Ivermectina y Abemectina
Ivermectina y Abemectina
Ivermectina y Abemectina
Campbell
Editor
Ivermectin and
Abamectin
With 66 Illustrations
Springer-Verlag
New York Berlin Heidelberg
London Paris Tokyo
William C. Campbell
Merck Institute for Therapeutic Research
Rahway, N.J. 07065
98765432 1
Dedicated
mectins has been obtained by MSDRL scientists, and they are therefore
in a good position to review the data. The toxicological and environmental
safety data that have been accumulated within our laboratories have been
made available to governmental regulatory agencies and other interested
parties in the form of official reports, but have seldom appeared in the
scientific literature. In other disciplines, much information already has
appeared in the scientific literature; but here, too, a large proportion of
the research has been carried out by, or in collaboration with, our own
scientists-and the names of those individuals inevitably came to mind as
prospective authors of review chapters. There was, moreover, a practical
advantage in the preponderance of MSDRL authors: the chapters could
be prepared synchronously. The editor knows, from hard-won experi-
ence, the difficulty of getting manuscripts from far-flung scribes with no
obligation other than a cheerfully and hastily given promise to write a
chapter for a particular volume. In some multiauthored books, a chapter
written by a conscientious contributor may be older by many months, or
even by a few years, than one written by some hopelessly delinquent
contributor. In the present case, the editor has been blessed with
colleagues who have been willing to make an extraordinary effort to bring
their labors to fruition at the same time. This book, then, comes close to
the objective of capturing a likeness of its subject at one instant in time.
Except in the context of synthetic and microbiological chemistry
(which must of necessity deal with structural diversity), this volume deals
almost exclusively with ivermectin and abamectin-not with the ex-
tended family of the avermectins or with the related family known as
milbemycins.
The expression of dosage in terms of micrograms (f.Lg) per kilogram of
body weight has become commonplace in accounts of the efficacy of
these compounds, and this form is generally used in this book. Toxicolog-
ical studies, however, usually deal with mUltiples of milligrams, rather
than fractions, and in that context dosages are expressed in terms of
milligrams (mg) per kilogram of body weight.
The chapters of the book have been arranged in three groups. The first
deals with the basic chemical, biochemical, and microbiological aspects;
the second deals with pharmacological, safety, and environmental
aspects; and the third covers practical use of the compounds as antipar-
asitic and pesticidal agents. In the case of human application, however,
the available safety data have emerged from the clinical experience, and
are to be found in Part III.
William C. Campbell
Acknowledgments
Preface vii
Part I
Chapter 1. Chemistry
M.H. Fisher and H. Mrozik 1
Chapter 4. Biosynthesis
S.T. Chen, O.D. Hensens, and M.D. Schulman 55
Part II
Chapter 6. Toxicology
George R. Lankas and Lea R. Gordon 89
Part III
Appendixes
J. DI NETTA, J.D.
Department of Regulatory Affairs, Merck Sharp & Dohme Research
Laboratories, Rahway, New Jersey 07065
H. MROZIK, Ph.D.
Department of Synthetic Chemistry Research, Merck Sharp & Dohme
Research Laboratories, Rahway, New Jersey 07065
NATURAL A VERMECDNS
ORS
components a : R26= C2 H5
components b : R26= CH 3
ARAMECDN
Abamectin contains at least 80% of avermectin B 'a and not more than 20% of avermectin B,b.
Avermedin R..
C48Hn O ,4 ; mol wt: 872. otT-white powder; [alo + 55.7 ± 2° (c = 1.06 in chloroform); uv max
(methanol): 237, 243, 252 nm (E 29, 120; 31,850; 20,510).
Avermedin Rib
OH
Semisynthetic derivative of the avermectins. q.v. Ivermectin contains at least 80% of 22.23-
dihydroavermectin Bla and less than 20% of 22.23-dihydroavermectin BIb. Off-white powder;
[aID + 71.5 ± 3° (c = 0.755 in chloroform); uv max (methanol): 238. 245 nm (f; 27.100; 30.100).
Composeat 8110
Chemistry
M.H. Fisher and H. Mrozik
""
DRS
A2b
CH3
CH3
C2HS
CH3
,
-CH2-CH-
OH
-CH:rCH-
OH
B20
B2b
H
H
C2HS
CH 3
,
-CH2-CH-
OH
-CH 2-CH-
-CH~H
127
CH~+
1 COOH
<71
o CH
OR 3
275/261
~
CH 3 ~CH3 3
HO0 CHz
HOWCHs- Oto......H +H
CH3
169 111 179
FIGURE 1.2. Mass fragments ofavermectins. (G. Albers-SchOnberg, B.H. Arison,
J.e. Chabala, A.W. Douglas, P. Eskola, M.H. Fisher, A. Lusi, H. Mrozik, J.L.
Smith and R.L. Tolman, J. Am. Chern. Soc. 103, 4216 [1981]).
1. Chemistry 5
28 12.0 12.1
26a 12.9 12.4
14a 15.1 15.1
24a 16.4 17.4
6' 17.7 17.7
6" 18.4 18.4
4a 19.9 19.9
12a 20.2 20.2
ICHJ 8
27 27.5 27.3
24 30.6 31.2
16 -34.3 34.5
2' -34.3 34.2
2" -34.3 34.2
26 35.2 35.5
18 36.6 36.9
12 39.8 39.7
20 40.5 41.2
2 45.7 45.7
-CHz- 6
>CH- 4
I(CH 2 /CH) 10
, /0-
C
/ '0-
I(CHx°:z) 3
15 1ll8.1 118.1
3 118.4 118.3
9 120.4 120.4
10 124.8 124.7
23 127.9 28.1
14 135.2 135.0
22 136.2 35.7
4 137.9 137.8
11 -138.0 138.0
8 139.7 139.6
-cH = C 7
>C=C 3
I(CHx = C) 10
~o-
-C 173.6 173.8
'0-
I(C02) 1
overall I 48
8,9 - Z
H:'x5
· ."0x5
/
3
3
3 1,_
He·"
3 0".
UV LIGHT
~
10,11 - Z
III. Chemistry
A. HYDROXY GROUP CHI\RACTERISTICS
O~H
Clb7""--o~tb~Oi CIb
HO~" 0"
ClbO
CHI'
I,
HtSO
(CHs'ICHOH
l%
18"c.J6h
CIHs H
"
Rs %HtS04
~
CHsOH
lS"C)6h
H ):--CHs
Rn' s
18"C,16h
Rs Rs
Monosocchoride Aglycone
RS'OCHs 01 OH
Ru=OHOIH
C. OXIDATIONS
E. RADIOLABELED DERIVATIVES
Because ivermectin (= 22,23-dihydroavermectin B\) is obtained by
catalytic reduction of avermectin B\ (vide infra), the same procedure
using tritium gas conveniently affords tritiated ivermectin (22,23- 3[H]-
xS'
H:" "'0xS'
J
He
I"
-'I
RO",
3 0",
FIGURE 1.6. Avermectin B\ 8,9-oxide and its hydrolysis to the 8,9-diol. (Adapted
from Smith 1985.)
4,OCH3 C2H!lJi 4'OCH3
H "C:--CH3
~-H .,' .CH3
OCH3~~1 CH3 0~HCH3
CH3~ OJ:H 0'''fi3I4~ 'H CH3~ oJ.-: H 0''''r;3f4~
HO~ Iii HO~· li2
CH30 CH30 CHi
H2 (1 ATM)(Ph3PbRhCI
OH
~\
0,\
CH3
FIGURE 1.7. Synthesis of ivermectin by selective hydrogenation of avermectin B 1 • (Adapted from Chabala et aI. 1980.)
1. Chemistry 15
F. ALKYLATIONS
G. INTERCONVERSIONS OF A VERMECTINS
H. GLYCOSIDE SYNTHESES
Avermectin aglycones, monosaccharides, and the naturally occurring
disaccharides themselves have been further modified by attaching various
sugars to the different hydroxy groups. Most of these methods have used
I-bromo sugars via the Konigs-Knorr procedure (Fisher and Tolman
1979, 1980). More recently, the use of I-phenylthio and I-fluoro sugars
has resulted in better yields of those glycosides (Nicolaou et al. 1984).
There are also derivatives of avermectin B\ containing fluoro groups in
the sugars (Bliard et al. 1987). A mixture of the two anomeric methyl
glycosides of oleandrose is obtained by acid methanolysis of avermectins,
and these have been converted to L-oleandrose (Els, Celmer, and Murai
1958) and the I-phenylthio glycoside (Nicolaou et al. 1984). The 4-a-L-
oleandrosyl-L-oleandrose disaccharide was obtained through an oxida-
tive degradation of avermectin B\ (Hanes sian et al. 1987). L-Oleandrose
and the 4-alfa-L-oleandrosyl-L-oleandrose disaccharide was also ob-
tained by total synthesis (Wuts and Bigelow 1983; Danishefsky et al.
1987).
16 M.H. Fisher and H. Mrozik
IV. Structure-Activity
The avermectins, when administered orally to sheep, demonstrate activ-
ity against gastrointestinal parasites. A study was done to compare the
anthelminitic activities of the principal naturally occurring avermectins,
the monosaccharides and aglycones, and some reduced derivatives
(Chabala et al. 1980). The data are shown in Table 1.3.
In general, compounds of the B-series, containing a 5-hydroxy group,
are more potent than those of the A-series, containing a 5-methoxy group.
The I-series and 2-series have similar potency against many parasites, but
the 2-series compounds showed some important deficiencies which made
them less interesting. For example, avermectin B2 was inactive against
adult Haemonchus contortus in sheep at 0.1 mg/kg per os. Reduction of
the 22,23-0Iefin had only a small effect on potency but conferred sufficient
overall improvement in spectrum and safety that 22,23-dihydro avermec-
tin B) was selected for commercial development under the nonproprie-
tary name ivermectin. The monosaccharides were two to fourfold less
active than the disaccharide, and dihydroavermectin B) aglycone was
thirtyfold less potent.
A study of the activities of various acylated avermectins against
Trichostronglyus colubriformis in gerbils is shown in Tables 1.4 and 1.5
(Mrozik et al. 1982a).
The 4"-O-acetates have the same potencies as the un substituted com-
TABLE 1.4. Derivatives of avermectin A2a and B2a and anthelmintic activity
against trichostrongylus colubriformis in gerbils.
Anthelmintic
No. ~" R2 Rn act.'
1 H CH l H 0.005
3 H H H 0.0125
4 CHlCO CH3 H 0.0625
5 H CH 3 CH 3CO 0.25
6 CH 3CO CH3 CH3CO 0.5
10 CH3CO CH3CO CH 3CO 0.5
Reprinted from H. Mrozik et al. 1982a, by permission of The American Chemical Society.
Anthelmintic
No. ~" Rs actb
2 H H 0.025
7 CH3CO H 0.031
8 H CH3CO 0.125
9 CH3CO CH3CO 0.25
12 H SI(CH3hC(CH 3h >2.5
13 Si(CH3hC(CH3h Si(CH3hC(CH3)3 >2.5
14 (CH3hCCO Si(CH3hC(CH3)3
15 (CH3)3 CCO H 0.5
16 CH3(CH2)6CO Si(CH3hC(CH3)3
17 CH3(CH2 )6CO H 0.125
18 CCl3CH2OOCCH2CH2CO Si(CH3hC(CH3)3
19 CCl3CH2OOCCH2CH2CO H
20 HOOCCH2CH2CO H 0.03
21 (4-N02C6H 4O)CO Si(CH3hC(CH3)3
22 H 2NCO Si(CH3hC(CH3)3
23 H 2NCO H 0.025
24 (CH3hNCO Si(CH3hC(CH 3h
25 (CH3hNCO H 0.06
26 CH 3CONHCH2CO Si(CH3hC(CH 3h
27 CH 3CONHCH2CO H 0.025
a From Mrozik et al. 1982a.
b Minimal doses (mg/kg needed to remove > 83% of the worm burden.
TABLE 1.6. Anthelmintic efficacy by oral administration in experimentally infected sheepa.
H.c. O.c. C.o.
See tables 4 & 5 Dose No. of
treatment mg/kg sheep EL4 Adult EL4 Adult T.a. T.c. EL4 Adult Oe.c.
none 6 (7W (442) (1421) (1137) (2852) (4110) (193) (1761) (61)
1 0.1 3 3c 3 0 3 3 3 1 0 3
0.05 3 2 2 0 2 2 2 0 2
0.025 3 0 0 2 0 0 0 2
4 0.25 3 3 3 3 3 3
5 0.25 1 0 0 0 2 1
6 0.25 1 3 2 1 2 2 3
2 0.1 3 3 3 3 3 3 3 3 3 3
0.05 3 3 3 3 3 3 3 3 3 3
0.025 3 3 3 1 2 2 2 0 1 3
7 0.1 3 3 2 3 3 3 3 2 3
0.05 3 3 0 3 3 3 2 0 3
0.025 3 3 0 3 1 3 2 0 3
9 0.25 1 3 3 3 3 3 3
0.15 2 3 3 3 3 3 3
20 O.lsc 3 0 2 3 3d 3d 3
a H.c. = Haemonchus contortus; O.c. = Ostertagia circumcincta; T.a. = Trichostrongylus axei; T.c. = Trichostrongylus colubriformis; C.o. = Coop-
eria oncophora; Oe.c. = Oseophagostomum columbianum.
b Geometric mean of the number of worms per untreated experimentally infected lamb, representative of' 'typical" infections encountered under standard
experimental procedures.
c 3 = > 90% efficacy; 2 = 60-89% efficacy; I = 20-59% efficacy; 0 = 0-19% efficacy.
d C. curticei.
20 M.H. Fisher and H. Mrozik
It has been reported elsewhere (Fisher 1985) that the aromatic deriv-
ative of avermectin B] is devoid of biological activity.
A comparison of the activities ofavermectins in anthelmintic, miticidal,
and insecticidal assays has been reported Mrozik 1985b). The results are
shown in Table 1.8.
These data illustrate the complex structure-activity relationships
among the avermectins when measured over several species. For exam-
ple, while 22,23-dihydroavermectin B] monosaccharide is three to four-
fold less active than avermectin B] against helminths, it is sixteen fold
more active than avermectin B] against the southern armyworm.
REFERENCES
Tway PC, Downing GV, Slayoack JRB, Rahn GS, Isensee RK (1984) Confirma-
tory assay for ivermectin in cattle tissue using chemical ionization
mass spectrometry/mass spectrometry (MS/MS). Biorned. Mass Spectrorn.
11:172-176
Tway PC, Wood JS, Downing GV (1981) Determination of ivermectin in cattle and
sheep tissues using high-performance liquid chromatography with fluorescence
detection. J. Agric. Food Chern. 29:1049-1063
Wuts PGM, Bigelow SS (1983) Total synthesis of oleandrose and the avermectin
disaccharide, benzyl alfa-L-oleandrosyl-alfa-L-4-acetoxyoleandroside. J. Org.
Chern. 48:3489-3493
CHAPTER 2
is described as having the brownish-gray spore mass color, smooth spore surface,
spiral sporophore structure, production of melanoid pigments and cultural
and carbon utilization patterns that together comprise the distinctive species
S. avermitilis.
The culture was named Streptomyces avermitilis because of its ability to
produce anthelmintic activity.
"All cultures were grown on nutrient agar + 0.2% yeast extract except Streptococcus
faecium MB-2820 and Streptococcus agalactiae MB-2875, which were grown on brain heart
infusion agar. Incubation was at 25° or 37° C.
b Filter paper discs (6.2 mm) were dipped into whole broth and applied to the assay plates.
C Filter paper discs (6.2 mm) were dipped into acetone solutions of abamectin. The disks
REFERENCES
I. Introduction
II. Process Improvement-Shake Flask Scale
A. Culture Development
1. Mutational Procedures
2. Isolation/Detection of Mutants
a. morphological variants
b. nonproducers
c. auxotrophs
d. altered structural mutants
e. compositional mutants
f. superior producers
3. Cross-Feeding Studies
4. Cultural Genealogy
B. Seed Media Development
C. Production Media Development
1. Medium Constitutent Substitutions
2. Medium Optimization Studies
a. titration studies
b. surface response methodology
III. Process Scale-Up
A. Fermentation Kinetics: Stirred Tanks
B. Model to Predict Cell Mass On-Line
C. Oxygen Transfer
1. Correlation of Power Input to Oxygen Transfer Coefficient
2. Use of Hydrofoil Impellers to Improve Oxygen Transfer
Coefficient
D. Strategy Used for Successful Scale-Up
IV. Downstream Extraction
34 Mary Nallin Omstead, Louis Kaplan, and Barry C. Buckland
I. Introduction
Streptomyces avermitilis was originally isolated from soil as described in
Chapter 2. The first fermentations of the culture used a standard screening
approach: a loopful of slant growth was inoculated into a seed flask which
was incubated and then transferred into two types of production flasks.
Seed cultures were grown for 48 to 72 hours in baffled flasks containing
50 ml seed medium I (Burg et al. 1979). Production flasks were inoculated
by transferring 2.0 ml of seed growth into 40 ml production medium A
(Burg et al. 1979) and incubated for 4 days. The production cultures were
then harvested and assayed for activity as indicated in Chapter 2.
This chapter describes the development of an optimized process for the
large-scale fermentation of Streptomyces avermitilis, beginning with the
original strain. Improvements in the process involved mUltiple pa-
rameters-including strain improvement, seed and production media
development, and modification of physical fermentation parameters (such
as aeration, temperature, pH, and length of incubation). Furthermore,
scaling up the avermectin process to large fermentation tanks demanded
the optimization of yet another set of parameters-such as oxygen
transfer and media sterilization.
demonstrated by the low level of kill and the low frequency of morpholog-
ical variants. EMS is also a less effective mutagen for other organisms
(Sega 1984). In all procedures using alkylating agents the mutagen either
was removed from the suspension by centrifugation or was inactivated by
adding 10% sodium thiosulfate to the reaction mixture.
Treating S. avermitilis with these alkylating agents resulted in various
types of mutants. Morphological variants were easily detected. These
survivors consisted of the following types: non sporulating (i.e., "bald")
colonies, sporulating colonies which displayed altered spore pigmenta-
tion, colonies which failed to produce the melanin pigment typical of the
S. avermitilis parent, and colonies which displayed various combinations
of these characteristics. Mutants categorized within each type of variant
class exhibited a range in the ability to produce avermectin: many
exhibited lowered production as compared to the parental type, while
some had become complete nonproducers. Auxotrophic strains were also
detected and purified. They, too, varied in the amount of avermectin
produced, although generally their titers were significantly reduced.
Other mutants produced structurally modified avermectin components,
including the aglycone or monosaccharide avermectins (Ikeda, Kotaki,
and Omura 1987; Schulman et al. 1985); components which no longer
contained completely closed furan ring structures (Goegelman, Gullo, and
Kaplan 1983); and des-methyl avermectins (either on the macrolide ring
or on the oleandrose units) (Schulman etal. 1985; Schulman et al. 1986).
Once again, these mutants produced reduced total avermectin com-
ponents. In addition, other mutants were isolated which synthesized all
the avermectin components (a total of 8 structural types) (Miller et at.
1979) but produced them in different ratios than the original parental
strain.
Treating various parental strains by the procedures described resulted
in the accumulation of many mutants of each type. They were subse-
quently tested as to whether any of them were capable of cross-feeding
each other. These tests were done either by cofermenting the mutant
strains in a mixed fermentation or by feeding to one fermentation an
extract prepared from another fermentation broth. The cofermentations
were generally difficult to interpret, although they were adequate in
evaluating whether strains that had independently lost the ability to
produce melanin could regain that function if cocultured. Experiments
that involved feeding one strain with an extract from another strain
proved to be more useful. This type of feeding study enabled us to
categorize our nonproducing mutants into several classes based on their
ability to convert a given substrate (e.g., an aglycone avermectin) into an
easily distinguishable product (in the example cited, a standard avermec-
tin). In this fashion, it was possible to differentiate nonproducing mutants
even further.
There remains one final type of mutant, that which is most important to
3. Process Development and Improvement 37
1979). This set specified a seed medium (II) grown for 24 to 48 hrs, with
subsequent transfer to production medium (B) which fermented at 28° C
for 5 to 6 days. As the S. avermitilis strain was being developed, we
wanted to be sure that the fermentation conditions (including media
composition, temperature, aeration, and length of fermentation) would
not become limiting to the new, improved strains. To start, we examined
the initial seed medium. As reported (Burg et al. 1979), seed medium II
contained distiller's solubles (a particulate raw material), lactose, and
Ardamine PH. Our first manipUlation of this medium involved testing
the effect of various carbon sources. Of those, cerelose is optimal for
S. avermitilis in this medium. Seed medium II is one in which cerelose
was the primary source of carbon. As shown in Table 3.3, this single seed
medium change, combined with superior mutants (note culture improve-
ment in Table 3.3), resulted in five- to elevenfold improvement in
avermectin production. The original seed medium (II) demonstrated
about the same degree of improvement (six to tenfold) based on improved
cultures alone. However, simultaneously modifying the seed and the
production media produced even better results. Seed medium III, com-
bined with production medium C, resulted in sixteen- to twenty-onefold
improved avermectin production, whereas seed medium II with produc-
tion medium C netted only thirteen- to fourteenfold improvement.
Therefore, the combined effects of culture improvement and modification
a: Values shown are total fold improvements in B1(a+b) titers, including increases
due to the following factors: strain improvement, seed and production media
modifications, and changes in the physical fermentation parameters
b: Not determined
3. Process Development and Improvement 39
As the seed medium was being developed, so too was the production
medium. Early studies involved classical titration experiments: the
concentration of a given component of production medium B (Burg et al.
1979) was varied while the remaining ingredients were held constant. In
this approach, one looks for the occurrence of maxima (in this case,
highest avermectin titers) as they are collected from tests of each
ingredient assessed independently (Box, Hunter, and Hunter 1978). It
was exactly this approach that led to the development of production
medium C from production medium B (Table 3.3). Individual ingredients
of production medium B were titrated while the remaining ingredients
were maintained at constant levels. Concentrations of each individual
component that resulted in maximum avermectin production were then
simply combined to yield the composition of production medium C. As
seen in Table 3.3, combining any of the seed media and any of the cultures
fermented in production medium C increased the avermectin titer com-
pared to that obtained under the identical conditions in production
medium B. Focusing on culture 5 (Table 3.4), avermectin titers improved
by a factor of 24 when fermented in production medium C. Obviously,
this approach does have merit; its use can have a definite positive impact
on process development.
We then pursued a somewhat different but equally "classical" ap-
proach to medium development. These tests substituted various nitrogen
sources for the peptonized milk ingredient in medium C. Protein sources
such as soy, pork, cottonseed, and fish, as well as soybean and cot-
tonseed meals, were substituted based on their nitrogen equivalence. The
best avermectin titer obtained from any of these substituted media was
only 42% of that obtained from the control medium. The avermectin
3. Process Development and Improvement 41
a: Values shown are total fold improvements in B 1(a+b) titers including increases due to the
following factors: strain improvement, seed and production media modifications, and
changes in the physical fermentation parameters
b: Not determined
G (41X) than in medium E (56X). Obviously, media G and J had not been
optimized for use with this culture. In general, these data definitely prove
that production medium development, coupled with culture improvement
and modifications in the physical fermentation parameters, can and does
result in improved processes for the fermentation of secondary metabo-
lites.
Figure 3.1 illustrates the shake flask fermentation of culture 8 in
production medium J. Avermectin production began sometime between
the second and third day of the fermentation, after logarithmic growth had
ceased. Although avermectin production has been reported to be catabo-
lite repressible (Ikeda, Kotaki, and Omura 1987; McCann-McCormick et
al. 1981), using the cultures and conditions described herein avermectin
production was unaffected by glucose. Neither ammonia nor pH levels
rose until relatively late in the fermentation cycle, indicating that the
culture had not lysed. Now that we had developed superior cultures and
25 I
"- 20
-. "'- ......
~ DCW .".",. .......
-------- ....
- - .............. .,..,..,....
7
.c ... ~-----
!
",.
,!!t15
..
C
~
_ 10
I;:)
:z:Do
U 5 5
~
Q
0
r-----____________________________________ ~4
100
80
..........................
80
~
c
;:)
•... ·······~Bl
40 ...........
20 ......................
....
0
............
0 20 40 60 80 100
Percent of Cycle
FIGURE 3.1. Avermectin formation and growth of Streptomyces avermitilis in
shake flasks.
44 Mary Nallin Omstead, Louis Kaplan, and Barry C. Buckland
20
/ -
[0
Cii 10 $:
--
OJ
Titer 8 1
~ ()
0
0
5
L-__~~~____L-__~__~____L -_ _~_ _~ 0
25 50 75 100
Percent of Cycle (%) --..
stirred tanks is similar to that obtained in the shake flask process. This
presents a special challenge for downstream extraction; the desired B'a
and BIb components must be separated during the purification process.
An additional challenge to the purification is that S. avermitilis also
synthesizes lipids in large amounts. These consist primarily of a mixture
of triglycerides (Metz et al. 1988) possessing C14-C17 free fatty acids.
Where t time
OUR oxygen uptake rate (mmoles/lIh)
Y o/x ratio of total oxygen consumed to cell concentration
X = cell concentration (g dry cell weightll)
Therefore the function OUR· t::.. t denotes the total oxygen consumed,
easily calculated by on-line computer summation of the oxygen uptake
rate with respect to time.
Assuming that there is a constant yield coefficient between cell mass
and total oxygen consumed during this time period, it is possible to extend
this model further by dividing the oxygen uptake rate by the total oxygen
consumed; this provides an estimation of the specific growth rate of the
culture (gram increase in cell mass/ total cell mass in grams/hour). Using
the power of the minicomputer, this estimation can be done on-line.
1 dx OUR
f.L=--=
x dt ±
t=O
OUR. At
:£
-
.5
~ o Tolol 02 (Moles/L) (J d.c.w.
'*
.3 10 .2'
cC j
::::r
.....til
.2 ~
&
CP
-
(5
~
C\I
0 .1
if 5
(5 d~
~ (y/
00 10 20 30 40
Hours
FIGURE 3.4. On-line estimation of cell concentration and specific growth rate
(from Buckland et al. 1985).
3. Process Development and Improvement 47
c. OXYGEN TRANSFER
As the cell mass increases, the cells' mycelia begin to tangle, making the
broth viscous (Figure 3.6). This increase in viscosity adversely affects
oxygen transfer. It has been determined experimentally (Buckland et al.
1988) that the oxygen transfer coefficient is approximately proportional to
the reciprocal of the square root of the viscosity. This effect limits the
maximum achievable cell concentration and therefore the reactor's
volumetric productivity. Consequently, the study of oxygen transfer
efficiency was especially important for this process.
Computer-controlled, highly instrumented fermentors can calculate
on-line a number of different parameters for a single fermentation batch.
In one series of experiments (Buckland et al. 1988), the performance of a
large-diameter axial flow hydrofoil impeller was compared to that of a
conventional Rushton radial flow impeller. Two fermentations were run
side by side and the oxygen uptake rate profiles for the 2 different agitator
~,,
,,
"
~\ •• A. .. Itt
i .6 '.....
.~ \
.,.
\,.," \
A
"
i .4
·0
....'
...........
)a. ...,
T+4C
'A..........lt'·... -.........• ...............~..
ict .2
\ T
·o·· .. ··~ .. ~ ......~ .. ~...... ~ ..•.. +.·~ .. ~ .. +.~ ..
o~------~------~--------------~------~------~
o 2 6 8 10 12
Hours
FIGURE 3.5. Specific growth rate (estimated on-line) for two fermentations at
different temperatures. Standard temperature (T) and standard temperature plus
4° C (T + 4C) (from Brix, Drew, and Buckland 1988).
48 Mary Nallin Omstead, Louis Kaplan, and Barry C. Buckland
Brookfield Viscometer
--
G-
o
Spindle LVT#2
1000
20 40 50 60 70
types are compared (Figure 3.7). The dissolved oxygen profiles are also
similar (Figure 3.8). The on-line calculation of the oxygen transfer
coefficient (KLa) is made using the data from Figures 3.7 and 3.8; not
surprisingly, the profiles are also similar (Figure 3.9). The most striking
difference is in the relative power consumption of the 2 different agitator
types (Figure 3.10). The results of this study (and others) demonstrated
that-for a viscous mycelial fermentation of this type-a large-diameter
axial flow impeller is much more efficient at oxygen transfer than the more
conventional Rushton radial flow impeller.
150
.... <).... PROCHEM
140 -a- RUSHTON
130
120
z 110
0
i= 100
<
ct:
:::> 90
~
< 80
(I)
ct:
70
< 60
~ 50
ci 40
c:i 30
20
10
0
0 10 20 30 40
HOURS
FIGURE 3.8. Dissolved oxygen profiles for same two fermentation described in
Figure 3.7 (from Buckland et af. 1988).
400
o Prochem
o Rushton
300
....
:::l
0
~
::::: 200
0
..J
::.::::
100
OL-----~----------~-----------J
20 30 40
Hours
FIGURE 3.9. On-line calculation ofthe oxygen transfer coefficient for the same two
avermectin fermentations described in 3.7 (from Buckland et al. 1988).
Prochem
2
O~----------~----------~----------~--------~
o 10 20 30 40
Hours
FIGURE 3.10. Agitator power draw for the same two avermectin fermentations
described in Figure 3.7 (from Buckland et al. 1988).
3. Process Development and Improvement 51
Environmental Containment
Streptomyces avermitilis is not known to be unusually harmful; neverthe-
less, it was necessary to maintain a high level of containment because the
product-avermectin-is very potent and could have a potential environ-
mental impact on aquatic life forms. Containment concerns extended to
both the fermentation operations and to the downstream purification
facility. These considerations were incorporated into the factory design
from the beginning and were resolved successfully: all possible exit
streams from the process are captured and the avermectin therein
degraded chemically.
Media Sterilization
As with many processes of this type, media sterilization is an important
variable (Jain and Buckland 1988) for both the seed tank stages and the
production stage. By necessity, more stringent conditions are required
as scale is increased to achieve the same assurance of complete steriliza-
tion (Buckland 1985). With a batch procedure, this is normally accomp-
lished by increasing time at sterilization temperature; with a continuous
sterilizer either the time or the temperature can be easily increased.
Superimposed on the requirement for sterility is the fact that a number
of chemical changes occur in the medium during sterilization, some of
which can have a great impact on culture productivity. This therefore
becomes an important area for optimization and, if possible, chemical
analysis.
Oxygen Transfer
Because of the viscous characteristics of the broth, the ability to transfer
sufficient oxygen to the individual microbial cell ultimately limits the cell
mass and hence productivity of the fermentation, assuming that the other
critical physical and chemical parameters that influence metabolism have
been chosen correctly to achieve high levels of product expression. When
evaluating a proposed process change in the pilot plant, it is necessary to
be sure that the oxygen requirements of the new process can be met
adequately in the larger-scale equipment. One simple method for doing
this is to run the pilot-scale fermentor in a way that mimics the limitations
ofthe factory equipment. An example is given in Figure 3.11, in which a
new medium formulation was evaluated in the pilot plant (medium 2). It
was immediately apparent that the oxygen demand of the new process
was in fact less than that for the standard process; given that the broth
viscosities were similar, introducing this process into the factory would
not result in starving the cells for oxygen.
52 Mary Nallin Omstead, Louis Kaplan, and Barry C. Buckland
Scale-Up of Avermectin
40.---------------------------------------~
...
::J
2
~
......
~ 30
o
~
E
-
~
Q)
20
~
o
~
a
&i 10
g
01
30 40 50
Hours
FIGURE 3.11. Scale-up information for new avermectin process (from Buckland
1985).
broth is first acidified and then mixed with an extractant such as toluene at
high temperature. The extractant is then decanted away from the aqueous
phase and, after just a few more processing steps, transferred to the final
crystallization tank.
REFERENCES
Biosynthesis
S. T. Chen, O. D. Hensens, and M. D. Schulman
I. Introduction
II. Incorporation of Labeled Precursors
III. Bioconversion of Intermediates
IV. Enzyme Studies
V. Effect of Specific Inhibitors
VI. Conclusions
I. Introduction
The avermectins are a family of oleandrose disaccharide derivatives of
pentacyclic lactones. Figure 4.1 presents a generalized structure of the
avermectins produced by Streptomyces avermitilis. In addition, a number
of key intermediates in the biosynthesis of the avermectins have been
identified. These structures are as follows: aglycones lack the oleandrose
disaccharide and possess a hydroxyl at C13; demethylavermectins lack
one or more methoxyls on the oleandrose disaccharide and possess a
hydroxyl at either or both the C3" and C3'; desfurano avermectins lack
the oxygen at C8a and C6 and possess a CH3 at C8a and an H at C6; and
5-keto avermectins lack the hydroxyl (or methoxyl) at C5 and possess a
keto group at C5.
Presently, the pathway for avermectin biosynthesis is not completely
understood. The pathway proposed in this chapter is based on evidence
obtained from four types of studies: the incorporation of labeled pre-
cursors into the avermectins; the conversion of proposed intermediates
into avermectins by producing strains and blocked mutants; the in vitro
measurement of enzymes involved in the biosynthesis of the avermectins;
and the production of incomplete avermectins by administration of
specific enzyme inhibitors.
56 T.S. Chen, O.D. Hensens, and M.D. Schulman
OR,
FIGURE 4.1. Avermectin terminology is as follows: X = CH = CH for "1"
components; X = CH 2CHOH for "2" components; RI = H for "B" com-
ponents, RI = CH3 for "A" components; R2 = CH 2CH3 for "a" components;
R2 = CH 3 for "b" components (from Schulman, Ruby 1987).
Precursors
---- Acetate
o (CH 3 )
Y
28
HaC
26 25
26a
0
L Isoleucine
or
27
\62~O L Valine
~
FIGURE 4.2. Incorporation of labeled precursors 10 the avermect1Os.
(CH3) = S-CH3 of methionine; ~ = acetate unit; .-~ = propionate unit;
• = carbons derived from isoleucine found in "a" components; • = carbons
derived from valine found in "b" components.
[14C] valine into the "b" components (Table 4.2). In addition, there was
high enrichment (twenty-fivefold) at C25 of avermectin B la by [I_I3C]
2-methylbutyrate (Table 4.1) and of avermectin BIb [l-I3C] by isobu-
tyrate. The metabolism of [1-13C] 2-methylbutyrate to [1-13C] propionate
accounts for the enrichment at C3, C7, Cll, C13, C23 of B la .
The methoxyl substituents at C5 of the macrolide moiety and C3' and
C3" of the oleandrose disaccharide are derived from the methyl of
methionine. It was shown using both 14C and 13C-methyl methionine and
[2_14C] methionine that the S-methyl and not the carbon backbone is
incorporated into avermectin (Tables 2.3, 2.4, and 2.5). The methyl is
incorporated equally into all 3 methoxyl groups.
The origin of the oxygen atoms in the macrolide moiety was investi-
gated by measuring the incorporation of [I_ 13 C, 1- 180] acetate and [I_ 13C,
I_ISO] propionate (Cane et al. 1983). The oxygens at C1, C5, C7, C13,
C17, C19, and C23 (for the "2" components) retained their isotope
TABLE 4.1. Incorporation of [Be] precursors in avermectins by S. avermitilis.
Relative 13C abundance in
Carbon avermectin B I • biosynthesized from ICC in avermectin B I • from
No. [2- 13] acetate [3- 13C]propionate [1- 13C]2-methylbutyrate [l,2- 13C 2]acetate [2,3- 13C 2]propionate
Col 1.4 1.0 1.1 59.3Hz N.D.2
C-2 3.2 1.2 0.9 59.3Hz N.D.
C-3 I.S 1.4 6.7 N.D.' N.D.
C-4 3.0 1.2 1.3 N.D. 43.4 Hz
C-4a 2.2 5.S 1.2 N.D. 43.4 Hz
C-5 1.0 1.3 O.S 40.7 Hz N.D.
C-6 3.S 1.2 1.0 40.7 Hz N.D.
C-7 1.6 1.4 7.7 N.D. 43.2 Hz
C-S 2.S 1.3 1.2 N.D. 43.2 Hz
C-Sa 2.4 6.2 1.0 N.D. N.D.
C-9 1.1 1.0 1.2 55.9 Hz N.D.
ColO 3.2 1.3 0.9 55.9 Hz N.D.
C-1I I.S 1.5 6.4 N.D. N.D.
C-12 2.6 1.5 1.0 N.D. 35.5 Hz
C-I2a 2.4 6.S 1.1 N.D. 35.5 Hz
C-l3 1.2 1.4 5.9 N.D. N.D.
C-14 2.9 0.9 0.9 N.D. 44.0 Hz
C-14a 2.3 6.6 1.1 N.D. 44.0 Hz
C-15 1.1 1.2 1.0 43.7 Hz N.D.
C-16 3.0 1.1 1.1 43.7 Hz N.D.
