ArticlePDF Available

Ivermectin induced blindness treated with intravenous lipid therapy in a dog

Authors:
Case Report
Journal of Veterinary Emergency and Critical Care
23(1) 2013, pp 58–62
doi: 10.1111/vec.12016
Ivermectin-induced blindness treated with
intravenous lipid therapy in a dog
Steven E. Epstein, DVM, DACVECC and Steven R. Hollingsworth, DVM, DACVO
Abstract
Objective – To report a case of blindness due to the ingestion of ivermectin and subsequent successful treatment
with intravenous lipid (IVL) therapy.
Case Summary – A female neutered Jack Russell Terrier was examined for acute onset of apparent blindness
after being exposed to ivermectin the previous day. The dog appeared to be blind during initial examination.
Pupillary light reflex, menace response, and dazzle reflex were not present in either eye. Fundic examination
revealed small areas of linear retinal edema. Electroretinography (ERG) showed diminished activity in both
eyes. Ivermectin was present in the serum on toxicological assay. Approximately 20 hours after exposure, IVL
was infused. Within 30 minutes of initiating the infusion, the pupillary light reflexes returned in both eyes, and
by the end of the infusion the patient behaved as if sighted. Fundic examination and ERG were unchanged at
this time. The dog was tested for the multidrug resistance gene mutation and was unaffected.
New or Unique Information Provided – Ivermectin toxicity occurs in dogs with apparent blindness being a
common clinical sign. This is the first case report of ivermectin-induced blindness evaluated with ERG before
and after treatment with IVL in a dog unaffected by the multidrug resistance gene mutation. Treatment with an
infusion of IVL therapy appeared to shorten the clinical course of disease in this patient without affecting ERG
results.
(J Vet Emerg Crit Care 2013; 23(1): 58–62) doi: 10.1111/vec.12016
Keywords:
intralipid, neurotoxin, toxicology
Introduction
An 11-year-old 10 kg female neutered Jack Russell Ter-
rier presented to the Small Animal Emergency Service
at the William R. Pritchard Veterinary Medical Teaching
Hospital of the University of California, Davis, for acute
onset blindness. The owners reported that the evening
prior to presentation the farrier was de-worming horses
with a product containing 1.87% ivermectinawith the
dog present in the pen. No abnormalities were noted
with the dog on the previous day. General physical ex-
amination findings on presentation were unremarkable.
The initial ophthalmic examination revealed a patient
who behaved as if unsighted; no menace response was
present in either eye and the patient ran into obstacles un-
der both photopic and scotopic conditions. Both pupils
From the Department of Surgical and Radiological Sciences, School of Vet-
erinary Medicine, University of California, Davis, CA.
The authors declare no conflict of interest.
Address correspondence and reprint requests to
Dr. Steven E. Epstein, Department of Surgical and Radiological Sciences,
School of Veterinary Medicine, University of California, Davis, CA 95616,
USA.
Email: seepstein@ucdavis.edu
Submitted September 25, 2011; Accepted November 25, 2012.
Abbreviations
ERG electroretinography
IVL intravenous lipid
GABA gamma-aminobutyric acid
MDR-1 multidrug resistance gene
PLR pupillary light reflex
were mydriatic in ambient light and the pupillary light
reflexes (PLRs) were absent.
A board certified ophthalmologist performed a com-
plete ophthalmic examination approximately 2 hours
after presentation to the emergency service, including
slit lamp biomicroscopy, indirect ophthalmoscopy, and
electroretinography (ERG).bBoth eyes were open and
appeared comfortable. No PLRs (either direct or consen-
sual), menace responses, nor dazzle reflexes were present
in either eye. No abnormalities were detected in the an-
terior segment or vitreous humor of either eye.
Fundic examination revealed small areas of linear reti-
nal edema ventral and medial to optic disk in the non-
tapetal area of both eyes (Figure 1). Following a 25-
minute period of dark adaption, an ERG was performed
58 CVeterinary Emergency and Critical Care Society 2013
Ivermectin-induced blindness treated with intralipid therapy
Figure 1: Retinal examination of dog with ivermectin-induced
blindness. The image depicts areas of retinal edema, right eye.
without sedation. Retinal responses from each eye were
assessed separately by use of a series of 8 red-light flashes
(findings were averaged). The b-wave amplitude was
75.56 v in the right eye and 33.89 v in the left (nor-
mal retinal function for this unit is >100 v). While the
specific origin of the ERG b-wave is controversial, it is
currently thought to be the result of retinal bipolar cell
activity.1, 2 In common practice, b-wave amplitude is con-
sidered the key indicator of retinal function.1–3
Results of a CBC were unremarkable. Serum biochem-
ical analyses showed no clinically significant abnormal-
ities. Serum was analyzed for macrocyclic lactones via
liquid chromatography-mass spectrometry with results
returning the following day positive for ivermectin at
1,500 parts per billion, but negative for abamectin, mox-
idectin, and selamectin. Testing for ABCB1-1or multi-
drug resistance (MDR)-1 allele mutation later revealed
that the dog was unaffected.
Approximately 20 hours after exposure and 3.5 hours
after clinical signs were noted, an IV catheter was
placed, and a 20% intralipid emulsioncwas administered
(1.5 mL/kg over a 10-minute period, then 0.25 mL/kg/
min for a 90-minute period). Thirty minutes after initi-
ation of the infusion, slight PLRs and a positive dazzle
reflex were noted. At the end of the infusion the patient
appeared to be sighted navigating around obstacles and
avoiding walls, had a positive menace response, but still
had bilateral mydriasis in ambient light. The linear ar-
eas of retinal edema were also still present. The ERG
was repeated and the b-wave amplitudes were similar
to the previous recordings with 55.83 v for the right eye
and 39.44 v for the left eye. The dog was discharged to
the owner the same day and has remained sighted ever
since.
Figure 2: Follow-up retinal examination of dog with ivermectin-
induced blindness. Same area of right eye as depicted in
Figure 1, 9 months after toxic event.
