Sei sulla pagina 1di 5

1340 SCHENCK & LAGMAN: JOURNAL OF AOAC INTERNATIONAL VOL 82, NO.

6, 1999

DRUGS, COSMETICS, FORENSIC SCIENCES

Multiresidue Determination of Abamectin, Doramectin,


Ivermectin, and Moxidectin in Milk Using Liquid
Chromatography and Fluorescence Detection
SCHENCK & LAGMAN: JOURNAL OF AOAC INTERNATIONAL VOL 82, NO. 6, 1999
FRANK J. SCHENCK1
U.S. Food and Drug Administration, Baltimore District Laboratory, 900 Madison Ave, Baltimore, MD 21201
LAWRENCE H. LAGMAN
U.S. Department of Agriculture, Agricultural Marketing Service, Eastern Laboratory, 645 Cox Rd, Gastonia, NC 28054

Abamectin, doramectin, ivermectin, and moxidectin drogenation of avermectin B1 (6). Doramectin is produced by
are macrocyclic lactones derived from soil dwell- mutational biosynthesis, feeding cyclohexanecarboxylic acid
ing actinomycetes, and are very effective against to a mutant strain of S. avermitilis (7, 8). Moxidectin is ob-
nematode, insect, and arthropod infestations. tained by the chemical modification of the milbemycin,
These compounds, known as endectocides, have nemadectin, which is produced by S. cyanogriseus (9). (See
been approved for use in beef cattle in the United Figure 1 for chemical structures.) An excellent review of the
States; however, they are currently not approved avermectins and milbemycins has been presented by McKel-
for use in dairy cattle. Abamectin, doramectin, lar and Benchaoui (10).
ivermectin, and moxidectin residues were isolated The avermectins and milbemycins are highly lipophilic
from milk by a series of liquid–liquid extraction and are stored in fat tissue, which acts as a reservoir, contribut-
steps, derivatized with trifluoroacetic anhydride, ing to their long-term persistence in the body (11). Ivermectin
and determined by liquid chromatography with flu- and moxidectin have been found in the milk of lactating ani-
orescence detection. Recovery studies were per- mals many weeks after dosing (12–14). None of the
formed in 2 laboratories. Recoveries of >80% 4 endectocides discussed above are approved for use in lactat-
(1–30 ng/mL) were achieved for all 4 compounds. ing dairy cattle in the United States (15).
Methods for the determination of ivermectin in milk have
been reported in the literature (16–21). Lagman (22) demon-
he avermectins and milbemycins are closely related strated that abamectin, doramectin, and moxidectin could be

T 16-membered macrocyclic lactones derived from


soil-dwelling actinomycetes of the genus Streptomyces.
They are extremely potent against nematode and arthropod
extracted from milk by using a method first developed for
ivermectin in milk. A multiresidue liquid chromato-
graphic (LC) method for the determination of abamectin,
species as well as acarine and insect plant pathogens. Their doramectin, ivermectin, and moxidectin in milk is described
use in the treatment of endoparasitic infections and here.
ectoparasitic infestations gave rise to the name endectocide.
The primary structural difference between the groups is that METHOD
the avermectins have a disaccharide moiety attached to the
13-position of the macrocyclic ring, whereas the milbemycins Apparatus
do not (1).
Eight avermectin compounds have been isolated by ex- (a) Centrifuge.—Equipped with rotors for handling 15
tracting the mycelia of S. avermitilis. These consist of 4 major and 50 mL centrifuge tubes.
“a” compounds and 4 minor (5–10%) “b” homologues, desig- (b) Centrifuge tubes.—Glass, 15 mL graduated and
nated A1a through B2b (2). Abamectin, a mixture of >80% 50 mL, with polyethylene stoppers.
avermectin B1a and <20% avermectin B1b, was first intro- (c) Ultrasonic bath.—Bransonic Model 2210 (Branson
duced as an agricultural pesticide and later as an animal Ultrasonic Corp., Danbury, CT) or equivalent.
endectocide (3–5). Ivermectin, a mixture of >80% (d) Liquid chromatograph.—Model 650-15 fluorescence
22,23-dihydroavermectin B1a, and <20% spectrophotometer, Series 410 LC pump, Model
22,23-dihydroavermectin B1b, is produced by the selective hy- ISS-100 autoinjector (Perkin-Elmer Corp., Norwalk, CT);
4.6 × 150 mm Chromegabond MC18 LC column (ES Indus-
Received January 29, 1999. Accepted by JM June 29, 1999. tries, W. Berlin, NJ) with Brownlee Newguard RP-18 guard
1 column (Rainin Instrument Co., Woburn, MA). Operating
Current address: U.S. Food and Drug Administration, 60 Eighth St,
NE, Atlanta, GA 30309. conditions: excitation wavelength, 364 nm; emission wave-
SCHENCK & LAGMAN: JOURNAL OF AOAC INTERNATIONAL VOL 82, NO. 6, 1999 1341