C-17 1.2 1.1 1.3 36.3 Hz N.D.
C-lS 4.4 1.3 1.0 36.5 Hz N.D.
C-19 1.2 1.1 1.3 36.3 Hz N.D.
C-20 3.8 1.3 0.9 36.3 Hz N.D.
C-21 1.3 0.9 1.2 54.9 Hz N.D.
C-22 3.7 1.3 1.0 54.9 Hz N.D.
C-23 1.8 1.5 5.6 N.D. N.D.
C-24 2.7 1.7 0.9 N.D. 35.2 Hz
C-24a 2.8 6.7 1.2 N.D. 35.2 Hz
C-25 2.2 1.0 24.5 N.D. N.D.
C-26 1.3 1.1 0.8 N.D. N.D.
C-26a 1.3 1.0 0.9 N.D. N.D.
C-27 1.4 1.0 1.1 N.D. N.D.
C-28 1.3 1.0 0.9 N.D. N.D.
C-l' 1.2 1.0 0.9
C-l" 1.1 1.0 1.0
C-2' 1.3 1.0 1.0
C-2" 1.2 1.0 1.0
C-3' 1.2 1.0 1.0
C-3" 1.1 1.1 1.1
C-4/ 1.1 1.1 1.1
C-4" 1.1 1.1 1.0
C-5' 1.1 1.3 0.8
C-5" 1.0 1.1 1.0
C-6' 1.0 1.1 1.0
C-6" 1.0 1.1 1.0
3/,3"O-CH 3 1.7 1.2 1.3
': Not determined.
60 T.S. Chen, D.D. Hensens, and M.D. Schulman
Labeled avermectin
Spec.
Spec. incorporation
Spec. radioact. into C25
radioact. (p.Ci/mmole) sidechain (%)
Substrate Conc (uM) ("Ci/mmole) B la BIb B la BIb
Valine 4.1 5.85 0.54 1.57 12.3 21.3
Isoleucine 7.3 3.28 2.32 0.40 70.4 14.7
Isobutyrate l.1 11.00 0.17 0.57 1.5 5.0
2.2 6.36 0.14 0.69 6.4 10.9
3.3 3.62 0.71 0.82 4.7 22.7
4.4 2.58 0.27 0.77 10.5 29.8
content (Table 4.6). Several conclusions were drawn from these results:
(1) the avermectin "1" components which contain a double bond at
C22-23 arise from the "2" components by dehydration of the hydroxyl at
C23; (2) the oxygen at C21 is probably derived from the 2-methylbutyryl
or isobutyryl sidechain (C25); (3) the avermectins are not derived from a
milbemycin type intermediate by late-stage oxidation at C13 (Ono et al.
1983).
To investigate the origin of the oleandrose backbone, studies with
3H/ 14C and 13C labeled glucose were conducted. Comparison of the
3H/ 14C ratio in the labeled avermectin and the methyl oleandrose obtained
by methanolysis (Table 4.7) indicated a direct conversion of glucose to
oleandrose (Chen et al. in press). This was confirmed by studies with
[1_I3C] glucose and [U-I3CJ glucose which showed high enrichment of the
oleandrose units (Schulman, Valentino, and Hensens 1986).
Studies with labeled glucose provided some insight into the metabolic
origin of the avermectin precursors (Schulman, Valentino, and Hensens
1986). Equal incorporation of [1- 14CJ, [2_ 14CJ and [6_ 14C] glucose into the
avermectin aglycone provided strong evidence for the symmetrical cleav-
age of glucose via the Embden-MyerhofIpathway. A parallel study with
[U-I3C] glucose demonstrated that the entire avermectin molecule is
synthesized from glucose (Figure 4.3). The oleandrose units derived
directly from glucose had the highest incorporation rate. The methoxyls
of the oleandrose units which are derived from the methyl of methionine
were also enriched. Methyl-labeled methionine can be synthesized from
glucose via a known microbial pathway that involves serine and serine
transhydroxymethylase. The carbons derived from acetate were enriched
3.3-fold over those derived from propionate. This is due to a dilution
of acetate by the passage through the tricarboxylic acid cycle to
yield propionate (via succinate). The asymmetric labeling of the
2-methylbutyrate-derived sidechain is explained by assuming isoleucine is
formed from oxalacetate via the known pathway in the presence of
threonine aldolase.
CH
R X
hypothesis that reduction of the C5 keto occurs after the formation of the
furan ring.
Glycosylation to the mono- and disaccharide was observed with a
variety of intermediates, indicating that the glycosyltransferases lacked
specificity. When [5-methoxyl-14C] A2a bisdemethylavermectin and un-
labeled methionine were fed to wild-type S. avermitilis, the unaltered
substrate was the only radioactive compound recovered (Schulman and
Ruby 1987). Its specific radioactivity was virtually identical to that added.
This demonstrates that methylation of the oleandrose units is not a
terminal step in avermectin biosynthesis and that it occurs before
attachment of the sugars to the macrolide ring. The addition of 6,8a-
deoxy-5,13,23-triketo avermectin to a nonproducing blocked mutant did
not yield any natural avermectin compound, indicating this compound is
not a biosynthetic intermediate.
66 T.S. Chen, O.D. Hensens, and M.D. Schulman
A VERMECTIN 5-KETOREDUCTASE
CH 2CH 3 CH 2 CH 3
H3 C •
S-Adenosyl S-Adenosyl
methionine homocysteine
OH OCH 3
CH 2CH 3 CH 2CH 3
H3 C
NADPH NADP+
+
H+
o OH
20
19
18
0'
co
17
~ 16
< 15
~ 14
13
l!!
:J
c
=:J 12
2. 11
f 10
9
~ 8
Ql
1/1
III
7
g 6
'tI
l!! 5
4
~
lI:: 3
.n 2
1
0
3 5 7
It has not yet been determined if both sugars are transferred by a single
enzyme or if two enzymes are required. It is clear, however, that the
sugars are transferred sequentially.
Oleandrosyl
Diphosphonucleotide
I
6, 8a-Seco-6, 8a·deoxy·
~
6, 8a-Seco-6, 8a-deoxy-
5 Keto "2a" AG 5 Keto "1a" AG
~
5-Keto "2a'· AG
~
5-Keto "1 a" AG
SAM ~
I---~).::-.--
A2.AG .... 8 2.AG
~
8 1aAG - - - - -•• A1.AG
SAM
SCHEME 1. Proposed pathway for biosynthesis of the avermectins in S. avermi-
tiUs.
72 T.S. Chen, O.D. Hensens, and M.D. Schulman
VI. Conclusions
The proposed pathway for biosynthesis of the avermectin "a" com-
ponents is presented in Figure 4.9. An exactly analagous pathway
is envisioned for the "b" components, with valine, isobutyrate, and
isobutyryl-CoA replacing isoleucine, 2-methylbutyrate, and 2-methyl-
butyryl-CoA. Isoleucine undergoes transamination and decarboxylation
to yield 2-methylbutyryl-CoA, which serves as the primer for chain
elongation. 2-methylbutyryl-CoA can also arise from fed 2-methylbuty-
rate by activation via a thiokinase. Chain elongation proceeds by addition
of acetate and propionate via their CoA esters in a fashion analagous to
fatty acid synthesis. At some point during the process, a precursor of the
"2" components is dehydrated at C22-23 to yield a precursor of the" 1"
compounds. From this point on, biosyntheses of the "1" and "2"
components proceed in parallel to produce the respective aglycones.
Because the methyltransferase and the glycosyltransferase display little
specificity, the aglycones are either methylated or glycosylated to pro-
duce "A" and "B" disaccharides. At the disaccharide level, B2a can be
methylated to A2a but B la cannot be methylated to Ala.
REFERENCES
Cane DE, Liang T-C, Kaplan LK, Nallin MK, Schulman MD, Hensens OD,
Douglas AW, Albers-Schonberg G (1983) Biosynthetic origin of the carbon
skeleton and oxygen atoms of the avermectins. J. Am. Chem. Soc. 105:4110-
4112
Chen TS, Arison BH, Gullo VP, Inamine ES (in press) Further studies on the
biosynthesis of the avermectins. J. Indust. Microbiol.
Chen TS, Inamine ES (in press) Studies on the biosynthesis of avermectins. Arch.
Biochem. & Biophys.
Ono M, Mishima M, Takiguchi Y, Teroa M (1983) Milbemycins, a new family of
macrolide antibiotics. Studies on the biosynthesis of milbemycins a2, a4 and D
using BC labelled precursors. J. Antibiot. 36:991-1000
Schulman MD, Ruby C (1987) Methylation of demethylavermectins. Antimicrob.
Agents Chemther. 31:964-965
Schulman MD, Valentino D, Hensens OD (1986) Biosynthesis of the avermectins
by Streptomyces avermitilis. J. Antibiot. 39:541-549
Schulman MD, Valentino D, Hensens OD, Zink D, Nallin MK, Kaplan LK,
Ostlind DA (1985) Demethylavermectins. Biosynthesis, isolation and charac-
terization. J. Antibiot. 38:1494-1498
Schulman MD, Valentino D, Nallin MK, Kaplan LK (1986) Avermectin B2
O-methyltransferase activity in Streptomyces avermitilis mutants that produce
increased amounts of the avermectins. Antimicrob. Agents Chemother. 29:620-
624
Schulman MD, Valentino D, Ruby C (1985) Avermectin B O-methyltransferase of
Streptomyces avermitilis. Fed. Proc. 44:931
CHAPTER 5
I. Introduction
II. Invertebrates
A. Electrophysiological Studies
B. Biochemical Studies
1. IVM Binding Sites
2. Chloride Uptake
3. Acetylcholine Release
4. Interaction with Retinal Binding Proteins
5. Inhibition of Chitin Synthesis
III. Vertebrates
A. A VM Stimulated Neurotransmitter Release
B. A VM Binding Sites
C. A VM Effect on [3H]GABA Binding
D. A VM Effect on Benzodiazepine Binding Sites
E. Chloride Uptake
F. Other Actions of AVM
IV. Conclusions
I. Introduction
The anthelmintic activity of the avermectins was first described in 1979
(Burg et al. 1979; Egerton et al. 1979; Miller et al. 1979). The mode of
action of these compounds, however, has remained elusive. Identifying
the mode of action is all the more difficult because the avermectins have
been studied in so many different model systems with an array of
experimental protocols. For example, direct injection of avermectin
(AVM) into Ascaris suum results in a rapid paralysis that is neither flaccid
74 M.J. Turner and J.M. Schaeffer
II. Invertebrates
A. ELECTROPHYSIOLOGICAL STUDIES
The earliest studies designed to elucidate the mode of action of A VM
were reported by Fritz, Wang, and Gorio (1979). They demonstrated that
when lobster stretcher muscle is perfused with 1O-5-10-6M AVM, the
inhibitory postsynaptic potentials are rapidly eliminated, followed by a
more gradual reduction in the amplitude of excitatory postsynaptic
potentials. In these preparations, AVM reduces the input resistance of the
muscle fiber by increasing its permeability to chloride (Cl-) ions. These
effects were not reversed by washing. The reduction of both excitatory
potentials and input resistance, however, was reversed by picrotoxin (a
GAB A antagonist active at the chloride channel). It was hypothesized
that these responses to AVM were caused by an increase of membrane
permeability to chloride ions, perhaps due to its interaction with GAB A
binding sites or by regulating the release of endogenous GABA. These
findings have subsequently been confirmed (Mellin, Busch, and Wang
1983; Albert et al. 1986).
The motor nervous system of Ascaris lumbricoides is an excellent
experimental system to study avermectin's mode of action. An elegant
description of the nervous system is available (Stretton et al. 1978) which
outlines the connections and electrical characteristics of the neurons. The
neurons, moreover, are large enough to be penetrated with microelec-
5. Mode of Action of Ivermectin 75
trodes. Using this model system, Kass and his collaborators (1980, 1984)
demonstrated that A VM (=5 x 1O-6M) inhibits transmission between
interneurons and excitatory motoneurons in the ventral nerve cord
(Figure 5.1), as well as inhibiting transmission between inhibitory
motoneurons and muscle; it has little effect, however, on excitatory
neuromuscular transmission. Picrotoxin can reverse the AVM-induced
block of interneuron-excitatory motoneuron transmission but has no
effect on the inhibitory motoneuronal synapse in either the presence or
absence of AVM. These findings suggest that AVM can function either as
a GABA agonist or as a stimulator of GABA release from presynaptic
inhibitory terminals.
Duce and Scott (1983, 1985) demonstrated that within the extensor
tibiae muscle of the locust (Schistocerca gregaria) , specific muscle
bundles are sensitive to GABA, whereas a separate population of muscle
bundles, which receive only excitatory innervation, are GABA-
insensitive. This specificity within a single preparation of muscle fibers
was exploited to investigate the interaction of AVM with the GABA
receptor-ionophore complex in insect muscle. The muscle bundle that is
sensitive to GABA receives inhibitory innervation and exhibits both
reversible and irreversible responses to AVM. There is a reversible
dose-dependent increase in CI- permeability in response to very low
doses of AVM (= 10-IOM); these effects appear to be due to an interaction
of AVM with the GABA receptor-CI- ion channel complex. Higher AVM
concentrations (=10- 8M) evoke an irreversible increase in Cl- conduc-
tance that continues to rise after removal of AVM. GABA-insensitive
neurons with no inhibitory innervation were also examined; in this case
AVM
8 5 p,g/ml
> 6
E
a) I I tI It Picrotoxin Wash
If
L!
en 10 p,g/ml
c: 4
I
,
0
a.
en
Q)
a: 2 I ! I
0
•I
0 20 40 60 80 100 120 140 160
Time, min.
FIGURE 5.1. Responses to indirect stimulation of DEL Each point represents the
mean of five responses. All recordings are from the same dorsal muscle cell.
Drugs were added at arrows. (From Kass et al. 1980).
76 M.J. Turner and J .M. Schaeffer
6. BIOCHEMICAL STUDIES
3.0
0
~O-
0> /0 :
/
E Ko=0.26nM
.......
(5 2.0 :,; 7
E
0..
.,en
c
6
BMa.=3.53
"0
E
0
/
c: (5 5
:J E
0-
m cD
4
II)
~ !l: 3
>
...... "-
0
"C
.-----. c
1.0
•
::J 2
I 0
'\
In
~
0
3.0
Bound. pmol/mg
[IVM], nM
FIGURE 5.2. Equilibrium binding of [3H] IVM to C. elegans membranes. Increas-
ing concentrations of PH] IVM were incubated with C. elegans membranes and
specific binding was determined as described in the text. Each point is the average
of 4 determinations. Replicate experiments gave similar results. A scatchard
analysis (inset) of the saturation data is shown. (From Schaeffer and Haines, in
press).
250
8/
100
/
-7 -
:::E
c
~
0
w 50
25
10 ~--------~------~--------~--------~~
0.1 0.25 0.5 1.0 2.5
KI,nM
FIGURE 5.3. Correlation between binding affinities of AVM analogs and their
biological potencies of C. elegans motility in vivo. EDso is the concentration of
AVM needed to produce immotility in 50% of the worms. The results are the mean
values from 2 to 10 experiments (SEM less than 15%). A correlation coefficient (r)
of 0.975 was calculated by linear regression analysis. For the log-log plot,
r = 0.923. The compounds tested were (1) IVM; (2) AVM B2a ; (3) AVM Bla
4'-0-phosphate; (4) 22,23-dihydro-AVM B 1a aglycone; (5) 22,23-dihydro-AVM BI
monosaccharide; (6) AVM BI monosaccharide; (7) 2-dehydro-4-hydro-a-2,3-
AVM B 1; and (8) AVM B 1a-5-ketone. (From Schaeffer and Haines, in press).
2. Chloride Uptake
The action of AVM on chloride uptake was studied on the perfused leg
muscle of the American cockroach, Periplaneta americana (Tanaka and
Matsumura 1985). AVM stimulated chloride uptake at concentrations as
low as to- 8M. The AVM-stimulated chloride uptake is antagonized by
picrotoxin and to a lesser extent by bicuculline methiodide. At 10-7M
AVM the leg muscles fail to respond to external stimuli. This inhibition
was postulated to be due to an increase of chloride uptake by the leg
muscle. These results suggest that AVM is opening the chloride channel
5. Mode of Action of Ivermectin 79
3. Acetylcholine Release
Nicholson and colleagues (1988) measured AVM-induced presynaptic
acetylcholine release in a cockroach CNS synaptosomal preparation. The
synaptosomes were preloaded with [3H]choline and pp-rfused under
various conditions. A VM causes a release of radioactivity presumed to be
acetylcholine (this was not verified). The threshold AVM concentration
for acetylcholine release was 10- 10M, the EDso value was lO- 8M, and
maximum stimulation required lO- 6M. The effect was chloride dependent
and could be antagonized by picrotoxin, trioxabicyclooctaines, and
hexachlorocyclohexane. In contrast, the ,a-isomer of hexachlorocy-
clohexane potentiated the A VM effect. The authors conclude that in the
insect CNS, AVM induces the opening of a presynaptic chloride ion
channel (not GABA-activated), leading to efflux of chloride ions, depolar-
ization of the nerve terminal, and hence, to neurotransmitter release.
III. Vertebrates
Avermectins have been tremendously successful anthelmintic agents
because of their ability to kill parasites without affecting the host
organism. Many of the reports describing AVM effects in vertebrates
require concentrations of A VM far in excess of those that will ever be
attained outside of the laboratory. For this reason, the effects of AVM on
nontarget vertebrate species cannot necessarily be related to the mode of
action against invertebrate target species.
2.0r-------r-------~------~------~
-
c-
.Qj
a
0.. 1.5
Ol
--'0Een
Q)
E
.e,
"0
C
::l 1.0
a
[D
~
Cii
~
·0
Q)
Q.
w 0.5
«
[D
«
<D
O~------~------~-------L------~
o 2 4 6 8
AVM (~M)
13
0>
c::
=SOP
c::.c:
'CO.-
- 0> 11
EQ)
",3=
a.Ci)
Q)3=
~Q)
-::::I
'2 ~ 9
.2 ..."
-0>
IE 0--0 + NaCI
C')U5 A--A + NaCI + Picrotoxinin
-
.0
Q)
_ -
0
7 ....... - NaCI
13.5
Q) -
A--A - NaCI + Picrotoxinin
a.
(J)
5 i I
10 100
Avermectin (nM)
FIGURE 5.5. Effect of AVM on [3H]flunitrazepam binding and modulating'of
AVM-stimulated [3H]flunitrazepam binding by chloride ions and picrotoxinin.
Membranes from rat cerebellum were incubated with 1 nM [3H]flunitrazepam and
various concentrations of AVM in the absence or presence of p.M diazepam in
a solution containing 50 mM Tris-citrate, pH 7.3, in the absence or presence of
150 mM NaCI and/or 30 p.M picrotoxinin. Incubations were performed at 23°C for
30 minutes, and membranes were then filtered through Whatman GF/B filters and
washed as described in Materials and Methods. Results are from a typical
experiment which was repeated three times with identical results (from Williams
and Yarbrough 1979).
E. CHLORIDE UPTAKE
10
8
'E
....
Ql
....
::J
u 6
Ql
.~
Cij
(j) 4
a:
l
0 I I I I
10-8 10-7 10-6
Avermectin concentration (M)
FIGURE 5.6. Concentration dependence of the stimulatory effect by avermectin
B'a. The measurements were standardized by assigning the value of I to the
current amplitude elicited by a control application of 5 JLM GABA. Different
concentrations of avermectin B'a were applied for 3 minutes alone and then in
combination with 5 JLM GABA. For each point the Mean ± standard deviation is
given for at least three determinations performed on different oocytes (from Sigel
and Baur 1987).
F. OTHER ACTIONS OF A VM
IV. Conclusions
It is not possible to assign a single mechanism of action for A VM in the
various systems that have been studied. In target organisms, the effect of
AVM is mediated via a specific, high-affinity (=10- IOM) binding site. The
physiological response to A VM binding is an increase in membrane
86 M.J. Turner and J.M. Schaeffer
Toxicology
George R. Lankas and Lea R. Gordon
I. Introduction
Abamectin (MK-0936) is a natural fermentation product of Streptomyces
avermitilis. Ivermectin (MK-0933) is a synthetic derivative of abamectin.
The chemical structure of abamectin differs from ivermectin only in the
bond between carbons 22 and 23; abamectin has a double bond where
ivermectin has a single bond and additional hydrogens on C-22 and C-23
(Figure 6.1). Both compounds are a mixture of homologous products with
B la and BIb components. The BIb component differs chemically from
the B la component by only 1 methylene (CH2) unit at the 26-carbon
position: the ethyl group (C2H5) is a methyl group (CH3) in the BIb form.
Abamectin and ivermectin are defined as containing a minimum of
80% B la and a maximum of 20% BIb components. Studies in our
laboratories have clearly demonstrated that the individual components
have very similar biological and toxicological properties and, for all
practical purposes, can be considered equivalent.
GAB A receptors. Abamectin and ivermectin increase calcium perme-
90 George R Lankas and Lea R Gordon
0 0
MK-0933 (Iverme~~!~)
(Bla component
093 6 (Abamectin) )
t1lC-
(B1a componen t shown
this expression period, TK'- mutants were detected by cloning the cells
in selection medium containing bromodeoxyuridine for 10 to 14 days. The
surviving cell population was determined by plating diluted aliquots in
nonselective growth medium.
The results showed that ivermectin at concentrations up to 60 ug/ml did
not produce any significant increase in mutation frequency (with or
without an S-9 metabolic activation system) when compared with appro-
priate negative controls.
Unscheduled DNA synthesis (UDS) occurs after cellular DNA is
damaged by agents that induce either base, short-patch, or long-patch
excision repair (Wilkins and Hart 1973; Setlow and Carrier 1964). The
damaged bases are enzymatically excised from their DNA strand, creat-
ing a gap; new bases are inserted and polymerized to fill the gap, using the
intact opposite strand as template. Since many carcinogenic and muta-
genic agents have been demonstrated to induce UDS, the assay is
considered to be a measure of genotoxicity (Williams, Laspia, and Dunkel
1982).
Ivermectin was assayed for its ability to induce UDS in an in vitro assay
using a normal human embryonic lung fibroblast cell strain, IMR-90, at
concentrations of 10 to 1000 ug/ml. The compound was tested with and
without the addition of a rat liver S-9 metabolic activation system.
Ivermectin did not induce UDS in human cells at any dose level.
In conclusion, ivermectin was tested in vitro for genotoxic activity in
microbial and mammalian cell mutagenesis assays and in human fibro-
blasts for DNA damage as measured by unscheduled DNA synthesis. In
all instances, ivermectin had no genotoxic activity.
The drug's effects in mice were similar after either oral or intraperi-
toneal administration: they consisted of ataxia (uncoordinated move-
ment), bradypnea (abnormally slow respiration rate), and tremors at all
dosage levels within approximately 1 hour of treatment. Death, preceded
by loss of righting, occurred from 75 minutes to 6 days after dosing, with
most occurring within 24 hours. There were no significant sex-related
differences in acute toxicity in mice after either route of administration.
In rats treatment-related physical signs consisted of ataxia, ptosis
(drooping upper eyelids), and decreased activity; the symptoms appeared
from about 4 to 24 hours after oral or intraperitoneal administration.
Death, generally preceded by loss of righting, occurred overnight to the
second day. Body weight gains during the 2-week observation period
appeared unaffected by drug administration. There were no significant
sex-related differences.
Ivermectin was more toxic in infant rats (1-2 days old) than in young
adult rats. As discussed in more detail in Section D, this enhanced
sensitivity is believed due to postnatal development of the blood-brain
barrier in rats (Betz and Goldstein 1981) (in humans this structure is
formed prenatally; Bohr and Mollgard 1974). This conclusion is supported
by significantly higher brain/plasma drug concentration ratios in neonatal
rats compared to adults.
In rats and rabbits acute dermal LD50 values for ivermectin after 24
hours of occluded exposure were >660 and 406 mg/kg, respectively.
These values are significantly greater than those discussed previously for
oral exposure in rats and mice, suggesting that ivermectin is poorly
absorbed percutaneously. The poor dermal absorption of abamectin has
been confirmed in rhesus monkey (Macaca mulatta) studies, demonstrat-
ing that 0.5% or less of a dermally applied dose is absorbed (see section
III A below).
The acute oral toxicity of ivermectin in dogs was assessed by adminis-
94 George R. Lankas and Lea R. Gordon
C. SUBCHRONIC TOXICITY
effect level was established in this study. Similar changes were not seen in
3-month, I-year, or 2-year studies with abamectin in rats at comparable
and higher dose levels, nor in other species given ivermectin.
In a 3-month oral toxicity study in beagle dogs the compound was
administered as a solution in sesame oil by gavage at doses of 0.5, 1.0, or
2.0 mg/kg/day. Mydriasis and slight weight loss were observed at doses of
1.0 and 2.0 mg/kg/day. Four of 8 dogs in the 2.0 mg/kg/day group
developed tremors, ataxia, anorexia, and dehydration, and were sacri-
ficed prior to the scheduled necropsy. No gross abnormalities were
observed in these dogs. Mydriasis was observed in dogs receiving 1.0 or
2.0 mg/kg/day. No other treatment-related effects were found.
Postmortem studies conducted on all animals disclosed no treatment-
related gross, microscopic, or organ weight changes. A no-effect level of
0.5 mg/kg/day was established for this study.
A 16-day oral toxicity study with ivermectin was conducted to deter-
mine its toxicity in immature domestic-bred rhesus monkeys (approxi-
mately 1 to 1.5 years old). Each of the 3 treatment groups were dosed
daily via nasogastric intubation at dosage levels of 0.3, 0.6, and
1.2 mg/kg/day. These dosage levels were arbitrarily chosen to provide an
approximate sixfold safety margin relative to the human clinical dose of
ivermectin; based on the acute toxicity in rhesus monkeys, these doses
would not be expected to produce clinical signs of toxicity. The control
group received the vehicle, sesame oil, at the same 1.0 mllkg dose volume
as the treated groups.
No drug-related physical signs were noted in any of the treated animals.
All animals survived the duration of the study with similar body weight
gains for all treated groups as compared to the controls. No treatment-
related ocular lesions or changes in hematology or serum biochemical
parameters were found during the study. There were no organ weight,
gross, or microscopic changes attributed to treatment. Therefore, treat-
ment of immature monkeys with 1.2 mg/kg/day for 14 days failed to
produce any treatment-related effects.
Because ivermectin is excreted in the milk of lactating animals, it was
necessary to generate appropriate preclinical data on the safety of
low-level exposure to nursing infants via maternal milk. Based on a
clinical study in lactating women treated with ivermectin, a maximum
milk level of the drug of 23 ng/ml was found on the day following
treatment, with levels depleting to below the 0.1 ng/ml detection limit by
approximately 1 week after treatment (M. Dobrinska, personal commu-
nication). This level of drug in human milk would result in an estimated
maximum-acute dosage of about 3 to 4 ug/kg in a nursing infant.
To assess the potential significance of a low-level neonatal exposure to
ivermectin, a study of neonatal rhesus monkeys was conducted. At
initiation of the study all animals were 7 to 13 days old. The animals were
divided into 2 treatment groups of 5 males and 3 females each; they
98 George R. Lankas and Lea R. Gordon
2&
• Maternal death •
• Litter. "lth cleft palate
20
The results of the multigeneration study and the acute toxicity study in
infant rats (see Section II B above) indicated that noenatal rats were more
sensitive to the drug than parental animals. To determine the reason for
this difference in sensitivity, female rats were administered tritium-
labeled ivermectin Bla at a dosage level of 2.5 mg/kg/day continuously
for 61 days prior to mating and during mating, gestation, and lactation.
Plasma and milk samples from dams and plasma samples from offspring
were analyzed for total radioactivity on days 1, 4, and 6 postpartum. On
day 10 postpartum all dams and offspring were sacrificed and total
radioactivity determined in plasma and brain samples.
The total drug residues in plasma and milk of dams and plasma from
their offspring are shown in Figure 6.3. The increase in maternal plasma
radioactivity at parturition relative to days 4 to 10 postpartum is likely due
to increased utilization of fat at parturition in the rat (Amano 1967; Scow
et al. 1964), resulting in release ofivermectin from adipose tissue (Chiu et
al. 1986). The concentrations of ivermectin in milk were consistently
three- to fourfold higher than those obtained from maternal plasma
samples on comparable days postpartum. Plasma drug levels in offspring
were relatively low on day 1 postpartum but increased rapidly from days 6
to 10 such that they equaled or exceeded the values in the dams. It should
be noted that the 0.8 ug/ml mean plasma concentration in offspring on day
10 postpartum is approximately 40 times the therapeutic plasma concen-
tration in humans and other species. Of greater importance is the finding
that the brain concentrations of ivermectin residues in offspring by day 6
postpartum were approximately tenfold the maternal brain concentrations
(Figure 6.4).
The results of this study indicated that the high drug concentrations in
milk from lactating dams chronically administered the drug were probably
responsible for the toxicity observed among neonatal offspring in multi-
generation studies. This conclusion is supported by the high drug levels
found in the plasma and brain of offspring relative to adult rats. In
particular there was a greater concentration of drug in the brain of
neonatal rats during the early neonatal period of enhanced sensitivity, and
this may explain the increased toxicity. This early neonatal period of
increased sensitivity to ivermectin toxicity in rats correlates with the
postnatal completion of the blood-brain barrier in this species (in humans
it is formed prenatally; Betz and Goldstein 1981; Bohr and Mollgard 1974;
Saunders 1977). Further support for the importance of postnatal exposure
in the toxicity observed in neonatal rats is derived from a cross-fostering
study, in which control pups cross-fostered to ivermectin-treated dams
exhibited the same degree of neonatal mortality and toxicity as rats
exposed in utero and fostered to drug-tested dams (R. T. Robertson,
personal communication). Therefore, postnatal exposure via milk-not a
combination of prenatal and postnatal exposure-explains the neonatal
toxicity observed in multigeneration studies conducted with ivermectin.
102 George R. Lankas and Lea R. Gordon
1.2
a
1
0.8
Plasma
cone 0.8
(p,g/ml)
0.4
0.2
5
b
Milk 3
cone
(p,g/ml) 2
1.4
C
1.2
1
F1
plasma 0.8
cone 08
(p,g/ml) .
0.4
0.2
0
1 4 6 10
Days postpartum
FIGURE 6.3. Mean concentrations of 3H-ivermectin in (a) plasma, (b) milk from
lactating rats, and (c) plasma from offspring of treated dams. Error
bars = ± standard deviation.
0.5
0.4
0.3
Brain
cone
(ppm)
0.2
0.1
- - -
0.0
1 4 6 10
Days postpartum
FIGURE 6.4. Mean 3H-ivermectin levels in brain from dams and offspring. Dashed
line represents steady state brain concentration of dams measured on day 10
postpartum. Error bars = ± standard deviation.
is genotoxic. The oral LDso in mice is 14-24 mg/kg for abamectin and
25-40 mg/kg for ivermectin; the minimal effect level for toxicity in dogs is
0.50 mg/kg/day for abamectin and 1.0 mg/kg/day for ivermectin. In
developmental toxicology studies on abamectin, as for ivermectin, cleft
palates were seen at or near matemotoxic doses. In multigeneration
TABLE 6.5. Comparative toxicity of abamectin and ivermectin in laboratory
animals.
Type of Abamectin Ivermectin
Study Species (mg/kg/day)
Oral LD50 CFt mouse 13.6-23.8 24.6-40
Oral LD50 CRCD rat 10.6-11.3 42.8-52.8
Oral LD50 CRCD 1.52 2.3
neonatal
rat
Acute toxicity Monkeys MEL = 2 mg/kg MEL = 2 mg/kg
Minimum Effect Level (MEL)
(mg/kg/day)
Abamectin Ivermectin
Suckling CRCD rat 0.12 0.4
neonatal
pup
Subacute oral Beagle dog 0.50 1.0
Chronic and CRCD rat 2.0' 0.8
subchronic
• 105-week study
104 George R. Lankas and Lea R. Gordon
TABLE 6.6. Results of teratology studies with abamectin and ivermectin in three species.
Mouse Rabbit Rat
Abamectin Ivermectin Abamectin Ivermectin Abamectin Ivermectin
Maternotoxicity
NEL" 0.05 0.1 1.0 3.0 1.6 5.0
MELb 0.075 0.2 2.0 6.0 2.0 to.O
Fetotoxicity
NEL 0.2 0.8 1.0 1.5 1.6 10.0
MEL 0.4 2.0 3.0
Developmental
toxicity
NEL 0.2 0.2 1.0 1.5 1.6 5.0
MEL 0.4 0.4 2.0 3.0 to.O
studies pup toxicity was the most sensitive parameter, with minimum
effect levels of 0.4 mg/kg/day for both compounds. Additional long-term
studies of abamectin are discussed below (Table 6.7).
male and 65 female rats each: Control I, Control II, and abamectin given
in the feed at 0.75, 1.50, and 2.0 mg/kg/day. The levels of abamectin in the
feed were adjusted and mixed biweekly to attain the targeted dose levels.
Treatment-related increases in body weight gain were seen in both
males and females in all dosage groups during the first year on study. At
106 George R. Lankas and Lea R. Gordon
drug week 60, when the peak difference in body weight gain was apparent
between treated and control rats, the treated females had 30%, 14%, and
13% increased body weight gain in the 0.75, 1.5, and 2.0 mg/kg/day
groups, respectively, when compared with controls. By the end of the
study (drug week 105) there were comparable mean body weight gain
values for all groups of females when averaged over the entire study
(P > 0.05); however, the increases in body weight gain over the com-
bined controls among the treated males were still statistically significant
(P s 0.05) (Robson 1959) at drug week 105 (21%, 9%, and 6% increases in
the 0.75, 1.5, and 2.0 mg/kg/day groups, respectively).
Clinical signs of toxicity were limited to tremors seen in a few rats in the
high-dose group and an occasional rat in the mid-dose group that
consumed enough feed to exceed the 2.0 mg/kg/day dose ofthe high-dose
group. Although some of the rats died with tremors, there were no gross
or microscopic changes seen in the nervous or muscular systems to
account for these tremors. This has been true for other species as well.
Abamectin was not carcinogenic in rats when given in the diet at doses
of 0.75, 1.5, and 2.0 mg/kg/day for 105 weeks. There was no statistically
significant (P > 0.05) increase in tumor incidence resulting from treatment
with abamectin (Mantel 1963, 1980; Mantel et al. 1982; Peto 1974; Peto et
al. 1980; Harter 1957; Cutler and Ederer 1958; Tukey, Ciminera, and
Heyse 1985). There were also no treatment-related nonneoplastic histopa-
thologic changes in this study. The no-effect level for toxicity was
1.5 mg/kg/day.
In a chronic toxicity and carcinogenicity study, Crl:CD-l (ICR) BR
mice, about 6 weeks old, were assigned to 5 groups: Control I, Control II,
2,4, and 8 mg/kg/day of abamectin in the feed.
Treatment-related tremors were seen in drug weeks 89 and 91 in 2
female mice in the high-dose group.
Increased mortality was seen among the high-dose males (P < 0.05) but
not the females (Mantel and Ciminera 1979). Dosing was stopped among
the high-dose males in drug week 90 due to the low number of survivors
(40%). All other mice were treated until sacrifice in drug week 94.
Treatment-related decreases in body weight gain (P < 0.05) were seen
among both the high-dose males and females (7% and 21%, respectively)
(Robson 1959). A dose-related trend toward increased food consumption
(g/kg) was seen among the females but not the males, and the high-dose
females had 20% decreased feed efficiency compared to controls (Tukey,
Ciminera, and Heyse 1985).
There were no treatment-related ophthalmic changes. There were no
effects of treatment on the hematologic or serum biochemical findings.
There were no treatment-related organ weight changes or gross lesions.
Abamectin was not carcinogenic to mice when given in the diet for 94
weeks at doses of 2, 4, and 8 mg/kg/day. There was no statistically
significant (P > 0.05) increase in tumor incidence reSUlting from abamec-
6. Toxicology 107
tin treatment (Mantel 1963, 1980; Mantel et al. 1982; Peto 1974; Peto et al.
1980; Harter 1957; Cutler and Ederar 1958; Tukey, Ciminera, and Heyse
1985). The no-effect level for clinical evidence of toxicity is 4 mg/kg/day.
These results demonstrate that abamectin, the close structural analog of
ivermectin, is not carcinogenic in rodents when administered at maximum
tolerated doses for 22 to 24 months.
Amano Y (1967) Changes of the levels of blood glucose during pregnancy in the
rat. lap. l. Pharmacol. 17:105-114
Ames BN, McCann J, Yamasaki E (1975) Methods for detecting carcinogens and
mutagens with the Salmonella/mammalian microsome mutagenicity test.
