Next Article in Journal
Impact of the Degree of Maturity of Walnuts (Juglans regia L.) and Their Variety on the Antioxidant Potential and the Content of Tocopherols and Polyphenols
Next Article in Special Issue
Application of Spectroscopic UV-Vis and FT-IR Screening Techniques Coupled with Multivariate Statistical Analysis for Red Wine Authentication: Varietal and Vintage Year Discrimination
Previous Article in Journal
How Far Can One Push the Noble Gases Towards Bonding?: A Personal Account
Previous Article in Special Issue
Ion Mobility Spectrometry in Food Analysis: Principles, Current Applications and Future Trends
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Transfer of a Multiclass Method for over 60 Antibiotics in Food from High Resolution to Low Resolution Mass Spectrometry

1
Istituto Zooprofilattico Sperimentale dell’Umbria e delle Marche “Togo Rosati”, 06126 Perugia, Italy
2
Department of Pharmaceutical Sciences, University of Perugia, 06123 Perugia, Italy
*
Author to whom correspondence should be addressed.
Molecules 2019, 24(16), 2935; https://doi.org/10.3390/molecules24162935
Submission received: 21 July 2019 / Revised: 8 August 2019 / Accepted: 10 August 2019 / Published: 13 August 2019
(This article belongs to the Collection Advances in Food Analysis)

Abstract

:
A multiclass method has been developed to screen and confirm a wide range of anti-microbial residues in muscle and milk, and validated using liquid-chromatography coupled to (low-resolution, LR) tandem mass spectrometry (LC-QqQ). Over sixty antibiotics, belonging to ten distinct families, were included in the method scope. The development process was rapidly concluded as a result of two previously implemented methods. This consisted of identical sample treatments, followed by liquid chromatography, and coupled with high-resolution (HR) mass spectrometry (LC-Q-Orbitrap). The validation study was performed in the range between 10–1500 μg·kg−1 for muscles and 2–333 μg·kg−1 for milk. The main performance characteristics were estimated and, then, compared to those previously obtained with HR technique. The validity of the method transfer was ascertained also through inter-laboratory studies.

1. Introduction

Antibiotics are widely used in livestock breeding to treat several diseases that appear in all the food producing animal species. To guarantee public health protection, the European Union requires member states to implement yearly monitoring plans to control the presence of antibiotic residues in food. Therefore, surveillance should be aimed particularly at controlling compliance with the maximum residue limits (MRLs), fixed in Table 1 of the Annex of Regulation (EC) No 37/2010 [1]. For several antibiotics, MRLs have been set in various matrices, such as eggs, fat, honey, kidney, liver, milk, and muscles and still, today, new MRLs are being fixed. In the early 2000s, the liquid chromatography coupled to tandem mass spectrometry technique (LC-QqQ) became essential in the routine analysis of single class of veterinary drug residues in food. Indeed triple quadrupole mass spectrometry analyzers were able to assure both greater sensitivity and selectivity than the traditional LC detectors, based on UV-Vis and fluorescence spectroscopy. In addition, for some important classes, such as aminoglycosides or avermectins, the need of a derivation step could be avoided. In the last ten years, the improvement of LC-QqQ systems allowed the realization of a further step in drug residue analysis, introducing procedures that are able to determine simultaneously more than one drug class [2,3,4]. As consequence, a remarkable effort has been made to progressively replace single-class with multiclass protocols, since this is a cost-effective way to improve the current residue control programs, thereby ensuring the determination of a wide number of compounds, with only few methods. Reviewing the main relevant published papers, some research groups recurred (Table 1). Among the control laboratories, the Official Food Control Authority of Zurich (Zurich, Switzerland), the RIKILT (Wageningen, Netherlands), the European Union Reference Laboratory for Antimicrobial Residues in Food (EURL, Fougères, France), the National Institute for Agrarian and Veterinary Research (INIAV, Vila do Conde, Portugal), the Istituto Zooprofilattico Sperimentale dell’Umbria e delle Marche (IZSUM, Perugia, Italy), the Canadian Food Inspection Agency (Calgary, Canada), the Residue Analysis Laboratory of Laboratório Nacional Agropecuário (LANAGRO, Porto Alegre, Brazil), and the US Department of Agriculture (USDA, Wyndmoor, PA, USA) are mentioning.
The universities of Barcelona (Spain), Almeria (Spain), and Athens (Greece) have been the most active in this analytical field. LC-QqQ techniques are the most consolidated and most common multiclass procedures for veterinary drugs. These techniques have been mainly developed using this type of equipment [5,6,10,11,12,16,17,19,20,23,27]. In 2008–2009, the Official Food Control Authority of Zurich and the Dutch RIKILT Institute proposed, for the first time, the application of high-resolution (HR) mass spectrometry, based on time-of-flight (TOF) technology [7,8,9]. About three years later, the same Laboratory of Zurich, and the research group of Almeria University developed multiclass procedures for veterinary drugs, respectively, in meat, and milk, using LC-Orbitrap technique, a new MS analyzer, that was commercialized in 2005 [13,29]. Later, the introduction of benchtop hybrid high-resolution mass spectrometers (mainly, Q-TOF and Q-Orbitrap) produced further advantages in terms of selectivity and accuracy and, accordingly, these kinds of equipment has been more commonly applied (Table 1) [14,15,18,21,22,24,25,26,28,29].
Based on all the above, multiclass methods are no longer innovative procedures, and there is interest in their wide diffusion. The possibility of easy implementation and sustainable daily management, independent from the available LC-MS equipment. The aim of this work was to discuss the transfer of previously developed multiclass methods for more than sixty antibiotics in meat and milk from an LC-Q-Orbitrap platform to an LC-QqQ one [24,25]. The performance characteristics of the new LC-QqQ methods were estimated by means of full validation studies carried out according to European Commission Decision 2002/657/EC [30]. Finally, a comparison between the two techniques was carried out in the light of their cost-effectiveness in routine analysis of veterinary drug residues.

2. Results and Discussion

2.1. Optimization of LC-MS/MS Conditions

The choice of analytes has been carried out using the most administered antibiotics in farm. Only the classes of aminoglycosides and colistins were excluded, as their high polarity hampers the chromatographic retention, based on the reversed-phase mechanism (C18 column). On the other hand, the addition of ion-pairing agents on the mobile phase produced remarkable ion suppression, with detrimental effects on all the other analytes [24]. The chromatographic conditions were optimized starting from the parameters set for the LC-Q-Orbitrap methods. In order to profitably increase analyte retention, the percentage of methanol (eluent B) was reduced from 5% down to 2% (by volume). According to a typical reversed-phase mechanism, this change allowed us to obtain retention times of about 0.5 min higher than the initial tested conditions (Figure S1).
The MS conditions were established without the infusion of the individual solutions of analytes, but by setting the transitions on the basis of the ion fragments previously studied [24]. As shown in Table 2, apart few exceptions, such as some beta-lactams ([M + Na]+), sulfanilamide ([M + H − NH3]+), spiramycin, neospiramycin, cefquinome, tildipirosin, tilmicosin, tulathromycin marker, and tulathromycin ([M + 2H]++), the selected precursor ion species were generally the protonated molecular ions ([M + H]+). For macrolides, it is not uncommon for the choice of bi-charged ions to be used as a precursor, due to their favorable abundance among the formed charged species [31]. The sample preparation was exactly the same as that previously optimized by Moretti et al. [24,25]; however, two internal standards (ISs) were replaced, in order to either, decrease costs (metacycline instead of tetracycline-d6), or to improve the MS response (ceftiofur-d3 instead of cefadroxil-d4). In this context, the ISs were not used for quantification purposes, but only to perform the internal quality control by checking the success of the analytical operations, during the routine application of the procedure as well as to monitor the run-to-run differences in the retention times [8]. For this purpose, at the beginning of sample treatment, IS were added at 10 µg·kg−1 and, before the release of the results, the presence (S/N > 3) of all eight compounds must be verified. The analyte quantification was achieved by matrix-matched curves (external standardization), which corrected the concentration for the relevant recovery factor [32]. The LC-QqQ chromatograms of a blank muscle, and of the same spiked at 10 µg·kg−1, are reported in Figure 1 and Figure 2, respectively. Eight representative analytes are shown, starting from the polar metabolite of florfenicol (florfenicol amine, RT = 3.4 min) to the last eluting compound (rifaximin, RT = 20.7 min). The analogous chromatograms are shown also for milk (Figure 3 and Figure 4).