Figure 1. Chemical structures of abamectin, doramectin, ivermectin, and moxidectin.

length, 475 nm; injection volume, 100 µL; column tempera- Standards
ture, 30°C; solvent flow rate, 0.7 mL/min.
(a) Abamectin reference standard.—0.819% (w/w)
Reagents abamectin B1a and 0.054% (w/w) abamectin B1b in glycerol
formal (Merial Ltd., Three Bridges, NJ).
(a) Solvents.—LC grade acetonitrile, ethyl acetate, hexane, (b) Doramectin reference standard.—Pfizer Research
2,2,4-trimethylpentane (isooctane), and tetrahydrofuran (THF); Laboratories, Groton, CT.
USP ethanol; ACS reagent grade ammonium hydroxide. (c) Ivermectin reference standard.—1.38% (w/w)
(b) Water.—Deionized (Milli-Q Water System, Waters ivermectin B1a and 0.21% (w/w) ivermectin B1b in glycerol
Corp., Milford, MA) or equivalent. formal (Merck Sharpe and Dohme Research Laboratories,
(c) LC mobile phase.—Acetonitrile–THF–water Rahway, NJ).
(90 + 6 + 4; v/v/v). Add 100 mL acetonitrile, 40 mL water, (d) Moxidectin reference standard.—Agricultural Re-
and 60 mL THF to 1.0 L volumetric flask. Add acetonitrile to search Division, American Cyanimid Corp., Princeton, NJ.
volume and mix.
Preparation and Handling of Silylated Tubes
(d) Silylation reagent.—Sylon-CT (Supelco, Inc.,
Bellefonte, PA). Graduated centrifuge tubes (15 mL) were thoroughly
(e) N-methylimidazole.—99% pure (Aldrich Chemical cleaned, silylated with Sylon CT for 30 min, rinsed with tolu-
Co., Milwaukee, WI). ene and methanol, soaked with methanol for 30 min, and
(f) Trifluoroacetic anhydride (TFAA).—Pierce, Rockford, IL. rinsed with acetone. The tubes may be used for 2 months be-
(g) Derivatizing reagent.—Mix 4 mL acetonitrile with fore the silylation procedure is repeated.
2 mL TFAA immediately before use. Silylated Tube Cleaning
(h) Milk.—Raw milk was obtained from the U.S. Depart-
ment of Agriculture, Beltsville, MD, and stored at –70ºC. Ho- After use the tubes are soaked for several hours in methy-
mogenized, grade A milk was obtained from local grocery lene chloride and then in detergent solution. They are rinsed
stores and stored at 4ºC. with hot water, distilled water, and acetone. They must be
1342 SCHENCK & LAGMAN: JOURNAL OF AOAC INTERNATIONAL VOL 82, NO. 6, 1999