Mutat. Res. 31:347-364
Betz L, Goldstein GN (1981) Developmental changes in metabolism and transport
properties of capillaries isolated from rat brain. l. Physiol. 312:365-376
Bohr V, Mollgard K (1974) Tight junctions in human fetal choroid plexus
visualized by freeze-etching. Brain Res. 81:314-318
6. Toxicology 111
Tukey JW, Ciminera JL, Heyse JF (1985) Testing the statistical certainty of a
response to increasing doses of a drug. Biometrics 41:295-301
Wester RC, Maibach HI (1975) Percutaneous absorption in the rhesus monkey
compared to man. Toxieol. & App. Pharm. 32:394-398
Wester RC, Maibach HI (1983) Cutaneous pharmacokinetics. 10 steps to percuta-
neous absorption. Drug Metab. Rev. 14(2): 169-205
Wilkins RJ, Hart RW (1973) Preferential DNA repair in human cells. Nature
247:35-36
Williams GM, Laspia MF, Dunkel, VC (1982) Reliability of the hepatocyte
primary culture/DNA repair test in testing coded carcinogens and non-
carcinogens. Mutat. Res. 97:359-370
CHAPTER 7
Pharmacokinetics of Ivermectin in
Animals and Humans
David W. Fink and Arturo G. Porras
I. Introduction
II. Cattle
III. Sheep
IV. Dogs
V. Swine
VI. Horses
VII. Humans
Introduction
The pharmacokinetic properties of ivermectin are a function of the
species in which the compound is studied. Ivermectin is effective against
parasites in a wide variety of hosts-including cattle, sheep, dogs, swine,
and horses. A previous review of ivermectin has described the effects of
formulation and route of administration on its pharmacokinetic properties
in animals (Lo et al. 1985). That publication included examples of
formulation modifications directed toward the development of oral and
parenteral dosage forms; it also illustrated the use of drug plasma
concentrations for characterizing the drug and for modifying formulations
for specific efficacy. The following summary presents the results of
representative pharmacokinetic and bioavailability studies of this drug in
different animal species and in humans. It includes descriptions of the
experimental procedures as well as the various measurement techniques
used in these studies to describe the pharmacokinetic properties of
ivermectin.
114 David W. Fink and Arturo G. Porras
Cattle
Commercially, ivermectin is most widely used for treating cattle. Bio-
availability studies have been run in this species using intravenous and
subcutaneous injectable formulations. To measure the intrinsic pharma-
cokinetic properties of ivermectin in cattle, the tritium-labeled compound
was formulated in a solution of 0.07 mCi/mg specific activity at a
concentration of 10.0 mg/ml; the solvent consisted of 60% (v/v) propylene
glycol and 40% (v/v) glycerol formal (Hibbert and Carter 1928) containing
5% polyvinylpyrrolidone. This solution was administered to cattle in a
single intravenous bolus at a dose of 300 ILg/kg body weight.
The pharmacokinetic results are shown in Figure 7.1. The data exhibit a
typical biexponential decay based upon the usual 2-compartment open
model.Figure 7.1 is a plot of the total concentration in plasma over time
following IV administration as determined by HPLC and radiochemical
analyses. The close agreement between the radioactivity data and the
HPLC data (the average relative difference is 5%) confirms that there are
no metabolites of ivermectin in the plasma over this time interval. This is
important in bioavailability studies, because the intact drug must be
discriminated in the plasma from possible degradates and metabolites of
similar structures. This selectivity is especially important for ivermectin,
because the structure is so complex that there are many structural
changes and degradation processes that can occur. For example, one
metabolic transformation of ivermectin (resulting from incubation at 37°C
with rat or steer liver microsomes) is the formation of the C(24)-
hydroxymethyl compounds and their respective monosaccharides (Miwa
et al. 1982).
The bioavailability analyses in this study used a sensitive high-pressure
liquid chromatographic (HPLC) analytical method. This method uses
fluorescence detection after preliminary sample preparation by liquid-
liquid partitioning and conventional gravity-fed column chromatography
(Tolan et al. 1980; Fink 1982). The potency ofivermectin requires the use
of extremely sensitive analytical procedures. This derivatization tech-
nique was developed to increase sensitivity for detection by forming an
intensely fluorescent compound based upon an analytical aromatization
reaction.
The results of the experiment depicted in Figure 7.1, in addition to the
biexponential decay, reveal a very short distributive phase in cattle, k = 6
day-I, a biological half-life t1l2 of 2.8 days, a very large volume of
distribution, 1.911kg (consistent with the lipophilicity of this compound),
and an area under the curve (AUC) of about 700 day-ng/ml. In an
independent study, application of a 3-compartment model to the data
obtained from 6 cattle also yielded an equal biological half-life t1l2 = 2.7
days. In this study, the 3-compartment (triexponential) model provided a
statistically significantly closer fit to the data than the 2-compartment
7. Pharmacokinetics ofIvermectin in Animals and Humans 115
1500 r----r----.---~----,----.r---._----._--~
o HPLC
1000
[J Radiotracer
700
E
.....
C>
c:
C 500
0
~
....
E
CI)
0
400
c:
8
c:
U
CI) 300
E
....
CI)
~
200
o 2 4 6 8 10 12 14
Time (hr)
FIGURE 7.1. Comparison of bioavailability analyses and radiotracer counts in
cattle plasma following intravenous dose. Data for 0.5 day following injection.
Dose rate: 300 ILg/kg body weight. Specific Activity: 0.07 mCi/mg.
demonstrated that the drug is carried mainly in the plasma (80%), and that
this distribution equilibrium between plasma and red blood cells remains
relatively constant with time.
Figure 7.2 depicts the results of a replicate study in 2 cattle measured
by chemical analysis and extending out to 4 weeks following the intrave-
nous dose. The plasma samples again were collected at specific time
intervals and analyzed as described previously (Tolan et al. 1980). The
insert in this figure shows the rapid distribution phase in cattle: the
biexponential decay mode is more clearly evident in this expanded scale.
1000
~,0,0...
300 300
0.......,
0-----"0
-E01
C 60
C
0
-:0=
c
~
30
C
CI)
(.)
c
0
(.) 10
c 6
-:0=
(.)
CI)
E
~ 3
~
1.0
0.6
0.3
0
Time, days
FIGURE 7.2. Plasma concentration of ivermectin in cattle for 1 month following
intravenous administration. Mean of 2 cattle.
7. Pharmacokinetics of Ivermectin in Animals and Humans 117
52
~t:Jl •
c:
C 39
('-\
0
+= • •
....
0
cCD
()
c: 26
0
()
c:
"-"-
+=
13r
()
CD
....E
CD
.2
•
00
0
I 6
I
12
----...-.-
18
.
24 • 30
Time, days
FIGURE 7.3. Bioavailability of ivermectin in cattle dosed subcutaneously with
IVOMEC propylene glycol-glycerol formal [60: 40 (v/v)] vehicle at 200 ~g/kg
body weight. Formulation concentration: 10 mg/ml ivermectin. Each point is the
average plasma concentration obtained from 10 cattle.
118 David W. Fink and Arturo G. Porras
90
0
80
70
~01 4-'
c: 60
--e
C
0
.2
50
\
cQ):
u
c:
8
0
40
c:
:;::
u
/0,\
Q)
E
~
30
~
0t2 '---
20
10
-
o~o ___
12 14
Time,doys
FIGURE 7.4. Effect of aqueous/nonaqueous solvent ratio on bioavailability of
ivermectin following subcutaneous administration to cattle at 200 ~g/kg body
weight. Formulation 4-1 is an aqueous (micelle) solution; 4-2 is an aqueous-
glycerol formal 50 : 50 (v/v) mixed solvent vehicle. (Modified from Lo et al. 1985.)
7. Pharmacokinetics of Ivermectin in Animals and Humans 119
Sheep
For administration to sheep, ivermectin was dissolved in a mixed solvent
of 60% (v/v) propylene glycol-40% (v/v) glycerol formal containing 5%
polyvinylpyrrolidone; it was given as a single intravenous bolus. The
concentration of the solution for this species was 2 mg/ml ivermectin and
the dose rate was 300 JLg/kg body weight. Plasma samples were collected
at specific time intervals and analyzed for ivermectin.
The points shown in Figure 7.5 are the averages over 4 animals. At each
sampling point the range of results was ± 15% or less. The results in sheep
were analogous to those for cattle. Plasma concentration as a function of
time clearly exhibited a biexponential decay for sheep. The biological
half-life was again 2.7 days, very similar to that obtained in cattle.
However, there is a significant difference in the volume of distribution of
ivermectin between the 2 species. Volume of distribution, Vd, was
120 David W. Fink and Arturo G. Porras
,
1000
700
•
4oo~
300
200 2 4 6 8 10
'C
E Time (hrl
.....Cl
-c:
100 0,
--e
c:
.2
c:
CD
80
70
60
40
Q
4
3
2
•
10 14
Time (days)
FIGURE 7.5. Biexponential decay of ivermectin concentration in plasma follow-
ing intravenous administration to cattle (0), sheep (e), and dogs (.6.). Dose rates:
300 ~g/kg body weight for cattle and sheep; 200 ~g/kg for dogs.Insert depicts the
rapid distribution phase in cattle. (Points are average from: 2 cattle, 4 sheep, and
5 dogs, respectively.) (Reproduced, with permission, from Lo et af. 1985.)
7. Pharmacokinetics of Ivermectin in Animals and Humans 121
Dogs
The dosing solution for intravenous administration to dogs was a 4-mg/ml
solution of ivermectin in the aqueous micellar formulation, which uses a
surface active agent to dissolve the drug (Lo and Williams 1983). This
solution contained 8% (w/v) polyoxyethylene sorbitan monooleate sur-
factant, 20% (w/v) glycerol formal cosolvent, and 1% benzyl alcohol; pH
was regulated to 6.2 with IN He 1. The injection was given to 5 dogs, each
at a dose of 200 JJ.g/kg body weight.
The procedure used for these bioavailability studies in dogs was a direct
method, based upon ultraviolet photometric detection of the drug using a
chemical derivative (the a2 isomer) of ivermectin as an internal standard
(Pivnichny, Shim, and Zimmerman 1983). This procedure was adapted to
a laboratory robotic system to increase the number of data points
available in these pharmacokinetic studies (Pivnichny, Lawrence, and
Stong 1987).
The curve obtained for the plasma concentration in dogs over time
(Figure 7.5) shows a greater slope than that obtained from the ruminants.
The results in dogs reveal a significant species difference in that iver-
mectin is eliminated (biotransformation, distribution, and excretion) more
rapidly (t1l2 = 1.8 days) than in either cattle or sheep. This measurement
of the terminal half-life in dogs is in accord with results recently reported
by Dainippon in Japan, which yielded a t1/2 = 1.6 days in dogs (Kojima,
Yamamoto, and Nakanishi 1987). The pharmacokinetic properties be-
tween species are summarized for comparison in Table 7.2.
The widespread use of ivermectin in a tablet formulation
(HEARTGARD-30) to prevent heartworm disease in dogs in a monthly
dosage regimen of only 6 JJ.g/kg is a dramatic illustration of the potency of
this drug. Bioavailability studies of these tablets document the rapid oral
absorption achieved by this dosage form. The results, shown in Figure
7.6, demonstrate that the plasma level obtained from the tablets peaks
within 2 to 4 hours and decays exponentially beyond the peak. The dose
used for this study was 100 JJ.g/kg, to provide sufficient precision of the
replicates relative to the sensitivity of the measurement process.
50
45
40
E
0, 35
c:
r::
0 30
~
E
~ 25
c:
0
0
c: 20
ts
II)
...
E
II)
15
~
10
0
0 5 10 15 20 25 30 35 40 45 50
Time, (hours)
FIGURE 7.6. Bioavailability of ivermectin in dogs dosed with HEARTGARD
tablet formulation at 100 #Lg/kg body weight. Each point is the average of 8 dogs.
Swine
One marked example of the effect of route of administration on bioavail-
ability can be found in a bioequivalence study in swine treated with
ivermectin by subcutaneous injection and by oral administration. Two
groups of 4 pigs were each given ivermectin at a dose of 200 JLg/kg
parenterally in a propylene glycol-glycerol formal (60: 40 v/v) vehicle or
orally in propylene glycol solution. The drug concentration in these
dosing solutions was 10 mg/ml ivermectin. Plasma samples were collected
at appropriate time intervals and analyzed using the fluorogenic-
derivatization technique described previously (Tolan et al. 1980). The
results are shown in Figure 7.7.
Mter either subcutaneous or oral administration of ivermectin to swine,
the plasma concentration of the drug reached a peak and then declined
exponentially with time. It is significant that the terminal portions
resulting from these 2 dosing regimens are nearly parallel, suggesting that
ivermectin is eliminated rapidly in swine following either route of
absorption. The concentration of ivermectin in the plasma, however,
attains its peak, tp , faster following the oral dose than the parenteral: that
these peaks appeared at 0.5 days and 2 days, respectively, demonstrates a
higher absorption rate via oral administration than subcutaneous. These
results in swine are in accord with the bioavailability obtained with
subcutaneous injections in cattle and sheep. The slower absorption
associated with the parenteral route has been attributed to precipitation of
the drug at the injection site (Lo et al. 1985). The area under the plasma
concentration-vs.-time curve (AUC) was larger after subcutaneous ad-
ministration, indicating that a greater fraction of the dose is absorbed than
after oral dosing. The bioavailability of ivermectin after oral administra-
tion, relative to that after subcutaneous administration, was estimated to
be 41%.
The feed route offers another opportunity to study the bioavailability of
ivermectin in swine following oral administration. Two groups of 6
Yorkshire barrows were each given a single oral dose of 400 JLg/kg body
weight ivermectin in premix formulations prepared for homogeneous
distribution in the swine feed. One premix formulation was composed of
0.22% ivermectin adsorbed on ground corn cobs. The second premix was
prepared to contain 0.6% ivermectin adsorbed on the same ground corn
cob substrate. Plasma samples were collected from the pigs during 14
7. Pharmacokinetics ofIvermectin in Animals and Humans 125
22
20
18
E
0,
c 16
c·
gc
0
14
Q)
0
12
c
8 10
til
E
t/)
til 8
a.
c
U 6
Q)
E
Q) 4
~
2
0
0 2 3 4 5 6 7 8
Time, (days)
days following the dosing, and these samples were analyzed for iver-
mectin. These bioavailability studies were also used to evaluate the
efficacy of ivermectin in feed formulations. Two studies were run, with
swine dosed at 400 I-tg/kg body weight in all the feed consumed: in both
studies, the drug reached its peak concentration in the plasma within 4 to
8 hours, in agreement with the results of oral administration shown in
Figure 7.7. These results are shown in Figure 7.8. Note that the drug is
rapidly adsorbed by the oral route, that it is eliminated rapidly from swine
(t1l2 ~ 0.5 day), and that good experimental agreement in the bioavail-
ability results was found between these 2 independent swine oral studies.
Horses
One additional clear example of the significant effect of formulation and
route of administration on the bioavailability of ivermectin has recently
been reported in a study in pregnant mares. A paste formulation and an
aqueous micellar formulation of this drug were administered (Asquith et
126 David W. Fink and Arturo G. Porras
20~---r----'-----r---~-----r----~
15
c
o
-
:;:
2
c
CD
u 10
c
o
u
c
~
E
~
CD
..:i 5
Time (hours)
al. 1987). The dose for both treatments was 200 JLg/kg, and the liquid
micellar solution (1%) was administered by nasogastric intubation. A
striking difference in bioavailability was found; the solution is absorbed
much more rapidly than the paste product. Although the liquid formula-
tion attained its peak concentration within 4 to 5 hours, the paste product
took 15 hours to reach its maximum plasma concentration. Accordingly,
the peak plasma concentration obtained from the solution was higher than
that offered by the paste, and the bioavailability (AUC) was 20% higher
following the nasogastric oral liquid dose.
Humans
Disposition of ivermectin in humans was studied in 4 healthy volunteers
after an oral dose (no i. v. formulation exists) of 3H-ivermectin labeled in
7. Pharmacokinetics of Ivermectin in Animals and Humans 127
1------.
100 . - - - - - - - - - - { I--------------------~
---;\~ ....'-----......
E
-.!!!.c-
....... 10 Metabolites
Cl
c:
Ivermectin
o 10 20 40 60 80
Time (hours)
FIGURE 7.9. Mean concentrations ofivermectin and metabolites in human plasma
after administration ofa 14-mg dose of 3H-labeled drug (average from 4 subjects).
128 David W. Fink and Arturo G. Porras
E
c,
c: 80
c::0
~ 60
'E
Q)
0
c: 40
8
c:
U
Q) 20
E
~
Q)
~
Time (hours)
60
E
-- CI
c:
50
.2
c:: 15mg
40
iU
-=c:
Q)
0 30
c:
0
0
c: 20
U
CD
E
~
CD 10
~
0
0 2 4 6 8 24 36 48 72
Time (hours)
REFERENCES
Asquith RL, Lane TJ, Plue RE, Seward RL, Kivipelto J (1987) The bioavailability
of ivermectin in horses when administered in a liquid formulation by nasogastric
intubation versus in an oral paste. Equ. Vet. Sci. 7:28-30
Campbell WC, Benz GW (1984) Ivermectin: a review of efficacy and safety.
J. Vet. Pharrn. & Therap. 7:1-16
Fink DW (1982) Some specific ftuorogenic reactions in pharmaceutical and
environmental applications. Trends in Anal. Chern. 1:254-258
Fink DW (in press) Ivermectin: analytical profile. In Florey K (ed) Analytical
Profiles of Drug Substances, Academic Press, New York
Hibbert H, Carter NM (1928) Studies on the reactions relating to carbohydrates
and polysaccharides. XVII. Structure of the isomeric methylidene glycerols.
J. Am. Chern. Soc. 50:3120-3127
Kojima K, Yamamoto K, Nakanishi Y (1987) Determination of 22,23-
dihydroavermectin B'a in dog plasma using solid-phase extraction and high-
performance liquid chromatography. J. Chrornatogr. 413:326-331
Kojima K, Yamamoto K, Katae H, Nakanishi Y (1987) Bioavailability of oral
ivermectin in dog [sic]. Nippon Juigaku Zasshi 49:899-900
Lo P-KA, Fink DW, Williams JB, Blodinger J (1985) Pharmacokinetic studies of
ivermectin: effects of formulation. Vet. Res. Cornrnun. 9:251-268
Lo P-KA, Williams JB (1983) Solubilization of ivermectin in water. U.S. Patent
4,389,397
Marriner SE, McKinnon I, Bogan JA (1987) The pharmacokinetics of ivermectin
after oral and subcutaneous administration to sheep and horses. J. Vet. Pharrn.
Therap. 10:175-179
Miwa GT, Walsh JS, VandenHeuvel WJA, Arison B, Sestokas E, Buhs R,
Rosegay A, Lu AYH, Walsh MAR, Walker RW, Taub R, Jacob TA (1982) The
metabolism of avermectins B'a, H 2B'a, and H 2B'b by liver microsomes. Drug
Metab. Dispos. 10:268-274
Pivnichny JV, Lawrence AA, Stong JD (1987) A robotic sample preparation
130 David W. Fink and Arturo G. Porras
I. Introduction
II. Tissue Residues in Cattle, Sheep, and Swine
III. Absorption, Distribution, and Excretion in Rats
IV. In Vitro Liver Metabolism of Ivermectin
V. In Vivo Liver Metabolism of Ivermectin
VI. In Vivo Fat Metabolism of Ivermectin
VII. Conclusion
I. Introduction
Ivermectin is used widely as an antiparasitic agent in food-producing
animals. As in the case of any such drug, the residual tissue concentration
of the therapeutic agent, or tissue residue, is a safety concern to the
meat-consuming public. To evaluate the toxic potential of the residual
tissue concentration of ivermectin and its metabolites, metabolism studies
have been carried out in target species (cattle, sheep, swine) using the
radiolabeled drug. Comparative metabolic studies were done in a labora-
tory animal, the rat, and in liver microsomes from various species.
Radiolabeled ivermectin was synthesized either by incorporating tri-
tium atoms in the C5 position by oxidation of the C5-alcohol to ketone
with manganese dioxide followed by reduction back to the alcohol with
sodium borotritide, or in the C22-23 positions by catalytic tritiation of
avermectin. In these synthetic reactions, the 2 components ofivermectin,
dihydro-B 1a (H 2B 1a) and dihydro-B 1b (H 2B 1b) are synthesized and purified
separately and then mixed in a ratio of 80: 20 with respect to H 2B 1a to
HzB 1b for dose preparation. Most of the studies were carried out with
C22-23-labeled drug (22,23-[3H]ivermectin). The tritium label has been
shown to be stable by volatility studies (Jacob et al. 1983).
This chapter addresses only those studies on ivermectin tissue residue
132 Shuet-Hing Lee Chiu and Anthony Y.H. Lu
a Results are from studies in which all species except swine were dosed at 0.3 mg/kg body
weight, subcutaneously (steers), intraruminally (sheep), or orally (male rat). Swine were
dosed SC at 0.4 mg/kg body weight. Unchanged drug H2B1a and H 2B1b were analyzed by
RIDA.
b Not calculated due to insufficient data.
134 Shuet-Hing Lee Chiu and Anthony Y.H. Lu
and sheep, probably because the fat tissues of these species eliminate
nonpolar metabolite conjugates less efficiently. This phenomenon will be
discussed at more length in the section on In Vivo Fat Metabolism
of Ivermectin. There is no difference between liver and fat residue
depletion in swine.
The distribution pattern of tissue residue remains unchanged when the
drug is dosed via different routes. Of the 3 routes-subcutaneous,
intraruminal, and percutaneous-in steers, the intraruminal dosing gives
the lowest residue levels in liver and fat. The depletion half-lives,
however, vary with the dosing method. Residue levels in the brain re-
main the lowest by all dosing routes (S.H.L. Chiu, M. Green, F.P. Baylis,
D. Eline, and T.A. Jacob, unpublished results).
Radiolabeled ivermectin is excreted mainly in feces in all 4 species
studied. Regardless of whether the drug was administered intraruminally
or subcutaneously to cattle, only 0.5% to 2.0% of the dose was eliminated
in urine; the remainder appeared in feces (Jacob et al. 1983). Recovery of
the radioactive dose 7 days after dosing varies between 62% to 83% in
cattle, sheep, and rats.
2 '·'v'lung
• OL"...
"- Hearl
1 X
X
, ". o"::::-L "'v• -___
--ol.:;:-r.~.
X"_.('
0_
~.
",.....
--"'[01 __
,..... ~'7-.X·-·-.- __ x........ --_
..~ .............. -0
.6 /
/ Kidney "> . ~..
' ........ ~
Kidney
o llvi'f·...
..... ...... ..;:....... -- Hear I
'. /
.3
I ~
f-X~ .... ~o
E
-11
I
----....x Lung jt
0.
0.
o
..
ClJ
.1
~
I.'
"'C
II>
ClJ X
0::
.06
I '
' ......
....... _ ---....... Blood
' ...
.03 ..........................
"x- --x-···_·-·x·- __ -•• .x.,.........Br·oln ...
.....
X ."',
' . ...., .. ,
.01~--~--~----~--~--~~--~----u
o 4 8 12 16 20 24 28
TABLE 8.3. Total radioactive residues in blood and tissues of male Sprague Dawley rats
dosed subcutaneously at 0.3 mg/kg with 22,23-[3H]H 2B ,a or 22,23-CH]H 2B ,b . a
Tissue residues (ppm) Depletion til'
Post-dose I day 2 days 4 days (day)
Tissues H,B'a H,B'b H,B,. H,B'b H,B,. H,B'b H,B,. H,B'b
Liver 0.320 0.229 0.130 0.082 0.043 0.010 1.06 0.66
Kidney 0.220 0.185 0.118 0.068 0.041 0.009 1.25 0.69
Fat pad 0.493 0.497 0.274 0.168 0.110 0.027 1.40 0.72
Muscle 0.109 0.067 0.044 0.023 0.017 0.004 1.16 0.75
Lung 0.166 0.108 0.064 0.041 0.027 0.004 1.19 0.63
Heart 0.126 0.105 0.073 0.044 0.027 0.005 1.36 0.68
Blood 0.019 0.013 0.009 0.004 0.003 0.000 1.14 0.42
Plasma 0.023 0.017 0.012 0.005 0.004 0.001 1.20 0.75
• Three rats (200 g each) were dosed at each time point with radiolabeled drug at specific activity of
0.1 mCi/mg. Blood was collected by heart puncture. Fat pad was removed from the testicular areas
of the animals.
OH
6
OCH,
HO
HOn
A· B·
HlC 0 H,c 0
R, Rz R,
HaB,. A CHIQi, H
II HaB,. A CH, H
m HaB ,. - Monosacchoride H CHICH, H
m Ha B,. - Monosotc.horide H CH, H
'lZ' 24 -OH-HIB,. A CHaCH, OH
1ZI 24-0H-HzB,. A CHl OH
1ZII 24-0H-HzBl.-Mono~horide H CHICH, OH
"illI 24-0H-HzB,.-Mono,oCChoride H CH, OH
IX 3--0- [)nmelhyl-HIB,. B CHaCHl H
X 3--0- Oumethyl-HaB,. B C~ H
account for 69% of total metabolites (or 45% oftotal residue) in composite
liver-tissue samples from 7 and 14 days post dose animals. Confirmation
of in vivo metabolite structures has been achieved by chromatographic
comparison with in vitro metabolites either directly (in at least 2 modes of
HPLC separation), or after chemical derivatization (such as acid hydroly-
sis or fluorescence derivatization) (Chiu et al. 1987). These rigorous
comparative tests are carried out for the identification of all major
metabolites and some of the minor metabolites from cattle, sheep, rat,
and swine.
exception of swine, a group of metabolites less polar than the parent drug
is present in the fat tissue of these animals; they are particularly notable in
tissues of sheep and cattle, accounting for 60% to 70% of total residue in
the fat when these animals were withdrawn from dosing for a period of 5
or 28 days, respectively. Upon saponification or esterase treatment, these
nonpolar metabolites regenerate polar products that resemble chromat-
ographically the polar liver metabolites of ivermectin (e.g., 24-0H-
H2BIJ. Furthermore, the nonpolar metabolites give rise to products
identical to monosaccharide and aglycone of 24-0H-H2Bla when sub-
jected to p-toluenesulfonic acid-catalyzed methanolysis conditions (Fig-
ure 8.3). These results lead to the hypothesis that the polar ivermectin
metabolites produced in the liver are esterified as fatty acid esters and
stored in fat tissue as nonpolar entities. These nonpolar compounds can
be converted back to the polar metabolites chemically or enzymatically,
as shown in Figure 8.4 (Chiu et al., 1988). Two approaches were taken to
substantiate this hypothesis. First, the regeneration of polar metabolites
from the nonpolar metabolites was studied with cholesterol esterase, and
the structures of the hydrolysis products were confirmed as identical to
known liver metabolites (24-0H-H2Bla, -H2B 1b , -H2Bla-MS). Secondly,
samples of fatty acid (oleic acid) esters of 24-0H-H2Bla and -H2B 1b were
synthesized and directly compared with the nonpolar metabolites isolated
from a 2-kg fat sample from steers. These studies have established the
general structural class of the ivermectin nonpolar metabolites as being
the fatty acid conjugates of 24-hydroxymethyl derivatives of ivermectin.
The ivermectin metabolism pathway in fat tissue presents a rather
uncommon, but not unique, phenomenon in xenobiotic metabolism.
Although formation of less polar metabolites deviates from the most
common metabolic pathway-in which more polar metabolites are
formed for readiness of eliminations-several precedents have been
reported in recent years (Caldwell and Marsh 1983). Formation of fatty
acid esters of xenobiotics containing an alcoholic hydroxyl group, in
particular, have been reported for tetrahydrocannabinol (Leighty,
Fentiman, and Foltz 1976), pyridopyrimidine (Schmid et al. 1980), and
etofenamate (Dell et al. 1982). The metabolic pathway of ivermectin is an
interesting addition to these examples.
In contrast to cattle, sheep, and rat, swine do not follow the fat
metabolism pattern observed in the other three species. The fat metabo-
lite profile of swine resembles that of the liver tissue, i.e., 2 major
metabolites slightly more polar than the parent drug are present. These
are confirmed to be the 3"-0-desmethyl derivatives of H 2B 1a and H 2B 1b by
chromatographic comparison with authentic samples generated by swine
liver microsomes. The absence of ester conjugation of swine liver
metabolites in fat tissue can probably be attributed to the fact that there is
nO primary hydroxyl functional group in these metabolite structures. As a
result, these molecules are less favorable substrates for esterification
compared to 24-hydroxymethyl- H 2B 1a and -H2B1b produced in the livers
8. Metabolism and Tissue Residues 141
OH
polar metabolite
011
non-polar metabolite
in fat tissue
011
of the other 3 species. The similarity of liver and fat metabolite profiles in
swine explains the close resemblance of the residue depletion half-lives in
the two tissues of swine (Table 8.2).
VII. Conclusion
The absorption, excretion, distribution, and metabolism of tritium-labeled
ivermectin have been studied in target food-producing species (cattle,
sheep, swine) and a laboratory species (rat). The drug is absorbed rapidly
via subcutaneous, intraruminal, or oral dosing routes. Fecal excretion is
142 Shuet-Hing Lee Chiu and Anthony Y.H. Lu
.---------------- --- .
(In vitro) (In vivo)
I I
I I Steer, Sheep, Rat
,
I I
I I
I I
I
,
I
I I
I
.
I
I
Polar Non-polar
Ivermectin Metabolites Metabolites
Steer (Liver) (Fat)
Sheep
Rat A
I
I
I
:
I
I I :
I I I
I I I
I I I
I IL ________________________ ~I
I
- _____________ - _ - ____ 1I I :
FIGURE 8.4. Relationship of ivermectin metabolites in the liver and fat tissues of
sheep, rat, and steer with in vitro metabolites by steer and rat liver microsomes.
the major pathway of drug elimination in all species studied. Only about
2% of dose is excreted via urine. Highest residual tissue concentrations of
radioactivity are present in the liver and fat tissues, while lowest levels
are observed in the brain. Metabolism of radiolabeled ivermectin in liver
and fat of cattle, sheep, and rat is similar. Some differences are noted in
swine. The parent drug is the major liver residue in all four species in the
early withdrawal time period. The major liver metabolite in cattle, sheep,
and rat is 24-hydroxymethyl-H2B 1a , which is further conjugated to fatty
acids as esters and deposited in the fat tissue. The major liver metabolites
in swine are 3"-O-desmethyl-H2B 1a and 3"-O-desmethyl-H2B 1b • These
drug-like compounds are also present in fat tissue as major metabolites. In
vivo metabolism of ivermectin has been found to correlate closely with
that by in vitro liver microsomes of the same species.
REFERENCES
Liver 9.1 99
14 86,86
18 83
27 85,81
45 87
91 88
Range = 77%-110%
Average = 90%
Coefficient of Variation = ± 7%
Horse 1M 72 4.2,7.1,2.1
Oral 6 3.5
Pig SC 48 4.6
Acknowledgments
The development and application of the ivermectin tissue assay represent
the work of many people.
I would like to acknowledge the contributions of the following people:
Dr. Patricia C. Tway, Mr. James S. Wood, Jr., Dr. Teresa A. Wehner,
Ms. Krystyna T. Czeh, Ms. Eugenia H. Greff, Mr. William E. Tait, and
Mr. Richard J. Varsolona.
REFERENCES
Safety of Ivermectin
in Target Animals
J.D. Pulliam and J.M. Preston
I. Introduction
II. Safety in Horses
III. Safety in Cattle
IV. Safety in Sheep and Goats
V. Safety in Swine
VI. Safety in Dogs
I. Introduction
As part of the overall avermectin development program, target animal
safety studies were conducted in many countries by Merck personnel and
by independent investigators in universities and other research institu-
tions. In many instances anticipated commercial formulations were
studied, but during early development and when excessive dosing de-
manded, alternative or more concentrated formulations were used. The
various test formulations were compared for safety by bridging studies
involving assessment of tolerance and bioavailability. Control groups
were given vehicle, saline, or left untreated; many studies included both
treated and untreated controls.
V. Safety in Swine
The signs of acute toxicity to ivermectin in swine were observed in a
study where groups of swine were given 1, 10, 50, and 100 times the use
level of 300 ltg/kg. At 30.0 mg/kg, the pigs were lethargic and ataxic
within 24 hours following treatment. Bilateral mydriasis, intermittent
156 J.D. Pulliam and J.M. Preston
In a 14-week study with daily oral dosing, dogs given 1.0 mg/kg or more
developed mydriasis. At 2.0 mg/kg, 4 of 8 dogs additionally lost a small
amount of weight and developed tremors, ataxia, anorexia, and dehydra-
tion. Euthanasia and necropsy of these 4 dogs revealed agonal gastro-
intestinal hemorrhage and/or congestion. No other treatment-related
gross or histological changes were found (Norbury, unpublished). Sub-
cutaneous injection of ivermectin at 4.7 mg/kg produced mydriasis and
salivation, while depression, ataxia, and deaths occurred at 9.4 mg/kg
(Seward, Brokken, and Plue 1986).
In assessing the physiologic effects of ivermectin, an intravenous dose
of 0.5 mg/kg had no significant effect on blood pressure or heart rate of
anesthetized dogs, nor did it modify blood pressure or heart rate
responses to autonomic drugs in a standard assay. An intragastric dose of
0.5 mg ivermectin/kg did not affect evoked or basal gastric secretions in
dogs with chronic gastric fistula (Clineschmidt, unpublished).
Safety in breeding bitches was evaluated in 2 studies. A dose of
0.5 mg/kg was administered to 14 pregnant bitches on Days 5, 15,25, and
35 of gestation and to 15 pregnant bitches on Days to, 20, 30, and 40 of
gestation. No teratogenic effects were observed in 48-day fetuses from
these bitches treated repeatedly with ivermectin during the first 40 days of
gestation. Ivermectin also had no effect on the reproductive status of the
pregnant bitches, as measured by the number of implants, resorptions,
and live and dead fetuses per pregnant female (Robertson, unpublished).
In the second study, bitches were given repeated oral doses of
ivermectin at 600 JLg/kg at least twice at monthly intervals before
breeding, and then on Days 10,25, and 45 of gestation, and again monthly
after whelping. No adverse effect on reproduction was observed in
treated dogs as compared to the controls (Gilman, unpublished).
The effect of ivermectin on spermatogenesis, fertility, and the re-
productive performance of 12 Beagle studs was evaluated. Ivermectin
was administered orally at 0.6 mg/kg at monthly intervals for 8 months
while controls were given water. Semen was collected every 3 days from
28 days before treatment until 83 days after (38 collections/dog). All studs
were then bred to 2 nontreated bitches each; litter size, birth weight, and
pup abnormalities and mortalities were evaluated. After all pups were
whelped, each stud was euthanized and necropsied, and the testes and
epididymides were examined microscopically. No adverse effect on
reproduction was observed in treated dogs as compared to the controls
(Daurio et al. 1987).
Following extra-label use of ivermectin products formulated and
marketed for horses (EQVALAN®), and for sheep, cattle, and swine
(IVOMEC®), it appeared that, within the Collie breed, some dogs were
unusually sensitive to the toxic effects of ivermectin (Campbell, Blair,
and Seward, 1983; Seward, Blair, Plue, and Brokken, 1983). In a
10. Safety of Ivermectin in Target Animals 159
controlled study, 8 Collies affected with Collie eye anomaly (CEA) and 8
Collies clinically free of CEA were studied to determine whether iver-
mectin was more toxic to members of the Collie breed and if CEA was
related. Groups were given 50, 200, or 600 p,g ivermectin/kg orally. One
dog given 200 p,g/kg and 1 given 600 p,g/kg developed severe signs of
toxicity including ataxia, depression, tremors, recumbency, and mydria-
sis: 1 died and 1 was euthanized. Two other dogs in these groups had mild
transitory signs. Ivermectin tissue assays showed high concentrations of
ivermectin in the central nervous system of the 2 Collies that died. Collie
eye anomaly was not related to the response to ivermectin (Pulliam,
Seward, and Henry 1985). The range of Collie sensitivity to ivermectin
was further studied by Paul et al. (1987), who found that Collies are
sensitive to doses from 100 to 2500 p,g/kg, and that the length of time
between dosing and onset of clinical signs may indicate the eventual
severity of toxicosis and serve as an important prognosticator for the
clinician. Physostigmine, 1 mg b.i.d., was found to have limited benefit in
treating these dogs, and then only in the more severely depressed and/or
comatose stages of ivermectin toxicity. Its use as the sole therapeutic
agent or complete antidote for ivermectin toxicosis was not recommended
(Tranquilli et al. 1987).
The unapproved (extra-label or off-label) use of products designed for
large animals continues to result in iatrogenic toxic deaths in dogs
(Seward, Brokken, and Plue 1986). Many of these reactions appear to be
due to the accidental or erroneous administration of excessive amounts of
ivermectin (Preston 1983).