Approximately 9 months after initial evaluation the
dog was reexamined. General physical examination was
unremarkable except for a healing wound on the right
antebrachium. Ophthalmic examination revealed nor-
mal pupil size in ambient light, present and brisk direct
and consensual PLRs, and a positive menace response
and dazzle reflex in both eyes. There was no evidence
of any intraocular inflammation and the areas of retinal
edema had resolved (Figure 2). The patient easily navi-
gated a maze under both photopic and scotopic condi-
tions. An ERG was not repeated at this time due to equip-
ment malfunction. The patient subsequently returned for
an ERG 2 months later (approximately 11 months after
the initial evaluation). The b-wave amplitudes had im-
proved in both eyes to 86.39 v in the right eye and
84.17 v in the left.
Discussion
Ivermectin toxicosis is well described in dogs.4–10 A wide
variety of clinical signs can occur including blindness,
mydriasis, hypersalivation, ataxia, tremors, respiratory
failure, obtundation, and death. Some intoxicated ani-
mals may show apparent blindness with or without the
other generalized signs. The first reported retinal lesions
with presumed ivermectin toxicosis were in 2 dogs in
198911 and have been subsequently reported in another
dog with presumed ivermectin toxicosis12 and in 2 dogs
with confirmed ivermectin toxicosis.13 Blindness due to
suspected ivermectin ingestion has also been reported in
a mule foal.14
Ivermectin, avermectin, milbemycin, and moxidectin
are macrocyclic lactones and are commonly used in
CVeterinary Emergency and Critical Care Society 2013, doi: 10.1111/vec.12016 59
S. E. Epstein and S. R. Hollingsworth
veterinary medicine. Gamma-aminobutyric acid
(GABA) is an inhibitory amino acid neurotransmitter.
GABA receptors, when activated by either endogenous
GABA or ivermectin, cause cell membrane hyperpo-
larization via increased postysynaptic permeability to
chloride ions. This effect is resolved with the application
of picrotoxin, a GABA antagonist.16 A second proposed
mechanism for the action of ivermectin is by binding
glutamate gated chloride channels which similarly
causes inhibition of excitatory motor neurons.16 In
nematodes ivermectin results in rapid paralysis of
movement and inability to feed due to pharyngeal mus-
cle weakness resulting in death of the parasite.15 Clinical
signs of ivermectin toxicity in mammals are attributed
to this enhancement of neuronal inhibition.17,18
In mammals GABA mediated interneurons are largely
present within the CNS, but may also be present on
skeletal muscle and intestines.19 P-glycoprotein (P-gp)
is a large transmembrane protein encoded by the MDR-
1 gene, and is responsible for limiting the penetration
of ivermectin into the CNS. Toxicity in mammals re-
quires a much larger dose as compared with that ob-
served in nematodes.15 The exceptions are dogs with a
homozygous mutation in the ABCB1-1allele that show
increased sensitivity to ivermectin toxicosis.20 In dogs
without the ABCB1-1polymorphism, clinical signs of
toxicosis can occur at doses of 2.5 mg/kg.21 In this Jack
Russell Terrier who was negative for the ABCB1-1
polymorphism, the ingested dose was unknown.
The mechanism for ivermectin-induced blindness is
not known at this time. Reports in dogs suggest that reti-
nal pathology is involved in the process, but there may
be a nerve and cortical component as well. Most cell
types within the retina express GABA-ergic receptors22
and GABA is considered the primary inhibitory neuro-
transmitter of the mammalian retina.23 It is possible that
if ivermectin crosses the blood-retinal barrier, neurons
originating in the retina may be affected in a manner
similar to the CNS.
This case report is of particular interest for a number of
reasons. Because the dog had participated in a noninva-
sive clinical study about 6 months prior to the toxic event,
the dog had received a complete ophthalmic examina-
tion by the same veterinary ophthalmologist who later
examined this dog in connection with the toxic episode.
The dog had been found to be free of any ophthalmic
abnormalities at that time, although an ERG was not
performed. Similar to 1 dog in a previous report,13 and
a mule foal14 with suspected ivermectin-induced blind-
ness, this patient did not exhibit an extinguished ERG
reading even at presentation when she was blind and
lacked PLRs. This would seem to eliminate the retina
as the sole location for the changes in PLRs and vi-
sion loss and imply a multifactorial anatomic basis of
this disease. Additionally, after the administration of
intravenous lipid (IVL) therapy and subsequent return
of vision, this patient did not demonstrate a significant
change in fundic signs or ERG readings, and only a min-
imal improvement in PLRs. The fact that the dog had
a return to vision without a significant change in PLRs
would tend to rule out cranial nerve II as the location
of the toxic effect as a lesion here would probably af-
fect both PLRs and vision. Cranial nerve III dysfunction
could cause PLR abnormalities without affecting vision.
However, this would necessitate a lesion relatively dis-
tal in the course of the nerve, as more proximal lesions
would also cause ptosis and strabismus, or ivermectin
only affecting the superficial and medial layers of the
nerve. Taken together, this would suggest that the blind-
ness associated with ivermectin toxicity is not entirely
retinal in origin and likely partially cortical in origin,
with the PLR effects due to effects elsewhere.
Ivermectin-induced blindness occurred in 22% of sus-
pected or diagnosed canine ivermectin toxicosis cases re-
ported to the Animal Poison Control Center.24 The dog
in this report had exposure to ivermectin with simul-
taneous positive plasma levels for ivermectin confirm-
ing ingestion. Positive plasma levels may only be used
to confirm ingestion and are less important than brain
concentrations because the latter correlate more directly
with the clinical consequences of intoxication.25, 26 Cur-
rent treatment recommendations for ivermectin toxicity
are supportive in nature. Picrotoxin, a GABA antagonist,
was reported to aid the recovery of 1 dog with ivermectin
toxicity, but caused violent seizures and tachycardia and
is not currently recommended.27 Vision loss associated
with ivermectin toxicity is temporary, and anecdotally,
recovery is expected in 2–10 days28 although the exact
recovery time is unknown. As blindness can last many
days, animals are typically discharged to owners to wait
for vision to return which can create anxiety for owners
and potentially the patient. In this case, IVL therapy was
used, which was associated with a rapid return of vision
in less than 90 minutes.