cleaned by hand. If they are machine washed, the silylation fer hexane to graduated 15 mL centrifuge tube, evaporate to
procedure must be repeated. <3 mL under stream of nitrogen, and adjust volume to 3.0 mL
with hexane. Add 4.0 mL acetonitrile, shake 1 min, centrifuge
Standard Solutions
4 min, transfer lower acetonitrile phase to 15 mL silylated cen-
(a) Stock standard solutions.—Solutions (100 µg/mL) of trifuge tube. Repeat acetonitrile extraction. Shake combined
abamectin, doramectin, ivermectin, and moxidectin in acetonitrile extracts with 1 mL hexane, centrifuge 4 min, and
acetonitrile were prepared and stored in actinic glassware at discard hexane. Evaporate acetonitrile to dryness under nitro-
–20°C. gen at 70°C.
(b) Mixed working standard solution (1.0 mg/mL).—From
each of the stock standard solutions 1.0 mL was added to a Derivatization
100 mL actinic glass volumetric flask. Acetonitrile was added
to volume. The solution was stored at –20°C. Less than 0.050 mL of residue should remain. (Note: The
presence of moisture, methanol, or excess milk matrix resi-
Sample Extraction due, caused by incomplete removal of the hexane layer during
Add 10.0 mL milk to 50 mL centrifuge tube. For recovery sample preparation, can interfere with the derivatization reac-
studies, add appropriate amount of working standard to milk, tion.) Reconstitute sample extract in 2.0 mL acetonitrile. Pre-
Vortex mix, and wait 15 min. Add 2.5 mL concentrated am- pare series of standards for standard curve by adding mixed
monium hydroxide, and mix. Add 10 mL ethanol, 10 mL ethyl working standard to 15 mL silylated centrifuge tubes. (Note: If
acetate, and 10 mL isooctane sequentially, shaking mixture methanolic working standards are used, they must first be
30, 30, and 20 s after each respective addition. Centrifuge evaporated to dryness.) Add acetonitrile to the 2 mL mark.
sample 4 min to separate liquid phases; transfer upper layer to Add 0.2 mL N-methylimidazole and Vortex mix.
second 50 mL centrifuge tube with pipette. Add additional Add 0.6 mL freshly prepared derivatization reagent to
10 mL ethyl acetate and 10 mL isooctane sequentially to first silylated tubes containing samples and standards. Stopper
tube, shaking mixture as above; repeat centrifugation. Com- tubes, Vortex briefly, and store in the dark. After 15 min, di-
bine top layers in second tube, and evaporate under stream of lute solutions to 5.0 mL with acetonitrile, Vortex mix, and
nitrogen at 70°C. Dissolve oily residue by adding ca 4 mL store shielded from direct light. Inject 100 µL of each sample
hexane, sonicating, and Vortex mixing. Quantitatively trans- and standard into LC system. Measure peak heights at reten-

Figure 2. Typical chromatograms of (A) control raw milk and (B) control raw milk fortified with 2.0 ng/mL abamectin,
doramectin, ivermectin, and moxidectin. Peak identities are (1) moxidectin, (2) abamectin B1b, (3) abamectin B1a,
(4) doramectin, (5) ivermectin B1b, (6) ivermectin B1a.
SCHENCK & LAGMAN: JOURNAL OF AOAC INTERNATIONAL VOL 82, NO. 6, 1999 1343

Table 1. Recovery of fortified abamectin, doramectin, ivermectin, and moxidectin residues from milk
Recovery, %a

Fortification, ng/mL Abamectinb Doramectin Ivermectinb Moxidectin

Homogenized milk

3 90.9 (9.4) 95.5 (8.5) 90.7 (13.6) 104.7 (23.7)


6 91.1 (17.4) 91.6 (20.3) 93.7 (21.2) 100.1 (15.3)
15 84.6 (10.7) 87.1 (12.1) 86.9 (14.5) 91.1 (11.9)
30 83.5 (3.1) 84.1 (4.4) 83.7 (7.0) 89.8 (7.2)
Raw milk

1 109.9 (3.3) 108.7 (5.2) 111.3 (3.6) 113.8 (17.4)


2 99.7 (0.7) 101.5 (1.7) 102.6 (1.7) 102.3 (2.4)
5 99.7 (2.9) 98.2 (2.7) 99.8 (2.8) 98.5 (2.4)
10 97.6 (2.4) 99.4 (2.0) 101.9 (0.6) 98.4 (1.8)
15 98.7 (2.3) 100.6 (1.8) 99.5 (1.5) 100.3 (0.6)
30 100.8 (2.3) 103.0 (2.1) 102.4 (2.1) 103.1 (2.3)

a
n = 4; values in parentheses are coefficients of variation.
b
Recoveries are for abamectin B1a and ivermectin B1a.