The only currently available formulation for the dog is a tablet to be
administered monthly at a dose rate of 6 p,g/kg for the prevention of
canine heartworm disease. This represents a wide margin of safety, not
only in normal dogs-where a single dose of 2.0 mg/kg and daily oral
dosing of 0.5 mg/kg for 14 weeks had no effect-but also in ivermectin-
sensitive Collies, which tolerate doses of 50 p,g/kg without effect (Pul-
liam, Seward, and Henry 1985; Paul et al. 1987).
In dogs with circulating microfilariae, vomiting, excess salivation,
blood in feces, soft stool/diarrhea, and depression have been observed
after ivermectin treatment (Schlotthauer et al. 1986). Although the
shocklike reaction in dogs infected with Dirofilaria immitis that develops
after administration of diethylcarbamazine did not occur following iver-
mectin treatment (Boreham and Atwell 1983), reactions following iver-
mectin treatment ofmicrofilaremic dogs may occur, and deaths have been
seen (Jackson, Seymour, and Beckett 1986).
The incidence of incidental concurrent adverse reactions, as reported
to the manufacturer and based on the sale of tens of millions of doses of
ivermectin tablets for use in dogs, is 0.001%. None of these reported
reactions has been confirmed to be caused by ivermectin.
160 J.D. Pulliam and J.M. Preston
REFERENCES
Asquith RL, Kivipelto J, Harvey JW, Bauer JE (1988) Comparative effects and
safety of ivermectin in pregnant mares. J. Equ. Vet. Sci. 8:32-35
Asquith RL, Kulwich R (1981) Safety and therapeutic activity of ivermectin as an
equine anthelmintic. J. Equ. Vet. Sci. 1: 18-20
Boreham PFL, Atwell RB (1983) Absence of shock-like reactions to ivermectin in
dogs infected with Dirofilaria immitis. J. Helminthol. 57:279-281
Brem JJ, Bulman GM (1986) An assay of biochemical parameters in cattle
medicated at therapeutic level with ivermectin. Vet. Argent. 3:365-373
Brokken ES, Barth D, Foster AG, Pulliam JD, Wallace DH (1983) Ivermectin: a
new broad-spectrum antiparasitic agent for swine. In Proceedings of the MSD
AGVET Symposium on Recent Developments in the Control of Animal Para-
sites, XXII World Veterinary Congress, Perth, Australia, pp 239-258
Button C, Barton R, Honey P, Rickford P (1988) Avermectin toxicity in calves
and an evaluation of picrotoxin as an antidote. Austral. Vet. J. 65:157-158
Campbell WC, Benz GW (1984) Ivermectin: a review of efficacy and safety.
J. Vet. Pharm. & Ther. 7:1-16
Campbell WC, Blair LS, Seward RL (1983) Ivermectin vs. heartworm: the present
status. Proc. 1983 Heartworm Symp., Orlando, Fla., pp 146-149
Daurio CP, Gilman MR, Pulliam JD, Seward RL (1987) Reproductive evaluation
of male Beagles and the safety of ivermectin. Am. J. Vet. Res. 48: 1755-1760
Egerton JR, Seward RL, Robin B (1983) Ivermectin as an antiparasitic agent for
horses. In Proceedings of the MSD AGVET Symposium on Recent Develop-
ments in the Control of Animal Parasites, XXII World Veterinary Congress,
Perth, Australia, pp 49-55
Herd RP, Donham JC (1983a) Efficacy of ivermectin against Onchocerca cervica-
lis microfilarial dermatitis in horses. Am. J. Vet. Res. 44:1102-1105
Herd RP, Donham JC (1983b) Control of equine cutaneous nematodiasis by
ivermectin. In Proceedings of the MSD AGVET Symposium on Recent Devel-
opments in the Control of Animal Parasites, XXII World Veterinary Congress,
Perth, Australia, pp 286-295
Herd RP, Kociba GJ (1985) Effect of ivermectin on equine blood constituents.
Equ. Vet. J. 17:142-144
Hotson IK (1983) The development of ivermectin as an antiparasitic agent in
sheep. In Proceedings of the MSD AGVET Symposium on Recent Develop-
ments in the Control of Animal Parasites, XXII World Veterinary Congress,
Perth, Australia, pp 42-48
Jackson RF, Seymour WG, Beckett RS (1986) Lower dose of ivermectin as
a microfilaricide, 0.05 mg/kg. In Proc. 1986 Heartworm Symp., New Orleans,
pp 15-18
Leaning WHD, Roncalli RA, Brokken ES (1983) The efficacy and safety
evaluation of ivermectin: A new injectable antiparasitic agent for cattle. In
Proceedings of the MSD AGVET Symposium on Recent Developments in the
Control of Animal Parasites, XXII World Veterinary Congress, Perth, Austra-
lia, pp 25-41
McKissick GE, Sutherland IH, Foix J, Olson G (1987) The safety of ivermectin
administered orally to pregnant mares. Equ. Vet. Res. 7:357-367
10. Safety of Ivermectin in Target Animals 161
Njanja JC, Bell JF, Wescott RB (1985) Apparent lack of toxicity in adult East
African goats on parenterally administered ivermectin. Bull. Anim. Health
Prod. Afr. 33:123-127
Paul AJ, Tranquilli WJ, Seward RL, Todd KS, DiPietro JA (1987) Clinical
observations in Collies given ivermectin orally Am. J. Vet. Res. 48:684-685
Preston JM (1983) Adverse reactions to unapproved applications (Letters to the
Editor) Vet. Rec. 112:286
Pulliam JD, Seward RL, Henry RT (1985) Investigating ivermectin toxicity in
Collies. Vet. Med. 80:33-40
Schlotthauer JC, Stromberg BE, Paul AJ, Todd KS, McCall JW, Dzimianski MT,
Blagburn BL, Hendrix CM (1986) Safety and acceptability ofivermectin in dogs
with naturally acquired patent infections of Dirofilaria immitis. Proc. 1986
Heartworm Symp., New Orleans, pp 29-32
SchrOder J, Swan GE, Barrick RA, Pulliam JD (1986) Effect ofivermectin on the
reproductive potential of breeding rams. J. So. Afr. Vet. Assoc. 57:211-213
Seaman JT, Eagleson JS, Carrigan MJ, Webb RF (1987) Avermectin B\ toxicity in
a herd of Murray Grey cattle. Austral. Vet. J. 64:284-285
Seward RL, Blair LS, Plue RE, Brokken ES (1983) The efficacy and safety of
ivermectin in dogs. In Proceedings of the MSD AGVET Symposium on Recent
Developments in the Control of Animal Parasites, XXII World Veterinary
Congress, Perth, Australia, pp 259-266
Seward RL, Brokken ES, Plue RE (1986) Ivermectin vs heartworms-A status
update. Proc. 1986 Heartworm Symp., New Orleans, pp 1-8
Tranquilli WJ, Paul AJ, Seward RL, Todd Jr. KS, DiPietro JA (1987) Response to
physostigmine administration in Collie dogs exhibiting ivermectin toxicosis.
J. Pharm. & Ther. 10:96-100
CHAPTER 11
Environmental Aspects of
Ivermectin Usage in Livestock:
General Considerations
B.A. Halley, R.J. Nessel, and A.Y.H. Lu
I. Introduction
II. Excretion of Ivermectin
III. Environmental Burden
IV. Soil Binding and Translocation
V. Stability in Soil and Photodegradation
VI. Toxicity to Soil Microorganisms
VII. Toxicity to Earthworms
VIII. Acute Toxicity to Freshwater Organisms
IX. Environmental Safety Assessment
X. Cattle Feedlot Environmental Safety Study
I. Introduction
A detailed analysis of ivermectin's effect on the environment was an
integral component of the overall program to develop ivermectin as an
antiparasitic drug for food-producing animals. The analytical studies were
designed to determine whether using ivermectin in animals would result in
any harmful or undesirable effects on the environment. These studies
measured ivermectin's physical properties and its mobility, distribution,
and stability in soil and water. Additional studies investigated the drug's
effect on a variety of environmentally important organisms. These
studies, combined with the clinical use pattern of ivermectin in cattle,
sheep, and swine, provided the means to assess ivermectin's environmen-
tal impact.
11. Environmental Aspects of Ivermectin Usage 163
100
-----------------{]
o 8 16 24
Cumulative exposure, hours
FIGURE 11.1. Photodegradation of H 2B1a • The half-life of tritium-labeled H 2B1a ,
exposed to sunlight ( • ) as a thin, dry film on glass in July in Rahway, New
Jersey, was about 3 hours. Foil-covered sample ( 0 ) was subjected to the same
conditions.
column or feces leachates limited the toxicity testing at only 1 or 2 concentrations. Not
enough data could be collected to accurately calculate the LC 50 values or the nontoxic
levels.
d Extrapolated value.
e Not determined. Toxicity observed at all concentrations in definitive test.
170 B.A. Halley, R.J. Nessel, and A.Y.H. Lu
safety. These levels are calculated from the ivermectin use pattern and
the rate of manure application. Based on the negligible effect at 30 ppb on
respiration and nitrification, and the no-effect level of 12 ppm on
earthworms, ivermectin would not adversely affect soil microbes and
other organisms. Phytotoxicity is also not expected, due to ivermectin's
lack of toxicity to freshwater algae over the concentration range of 1 to
10 ppm. Nor has phytotoxicity been observed with abamectin, the related
compound now being developed as a pesticide.
Aquatic organisms, such as water fleas and fish, are highly sensitive to
ivermectin toxicity. However, due to its low water solubility, high
octanol-water partition coefficient, and tight soil binding, ivermectin will
be immobile in soil and will not translocate or leach into ground water.
Based on the soil-binding data and the Freundlich equation, Halley and
colleagues (1989b) have calculated that the ivermectin concentration in
runoff and ground water should be below the no-effect level of 0.01 ppb
observed for D. magna (Table 11.5). Thus, in the environment, the
concentration of ivermectin in water should be sufficiently low to cause
no adverse effects on freshwater organisms. Other studies have shown
that ivermectin (or abamectin) does not appear to be very toxic to birds
(Kelso 1984), chickens (Mousa et al. 1986; Zeman 1987), ducks (see
Chapter 13), and mammals (Nessel, Jacob, and Robertson 1983). The
effect of ivermectin on manure-dwelling or -feeding insects is discussed in
Chapter 12.
11. Environmental Aspects of Ivermectin Usage 171
Acknowledgements
We would like to thank Drs. Peter Wislocki, William VandenHeuvel and
S.H.L. Chiu for their constructive comments on this manuscript.
REFERENCES
Environmental Aspects of
Use of I vermectin and Abamectin
in Livestock:
Effects on Cattle Dung Fauna
R.A. Roncalli
A series of trials (see Tables 12.1 and 12.2) was conducted in Australia to
examine the effects of ivermectin and abamectin fecal residues on larval
and adult stages of Onthophagus gazella. Cattle were treated once,
subcutaneously, at 200 or 300 I-'g/kg body weight. The trials were carried
out in sequential order, to assess the effect of the drugs' residues in feces
at different time intervals following the placement of the beetles.
In these trials feces from treated and untreated animals were placed in
buckets, along with pairs of O. gazella, and observed for the beetles'
viability, production of brood balls, and emergence of new beetle
populations for up to 35 days after treatment. The feces were not exposed
to sunlight, eliminating any effects resulting from degradation of iver-
mectin and abamectin (which may occur under field conditions). The
results of these trials indicate that neither ivermectin nor abamectin at the
above doses had any effect on adult O. gazella. Both compounds did,
however, prevent larval development: for ivermectin, up to 21 days
following treatment, for abamectin, up to 28 days.
B. ONTHOPHAGUS BINODIS
VI. Conclusions
Ivermectin and abamectin have an effect on some important dung fauna,
such as the larval stages of Diptera and of some dung beetles. The effect
on biting flies (i.e., the horn fly) could be considered beneficial, but its
practical impact under field conditions remains to be determined. Iver-
mectin and abamectin appear to affect dung beetles mainly in the larval
stages, and this could be considered harmful. The importance of these
180 R.A. Roncalli
REFERENCES
Bastiman B (1970) Problems of pasture contamination and herbage rejection under
intensive grazing. In 11th Progress Report of Experimental Husbandry and
Experimental Horticultural Stations, NAAS, pp 62-69
Doube BM (1986) Biological control of the buffalo fly in Australia: the potential of
the Southern Africa dung fauna. Misc. Pubs. Entomol. Soc. of Amer. (No.
61):16-34
Drummond RO (1987) Economic aspects of ectoparasites of cattle in North
America. In The Economic Impact of Parasitism in Cattle, MSD AGVET
Symposium, Montreal, Quebec, Canada, pp 9-24
Fincher GT (1986) Importation, colonization, and release of dung-burying scar-
abs. Misc. Pubs. Entomol. Soc. of Amer. (No 61):69-76
Halley BA, Nessel RJ, Lu AYH (1988) Environmental aspects of ivermectin
usage in livestock: general considerations. (Chapter 11)
Holm E, Wallace MMH (1987) The influence of superphosphate on the establish-
ment of introduced dung beetles in south-eastern Australia. J. Aust. Inst. Agric.
Sci. 5:202-203
Legner EF, Warkentin RW (1983) Questions concerning the dynamics of Ontho-
phagus gazella (Coleoptera:Scarabaeidae) with symbovine flies in the lower
Colorado desert of California. Proc. 51st Ann. Con! Calif. Mosquito & Vector
Contr. Assoc. pp 99-101
12. Effects on Cattle Dung Fauna 181
Environmental Aspects of
Abamectin Use in Crop Protection
P.G. Wislocki, L.S. Grosso, and R.A. Dybas
I. Introduction
II. Environmental Fate Of Abamectin
A. Photolysis
1. Water
2. Soil
3. Thin Film
B. Hydrolysis
C. Soil Metabolism
D. Solubility
E. Soil Binding
F. Plant Uptake
G. Bioconcentration in Fish
H. Fate in the Aquatic Environment
I. Fate in the Terrestrial Environment
J. Conclusions
III. Effects Of Abamectin On Non-Target Organisms
A. Aquatic Organisms
1. Invertebrates: Acute Toxicity
2. Fish: Acute Toxicity
3. Chronic Toxicity
4. Plants
5. Conclusions
B. Terrestrial Organisms
1. Soil micro-and macroorganisms
2. Bees
3. Birds
4. Mammals
5. Conclusions
IV. Risk Assessment
A. Aquatic organisms
B. Terrestrial organisms
V. Conclusions
13. Environmental Aspects of Use in Crop Protection 183
I. Introduction
The use of any chemical as a pesticide results in the exposure of the
environment to the chemical. The extent of this exposure depends on the
way the chemical is applied-pattern, rate, and frequency-as well as its
persistence in the environment. Humans can control all these parameters
save the last. A chemical's persistence is determined by its physico-
chemical properties: half-life in soil and water, photolytic stability, and
soil-binding ability. These same factors also affect the chemical's bio-
availability to living organisms.
In Chapter 20 Dr. Dybas discusses the agricultural uses of abamectin. It
will be used at a maximum rate of 0.025 lb active ingredient/acre
(28 g/hectare) 3 times/season, or at a maximum rate of 0.02 lb active
ingredient/acre (22 g/hectare) up to 10 times/season. Abamectin is
also being used by homeowners to control fire ants at a use rate of only
0.00011 lb active ingredient/acre (123 mg/hectare). The compound is
formulated in a bait used at a rate of more than a million particles/acre.
The bait particles are rapidly scavenged by foraging fire ants. This use will
not lead to significant exposure to the environment and therefore is not a
topic of discussion in this chapter.
The risk to nontarget organisms associated with the use of a pesticide
depends upon both its toxicity to nontarget organisms and their exposure
to the pesticide. As a pesticide, abamectin is very toxic to target pests. It
is also toxic to certain nontarget organisms; however, the pesticide's
properties and use patterns reduce the exposure of nontarget organisms.
In this regard abamectin has many positive attributes. It degrades rapidly
and its physiocochemical properties prevent it from bioconcentrating or
bioaccumulating in the environment. An analysis of abamectin's physio-
chemical properties and their effect on its environmental fate per se, as
well as the chemical's toxicity and risk to nontarget organisms, follows.
The nomenclature used in this chapter should be clarified. The product
that is being developed for agricultural use is designated abamectin. It
refers to a mixture of not less than 80% avermectin B la and not more than
20% avermectin Bib. (Figure 13.1). Certain studies presented here used
radioactively labeled avermectin Bla ; they are designated accordingly.
4' OCH 3
CH~ oJ-: H
O~:CH3 O. H
HO~ ~ "1314~
CH 30 12
CH3'
ABAMECTIN = AVERMECTIN B1
FIGURE 13.1. The structure of abamectin.
A. PHOTOLYSIS
1. Water
In a water photolysis experiment avermectin B1a was exposed to sunlight
and its degradation was followed by HPLC. Under these conditions
avermectin B 1a degraded with a half-life of less than 12 hours. Products
formed during the course of photolysis included a less polar degradate
identified as a photo isomer, the delta-8,9 Z isomer of avermectin B 1a , and
2 fractions more polar than avermectin B 1a, designated the moderately
polar and polar fractions. During the course of this degradation the
characteristic UV absorption at 245 nm is lost. (Absorption is due to the
presence of a diene moiety at carbons 8 through 11 in the molecule.) This
loss of absorbance also occurs when abamectin is photolyzed in organic
solvents (Mrozik et al. 1988). The terminal degradation products of water
photolysis were contained in the polar fraction. Its toxicity and that of the
other fractions are discussed later.
13. Environmental Aspects of Use in Crop Protection 185
2. Soil
When soil containing avermectin B la was exposed to sunlight on glass
plates, it degraded to mUltiple products rapidly, with a half-life of 21
hours.
3. Thin Film
Abamectin is exceptionally susceptible to degradation when it is present
on a surface as a thin film. Such degradation occurs even in the absence of
light; however, it is greatly accelerated in the presence of light. Both
under laboratory light and in simulated sunlight, a thin film of abamectin
degrades with an initial half-life of 4 to 6 hours. Both avermectin Bla
and avermectin BIb degrade at the same rate (J. MacConnell and R.
Demchak, personal communication). The degradation products include
the delta-8,9 isomer of avermectin B la and a polar and moderately polar
fraction. HPLC profiles of the polar degradates generated on plants and
on glass demonstrated that these degradates were similar. The degradates
on glass contained a multitude of components, some of which had from 1
to 6 additional oxygens compared to the parent compound. Other
components had lost the features of the macrocyclic ring, including the
loss of UV absorbance at 245 nm; only the sugar portion of the molecule
was recognizable by NMR or mass spectroscopy. These data indicate that
extensive degradation occurs when abamectin is exposed to light.
B. HYDROLYSIS
C. SOIL METABOLISM
D. SOLUBILITY
A major factor which determines the behavior of a chemical in the
environment is its solubility. Because abamectin is quite insoluble in
water, determining its actual solubility was very difficult. For this reason
the method of May and Wasik (1978) was used. By this dynamic method it
was determined that the solubility of avermectin B1a was 7.8 ppb in water.
E. SOIL BINDING
F. PLANT UPTAKE
used to treat previous crops. A rotational crop study (Moye et al. 1987)
was peJformed using 3 different soil types that had organic matter
contents from 0.14% to 38.3%. Although total applications of up to
0.36 lbs active ingredient per acre were used, there was no biocon-
centration of residues. Indeed, the highest average level of total radioac-
tive residues of avermectin B 1a found in any plant part was only 11.6 ppb.
This level is quite low compared to total radioactive residues in soil of
1070 ppb. Examination of the properties of the plant residues by
extraction indicated that 95% of the residues were not avermectin
B1a-related: they could not be extracted with acetone, which removes
avermectin B 1a and its degradates. Plant metabolism studies have shown
that radioactive fragments from avermectin B1a can be incorporated into
the natural products of plants. Therefore, the radioactive residue in the
plants is due to their uptake of small fragments of the avermectin B1a
molecule and the incorporation of 1- and 2-carbon fragments into natural
plant constituents.
G. BIOCONCENTRATION IN FISH
3.0
2.5
i='
0-
~
2.0
(.)
ABAMECTIN
z 1.5
0
(.)
(!) 1.0
0
....J
.5
0
0 1 2 4 8 15
TIME (DAYS)
FIGURE 13.2. Decrease in the concentration of abamectin (ppt) over time in the
water of tanks subjected to drift simulation.
of soil that had been sprayed with abamectin was analyzed at 0, 1,2, and 4
days. Abamectin degraded rapidly, with a half-life of 8 hours (Figure
13.4).
This simulated field study demonstrates that abamectin degrades
readily both on soil and in the aquatic environment.
2.0
1.6
1.2
i='
0- .8
0-
(.)
.4
z 0
0
(.) -.4
(!)
0 -.8
....J
-1.2
-1.6
-2.0
0124 8 15 31 52
TIME (DAYS)
FIGURE 13.3. Decrease in the concentration of abamectin (ppt) over time in the
water of tanks subjected to runoff simulation.
190 P.G. Wislocki, L.S. Grosso, and R.A. Dybas
2.0
1.6
CO 1.2
a..
-z
a..
() .8
0 .4
()
C!J
0
9
-.4
-.8
o 2 4
TIME (DAYS)
FIGURE 13.4. Decrease in the concentration of abamectin (ppt) over time in soil
sprayed directly with abamectin.
Soil dissipation studies and crop residue trials were petformed to deter-
mine the residues of abamectin on soil and crops (Tway et aI., unpub-
lished data). Because these studies are actual field studies, they most
accurately depict the residue levels to which terrestrial organisms will
actually be exposed. In the soil dissipation studies the initial levels of
abamectin in the top 2 inches of soil were from 10 to 20 ppb. This is V. to ~
of the theoretical level of 42 ppb expected to be found at zero time when
0.025 lb active ingredient/acre is applied to soil. Minimal residues were
found at the 2-to-4-inch level, and no residues were found at a soil depth
of 4 to 6 inches. Residues depleted rapidly and did not accumulate, even
after 12 weekly applications.
Residue data on crops were determined to establish tolerances in raw
agricultural commodities and food and feed additives. These residue
values can also be used to estimate the exposure of terrestrial organisms
to abamectin. Residue data are available for 4 crops: cotton foliage, celery
plants, citrus fruits, and tomatoes. The cotton foliage trials were per-
formed in South Africa at application rates different from those in the
United States when calculated at the U.S. rate of 0.02 lb active in-
gredient/acre, the initial residue values were approximately 1 ppm.
Within 3 days the residue values were approximately 5% of the initial
values. In 20 residue trials on celery plants, the plants were treated at 10
weekly intervals at a rate of 0.02 lb active ingredient/acre. At zero time
after the last of 10 weekly applications the average residue value in the
whole celery plants was 170 ppb. By 7 days after the application the
13. Environmental Aspects of Use in Crop Protection 191
residues had declined to 9.5 ppb, which is less than 6% of the initial
residue levels. These data demonstrate that even repeated applications of
abamectin do not result in accumulated residues-they deplete rapidly.
Residue results from cotton foliage and celery plants can be used as
surrogate data for grasses and leaves and leafy crops, respectively, in risk
assessments. Due to their small surface-to-mass ratios, citrus fruits and
tomatoes have lower initial residues of abamectin. The zero-time residue
levels in citrus fruits treated with 0.025 lb active ingredient/acre average
from 14 to 45 ppb. The residues declined to approximately 20% of initial
values by 7 days. The initial residue level of abamectin on tomatoes was
less than 17 ppb. By 3 days after the application the residue levels were
less than 5 ppb and usually less than 2 ppb, the limit of detection of the
analytical method. These data on citrus fruits and tomatoes can be used as
surrogate data for other fruits. Metabolism studies on citrus fruits (Iwata
et al. 1985; Maynard et al. 1989), cotton (Bull et al. 1984), and celery
(Moye et al., unpublished data) have also demonstrated the rapid
degradation of avermectin B'a on plants. These studies have shown that
the degradation products formed are the delta 8,9 isomer of avermectin
B'a, the moderately polar fraction, and the polar fraction. These fractions
are made up of multiple components. Because the polar fraction is the
major degradate, and because it is similar to the polar fraction generated
on glass, the glass-derived material was used for toxicology studies. The
results of these studies is discussed below. The delta 8,9 isomer is present
on plants at 10% to 20% of the total residue of it and its parent compound.
It also degrades at the same rate as its parent compound. Because the
delta 8,9 isomer is closely related to its parent, it was also tested for
toxicity in mice.
The soil dissipation and residue data indicate that, under field use
conditions, the residue levels of abamectin do not accumulate, even when
multiple applications are made; moreover, the residue levels deplete
rapidly, greatly limiting exposure.
J. CONCLUSIONS
A. AQUATIC ORGANISMS
3. Chronic Toxicity
The results of the environmental fate studies indicate that chronic
exposure to abamectin should be minimal and without effects. Its rapid
photolytic degradation and strong binding to sediment preclude its being
bioavailable to aquatic organisms in a chronic manner. Nonetheless, 3
chronic exposure studies have been performed (Table 13.4). Full life-
cycle studies have been done with daphnia and mysid shrimp. (Studies
were performed by Springborn Life Sciences, Wareham, MA.) An early
4. Plants
Abamectin is essentially nonphytotoxic, permitting its use even on
sensitive ornamental plants. Its phytotoxicity to nontarget terrestrial
plants will be discussed in Chapter 20. However, 2 studies were per-
formed to assess its phytotoxicity to aquatic plants. (Studies were
performed by Springborn Life Sciences, Wareham, MA.) These studies
indicated that abamectin was relatively nontoxic to duckweed (Lemna
gibba) and to freshwater algae (Selenastrum capricornatum). The con-
centration of abamectin calculated to decrease frond production in
duckweed by 50% was 3.9 ppm. A concentration greater than 100 ppm
would be required to reduce the algae cell proliferation by 50%.
5. Conclusions
Aquatic organisms vary greatly in their sensitivity to abamectin. Aquatic
plants are only minimally affected by high concentrations of abamectin.
Certain invertebrates, however, are quite sensitive, with LC so values in
the ppt range, while other invertebrates are much less sensitive. Fish are
fairly uniform in their response to abamectin and are 2 to 3 orders of
13. Environmental Aspects of Use in Crop Protection 195
B. TERRESTRIAL ORGANISMS
2. Bees
The toxicity of abamectin to bees was assessed in laboratory studies by
applying it directly to the bees, by putting it in their food, and by exposing
the bees to foliage that had been treated at various times prior to
harvesting (Atkins, personal communications). Abamectin was found to
be quite toxic to the bees. In 2 studies it had contact LC50 values of 0.002
and 0.017 JLg/bee at 24 and 48 hours, respectively. When abamectin was
fed to bees, it had an LD50 of 0.009 JLg/bee. However, citrus or alfalfa
foliage that had been treated with abamectin 24 to 48 hours earlier was not
toxic. Abamectin rapidly loses its toxicity to bees upon exposure to the
environment.
3. Birds
The bobwhite quail and mallard duck are the species normally chosen for
avian toxicity tests. In Table 13.5 the results of a number of studies with
196 P.G. Wislocki, L.S. Grosso, and R.A. Dybas
these two species are presented. The studies were performed by Wildlife
International (Easton, MD). Abamectin was not very toxic to the
bobwhite quail. The LD50 was greater than 2 g/kg. When quail were fed
abamectin in their diet for 5 days, followed by 3 days on untreated feed,
the LC50 was 3102 ppm. Mallard ducks were somewhat more sensitive:
the LC50 after 5 days of dietary exposure was determined to be 383 ppm.
An avian reproduction study was performed in the mallard duck. When
ducks were fed 3,6, or 12 ppm abamectin in their diet for 18 weeks, there
was no statistically significant effect on any parameter of reproduction.
These dietary levels are greater than those which would occur in the
environment.
4. Mammals
Mammalian toxicity data (discussed in detail in Chapter 6) indicate that
abamectin has an LD50 of approximately 10.0 mg/kg in mice. Studies
were also performed on the two degradation products, the delta 8,9
isomer of avermectin B 1a and the polar fraction. The delta 8,9 isomer is at
least eightfold less acutely toxic than its parent compound, but it
possesses the same level of subchronic toxicity. For this reason residue
data now include the level of this isomer. The polar fraction isolated from
the degradation of abamectin on glass is more than 500 times less acutely
toxic than abamectin and is greater than 20 times less toxic in the most
sensitive subchronic test.
5. Conclusions
Abamectin does not affect nitrogen-fixing bacteria, even at levels higher
than those which would be present in the environment. At high concen-
trations abamectin can have a toxic effect on earthworms and birds.
However, these effects occur at concentrations which are higher than
those found in the environment. The toxicity of abamectin to bees is
13. Environmental Aspects of Use in Crop Protection 197
A. AQUATIC ORGANISMS
B. TERRESTRIAL ORGANISMS
The lowest acute toxicity value, obtained in the weanling rat, was an LD50
of 1.5 mg/kg. If one assumes that a wild mammal will consume 20% of its
body weight in food/day, it would have to ingest residue levels of
7.5 ppm, 44 times higher than the average found on celery, to reach the
LD50 level. Several other factors should be mentioned. The exposure
levels assume that all mammalian foodstuffs have the same residue level
as the target crop. In actuality, this is not the case. Animals would also
consume foodstuffs outside the direct application area, where residue
levels would be lower. Also, laboratory animals were dosed by gavage,
which has been shown to yield a twofold greater toxicity to abamectin
than in feed administration. Thus an additional twofold safety factor
exists for wild mammalian species.
The lowest subchronic toxicity value was obtained in the materno-
toxicity-teratogenicity study in the CF-l mouse. The NOEL was
0.05 mg/kg by gavage and 0.1 mg/kg in the diet. In this study mUltiple
doses of abamectin were given. Using the dietary NOEL, a mammal
which ingests 20% of its body weight in food/day would have to consume
greater than 0.50 ppm of abamectin in the diet for several days. The low
initial residue levels-which are 3 times lower than this amount-and the
rapid degradation eliminate the possiblility of continuous exposure: no
maternotoxic or teratogenic effects should occur.
Residues in fruit are much lower than those in celery, and, therefore, no
effects from fruit consumption should occur. Insects are a major food
source for certain mammals. Given the low residue levels on plants and
the rapid degradation of abamectin, insects will not have high residues of
abamectin. Insects will not bioconcentrate residues of abamectin. A study
by Bull (1986) demonstrated that the half-life of abamectin in insects is
less than a half a day. Insects should not contain abamectin residues at
levels sufficient to be toxic to those animals consuming them.
200 P.G. Wislocki, L.S. Grosso, and R.A. Dybas
V. Conclusions
Abamectin neither persists nor accumulates in the environment. It rapidly
photolyzes in water, on soil, and as a thin film on both inert and biological
surfaces. Soil metabolizes abamectin at a moderate rate. Its strong
binding to soil prevents it from entering the aquatic environment, reduces
its concentration in water, and limits its bioavailability. These character-
istics reduce the environmental exposure to abamectin to a level below
that which would cause effects on aquatic or terrestrial organisms.
Therefore, when label directions are followed, abamectin can be used in
an environmentally safe manner.
REFERENCES
Brooke DN, Dobbs AJ, Williams N (1986) Octanol water partition coefficients
(P): measurement, estimation and interpretation, particularly for chemicals with
P lOS. Ecotox. & Envir. Safety 11:251-260
Bull DL (1986) Toxicity and pharmacodynamics of avermectin in the tobacco
budworm, corn earworm and fall armyworm (Noctuidae Lepidoptera). J. Agric.
Food Chem. 34:74-78
Bull DL, Ivie GW, MacConnell JG, Gruber VF, Ku CC, Arison BH, Stevenson
JM, VandenHeuval WJA (1984) Fate of avermectin Bla in soil and plants.
J. Agric. Food Chem. 32:94-102
Hoerger FD, Kenaga EE (1972) Pesticide residues in plants. Correlation of
representative data as a basis for estimation of their magnitUde in the environ-
ment. In F. Coulston and F. Korte (eds) Environmental Quality, Vol 1,
Academic Press, New York, pp 9-28
Iwata I, MacConnell JG, Flor JE, Putter I, Dinhoff TM (1985) Residues of
avermectin B 1a on and in citrus fruits and foliage. J. Agric. Food Chem.
33:467-471
Kenaga EE (1973) Factors to be considered in the evaluation of the toxicity of
pesticides to birds in their environment. In F. Coulston and F. Korte (eds)
Environmental Quality and Safety, Vol 2, Academic Press, New York,
pp 161-181
May WE, Wasik SP (1978) Determination of the aqueous solubility of the
polynuclear aromatic hydrocarbons by a coupled liquid chromatography tech-
nique. Anal. Chem. 50:175-182
Maynard MS, Iwata Y, Wislocki PG, Ku CC, Jacob TA, (1989) The fate of
avermectin B 1a on citrus fruits: 1. Distribution and magnitude ofthe 14C residue
and avermectin B 1a on citrus fruits from a field study. J. Agric. Food Chem.
37:178-183
Moye HA, Malagodi MH, Yoh J, Leibee GL, Ku CC, Wislocki PG (1987)
Residues of avermectin B 1a in rotational crops and soils following soil treatment
with [14C] avermectin B 1a. J. Agric. Food Chem. 35:859-864
Mrozik H, Eskola P, Reynolds GF, Arison BH, Smith GM, Fisher MH (1988)
Photoisomers of avermectins. J. Org. Chem. 53:1820-1823
CHAPTER 14
I. Introduction
II. Worker Exposure to Abamectin During Airblast Treatment of Citrus
Groves
III. Worker Exposure to Abamectin during Reentry
I. Introduction
Abamectin is the avermectin selected for agricultural use. Its various
formulations, and the uses that have been developed, are described in
Chapter 20.
Although abamectin is applied to ornamental plants, crops, and trees at
extremely low rates, its high oral mammalian toxicity (Chapter 6) initially
raised concern regarding the safety of workers mixing, loading, and
applying the product, as well as those harvesting treated crops. An
established quantitative exposure model developed for registered pesti-
cides indicated a significant correlation between dermal exposure and
application rate (Reinert and Severn 1985). The exposure model was
based on data generated from exposure monitoring of workers in orchards
during ground application of pesticides using high-pressure or airblast
equipment. A linear regression analysis of the data failed to intercept the
origin, thus predicting substantial exposure to such products used at low
rates of application. The reported values are normalized to 3000 cm2 of
exposed skin, the amount for an applicator wearing long pants, a
short-sleeved open-necked shirt, and no gloves or facial protection. The
authors argue that the correlation (r=0.7) permits an estimate of exposure
during airblast application of any pesticide with a certain level of
confidence. It should be pointed out that the model does not consider the
bioavailability of individual products (such as dermal penetration), and
thereby again overestimates exposure.
The studies on which the exposure model was based were conducted
with pesticide products used at conventional rates of application (0.25 to
202 L.S. Grosso, R.A. Dybas, and S.F. Rickard
a As an estimator of the variance of the data, the geometric mean is multiplied and divided
by the geometric standard deviation (GSD).
clearly show that the existing model available for applicators grossly
overestimates exposure. Based on the available safety data on the
avermectins in the most sensitive model (CF-l mouse), in addition to
human data and exposure data, we believe that there are adequate
margins of safety for workers who mix/load or spray abamectin using
airblast equipment with or without the use of conventional clothing.
tion curve for both residue species is similar (Figure 14.1). The "prior
to last application" samples for all 4 treated plots had average aver-
mectin Bla/ a8,9-isomer residues of 1.2 ng/cm2 and 0.14 ng/cm2 avermec-
tin BIb. The residues at 2 hours following the last application aver-
aged 19.2 ng/cm 2 for the combined residues of Bla/ a8,9 isomer and
2.9 ng/cm2 avermectin BIb. Abamectin is unstable in the presence of
sunlight, but these residues did not decay through Hour 16, as the treated
chrysanthemums were protected from light in the late afternoon by
drawing close the greenhouse thermal shades. Between Hours 16 and 30,
after the thermal shades were opened, the residues decayed, with average
residues at 30 hours of 10.1 ng/cm2 avermectin B la and 1.5 ng/cm2
avermectin BIb. At 7 days after the last application, residues averaged 1.9
ng/cm2 avermectin B la and 0.23 ng/cm2 avermectin BIb, essentially
identical to the residues found at 7 days following the second of 3
applications. No buildup or accumulation of residues was observed
following multiple applications in greenhouse conditions. No difference
was found in the recoveries from leaf disc samples spiked in the field and
those spiked in the laboratory. Application of the Zweig-Popendorf
Factor to the DFR data from this study results in the following estimate of
worker exposure to abamectin residues. The average for total DFR
residues of abamectin and its a8,9-isomer was 22 ng/cm2 at 16 hours after
the third of 3 weekly sprays at the recommended rate of 27 g abamectin/
hectare, immediately prior to the reentry phase.