2.2. Method Validation

Selectivity requirements are reported in Commission Decision 2002/657/EC [30]. The ion ratio of the two selected transitions (Table 2), and their relative retention times (<2.5%), were checked to confirm analyte identification. Linearity in the matrix was evaluated with five-points matrix-matched curves: 2, 10, 33, 100, and 150 µg·kg−1. Therefore, levels higher than 150 µg·kg−1 had to be tested, and the final extract was diluted ten-fold or more, as reported in Tables S1 and S2. The linearity data are summarized in Table S3. For several analytes, the first calibration point (2 µg·kg−1) had to be discarded, due to the scarce response. In other more critical cases (e.g., cefacetrile in meat/muscle, tildipirosin and tulathromycin markers in milk) additional points have been removed. Since Commission Decision 657/2002/EC [30] does not furnish precise criteria for evaluating linearity, the “Guidance document on analytical quality control and validation procedures for pesticide residues analysis in food and feed” was followed [33]. The percentage deviation of the back-calculated concentrations (Cmeasured) from the true concentrations (Ctrue) was calculated (1):
D e v i a t i o n   ( % ) = ( C m e a s u r e d C t r u e ) C t r u e 100
For each calibration point, it was verified that its value was not more than ± 20%. As an example, the calibration data of six analytes, belonging to different antibiotic families, are reported in Table S4 and Figures S2 and S3.
The overall recoveries and precisions data are listed in Table 3. For meat, seven validation levels were performed in the range 10–1500 µg·kg−1, whereas, in milk, five levels were investigated (10–333 µg·kg−1), with an additional concentration at 2 µg·kg−1 to check amoxicillin, ampicillin, and penicillin G accuracy at ½ MRL, as required by the Commission Decision 2002/657/EC [30]. Moreover, in milk, two additional molecules were tested with respect to the original group of 62 compounds, that is, the two quinolones, nalidixic acid, and norfloxacin.
In this work, the classical validation scheme (0.5 MRL, MRL and 1.5 MRL), described in Commission Decision 2002/657/EC, was not applied. Paragraph 3.1.3. of the same decision allows the introduction of alternative models [30] and, since the validation studies of multiclass procedures have to consider dozens of MRLs, which can vary also in function of the animal species, the adoption of progressive validation levels, that are equal for all the analytes, was fully justified. On the other hand, when the Commission Decision was issued (2002), the development of multiclass procedures for the control of veterinary drug residues was not initiated. The spiking ranges were chosen, considering, on the one side, the reachable concentrations, and, on the other side, the relevant MRLs [1] in all the couplings analyte/matrix. Preliminary experiments demonstrated that, at levels lower than 10 µg·kg−1, the precision of several amphenicols, macrolides, beta-lactams, and tetracyclines became unsatisfactory.
The recovery factors (Rec) were established by comparing the peak area of each compound in the spiked samples against the peak area in the matrix matched standards, by which the antibiotics were added immediately prior to LC injection. The data in Table 3 summarize the average recoveries obtained in the whole validation range. In muscles, all recoveries were higher than 70%, except the majority of tetracyclines (62–69%), three beta-lactams (64–68%), and two macrolides (neospiramycin, 67% and tulathromycin, 69%). Analogously, in milk, recoveries higher than 70% were generally obtained, except for the more polar sulfonamides (61–70%) and one macrolide (erythromycin A, 67%). Since the quantification was performed with an external standardization, the raw results were always corrected for the relevant recovery factor, in order to correct the systematic error [32]. With regard to precision, the coefficients of variations (CVs) were calculated at each validation level both, repeatedly, and in intra-lab reproducibility conditions (CVr, and CVwR, respectively), by applying ANOVA. Moreover the CVwR (and CVr) were pooled to obtain an overall precision index, namely CVwR,pooled (2),
C V w R , p o o l e d = ( n 1 1 ) C V w R 1 2 + ( n 2 1 ) C V w R 2 2 + ... ( n n 1 ) C V w R n 2 ( n 1 1 ) + ( n 2 1 ) + ...
where CVwR1, CVwR2 ... CVwRn were the coefficients of variation at the increasing levels 1, 2 … n; n1, n2…n were the number of replicates at each level [34]. For a certain analyte, the CVwR,pooled give a single estimate of precision, which can be applied to calculate decision limits (CCα) and detection capabilities (CCβ) at whatever MRL value. Therefore, the CVwR,pooled can also be used to obtain CCα and CCβ, where new MRLs were fixed in Regulation 37/2010 [1], maximizing the cost-effectiveness of the validation study. The decision limit and detection capability were calculated as follows (equations 3 and 4):
C C α M R L = M R L + 1.64 C V w R , p o o l e d M R L
C C β M R L = C C α M R L + 1.64 C V w R , p o o l e d C C α M R L
In Tables S4 and S5, the MRLs of the 64 antibiotics, in various food-producing animal species, and in bovine milk, together with the relevant CCαs and CCβs, are listed. The MRLs are those reported in the last consolidated text of Regulation 37/2010 [1,35]. In muscles, valnemulin demonstrated high imprecision (CVwR,pooled = 31%) and, therefore, for these compounds, the developed procedure could be only used for screening purposes (Table 3). For all the other antibiotics, CVwR,pooled was always lower than 20%. On the other hand, in milk, cefacetrile and tulathromycin marker demonstrated insufficient precision (CVwR,pooled > 22%). Matrix effects (ME%) listed in Table 3 were calculated as follows (equation 5),
M E ( % ) = b M M b S × 100
where bMM, and bS were the slopes of matrix matched curves, and solvent standard curves, i.e., curves prepared in ammonium acetate 0.2 M, respectively. In the whole, although the sample purification was scarce, the matrix effects (suppression or enhancement) were limited and very few compounds demonstrated ME (%) higher than |50|%. This was probably because the long chromatographic run (30.5 min) allowed the distribution of matrix-interfering compounds and analytes, from preventing excessive bunching [26].
Since the signal-to-noise approach (S/N) is rather subjective [36], “operative” (fit for purpose) LOD and LOQ were fixed by examining the precision at each validation level (Table S7). All the compounds were detectable at the first concentration, i.e., 2 µg·kg−1 for amoxicillin, ampicillin, and penicillin G in milk and 10 µg·kg−1 for all the others. The only exception was cefacetrile, which is a scarcely ionizable molecule and, therefore, detectable and quantifiable from 33 µg·kg−1 in muscle (CVr = 13%, CVwR = 14%, recovery = 76%) and only detectable in milk (insufficient precision). A satisfactory accuracy for both amoxicillin and ampicillin was obtained at 2 µg·kg−1, whereas penicillin G demonstrated unsatisfactory precision at this level (CVr = 23%, CVwR = 26%) and, therefore, this beta-lactam could be quantified only starting from 10 µg·kg−1 (CVr = 7.5%, CVwR = 11%). Therefore, penicillin G, a fundamental drug for the treatment of sub-clinical mastitis in lactating cows, could not be quantified in milk at ½ MRL (2 µg·kg−1) and the method was suitable only for screening purpose [30]. In Figure S4 the LC-QqQ chromatograms of these three compounds are shown in a blank milk sample and in the same sample spiked at 2 µg·kg−1.