The treatment of ivermectin toxicosis with IVL has re-
cently been described29, 30 although there are anecdotal
reports of its use for years. In one case of an ABCB1-1
unaffected Border Collie31 that presented with both oph-
thalmologic and neurologic symptoms, the dog’s dazzle
response returned within 6 hours. The PLR was incom-
plete and visual ability improved over the next 24 hours.
The return to vision appeared to take longer than the
time course for the case reported here, but in both cases
the dazzle response returned within hours of initiating
IVL therapy.
This seemingly positive response to lipid therapy was
not appreciated in 3 dogs that were homozygous for
the ABCB1-1gene mutation.30 This patient and the
60 CVeterinary Emergency and Critical Care Society 2013, doi: 10.1111/vec.12016
Ivermectin-induced blindness treated with intralipid therapy
Border Collie were negative for the mutation, yet both
appeared to have a clinical response to IVL therapy,
while 3 homozygous dogs did not. This raises the pos-
sibility that IVL therapy may only be helpful if the dog
is not a homozygous mutant. Possible support for this is
that P-gp is expressed in renal tubular epithelial and bil-
iary canalicular cells,31 which may reduce the excretion
rates during ivermectin toxicosis in ABCB1-1homozy-
gous mutant dogs. P-gp also transports ivermectin from
the brain into the blood. Without this ability, homozy-
gous mutant dogs would rely solely on a concentration
gradient for diffusion of ivermectin from the brain into
the infused lipid. Without P-gp, the ivermectin parti-
tioned in the circulating lipid could back diffuse into
the brain, limiting the potential helpful effects of IVL
therapy. In wild type dogs, ivermectin transport into cir-
culating lipid may be an active process and back diffu-
sion into the brain would be limited. Further investiga-
tions are needed to investigate the role of IVL therapy in
ABCB1-1homozygous mutant dogs.
Intravenous lipid has also been used in the treatment
of moxidectin toxicosis in a puppy, which may have
shortened the clinical course of disease,32 and recently
IVL therapy was used to treat successfully lidocaine tox-
icity in a cat.33 In human medicine, IVL infusions have
been used to treat toxicities for lipid soluble drugs such as
local anesthetics,34, 35 a severe case of organophosphate
poisoning,36 antidepressants,37–40 and cardiac drugs.41–43
The mechanism by which lipids improve clinical signs
of toxicity associated with the macrocyclic lactones is
unclear. A proposed mechanism is through a “lipid
sink” where ivermectin is removed from the CNS or
retina by sequestering it into the plasma lipid phase. If
a drug is lipophilic, theoretically this allows a higher
drug concentration in the plasma with less available
to be toxic in the tissues. A drug is deemed lipophilic
if its log Pis >1.0. This “lipid sink” theory is sup-
ported with increasing plasma levels of clomipramine,37
mepivacaine,44 bupropion,39 and ivermectin29 when tox-
icoses are treated with lipid emulsions. The log Pfor
these drugs are 3.30, 1.89, 3.47, and 3.50, respectively.45
Although 20% intralipid solutions have been used in
parenteral nutrition safely, there is no clinical evidence
on safety of short-term large boluses of lipid solutions.
Short-term infusions of soybean oil-based lipid emul-
sions have been shown to decrease neutrophil function
in dogs.46 Other potential adverse effects are pancreati-
tis, fat emboli, phlebitis, and hypersensitivity reactions.
However, no adverse outcome related to IVL therapy
for the treatment of intoxication has been reported ex-
cept for laboratory difficulty in analyzing lipemic blood
samples.47
The optimal dose of intralipid for IVL therapy
is unknown. The dose of 1.5 mL/kg bolus then
0.25 mL/kg/min was chosen based on this being the
most commonly cited dose in human case reports. A re-
view of IVL therapy to treat local anesthetic toxicity in
humans suggests a dose of 1.5 mL/kg bolus that can be
repeated up to 3 times in cardiac arrests then 0.25–0.5
mL/kg/min.48 On this basis it is reasonable to use this
dose in veterinary medicine as well until further studies
show an optimal dose.
The present case report shows a definitive diagnosis of
ivermectin ingestion with acute onset of blindness that
resolved temporally after a lipid infusion. The patient’s
fundic changes were not altered with lipid therapy, nor
were the ERG results different, despite an apparent clin-
ical improvement. The improvement observed in this
patient however cannot definitively be attributed to the
IVL therapy. Further assessment of plasma levels would
have been useful to support a “lipid sink” theory, but
as they do not necessarily correlation to brain levels or
clinical signs, they were not performed. At this time IVL
therapy appears to be a reasonable treatment option for
ivermectin-induced blindness although its safety and ef-
ficacy in canine patients has not been fully determined
at this time. Further studies on the benefit of lipid emul-
sions to treat lipid soluble toxicities such as ivermectin
are indicated.
Footnotes
aDuramectin Paste, Durvet, Blue Springs, MO.
bRetinoGraphics, Inc., Norwalk, CT.
cIntralipid, for Baxter Healthcare Corporation by Fresenius Kabi, Uppsala,
Sweden.
References
1. Ekesten B. Ophthalmic examination and diagnostics, part 4: electro-
diagnostic evaluation of vision, In: Gelatt KN. ed. Veterinary Oph-
thalmology, 4th ed. Ames, IA: Blackwell; 2007.
2. Ofri R. Retina, In: Maggs DJ, Miller PE, Ofri R. eds. Slatter ’s Fun-
damentals of Veterinary Ophthalmology, 4th ed. St. Louis: Elsevier;
2008.
3. Acland GM. Diagnosis and differentiation of retinal diseases in small
animals by electroretinography. Semin Vet Med Surg Sm Anim 1988;
3:15–27.
4. Easby SM. Ivermectin in the dog. Vet Rec 1984; 115:45.
5. Paul AJ, Tranquili WJ, Seward RL, et al. Clinical observations in
Collies given ivermectin orally. Am J Vet Res 1987; 48:684–686.
6. Houston DM, Parent J, Matushek KJ. Ivermectin toxicosis in a dog.
J Am Vet Med Assoc 1987; 191:78–80.
7. Hopkins KD, Marcella KL, Strecker AE. Ivermectin toxicosis in a
dog. J Am Vet Med Assoc 1990; 197:93–94.