25
23
•••
21
18
•
17
II)
:;jll
....'tI
III
II)
II: 8
2 3 4 5 6 7
TIllE (Days)
NOEL = 50 ILg/kg/day
Exposure = 14.7 ILg/kg/day
MOS = NOEL/exposure x 100 (dpf)
= 50 x 100 = 340
14.7
MOS = 340 for maternotoxicity
It was believed that this worker exposure of 14.7 ILg/kg/day from foliar
residues of abamectin estimated via the Zweig-Popendorf Factor was an
overestimate of exposure, resulting in a very conservative MOS.
To confirm the accuracy ofthe Zweig-PopendorfFactor with abamectin
under the same greenhouse conditions, an actual worker reentry study
was conducted in the Netherlands. This study measures the dermal
exposure of workers to foliar residues of abamectin during harvest of
propagation cuttings from greenhouse-grown chrysanthemums that had
been treated with abamectin (0.15 EC) at its maximum recommended use
rate. Residues from coveralls, hats, gloves, and hand rinses of harvesters
were used to estimate exposure, which was then used in a risk assessment
to determine margins of safety for workers involved in the culture and
production of greenhouse ornamentals.
The study was conducted in the same commercial chrysanthemum
propagation greenhouse at DeLier, Netherlands as that used for the DFR
study. Each of 4 workers was dressed in a disposable Tyvek spun-olefin
coverall and baseball hat with prepared dosimeters attached to the front
and the back of the hat, the chest, back, upper and lower arms, and upper
and lower legs. The workers wore cotton gloves. Each worker was
assigned to a separate treated plot for harvesting. At the end of each of
four I-hour work periods, the dosimeters and gloves were removed into
separate numbered bags. Mter the gloves were removed, each worker's
hands were rinsed 3 times. Control and fortified (spiked) samples from the
field, along with the experimental samples, were immediately frozen
following their collection for subsequent analysis using an HPLC-
fluorescence method which determines abamectin (avermectin B la and
avermectin Bib) and the ~8,9-isomer.
Abamectin residues were found only on the gloves, lower-leg do-
simeters, and in the hand-rinse solutions; in 1 case low-level residues
were found on the chest dosimeter. The data are presented in Table 14.5.
Average exposure was 70,863 ng/hr with a coefficient of variation (CV)
of 5.4%. Approximately 97% of the total exposure was on the hands, 3%
on the clothing, and less than 1% on the face and clothed body. Because
50% of the limit of quantitation was used when no residues were
measured on a given dosimeter, calculated exposures of the face and neck
(except in 1 case) and the clothed body were calculational artifacts that
were insignificant in estimating exposure. Body protection by long pants
212 L.S. Grosso, R.A. Dybas, and S.P. Rickard
REFERENCES
I. Introduction
II. Use in Cattle
A. Injectable Formulation
B. Oral Formulations
C. Topical Formulation
D. Sustained-Release Bolus
III. Use In Sheep
A. Oral Formulation
B. Injectable Formulation
IV. Use In Goats
A. Oral Formulation
B. Injectable Formulation
V. Use In Swine
A. Injectable Formulation
References
I. Introduction
This chapter presents a summary of the efficacy data that form the basis
for the clinical use of ivermectin in cattle, sheep, goats, and swine. At
least 2 adequate and well-controlled trials investigated each ivermectin
formulation's effect on each parasite species. Most of the trials were
conducted by MSDRL investigators, though investigators at other labora-
tories also contributed. The efficacy percentages presented here are those
upon which the regulatory approvals were based. Animal health applica-
tions of ivermectin have been reviewed (Campbell et al. 1983; Campbell
and Benz 1984; Benz 1985; Campbell 1985; Bennett 1986; Barragry 1987).
Clinical usages have also been summarized concerning cattle (Leaning,
Roncalli, and Brokken 1983), sheep (Hotson 1983), and swine (Brokken et
al. 1983). Recent reports that expand the basis of the label claims are
included.
216 G.W. Benz, R.A. Roncalli, and S.J. Gross
I. Use in Cattle
A. INJECTABLE FORMULATION
Endoparasite Activity
The recommended dose of 200 /Lg ivermectin/kg was selected upon
completion of numerous dose-titration trials. Adult Cooperia oncophora,
C. punctata, Trichostrongylus colubriformis, and Nematodirus helvetia-
nus each required a dose between 100 /Lg/kg and 200 /Lg/kg; fourth-
stage larvae of Haemonchus placei, C. oncophora, N. helvetianus, and
T. colubriformis required a dose of 200 /Lg/kg.
Trial data used for registration of label efficacy claims are summarized
in Table IS.I. In work performed subsequent to the original registrations,
hemorrhagic dermal lesions caused by Parafilaria bovicola were observed
to require at least 70 days for regression (Swan et al. 1983). Efficacy of
more than 99% was found for Thelazia spp. adults (Carmichael, SolI, and
Scherer 1985).
Infections with Ostertagia and Cooperia acquired up to at least 7 days
after an ivermectin injection, as well as with Dictyocaulus viviparus
acquired up to at least 14 days later, are controlled (Barth 1983; Armour et
al. 1985), presumably because the drug persists following injection. Based
on this persistent effect, control programs utilizing ivermectin injections
at 3, 8, and 13 weeks after turnout onto spring pasture have been effective
in controlling gastrointestinal parasites as well as lungworms (Ryan et al.
1986; Taylor, Mallon, and Kenny 1985; Jacobs, Fox, and Ryan 1987). In
such programs, cattle on pastures contaminated with D. viviparus infec-
tive larvae have been found to be resistant to lungworm disease in the
next grazing season, probably because infective larvae ingested between
treatments induced immunity; gastrointestinal nematodes in the second
season were not apparent but inhibited Ostertagia larvae did accumulate
(Armour et al. 1988).
Ectoparasite Activity
The dose rate confirmed as optimal against nematode parasites was also
found to be highly effective against several economically important
ectoparasites.
15. Use of Ivermectin in Cattle, Sheep, Goats, and Swine 217
Grubs
The common cattle grubs in the Northern Hemisphere, Hypoderma bovis
and H. lineatum, are highly susceptible to ivermectin. All 3 larval stages
are killed following treatment. Efficacy in confirmation trials was 100% in
most instances, although a few third instars (larvae), which were near to
emergence from their host at the time of treatment, were expelled even
though they had been grossly affected and were probably dead. Killing
Hypoderma first instars during their normal migratory route may cause
adverse host-parasite reactions-such as eosinophilic esophagitis leading
to bloat (H. linea tum) or hemorrhage, (H. bovis) which affects the spinal
cord and most often leads to hindquarter paralysis. These reactions can
be avoided by not treating cattle with ivermectin during this migratory
season (as determined by local epidemiologic patterns).
The tropical cattle grub affecting cattle in Latin America, Dermatobia
hominis, is also controlled effectively (Roncalli 1984b).
218 G.W. Benz, R.A. Roncalli, and S.J. Gross
Lice
Lice that ingest blood or body fluids from their host (i.e., sucking lice),
including Haematopinus eurysternus, Linognathus vituli, and Soleno-
potes capillatus, are highly susceptible to therapeutic treatment. Iver-
mectin given subcutaneously at 200 ILg/kg kills adult sucking lice as they
feed upon blood or its constituents; the drug persists sufficiently long so
that newly emerged larvae as well as nymphs are also killed as they begin
to feed. In the registration trials, viable lice were recovered from cattle
1 week after treatment but none thereafter (up to 8 weeks). Lice surviving
on ivermectin-treated cattle during the first week after treatment should
be considered infective to nonmedicated cattle. To avoid reinfestation,
ivermectin-medicated cattle should not be mingled with those untreated
during the balance of the winter season, when these parasites are still
active on their host.
Biting lice (Damalinia bovis) are not eliminated consistently from
cattle given a therapeutic injection of ivermectin. This may be related
to their superficial feeding pattern. A claim stated as "aid in the
control" of D. bovis is registered in Europe and some Southern
Hemisphere countries. In the United States, treatment of infested
cattle was ineffectual; whether there are susceptibility differences
among D. bovis populations from various geographic localities
has not been determined.
Mites
Among mange mites, Sarcoptes scabiei var. bovis is the most sensitive to
ivermectin. This species burrows more deeply into the dermis than do
other species and evidently consumes blood or body fluids directly. In
registration trials, live sarcoptic mites from medicated animals were
recovered at 1 week following injection with ivermectin, but not there-
after (up to 8 weeks).
Psoroptes ovis does not burrow into the skin as deeply as Sarcoptes.
Nonetheless, no live psoroptic mites were recovered during confirmation
trials after 2 weeks following treatment although, in subsequent trials, a
few P. ovis have been found on cattle treated 20 days previously (Wright
and Guillot 1984). An isolation period of at least 14 days following
treatment is adequate for use in control programs (Strickland and Gerrish
1987). Very few live mites can be recovered subsequent to treatment; the
persistence of ivermectin following injection presumably accounts for the
few recovered mites not being able to reestablish viable populations
(Guillot, Wright, and Oehler 1986).
Chorioptes bovis, living on the superficial dermal surface, is the least
responsive of the cattle mites studied; only an "aid in control" claim is
registered in certain countries.
15. Use of Ivermectin in Cattle, Sheep, Goats, and Swine 219
Ticks
Ticks also are affected by ivermectin. Boophilus microplus and B.
decoloratus, which are single-host species, are more sensitive than
multihost ticks. Ingestion of blood by adult females is interfered with;
eventually they falloff, usually only partially engorged, and few eggs are
produced. The efficacy of therapeutic doses against Boophilus spp. is
sufficient for the registration of "aid in control" claims in several
countries. Boophilus strains resistant to organophosphates, DDT, synthe-
tic pyrethroids, and amidine were sensitive to ivermectin (Roncalli et al.
1984).
Effects against most multihost ticks are too transient for meaningful
efficacy claims; however, a claim has been registered for the sand tampan
(Ornithodoros savignyi) present in South Mrica (SolI et al. 1984).
Screwworms
Among fly larvae (screwworms) that develop in the wounds of cattle, a
therapeutic injection of ivermectin effectively controls Old World screw-
worm fly (Chrysomya bezziana) larvae for 14 days following treatment.
Established infestations are less effectively controlled and may require
additional measures. Screwworms of Cochliomyia hominivorax are not
controlled by such ivermectin treatment.
ORAL FORMULATIONS
IVOMEC oral solution for cattle contains 0.4% w/v ivermectin and
should be administered at 2.5 m1l50 kg body weight using standard dosing
equipment. IVOMEC cattle paste contains 0.153% w/v ivermectin and is
administered orally by depressing the handle of the Medigun applicator
completely to deliver 23 mg ivermectin per 113.5 kg (250 lb) body weight.
These formulations are indicated for the treatment and control of gastro-
intestinal nematodes, lungworms, grubs, and sucking lice.
Endoparasite Activity
Efficacy data used for registration of nematode efficacy claims for both
oral formulations are listed in Table 15.2.
Ectoparasite Activity
Complete efficacy is lacking against mange mites following oral adminis-
tration of ivermectin to cattle, presumably because less ivermectin is
absorbed systemically and it persists for a shorter duration compared to
the subcutaneous route.
The common cattle grubs in the Northern Hemisphere, Hypoderma
220 G.W. Benz, R.A. Roncalli, and S.l. Gross
TOPICAL FORMULATION
IVOMEC Pour On for cattle contains 0.5% w/v ivermectin and is applied
along the topline in a narrow strip extending from the withers to the
tailhead. The volume of application is 1 mlllO kg body weight. This
formulation is for the treatment and control of gastrointestinal nematodes,
lungworms, grubs, sucking and biting lice, chorioptic and sarcoptic
mange mites, and as an "aid in control" of ticks (Boophilus microplus).
Endoparasite Activity
The recommended dose rate of 500 j.tg/kg was selected following a series
of dose-titration trials. Dose-confirmation data against gastrointestinal
nematodes and lungworms (Hotson et al. 1985; Alva-Valdes et al. 1986;
Yazwinski et al. 1986) are summarized in Table 15.3. The formulation has
15. Use of Ivermectin in Cattle, Sheep, Goats, and Swine 221
Ectoparasite Activity
Ivermectin applied topically to cattle effectively controls several ecto-
parasites (Barth et al. 1986; Alva-Valdes et al. 1986; Hotson et al. 1985).
Infestations of Chorioptes bovis as well as Sarcoptes scabiei var. bovis
are controlled. However, the formulation does not completely eliminate
Psoroptes ovis and, in such cases, the use of IVOMEC Injection should be
considered. Sucking lice (Haematopinus eurysternus, Linognathus vituli,
and Solenopotes capillatus) as well as biting lice (Damalinia bovis) are
effectively controlled. Hom flies (Haematobia irritans) are controlled for
up to 35 days following product application. Northern Hemisphere grubs
(larvae of Hypoderma bovis and H. linea tum) and tropical grubs (Der-
matobia hominis) are controlled. The topical formulation "aids in the
control" of Boophilus microplus (Cramer et al. 1985).
SUSTAINED-RELEASE BOLUS
Use in Sheep
ORAL FORMULATION
Endoparasite Activity
The recommended dose rate of 200 JLg/kg for sheep was selected
following a series of dose-titration trials. Dose-confirmation data against
gastrointestinal nematodes and lungworms, used for registration, are
summarized in Table 15.4. Strains of Haemonchus contortus, Ostertagia
circumcincta, and Trichostrongylus colubriformis tolerant to benzim-
idazoles, levamisole, and morantel (and an H. contortus strain tolerant to
rafoxanide) were fully susceptible to ivermectin. Haemonchus resistance
to ivermectin has been reported following repeated treatments in labora-
tory (Egerton, Suhayda, and Eary 1988) and, at short intervals, field
situations (Carmichael et al. 1987; van Wyk and Malan 1988; van Wyk et
al. 1987; Echevarria and Trindade, 1989). Trichostrongylus colubriformis
resistance under laboratory conditions also has been reported (Giordano,
Tritschler, and Coles 1987).
Ectoparasite Activity
Efficacy also was demonstrated against all three instars (larvae) of nasal
bots, Oestrus ovis (Roncalli 1984a), and against itch mites, Psorergates
15. Use of Ivermectin in Cattle, Sheep, Goats, and Swine 223
ovis (Hotson 1983). Ivermectin delivered orally to sheep does not control
other mange infestations.
INJECTABLE FORMULATION
IVOMEC Injection, the same 1.0% w/v solution used in cattle, is also
employed for sheep. The product is administered subcutaneously at
0.5 ml/25 kg body weight, using care to ensure that the skin has been
penetrated before attempting delivery of the dose. It is indicated for the
treatment and control of gastrointestinal nematodes, lungworms, nasal
bots, mange mites, and itch mites.
Endoparasite Activity
The dose of 200 p,g ivermectin/kg utilized orally in sheep was selected for
this formulation based on other usages. Registration trials subsequently
confirmed high efficacy against gastrointestinal nematodes and lung-
worms (Table 15.5). Efficacy against Protostrongylus rufescens has also
been demonstrated (Dakkak et al. 1986).
224 G.W. Benz, R.A. Roncalli, and S.J. Gross
Ectoparasite Activity
Efficacy was confirmed in registration trials against the parasitic larval
stages of the nasal bot, Oestrus ovis.
Infestations caused by mange mites, Sarcoptes scabiei, and itch mites,
Psorergates ovis, also are controlled by ivermectin given subcutaneously.
In contrast to the oral formulation, which does not control Psoroptes ovis,
sheep given the injectable formulation do experience marked reductions
in Psoroptes numbers and clinical signs. A second injection 7 days later
may be required to eliminate all the living mites (Roncalli and Sutherland
1986). These reductions in psoroptic mites on sheep are presumably
related to the longer persistence of ivermectin following subcutaneous
administration, compared to oral administration, as well as to the feeding
patterns of Psoroptes on sheep (Kirkwood 1985).
Use in Goats
ORAL FORMULATION
IVOMEC or ORAMEC Liquid for goats is the same 0.08% w/v solution of
ivermectin as used in sheep, and is to be given at 2.5 mlll0 kg body
15. Use of Ivermectin in Cattle, Sheep, Goats, and Swine 225
INJECTABLE FORMULATION
Use in Swine
INJECTABLE FORMULATION
Endoparasite Activity
Efficacy data used for the registration of nematode efficacy claims are
summarized in Table 15.7. Kidney worms, Stephanurus dentatus, are
controlled (Plue et al. 1986). Further, ivermectin given to sows 7 to 14
days before farrowing effectively controls transmission of Strongyloides
ransomi larvae to newborn pigs.
Ectoparasite Activity
Infestations of lice (Haematopinus suis) and mange mites (Sarcoptes
scabiei var. suis) were effectively controlled during registration trials. In
mange control programs, all animals in a herd must be treated and a
consistent program of retreatment adhered to (Hogg 1984). Sows prefera-
bly should be treated prior to breeding and 7 to 14 days prior to farrowing
(Courtney, Ingalls, and Stitzlein 1983). Boars should be treated at least
twice annually, although frequency of and need for treatments depend on
exposure. All feeder pigs received for finishing should be treated before
placement in clean quarters; pigs exposed to contaminated soil, premises,
or other pigs may need retreatment if reinfestation occurs.
IVOMEC Injection in swine has a persistent drug level sufficient to
control sarcoptic mite infestations throughout the life cycle, from egg to
adult. Because the ivermectin effect is not immediate, however, care
must be taken to prevent reinfestation from exposure to untreated animals
or contaminated facilities. Generally, pigs should not be moved to clean
REFERENCES
Alva-Valdes R, Wallace DH, Holste JE, Egerton JR, Cox JL, Wooden JW,
Barrick RA (1986) Efficacy of ivermectin in a topical formulation against
induced gastrointestinal and pulmonary nematode infections, and naturally
acquired grubs and lice in cattle. Am. J. Vet. Res. 47:2389-2392.
Armour J, Bairden K, Batty AF, Davison CC, Ross D (1985) Persistent
anthelmintic activity of ivermectin in cattle. Vet. Rec. 116:151-153.
Armour J, Bairden K, Ryan WG (1988) Immunity of ivermectin treated cattle to
challenge from helminth parasites in the following season. Vet. Rec. 122:223-
225.
Baggott DG, Batty AF, Ross DB (1986) The control of mature nematode
infections in cattle by sustained delivery ofivermectin. Proceedings of the 14th
World Congress on Diseases of Cattle, Vol. 1, Dublin, Ireland, pp 160-165.
Barragry TB (1987) A review ofthe pharmacology and clinical uses ofivermectin.
Can. Vet. J. 28:512-517.
Barth D (1983) Persistent anthelmintic effect of ivermectin in cattle. Vet. Rec.
113:300.
Barth D, Batty AF, Robin B, Preston JM (1986) Efficacy of a topical formulation
of ivermectin against cattle ectoparasites. Proceedings 14th World Congress of
Diseases of Cattle, Vol 1, Dublin, Ireland, pp 157-159.
Barth D, Preston JM (1985) Efficacy against somatic Strongyloides ransomi
larvae. Vet. Rec. 116:366-367.
Bennett DG (1986) Clinical pharmacology ofivermectin. J. Am. Vet. Med. Assoc.
189:100-104.
Benz GW (1985) Animal health applications of ivermectin. Southwest. Entomol.
(Suppl.) 7:43-50.
Brokken ES, Barth D, Foster AG, Pulliam JD, Wallace DH (1983) Ivermectin, a
new broad-spectrum antiparasitic agent for swine. In Proceedings of the MSD
AGVET Symposium "Recent Developments in the Control of Animal Para-
sites," in association with the XXII World Veterinary Congress, Perth,
Australia, pp 239-258.
Campbell WC (1985) Ivermectin: An update. Parasitol. Today 1:10-16.
Campbell WC, Benz GW (1984) Ivermectin: A review of efficacy and safety. J.
Vet. Pharm. Therap. 7:1-16.
Campbell WC, Fisher MH, Stapley EO, Albers-Schonberg G, Jacobs TA (1983)
Ivermectin: A potent new antiparasitic agent. Science 221:823-828.
Carmichael IH, Soll MD, Scherer H (1985) The use ofivermectin in the control of
bovine thelaziosis. 11th Conference World Association for the Advancement of
Veterinary Parasitology, Rio de Janeiro, Abstract 106.
Carmichael IH, Visser R, Schneider D, Soll MD (1987) Haemonchus contortus
resistance to ivermectin (Letter to Editor). J. S. Afr. Vet. Assoc. 58:93.
Courtney CH, Ingalls WL, Stitzlein SL (1983) Ivermectin for the control of swine
228 G.W. Benz, R.A. Roncalli, and S.J. Gross
I. Introduction
II. Efficacy
A. Endoparasite Activity
B. Ectoparasite Activity
I. Introduction
This chapter presents a summary of the efficacy data that form the basis
for the clinical use of abamectin (avermectin B t ) in cattle. The product is
formulated in the same vehicle as used for IVOMEC (ivermectin)
Injection. At the present time, AVOMEC (abamectin) Injection is regis-
tered only in Australia.
At least 2 adequate and well-controlled trials investigated the effect on
each parasite species. Most of the trials were conducted by MSDRL
researchers, though investigators at other laboratories also contributed.
The efficacy percentages presented here are those upon which regulatory
approval was based. Other efficacy data obtained with abamectin but
formulated in a different vehicle, and not relied upon for registration,
have been presented (Benz and Ernst 1979; Egerton et al. 1979; Wescott
et al. 1980).
II. Efficacy
AVOMEC Injection is a sterile solution containing 1.0% w/v abamectin.
The product is indicated for the treatment and control of gastrointestinal
nematodes (including inhibited immature Ostertagia ostertagl), lung-
worms (Dictyocaulus v;v;parus), and sucking lice (Linognathus vitum
(Scott et al. 1985). It is to be administered subcutaneously in front of or
behind the shoulder at 1 mil 50 kg body weight.
16. Use of Abamectin in Cattle 231
A. ENDOPARASITE ACTIVITY
B. ECTOPARASITE ACTIVITY
engorged female ticks dropped from treated cattle for at least 21 days, and
the reproductive potential among surviving female ticks is reduced by
interference with egg production.
REFERENCES
Armour J, Bairden K, Batty AF, Davison CC, Ross D (1985) Persistent anthel-
mintic activity ofivermectin in cattle. Vet. Rec. 116:151-153.
Barth D (1983) Persistent anthelmintic effect of ivermectin in cattle. Vet. Rec.
113:300.
Benz GW and Ernst JV (1979) Anthelmintic activities of the B.a fraction of
avermectin against gastrointestinal nematodes in calves. Am. J. Vet. Res.
40: 1187-1188.
Bremner KC, Berrie DA, Hotson IK (1983) Persistence of the anthelmintic
activity of ivermectin in calves. Vet. Rec. 113:569.
De Chaneet GC, Casey R, Dixon FF, Besier RB, Mitchell RK (1988) Effect of
avermectin B. and benzimidazole anthelmintics on worm egg output of treated
cattle. Austral. Vet. J. 65:85-86.
Eagleson JS, Bowie JY (1986) Oxfendazole resistance in Trichostrongylus axei in
cattle in Australia. Vet. Rec. 119:604.
Egerton JR, Ostlind DA, Blair LS, Eary CH, Suhayda D, Cifelli S, Riek RF,
Campbell WC (1979) Avermectins, a new family of potent anthelmintic agents:
efficacy of the B.a component. Antimic. Agents Chemother. 15:372-378.
Scott PG, Burrows RO, Hotson IK, Cox JL (1985) Avermectin B. as an
anti-parasitic agent for cattle. 11th Conference World Association for Ad-
vancement of Veterinary Parasitology, Rio de Janeiro, Abstract 83.
Tahir MS, Holroyd RG, Copeman DB (1986) Treatment of beef calves with
ivermectin and avermectin B. in dry tropical Australia. 6th International
Congress of Parasitology, Brisbane, Australia, Abstract 651.
Wescott RB, Farrell CJ, Gallina AM, Foreyt WJ (1980) Efficacy of avermectin
B.a for treatment of experimentally induced nematode infections in cattle. Am.
J. Vet. Res. 41:1326-1328.
CHAPTER 17
I. Products
A. Eqvalan paste™, Zimecterin paste™
B. Eqvalan liquid ™
c. Eqvalan injectable ™
II. Antiparasitic efficacy
A. General
B. Parascaris equorum
C. Strongylus vulgaris, S. edentatus, S. equinus
D. Small strongyles
E. Dictyocaulus arnfieldi (Lungworm)
F. Draschia sp. and Habronema sp.
G. Onchocerca cervicalis
H. Other helminths
I. Gastrophilus spp. (bots)
J. Sarcoptes scabei
I. Products
A. EQvALAN PASTE TM; ZIMECTERIN PASTE ™
This is a paste, for oral use, containing 1.87% w/w ivermectin. In the
United States, it is available in a prefilled syringe, calibrated so that each
increment represents the amount of drug needed for 250 lb (114 kg) of
body weight at the rate of 200 ltg/kg. Each syringe contains enough paste
to treat one mature horse. The syringe sold in international markets is
calibrated to deliver the amount of drug needed for 100 kg (220 lb) of body
weight at 200 ltg/kg.
B. PARASCARIS EQUORUM
(Corba et al. 1986), illustrating that the oral paste, like the injectable
solution, is active against adult P. equorum. An effect on the passage of
eggs was also observed by Puccini and colleagues (1984). In another
study, fecal ascarid egg counts in 14 naturally infected horses were
negative following treatment with ivermectin oral paste and remained at
zero for 10 to 12 weeks following treatment, while egg counts in the 6
control horses were substantially unchanged (E.T. Lyons, unpublished
data). Efficacy against adult ascarids was demonstrated more directly and
definitively in controlled studies in which ascarid burdens were deter-
mined at necropsy (French et al. 1988b; DiPietro et al. 1987). In both
studies ivermectin paste at 200 jLg/kg was 100% effective against the adult
worms.
Ivermectin is also active against the immature stages of P. equorum. In
two studies in naturally infected ponies, larval burdens were determined
at necropsy following treatment with the paste at 200 jLg/kg. When
necropsy was performed 2 weeks after treatment, the reduction in lung
larval burden was 100% (French et al. 1988b) and the reduction in
intestinal larval burden was also 100% (French et al. 1988b; DiPietro et al.
1987). When necropsy was delayed until 5 weeks after treatment, larvae
were recovered from the intestines; and, since treatment is highly active
against the early migrating stage, this probably reflected reinfection,
17. Use of Ivermectin in Horses 237
despite effort taken in one of the trials to prevent it. In one of the trials,
treatment was not associated with clear-cut improvement in clinical
condition but was associated with improved weight gain (French et al.
1988b).
To define the efficacy ofivermectin against immature P. equorum more
precisely, 2 trials were conducted on experimentally induced infections.
Groups of 6 horses were treated with ivermectin paste or liquid at
200 JLg/kg, or were maintained as untreated or vehicle-untreated controls
(DiPietro et al., in press; French et al., in press). Treatment was
administered on Day 11 of infection, when the majority of the parasites
would be in the migratory L3 larval stage. (Necropsy of indicator horses
at that time revealed larvae in the lungs but not in the small intestine).
At necropsy, 14 days after treatment, the efficacy of ivermectin against
P. equorum was determined to be 100% in both trials and with both
formulations (P < 0.001). It is not possible to conclude from the data
whether all of the larvae were killed in the lungs or on early entry into the
small intestine.
To determine efficacy against the later (L4) stage of P. equorum, 4 trials
were conducted with experimentally induced infections (A. Batty, unpub-
lished data; J.L. Duncan, unpublished data; T.R. Bello 1985; and
DiPietro et al., in press). Treatment was given on Day 28 or 29 of
infection. At necropsy on Day 42 or 43 of infection (14 days after
treatment), the immature ascarids in the small intestine were counted,
revealing reductions of 62%,95%, greater than 99%, and greater than 99%
in the paste-treated groups in the 4 trials, respectively. In 2 of the trials,
ivermectin liquid was also tested, and reductions of greater than 99%
were recorded in both (DiPietro et at., in press; J.L. Duncan, unpub-
lished data). Using an appropriate statistical method (Friedman's test) to
allow for heterogeneity of variances among treatments, the overall
reduction was calculated as 98.4% (P > 0.001). In the trial by DiPietro and
colleagues (in press), gross examination oflung and liver revealed similar
lesions in the treated and untreated foals, but the ascarids recovered from
the treated animals were significantly smaller than those from the
controls. Thus treatment evidently affected the parasites adversely,
perhaps by interfering with feeding, but did not kill them.
Thus the paste and liquid ivermectin formulations have been shown to
be highly active against both the early (L3) tissue phase and against the
later intestinal (L4) phase. The significance and differences of opinion
associated with this efficacy have been discussed by Boraski (1987).
The oral paste formulation, used at 200 JLg/kg, was 100% active against
adult S. vulgaris, as was the injectable formulation which was tested in
the same experiment (Klei et al. 1984).
The migratory fourth stage larvae of S. vulgaris are of special im-
238 W.C. Campbell, W.H.D. Leaning, and R.L. Seward
portance because (1) they cause lesions of certain arteries (especially the
mesenteric), often giving rise to serious clinical signs; and (2) they are
particularly hard to eliminate by chemotherapeutic means. Extensive
studies by Slocombe and his colleagues (Slocombe et al. 1982; Slocombe
and McGraw 1984; Slocombe et al. 1984) showed that the injectable
formulation of ivermectin is highly active against the migratory fourth
stage larvae. He further showed that efficacy against late fourth stage
larvae may not be demonstrable if necropsy is performed only 2 weeks
after treatment; and he cautioned that treatment appeared to cause some
larvae to become situated more deeply in the wall of the ileocolic artery,
prompting a concern about the eventual fate of such larvae and the impact
of their putative slow disintegration within the arterial wall-especially
after repeated cycles of infection and treatment. Studies by Klei and his
colleagues (Klei et al. 1984a; Klei et al. 1984b) confirmed that the interval
between treatment and necropsy is critical for demonstrating the efficacy
of ivermectin against the migratory larvae of S. vulgaris. In their
experience, the oral paste (and the injectable formulation) at 200 #Lg/kg
was 99% effective against 8-week-old larvae, as revealed by necropsy 5
weeks after treatment. Klei also commented on the potential impact of
dead or dying larvae on the arterial walls. He noted,
Killing S. vulgaris larvae with ivermectin not only promoted a remarkable
resolution of arterial lesions, but also prevented their progression into the lesions
observed in the nontreated controls. Although ivermectin clearly kills intravascu-
lar S. vulgaris larvae, nematode bodies persist for some time following treatment,
as indicated by the numbers of dead larvae recovered. It is interesting to note that
the presence of the inactive or paralyzed worms alone does not provoke the
arterial lesions associated with S. vulgaris migrations. Observations of lesions in
livers and lungs in control animals confirm and extend previous observations that
S. vulgaris migrations are not restricted to the predilection sites in the mesenteric
vasculature. The severity and occurrence of lesions in extravascular sites were
markedly reduced in the ivermectin-treated foals, indicating that resolution of
these lesions also occurs within the 5-week period following treatment.
Repeated cycles of infection and ivermectin therapy have not resulted in
clinical disease or unique lesions (T.R. Klei, unpublished data). It is not
known whether ivermectin is active against the earliest parasitic (L3)
stage of S. vulgaris, but there is evidence that the drug (at least when
given intramuscularly) is effective prophylactically. A foal treated 2
weeks before inoculation with larvae and necropsied 4 weeks after
inoculation had no evidence of arterial infection, whereas the arteries of
control foals showed clear signs of the infection (Slocombe et al. 1984).
Apparently the ivermectin persisted in sufficient concentration to affect
the third or fourth stage larvae at some time during the 4-week period
following inoculation with infective larvae.
Ivermectin was tested at O.2#Lg/kg against adult S. edentatus in horses.
In 5 trials, side-by-side comparison of the paste and injectable formula-
17. Use of Ivermectin in Horses 239
tions gave reductions in worm burden of greater than 99% and 100%,
respectively (T.R. Klei, D.H. Wallace, T.A. Yazwinski, and J.L. Dun-
can, unpublished data). Against the L4 migratory stage of this species, an
efficacy of 100% was recorded for both formulations (J.L. Duncan,
unpublished data).
In 4 trials, the paste and injectable formulations were 100% effective in
removing adult S. equinus at a dosage of 0.2 ILg/kg (T.R. Klei, T.A.
Yazwinski, and D.H. Wallace, unpublished data).
D. SMALL STRONGYLES
G. ONCHOCERCA CERVICALIS
Ivermectin is not known to be active against the adult stage of any species
of Onchocerca in any host species, and its lack of efficacy in the case
of Onchocerca spp. in the horse has been documented (Lyons, Drudge,
and Tolliver 1988). However, its activity against the skin-dwelling micro-
filariae of the genus Onchocerca has been known since Egerton and
colleagues (1981) demonstrated that a single subcutaneous injection at
200 JLg/kg was highly effective against the microfilariae of o. cervicalis in
horses. Subsequent studies have shown that intramuscular administration
of the same dosage is highly effective; and Herd and Donham (1983) have
reviewed the parasitological and clinical significance of the finding.
The efficacy of the paste formulation against the microfilariae of O.
cervicalis has also been reported (French et al. 1988a; Foil et al., 1985;
and Pollitt et al. 1986). In these studies, treatment resulted in both the
disappearance of microfilariae from the skin and the resolution of the
associated lesions. In the experience of Pollitt and colleagues (1986), "a
single oral dose of ivermectin caused all microfilariae of Onchocerca spp.
to disappear from the skin and all the horses showed marked clinical
improvement of their dermatitis 1 to 2 weeks after treatment. By 4 to 6
weeks it was impossible to determine the original sites of the lesions."
These authors went on to point out that ivermectin did not reduce lesions
attributed to hypersensitivity to midge bites, and therefore it could be
used in horses to differentiate between that form of dermatitis and
onchocercal dermatitis. The presence of o. cervicalis microfilariae in the
skin is by no means always associated with dermatitis, but where
dermatitis occurs, it can be treated successfully with ivermectin.
17. Use of Ivermectin in Horses 241
H. OTHER HELMINTHS
Ivermectin at 200 p.g/kg, given orally in the paste formulation, was
effective against Strongyloides westeri in foals (Ryan and Best 1985;
Schlichting and Stoye 1985). This observation is in accord with earlier
studies using the injectable formulation (see reviews cited previously).
Ponies with naturally acquired infections of Setaria equina were treated
intramuscularly with ivermectin. Dosages of 200 to 500 p.g/kg gave
moderate efficacy (80%-88%) (Klei, Torbert, and Ochoa 1980). Iver-
mectin was given to a pony with severe jaw disease attributed to invasion
of the tissue by Micronema deletrix, but the disease was not alleviated
(Keg et al. 1984). A pony continued to pass Anoplocephala spp. eggs in
the feces after treatment with ivermectin paste (Torbert, Kramer, and
Klei 1982), as would be expected in view of the lack of efficacy of this
drug against tapeworms in general.
J. SARCOPTES SCABIEI
Ivermectin was highly effective against Sarcoptes scabiei (of goat origin)
in experimentally infected donkeys, Equus asinus (Abu-Samra et al.
1985). Similarly, systemic ivermectin treatment in conjunction with a
topical acaricidal dressing was reported curative against sarcoptic mange
(perhaps of fox origin) in horses (Christensson et al. 1984).
REFERENCES
Abu-Samra MT, Ali BH, Musa BE, Ibrahim KEE (1985) Experimental infection
of the domestic donkey (Equus asinus asinus) with a goat strain of Sarcoptes
scabiei, and treatment with ivermectin. Acta Tropica 42:217-224.
Asquith RL, Plue RE, Seward RL (1983) Field performance of the equine
parasiticide, ivermectin, in an oral paste. Equ. Vet. J. 3:90-91.
242 W.C. Campbell, W.H.D. Leaning, and R.L. Seward
Baker NF, Miller IE, Madigan JE, Fulton R, Seward RL (1984) Anthelmintic
action of ivermectin, oxibendazole, and pyrantel pamoare against thiaben-
dazole-resistant small strongyles of horses. Equ. Pract. 6:8-19.
Bello TR (1985) Proc. 30th Ann. Mtg., Am. Assoc. Vet. Parasit., University of
North Carolina.
Boraski EA (1987) Ivermectin, efficacy against ascarids-new information. Calif.
Vet. 41:16-17.
Britt DP, Preston JM (1985) Efficacy of ivermectin against Dictyocaulus arnfieldi
in ponies. Vet. Rec. 116:343-345.
Burrows RO, Thomson BM, Lindsey MJ (1985) Efficacy of ivermectin against
nematodes of horses including small strongyles resistant to benzimidazole.
Austral. Vet. J. 61:343-344.
Campbell WC (1985) Ivermectin: An update. Parasit. Today 1:10-16.
Campbell WC, Benz GW (1983) Ivermectin: A review of efficacy and safety.
J. Vet. Pharm. & Therap. 7:1-16.
Christensson D, Lindstedt E, Wierup M, Aropsenius J (1984) Sarcopotic mange in
horses in Sweden. Svensk Vet. 36:15-17 (in Swedish).