2.3. Comparison of LC-QqQ and LC-Q-Orbitrap Methods

Comparing the recovery factors of the methods developed in meat, similar results were always obtained (differences < 15%), except for cefquinome, which demonstrated a higher recovery when LC-QqQ was applied (+ 26%: 97% LR vs. 71% HR) and valnemulin which, nevertheless, was not accurately quantifiable with the LR procedure, as discussed above. The recovery differences are visualized in Figure S5. Examining the data of the more polar cephalosporins (desacetylcephapirin, cephapirin, cefacetrile and cefalonium), cefquinome recovery appeared suspect (over-estimated) using QqQ technique. Since the sample preparation protocol was the same, this difference should be attributed to an instrumental technique. Interestingly, cefquinome co-eluted with sulfamerazine and, in addition, accidentally their precursor ion had the same nominal mass charge ratio i.e., m/z 265 (Table 2). In our laboratory, at the beginning of the development of multiclass methods for antibiotics in food (2014), two separate matrix-matched curves were prepared, one for beta-lactams and another for all other analytes [24]. This measure was precautionary in order to avoid the possible negative effects of methanol, contained in the intermediate solution of antibiotics other than beta-lactams (see Section 2.2) on beta-lactam stability, as described in the literature [37,38]. In saying that, the artefacts in the quantification of cefquinome and/or sulfamerazine could occur if “hidden” transitions were shared between these two compounds. This means that a transition, monitored only for one of the two analytes (Table 2), was shared by the other one, too. In order to verify this hypothesis, two individual solutions (50 ng·mL−1) were separately injected, by simultaneously monitoring the four relevant SRMs (Figure S6). It was evident that sulfamerazine shared a “hidden” transition with cefquinome (m/z 265 > m/z 199—left side of Figure S6), but also viceversa (m/z 265 > m/z 156—right side of Figure S6). For sulfamerazine the fragment ion species at m/z 199 was formed by the rearrangement ion, by losing H2SO2 from the protonated molecule [39]. On the other hand, for cefquinome, the ion m/z 156 derived by the cleavage of C-C bond between oxime and the carbonyl group of C7 amide ([C5H6N3OS]+) of the beta-lactam ring [40]. However, from a quantitative point of view, sulfamerazine could notably affect the peak area of cefquinome, but the contrary was much less, since sulfamerazine responded more significantly than cefquinome (about 4.9 × 106 vs. 1.1 × 106). Accordingly, when sulfamerazine was co-present, cefquinome concentration was significantly over-estimated. Later, observing that, in the adopted experimental conditions, beta-lactams were not deteriorated by methanol, the validation study in milk was performed by preparing only one matrix-matched calibration curve with all the analytes, including beta-lactams. In summary, although the development of LR procedures has been very fast and effective, due to previously-studied conditions [24,25], for one analyte, i.e., cefquinome, the choice of the precursor ion should be re-evaluated.
In Figure S7 the differences between LR and HR recovery factors in milk are shown. Tulathromycin marker and cefquinome had better recovery rates using HR detection, whereas valnemulin, cloxacillin, and rifaximin, showed better results using the LR system. As reported in our previous paper [25], LC-Q-Orbitrap suffers from “post interface ion suppression”, which consists of instrument saturation when intense matrix-related compounds are present [41]. This phenomenon was more pronounced for the last eluting compounds, such as valnemulin, cloxacillin, and rifaximin, which explains the observed data in milk extracts. These have more interfering substances with respect to muscle [25]. Comparing the precision data (intra-laboratory reproducibility, CVwR,pooled, see Tables S8 and S9), remarkable differences (≥15%) between the two techniques were observed only in milk and, again, tulathromycin marker and cefquinome revealed the worse performances when determined by LC-QqQ. CwR,pooled of tylosin, cloxacillin, and tylvalosin were about 10% higher when analyzed by LC-Q-Orbitrap (Figure S9).
According to accreditation rules [42], since 2014 our laboratory participated in Proficiency Test Schemes in meat and milk, by applying the LC-Q-Orbitrap methods in these products. Some of the stored test materials were then re-tested, by applying the new developed procedures. The results, together with the consensus values assigned by the Organizers, are listed in Table 4. Examining the acceptability ranges, satisfactory z-scores would have been always obtained, except in the case of amoxicillin in milk (sample code: MI1532-A1). This latter result was explainable with the well-known instability of penicillin antibiotics [43,44].
In summary, the main advantages and disadvantages of the two techniques are undoubtedly, the QqQ analyser, which only involves only a kind of acquisition, i.e., SRM mode. On the contrary, Q-Orbitrap forces more complex experiments, also because the detection of analytes at trace levels is complicated by the “post interface ion suppression” phenomenon. On this subject, we demonstrated that, in certain chromatographic regions, for milk it is not possible to reach the required limits using full-scan experiments, since the massive presence of interfering substances can drastically worsen the sensitivity [25]. With regard to the sample throughput, the sample preparation protocol is identical and, therefore, there is no great difference. Moreover, it must be highlighted that some performances of LR system are due to the obsolescence of the available equipment. For example, most likely, a more recent LR platform could reach comparable LODs with HR, i.e., lower than 10 µg·kg−1. All that said, the LC-QqQ technique is more suitable for routine laboratories, considering its user-friendliness and lesser cost (about three time lesser than LC-Q-Orbitrap).

3. Experimental

3.1. Chemical and Reagents

Acetonitrile (ACN) and methanol (LC-MS grade) were from Carlo Erba Reagents (Milan, Italy). Formic acid (50%) and N,N’-dimethylformamide (DMF) were purchased from Sigma-Aldrich (St. Louis, MO, USA). EDTA sodium salt dehydrate and ammonium acetate were provided by Sigma-Aldrich. Ultra-pure deionized water was generated by a Milli-Q purification apparatus (Millipore, Bedford, MA, USA). Amoxicillin, ampicillin, cloxacillin, dicloxacillin, nafcillin, oxacillin, penicillin G (benzylpenicillin), penicillin G-d7, penicillin V (phenoxymethylpenicillin), cefalonium, cefoperazone, cefquinome, ceftiofur, cephalexin, ciprofloxacin, danofloxacin, difloxacin, enrofloxacin, flumequine, marbofloxacin, nalidixic acid, norfloxacin, oxolinic acid, sarafloxacin, erithromycin A, spiramycin I, tylosin A, tilmicosin, sulfadiazine, sulfaguanidine, sulfadimethoxine, sulfamerazine, sulfamethazine (sulfadimidine), sulfamethoxazole, sulfanilamide, sulfapyridine, sulfaquinoxaline, sulfathiazole, trimethoprim, chlortetracycline, doxycycline, metacycline, oxytetracycline, tetracycline, florfenicol, florfenicol amine, thiamphenicol, lincomycin, rifaximin, tiamulin and valnemulin were obtained from Sigma-Aldrich. Sulfamonomethoxine was purchased from Dr. Ehrenstorfer (Augsburg, Germany); cefazolin, cefacetrile, ceftiofur-d3, cephapirin, desacetylcephapirin, florfenicol-d3, 3-O-acetyltylosin, gamithromycin, neospiramycin, spiramycin I-d3, tildipirosin, tulathromycin, tylvalosin, 4-epi-chlortetracycline, 4-epi-tetracycline, 4-epi-oxytetracycline, and tulathromycin marker (CP-60,300) were purchased from TRC Inc. (Toronto, Canada); sulfamethazine-13C6, sulfanilamide-13C6 and enrofloxacin-d5 were obtained from WITEGA (Berlin, Germany).

3.2. Standard Solutions

Individual stock standard solutions of 100 µg·mL−1 were prepared with methanol (amphenicols, lincosamides, macrolides, pleuromutilins, sulphonamides, tetracyclines and trimethoprim). The solubilization and storage conditions were previously studied [24]. Beta lactams were solubilized in H2O/ACN 75/25 (v/v), except ceftiofur (DMF). Quinolones in MeOH/H2O 80/20 (v/v), except ciprofloxacin, nalidixic acid, norfloxacin, enrofloxacin and oxolinic acid (DMF). Rifaximin was prepared in MeOH/H2O 50/50 (v/v). The stock solutions were stored at −20 °C with variable storage times: From 1 month (cefquinome) to 24 months (sulphonamides). Working solutions (10, 1 and 0.1 µg·mL−1) were prepared from the relevant stock solutions diluting with H2O/ACN 75/25 (v/v) for beta-lactams and with methanol for all the other antibiotics. The solutions of internal standards were prepared according to their native compound or class.