tion times of abamectin B1a, doramectin, ivermectin B1a, and of ivermectin in milk (18). The 4 lipophilic endectocides
moxidectin derivatives as indicated by standards. (abamectin, doramectin, ivermectin and moxidectin) were ex-
tracted from milk along with milk fat, and separated from fat
Generation of Standard Curves and Calculations
by acetonitrile/hexane partitioning.
For each of the 4 endectocides, calculate slope, a, and inter- Although all 4 of the endectocides have UV chromophores,
cept, b, of a standard curve by linear regression analysis of stan- the presence of coextracted milk matrix components pre-
dards based on peak height of the endectocide derivative peaks, cludes using UV detection at the low levels. Tway et al. (23)
using y = ax + b, where y = peak height and x = concentration first reported that fluorescent derivatives of ivermectin can be
of standard in ng/mL. Using peak heights of endectocide deriv- formed by reacting it with acetic anhydride (AA) at 95°C in
atives in the milk sample, calculate concentration injected into the presence of a base catalyst, N-methylimidazole (NMIM).
the LC system using the following equation: The AA–NMIM derivatization reaction results in many re-
agent by-products; thus further cleanup of the derivatized
Analyte injected into LC system, ng/mL = standards and samples is required. De Montigny et al. (24) re-
(peak height – b)/a ported that when trifluoroacetic anhydride (TFAA) was sub-
stituted for AA, the derivatization of avermectins took place in
Calculate concentration of analyte in milk sample as fol-
<30 s at 25°C. Because substantially fewer reagent
lows:
by-products were formed by the TFAA derivatization, fur-
Milk sample, ng/mL = ther cleanup of the derivatized standards and samples was
(analyte injected, ng/mL) × (V1/V2 ) or not required.
The TFAA derivatives of the 4 endectocides are very hy-
Milk sample, ng/mL = drophobic. Liquid chromatographic separation is usually
(analyte injected, ng/mL) × 0.5 achieved by reversed-phase octadecylsilica (C18) or
octylsilica (C8) columns with mobile phases containing a very
where V1 = final volume of derivatized extract (5.0 mL) and high percentage of organic solvent. Typically, mixtures of
V2 = volume of milk extracted (10 mL). methanol–water (95 + 5) or acetonitrile–water (97 + 3), with
flow rates ranging from 1.2 to 2.0 mL/min, are used. Even
Results and Discussion with these high percentages of organic modifier and relatively
high flow rates, retention times of 12–15 min for ivermectin
The liquid chromatographic method with fluorescence de- are common. We achieved similar retention times using a
tection is both specific and sensitive. The extraction and 0.7 mL/min flow rate with an acetonitrile–THF–water
cleanup is similar to the method presently used by the U.S. (90 + 6 + 4) mobile phase. The lower flow rate resulted in sig-
Food and Drug Administration (FDA) for the determination nificantly better chromatography, with a greater than 2-fold
1344 SCHENCK & LAGMAN: JOURNAL OF AOAC INTERNATIONAL VOL 82, NO. 6, 1999