Corba J, Andrasko H, Stoffa P, Holakovsky P (1986) Efficacy of Eqvalan™ and
Panacur™ paste against gastrointestinal nematodes of horses. Veterinarstvi
36:79-80 (in Slovak).
DiPietro JA, Lock TF, Todd KS, Davis JL (in press) Evaluation of ivermectin for
larvacidal effect in experimentally induced Parascaris equorum infections in
pony foals. Am. J. Vet. Res.
DiPietro JA, Lock TF, Todd KS, Reuter VE (1987) Evaluation ofivermectin paste
in the treatment of ponies for Parascaris equorum infections. J. Am. Vet. Med.
Assoc. 190:1181-1183.
Egerton JR, Brokken ES, Suhayda D, Eary CH, Wooden JW, Kilgore RL (1981)
The antiparasitic activlty of ivermectin in horses. Vet. Parasit. 8:83-88.
Egerton JR, Seward RL, Robin B (1984) Ivermectin as an antiparasitic agent for
horses. Proc. MDS AGVET Symposium: Recent Developments in the Control
of Animal Parasites, XXII World Veterinary Congress, Perth, Australia, Aug.
25-26, 1983, pp. 49-55.
Foil LD, Klei TR, Miller RI, Foil CS, French DD (1985) Pathogenesis of equine
ventral midline dermatitis and its association with Onchocerca cervicalis and
Haematobia irritans. Abst. 196, 11th Conf. World Assoc. Adv. Vet. Parasitol.,
Rio de Janeiro, Brazil, Aug. 5-9, 1985.
French DD, Klei TR, Chapman MR, Torbert BJ (1985) Field evaluation of
ivermectin against benzimidazole resistant strongyles in Louisiana horses.
Abst. 197, 11th Conf. World Assoc. Adv. Vet. Parasitol.
French DD, Klei TR, Foil CS, Miller RI, Foil LD, Chapman MR, McClure JJ
(1988a) Efficacy of ivermectin in paste and injectable formulations against
microfilariae of Onchocerca cervicalis and resolution of associated dermatitis in
horses. Am. J. Vet. Res. 49:1550-1554.
French DD, Klei TR, Taylor HW, Chapman MR, Wright FR (1988) Efficacy of
ivermectin in the oral paste formulation against naturally acquired adult and
larval stages of Parascaris equorum in pony foals. Am. J. Vet. Res. 49:1000-
1003.
French DD, Klei TR, Chapman MR and Taylor HW (in press) Efficacy of
ivermectin in the oral paste and oral drench formulations against migrating
larvae of Parascaris equorum. Am. J. Vet. Res.
17. Use of Ivermectin in Horses 243
Torbert BJ, Kramer BS, Klei TR (1982) Efficacy of injectable and oral paste
formulations of ivermectin against gastrointestinal parasites in ponies. Am. J.
Vet. Res. 43:1451-1453.
Weiss J (1984) Value of ivermectin in treating certain parasitoses of horses. Rev.
Med. Vet. 135:425-433 (in French).
CHAPTER 18
I. DOGS
A. Products
1. Heartgard 30
2. Heartgard 30 chewable
B. Antiparasitic efficacy
1. Endoparasites
a. Dirofilaria immitis (heartworm)
b. Dipetalonema reconditum
c. Toxocara canis
d. Toxascaris leonina
e. Ancylostoma caninum (hookworm)
f. Uncinaria stenocephala (hookworm)
g. Strongyloides stercoralis
h. Filaroides osleri
i. Capillaria spp.
j. Trichuris vulpis (whipworm)
2. Ectoparasites
a. Sarcoptes scabei
b. Demodex canis
c. Otodectes cynotis
d. Ctenocephalides cati
II. CATS
A. Products
B. Antiparasitic efficacy
1. Endoparasites
a. Aelurostrongylus abstrusus
b. Toxocara cati
c. Ancylostoma spp.
d. Other helminths
2. Ectoparasites
a. Nodoedres cat;
b. Otodectes cynotis
c. Ctenocephalides cati
246 W.C. Campbell
I. Dogs
A. PRODUCTS
1. Heartgard 30™
This is a product for the prevention of heartworm disease in dogs. It is for
use by, or on the order of, a licensed veterinarian, and consists of
color-coded packages of white, hard tablets containing ivermectin in the
amount of 65 ILg (blue code, for dogs weighing up to 25Ib), 136 ILg (green
code, for dogs weighing 26 to 50 lb), or 272 ILg (brown code, for dogs
weighing 51 to 100 lb). When administered according to this schedule, the
tablets provide a minimum ivermectin dosage of 6 ILg/kg.
The product is to be administered at monthly intervals, beginning
within one month after the first expected exposure to mosquitoes and
ending within a month after the last potential exposure. The indication for
use in heartworm prevention is based on efficacy studies summarized
below.
The product is not recommended for dogs under 6 weeks of age.
Certain dogs, particularly Collies, are exceptionally susceptible to iver-
mectin toxicity (see Chapter 10). In such dogs, adverse reactions have
been reported when ivermectin was given under experimental conditions
at 200 ILg/kg but not when given at 50 ILg/kg.
2. Heartgard 30 Chewable ™
The chewable formulation consists of tablets that are designed to be
readily accepted by dogs when proffered to them, thus obviating the need
to insert the medication into the dog's mouth. In other respects the
product's use is similar to that of Heartgard 30™ .
B. ANTIPARASITIC EFFICACY
1. Endoparasites
a. Dirofilaria immitis (heartworm):
b. Dipetalonema reconditum
Lindemann and McCall (1983) gave ivermectin orally at 250 ltg/kg to
three Beagle dogs experimentally infected with D. reconditum. Treatment
was followed by a reduction of more than 99% in the number
of circulating microfilariae. When the dogs were necropsied, no adult
D. reconditum were found in the abdominal cavity or connective tissues,
but the authors cautioned that the absence of adult worms in this
experiment is not necessarily a reflection of drug efficacy.
c. Toxocara canis
Treatment of patent naturally acquired infections of T. canis in dogs
reduced the worm burden by 91% when the ivermectin dosage was
200 ltg/kg subcutaneously (s.c.) and 97% when the dosage was 400 ltg/kg
s.c. (Anderson and Roberson 1982).
The immature stages of T. canis are important in two quite different
contexts-the intestinal and the extraintestinal. Against the intestinal
fourth stage larvae (in dogs treated 2 weeks after inoculation with
infective eggs) ivermectin was 97% effective at 200 ltg/kg s.c. and 98%
effective at 400 ltg/kg s.c. (Anderson and Roberson 1982). Dosages of 50
and 100 ltg/kg s.c. gave no significant reduction in worm burden.
Yazwinski, Tilley, and Greenway (1982) also treated dogs 2 weeks after
inoculation with infective eggs of T. canis, but they reported 100%
efficacy at dosages of 100 ltg/kg s.c. and even 50 ltg/kg s.c. This
discrepancy may be due to the difference in intensity of infection, the
mean control worm burden being 232 in the former trial and only 5.4 in the
latter.
The importance of the extraintestinal (L2) larvae of T. canis lies in their
role in transuterine and transmammary transmission from bitch to pup;
and it has been notably difficult to block this transmission by chemo-
therapy. Using experimentally infected mice as a model, Abo-Shenada
(1982) found that L2 larvae were affected by ivermectin at high dosage
(2000 ltg/kg) given p.o. or s.c. on Days 2 to 7 post inoculation. In mice so
18. Use of Ivermectin in Dogs and Cats 251
treated the larvae failed to migrate from the host liver to the brain and
muscles; but when treatment was given after the larvae had completed
their migration (Days 8 to 13) even that very high dosage failed to reduce
their number. Studies in dogs, however, suggest that it is possible to
affect T. canis in the tissues in such a way that pups born to treated
bitches are essentially free of infection. Bitches inoculated with T. canis
eggs were treated with ivermectin around the 42nd day of gestation (when
the larvae are reactivated from the tissues) and the pups subsequently
born to those bitches were necropsied either at birth or at weaning (Shoop
et al. 1988). Treatment regimens that resulted in greater than 99%
reduction in worm burden and in the passage of eggs in the feces were:
1000 ~g/kg s.c. on Days 20 and 42 of gestation; 500 ~g/kg s.c. on Days 38,
41,44, and 47 of gestation; or 1000 ~g/kg s.c. on Day 20 followed by 500
~/kg on Days 42, 47, and 53 of gestation.
d. Toxascaris leonina
T. leonina appears to be less sensitive to ivermectin than T. canis, but
very little comparative information is available. Using experimentally
infected dogs, Anderson and Roberson (1982) found that dosages up to
400 ~g/kg s.c. did not give satisfactory activity against the adult worms.
Yazwinski, Tilley, and Greenway (1982) treated dogs 4 weeks after
inoculation with T. leonina eggs (when the worms would have been in the
L4 stage). They recorded 100% efficacy with a dosage of 200 ~g/kg s.c.,
but the mean number of worms recovered from control dogs was small.
when given orally but 90% effective when given subcutaneously (Egerton,
Eary, and Suhayda 1985). When given by either route, a dosage of
24 JLg/kg was more than 96% effective against the adults and against L4
larvae (Egerton, Eary, and Suhayda 1985).
g. Strongyloides stercoralis spp.
In a preliminary probe, ivermectin was given as a single oral dose at
800 JLg/kg to 1 of 2 dogs that had severe (corticosteroid-enhanced)
strongyloidiasis. The untreated dog, when necropsied 6 days after treat-
ment, yielded 56,556 adult intestinal worms and 976,185 intestinal larvae.
The treated dog yielded no intestinal adults and only 2 larvae (L.M.
Aikens and G.A. Schad, personal communication 1988). Negligible
numbers of worms or larvae were recovered from extraintestinal sites in
either dog.
h. Filaroides osleri
There seems to be no published evidence of the efficacy of ivermectin
against F. osleri, but Clayton (1983) mentioned en passant that a single
injection at 400 JLg/kg had shown "promising results in a limited number
of experimental F. osleri infections."
i. Capillaria spp.
A single dog with nasal capillariasis (C. aerophilia) was given ivermectin
orally at 200 JLg/kg. The nasal discharge stopped within 7 days and the
feces became free of eggs within 14 days. There was no relapse for at least
8 months (Evinger, Kazacos, and Cantwell 1985). A single dog with
urinary capillariasis (c. plica) was injected subcutaneously with iver-
mectin at 200 JLg/kg (after temporary alleviation of clinical signs had been
achieved by fenbendazole therapy). The ivermectin treatment was fol-
lowed by apparent clinical and parasitological cure (passage of eggs in
urine ceased within 7 days) (Kirkpatrick and Nelson 1987).
j. Trichuris vulpis (whipworm)
When tested against immature (4-week-old) T. vulpis in experimentally
infected dogs, ivermectin was 100% effective at 100 JLg/kg s.c. (Yaz-
winski, Tilley, and Greenway 1982), but the control dogs had a mean of
only 6.0 worms. Against mature T. vulpis, treatment was more than 99%
effective at 100 JLg/kg s.c., and more than 95% effective at 200 JLg/kg p.o.
(J. R. Egerton, unpublished data).
2. Ectoparasites
a. Sarcoptes scabiei
Dogs with natural infestations of Sarcoptes scabiei were treated sub-
cutaneously with ivermectin at various dosages. At 200 JLg/kg or higher,
treatment resulted in complete cure by Day 7 after treatment. Dosages as
254 W.C. Campbell
low as 50 p,g/kg were also effective but took 2 to 3 weeks to achieve their
full effect (Yazwinski et al. 1981). The efficacy of ivermectin was
confirmed in a clinical trial involving almost 300 dogs. Despite confine-
ment of the dogs "in a heavily contaminated environment which was
conducive to repeated reinfestation during treatment," 2 doses of the
drug at 200 p,g/kg s.c., given 14 days apart, provided marked clinical
improvement and complete parasitological cure 2 weeks after the second
injection (Scheidt et al. 1984). Additional evidence of efficacy was
provided by Gravino, de Caprariis, and Agresti (1985), who used
400 p,g/kg s.c., and gave 3 treatments at 8-day intervals.
b. Demodex canis
Despite anecdotal reports of ivermectin's efficacy in demodectic mange,
there is little or no scientific evidence to support it. In one clinical study,
ivermectin was administered subcutaneously to 4 dogs with generalized
amitraz-resistant demodecosis. The dosage was 400 p,g/kg and was
repeated weekly for a total of 8 treatments. Despite this rather intensive
treatment, and despite the apparent clinical and parasitological cure of 1
dog, no reduction in mite number could be demonstrated in the remaining
3 dogs (Scott and Walton 1985). All 4 dogs, however, showed some
clinical improvement, and the authors suggested that this might be a
reflection of decreased mite activity in the skin. Perhaps the dosage used
in that study was not high enough, because Gravino, de Caprariis, and
Giglio (1985) reported a favorable response in an (uncontrolled?) study in
which 20 dogs were given 600 p,g/kg s.c. at weekly intervals for a total of 5
treatments. On the other hand, Belot, Parent, and Pangui (1984) reported
that demodectic mange responded to 2 doses of ivermection at 400 p,g/kg
s.c. given 15 days apart. These 3 trials were conducted in the United
States, Italy, and France, respectively, so there may be geographical
strains with different degrees of susceptibility. Additional controlled
studies are needed to clarify the matter.
c. Otodectes cynotis
The efficacy of ivermectin versus O. cynotis was demonstrated by
Yazwinski and colleagues (1981) in a controlled clinical trial using natural
infections. A single subcutaneous injection at 200 p,g/kg gave complete
cure in 14 days, while a single treatment at 400 p,g/kg resulted in cure in 7
days. Similarly, Molina (1986) found that a single injection at 200 p,g/kg
gave complete cure in 14 days, and Chauve (1984) also found that dosage
to be fully effective.
d. Ctenocephalides cati
Despite anecdotal reports of efficacy, neither abamectin nor ivermectin
has been shown to have significant activity against fleas on dogs. In
a controlled study, ivermectin was given orally, at 50 p,g/kg/day or
18. Use of Ivermectin in Dogs and Cats 255
II. Cats
A. PRODUCTS
Ivermectin has not been approved by regulatory agencies for use in cats in
any form, nor has any application for such use been submitted by the
manufacturer.
B. ANTIPARASITIC EFFICACY
1. Endoparasites
a. Aelurostrongylus abstrusus
Blagbum and colleagues (1987) gave ivermectin orally at various dosages
to cats with natural infection of A. abstrusus, but there was little if any
evidence of efficacy. Kirkpatrick and Megella (1987) tested ivermectin
against the same parasite in a single naturally infected cat. After an
injection of ivermectin at 200 JLg/kg, larvae continued to be detectable in
the feces. After a second injection at 400 JLg/kg larvae could no longer be
found in the feces, but no improvement in the pulmonary disease could be
demonstrated radiographically, and the findings in this single animal are
of doubtful significance.
b. Toxocara cati
In a single treated cat, the passage of T. cati eggs in the feces ceased after
treatment of the cat with ivermectin at 200 JLg/kg s.c. (Kirkpatrick and
Megella 1987). The limited evidence of efficacy provided by this single
instance is supported by observations made by Blagbum and colleagues
(1987) in several cats with very light natural T. cati infection. In those
cats, oral treatment at 100 or 300 JLg/kg appeared to be highly effective,
but additional studies would clearly be needed to determine the efficacy of
ivermectin against this common ascarid.
c. Ancylostoma spp.
Hookworms in cats, like those in dogs, appear to be very sensitive
to ivermectin. Using cats with very light natural infections, Blagbum
and colleagues (1987) obtained evidence that dosages ranging from 10 to
300 JLg/kg p.o. and s.c. were active against A. braziliense; and in one cat
infected with A. tubaeforme, a dosage of 10 JLg/kg p.o. appeared to be
effective.
256 W.C. Campbell
d. Other Helminths
It is not known whether ivermectin is effective against Capillaria sp. in
cats. Ivermectin is not known to be active against any species of flukes or
tapeworms, and the incidental observations of Blagburn and colleagues
(1987) on several tapeworm species in cats are in accord with this general
finding.
2. Ectoparasites
a. Nodoedres cati
According to Bigler, Waber, and Pfister (1984), a single s.c. injection of
ivermectin at 1000 ILg/kg was effective in controlling an outbreak of
notoedric mange. This was a clinical report, not the record of a laboratory
trial, but all 17 treated cats were judged completely cured. Suarez and
Bertero (1986) used the same high dosage successfully against N. cati and
reported that all skin scrapings were negative for mites 7 and 30 days after
treatment.
b. Otodectes cynotis
Chauve (1984) reported that ivermectin at 200 ILg/kg s.c. was effective
against naturally acquired o. cynotis infection in cats. Chauve and
Reynaud (1984) extended this observation to experimentally induced
infestations, and showed that mites disappeared 14 days after the cats had
received a single injection at 666 ILg/kg. Further clinical reports of the
effectiveness of the 200 ILg/kg s.c. dosage, and of a dosage of 400 ILg/kg
s.c., were provided by Church (1985) and Franc, Dorchies, and Soubey-
roux (1985), respectively.
c. Ctenocephalides cati
In a clinical note, Bigler, Waber, and Pfister (1984) reported that adult
populations of fleas (presumably C. cati) were "decimated" by an
injection of ivermectin at the very high dosage of 1000 ILg/kg. However,
there appears to be no evidence, obtained under controlled conditions, of
the efficacy of ivermectin against fleas on cats.
REFERENCES
mectina nel trattamento dello rogna sarcoptica del cane. Acta Med. Vet.
31:43-47.
Gravino AE, deCaprariis D, Giglio M (1985) Impiego della 22,23-diidroaver-
mectina nel trattamento della Demodicosi del cane. Acta Med. Vet. 31:185-189.
Jackson RF (1987) Microfilaricides. Sem. in Vet. Med. & Surg. (Sm. Anim.)
2:44-47.
Jackson RF (1984) Ivermectin again. Am Heartworm Soc Bull 10:9.
Jackson RF, Seymour GW (1981) Efficacy of avermectins against microfilariae of
Dirofilaria immitis. In Otto GF (ed) Proceedings of the Heartworm Symposium
1980. Vet. Med. Pub. Co., Edwardsville, KS, pp 131-136.
Jackson RF, Seymour WG, Beckett R (1986) Routine use of 0.05 mg/kg
ivermectin as a microfilaricide, in Otto GF (ed): Proceedings of the Heartworm
Symposium, 1986. Washington, DC, American Heartworm Society, pp 37-39.
Kirkpatrick CE, Megella C (1987) Use of ivermectin in treatment of Aeluro-
strongylus abstrusus and Toxocara cati infections in a cat. J. Am. Vet. Med.
Assoc. 190:1309-1310.
Kirkpatrick CE, Nelson GR (1987) Ivermectin treatment of urinary capillariasis in
a dog. J. Am. Vet. Med. Assoc. 191:701-702.
Knight DH, Campbell WC, Weiner DJ, Washabau RJ (1986) Microfilaricidal
efficacy of ivermectin in adulticide treated and untreated heartworm infected
dogs. In Otto GF (ed) Proceedings of the Heartworm Symposium 1986.
American Heartworm Society, Washington, D.C., pp 19-27.
Lindemann BA, McCall JW (1983) Microfilaricidal activity ofivermection against
Dipetalonema reconditum. J. Vet. Pharm. & Therap. 6:75-76.
Lok JB, Harpaz T, Knight DH (1988) Abnormal patterns of embryogenesis in
Dirofilaria immitis treated with ivermectin. J. Helminthol. 62:175-180.
McCall JW, Lindemann BE, Porter CA (1981) Prophylactic activity of avermec-
tins against Dirofilaria immitis, in Otto GF (ed): Proceedings of the Heartworm
Symposium, 1980. Edwardsville, KS, Veterinary Medicine, pp 126-130.
McCall JW, Dziminanski MT, Plue RE (1986) Ivermectin in heartworm pro-
phylaxis: Studies with experimentally induced and naturally acquired infec-
tions. In Otto GF (ed) Proceedings of the Heartworm Symposium 1986.
American Heartworm Society, Washington, D.C., pp 9-13.
McManus EC, Pulliam JD (1984) Histopathologic features of canine heartworm
microfilarial infection after treatment with ivermectin. Am. J. Vet. Res.
45:91-97.
Molina CG (1986) The activity of ivermectin against Octodectes cynotis in
naturally infested dogs. Vet. Mex. 17:39-40.
Paul AJ, Todd KS, Sundberg JP (1986a) Efficacy of ivermectin against Dirofilaria
immitis larvae in dogs 30 and 45 days after induced infection. Am. J. Vet. Res.
47:883-884.
Paul AJ, McCall JW, Todd KS (1986b) Efficacy of ivermectin against Dirofilaria
immitis larvae in dogs forty-five days after induced infection. In Otto GF (ed)
Proceedings of the Heartworm Symposium 1986. American Heartworm Soci-
ety, Washington, D.C., pp 33-35.
Plue RE, Seward RL, Acre KE, Cave JS, Schlotthauer JC, Stromberg BE (1983)
Clearance of Dirofilaria immitis, microfilariae in dogs using 200 mcg/kg
ivermectin subcutaneously, in Otto GF (ed): Proceedings of the Heartworm
Symposium, 1983, Edwardsville, KS, Veterinary Medicine, pp 153-160.
18. Use ofIvermectin in Dogs and Cats 259
I. Introduction
II. Laboratory Animals
A. Mice
1. Endoparasites
a. Aspiculuris tetraptera
b. Nematospiroides dubius
c. Strongyloides spp.
d. Syphacill obvelata
e. Toxocara canis
f. Trichinella spp.
g. Trichuris muris
2. Microfilariae
3. Ectoparasites
a. Mites
b. Triatomid bugs
c. Cuterebra fontinella
B. Rats
a. Angiostrongylus cantonensis
b. Trichinella spiralis
c. Syphacia muris
C. Guinea Pigs
1. Ticks
2. Tsetse flies
3. Mange mites
D. Hamsters
E. Gerbils
F. Rabbits
1. Mange mites
2. Tsetse flies
3. Ticks
G. Ferrets and Jirds
19. Use of Ivermectin in Mammals, Birds, Fish, and Reptiles 261
VI. Reptiles
A. Snakes
B. Chelonians
VII. Conclusion
I. Introduction
The first observations on the antiparasitic properties of the avermectins
were made in laboratory animal screens. Ivermectin is potent (allowing
for administration of very small doses); it can be administered either
orally or parenterally; and it is effective against both arthropod and
nemathelmintic endo- and ectoparasites. These attributes have led to the
extensive use of the compound for treating parasitic infections in a variety
of laboratory, wild, zoo, and exotic mammals, as well as in reptiles, birds,
and fish. Apart from domestic animals, ivermection has been approved by
various regulatory authorities for use in camels and reindeer only. The
treatment of any other species with the product is considered to be
"extralabel" usage.
1. Endoparasites
a. Aspiculuris tetraptera
Ivermectin added to the drinking water for 24 hours to provide a dose of
1.0 to 1.6 mg/kg body weight was fully effective in removing naturally
acquired infections of this parasite from laboratory mice (Hasslinger and
Wiethe 1987).
b. Nematospiroides dubius
Oral treatment at 5 mg/kg on Days 3 or 6 post infection was reported to
remove all worms present (Sayles and Jacobson 1983); Rajasekariah and
colleagues (1986) reported that a much lower dose of 0.3 mg/kg com-
pletely eliminated adult worms from mice.
c. Strongyloides spp.
Grove (1983) reported that, following ivermectin treatment of mice with
01 mg on Days 0 and 1 (migration phase) and 3 and 4 (intestinal phase)
after infection with S. ratti, the treated animals subsequently passed no
19. Use of Ivermectin in Mammals, Birds, Fish, and Reptiles 263
larvae in the feces. Moreover, he found that a single dose of .05 mg 6 days
after infection eradicated intestinal adult worms. Rajasekariah and col-
leagues (1986) reported that 2 treatments at 0.3 mg/kg are required to
eliminate adult parasites from mice.
Grove (1983) reported that S. stercora lis responds similarly to iver-
mectin: daily doses of .01 mg of ivermectin on Days 0, 1, 2, and 3 after
infection greatly reduced parasite numbers in the muscles, while a single
dose of 0.1 mg given 1 hour or 6 days after infection completely eradicated
larvae from the muscles.
d. Syphacia obvelata
Ostlind, Nartowicz, and Mickle (1985) reported that feeding a diet
containing 0.0005% ivermectin for 6 days to mice naturally infested with
pinworms was more than 99% effective against both adult and immature
worms; moreover, a single oral treatment at a dose rate of 2 mg/kg was
94% to 100% effective against adult and 86% to 97% effective against
immature S. obvelata.
e. Toxocara canis
Treating mice harboring immature T. canis with ivermectin at 2 mg/kg for
2 to 7 days after infection (Abo-Shenada and Herbert 1984), or 0.4 mg/kg
on Days 15 to 28 after infection (Carillo and Barriga 1987) alters the
distribution of the larvae: Larvae are retained in the liver and lungs, and
fewer migrate to the brain. None of these dose rates completely eradica-
ted the parasite from the tissues, however.
f. Trichinella spp.
Treating mice with avermectin at a dose rate of 3 mg/kg reduced the
number of intestinal T. spira lis 80% to 90% (Campbell, Blair, and Lotti
1979). The number of parasites was not reduced significantly, however,
following ivermectin treatment (5-10 mg/kg) of mice harboring the
migrating and muscle stages of a number of Trichinella spp. (Sanmartin-
Duran et al. 1986). Ivermectin was effective against intestinal forms of T.
nelsoni and T. nativa in this study, reducing their number by 91% to 96%
(relative to controls), following treatment at 2 and 5 mg/kg on Days 1 and
6 post infection.
g. Trichuris muris
Although some degree of efficacy was apparent after a repeated dose of
ivermectin at 10 mg/kg, mice treated for T. muris infestation were not
completely cured (Rajasekariah et al. 1986).
2. Microfilariae
Ivermectin's microfilaricidal activity has been demonstrated against
immature stages of a number of filarial parasites in mice.
264 M.D. SoU
3. Ectoparasites
The efficacy of ivermectin has been evaluated against some parasitic
arthropod species in the mouse.
a. Mites
Silverman, Blatt, and Lerro (1983) reported that feeding mice a commer-
cial diet containing 2 mg of ivermectin/kg of feed for two 6-day periods,
1 week apart, effectively eliminated Myobia musculi mites. Efficacy of
the product against murine mites was confirmed by Wing, Courtney,
and Young (1985), who demonstrated that 2 subcutaneous injections at
0.2 mg/kg, given 1 week apart, effectively eliminated Mycoptes musculi-
nus and Myobia musculi from laboratory mice. Single treatments reduced
infections only temporarily.
Baumans et al. (1988) reported that spraying mice with a .01% solution
of ivermectin was effective in eliminating symptoms of mite infection for
12 weeks after treatment.
b. Triatomid bugs
Treating mice with ivermectin at 0.2 mg/kg was reported to have caused
high mortality and a reduction in egg-laying potential of Rhodnius prolixus
and Hemiptera triatominae feeding on these animals (Azambuja et al.
1985).
19. Use ofIvermectin in Mammals, Birds, Fish, and Reptiles 265
c. Cuterebra fontinella
The C. fontinellalmouse model was used to demonstrate the systemic
insecticidal efficacy of the avermectins. Ostlind, Cifelli, and Lang (1979)
reported an avermectin mixture to be effective at dose levels of 0.15
mg/kg and higher. Drummond (1980) subsequently confirmed this activity
using ivermectin in mice with induced C. fontinella infections.
B. RATS
1. Ticks
In screening tests, ivermectin has been shown to be completely effective
against the lone star tick (Amblyomma americanum) when adult ticks are
266 M.D. SolI
fed on treated guinea pigs (Hunt 1982). Efficacy has also been demon-
strated against larval, nymphal, and adult Rhipicephalus sanguine us-as
well as A. americanum-when these ticks were fed on guinea pigs treated
at dose rates of up to 2.0 mg/kg (Wilkins et al. 1981).
2. Tsetse Flies
Mortality of tsetse flies (Glossina palpalis) occurred among flies fed
exclusively on guinea pigs treated with ivermectin at dose rates greater
than 2.0 mg/kg (Distelmans, D'Haeseleer, and Mortelmans 1983).
3. Mange Mites
Ivermectin has been used successfully to treat mange caused by the mite
Trixacarus caviae in a guinea pig colony. The compound was adminis-
tered at 0.2 mg/kg subcutaneously on 3 occasions with 7-day intervals
(Harvey 1987).
D. HAMSTERS
E. GERBILS
F. RABBITS
1. Mange Mites
Although subcutaneous injection of ivermectin at 0.2 mg/kg is reported to
be effective for treating rabbits naturally infected with psoroptic and
19. Use ofIvermectin in Mammals, Birds, Fish, and Reptiles 267
2. Tsetse Flies
The reproductive capacity of Glossina palpalis fed on rabbits treated with
ivermectin at 2.0 mg/kg has been shown to be significantly reduced
(Abbeele, D'Haeseleer, and Goossens 1986).
3. Ticks
In an assessment of the suitability of the rabbit as a model for testing
systemic ascaricides, ivermectin has been shown to be effective against
induced infestations of Ornithodoros moubata for up to 7 days after
treatment at dose rates of 0.1 to 0.4 mg/kg (Frossard 1981).
A. RODENTS
B. HEDGEHOGS
C. CARNIVORES
1. Foxes
a. Endoparasites
Blagbum and colleagues (1986) report a study in which wild foxes were
trapped and an assessment of helminth infection made by fecal flotation.
Animals were then divided into groups and treated with a number of
anthelmintics, including ivermectin at 0.4 mg/kg. Of the 9 foxes positive
for Ancylostoma spp. eggs prior to ivermectin treatment, none was
positive by 3 weeks after treatment.
In a study involving more than 500 female arctic and silver foxes, those
treated with ivermectin subcutaneously at 0.2 mg/kg had better concep-
tion and parturition rates than untreated animals (Kopczewski et al.
1987). The numbers of cubs with Toxocara canis eggs in the feces at 30,
60, and 75 days of age was approximately 50% lower for those cubs born
to ivermectin-treated vixens than for those born to controls.
19. Use ofIvermectin in Mammals, Birds, Fish, and Reptiles 269
b. Ectoparasites
The efficacy of ivermectin has been assessed against 2 mite species-
Sarcoptes scabiei var canis and Cheyletiella blakei-in foxes.
Sarcoptic mange mites were eradicated from foxes treated with iver-
mectin at dose rates of 0.2 or 0.4 mg/kg once or 0.2 mg twice with a
5-week interval (Berge and Smeds 1984). These authors recommend a
treatment regimen involving an initial dose of 0.4 mg/kg, followed by a
second dose of 0.2 mg/kg, 2 to 3 weeks later, to control this parasite in
foxes.
In a study involving polar foxes, Malczewski and colleagues (1984)
reported that 6 months after 2 subcutaneous ivermectin treatments at
dose rates up to 0.5 mg/kg, 6 weeks apart, no C. blakei mites were
recovered from treated animals; these foxes were also free from clinical
signs of infestation after treatment.
2. Wolves
Treatment of captive wolves with ivermectin by intramuscular injection,
the direct oral route, or in treated meat was found to kill the adult and
nymphal stages of the biting louse, Trichodectes canis (Taylor and
Spraker 1983). Free-ranging packs were subsequently successfully
treated with injections and treated meat. Offspring of previously infested,
treated adults were also reported to be free of lice.
D. UNGULATES
1. Camels
Ivermectin given at 0.2 mg/kg subcutaneously effectively treats and
controls gastrointestinal nematodes and sarcoptic mange mites in camels
(Camelus dromedarius) (Ibrahim et al. 1981; Tager-Kagan and Robin
1986; Jones 1987). Studies have demonstrated a wide safety margin, but
270 M.D. So11
3. Reindeer
Ivermectin, administered subcutaneously at 0.2 mg/kg, is approved for
treating and controlling warbles (Oedemagena tarandi) in reindeer (Rang-
ifer tarandus) in the United States. Under an IR4 (minor species)
development project conducted in Alaska, a high degree of efficacy was
established in extensive studies (Dieterich 1985). Safety was demon-
strated and no adverse reactions were elicited following treatment of
animals at 5 and 10 times the recommended dose. The withdrawal period
is 56 days.
Ivermectin's efficacy in reindeer is supported by a field study con-
ducted in Sweden (Nordkvist et al. 1984). Grazing reindeer treated with
ivermectin at 0.2 mg/kg were returned to pasture for 150 days during the
winter. Efficacy, determined by slaughtering and processing 5 treated and
5 control animals at the end of this time, was reported to be at or near
100% against Oedemagena tarandi, Cephenemyia trompe, Dictyocaulus
viviparus, and Elaphostrongylus rangiferi. The high level of efficacy,
despite the long treatment/slaughter interval, is due to the reindeers'
decreased exposure tei infective larvae in the winter months. The average
weight loss ofthe treated group over the winter was significantly less than
that of the controls.
4. Deer
Ivermectin's efficacy has been evaluated against various endo- and
ectoparasites in red deer (Cervus elaphus), white-tailed deer (Odocoileus
virginianus), and mule deer (0. hemionus).
272 M.D. SolI
a. Endoparasites
Dictyocaulus viviparus. The high level of efficacy of ivermectin against
lungworm in domestic animals has also been demonstrated in red deer.
Treatment at 0.1 mg/kg subcutaneously 1 to 4 weeks after induced
infections was 100% effective against this parasite (Mackintosh and
Mason 1985). Oral treatment (0.2 mg/kg) of red deer on pasture reduced
fecal larval counts to low levels by 20 days after each of 4 treatments
(Bowie, Mackintosh, and Mason 1987). Mean larval output and pro-
portion of deer shedding D. viviparus larvae at 27 and 33 days after
treatment were significantly lower for ivermectin-treated animals than for
those treated with oxfendazole.
Parelaphostrongylus. In white-tailed deer, ivermectin has been shown to
be effective against certain immature stages of Parelaphostrongylus
tenuis (the meningeal nematode), but not against the adults. Kocan (1985)
reports that treatment at 0.1 or 0.2 mg/kg subcutaneously is effective
against larvae penetrating the abomasum. Treatment on Days 10 and 30
after infection was ineffective. This could be due to the apparent inability
of the avermectins to cross the blood-brain barrier, and the fact that the
larvae of this parasite are generally located in the spinal cord by Day 10
after infection. Fecal larval output was reduced completely by 10 days
after treatment of deer with patent infections, but larvae reappeared in the
feces by Day 28. These findings, together with the absence of eggs and
larvae in the lungs of animals slaughtered 12 days after treatment, indicate
activity against first stage larvae in the lungs and possibly an effect on egg
production (Kocan 1985).
Samuel and Gray (1988) report that ivermectin treatment (0.2 or 0.4
mg/kg) of white-tailed deer artificially infected with the muscle nematode
Parelaphostrongylus andersoni, eliminated first stage larvae from the
feces for up to six weeks. This was attributed to suppressed larval
production by adult females and/or destruction of larvae in the lungs.
Oral ivermectin treatment had little effect on the adults of another
metastrongyloid nematode Elaphostrongylus cervi in red deer (Watson
1986).
b. Ectoparasites
Lice. Treatment of mule and white-tailed deer with ivermectin at
0.2 mg/kg intramuscularly was reported to reduce the number of Linog-
nathus africanus by 90% within 7 days of treatment. A second treatment
21 days later further reduced louse numbers but did not eradicate them
(Foreyt, Rice, and Kim 1986). The interval between treatments in this
case was probably too long relative to the life cycle of the parasite to
completely eliminate the infection.
Fly Strike. Treatment of a single red deer stag suffering from fly strike
with 40 mg of ivermectin subcutaneously (total dose) reportedly aborted
the strike within 3 days (Fletcher 1984).
19. Use of Ivermectin in Mammals, Birds, Fish, and Reptiles 273
5. Bighorn Sheep
Desert bighorn sheep (Ovis canadensis), naturally infested with Psorop-
tes ovis, were successfully treated for scabies with ivermectin adminis-
tered intramuscularly at 0.5 or 1.0 mg/kg-either as a single or repeated
dose with a 14-day interval (Meleney, Wright, and Guillot 1980; Kinzer et
al. 1983). Ivermectin was also effective against this parasite in free-
roaming sheep treated by administering 35 mg of the drug loaded in frozen
gelatin "bullets" and fired from a modified rifle (Meleney, Wright, and
Guillot 1980).
Activity against lungworm has also been demonstrated. Under labora-
tory conditions, Miller and colleagues (1987) report that a single sub-
cutaneous injection (0.2 mg/kg) eliminated first stage Protostrongylus
larvae from the feces of bighorn sheep examined 4 weeks after treatment.
Good efficacy was confirmed by monitoring fecal larval counts of
bighorns treated subcutaneously (0.5 mg/kg) at the time of translocation,
over an extended period of time (Miller and Hobbs 1988).
