with mass detection and corona charged-aerosol detection, [137] X. Zhang, Y. Zhang, H. Zhao, Y. He, X. Li, Z. Yuan, Highly sensitive and se-
J. Pharm. Biomed. Anal. 56 (2011) 271e279. lective colorimetric sensing of antibiotics in milk, Anal. Chim. Acta 778
[115] US Department of Agriculture, Food Safety and Inspection Service (USDA- (2013) 63e69.
FSIS), Office of public health science, Confirmation of aminoglycosides by [138] M. Gbylik-Sikorska, A. Posyniak, T. Sniegocki, J. Zmudzki, Liquid chro-
HPLC-MS/MS: SOP N_CLG-AMG1.02, www.fsis.usd/gov/PFD/CLG_AMG_1_03. matographyetandem mass spectrometry multiclass method for the
pdf, (accessed 01.03.15). determination of antibiotics residues in water samples from water
[116] K. Bousova, H. Senyuva, K. Mittendorf, Quantitative multi-residue method for supply systems in food-producing animal farms, Chemosphere 119
determination antibiotics in chicken meat using turbulent flow chroma- (2015) 8e15.
tography coupled to liquid chromatographyetandem mass spectrometry, [139] H. Kim, K. Seo, H. Kim, E. Jeong, J. Ghim, S. Lee, Y. Lee, D. Kim, J. Shin,
J. Chromatogr. A 1274 (2013) 19e27. Simple and accurate quantitative analysis of 20 anti-tuberculosis drugs in
[117] Y. Cui, N. Ma, X. Li, C. Lv, M. Li, M. Li, L. Song, M. Liu, Q. Li, K. Bi, Development human plasma using liquid chromatographyeelectrospray ion-
of an ultra-fast liquid chromatographyetandem mass spectrometry method izationetandem mass spectrometry, J. Pharm. Biomed. Anal. 120 (2015)
for simultaneous determination of cefazedone and etimicin in beagle dog 9e16.
plasma: application to the pharmacokinetic study of the combination of
cefazedone and etimicin injections, J. Chromatogr. B 973 (2014) 97e103.
[118] S. Toda, S. Nakagawa, T. Naito, H. Kawaguchi, Aminoglycoside antibiotics. XV
Chemical conversion of neomycin B to paromomycin I, 600 0 -deamino-600 0 -
hydroxyneomycin B and 600 0 -deamino-600 0 -hydroxy-paromomycin I,
J. Antibiot. 36 (1983) 87e91.
[119] A. Breaud, C. Henemyre-Harrisa, S. Schoolsa, N. Emeziennaa, W. Clarke, Rapid Faten Farouk Abdel Fattah is a lecturer of Pharmaceutical
quantification of the aminoglycoside arbekacin in serum using high perfor- Chemistry at Ahram Canadian University. Her research
mance liquid chromatographyetandem mass spectrometry, Clin. Chim. Acta focuses on the development of cost effective analytical
418 (2013) 102e106. methodologies for the analysis of antibiotic residues in
[120] R. Kumar, A. Rubies, R. Companyo , F. Centrich, Hydrophilic interaction food and beverage as well as development of analytical
chromatography for the analysis of aminoglycosides, J. Sep. Sci. 35 (2012) methods for detecting drug metabolism and interactions.
498e504.
[121] C. Chiaochan, U. Koesukwiwat, S. Yudthavorasit, N. Leepipatpiboon, Efficient
hydrophilic interaction liquid chromatographyetandem mass spectrometry
for the multiclass analysis of veterinary drugs in chicken muscle, Anal. Chim.
Acta 682 (2010) 117e129.
[122] R. Oertel, U. Renner, W. Kirch, Determination of neomycin by LC-tandem
mass spectrometry using hydrophilic interaction chromatography,
J. Pharm. Biomed. Anal. 35 (2004) 633e638.
[123] C. Díez, D. Guillarme, A. Spo € rri, E. Cognard, D. Ortelli, P. Edder, S. Rudaz,
Aminoglycoside analysis in food of animal origin with a zwitterionic
F. Farouk et al. / Analytica Chimica Acta 890 (2015) 21e43 43

Hassan Azzazy is a tenured professor of Chemistry at the Wilfried Niessen received his PhD from the University of
American University in Cairo. He was chairman of Amsterdam in 1986. Since 1996, he works as an indepen-
Chemistry department, the founding director of graduate dent consultant in his company hyphen MassSpec. Be-
Chemistry program, and the Associate Dean for Graduate t w e e n 2 0 0 2 a n d 2 0 14 , N i e s s e n w a s p a r t - t i m e
Studies & Research. He is the founder of Novel Di- extraordinary professor in bioanalytical mass spectrom-
agnostics & Therapeutics Research group and co-founder etry at VU University Amsterdam. His research interests
of d-Kimia, LLC. Dr. Azzazy was a postdoctoral fellow and involve principles, technology and applications of (hy-
assistant professor at University of Maryland School of phenated) mass spectrometry and fragmentation of
Medicine, Baltimore. Dr. Azzazy published over 60 even-electron ions. Niessen is (co-)author of more than
refereed articles, 55 conference abstracts, and 25 book 200 refereed papers. He was guest-editor on five MS-
chapters. He serves on the editorial boards of Clinical related special issues in Journal of Chromatography A, ed-
Biochemistry, Clin Chimica Acta, and Clinical Chemistry itor of Volume 8 of the Encyclopedia of Mass Spectrometry,
Laboratory Medicine. and author of Liquid Chromatography-Mass Spectrometry
(3rd Edition, 2006).

You might also like