8. Hadrick MK, Bunch SE, Kornegay JN. Ivermectin toxicosis in two
Australian shepherds. J Am Vet Med Assoc 1995; 206:1147–1150.
9. Hopper K, Aldrich J, Haskins SC. Ivermectin toxicity in 17 collies. J
Vet Intern Med 2002; 16:89–94.
10. Nelson OL, Carsten E, Bentjen SA, et al. Ivermectin toxicity in an
Australian shepherd dog with the MDR1 mutation associated with
ivermectin sensitivity in Collies. J Vet Intern Med 2003; 17:354–356.
11. Ketring K. In: Proceedings of presumed ocular toxicity of ivermectin.
13th Annu Kal Kan Symp 1989; 109–110.
CVeterinary Emergency and Critical Care Society 2013, doi: 10.1111/vec.12016 61
S. E. Epstein and S. R. Hollingsworth
12. Wolfer J, Grahn B, Lane S. Diagnostic ophthalmology. Can Vet J
1997; 38:729–730.
13. Kenny PJ, Vernau KM, Puschner B, et al. Retinopathy associated
with ivermectin toxicosis in two dogs. J Am Vet Med Assoc 2008;
233:279–284.
14. Plummer CE, Kallberg ME, Ollivier FJ, et al. Suspected ivermectin
toxicosis in a miniature mule foal causing blindness. Vet Ophthalmol
2006; 9(1):29–32.
15. Campbell WC, Fisher MH, Stapley EO, et al. Ivermectin: a potent
new antiparasitic agent. Science 1983; 221:823–827.
16. Wolstenholme AJ, Rogers AT. Glutamate-gated chloride chan-
nels and the mode of action of the avermectin/milbemycin an-
thelmintics. Parasitology 2005; 131(suppl):S85–S95.
17. Wang CC, Pong SS. Actions of avermectin B1a on GABA nerves.
Prog Clin Biol Res 1982; 97:373–395.
18. Pong SS, Wang CC. Avermectin B1a modulation of g-aminobutyric
acid receptors in rat brain membranes. J Neurochem 1982; 38:375–
379.
19. Trailovic SM, Nedeljkovic JT. Central and peripheral neurotoxic ef-
fects of ivermectin in rats. J Vet Med Sci 2011; 73(5):591–599.
20. Mealey KL, Bentjen SA, Gay JM, et al. Ivermectin sensitivity in
collies is associated with a deletion mutation of the mdr1 gene.
Pharmacogenetics 2001; 11:727–733.
21. Mealey KL. Ivermectin: macrolide antiparasitic agents, In: Peterson
ME, Talcott PA. eds. Small Animal Toxicology. 2nd ed. St. Louis:
Elsevier; 2006.
22. Robin LN, Kalloniatis M. Interrelationship between retinal ischemic
damage and turnover and metabolism of putative amino acid neuro-
transmitters, glutamate and GABA. Doc Ophthalmol 1992; 16:125–
130.
23. Yang XL. Characterization of receptors for glutamate and GABA in
retinal neurons. Prog Neurobiol 2004; 73:127–150.
24. Merola V, Khan S, Gwaltney-Brant S. Ivermectin toxicosis in dogs:
a retrospective study. J Am Anim Hosp Assoc 2009; 45:106–111.
25. Marques-Santos LF, Bernardo RR, de Paula EF, et al. Cyclosporin
A and trifluoperazine, two resistance modulating agents increase
ivermectin neurotoxicity in mice. Pharmacol Toxicol 1999; 84:125–
129.
26. Lankas GR, Minsker DH, Robertson RT. Effects of ivermectin on
reproduction and neonatal toxicity in rats. Food Chem Toxicol 1989;
27(8):523–529.
27. Sivine F, Plume C, Ansay M. Picrotoxin, the antidote to ivermectin
in dogs? Vet Rec 1985; 116:195–196.
28. Gelatt KN. Essentials of Veterinary Ophthalmology, 1st ed. Balti-
more, MD: Lippincott Williams & Wilkins; 2000.
29. Clarke DL, Lee JA, Murphy LA, et al. Use of intravenous lipid
emulsion to treat ivermectin toxicosis in a border collie. J Am Vet
Med Assoc 2011; 239(10):1328–1333.
30. Wright HM, Chen AV, Talcott PA, et al. Intravenous fat emulsion as
treatment for ivermectin toxicosis in three dogs homozygous for the
ABCB1–1gene mutation. J Vet Emerg Crit Care 2011; 21:666–672.
31. Mealey KL. Therapeutic implications of the MDR-1 gene. J Vet Phar-
macol Ther 2004; 27:257–264.
32. Crandell DE, Winberg GL. Moxidectin toxicosis in a puppy success-
fully treated with intravenous lipids. J Vet Emerg Crit Care 2009;
19:181–186.
33. O’Brien TQ, Clark-Price SC, Evans EE, et al. Infusion of a lipid
emulsion to treat lidocaine intoxication in a cat. J Am Vet Med Assoc
2010; 12:1455–1458.
34. Warren J, Thoma R, Georgescu A, et al. Intravenous lipid infusion
in the successful resuscitation of local anesthetic-induced cardiovas-
cular collapse after supraclavicular brachial plexus block. Anesth
Analg 2009; 108:1578–1580.
35. .Sonsino DH, Fischler M. Immediate intravenous lipid infusion in
the successful resuscitation of ropivicaine-induced cardiac arrest
after infraclavicular brachial plexus block. Reg Anesth Pain Med
2009; 34:276–277.
36. Zhou Y, Zhan C, Li Y, et al. Intravenous lipid emulsions combine
extracorporeal blood purification: a novel therapeutic strategy for
severe organophosphate poisoning. Med Hypotheses 2010; 74:309–
311.
37. Finn S, Uncles D, Willers J, et al. Early treatment of a quetiap-
ine and sertraline overdose with Intralipid. Anaesthesia 2009; 64:
191–194.
38. Harvey M, Cave G, Hoggett K. Correlation of plasma and peritoneal
dialysate clomipramine concentration with hemodynamic recovery
after intralipid infusion in rabbits. Acad Emerg Med 2009; 16(2);151–
156.