6. Impala
Horak and colleagues (1984) evaluated the efficacy of ivermectin against
the endo- and ectoparasites of free-living impala treated subcutaneously
at 0.2 mg/kg. Based on parasite recoveries, a high level of efficacy was
demonstrated against the nematode parasites Longistrongylus sabie,
Haemonchus krugeri, Trichostrongylus thomasi, T. colubriformis, Im-
palia tuberculata, Gaigeria pachyscelis, and Oesophagostomum colum-
bianum. Activity was variable against Cooperia hungi, Cooperoides
hamiltoni, and Strongyloides spp.
Of the 4 tick species present on these animals, only Boophilus
decoloratus was markedly affected by treatment. The efficacy of iver-
mectin at this dose was high against the sucking lice Linognathus
aepycerus and L. nevilli but poor against the biting lice Damalinia
aepycerus and D. elongata.
This spectrum of activity is similar to that observed against gastroin-
testinal nematodes and lice on goats treated at 0.2 mg/kg subcutaneously
(unpublished).
7. Other Antelope
Based on fecal egg counts, ivermectin at 0.2 mg/kg is reported to be
effective against gastrointestinal nematodes of the genera Nematodirus,
Trichuris, Trichostrongylus, Ostertagia, Cooperia, Haemonchus, and
274 M.D. Soil
8. Bison
Studies have been conducted to provide data on the use of ivermectin at
0.2 mg/kg subcutaneously for controlling and treating hypodermosis
(Hypoderma bovis) in the American buffalo (Bison bison), under the
auspices of the IR4 (minor species) system (Schillhom, Sikarskie, and
Braselton 1987). These studies demonstrated safety at doses at 0.2 mg/kg
and 1.0 mg/kg. Efficacy against Hypoderma bovis was evaluated under
field conditions, by treating animals at 0.2 mg/kg in the fall. The mean
number of grub lesions in treated animals the following spring was zero
compared to 11.3 for the control animals (Schillhom, Sikarskie, and
Braselton 1987). The efficacy of ivermectin against Hypoderma in the
bison is therefore comparable to that established in cattle for this parasite.
9. Buffalo
The efficacy of ivermectin against louse infection (Haematopinus tuber-
culatus) has been assessed in buffaloes (Lau and Singh 1985). Sub-
cutaneous treatment at 0.2 mg/kg was less effective (85%) than treatment
at 0.4 mg/kg (100%) on evaluation 7 to 14 days later. Efficacy waned to
45% to 50% by 33 days after treatment in this study. It was suggested that
a second treatment 7 to 14 days after the first may result in better efficacy
over a longer period of time.
E. ELEPHANTS
F. NONHUMAN PRIMATES
1. Mite Infections
Joseph and colleagues (1984) reported that rhesus macaques with pul-
monary acariasis (Pneumonyssus) treated with ivermectin at 0.2 mg/kg
subcutaneously had only dead, fragmented mites in their lungs at ne-
cropsy, while control monkeys had numerous live mites. Histopathologi-
cally, inflammatory lesions were most severe in control animals and
decreased progressively with time in treated monkeys. Efficacy against
lung mites in this species has also been reported by Brack and Rietschel
(1986).
Efficacy has been demonstrated against Psorergates mites in stump-
tailed macaques (Bowman and Griffith 1987).
2. Nematode Infections
Feeding thiabendazole-medicated pellets for 1 week to rhesus monkeys
infected with Strongyloides fuelleborni did not eliminate the infection.
Brack and Rietschel (1986) subsequently treated the animals with iver-
mectin at 0.2 mg/kg subcutaneously for lung mite infections and found
this treatment to be effective against both the mites and the persisting
Strongyloides infection. No eggs were present in the feces 1 month after
treatment. Efficacy has been reported against natural Strongyloides spp.
infection in squirrel-monkeys (Battles, Greiner and Collins 1988).
An apparently healthy cymologous monkey (Macacafusicularis) which
died after routine inhalation anesthesia was found to be infected with the
276 M.D. SolI
IV. Birds
Ivermectin, administered orally, topicallY, and by injection, has been
used for the treatment of mite and nematode infestations in wild and
domesticated birds. Coles (987) considers the drug to work equally well
in these species when given orally or by injection and recommends oral
dosing.
In chickens, somnolence has been observed at doses of 5.4 mg/kg
(Zeman 1987). Listlessness and ataxia occurred at 16.2 mg/kg, but a dose
of 48.6 mg/kg was required to cause mortality. Mousa and colleagues
(1986b) reported that doses of 0.1 to 15.0 mg/kg of ivermectin were
nontoxic to chickens.
A. NEMATODES
a. Ascaridia
A single oral ivermectin treatment at 0.1, 0.2, or 0.4 mg/kg was found to
be ineffective against the tissue phase of Ascaridia galli in chickens
infested 12 days before treatment. These dosages were, however, 96% to
100% effective against adult worms present 40 days after infection (Mousa
et af. 1986b). These authors therefore recommended a repeated treatment
with a 30-day interval for controlling this parasite. The inclusion of
ivermectin in poultry feed at a level of 4 ppm was 84% effective against a
challenge infection of A. galli (R. L. Kilgore, unpublished data).
Successful treatment of Ascaridia cofumbae infection in pigeons required
2 oral doses 0.5 mg) 3 weeks apart (Schepkens, Duchatel, and Vinde-
vogel 1985).
Okaema (1988) reports that subcutaneous injection of guinea fowl
19. Use of Ivermectin in Mammals, Birds, Fish, and Reptiles 277
B. MITES
b. Cnemidocoptes spp.
Ivermectin has been used by intramuscular injection (0.2 mg/kg) and
topical application to treat "scaly-leg" in cage birds ranging in size from
budgies to macaws (Kelso 1984; Tassi 1984; Hogan et al. 1984; Ryan
1986). Heavily infested birds may require a second injection (Kelso 1984;
Hogan et al. 1984) or 2 oral treatments at 0.2 mg/kg 10 to 16 days apart
(Ryan 1987). Topical application of a 1% solution to crusts on the beak,
eyelids, legs, and pericloacal areas of affected birds was reportedly
effective in all but the most severe cases (Tassi 1984). Kummerfeld and
Nolte (1987) reported effective spot treatment of budgerigars for Cne-
midocoptes pilae infestation at levels of 0.4 to 20.0 mg of ivermectin/kg.
c. Sternostoma tracheolum
Dead mites were recovered from the lungs and air sacs of canaries
injected intramuscularly with .02 to .06 mg of ivermectin per bird
(Brownell 1984). In this study, clinical recovery occurred 10 days after
treatment, and the birds began singing 4 days later. Although no mortality
occurred in birds treated at these high levels, immobility was reported.
d. Cytodites nudus
Subcutaneous injection of golden pheasants infested with this air-sac mite
was effective only at high doses (50 mg/kg) (Grimm and Centurier 1986).
e. Pterolichidae
Three doses of ivermectin (0.2 mg/kg orally) at 4-week intervals were
effective for the treatment of quill mite infestation in an adult ostrich
(Hoover, Lochner, and Mullins 1988).
v. Fish
Ivermectin has been used to control parasitic copepods on fish; the drug
has shown some efficacy against nematodes. Dosage levels in fish are
critical: the toxic level is close to the therapeutic level in some species.
Although no mortalities or toxic effects were reported following oral
ivermectin treatment of Atlantic salmon or rainbow trout at 0.2 mg/kg,
the margin of safety is narrow, with mortalities occurring at twice this
dose (Palmer et al. 1987). Mortality of mottled sculpin, 5.5 cm in length,
was reported to have occurred after injection of more than .05 ml of a
.05% ivermectin solution (Heckman 1985).
19. Use of Ivermectin in Mammals, Birds, Fish, and Reptiles 279
A. COPEPODS
B. NEMATODES
IV. Reptiles
Although adverse reactions were not recorded in snakes given ivermectin
at 0.2 mg/kg (Lawrence 1984; Stanchi and Grisolia 1986), and the corn
snake (Elapha guttata) tolerated doses of up to 1.0 mg/kg without
apparent adverse clinical signs, chelonians were found to be sensitive to
the effects of the compound at much lower levels (Teare and Bush 1983).
Red-footed tortoises were killed by treatment at 0.4 mg/kg; paralysis
occurred with doses as low as 0.05 mg/kg when it was repeated within
72 hours. Doses which induced minimal signs of toxicosis ranged from
0.025 mg/kg in leopard tortoises to 0.1 mg/kg in box tortoises. A dose
of 0.05 mg/kg was considered safe in red-footed tortoises if treatment
intervals were not less than 7 days (Teare and Bush 1983).
A. SNAKES
B. CHELONIANS
Conclusion
Ivermectin is efficacious against a wide-range of endo- and ectoparasites,
in a wide variety of hosts, and is formulated in a variety of dosage forms.
Although mammals tolerate ivermectin very well, it must be empha-
sized that the safety of the drug for nondomestic species-with the
exception of camels and reindeer-has not been proven in controlled
studies. Use in nonmammalian species is not without risk, especially for
reptiles and fish, where the therapeutic dose may be close to the toxic
level.
Similarly, because the compound's efficacy against the wide array of
parasites infecting laboratory, zoo, wild, and exotic animals has not been
proven in controlled studies, clinical response to "extra-label" usage of
the product should be monitored closely.
REFERENCES
ivermectin against natural infection of Syphacia muris in rats. Lab. Ani. Sci.
37:791-792.
Battles AH, Greiner EC, Collins BR (1988) Efficacy of ivermectin against natural
infection of Strongyloides spp. in squirrel-monkeys. Lab. Ani. Sci. 38:474-485.
Baumans V, Havenaar R, Van Herck H, Rooymans TP (1988) The effectiveness
ofIvomec and Neguvon in the control of murine mites. Lab. Anim. 22:243-245.
Berge GN, Smeds E (1984) Efficacy of ivermectin against S. scabiei infestations of
foxes. Nord. Vet. Med. 36:156-161.
Bianco AE, Nwachukwa MA, Townson S, Doenhoff MJ, Muller RL (1986)
Evaluation of drugs against Onchocerca microfilariae in an inbred mouse
model. Trop. Med. Parasit. 37:39-45.
Blagburn BL, Swango LJ, Hendrix CM, Lindsay DS (1986) Comparative effica-
cies of ivermectin, febantel, fenbendazole and mebendazole against helminth
parasites of gray foxes. 1. Am. Vet. Med. Assoc. 189:1084-1085.
Blair LS, Campbell WC (1980) Suppression of maturation of Dirofilaria immitis in
Mustela putorius by a single dose of ivermectin. 1. Parasit. 66:691-692.
Blair LS, Williams E, Ewanciw DV (1982) Efficacy of ivermectin against third
stage Dirofilaria immitis larvae in ferrets and dogs. Res. Vet. Sci. 33:386-387.
Bowie, JY, Mackintosh CG, Mason PC (1987) Effect of ivermectin and oxfen-
dazole on shedding of larvae of the lungworm Dictyocaulus viviparus by Deer.
New Zeal. Vet. 1. 35:8-10.
Bowman TA, Griffith JW (1987) Comparison of treatments for Psorergates mites
in stumptailed macaques. Lab. Ani. Sci. 37;100-102.
Boyce W, Kollias G, Courtney CH, Allen J, Chalmers E (1984) Efficacy of
ivermectin against gastrointestinal nematodes in dromedary camels. 1. Am.
Vet. Med. Assoc. 185:1307-1308.
Brack M, Rietschel W (1986) Ivermectin for the control of Strongyloides
fuelleborni in rhesus monkeys. Kleintierpraxis 31:29.
Brownell JR (1984) Sternostoma tracheolum-continuing studies. Proc. 33rd
Western Poultry Disease Con!, Davis, CA, pp 91-93.
Calc ott PH, Fatig RO (1984) Avermectin modulation of gamma-aminobutyric acid
binding to membranes of rat brain, brine shrimp and a fungus. J. Antibiot.
37:797-801.
Campbell WC (1982) Efficacy of the avermectins against filarial parasites. Vet.
Res. Commun. 5:251-262.
Campbell WC, Blair LS, Lotti VJ (1979) Efficacy of avermectins against Trichi-
nella spiralis in mice. 1. Helminthol. 53:254-256.
Carrillo M, Barriga 0 (1987) Anthelmintic effect oflevamisole hydrochloride and
ivermectin on tissue toxocariasis of mice. Am. 1. Vet. Res. 48:281-283.
Chiu SHL, Sestokas E, Taub R, Buhs RP, Green M, Sestokas R, VandenHeuval
WJA, Arison BH, Jacobs TA (1986) Metabolic disposition of ivermectin in
tissues of cattle, sheep and rats. Drug Metab. & Dispos. 14:590-600.
Coles BH (1987) Ivermectin for birds. Vet. Rec. 120:604.
Curtis SK, Housley R, Brooks DL (1988) The use of ivermectin in rabbits for the
treatment of ear mites. Proc. 125th Mtg. AVMA, Portland, Oregon, Abstr. 113.
Denham DA (1982) The effects of some avermectins on the growth of Brugia
pahangi. Meth. & Find. Exper. CUn. Pharmacol. 4:347-350.
Devaney E, Howells RE (1984) The Microfilaricidal activity ofivermectin in vitro
and in vivo. Trop. Med. Parasit. 35:47-49.
282 M.D. SoIl
Wilkins CA, Conroy JA, Ho P, O'Shanny WJ, Malatesta PF, Egerton JR (1980)
Treatment of psoroptic mange with avermectins. Am. J. Vet. Res. 41:2112-
2113.
Williams M, Yarbrough GG (1979) Enhancement of in vivo binding of diazepam
by a novel anthelmintic agent, avermectin B)a. Eur. J. Pharmacol. 56:273-276.
Wing SR, Courtney CH, Young MD (1985) Effect ofivermectin on murine mites.
J. Am. Vet. Med. Assoc. 187:1191-1192.
Wright FC, Riner JC (1985) Comparative efficacy of injection routes and doses of
ivermectin against Psoroptes in rabbits. Am. J. Vet. Res. 46:752-754.
Zahner H, Sanger I, Lammler G, Muller HA (1987) Effect of ivermectin on
Dipetalonema uiteae and Litomosoides carinii infections of Mastomys natalen-
sis. Trop. Med. Parasit. 38:117-122.
Zeman P (1987) Systemic efficacy of ivermectin against Dermanyssus gallinae in
fowls. Vet. Pathol. 23:141-146.
CHAPTER 20
I. Introduction
II. Products
A. AVID®, AGRI-MEK®, VERTIMEC®
B. AFFIRM®
III. Applications
A. Red Imported Fire Ant (Solenopsis invicta)
B. Citrus
1. Citrus Rust Mite (Phyliocoptruta oleivora)
2. Broad Mite (Polyphagotarsonemus latus)
3. Citrus Red Mite (Panonychus dtn)
4. Citrus Thrips (Sdrtothrips dtn)
5. Beneficial Organisms
C. Ornamentals
1. Twospotted Spider Mite (Tetranychus urticae)
2. Leafminer (Liriomyza trifoliz)
3. Thrips Species
D. Cotton
1. Spider Mites (Tetranychus urticae, T. turkestani, T. pacijicus,
T. dnnabarinus)
2. Lepidoptera
3. Cotton Aphid (Aphis gossypil)
E. Vegetables (celery, tomato, pepper)
1. Leafminers (Liriomyza trifolii; L. sativae)
2. Twospotted Spider Mite and Tomato Russet Mite (T. urticae
and Aculops lycopersicz)
3. Armyworms (Spodoptera exigua and S. eridania)
4. Tomato Pinworm (Keiferia lycopersicelia)
5. Beneficial Arthropods
F. Pears
1. Pear Psylla (Psylia pyricola)
2. Twospotted Spider Mite, European Red Mite, Pear Rust Mite
(T. urticae, Panonychus ulmi, Epitrimerus pyn)
G. Miscellaneous Crops
1. Strawberry
2. Deciduous Tree Nuts
288 Richard A. Dybas
I. Introduction
Initial indications that the avermectins possessed activity against arthro-
pods were obtained in studies with avermectin B'a against the confused
flour beetle (Tribolium confusum) and the sheep blowfly larva (Lucilia
cuprina). Evaluation of the 8 major natural avermectins and several
hundred semisynthetic avermectin analogs and derivatives showed that
avermectin B" the major component of the fermentation of Streptomyces
avermitilis, was extremely toxic to acarines and to a wide range of
insects, induding members of the orders Diptera, Homoptera, Coleop-
tera, and Lepidoptera (Putter et al. 1981). Avermectin B, (MK-936), with
the common name abamectin, was selected for use against arthropod
pests of economic importance in horticulture and agriculture.
II. Products
A. AVID®, AGRI-MEK®, VERTIMEC®
Abamectin for use in foliar spray applications is formulated as a 1.8% w/v
emulsifiable concentrate containing 0.15 lb abamectin per U.S. gallon.
The oral LD50 of this product in rats is 650 mg/kg body weight, while the
dermal LD50 in rabbits is greater than 2000 mg/kg body weight. Rabbits
exhibit slight-to-moderate eye irritation and very slight-to-slight skin
irritation. Skin sensitization was not observed in guinea pigs with the
emulsifiable concentrate formulation.
The product is being developed for use on a number of horticultural and
agronomic crops to control phytophagous mites and insects. It is recom-
mended at rates in the range of 0.005 to 0.0251b abamectin/acre (5.4 to 27
grams ai/hectare), in volumes of water ranging from 5 to 1000 gallons per
acre, depending on the crop to be treated and application equipment. For
certain crop applications, e.g., for control of mites and insects on citrus,
pear, and deciduous tree nuts, the product is recommended in combina-
tion with a 0.25% narrow-range paraffinic oil in dilute sprays or a
minimum of 1 gallon oil per acre (9.5 liters/ha) in concentrate sprays.
The maximum recommended application rate for citrus, pears, and
deciduous tree nuts is 0.025 lb active ingredient (ai)/acre (20 fl oz
AGRI-MEK®/acre). The maximum recommended rate for ornamental
plants, cotton, vegetables, and strawberries is 0.02 lb ai/acre (16 fl oz
AGRIMEK®/acre).
A maximum of 0.075 lb ai/acre per growing season will be allowed on
citrus, pears, and tree nuts, and 0.20 lb ai/acre on vegetable crops.
With the exception of mild symptoms of phytotoxicity to certain ferns,
and slight spotting of carnation foliage, crops tolerate the product well
when the recommended application directions are followed. This applies
to abamectin sprayed in water or, where recommended, in combination
with paraffinic spray oils (Green et al. 1985a).
20. Abamectin Use in Crop Protection 289
B. AFFIRM®
Abamectin for use in controlling the red imported fire ant (Soienopsis
invicla) is formulated as a bait containing 0.011% w/w abamectin in
soybean oil on pregelled defatted corn grits. The oral LDso in rats and
dermal LDso in rabbits is greater than 2000 mg/kg, and the product is
nonirritating to rabbit skin.
AFFIRM® is intended for use at a rate of 1.0 Ib of bait per acre for
broadcast application through ground or aerial equipment, or 5 to 7
tablespoons of bait for treatment of individual fire ant mounds. The
product is recommended for nonagricultural lands such as lawns, turf,
golf courses, and parks. The U.S. EPA-approved label restricts bait
application to land other than pastures, range lands and crop land.
III. Applications
The red imported fire ant is an important pest in the southern United
States. The worker ants inflict painful stings to humans, they feed directly
on crops, and the ant mounds cause damage to farm machinery. Any
successful control strategy requires eliminating the worker ants and/or
the physogastric queen, who is responsible for the reproductive capacity
of the colony.
Studies conducted in the USDA's Gainesville, FL, laboratory demon-
strated that abamectin exhibits a rate-related delayed toxicity to red
imported fire ant (RIFA) workers. This delayed effect is an important
criterion for efficacy of a bait toxicant against RIFA, since the worker
ants must gather the bait toxicant and distribute it throughout the colony
before the effects of treatment are observed.
In laboratory studies, abamectin at a concentration of 0.1 % killed more
than 75% of RIFA workers after 10 days exposure. Delayed toxicity to
worker ants was observed; less than 10% kill occurred 1 day after
treatment at that concentration (Lofgren and Williams 1982; Williams
1983). When entire colonies were treated in the laboratory, 0.0025%
abamectin in soybean oil caused loss of all brood production and sterility
of the RIFA queen. Abamectin effects at this concentration appeared
irreversible. Lower abamectin concentrations-for example, 0.00025% in
soybean oil-caused a reduction in brood production; however, the
effects were transient, with a return to normal egg production in 16
weeks.
Histological examination of the ovaries of sterile queens showed
several tissue and cell abnormalities. The damage was characterized by
hypertrophy of the squamous epithelium that sheaths the ovarioles, and
pycnosis of the nurse-cell nuclei. According to the authors, the lack of
eggs containing yolk, the very small number of eggs produced, and the
290 Richard A. Dybas
small size of the eggs all point to an effect on the neurosecretory cells
(Glancey, Lofgren, and Williams 1982).
Abamectin applied at lib of bait per acre (50 mg ai/acre) resulted in
virtually complete elimination in worker brood (> 95%) and a significant
reduction in colony ant populations (> 85%) at 6 to 12 weeks post
treatment. Consistent with the irreversible effects on RIFA queens noted
in the laboratory, the USDA researchers found no worker brood in
treated colonies 18 weeks after application (Lofgren and Williams 1982;
Williams 1985).
Treating individual fire ant mounds with abamectin bait provided high
colony mortality, in the range of 55% to 82%, 12 to 21 weeks after
treatment with 1,4, and 7 tablespoons bait per mound. The 7-tablespoon
rate provided the fastest kill, although by 21 weeks there was no
difference in colony mortality between the 4- and 7-tablespoon rates. The
7-tablespoon rate of AFFIRM® is recommended for use by homeowners,
since more rapid kill of worker ants and elimination of fire ant colonies is
expected in this market (Greenblatt et al. 1986). Apperson, Powell, and
Browne (1985) and Lemke and Kissam (1987) also reported that AF-
FIRM® provided greater than 80% reduction in colonies with brood and
colony mortality following treatment of individual mounds.
Preliminary evidence from field trials suggests that fire ants do not
rapidly recolonize abamectin-treated areas. The gradule elimination of
worker populations and the presence of live sterile queens in treated
colonies may prevent adoption and establishment of newly mated phy-
sogastric queens in abamectin plots (Greenblatt et al. 1986). Therefore,
the slow action of abamectin appears to offer the advantage of providing
high mortality to treated colonies while delaying or preventing reinfes-
tation of treated areas.
B. CITRUS
TABLE 20.1 Activity of abamectin against insect larvae and adult mites and
aphids.
Mite Species (Contact effect against adult mites) LC90 (ppm)A
Phyllocoptruta oleivora (Ashmead) (citrus rust mite) 0.02
Tetranychus urticae Koch (twospotted spider mite) 0.03
Panonychus ulmi (Koch) (European red mite) 0.04
Polyphagotarsonemus latus (Banks) (broad mite) 0.05
Tetranychus turkestani (Ugarov & Nikolski) (strawberry spider mite) 0.08
Tetranychus pacificus (McGregor) (Pacific spider mite) 0.16
Panonychus citri (McGregor) (citrus red mite) 0.24
Insect Species (Foliar residue bioassay)
Manduca sexta (L.) (tobacco hornworm) 0.02
Leptinotarsa decemlineata (Say) (Colorado potato beetle) 0.03
Heliothis virescens (F.) (tobacco budworm) 0.10
Epilachna varivestis Mulsant (Mexican bean beetle) 0.40
Trichiijjlusia ni (Huebner) (cabbage looper) 1.0
Heliothis zea (Boddie) (cotton bollworm) 1.5
Spodoptera eridania (Cramer) (southern armyworm) 6.0
Spodoptera jrugiperda (J .E. Smith) (fall armyworm) 25.0
A Mortality assessed 72-96 hours after exposure. Dybas, R.A., Green, A. St. J. (1984)
Avermectins: Their Chemistry and Pesticidal Activity. Proceedings, 1984 British Crop
Protection Conference-Pests and Diseases, Brighton, England 31:947-954.
20. Abamectin Use in Crop Protection 293
5. Beneficial Organisms
Abamectin exhibits a relatively low order of intrinsic toxicity to the
predatory mite Euseius tularensis (LC90 9.5 ppm) compared to its activity
on citrus red mite (LC90 0.30 ppm) in slide-dip bioassays. These data
(Brown, unpublished) provide a safety factor of 31.7 based on the LC90
ratio for predator to prey and a safety factor of 54.6 at the LC50 level. In a
field trial by Brown (unpublished), populations of the predatory mite
E. tularensis rebounded rapidly following an application of abamectin at
0.025 lb ai/acre plus 1% crop oil. The numbers of predatory mites had
declined at 3 days following application but were reestablished to
control-plot levels by 6 days.
Morse and colleagues (1987) reported that field-aged foliar residues of
abamectin were relatively innocuous to selected beneficial arthropods of
citrus. One-day-old residues of abamectin, applied to lemon foliage at
0.0251b ai/acre (the highest rate recommended on the product's label) in
combination with 0.5% narrow-range oil, showed less than 10% mortality
294 Richard A. Dybas
c. ORNAMENTALS
3. Thrips Species
D. COTTON
been observed in all trials. Results of limited studies have also shown
that, under certain conditions, the addition of the nonionic surfactant,
Leaf Act 80A, may improve leafminer control, presumably by enhancing
penetration of abamectin into the foliage and thereby extending the period
of residual protection (Merck data, unpublished).
2. Twospotted Spider Mite and Tomato Russet Mite
(T. urticae and Aculops lycopersici)
Laboratory studies have shown that abamectin is highly toxic to erio-
phyid and tetranychid mites (Dybas and Green 1984; McCoy, Bullock,
and Dybas 1982). The LC 90 of abamectin against T. urticae adults was
0.03 ppm. Royalty and Perring (1987) found that abamectin was very toxic
to A. lycopersici, with an LC 90 of 0.00959 ppm against adults. These
authors also noted that abamectin was much less toxic to the predatory
mite Homeopronematus anconai; thus, abamectin sprays should suffi-
ciently control tomato russet mite without reducing H. anconai popula-
tions.
Limited field studies have been conducted to date with abamectin for
control of mites on tomato with excellent results. Greater than 80%
control of mite populations was observed following abamectin applica-
tions at 0.01 to 0.021b ai/acre (Schuster 1984; Merck data, unpublished).
5. Beneficial Arthropods
Trumble (1985a) reported that weekly treatments of celery with abamec-
tin at 0.01 lb ai/acre (its recommended rate of application for leafminer
control) had a minimal effect on both adult and larval stages of the
leafminer parasite complex. Abamectin applications did not adversely
impact seasonal percent parasitism, adult parasite mortality and survival,
or emergence of immature parasites of Diglyphus intermedius, D. begini,
Chrysonotomyia punctiventris, Chrysocharis parksi, C. ainsliei, and
Halticoptera circulus from treated foliage.
Abamectin at application rates of 0.01 to 0.02 lb ai/acre also did not
adversely affect the beneficial leafminer parasite complex on celery,
tomato, and pepper under field use conditions (Zehnder and Trumble
1984; Carson, Nakakihara, and Trumble 1986; Trumble 1985b; Chandler
1985). Parasite mortality from abamectin treatments were not statistically
different from the controls.
Results of several field trials support the fact that abamectin has a good
safety margin for beneficials and is useful for Integrated Pest Management
programs on celery, tomato, and peppers.
F. Pears
1. Pear psylla (Psylla pyricola)
Abamectin, at rates of 0.0125 to 0.025 lb ai/acre, has provided significant
reduction in Psylla pyricola nymphal populations in postbloom sprays on
pears. The addition of 0.25% paraffinic oil in dilute sprays, or a minimum
of 1 gallon oil per acre, extends abamectin's residual control of psylla
nymphs for 4 weeks or longer, and provides effective control of fruit
damage.
Burts (1983, 1984) reported that abamectin provided good control of
304 Richard A. Dybas
G. Miscellaneous Crops
1. Strawberry
Limited studies have shown that abamectin is effective against spider
mite infestations on strawberry and tree nuts. Carson and colleagues
(1987) reported that 7 biweekly applications of abamectin on strawberry
at 0.02 lb ai/acre in 50 to 100 gallons of water per acre were effective in
reducing T. urticae populations throughout the trial. Abamectin's activity
was not improved by the addition of 0.1 % oil in the spray tank. The
authors also reported that abamectin controlled a moderate density of
aphids under this spray regime.
Earlier, Trumble and Nakakihara (1984) had reported that abamectin
applied to strawberries at 0.04 lb ai/acre, alone and in combination with
0.1 % oil, reduced motile spider mite populations by an average of 92.7%
and 95.8%, respectively, over an 8-week period following a single
application. A second application of abamectin at the same rates, alone
and in combination with paraffinic oil, reduced mite levels but was less
effective, providing only 30.9% and 72.8% control over a 3-week period.
More recent studies have shown that abamectin controls spider mites
very effectively on strawberry following 2 applications, timed on a 7-day
spray schedule to kill hatching larvae (Dunbar, unpublished). In 3
replicated field trials, abamectin provided an average of 79% and 68%
control of T. vrticae over a 35-day period when applied twice at 0.02 and
O.Ollb ai/acre, compared to only 52% control of mite populations over the
same period from a single application at 0.01 lb ai/acre.
2. Deciduous Tree Nuts
Abamectin applied at 0.00625 lb ai/lOO gallons water in a high volume
dilute spray to almonds was shown to reduce initial popUlations of
Tetranychus pacificus by 85% at 6 days post treatment. Residual control
of spider mite populations was obtained for 48 days and was enhanced by
the combination of abamectin with a narrow-range spray oil (Sanderson
and Barnes 1983). The authors also reported that abamectin treatment did
not have a negative impact on beneficial species, including the predatory
mite, Metaseiulus occidentalis, and six-spotted thrips, Scolothrips sex-
maculatus. Payne, Dutcher, and Ellis (1985) reported that abamectin
applied at a rate of 0.02 lb ai/acre to pecans provided excellent control
(95%-100%) of the hickory shuckworm, pecan leaf scorch mite, and
pecan weevil. Abamectin, however, provided poor control of yellow
aphids and the black aphid in this field trial.
REFERENCES
Anderson TE, Babu JR, Dybas RA, Mehta H (1986) Avermectin Bl: ingestion and
contact toxicity against Spodoptera eridania and Heliothis virescens (Lepidop-
306 Richard A. Dybas
Howitt AJ, Hays AR (1986) Pear, full season psylla block test. Insectic. & Acaric.
Tests 11:71-72.
Hoy MA, Cave FE (1985) Laboratory evaluation of avermectin as a selective
acaricide for use with Metaseiulus occidentalis (Nesbitt) Acarina: Phyto-
seiidae). Exper. & Appl. Acarol. 1:139-152.
Iwata I, MacConnell JG, Flor JE, Putter I, Dinoff TM (1985) Residues of
avermectin Bla on and in citrus fruits and foliage. 1. Agric. Food Chem.
33:467-471.
Leigh T, Wynholds P, Bentley W, Arrendondo C (1988) Insect pest status in the
San Joaquin Valley in 1987. Cal. Cot. Prog. Rep. March: 8-9.
Lemke LA, Kissam JB (1987) Evaluation of various insecticides and home
remedies for control of individual red imported fire ant colonies. 1. Entomol.
Sci. 22:275-281.
Lofgren CS, Williams CF (1982) Avermectin B1a: highly potent inhibitor of
reproduction by queens of the red imported fire ant (Hymenoptera: Formici-
dae). 1. Econ. Entomol. 75:798-803.
MacConnell JG, Demchak RJ, Preiser FA, Dybas RA (in press) A study of the
relative stability, toxicity, and penetrability of abamectin and its 8,9-oxide. 1.
Agric. Food Chem.
McCoy, CW, Bullock RC, Dybas RA (1982) Avermectin B1: a novel miticide
active against citrus mites. Proc. Fla. State Hort. Soc. 95:51-56.
Mink JS, Luttrell RG (1987) Chemical control of the fall armyworm on cotton.
Insectic. & Acaric. Tests 12:236-237.
Mizell RF, Schiffhauer DE, Taylor JL (1986) Mortality of Tetranychus urticae
Koch (Acari: Tetranychidae) from abamectin residues: effects of host plant,
light, and surfactants. 1. Entomol. Sci. 21:329-337.
Moar WJ, Trumble JT (1987) Toxicity, joint action, and mean time of mortality of
dipel 2X, avermectin BJ, neem and thuringiensin against beet armyworms
(Lepidptera: Noctuidae). 1. Econ. Entomol. 80:588-592.
Mollet JA, Nelson RD, Norton JA, Dybas RA (1986) Abamectin for control of
spider mites on greenhouse carnations and roses Entomological Society of
America National Conference, Reno, NV Dec. 7-11.
Morse JG, Bellows TS, Gaston LK, Iwata Y (1987) Residual toxicity of
acaricides to three beneficial species on California citrus. 1. Econ. Entomol.
80:953-960.
Morse JG, Brawner OL (1983) Citrus thrips pesticide efficacy tests in California.
Insectic. & Acaric Tests 8:37-39.
Morse JG, Brawner OL (1986) Toxicity of pesticides to Scirtothrips citri (Thysa-
noptera: Thripidae) and implications to resistance management. 1. Econ.
Entomol. 79:565-570.
Morse JG and Brawner OL (1987) Citrus thrips control trial in California. Insectic.
& Acaric Tests 12:91-92.
Morse JG, Elliott WG (1985) Citrus red mite pesticide efficacy tests in Riverside.
Insectic. & Acaric. Tests 10:55-56.
Morse JG, Elmer HS, Brawner OL (1984) Evaluation of unregistered materials for
control of citrus thrips in California. Insectic. & Acaric Tests 9:91-92.
Morse JG, Grout TG, Brawner OL (1986) Citrus thrips control trials in California.
Insectic. & Acaric Tests 11:101-102.
Oetting RD (1987) Laboratory evaluation of the toxicity and residual activity of
abamectin to Ecinothrips americanus. 1. Agric. Entomol. 4:321-326.
20. Abamectin Use in Crop Protection 309
Tuttle DM, Mullis CH (1984) Control of carmine spider mite on cotton. Insectic &
Acaric Tests 9:278.
Waddill V (1982) Evaluation of insecticides for control of insect pests of tomato.
Insectic & Acaric Tests 7: 126.
Williams DF (1983) The development of toxic baits for the control of the red
imported fire ant. Fla. Entomol. 66:162-171.
Williams DF (1985) Laboratory and field evaluation of avermectin against the
imported fire ant. Southwest Entomol. (Suppl.) No. 7:27-33.
Wright DJ, Loy A, Green A St J, Dybas RA (l985a) The translaminar activity
of abamectin (MK-936) against mites and aphids. Med. Fac. Landbouww.
Rijksuniv. Gent 50I2b:595-601.
Wright DJ, Roberts ITJ, Androher A, Green A St J, Dybas RA (1985b) The
residual activity of abamectin (MK-936) against Tetranychus urticae (Koch) on
cotton. Med. Fac. Landbouww. Rijksuniv. Gent 50I2b:633-637.
Wright JE, Jenkins IN, Villavaso, EJ (1985) Evaluation of avermectin Bl
(MK-936) against Heliothis spp. in the laboratory and in field plots and against
the boll weevil in field plots. Southwest. Entomol. (Suppl.) No. 7:11-16.
Wynholds PF, Leigh TF (1982) Control of spider mites on cotton. Insectic &
Acaric Tests 7:165.
Wynholds PF, Leigh TF (1986) Control of spider mites on cotton. Insectic &
Acaric Tests 11:307-308.
Zehnder GW, Trumble JT (1984) Insect control on celery. Insectic. & Acaric.
Tests 9:89-90.
CHAPTER 21
I. Introduction
II. Treatment of Onchocerciasis
A. Phase I Studies
B. Phase II Studies
C. Phase III Studies
D. Phase IV Studies
E. Ivermectin Effect on the Parasite O. volvulus
F. Ivermectin Effect on Transmission and as a Prophylactic Agent
G. Conclusion and Present Clinical Usage
III. I vermectin for Uses in Filarial Infections other than Onchocerciasis
A. Ivermectin in the Treatment of Filariasis Caused by Wuchereria
bancrofti
B. Ivermectin Use in Patients Infected with More than One Filarial
Species
C. Ivermectin Treatment for Human Nematodes
I. Introduction
Ivermectin (Campbell et al. 1983) has been extensively tested in human
onchocerciasis, and is now considered the drug of choice. In a single
yearly dose, it suppresses microfilariae in the skin and eyes, and in most
infected persons prevents disease progression. It also shows considerable
promise for treatment of lymphatic filariasis and strongyloidiasis.