3.3. LC-MS-MS Conditions

LC-MS/MS measurements were performed by a Surveyor LC pump, coupled with a triple quadrupole mass spectrometer (TSQ Quantum Ultra, Thermo Fisher, San Jose, CA, USA), an electrospray source included, and operating in a positive ionization mode. Separation was achieved on a Poroshell 120 EC-C18 column (3.0 × 100 mm; 2.7 μm particle diameter), which was connected to a guard column Poroshell (2.1 × 5 mm), both from Agilent Technologies (Santa Clara, CA, USA). The flow rate used was 0.25 mL·min−1 and the column temperature set at 30 °C. Mobile phase A was an aqueous solution 0.1% (v/v) formic acid and eluent B methanol. The gradient profile started at 2% eluent B for 1 min and increased linearly up to 95% B in 19.5 min; this condition was maintained for 5 min before returning to initial condition in 1 min (2% B) and held for 4 min to equilibrate the column. The sample temperature was kept at 16 °C and the injection volume 10 µL. For MS detection, the parameters were the follows: Capillary temperature 300 °C, vaporizer 320 °C, spray voltage 3 kV, and a resolution setting of Q1 and Q3 m/z 0.7. Sheath gas and auxiliary gas (nitrogen) pressures were set at 35, and 15 arbitrary units, respectively. Collision gas (argon) pressure: 1.5 mtorr. The collision energies, that were associated with each transition, are listed in Table 2.

3.4. Sample Preparation

The sample preparation was described elsewhere [24,25]. Briefly, (1.50 ± 0.01) g of minced muscle or milk were weighed in a 50 mL Falcon tube. The sample was spiked with: (i) 15 µL of the solution of the two internal standards (ISs) of beta-lactams at 1 µg·mL−1 (ceftiofur-d3 and penicillin G-d7); (ii) 15 µL of the solution of the other six ISs (sulfanilamide-13C6, sulfamethazine-13C6, enrofloxacin-d5, florfenicol-d3, spiramycin-d3 and metacycline) at 1 µg mL−1. For muscles, one hundred microliters of 0.1 M of EDTA was then added, and the sample was extracted with 3 mL of a mixture of acetonitrile/water 80/20 (v/v). Milk was extracted with one milliliter of 0.1 M of EDTA and 3 mL of acetonitrile. A second extraction with 3 mL of pure acetonitrile was performed for both matrices. After centrifugation, the reunited extracts were evaporated and solubilized in 1.5 mL of ammonium acetate 0.2 M. Ten µL was injected in the LC system.

3.5. Method Validation

The validation was carried out following the Commission Decision 2002/657/EC [30]. To test the selectivity, blank muscle samples, belonging to the main animal species (bovine, swine and poultry) and bovine milk samples from different origins, were analyzed. The linearity in the matrix has been evaluated in the range 2–150 µg·kg−1 (2, 10, 33, 100 and 150 µg·kg−1). The matrix-matched solutions were prepared by adding the analytes immediately prior to LC injection. The curves were constructed, including at zero (blank), and by plotting the average peak area of analyte (three injections for each concentration point) against its concentration. An unweighted linear regression model was applied to the calibration data. The precision (repeatability and within-laboratory reproducibility), recovery (trueness), decision limit (CCα), and detection capability (CCβ) were studied following the experimental plans, described in Table S1 (muscle) and Table S2 (milk) of the Supplementary Material. A blank bovine muscle or milk was spiked at the beginning of the extraction procedure with the appropriate standard solutions. Four replicates (n = 4) at each level were carried out on the same day, along with the relevant matrix-matched calibration curve. Each series was repeated on three different days, and at varying times, operator, and calibration status of the LC-MS system. The spiking levels in muscles were; 10, 33, 100, 150, 333, 1000, and 1500 µg·kg−1 and 10, 33, 100, 150, and 333 µg kg−1 in milk. In milk, penicillin G, amoxicillin, and ampicillin were also tested at 2 µg·kg−1. The Limits of Detection (LODs ) and Quantification (LOQs) were estimated on the basis of the observed accuracy (recovery and precision) at the first validation concentration or, if necessary, at the second one.

4. Conclusions

The validity of the transfer, from LC-Q-Orbitrap to LC-QqQ of two multiclass methods for veterinary drugs in food, has been demonstrated. Using the LR platform, valnemulin in muscle, cefacetrile, and tulathromycin marker in milk did not reach acceptable precision. In addition, examining LC-QqQ results, a series of accidental events (co-elution, selected precursor ions, fragmentation pathway etc) produced an over-estimation of cefquinome in meat, due to the co-presence of sulfamerazine. If neglected, this phenomenon could give false positive results. Since the quantification process of the LC-Q-Orbitrap system was based on a different principle (peak area of the precursor ion measured with MS accuracy < 5 ppm), this drawback was not observed. However, in milk, LC-Q-Orbitrap achieved worse recoveries for some of the more non-polar analytes, eluting in the chromatographic zone in which the interfering substances were more abundant. The “post interface ion suppression”, which is a specific phenomenon of Orbitrap mass analysers could explain this latter evidence. A further consideration concerns the chromatographic separation, which is fundamental when highly selective detectors are applied. A good separation of peaks, avoiding the overlapping and bunching of both analytes and endogenous substances, minimizes the risk of false positive results and reduces matrix effects. Finally, the described method transfer has been successfully performed on an obsolete LC-QqQ platform (fifteen year-old), encouraging the implementation of multiclass strategy also in routine laboratories with limited instrumental resources.

Supplementary Materials

The following are available online, Figure S1: LC-QqQ chromatograms of the three more polar analytes (sulfanilamide, sulfaguanidine and florfenicolamine) with different % MeOH at the beginning of the gradient: 2% (left) and 5% (right), Figure S2: Matrix-matched calibration curves of danofloxacin, doxycycline, oxacillin, spiramycin, sulfamethazine and tiamulin in meat, Figure S3: Matrix-matched calibration curves of of danofloxacin, doxycycline, oxacillin, spiramycin, sulfamethazine and tiamulin in milk, Figure S4: LC-QqQ chromatograms of a blank (a) and a milk sample spiked at 2 μg kg-1 (b) of the three penicillins with MRL at 4 μg kg-1, Figure S5: Recovery differences between the LC-QqQ and LC-Q-Orbitrap methods in meat. Positive values (%) indicate better recovery of LC-QqQ procedure and vice versa, Figure S6: LC-QqQ chromatograms of individual solutions (50 ng mL-1) of sulfamerazine (left) and cefquinome (right), Figure S7: Recovery differences between the LC-QqQ and LC-Q-Orbitrap methods in bovine milk. Positive values (%) indicate better recovery of LC-QqQ procedure and vice versa, Figure S8: Precision differences between the LC-QqQ and LC-Q-Orbitrap methods in meat. Positive values (%) indicate better precision of LC-QqQ procedure and vice versa, Figure S9: Precision differences between the LC-QqQ and LC-Q-Orbitrap methods in bovine milk. Positive values (%) indicate better precision of LC-QqQ procedure and vice versa, Table S1: Validation plan in bovine muscle, Table S2: Validation plan in milk, Table S3: Linearity studies in matrix (matrix-matched curves), Table S4: Calibration data (matrix-matched curves) of six representative analytes, Table S5: MRL (μg kg-1), decision limits (μg kg-1) and detection capabilities (μg kg-1) of the 64 tested antibiotics in the main food-producing species, Table S6: MRL (μg kg-1), decision limits (μg kg-1) and detection capabilities (μg kg-1) of the 64 antibiotics in aquaculture, rabbits, horses, Table S7: Estimated LODs and LOQs based on the observed accuracy at the first and at the second validation level.

Author Contributions

Conceptualization, D.G., S.M., G.S. and F.P.; methodology, G.S. and S.M.; validation, D.G., F.P. and C.B.; investigation, D.G., F.P., S.M, G.S. and C.B.; data curation, R.S. and F.I.; writing—original draft preparation, R.G.; writing—review and editing, R.G., R.S. and F.I.; supervision and funding acquisition, R.G.