39. Sirianni AJ, Osterhoudt KC, Calello DP, et al. Use of lipid emul-
sion in the resuscitation of a patient with prolonged cardiovascular
collapse after overdose of bupropion and lamotrigine. Ann Emerg
Med 2008; 51(4):412–415.
40. Weinberg G, Di Gregorio G, Hiller D, et al. Reversal of haloperidol
induced cardiac arrest by using lipid emulsion. Ann Intern Med
2009; 150(10):737–738.
41. Dolcourt B, Arron C. Intravenous fat emulsion for refractory vera-
pamil and atenolol induced-shock: a human case report [abstract].
Clin Toxicol 2008; 46:620.
42. Hurley WT, Hanalon P. Lipid emulsion as an antidote at the Wash-
ington Poison Control Center; use in carbamazepine, flecaninde,
hydrochloroquine, bupivicaine, and buproprion. North American
congress of clinical toxicology annual meeting. San Antonio, TX,
USA. Clin Toxicol 2009; 47:702–765.
43. Harvey MG, Cave GR. Intralipid infusion ameliorates propranolol-
induced hypotension in rabbits. J Med Toxicol 2008; 4(2):71–76.
44. Litz RJ, Roessel T, Heller AR, et al. Reversal of central ner-
vous system and cardiac toxicity after local anesthetic intoxica-
tion by lipid emulsion injection. Anesth Anal 2008; 106(5):1575–
1577.
45. Fernandez AL, Lee JA, Rahilly L, et al. The use of intravenous lipid
emulsion as an antidote in veterinary toxicology. J Vet Crit Care
2011; 21(4):309–320.
46. Kang JH, Yang MP. Effect of a short-term infusion with soybean oil-
based lipid emulsion on phagocytic responses of canine peripheral
blood polymorphonucreal neutrophilic leukocytes. J Vet Intern Med
2008; 22:1166–1173.
47. Jamaty C, Bailey B, Larocque A, et al. Lipid emulsions in the treat-
ment of acute poisoning: a systematic review of human and animal
studies. Clin Toxicol 2010; 48:1–27.
48. Cave G, Harvey M. Intravenous lipid emulsion as antidote beyond
local anesthetic toxicity: a systematic review. Acad Emerg Med 2009;
16:815–824.
62 CVeterinary Emergency and Critical Care Society 2013, doi: 10.1111/vec.12016
... Antidysrhythmic [30] Flecainide 1 0 20 Beta-blocker [41] Metoprolol 1 0 20 [28,44] Propranolol 2 0 34 [29] Atenolol 1 0 20 [49] Propanolol/Clonidine 1 0 36–48 Calcium channel blocker [81] Diltiazem 0 1 1 [35,83] Nifedipine 2 0 25 [24,54,56,57] Verapamil 4 0 133 Insecticide/antiparasitic [65] Avermectin 0 1 1 [53] Chlorpyriphos 1 0 49 [51] Diazinon 1 0 24 [37] Dichlorvos 1 0 48 [68,71,73,75,79,84,85] Ivermectin 0 7 17 [64] Ivermectin + Praziquantel 0 1 1 [64,72,77] Moxidectin + Praziquantel 0 3 4 [36] Parathion 1 0 18 [67,74,76,80,82] Permethrin 0 5 11 [25,33,58] Malathion 3 120 GABA agonists 3 [64,70,78] Baclofen 0 0 8 [46] Pentobarbital 1 0 NR [38] Propofol 1 0 NR [32,43,47] Thiopental 3 55 ...
... Five animal experiments report the effect of ILE in the treatment of either dichlorvos,[37] parathion,[36] and malathion toxicity.[25,33,58] Numerous animal case reports and case series describe ILE for the treatment of antihelminthic toxicity in particular, with ivermectin [64,68,69,71,73,75,84,85] and moxidectin [64,72,77] poisoning. Gang and colleagues evaluated the toxicity of intraperitoneal dichlorvos in a rodent model. ...
Article
Background: The use of intravenous lipid emulsion (ILE) therapy for the treatment of lipophilic drug toxicity is increasing. Despite this, the evidence for its effect in non-local anesthetic toxicity remains sparse. Furthermore, many case reports describe ILE use for substances in which no clear efficacy data exists. The American Academy of Clinical Toxicology established a lipid emulsion workgroup. The aim of this group is to review the available evidence regarding the effect of ILE in non-LA drug poisoning and develop consensus-based recommendations on the use of this therapy. Methods: A systematic review of the literature was performed to capture articles through 15 December 2014. Relevant articles were determined based upon a predefined methodology. Articles involving pre-treatment experiments, pharmacokinetic studies not involving toxicity, and studies not addressing antidotal use of ILE met pre-defined exclusion criteria. Agreement of at least two members of the subgroup was required before an article could be excluded. Results: The final analysis included 203 articles: 141 for humans and 62 for animals. These include 40 animal experiments and 22 case reports involving animal toxicity. There were three human randomized control trials (RCT): one RCT examined ILE in TCA overdose, one RCT examined ILE in various overdoses, and one study examined ILE in reversal of sedation after therapeutic administration of inhaled anesthesia. One observational study examined ILE in glyphosate overdose. In addition, 137 human case reports or case series were identified. Intravenous lipid emulsion therapy was used in the management of overdose with 65 unique substances. Conclusions: Despite the use of ILE for multiple substances in the treatment of patients with poisoning and overdose, the effect of ILE in various non-local anesthetic poisonings is heterogenous, and the quality of evidence remains low to very low.
... We found published data documenting retinal lesions associated with presumptive ivermectin intoxication in 2 dogs in 1989 [21]. Epstein and Hollingsworth [22] also reported a case of apparent blindness in a Jack Russell Terrier subsequent to an apparent ivermectin overdose. A detailed ophthalmic examination of the affected dog carried out using slit lamp biomicroscopy, indirect ophthalmoscopy and electroretinography revealed diminished pupillary light reflex, menace response, dazzle reflex and retinal edema in both the eyes. ...