B. PHASE II STUDIES
The Phase I clinical data made it obvious that ivermectin was a very
promising drug (Editorial 1984). This led to a series of Phase II studies,
which were conducted as double-blind, controlled clinical trials. They
focused particularly on assessing invermectin's safety and efficacy as
compared to the standard treatment for onchocerciasis, diethylcarbam-
azine (DEC); the control group received a placebo. The 4 studies used
essentially the same rigorous protocol: subjects were hospitalized, treated
with single-dose ivermectin (200 ltg/kg) or a I-week course of DEC, and
examined every day-some of the observations being done every 4 to 6
hours. Patients also had detailed eye exams, including fluorescein angio-
graphy. Each study had 10 to 15 subjects per group. Phase II studies were
conducted in Liberia, Senegal, Ghana, and Mali.
The trial in Liberia (Greene et al. 1985) showed that a single dose of
ivermectin was at least as effective in reducing the microfilaria counts as
DEC given daily for 8 days; also, ivermectin caused very little reaction as
compared to DEC. Subsequent results showed that the marked reduction
of skin microfilaria counts seen following ivermectin treatment persisted
to 12 months (Taylor et al. 1986). These results were confirmed in the
other 3 Phase II studies (Lariviere et al. 1985; Diallo et al. 1986; Awadzi
et al. 1986), Ivermectin was clearly microfilaricidal, but examination of
excised nodules showed that it did not kill the adult worms (Greene et ai,
1985), It did, however, produce a transient interruption of microfilarial
release (Schulz-Key et al. 1985).
Ivermectin treatment produced either no or a very mild clinical reaction
21. Use of Ivermectin in Humans 313
Phase III trials assessed the efficacy of 3 different dosages of the drug
(100, 150, and 200 ILg/kg) and collected further information on safety.
These trials were all double-blind, and, again, patients were followed for 1
year. Each group had at least 50 patients; all Phase III participants were
hospitalized for at least 2 days after treatment. At this point, the Liberian
study is the only one of the 6 published. Other studies following similar
protocols were undertaken in Ghana, Mali, Cote d'Ivore, Togo, and
Guatemala.
The 12-month follow-up data ofthe Liberian study show that, by Day 3,
each of the 3 doses of ivermectin produced quite a marked drop in skin
microfilaria counts (White et al. 1987). This decrease was more marked at
Day 3 for the 200 ILg/kg group than for the 100 ILg/kg dose, suggesting a
dose response at this early time point. At 3 months, however, the counts
for the 3 different doses were indistinguishable; they were still indistin-
guishable at 6 months and at 12 months.
In the Phase III trials, the clinical reaction score was assessed as it had
been in the Phase II studies. In the Liberian study, each of the 3 treatment
groups showed an increase in the clinical reaction score over the first few
days following treatment (White et al. 1987). Because there were now 50
people in each group (as opposed to only 10 in each group in the Phase II
314 B.M. Greene, K.R. Brown, and H.R. Taylor
study), this increase was statistically significant for both 200 and
150 ILg/kg compared to the placebo group. The reaction score for the
100 ILg/kg group was not significantly different from the placebo group.
The reaction was relatively mild in each case (except for 1 patient out of
150 who missed 1 day of work) and had essentially resolved by Day 4.
Again, no serious ocular lesions were precipitated by treatment, and the
ocular status was markedly improved 1 year after treatment (White et al.
1987). Similar results from the other Phase III trials have been reported
informally (Lariviere et al. 1986; Mossinger et al. 1987; Hussein, Bird,
and Jones 1987; Ranque et al. 1986).
The Phase III studies reconfirmed that ivermectin was both safe and
effective and induced only a minimal Mazzotti reaction without a
significant ocular reaction. These studies suggested that 150 ILg/kg was
probably the optimal dose, in terms of its antiparasitic activity and lack of
side effects.
At the end of the I-year follow-up in the Liberian Phase III study,
participants were reallocated for a dose-interval study. At 12 months, the
patients in the 3 treatment groups (100, 150, or 200 ILg/kg of ivermectin)
were rerandomized so that, in each treatment group, half the persons
were retreated with the same dose of ivermectin they had received
initially, and the other half were given placebo (Greene et al. 1987). The
original placebo group received 150 ILg/kg on a 6-monthly basis. At 24
months, all patients were again given ivermectin; the dose being the same
as they had received initially. This study provided information on 3
different retreatment schedules: 6-monthly treatments with 150 ILg/kg
with a I-year follow-up; annual treatment with 100, 150, or 200 ILg/kg with
a 2-year follow-up; and 2-yearly treatment with the 3 doses, also with a
2-year follow-up.
The 2-year follow-up data suggest that, even 24 months after initial
treatment, there is still a marked effect on skin microfilaria counts at each
treatment dose (Figure 21.1). Yearly retreatment with 150 ILg/kg,
however, appears to be an optimal regime for reducing microfilarial
counts (Figure 21.2) and for safety. Importantly, the clinical reaction seen
after retreatment at 12 months was less than that seen after the initial
treatment.
The ocular examination data at 24 months for the 3 different treatment
groups are shown in Figure 21.3. There was a marked improvement in the
ocular status of each group after treatment (Newland et al. 1988). Even
without annual retreatment, the ocular status showed marked improve-
ment over conditions prior to the initial treatment, 24 months earlier.
Ivermectin was also found to be safe and effective in patients with severe
ocular involvement (Taylor et al. unpublished). Interestingly, the im-
provement in ocular disease appeared to be even more prolonged than the
effect on the skin snip counts.
21. Use of Ivermectin in Humans 315
0--0 1OO1"J/1<9
.......
........
I5OI"JIKt
2OOI"J/KQ
Placebo at 12 months
20
II
l
-
2
c
II
/t,
I 12
~
J 8 / ........._- j
/" /
" t-
"" .. '
---;::.-<";>(,
--~i
1·1
'1
,::.--- I
V' ,/ _ '
4 I
I 12 18 24
t t
Time (Months)
D. PHASE IV STUDIES
c: 20
~
(J)
co
- "\
E
....... 16
~
c:
0
\
,'\,"\ .
cu
E
-
u 12
.~
cu
E " \
0
cu \\
,\
"
(!) 8 \
\\
\,
,\,
4
\\.11 . .-:::."':'..::-.,:_ .......-'1
\. \ { II
r.-· ------ I
o~--------~----------~-----------
6 12
Time (Months)
FIGURE 21.2. Comparison of the effect on skin microfilaria density load 6-monthly
and 12-monthly treatment with ivermectin 150 I-'g/kg.
Ivermectin's effect on the adult worm was examined in detail in both the
Phase II and Phase III trials. Ivermectin did not affect the viability of the
adult worm-male or female. In some studies, there appeared to be
slightly fewer adult males in the ivermectin nodules, but this was not
statistically significant (Greene et al. 1985; Schulz-Key et al. 1985). There
21. Use of Ivermectin in Humans 317
2 I: 95% C.l.
O~---L----~---------~---------~---------~
4 3 6 12 18 24
3 150 meg/kg
4
200 meg/kg
3
2
T
o 3 6 12 18
TIME (MONTHS)
FIGURE 21.3. Ocular reaction index results over 24 months. The broken line after
12 months shows the response of patients given only a single dose of ivermectin.
The solid line shows the response of patients retreated at 12 months.
ment. In contrast, DEC kills the microfilariae very effectively but has no
residual effect on adult worms, which continue to produce microfilariae
normally (Albiez et al. 1988) The rate of reappearance of microfilariae in
the skin after DEC is, therefore, different from that after ivermectin.
Pretreatment levels are usually reached 1 to 2 years after DEC (Duke
1968), whereas after treatment with ivermectin, the microfilaria counts in
skin snips are still low at 1 or even 2 years.
In October 1987, just over 5 years after the first report on ivermectin's use
in onchocerciasis was published, the French government approved the
drug's use in humans (Lindley 1987). Although not a macrofilaricide-and
therefore not a permanent cure for onchocerciasis-ivermectin given as a
single oral dose of 150 #Lg/kg annually shows promise of revolutionizing
the treatment of this major disease of humankind, which previously was
essentially untreatable.
Ivermectin is now considered the drug of choice for onchocerciasis. It
should not be taken during pregnancy nor by lactating females within 3
months of delivery. It induces a minimal reaction (Mazzotti-like) in 5% to
15% of infected adults and a more significant reaction in about 1%. Its
safety is unproven in children under the age of 5 and in persons with
meningitis, sleeping sickness, and other CNS disorders. Its use was
21. Use ofIvermectin in Humans 319
Nalin and colleagues (1987) reported efficacy results from studies of 294
infected patients treated with ivermectin. Mean percent cures were:
S. stercoralis, 82%; A. lumbricoides, 100%; E. vermicularis, 84%; and
T. trichiuris, 69%.
Naquira and colleagues (unpublished data on file at Merck Sharp &
Dohme Research Laboratories) have studied 100 patients ranging in age
from 11 to 74 who were infected with Strongyloides stercora lis . Diagnosis
was based on the presence of S. stercora lis larvae in the stool; ivermectin
was given either as a single oral dose (50, 100, 150, or 200 JLg/kg) or as a
single dose of 100 or 200 JLg/kg on each of 2 successive days. Follow-up
stool examinations were performed at least once between days 17 to 24
and between days 30 to 38 after dosing.
Patients who received a total of 50 ro 100 JLg/kg ivermectin had a
response rate which ranged from 69% to 75%, whereas the patients who
received a total of 150 JLg/kg or more had a cure rate of 88% to 100%.
Based on this study, ivermectin in a dose of 150 JLg/kg would appear to be
highly effective against S. stercoralis. Even for patients who were not
cured, the change in larval counts in the stool was considered to be very
beneficial.
Studies of other parasites in these patients and 131 additional persons
without strongyloides were performed; cure rates obtained for human
hookworms (either Necator or Ancylostoma) were unsatisfactory even
though hookworm egg counts in stools were reduced. Even at
200 JLg/kg/day, the 30-day cure rate for patients with hookworm was only
22%; at 30 days patients who were not cured had only a 42% drop in egg
counts in the stool. The mean cure rate in all patients at 30 days
(regardless of dose) was 85% for Trichuris trichiura, 100% for Ascaris
lumbricoides, and 85% for Enterobius vermicularis.
The preceding data demonstrate ivermectin's high degree of efficacy
and suggest its possible use as a replacement for thiabendazole in treating
Strongyloides stercoralis. Ivermectin could not be recommended as a
21. Use of Ivermectin in Humans 321
REFERENCES
Albiez EJ, Newland HS, White AT, Kaiser A, Greene BM, Taylor HR, Buttner
DW (1988) Chemotherapy of onchocerciasis with high doses of diethylcarbama-
zine or a single dose of ivermectin: microfilaria levels and side effects. Trap.
Med. Parasit. 39: 19-24.
Awadzi K, Dadzie KY, Schulz-Key H, Gilles HM, Fulford AJ, Azia MA (1986)
The chemotherapy of onchocerciasis XI. A double-blind comparative study of
ivermectin, diethylcarbamazine, and placebo in human onchocerciasis in
Northern Ghana. Ann. Trap. Med. Parasit. 80:433-442.
Awadzi K, Kadzie KY, Schulz-Key H, Haddock DRW, Gilles HM, Aziz MA
(1984) Ivermectin in onchocerciasis (letter). Lancet 2:2921.
Awadzi K, Dadzie KY, Schulz-Key H, Haddock DRW, Gilles HM, Aziz MA
(1985) The chemotherapy of onchocerciasis X. An assessment of four single
dose treatment regimes of MK-933 (ivermectin) in human onchocerciasis. Ann.
Trap. Med. Parasit. 79:63-78.
Aziz MA, Diallo S, Diop 1M, Lariviere M, Porta M (l982a) Efficacy and tolerance
of ivermectin in human onchocerciasis. Lancet 2:171-173.
Aziz MA, Diallo S, Lariviere M, Diop 1M, Porta M, Gaxotte P (l982b) Ivermectin
in onchocerciasis. Lancet 2:1456-1457.
Bird AC, EI-Sheikh H, Anderson J, Fuglsang H (1980) Changes in visual function
and in the posterior segment of the eye during treatment of onchocerciasis with
diethylcarbamazine citrate. Brit. 1. Ophthalmol. 64: 191-200.
Campbell WC, Fisher MH, Stapley EO, Albers-Schonberg G, Jacob TA (1983)
Ivermectin: a potent new antiparasitic agent. Science 221:823-828.
Coulaud JP, Lariviere M, Aziz MA, Gervais MC, Gaxotte P, Deluol AM, Cenac J
(1984) Ivermectin in onchocerciasis. Lancet 2:526-527.
Coulaud JP, Lariviere M, Gervais MC, Gaxotte P, Aziz MA, Deluol AM, Cenac J
(1983) Treatment of human onchocerciasis with ivermectin. Bull. Soc. Pathol.
Exot. Filiales 76:681-688.
Cupp EW, Bernardo MJ, Kiszewski AE, Collins RC, Taylor HR, Aziz MA,
Greene BM (1986) The effects of ivermectin on transmission of Onchocerca
volvulus. Science 231 :740-742.
Dadzie KY, Bird AC, Awadzi K, Schulz-Key H, Gilles HM, Aziz MA (1987)
Ocular findings is a double-blind study of ivermectin versus diethylcarbamazine
versus placebo in the treatment of onchocerciasis. Brit. 1. Ophthalmol.
71:78-85.
Diallo S, Aziz MA, Lariviere M, Diallo JS, Diop-Mar I, N'Dir 0, Badiane S,
Py D, Schulz-Key H, Gaxotte P, Victorius A (1986) A double-blind comparison
of the efficacy and safety of ivermectin and diethylcarbamazine in a placebo
controlled study of Senegalese patients with onchocerciasis. Trans. Roy. Soc.
Trop. Med. Hyg. 80:927-934.
Diallo S, Aziz MA, Ndir 0, Badiane S, Bah IB, Gaye 0. (1987) Dose-ranging
study of ivermectin in treatment of filariasis due to Wuchereria bancrofti.
Lancet 1:1030.
322 B.M. Greene, K.R. Brown, and H.R. Taylor
A. Summary
B. Reagents
C. Solutions
D. Apparatus
E. Chromatographic Apparatus
F. Standards for the Determinative Assay
G. Procedure for the Determinative Assay of Tissues
H. Suitable Stopping Places for Tissue Assays
I. Analytical Procedure for Asay in Plasma
A. Summary
The tissue sample is homogenized with acetone-water, and dihydro B 1a is
extracted from the tissue with isooctane. The isooctane is evaporated,
and solvent-solvent distributions into acetonitrile out of hexane and into
hexane out of acetonitrile-water are performed. Solvent is removed and
fluorescence produced by heating at 95° C with the imidazole-derivatizing
reagent. Mter adding chloroform, the reaction mixture is washed through
a silica gel SEP-PAK, the solvent is removed, and HPLC analysis
(fluorescence detection) on Zorbax CI8 is performed on the residue
dissolved in methanol. A flow diagram of the assay is shown in Figure
AI. I.
Proteins are precipitated from plasma by addition of acetone and then
water. The ivermectin dihydro B la is extracted with ethyl acetate from
the acetone-water-plasma mixture. The solvent is removed and fluo-
rescence produced by heating at 95° C with the derivatizing reagent. Mter
adding chloroform, the reaction mixture is washed through a silica gel
SEP-PAK, the solvent is removed, and the HPLC analysis (fluorescence
detection) on Zorbax CI8 is performed on the residue dissolved in
methanol.
Homogenize 5g t i _ Extract Into evaporate Dissolve In methanol centrtfuge Dissolve In
with -a-Iweter l - - laooctane end cool -20"C evaporate hexane
~ -
,
A
-- --
t
B
0.5 mI ellquot AdcI ..egent end Dilute with chlorofonn eveporate Dissolve In Inject Inlo LC
heet85-100·C r---- end pass through methanol ~ with IIU0f'8scenc:e
eveponte Sep·Pe~
for 1 hour cletec:llon
t
C
FIGURE ALl. Flow diagram of determinative assay. A, Band C are possible stopping points during the assay.
326 G.V. Downing
B. Reagents
1. Acetic anhydride-Fisher or Baker, reagent grade.
2. Acetone-Mallinckrodt, Nanograde.
3. Acetonitrile-Fisher, HPLC grade; Mallinckrodt, Nanograde; or
Burdick & Jackson, distilled in glass.
3. Chloroform-Burdick & Jackson, distilled in glass (1% ethanol
preservative).
5. Dimethyl Formamide (DMF)-Baker or Fisher, reagent grade; or
Mallinckrodt, AR.
6. Ethyl Acetate-Burdick & Jackson, distilled in glass; or Fisher,
pesticide grade.
7. Hexane-Burdick & Jackson, distilled in glass; or Mallinckrodt,
Nanograde.
8. Methanol-Burdick & Jackson, distilled in glass; or Mallinckrodt,
Nanograde.
9. Methylene Chloride-any grade available.
10. 1-Methylimidazole-99%, Aldrich Chemical Company.
11. Nitrogen-the equivalent of Matheson extra-dry compressed gas.
12. Sodium Chloride-Baker or Fisher, reagent grade.
13. Sylon-CT-Supelco, Inc.
14. 2,2,4-Trimethylpentane-isooctane, Burdick & Jackson, distilled in
glass; or Mallinckrodt, Nanograde.
15. Water, Filtered Millipore-Distilled water is treated with a Milli-Q
system and then filtered through a Millipore Type HA 0.451Lm disc.
C. Solutions
1. Derivatizing Reagent-Mix 0.9 ml of DMF with 0.3 ml of acetic
anhydride and 0.2 ml 1-methylimidazole. Make up just before use.
2. Acetone/Water-50% (v/v).
3. 95% Methanol/Water-Make 100 ml of filtered Millipore water to 2
liters with methanol. Mix. With the aid of vacuum, filter mixture
through Millipore FH 0.51Lm filters. Deaerate by slowly bubbling
nitrogen through for 10 minutes.
D. Apparatus
1. Balance-analytical, capable of weighing 1 mg accurately.
2. Balance-capable of weighing 5 g accurately into an approximately
50-g tube.
3. Bath, water-variable temperature 40° to 80° C.
4. Bath, oil-95° to 100° C.
5. Centrifuge, IEC Model HN-S-II, with 6-place rotor IEC #958 and 15-
Appendix I. Assaying Ivermectin Residues in Tissue and Plasma 327
and 50-ml cups. The centrifuge is run at 2000 to 2500 rpm. The
centrifuge used gives ca. 700 to 750 x g maximum centrifugal force
(RCF).
6. Centrifuge tubes-glass, 15 to 50 ml with polyethylene stoppers to fit.
7. Centrifuge tubes-50-ml polyproplyene, Corning#25331 (used only
for storing standard solutions).
8. Centrifuge tubes, 15 ml, silylated approximately once every 2 months
(used only for the derivatization reaction). Tube stoppers must fit
tightly. Fill each tube to the top with Sylon-CT. Let stand 20 minutes.
Immediately and quickly rinse, first thoroughly with toluene and then
with methanol. Fill with methanol. Let stand 20 minutes, rinse
thoroughly with acetone, and dry. All glassware used should be
completely free of all acidic and alkaline residues. (These tubes
should be cleaned by hand by first soaking in methylene chloride
immediately after use and then in detergent for at least several hours,
followed by thorough rinsing with hot water, distilled water, and
acetone before thorough drying. Variations in the washing regimen
are not recommended, since some analysts have had poor results
when the standard washing method was not followed.)
9. Dispensing pipettors-lO, 15, and 20 ml.
10. Filtration unit, complete with Millipore FHLP 04700 filter.
11. Freezer-capable of reaching temperatures of -200 C.
12. Gloves-disposable PVC from Fisher Chemical.
13. Graduate cylinder-25 ml, l00ml.
14. Graduate cylinder-2000 ml with ground-glass stopper.
15. Homogenizer, Polytron-Brinkmann Instruments, #27-11-200-5 with
PTA lOS generator #27-11-330-3.
16. Pipets-disposable, Pasteur.
17. Pipets-graduated 1.0 ml and 2.0 ml.
18. Pipets-volumetric 0.5 ml, 1 ml, 2 ml, 3 ml, 4 ml, and 5 ml.
19. Repipet-5 to 10 ml volumes.
20. SEP-PAK-silica cartridge, Part No. 51900-Waters Associates.
21. Spatula-stainless steel.
22. Syringe-50ILI, 250ILI, and 5 ml and 10 ml.
23. Tape-#13 2956, ~ inch from Ace Scientific.
24. Ultrasonic bath-Sonogen Automatic Cleaner-Branson Model 520
or equivalent.
25. Vortex mixer.
E. Chromatographic Apparatus
A Beckman-Altex Model IIOA liquid chromatographic pump complete
with Waters Wisp Autosampler and Kratos-Schoeffel Instruments Model
FS950 fluorescence detector with a I-millivolt recorder was used. A
328 G. V. Downing
51Lm, 4.6-mm ID, CI8 standard Brownlee Labs guard column (Spheri-5
RP-18 OD-GU, obtained from Rainin Instrument Co., Inc.) was used
before the analytical column. This guard column is replaced monthly
unless the pressure reaches 2000 psi, in which case it is replaced
immediately.
The ivermectin H2Bla derivative has a retention time of about 12 to 13
minutes when the indicated conditions are used.
All Tissues
1. Weigh accurately 5.0 grams of sample into a 50-ml centrifuge tube.
Fat Only
2. Add 15 ml of the acetone/water and 10 ml ofisooctane to the tube and
homogenize until a homogeneous mixture results.
3. Rinse probe with 10 ml isooctane in a clean 50-ml tube.
4. Pour isooctance into the 50-ml tube with the homogenate.
5. Add 1 gram of solid sodium chloride to the tube containing the
homogenate.
Extractions
All Tissues
6. Stopper the tube, shake well for 1 minute, and centrifuge for 10
minutes.
7. Transfer the upper or isooctane layer to a second 50-ml centrifuge
tube with a disposable pipet. Completely avoid the lower layer.
8. Break up the plug by using a vortex mixer and/or shaking; add 15 ml
of clean isooctane, repeating the extraction and combining isooctane
layers.
9. Evaporate the combined isooctane layers to a small volume (or
ca. 5 ml for fat) using an 80° C bath and nitrogen purge.
10. Repeat the extractions with 2 more passes of 15 ml each of isooctane,
adding the isooctane in each case to the same tube which had the
previous 2 isooctane layers.
330 G.V. Downing
11. Again evaporate as far as possible using the 80° C bath and purge.
12. Add 6 ml of methanol to all samples except fat and dissolve or
resuspend completely with an ultrasonic bath and/or vortex mixer.
13. Place all samples except fat in a refrigerator until thoroughly cooled
(at this point the sample is best left overnight in the refrigerator or
freezer).
Fat Only
12. Either add 2 ml of hexane and S ml of acetonitrile to the still hot and
molten fat or freeze overnight at -20° C. For samples frozen
overnight, remelt in a 80° C bath and add the hexane and acetonitrile
the next day.
13. Shake the still warm mixture thoroughly for ca. 1 minute and
immediately centrifuge.
14. Cool in ice water until the lower or fat layer congeals and the upper or
acetonitrile layer can be decanted completely into a fresh IS-ml
centrifuge tube.
IS. Repeat the melting, extraction with S ml of acetonitrile, and transfer
into the same IS-ml tube.
16. Add 2 ml of hexane to the IS-ml tube, shake thoroughly, centrifuge,
and remove the upper, hexane layer to waste by disposable pipet or
by aspiration. Go to Step 22.
fuge to clear the layers, and move the upper, hexane layer to waste by
disposable pipet or by aspiration.
All Tissues
22. Evaporate the acetonitrile solution to 1.0 ml using a ca. 60° C bath and
nitrogen flush.
23. If the acetonitrile is less than 1 ml, make up to 1 ml with fresh
acetonitrile.
24. Use an ultrasonic bath to get a homogeneous mix if necessary.
25. Add 4 ml of distilled water (2 ml for kidney samples) and 5 ml of
hexane; shake ca. 1 minute and centrifuge for 5 minutes.
26. Move the upper, hexane layer to a clean 15-ml centrifuge tube using a
disposable pipet. Avoid the lower layer.
27. Repeat with 5 ml and, then, 4 ml hexane extractions combining all 3
hexane extracts.
28. Evaporate to dryness (or as near dryness as possible) using a 40° C
bath and nitrogen purge. (To get the sample more completely dry,
more heat may need to be applied at the end of the evaporation. The
bath may go up to ca. 80° C at this point.)
29. Redissolve the residue in exactly 1.0 ml of methanol using a vortex
mixer and ultrasonic bath.
30. Mix thoroughly and centrifuge for 5 minutes.
Derivatization
31. Pipet exactly 0.5 ml of the solution from Step 30 into the bottom of a
silylated 15-ml centrifuge tube. Use a O.5-ml volumetric pipet.
32. Completely evaporate the methanol carefully, using at 60° C bath and
nitrogen purge. Avoid spattering.
33. With a 1.0-ml graduated pipet, add 0.1 ml of freshly made acetic
anhydride-methylimidazole-DMF mix to each tube and a series of
standard tubes (see standard section F preceding).
34. Stopper, vortex briefly, and centrifuge for a few seconds.
35. Tape the stoppers in place and put all samples and standards in a
well-stirred 95° C oil bath for 1 hour. The bottoms of the tubes should
be about 1 inch below the oil surface.
36. Remove the tubes, wash the oil off the outside of the tubes with
acetone in a wash bottle, and cool briefly. The residue should be
black.
37. Add about 1 ml of chloroform to each tube and vortex to mix.
38. Wash a SEP-PAK cartridge with 3 to 4 ml of chloroforM, using a
syringe to force the liquid through.
39. Add the sample by disposable pipet to the syringe and force it through
the cartridge.
332 G.V. Downing
All Tissues
40. Wash the centrifuge tube 3 times with 1 ml each of chloroform into the
syringe and through the cartridge.
41. Elute the column with a further 8 to 9 ml of chloroform (with aliquots
of 3, 3, 2, or 3 ml).
42. Collect all the chloroform eluate from the SEP-PAK in a 15-ml
centrifuge tube.
43. Evaporate the chloroform off with a 60° C bath and nitrogen purge.
Get the residue completely dry.
HPLC Analysis
44. Pipet 0.5 ml of methanol (or other suitable quantity) into the tube and
use a vortex mixer and ultrasonic bath to completely dissolve the
residue.
45. Centrifuge briefly and load WISP inserts if autosampler is used.
46. Inject 50 ILl of the supernatant of each sample and standard into the
HPLC.
47. Measure the peak heights at the retention time of the derivative of
dihydro-B 1a as indicated by the standards.
48. Generate a standard curve by performing a linear regression analysis
of response (peak height) versus concentration (ng/ml) for standards.
The curve should be linear with a regression coefficient of 0.97 or
better. Use the regression equation to determine the concentration of
unknowns with an observed peak height.
Calculate the sample concentration as follows:
VI VI
ppb = (ng/ml)s x V2 x DIG = V2 x (ng/ml)s x DI5
FIGURE AL2. Chromatographic tracings of (A) standard and (B) plasma sample containing ivermectin.
Appendix I. Assaying Ivermectin Residues in Tissue and Plasma 335
the syringe and through the cartridge, using the same disposable
pipet.
18. Elute the cartridge with a further 8 to 9 ml of chloroform (aliquots of 3,
3, 2, or 3 mI).
19. Collect all the chloroform, leaving the SEP-PAK from Steps 16, 17,
and 18 (eluent) in a IS-ml centrifuge tube.
20. Evaporate the chloroform completely off with a 60° C bath and
nitrogen purge. The residue must be completely dry.
21. Pipet exactly 0.5 ml of methanol (or other larger volumes for
high-assay plasmas) into the tube and use a vortex mixer and
ultrasonic bath to completely dissolve the residue.
22. Centrifuge and inject SO ILl of sample from each sample and each
standard in turn into the HPLC unit using the WISP Autosampler.
23. Measure the peak heights of the derivative at the retention time
indicated by the standards.
24. Calculate results as described in the tissue method starting at Step 48,
substituting ng/ml for ppb and S ml for G in the equation.
ApPENDIX II
B. Reagents
1. Acetic anhydride-Baker or Fisher, reagent grade.
2. Acetone-Mallinckrodt, Nanograde.
3. Acetonitrile-Fisher, HPLC grade; or Mallinckrodt, Nanograde.
4. Dimethyl Formamide (DMF)-Baker or Fisher, reagent grade.
5. Hexane-Burdick & Jackson, distilled in glass; or Mallinckrodt,
Nanograde.
Extract Into
Add IUIfurIc DIsecIM In G.1 mI
DIeeoMtIn 0.5 mt aliquot Lelstenelst { Methylene chloride Enporste
2.0 mI meIhenoI KId rugenta Hexe.,. rugent and hNt
".ponIte Iodrytubn RTovemlght oHlOhent
IDA IS-100°C 1 hour
and teke thInI .. aliquot
f
I
FIGURE All. I. Flow diagram of confirmatory assay. B is a possible stopping point during the assay.
338 G. V. Downing
C. Solutions
1. Derivatizing Reagent-Mix 0.2 ml of I-methylimidazole with 0.3 ml of
acetic anhydride and 0.9 ml of DMF .Make up just before use.
2. Acetone/Water-50% (v/v).
3. Methanol/Water Mobile Phase (90/I0)-Make 200 ml of doubly dis-
tilled and filtered water to 2 liters with methanol. Deaerate by slowly
bubbling nitrogen through for 10 minutes. Make other water content
mobile phases correspondingly.
4. 40% Methylene Chloride-Hexane-Isobutyl Alcohol-Fill a 500-ml
stoppered graduate to the 200 mark with methylene chloride. Add
hexane to the 500-ml mark and, then, 20 ml of isobutyl alcohol. Mix.
5. 1% Isobutyl Alcohol in Hexane-Make 5 ml of isobutyl alcohol to 500
ml in a graduated cylinder. Mix.
6. 1% Sulfuric Acid in Isopropanol-Pipet 0.5 ml of sulfuric acid carefully
into about 40 ml of isopropanol in a 50-ml volumetric flask. Mix. Dilute
to the mark with isopropanol and thoroughly mix. Make fresh just
before use.
7. 1% Sulfuric Acid in Methanol-Pipet 0.5 ml of sulfuric acid carefully
into about 40 ml of methanol in a 50-ml volumetric flask. Mix. Dilute to
the mark with methanol and thoroughly mix. Make fresh just be-
fore use.
D. Apparatus
1. Balance-analytical, capable of weighing 1 mg accurately.
2. Balance-capable of weighing 5 g accurately into an approximately
60-g tube.
Appendix II. Assaying Ivermectin Residues in Liver 339
E. Chromatographic Apparatus
A Beckman-Altex Model 110A liquid chromatographic pump complete
with Valco sample valve with syringe-loading sample loop or Waters
Wisp Autosampler and Kratos-Schoeffel Instruments Model FS950 fluo-
rescence detector with a I-millivolt recorder is used. A 51Lm, 4.6-mm 10,
C18 standard Brownlee Labs guard column (Spheri-5 RP-18 OD-G,
obtained from Rainin Instrument Co., Inc.) is used before the analytical
column. This guard column is replaced monthly unless the pressure
reaches 2000 psi, in which case it is replaced immediately.
Conditions: 15 cm lengthx4.6 mm 10 Zorbax ODS-CI8 column.
Mobile phase-IO% water in methanol (v/v).
Column temperature-43° C.
Flow-1.8 mIl minute (usual pressure 1000 psi, 500 to
2000 psi acceptable).
For FS950:
Excitation Lamp-FSA 110, standard 365 nm.
Standard Kratos Flowcell-FSA 210.
Excitation Filter-FSA 403, 365 nm band filters.
Emission Filter-418 nm.
Sensitivity Range-0.2 A or higher.
Retention Time-14 minutes (or greater with Modifications for I, the
parent compound)
Time Constant-about 6.
18. Add 1% isobutyl alcohol in hexane to the 7-ml mark and mix.
19. Add 4 ml of water from a graduated pipet, shake 1 minute, and
centrifuge for 5 minutes.
20. Move the upper phase as completely as possible to a fresh 15-ml tube.
Move no lower phase.
21. Repeate the extraction with a second 6 ml and combine extracts.
22. Blow completely to dryness with a nitrogen flush and a 40° C bath. At
the end of this evaporation, the temperature can be allowed to rise to
60-70° C.
23. Take up residue in exactly 0.5 ml of methanol, using vortex mixing
and ultrasound. In all above (17-23), the (M) samples should be
handled first and placed on the LC before handling the (A) or (I)
samples. After extraction, protect the (M) samples from light as much
as possible. (If the (A) and (I) samples are to be delayed signifcantly,
they should be stored in freezer at - 20° C before adding the
methanol.)
24. Centrifuge and inject 50 ILl of the clear phase (M samples) onto the
HPLC with the modified mobile phase doing the first standard of a
type, the unknowns, and finally the second standard, in that order.
25. Do the same with the (A) and (I) samples.
26. Examine the unknown sample chromatograms for the presence of the
(A), (M), and (I) peaks at the same elution time as the standard peak.
(That is, compare the unknown (A) to standard (A), etc.)
27. Average the 2 peak heights for each standard (A, M, and I) and
calculate each unknown as a precent of that standard average.
28. A value of 60% or more of all 3 (A, M, and I) is proof that that
particular level of ivermectin is present in the tissue (liver).
ApPENDIX III
List of Registrations
J. Di Netta
Ivermectin was released for registration by Merck & Co., Inc., in 1981
and first registered in France as IVOMEC Injection for Cattle that same
year.
Since that time ivermectin has been approved for use in over 60 countries.
It is currently registered for use in cattle, sheep, horses, goats, swine,
dogs, camels, reindeer, and bison.
The following lists the products and the countries in which ivermectin is
registered.
IVERMECTIN APPROVALS
IVOMEC' byection for Cattle
ALGERIA GUATEMALA PHILIPPINES
ARGENTINA HOLLAND POLAND
AUSTRALIN HONDURAS PORTUGAL
AUSTRIA HUNGARY ROMANIA
BELGIUM INDIA S. AFRICA
BRAZIL IRELAND SPAIN
BULGARIA ISRAEL SUDAN
CANADA ITALY SWEDEN
CHILE JAMAICA SWITZERLAND
COLOMBIA JAPAN THAILAND
COSTA RICA JORDAN TRINIDAD
C.S.S.R KENYA TUNISIA
DENMARK LUXEMBOURG TURKEY
ECUADOR MALAYSIA U.K.
EGYPT MEXICO URUGUAY
ELSALVADOR MOROCCO U.S.A.
FINLAND NEW ZEALAND U.S.S.R.
FRANCE NORWAY VENEZUELA
G.D.R. PANAMA YUGOSLAVIA
GERMANY PARAGUAY ZIMBABWE
GREECE PERU
IVOMEC' Oral Solution for Cattle
FRANCE NEW ZEALAND U.K.
HOLLAND SOUTHAFRICA
Appendix III. List of Registrations 345
A AGRI-MEK, 288
Abamectin Airblast treatment, 202, 203
antibacterial activity, 29 (Table 2.3) See also Worker exposure.
antifungal activity, 30 (Table 2.4) Alaska, 271
biological activity, 20 (Table 1.8) Algae, toxicity, 168
clinical trials, cattle, 216-222, Algeria, 268, 344, 346
230-233 Alkylating agents, use in mutational
compared to ivermectin, 102-103 procedures, 35-36
toxicity, 103 (Table 6.5), 110 Alkylations, 15
crop protection, 288-305 Almonds, 305
environmental effects, 184-198 Alpacas, clinical trials, 271
photoinstability, 292 AI Salvador, 344
structure of, 90 (Fig. 6.1), 184 (Fig. Amblyomma americanum, 265-266
13.1) Amidine,219
toxicity studies, 102-107 Amitraz, 254
worker exposure study, 201-213 Analytical techniques, tissue residues,
See also Specific topics. 137-138, 144-145
Acarines, 288 Ancylostoma caninum, 251-252
Acetylcholine release, ivermectin, 79 Ancylostoma spp., 255, 269, 320
Acid-sensitivity, avermectins, 8 Angiostrongylus cantonensis, 265
Aculops lycopersici, 302 Anguillicola crassus, 279
Acute toxicity Anorexia, 96, 108
abamectin, 193-194 (Table 13.4) Antelope, clinical trials, 273-274
ivermectin, 92-95, 169 Anthelmintic market, 24
Aelurostrongylus abstrusus, 255 Anthelmycin, 24
AFFIRM, 289-290 Anthelvencin, 24
nonagricultural land, 289 Antifungal/antibacterial activity
Africa, 275 abamectin, 29-30 (Tables 2.3, 2.4)
Agitators, and fermentation, 47-48, Streptomyces auermitilis, 28-31
49-50 (Figs. 3.7,3.8,3.9,3.10) Aphids, 298, 300
Aglycones Aphodius, 178
avermectin, 10-12, II (Fig. 1.5), 16 Aquatic life. See Environmental
ivermectin, 12 effects of abamectin
348 Index