increase in peak heights compared to acetonitrile–water or Parasitology, M.J. Howell (Ed.), Australian Academy of Sci-
methanol–water mobile phases at higher flow rates. Typi- ence, Canberra, Australia, p. 240
cal chromatograms of fortified and control milks are (4) De Chancet, G.C., Casey, R., Dixon, F.F., Besier, R.B., &
shown in Figure 2. Mitchell, R.K. (1988) Austr. Vet. J. 65, 85–86
Recovery studies of the 4 endectocides were performed in (5) Lasota, J.A., & Dybas, R.A. (1990) Acta Leiden. 59, 217–225
2 different laboratories. Recovery studies using homogenized (6) Chabala, J.C., Mrozik, H., Tolman, R.L., Eskola, P., Lusi, A.,
milk were first performed at one laboratory using an Peterson, L.H., Woods, M.F., & Fisher, M.H. (1980) J. Med.
Chem. 23, 1134–1136
acetonitrile–water (97 + 3) mobile phase. Recovery studies
(7) Dutton, C.J., Gibson, S.P., Goudie, A.C., Holdom, K.S.,
were later performed on raw milk, using the improved
Pacey, M.S., & Ruddock, J.C. (1991) J. Antibiot. 44,
acetonitrile–THF–water (90 + 6 + 4) mobile phase at the sec-
357–365
ond laboratory. Table 1 shows that recoveries ranging from 83
(8) Hafner, B.W., Holley, B.W., Holdom, K.S., Lee, S.E., Wax,
to 114% at 1–30 ng/mL levels were obtained for the R.G., Beck, D., McArthur, H.A.I., & Werner, W.C. (1991) J.
4 endectocides by the 2 laboratories. The CVs were lower for Antibiot. 44, 349–356
raw milk than for homogenized milk because of improved (9) Lanusse, C., Lifschitz, A., Virkel, G., Alvarez, L., Sánchez,
chromatography and the use of the acetonitrile–THF–water S., Sutra, J.F., Galtier, P., & Alvinerie, M. (1997) J. Vet.
mobile phase with raw milk samples. The limits of detection Pharmacol. Ther. 20, 91–99
(S/N = 3) for the 4 endectocides were ca 0.3 ng/mL when this (10) McKellar, Q.A., & Benchaoui, H.A. (1996) J. Vet.
mobile phase was used. Pharmacol. Ther. 19, 331–351
Two potential problems with the TFAA derivatization (11) Lanusse, C., & Prichard, R. (1993) Vet. Parasitol. 49,
have been reported. If any water is present in the injection ma- 123–158
trix, excess TFAA may be hydrolyzed to trifluoroacetic acid, (12) Toutain, P.L., Campan, M., Galtier, P., & Alvinerie, M.
resulting in breakdown of the derivative (25, 26). Therefore, (1988) J. Vet. Pharmacol. Ther. 11, 288–291
the derivatized standards and samples were diluted with (13) Alvinerie, M., Sutra, J.F., & Galtier, P. (1993) Vet. Res. 24,
acetonitrile and not with mobile phase which contains water. 417–421
De Groodt et al. (27) reported that TFAA derivatives of (14) Alvinerie, M, Sutra, J.F., Lanusse, C., & Galtier, P. (1996)
ivermectin are unstable in the presence of light and will un- Vet. Res. 27, 545–549
dergo 50% degradation in 3 h when exposed to daylight. Al- (15) Code of Federal Regulations (1998) Title 21, section 556,
though some degradation of doramectin TFAA derivatives Washington, DC
was noted after 3 h when exposed to laboratory fluorescent (16) Alvinerie, M., Sutra, J.F., Galtier, P., & Toutain, P.L. (1987)
lighting, no degradation of the TFAA derivatives of the Ann. Rech. Vet. 18, 269–274
4 endectocides was observed over 6 h when the derivatives (17) Chiou, R., Stubbs, R.J., & Bayne, W.F. (1987) J.
were shielded from direct light. Chromatogr. 416, 196–202
(18) Kijak, P.J. (1992) J. AOAC Int. 75, 747–750
(19) Heller, D.N., & Schenck, F.J. (1993) Biol. Mass Spectrom.
Acknowledgments
22, 184–193
(20) Schenck, F.J. (1995) J. Liq. Chromatogr. 18, 349–362
Funding for the work performed at the Agricultural Mar-
(21) Dusi, G., Curatolo, M., Fierro, A., & Faggionato, E. (1997)
keting Service Laboratory was provided by the USDA Pesti- Ital. J. Food Sci. 9, 337–342
cide Data Program. (22) Lagman, L. (1995) Poster No. 14-E-002, 109th AOAC IN-
TERNATIONAL Annual Meeting, Nashville, TN
References (23) Tway, P.C., Wood, J.S., & Downing, G.V. (1981) J. Agric.
Food Chem. 29, 1059–1063
(1) Shoop, W.L., Mrozik, H., & Fisher, M.H. (1995) Vet. (24) De Montigny, P., Shim, J.S.K., & Pivichny, J.V. (1990) J.
Parasitol. 59, 139–156 Pharm. Biomed. Anal. 8, 507–511
(2) Albers-Schonberg, G., Arison, B.H., Chabala, J.C., Douglas, (25) Rabel, S.R., Stobaugh, J.F., Heinig, R., & Bostick, J.M.
A.W., Eskola, P., Fisher, M.H., Lusi, A., Mrozik, H., Smith, (1993) J. Chromatogr. 617, 79–86
J.L., & Tolman, R.L. (1981) J. Am. Chem. Soc. 103, (26) Payne, L.D., Hicks, M.B., & Wehner, T.A. (1995) J. Agric.
4216–4221 Food Chem. 43, 1223–1237
(3) Tahir, M.S., Holroyd, G., & Copeman, D.B. (1986) in Para- (27) De Groodt, J.M., Wyhowski De Bukanski, B., & Srebrnik, S.
sitology—Quo Vadit, 6th International Congress of (1994) J. Liq. Chromatogr. 17, 1416–1426

Potrebbero piacerti anche