... Recently, ILEs have also been used as an antidote for ivermectin toxicity in various species of animals [15, 36, 37]. Previously, ILE has been successfully used to treat ivermectin intoxication in various breeds of dogs including Australian Shepherd [36], Jack Russell Terrier [22], Border Collie [16] as well as in a miniature Shetland pony [37]. Additionally, ILE has also been used to address moxidectin toxicosis in a puppy [28]. ...
Article
Full-text available
Ivermectin is widely used in veterinary practice for the treatment of ecto- and endo-parasites. In wildlife, an extra-label use this parasiticide is sometimes associated with toxicity. Different treatment regimens have been used in ivermectin toxicosis. The present report describes a successful reversal of ivermectin toxicity by intravenous administration of a commercially available lipid emulsion in a captive African lion (Panthera leo). Case presentation A 2-year old captive African lion (Panthera leo) weighing ~130 kg was presented with acute neurological impairment and bilateral blindness that had developed 24 h after ivermectin exposure. The animal was treated with a commercially available lipid emulsion along with supportive therapy and experienced complete recovery. To our knowledge, this is the first case report of the use of lipid emulsion in the management of ivermectin induced blindness in an African lion and it appears that intravenous lipid emulsion may be an effective therapy in ivermectin toxicity in lions. Further testing in expanded clinical trials is clearly warranted.
... Kintamani puppies were found to exhibit toxicity symptoms after treatment, including restlessness, depression, tremors, mydriasis, hypersalivation, anorexia, and polydipsia (Tabel 1). These symptoms are similar to clinical ivermectin poisoning findings reported in dogs (Hopper et al., 2002;Epstein and Hollingsworth, 2013), calves (Patel et al., 2018), horses (Norman et al., 2012), lions (Saqib et al., 2015), and rabbit (Branco et al., 2021). Until now, ivermectin is still the drug of choice in parasite control practices because of its broad spectrum of action. ...
Article
Full-text available
Pr Ivermectin toxicity is known to cause harmful side effects or even death in dogs intolerant to the medication. Intolerant dogs have a mutation in the MDR-1 (Multi-Drug Resistance) gene, so they lack the P-glycoprotein gene that removes drugs from the brain. Therefore, this study aimed to determine ivermectin toxicity in Kintamani Bali puppies by examining physiological responses based on hematological profiles and aminotransferase activity after a high-dose injection. A laboratory observational approach was used, and the samples were 25 healthy female Kintamani puppies based on a veterinary examination, aged 3-6 months, weighing 6.32 ± 1.18 kg, randomly divided equally into five treatment groups. The treatments included a placebo (1ml Aqua Pro Injection) as a control, as well as a single dose of ivermectin injection sequentially 200, 400, 800, and 1600 µg/kg subcutaneously. Blood samples were collected before treatment and after 7 and 14 days post-treatment. The hematologic parameters observed included levels of hemoglobin, erythrocytes, hematocrit, total leukocytes, neutrophils, lymphocytes, monocytes, eosinophils, and basophils, as well as blood biochemistry, namely aspartate aminotransferase (AST) and alanine aminotransferase (ALT) activities. Observation results after 4 hours of administration of ivermectin at doses of 800 and 1600 µg/kg of puppies showed changes in behavior, restlessness, depression, tremors, mydriasis, hypersalivation, anorexia, and polydipsia. Meanwhile, the results of hematological examination on the seventh day after ivermectin treatment showed a trend of erythropenia, leukocytosis, a decrease in hemoglobin levels, and an increase in aminotransferase enzyme activity. This condition continued until day 14, but the physiological parameter values showed that the puppy’s condition gradually improved compared to the seventh day after treatment. There were significant differences in the blood profile, AST, and ALT of Kintamani puppies injected with ivermectin at doses of 800 and 1,600 ug/kg compared to controls on days 7 and 14 after and before treatment. It was concluded that high-dose ivermectin injections in Kintamani Bali puppies caused toxicity with clinical signs of erythropenia, decreased hemoglobin, leukocytosis, and increased aminotransferase activity.
... The undesirable efficacy of ivermectin depends on the dose and duration of administration (Prichard, 1985). Ivermectin overdose may be produced variable side effects ranging from mild to extremely severe (Epstein and Hollingsworth, 2013). The most dominant clinical symptoms of ivermectin poisoning in domestic and wild animals are CNS depression and sometimes coma frequently resulting in death (Trailovic and Nedejkovic, 2011). ...
Article
Full-text available
Ivermectin (IVM), natural fermentation product derived from the soil bacterium Streptomyces avermitilis, is a broad spectrum anthelmintic, insecticide and acaricide. The present study was designed to detect the effect of therapeutic and double therapeutic dose of ivermectin on liver and kidney function parameters, sperm count and abnormalities as well as, histopathological alterations on liver, kidney and tests. Thirty male white albino rats were equally divided into three groups, group (A): control, group (b): have therapeutic dose of ivermectin (0.2 mg/kg BW/SC), group (C): have double therapeutic dose of ivermectin (0.4 mg/kg BW/SC). The rats were injected once weekly for eight weeks. In both treated groups a significant increase in aspartate aminotransferase (AST), alanine aminotransferase (ALT), urea, uric acid and creatinine were recorded. Albumin and total protein were significantly decreased than that of the control group. Moreover, significant decrease in total sperm count with significant increase in sperm abnormality was also demonstrated. Various pathological changes in liver, kidney and tests were also detected. The severity of these changes varied from mild to severe changes according to the dose as histopathological changes were more severe in male rats injected with double therapeutic dose than that injected with the therapeutic one. Following to the present results, the administration of either therapeutic or double therapeutic dose of ivermectin produced alterations in some biochemical parameters which correlated with histopathological changes as well as, it has deleterious effect on male fertility. Consequently, it could be concluded that, it is not preferable to use ivermectin particularly at double therapeutic dose mostly to breading males. [Rabab, R, Elzoghby, Aziza Amin, Ahlam, F, Hamouda, and Abdel Fatah, Ali. Toxicological and pathological studies of Ivermectin on male albino rats. J Am Sci 2015;11(3):73-83]. (ISSN: 1545-1003). http://www.jofamericanscience.org. 10