Funding

This research was supported by Ministero della Salute, Ricerca Corrente IZSUM 002 2015

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. European Communities Commission Regulation (EU) No 37/2010 of 22 December 2009 on pharmacologically active substances and their classification regarding maximum residue limits in foodstuffs of animal origin. Off. J. Eur. Communities 2010, L15, 1–72. Available online: https://eur-lex.europa.eu/legal-content/EN/TXT/?uri=CELEX%3A32010R0037 (accessed on 30 May 2019).
  2. Masiá, A.; Suarez-Varela, M.M.; Llopis-Gonzalez, A.; Picó, Y. Determination of pesticides and veterinary drug residues in food by liquid chromatography-mass spectrometry: A review. Anal. Chim. Acta 2016, 936, 40–61. [Google Scholar] [CrossRef] [PubMed]
  3. Mainero Rocca, L.; Gentili, A.; Pérez-Fernández, V.; Tomai, P. Veterinary drugs residues: A review of the latest analytical research on sample preparation and LC-MS based methods. Food Addit. Contam. - Part A Chem. Anal. Control. Expo. Risk Assess. 2017, 34, 766–784. [Google Scholar] [CrossRef] [PubMed]
  4. Rossi, R.; Saluti, G.; Moretti, S.; Diamanti, I.; Giusepponi, D.; Galarini, R. Multiclass methods for the analysis of antibiotic residues in milk by liquid chromatography coupled to mass spectrometry: A review. Food Addit. Contam. - Part A Chem. Anal. Control. Expo. Risk Assess. 2018, 35, 241–257. [Google Scholar] [CrossRef] [PubMed]
  5. Aguilera-Luiz, M.M.; Vidal, J.L.M.; Romero-González, R.; Frenich, A.G. Multi-residue determination of veterinary drugs in milk by ultra-high-pressure liquid chromatography-tandem mass spectrometry. J. Chromatogr. A 2008, 1205, 10–16. [Google Scholar] [CrossRef] [PubMed]
  6. Chico, J.; Rúbies, A.; Centrich, F.; Companyó, R.; Prat, M.D.; Granados, M. High-throughput multiclass method for antibiotic residue analysis by liquid chromatography-tandem mass spectrometry. J. Chromatogr. A 2008, 1213, 189–199. [Google Scholar] [CrossRef] [PubMed]
  7. Kaufmann, A.; Butcher, P.; Maden, K.; Widmer, M. Quantitative multiresidue method for about 100 veterinary drugs in different meat matrices by sub 2-microm particulate high-performance liquid chromatography coupled to time of flight mass spectrometry. J. Chromatogr. A 2008, 1194, 66–79. [Google Scholar] [CrossRef] [PubMed]
  8. Stolker, A.A.M.; Rutgers, P.; Oosterink, E.; Lasaroms, J.J.P.; Peters, R.J.B.; Van Rhijn, J.A.; Nielen, M.W.F. Comprehensive screening and quantification of veterinary drugs in milk using UPLC-ToF-MS. Anal. Bioanal. Chem. 2008, 391, 2309–2322. [Google Scholar] [CrossRef]
  9. Peters, R.J.B.B.; Bolck, Y.J.C.C.; Rutgers, P.; Stolker, A.A.M.M.; Nielen, M.W.F.F. Multi-residue screening of veterinary drugs in egg, fish and meat using high-resolution liquid chromatography accurate mass time-of-flight mass spectrometry. J. Chromatogr. A 2009, 1216, 8206–8216. [Google Scholar] [CrossRef]
  10. Stubbings, G.; Bigwood, T. The development and validation of a multiclass liquid chromatography tandem mass spectrometry (LC-MS/MS) procedure for the determination of veterinary drug residues in animal tissue using a QuEChERS (QUick, Easy, CHeap, Effective, Rugged and Safe) approac. Anal. Chim. Acta 2009, 637, 68–78. [Google Scholar] [CrossRef]
  11. Gaugain-Juhel, M.; Delépine, B.; Gautier, S.; Fourmond, M.P.; Gaudin, V.; Hurtaud-Pessel, D.; Verdon, E.; Sanders, P. Validation of a liquid chromatography-tandem mass spectrometry screening method to monitor 58 antibiotics in milk: A qualitative approach. Food Addit. Contam. - Part A Chem. Anal. Control. Expo. Risk Assess. 2009, 26, 1459–1471. [Google Scholar] [CrossRef] [PubMed]
  12. Martínez Vidal, J.L.; Frenich, A.G.; Aguilera-Luiz, M.M.; Romero-González, R. Development of fast screening methods for the analysis of veterinary drug residues in milk by liquid chromatography-triple quadrupole mass spectrometry. Analytical and Bioanalytical Chemistry. 2010, 397, 2777–2790. [Google Scholar] [CrossRef] [PubMed]
  13. Kaufmann, A.; Butcher, P.; Maden, K.; Walker, S.; Widmer, M. Development of an improved high resolution mass spectrometry based multi-residue method for veterinary drugs in various food matrices. Anal. Chim. Acta 2011, 700, 86–94. [Google Scholar] [CrossRef] [PubMed]
  14. Hurtaud-Pessel, D.; Jagadeshwar-Reddy, T.; Verdon, E. Development of a new screening method for the detection of antibiotic residues in muscle tissues using liquid chromatography and high resolution mass spectrometry with a LC-LTQ-Orbitrap instrument. Food Addit. Contam. - Part A Chem. Anal. Control. Expo. Risk Assess. 2011, 28, 1340–1351. [Google Scholar] [CrossRef] [PubMed]
  15. Wang, J.; Leung, D. The challenges of developing a generic extraction procedure to analyze multi-class veterinary drug residues in milk and honey using ultra-high pressure liquid chromatography quadrupole time-of-flight mass spectrometry. Drug Test. Anal. 2012, 1, 103–111. [Google Scholar] [CrossRef] [PubMed]
  16. Bittencourt, M.S.; Martins, M.T.; de Albuquerque, F.G.S.; Barreto, F.; Hoff, R. High-throughput multiclass screening method for antibiotic residue analysis in meat using liquid chromatography-tandem mass spectrometry: A novel minimum sample preparation procedure. Food Addit. Contam. - Part A Chem. Anal. Control. Expo. Risk Assess. 2012, 29, 508–516. [Google Scholar] [CrossRef] [PubMed]
  17. Martins, M.T.; Melo, J.; Barreto, F.; Barcellos Hoff, R.; Jank, L.; Soares Bittencourt, M.; Bazzan Arsand, J.; Scherman Schapoval, E.E. A simple, fast and cheap non-SPE screening method for antibacterial residue analysis in milk and liver using liquid chromatography-tandem mass spectrometry. Talanta 2014, 129, 374–383. [Google Scholar] [CrossRef] [PubMed]
  18. Kaufmann, A.; Butcher, P.; Maden, K.; Walker, S.; Widmer, M. Multi-residue quantification of veterinary drugs in milk with a novel extraction and cleanup technique: Salting out supported liquid extraction (SOSLE). Anal. Chim. Acta 2014, 820, 56–68. [Google Scholar] [CrossRef]
  19. Freitas, A.; Barbosa, J.; Ramos, F. Multidetection of antibiotics in liver tissue by ultra-high-pressure-liquid-chromatography-tandem mass spectrometry. J. Chromatogr. B Anal. Technol. Biomed. Life Sci. 2015, 976-977, 49–54. [Google Scholar] [CrossRef]