Article
Full-text available
Michael D Becker, Brian C YoungEmergency and Critical Care, Animal Specialty Group, Los Angeles, CA, USAAbstract: The objective of this retrospective study was to describe the responses to treatment with intravenous lipid emulsion (ILE) and the outcomes for a variety of severe intoxications. This case series includes 10 client-owned animals, 9 dogs and 1 cat, that underwent treatment with ILE for a variety of severe intoxications over a 4-year period. History, physical examination findings, clinical signs, clinicopathological test results, treatment, response to treatment, and outcome were recorded. Eight of the 10 patients survived to discharge. The toxicities included in this case series were baclofen, ivermectin and spinosad plus milbemycin oxime, baclofen and tadalafil, carbamate, methamphetamine, dextroamphetamine sulfate, amlodipine, bromethalin, and organophosphate. The two patients who died were intoxicated with bromethalin and an organophosphate. Six of the 10 patients developed lipemia secondary to ILE administration, and there were no other known adverse effects. Overall, ILE was a safe therapeutic option. This case series provides clinical evidence of successful treatment with ILE as an antidote for previously unpublished toxicities (amlodipine, carbamate, methamphetamine, and dextroamphetamine sulfate), additional evidence of success in treating baclofen and ivermectin toxicosis, as well as unsuccessful treatment of bromethalin and organophosphate toxicities.Keywords: intravenous lipid emulsion, toxicity, amlodipine
Article
Zusammenfassung Vergiftungen können zu lebensbedrohlichen Notfällen führen. Neben der allgemeinen Stabilisation des Patienten sollte eine Therapie eine schnellstmögliche Toxinelimination bewirken. Zunächst wird versucht, durch allgemeine Dekontaminationsmaßnahmen (gastrointestinal, dermal, okulär) die Giftaufnahme so gering wie möglich zu halten. Eine genauere Eingrenzung des Toxins bzw. seiner Eigenschaften kann bei der Auswahl spezifischerer Behandlungsansätze, wie beispielsweise Dialyse oder Gabe eines Antidots, Hilfestellung leisten. Die Therapie mit Lipidemulsionen zur raschen Elimination lipophiler Substanzen aus dem Körper hat in den letzten Jahren zunehmend an Bedeutung gewonnen. Sie kann das Allgemeinbefinden des Patienten schnell verbessern und ist mit wenig Nebenwirkungen verbunden.
Article
Full-text available
Objective: To describe the outcome of 20 cats treated with intravenous lipid emulsion (IVLE) after an accidental parenteral ivermectin overdose. Case series summary: Twenty adult cats presented after receiving a 4 mg/kg accidental subcutaneous overdose of ivermectin. After admission, two IVLE treatments were initiated in asymptomatic cats: a single bolus (1.5 mL/kg; n = 16) versus a bolus followed by a 30-minute constant rate infusion (0.25 mL/kg/min; n = 4). Six out of the 16 cats that received only the single bolus developed clinical signs of ivermectin intoxication. Based on the severity of the clinical signs and their duration (approximately 48 hours), these 6 cats were retrospectively considered either moderately (n = 3) or severely (n = 3) intoxicated by ivermectin. Cats with a low body condition score (BCS) had more severe signs of ivermectin toxicity. Additional IVLE was administered until clinical resolution was complete. Median (min to max) cumulative dose of IVLE per cat was 4.5 (3.0-4.5) mL/kg for 36 (12-36) hours and 19.5 (7.5-37.5) mL/kg for 96 (72-168) hours for moderately and severely intoxicated cats, respectively. New or unique information provided: Our series describes the treatment of accidental ivermectin parenteral overdose in 20 cats with early initiation of IVLE therapy. An early bolus followed by a 30-minute constant rate infusion of IVLE appeared to mitigate the signs of ivermectin toxicosis in cats compared to a single treatment bolus. Our observations also suggest that cats with a low BCS given only a bolus of IVLE treatment were more likely to develop signs of ivermectin intoxication and require a greater amount of IVLE for the resolution of clinical signs. Based on our observations, BCS appears to influence the severity of ivermectin toxicity with a low BCS being associated with more severe signs of ivermectin toxicity.
Article
Full-text available
Lipid resuscitation therapy was identified in 1998 as an effective treatment for local anesthetic systemic toxicity in an animal model. Since the original observation, the field has progressed tremendously with successful clinical translation and expansion of use to treatment of other types of drug overdose. Recent work has expanded our understanding of the mechanism of this novel treatment, one that includes both a dynamic scavenging component and direct cardiotonic effect. In this review, we discuss the past, present, and future of lipid resuscitation therapy with a focus on our understanding of the mechanism and directions that the field is moving, both from a clinical and basic research side. © 2015 American Society for Parenteral and Enteral Nutrition.
Article
Full-text available
Ivermectin is considered a very safe drug; however, there are reports of toxic effects in particularly sensitive populations or due to accidental overdose. The aim of this study was (1) to further characterize the central and peripheral toxic effects of ivermectin in animals and (2) to determine possible therapeutic strategies for use in cases of ivermectin poisoning. We tested the effects of experimental doses of ivermectin previously reported to cause various intensities of CNS depression. However, in our study, ivermectin at 2.5, 5.0 and 7.5 mg/kg i.v. did not produce visible CNS depression in rats and 10 mg/kg resulted in sleepiness and staggering 10 to 40 min after application, while a dose of 15 mg/kg caused CNS depression very similar to general anesthesia. Ivermectin dose-dependently potentiates thiopentone-induced sleeping time in rats. Flumazenil (0.2 mg/kg), the benzodiazepine antagonist, did not affect the action of thiopentone; however, it significantly reduced sleeping time in rats treated with a combination of ivermectin (10 mg/kg) and thiopentone (25 mg/kg; from 189.86 ± 45.28 min to 83.13 ± 32.22 min; mean ± SD). Ivermectin causes an increase in the tonus (EC(50)=50.18 µM) and contraction amplitude (EC(50)=59.32 µM) of isolated guinea pig ileum, very similar to GABA, but without the initial relaxation period. These effects are dose-dependent and sensitive to atropine. Our results confirm the central and peripheral GABAergic properties of ivermectin in mammals and also indicate involvement of the cholinergic system in its toxicity. In addition, the results suggest that flumazenil and atropine have potential clinical roles in the treatment of ivermectin toxicity.