  20. Martins, M.T.; Barreto, F.; Hoff, R.B.; Jank, L.; Arsand, J.B.; Feijó, T.C.; Schapoval, E.E.S. Determination of quinolones and fluoroquinolones, tetracyclines and sulfonamides in bovine, swine and poultry liver using LC-MS/MS. Food Addit. Contam. - Part A Chem. Anal. Control. Expo. Risk Assess. 2015, 32, 333–341. [Google Scholar] [CrossRef]
  21. Wang, J.; Leung, D.; Chow, W.; Chang, J.; Wong, J.W. Development and Validation of a Multiclass Method for Analysis of Veterinary Drug Residues in Milk Using Ultrahigh Performance Liquid Chromatography Electrospray Ionization Quadrupole Orbitrap Mass Spectrometry. J. Agric. Food Chem. 2015, 63, 9175–9187. [Google Scholar] [CrossRef] [PubMed]
  22. Dasenaki, M.E.; Thomaidis, N.S. Multi-residue determination of 115 veterinary drugs and pharmaceutical residues in milk powder, butter, fish tissue and eggs using liquid chromatography-tandem mass spectrometry. Anal. Chim. Acta 2015, 880, 103–121. [Google Scholar] [CrossRef] [PubMed]
  23. Dasenaki, M.E.; Michali, C.S.; Thomaidis, N.S. Analysis of 76 veterinary pharmaceuticals from 13 classes including aminoglycosides in bovine muscle by hydrophilic interaction liquid chromatography–tandem mass spectrometry. J. Chromatogr. A 2016, 1452, 67–80. [Google Scholar] [CrossRef] [PubMed]
  24. Moretti, S.; Dusi, G.; Giusepponi, D.; Pellicciotti, S.; Rossi, R.; Saluti, G.; Cruciani, G.; Galarini, R. Screening and confirmatory method for multiclass determination of 62 antibiotics in meat. J. Chromatogr. A 2016, 1429, 175–188. [Google Scholar] [CrossRef] [PubMed]
  25. Moretti, S.; Cruciani, G.; Romanelli, S.; Rossi, R.; Saluti, G.; Galarini, R. Multiclass method for the determination of 62 antibiotics in milk. J. Mass Spectrom. 2016, 51, 792–804. [Google Scholar] [CrossRef] [PubMed]
  26. Anumol, T.; Lehotay, S.J.; Stevens, J.; Zweigenbaum, J. Comparison of veterinary drug residue results in animal tissues by ultrahigh-performance liquid chromatography coupled to triple quadrupole or quadrupole–time-of-flight tandem mass spectrometry after different sample preparation methods, including use of. Anal. Bioanal. Chem. 2017, 409, 2639–2653. [Google Scholar] [CrossRef] [PubMed]
  27. Lehotay, S.J.; Lightfield, A.R. Simultaneous analysis of aminoglycosides with many other classes of drug residues in bovine tissues by ultrahigh-performance liquid chromatography–tandem mass spectrometry using an ion-pairing reagent added to final extracts. Anal. Bioanal. Chem. 2018, 2018. 410, 1095–1109. [Google Scholar] [CrossRef]
  28. Gaspar, A.F.; Santos, L.; Rosa, J.; Leston, S.; Barbosa, J.; Vila Pouca, A.S.; Freitas, A.; Ramos, F. Development and validation of a multi-residue and multi-class screening method of 44 antibiotics in salmon (Salmo salar) using ultra-high-performance liquid chromatography/time-of-flight mass spectrometry: Application to farmed salmon. J. Chromatogr. B Anal. Technol. Biomed. Life Sci. 2019, 1118–1119, 78–84. [Google Scholar] [CrossRef]
  29. Romero-González, R.; Aguilera-Luiz, M.M.; Plaza-Bolaños, P.; Frenich, A.G.; Vidal, J.L.M. Food contaminant analysis at high resolution mass spectrometry: Application for the determination of veterinary drugs in milk. J. Chromatogr. A 2011, 1218, 9353–9365. [Google Scholar] [CrossRef]
  30. Commission Decision (2002/657/EC) of 12 August 2002 implementing Council Directive 96/23/EC concerning the performance of analytical methods and the interpretation of results. Off. J. Eur. Communities 2002, L221, 8–36.
  31. Dickson, L.C. Performance characterization of a quantitative liquid chromatography-tandem mass spectrometric method for 12 macrolide and lincosamide antibiotics in salmon, shrimp and tilapia. J. Chromatogr. B Anal. Technol. Biomed. Life Sci. 2014, 967, 203–210. [Google Scholar] [CrossRef] [PubMed]
  32. SANCO/2004/2726-rev 4-December 2008 Guidelines for the Implementation of Decision 2002/657/EC. European Commission Health & Consumer Protection Directorate-General. 2008. Available online: https://ec.europa.eu/food/sites/food/files/safety/docs/cs_vet-med-residues_cons_2004-2726rev4_en.pdf (accessed on 30 May 2019).
  33. SANTE/11813/2017. Guidance document on analytical quality control and validation procedures for pesticide residues analysis in food and feed. In European Commission Health & Consumer Protection Directorate-General. 2017. Available online: https://ec.europa.eu/food/sites/food/files/plant/docs/pesticides_mrl_guidelines_wrkdoc_2017-11813.pdfwebsite (accessed on 30 May 2019).
  34. Barvick, V.J.; Wllison, S.R.L. Part (d): Protocol for uncertainty evaluation from validation data. In VAM Project 3.2.1. Development and Harmonisation of Measurement Uncertainty Principles; LGC: Teddington, UK, 2000. [Google Scholar]
  35. European Communities Commission Regulation (EU) No 37/2010 of 22 December 2009 on pharmacologically active substances and their classification regarding maximum residue limits in foodstuffs of animal origin. Consolidated test. 3 March 2019. Available online: https://eur-lex.europa.eu/collection/eu-law/consleg.html (accessed on 30 May 2019).
  36. Galarini, R.; Moretti, S.; Saluti, G. Quality Assurance and Validation General Considerations and Trends. In Chromatographic Analysis of the Environment Mass Spectrometry Based Approaches, Fourth Edition; Leo, M.L., Nollet, D.A.L., Eds.; CRC Press: Boca Raton, FL, USA, 2017; ISBN 9781315316208. [Google Scholar]
  37. Grujic, S.; Vasiljevic, T.; Lausevic, M.; Ast, T. Study on the formation of an amoxicillin adduct with methanol using electrospray ion trap tandem mass spectrometry. Rapid Commun. Mass Spectrom. 2008, 22, 67–74. [Google Scholar] [CrossRef] [PubMed]
  38. Mastovska, K.; Lightfield, A.R. Streamlining methodology for the multiresidue analysis of beta-lactam antibiotics in bovine kidney using liquid chromatography-tandem mass spectrometry. J. Chromatogr. A 2008, 1202, 118–123. [Google Scholar] [CrossRef] [PubMed]
  39. Asteggiante, L.G.; Nunez, A.; Lehotay, S.J.; Lightfield, A.R. Structural characterization of product ions by electrospray ionization and quadrupole time-of-flight mass spectrometry to support regulatory analysis of veterinary drug residues in foods. Rapid Commun. Mass Spectrom. 2014, 28, 1061–1081. [Google Scholar] [CrossRef] [PubMed]
  40. Niessen, W.M.A.; Correa, R.A.C. Interpretation of MS-MS Mass Spectra of Drugs and Pesticides; John Wilewy and Sons: Hoboken, NJ, USA, 2017. [Google Scholar]
  41. Kaufmann, A.; Walker, S. Extension of the Q Orbitrap intrascan dynamic range by using a dedicated customized scan. Rapid. Commun. Mass Spectrom. 2016, 30, 1087–1095. [Google Scholar] [CrossRef] [PubMed]
  42. General requirements for the competence of testing and calibration laboratories, Second ed.; International Standard Organization: Geneva, Switzerland, 2005.
  43. Berendsen, B.J.A.A.; Elbers, I.J.W.W.; Stolker, A.A.M.M. Determination of the stability of antibiotics in matrix and reference solutions using a straightforward procedure applying mass spectrometric detection. Food Addit. Contam. - Part A Chem. Anal. Control. Expo. Risk Assess. 2011, 28, 1657–1666. [Google Scholar] [CrossRef]
  44. Gaugain, M.; Chotard, M.P.; Verdon, E. Stability study for 53 antibiotics in solution and in fortified biological matrixes by LC/MS/MS. J. AOAC Int. 2013, 96, 471–480. [Google Scholar] [CrossRef]
Sample Availability: Not available.
Figure 1. LC-QqQ chromatograms of a blank bovine muscle.
Figure 1. LC-QqQ chromatograms of a blank bovine muscle.
Molecules 24 02935 g001
Figure 2. LC-QqQ chromatograms of a spiked bovine muscle (10 µg·kg−1).
Figure 2. LC-QqQ chromatograms of a spiked bovine muscle (10 µg·kg−1).
Molecules 24 02935 g002
Figure 3. LC-QqQ chromatograms of a blank bovine milk.
Figure 3. LC-QqQ chromatograms of a blank bovine milk.
Molecules 24 02935 g003
Figure 4. LC-QqQ chromatograms of a spiked bovine milk (10 µg·kg−1).
Figure 4. LC-QqQ chromatograms of a spiked bovine milk (10 µg·kg−1).
Molecules 24 02935 g004
Table 1. Overview of multiclass methods for the determination of veterinary drug residues in tissues and milk.
Table 1. Overview of multiclass methods for the determination of veterinary drug residues in tissues and milk.
N° of Veterinary DrugsMatrixEquipmentReferenceLaboratory/Centre a
118MilkLC-QqQAguilera-Luiz et al. 2008 [5]Almeria University (Spain)
239Chicken muscleLC-QqQChico et al. 2008 [6]Barcelona University (Spain)
3>100MuscleLC-TOFKaufmann et al. 2008 [7]OFCA-Zurich (Switerland)
4>100MilkLC-TOFStolker et al. 2008 [8]RIKILT (The Netherlands)
5Ca 100Meat and other foodLC-TOFPeters et al. 2009 [9]RIKILT (The Netherlands)
6Ca 26Animal tissuesLC-QqQStubbings et al. 2009 [10]FERA (UK)
758MilkLC-QqQGaugain-Juhel et al. 2009 [11]EURL (France)
821MilkLC-QqQMartinez-Vidal et al. 2010 [12]Almeria University (Spain)
930MilkLC-Orbitrap, LC-Q-TOF, LC-QqQRomero-González et al. 2011 [5]Almeria University (Spain)
10>100Meat and other foodLC-OrbitrapKaufmann et al. 2011 [13]OFCA-Zurich (Switzerland)
11>60MeatLC-LTQ-OrbitrapHurtaud-Pessel et al. 2011 [14]EURL (France)
1259Milk and honeyLC-Q-TOFWang et al. 2012 [15]CFIA-Calgary (Canada)
1321MeatLC-QqQBittencourt et al. 2012 [16]LANAGRO (Brazil)
1424Milk and liverLC-QqQMartins et al. 2014 [17]LANAGRO (Brazil)
15>100MilkLC-Q-OrbitrapKaufmann et al. 2014 [18]OFCA-Zurich (Switzerland)
1639LiverLC-QqQFreitas et al. 2015 [19]INIAV (Portugal)
1723LiverLC-QqQMartins et al. 2015 [20]LANAGRO (Brazil)
18>100MilkLC-Q-OrbitrapWang et al. 2015 [21]CFIA-Calgary (Canada)
19>100Various foodLC-Q-TOFDasenaki et al. 2015 [22]University of Athens (Greece)
2076Bovine muscleLC-QqQDasenaki et al. 2016 [23]University of Athens (Greece)
2162Animal muscleLC-Q-OrbitrapMoretti et al. 2016 [24]IZSUM (Italy)
2262MilkLC-Q-OrbitrapMoretti et al. 2016 [25]IZSUM (Italy)
23>120Animal tissuesLC-QqQ/LC-Q-TOFAnumol et al. 2017 [26]USDA (USA)
24174Bovine tissuesLC-QqQLehotay et al. 2018 [27]USDA (USA)
2544SalmonLC-Q-TOFGaspar et al. 2019 [28]INIAV (Portugal)
a OFCA = Official Food Control Authority; CFIA:Canada Food Inspection Agency; FERA: The Food and Environment Research Agency; LANAGRO: Laboratório Nacional Agropecuário; INIAV: Instituto Nacional de Investigação Agrária e Veterinária; USDA: United States Department of Agriculture; IZSUM: Istituto Zooprofilattico Sperimentale dell’Umbria e delle Marche.
Table 2. Summary of the selected reactions transitions (SRM) monitored for the sixty-four targeted analytes.
Table 2. Summary of the selected reactions transitions (SRM) monitored for the sixty-four targeted analytes.
AnalyteRetention Time (min)Adduct (m/z)Precursor Ion (m/z)Product Ions (m/z)Collision Energy (eV)
1Sulfaguanidine2.85[M + H]+215.192.015
156.020
2Florfenicolamine3.20[M + H]+248.1230.110
130.130
3Sulfanilamide3.30[M + H − NH3]+156.092.012
108.110
Sulfanilamide-13C63.30[M + H − NH3]+162.098.113
114.113
4Desacetylcephapyrin 6.80[M + H]+382.1152.030
226.020
5Amoxicillin8.30[M + H]+366.1349.110
114.020
6Sulfadiazine8.50[M + H]+251.1108.026
156.015
7Sulfathiazole9.20[M + H]+256.092.128
156.015
8Cephapyrin9.45[M + H]+424.1292.120
152.030
9Sulfapyridine9.50[M + H]+250.1108.026
156.017
10Tildipirosin9.90[M+2H]++367.7281.220
98.118
11Sulfamerazine9.90[M + H]+265.1108.027
156.017
12Cefquinome10.00[M + 2H]++265.1134.220
199.120
13Cefacetrile10.15[M + Na]+362.0258.010
302.010
14Cefalonium10.50[M + H]+459.1337.010
152.020
15Lincomycin10.50[M + H]+407.2126.130
359.210
16Tulathromycin marker10.60[M + 2H]++289.0158.317
420.517
17Thiamphenicol10.60[M + H]+356.0308.020
229.020
18Epitetracycline10.60[M + H]+445.2410.220
392.130
19Trimethoprim10.70[M + H]+291.1261.130
230.130
20Marbofloxacin10.80[M + H]+363.1276.114
320.114
21Sulfamethazine11.10[M + H]+279.192.131
124.128
Sulfamethazine-13C611.10[M + H]+285.1186.117
22Epioxytetracycline11.35[M + H]+461.2426.120
337.130
23Norfloxacina11.50[M + H]+320.1231.239
282.129
24Tetracycline11.50[M + H]+445.2410.220
392.130
25Cefalexin11.70[M + H]+348.1158.010
174.120
26Oxytetracycline11.80[M + H]+461.2426.120
337.130
27Ciprofloxacin11.80[M + H]+332.1245.123
288.117
28Enrofloxacin11.90[M + H]+360.2245.026
316.119
Enrofloxacin -d511.90[M + H]+365.2321.418
29Tulathromycin11.90[M + 2H]++404.0158.120
116.120
30Danofloxacin11.95[M + H]+358.2283.124
340.122
31Cefazolin12.00[M + H]+455.0323.110
156.020
32Sulfamethoxazole12.10[M + H]+254.1108.128
156.016
33Difloxacin12.30[M + H]+400.1299.128
356.118
34Ampicillin12.30[M + H]+350.1106.120
160.020
35Sulfamonomethoxine12.30[M + H]+281.1108.128
156.016
36Florfenicol12.40[M + H]+358.0241.020
340.010
Florfenicol -d312.40[M + H]+361.0241.016
37Cefoperazone12.60[M + H]+646.1530.310
143.130
38Sarafloxacin12.60[M + H]+386.1342.118
299.126
39Epichlortetracycline12.85[M + H]+479.1444.120
154.030
40Neospiramycin13.40[M + 2H]++350.2160.110
174.120
41Chlortetracycline13.80[M + H]+479.1441.120
154.030
42Spiramycin14.05[M + 2H]++422.3702.410
174.120
Spiramycin -d314.05[M + 2H]++423.8174.020
43Sulfadimethoxine14.40[M + H]+311.1108.129
156.021
44Sulfaquinoxaline14.80[M + H]+301.192.130
156.021
45Oxolinic Acid15.00[M + H]+262.1216.029
244.018
46Ceftiofur15.10[M + H]+524.0241.020
126.030
Ceftiofur-d315.10[M + H]+527.0244.115
Metacycline15.15[M + H]+443.1426.216
47Gamithromycin15.40[M + H]+777.5158.139
619.732
48Tilmicosin15.70[M + 2H]++435.3695.520
174.130
49Doxycycline15.50[M + H]+445.2428.110
321.135
50Nalidixic Acida16.80[M + H]+233.1159.030
187.025
51Tiamulin17.10[M + H]+494.3192.120
119.030
52Penicillin G17.15[M + Na]+357.1198.120
182.020
Penicillin G-d717.15[M + Na]+364.0205.213
53Flumequine17.20[M + H]+262.1202.033
244.019
54Tylosina A17.40[M + H]+916.5174.136
772.528
55Erythromycin17.60[M + H]+734.5576.420
158.130
563-O-Acetyltylosin17.75[M + H]+958.5174.036
772.528
57Oxacillin18.20[M + Na]+424.1265.120
182.020
58Penicillin V18.20[M + Na]+373.1182.020
214.020
59Cloxacillin18.50[M + Na]+458.1299.020
182.020
60Valnemulin19.10[M + H]+565.4263.120
72.130
61Dicloxacillin19.20[M + Na]+492.0333.020
182.020
62Nafcillin19.30[M + H]+415.1171.134
199.113
63Tilvalosin19.45[M + H]+1042.6174.039
814.530
64Rifaximin20.60[M + H]+786.4754.320
736.330
a Acquired only in milk.
Table 3. Precision, recovery and matrix effect (muscle and milk).
Table 3. Precision, recovery and matrix effect (muscle and milk).
MuscleMilk
Analyte a,bCVr,pooled (%)CVRw, pooled (%)Rec (%)ME c (%)CVr,pooled (%)CVRw, pooled (%)Rec (%)ME c (%)
Sulfaguanidine5.81083−145.01461253
Florfenicol Amine3.26.185−222.53.894−13
Sulfanilamide6.68.974−355.81266−21
Desacetylcephapirin7.67.376−15.96.091−21
Amoxicillin4.56.364−15.85.889−10
Sulfadiazine5.18.286−184.88.674−16
Sulfathiazole5.28.083−65.28.371−15
Cephapirin6.97.8751121294−5
Sulfapyridine4.67.285135.89.567−13
Tildipirosin6.210 72−86.020875
Cefquinome6.17.297−4111978−16
Sulfamerazine3.85.989116.78.770−13
Cefacetrile121380−26222792−5
Cefalonium6.77.37838.78.793−6
Lincomycin4.37.283−122.34.49230
Epitetracycline6.29.266224.77.6965
Trimethoprim3.47.890−32.74.9944
Thiamphenicol101187−13131392−13
Tulathromycin marker5.68.681−138.72681−18
Marbofloxacin6.17.087−274.34.996−11
Sulfamethazine4.17.285−17.81468−13
Epioxytetracycline8.8136240131590−3
Norfloxacin----7.08.0943
Tetracycline6.39.471274.55.09140
Cefalexin6.28.46416.26.790−7
Oxytetracycline6.78.163104.45.2901
Ciprofloxacin7.58.481−256.17.595−10
Enrofloxacin4.76.693−164.15.999−1
Tulathromycin8.31669107.513945
Danofloxacin5.77.290−174.25.0973
Cefazolin8.49.083−8141494−11
Sulfamethoxazole7.79.788−166.76.784−24
Difloxacin5.07.493−153.34.296−13
Ampicillin6.68.068−29.41088−10
Sulfamonomethoxine4.67.48606.49.978−19
Florfenicol9.81690−17131394−28
Cefoperazone6.88.485−131618103−25
Sarafloxacin5.87.486−264.86.699−22
Epichlorotetracycline9.31371657.41110022
Neospiramycin7.81667127.3128613
Chlortetracycline5.87.769475.77.29248
Spiramycin8.41774204.57.69121
Sulfadimethoxine4.58.188−123.93.990−29
Sulfaquinoxaline6.17.786−264.64.891−38
Oxolinic Acid4.96.79742.66.696−3
Ceftiofur7.59.772−216.77.195−24
Gamithromycin5.67.194553.24.89826
Tilmicosin6.91088493.24.49538
Doxycycline7.09.56916.67.49510
Nalidixic Acid----3.66.094−3
Penicillin G7.99.085−2112139237
Tiamulin9.31388−212.54.198−5
Flumequine4.37.395−32.94.59552
Tylosin A7.71585292.54.89654
Erythromycin A4.98.689−274.71267−21
3-O-Acetyltylosin9.31688423.75.09553
Oxacillin6.01183−298.08.51001
Penicillin V7.91185−287.57.7996
Cloxacillin8.21184−279.812110−25
Valnemulin193175−262.75.410855
Dicloxacillin6.71081−25131399−10
Nafcillin5.08.284−75.05.499−19
Tylvalosin9.01993505.46.910190
Rifaximin9.01291334.89.6987
a Nalidixic acid and norfloxacin were not included in muscle method since these antibiotics were introduced later; b For valnemulin (muscle), cefacetrile (milk) and cefquinome (milk) the method can be used only for screening purposes (inadequate accuracy); c Values of matrix effect (ME) in bold are considered significant (>ǀ20ǀ%).
Table 4. Participation in Proficiency Test Schemes: comparison between methods.
Table 4. Participation in Proficiency Test Schemes: comparison between methods.
MethodLC-Q-OrbitrapLC-QqQ
Sample Code/YearMatrixAnalyteFound Concentration (µg·kg−1)Found Concentration (µg·kg−1)Consensus Value (µg·kg−1)Acceptability Range (µg·kg−1)
MI1432-A1/2014 aMilkSulfamethazine1449610357–150
MI1432-A2/2014 aMilkAmoxicillin5NDNot assigned-
M1435-A1/2014 aPig muscleSulfamethazine88756936–102
Pig muscleSulfadimethoxine32232712–42
M1433-A2/2014 aTurkey muscleCiprofloxacin5.555.63.2–8.1
Turkey muscleEnrofloxacin17315216092–227
MI1532-A1/2015 aMilkAmoxicillin16ND145.6–22
MI1532-A2/2015 aMilkSulfamethazine16513113475–191
MI1623-A1/2016 aMilkFlumequine911118847–129
MI1623-A2/2016 aMilkOxytetracycline93559149–132
MI1715-A2/2017 aMilkDanofloxacin91807439–109
484 (material C)/2018 bBovine MuscleMarbofloxacin178193170100–240
484 (material C)/2018 bBovine muscleOxytetracycline897910659–152
334/2019 bBovine muscleCiprofloxacin1010NAc-
334/2019 bBovine muscleEnrofloxacin8284NAc-
544/2019 bBovine muscleTylosin A5487NAc-
a Test Veritas, Padova, Italy; b RIKILT, Wageningen, Netherlands; c The final Report is not yet available.

Share and Cite

MDPI and ACS Style

Giusepponi, D.; Paoletti, F.; Barola, C.; Moretti, S.; Saluti, G.; Ianni, F.; Sardella, R.; Galarini, R. Transfer of a Multiclass Method for over 60 Antibiotics in Food from High Resolution to Low Resolution Mass Spectrometry. Molecules 2019, 24, 2935. https://doi.org/10.3390/molecules24162935

AMA Style

Giusepponi D, Paoletti F, Barola C, Moretti S, Saluti G, Ianni F, Sardella R, Galarini R. Transfer of a Multiclass Method for over 60 Antibiotics in Food from High Resolution to Low Resolution Mass Spectrometry. Molecules. 2019; 24(16):2935. https://doi.org/10.3390/molecules24162935

Chicago/Turabian Style

Giusepponi, Danilo, Fabiola Paoletti, Carolina Barola, Simone Moretti, Giorgio Saluti, Federica Ianni, Roccaldo Sardella, and Roberta Galarini. 2019. "Transfer of a Multiclass Method for over 60 Antibiotics in Food from High Resolution to Low Resolution Mass Spectrometry" Molecules 24, no. 16: 2935. https://doi.org/10.3390/molecules24162935

Article Metrics

Back to TopTop