Article
Full-text available
A 5-year-old castrated male domestic shorthair cat was examined because of presumptive lidocaine intoxication. Thirty minutes earlier, the cat had received an SC injection of approximately 140 mg of lidocaine hydrochloride (20 mg/kg [9.1 mg/lb]) to facilitate closure of a wound on the left pelvic limb. Initial physical examination revealed severe lethargy and respiratory distress; erratic, poor-quality pulses with severe hypotension; and pulmonary edema. Initial supportive treatment included administration of oxygen and IV administration of lactated Ringer's solution. Additional treatment with a 20% lipid emulsion (1.5 mL/kg [0.68 mL/lb], IV) delivered over a 30-minute period resulted in dramatic improvement in cardiovascular and behavioral variables. No adverse effects from lipid emulsion were detected on routine hematologic evaluation, thoracic radiography, or computed tomography. IV administration of a lipid emulsion was used in the treatment of lidocaine intoxication in a cat. Rapid infusion of a lipid emulsion may be a therapeutic option for veterinary patients with toxicosis attributable to local anesthetics or other lipid-soluble drugs.
Article
: Avermectin B1a stimulates high-affinity binding of [3H]-γ-aminobutyric acid (GABA) to receptors in washed rat brain membranes. Scatchard analysis of the data indicates that the drug does not significantly alter the apparent dissociation constant of GABA binding, but increases the detectable number of binding sites from 3.2 to 5.1 pmol/mg protein. (+)-Bicuculline completely blocks control and avermectin B1a-stimulated GABA binding, whereas picrotoxin antagonizes specifically the avermectin B1a-stimulated GABA binding. The avermectin B1a-stimulated GABA binding is also chloride ion-dependent, whereas GABA binding in the control is not. These observations suggest that the mechanism of avermectin B1a stimulation of GABA binding may involve the chloride ion channel.
Article
To describe the outcome of 3 cases of ivermectin toxicosis in dogs homozygous for the ABCB1–1Δ gene mutation treated with intravenous fat emulsion (IFE). One Australian Shepherd and 2 Miniature Australian Shepherds were treated for naturally occurring ivermectin toxicosis with IFE. All 3 dogs were homozygous for the ABCB1–1Δ gene mutation. Serum ivermectin concentrations confirmed ivermectin exposure in each case. All 3 dogs exhibited tremors, ptyalism, and central nervous system depression, which progressed over several hours to stupor in 2 dogs, and to a comatose state requiring mechanical ventilation in the remaining dog. A 20% formulation of IFEa was administered as an IV bolus (1.5 mL/kg) followed by a slow IV infusion (7.5–15 mL/kg [0.25–0.5 mL/kg/m], over 30 minutes). No change was observed in the neurologic status of any patient. Lipemia visible upon blood sampling persisted for 36 hours in 1 dog however, no other adverse effects were noted. Flumazenil (0.01 mg/kg IV), followed by a constant rate infusion(CRI) of 0.01 mg/kg/h IV was administered in 1 case, without any apparent clinical benefit or adverse effect. IFE was ineffective in the treatment of ivermectin toxicosis in these ABCB1–1Δ homozygous mutant dogs. Further investigation is necessary to determine why IFE treatment was unsuccessful in these cases and whether its use can be optimized to yield better results.
Article
A 2-year-old spayed female Border Collie was treated with IV lipid emulsion (ILE) after ingesting 6 mg/kg (2.73 mg/lb) of an equine ivermectin anthelmintic paste 8 hours prior to examination. On initial examination, the dog had stable cardiovascular signs but had diffuse muscle tremors and was hyperthermic. Neurologic evaluation revealed that the dog was ataxic and had mydriasis with bilaterally absent menace responses and pupillary light reflexes. The remaining physical examination findings were unremarkable. Results of CBC, serum biochemical analysis, venous blood gas analysis, and measurement of plasma lactate concentration were also within reference limits. The dog was treated with ILE in addition to supportive care with IV fluid therapy and cardiovascular, respiratory, and neurologic monitoring. The use of ILE treatment was initiated in this patient on the basis of previous clinical and experimental evidence supporting its use for toxicosis resulting from lipid-soluble agents. An initial bolus of 1.5 mL/kg (0.68 mL/lb) of a 20% sterile lipid solution was administered IV over 10 minutes, followed by a constant rate infusion of 0.25 mL/kg/min (0.11 mL/lb/min) over 60 minutes that was administered twice to treat clinical signs of ivermectin toxicosis. The dog was discharged from the hospital 48 hours after admission and was clinically normal within 4 days after ivermectin ingestion. Further diagnostic evaluation subsequently revealed that this dog was unaffected by the multidrug resistance gene (MDR-1) deletion, known as the ATP-binding cassette polymorphism. Ivermectin toxicosis in veterinary patients can result in death without aggressive treatment, and severe toxicosis often requires mechanical ventilation and intensive supportive care. This is particularly true in dogs affected by the ATP-binding cassette polymorphism. Novel ILE treatment has been shown to be effective in human patients with lipid-soluble drug toxicoses, although the exact mechanism is unknown. In the patient in the present report, ILE was used successfully to treat ivermectin toxicosis, and results of serial measurement of serum ivermectin concentration supported the proposed lipid sink mechanism of action.
Article
To review the use of IV lipid emulsion (ILE) for the treatment of toxicities related to fat-soluble agents; evaluate current human and veterinary literature; and to provide proposed guidelines for the use of this emerging therapy in veterinary medicine and toxicology. Human and veterinary medical literature. Human data are composed mostly of case reports describing the response to treatment with ILE as variant from mild improvement to complete resolution of clinical signs, which is suspected to be due to the variability of lipid solubility of the drugs. The use of ILE therapy has been advocated as an antidote in cases of local anesthetic and other lipophilic drug toxicoses, particularly in the face of cardiopulmonary arrest and unsuccessful cardiopulmonary cerebral resuscitation. The use of ILE therapy in veterinary medicine has recently been advocated by animal poison control centers for toxicoses associated with fat-soluble agents, but there are only few clinical reports documenting successful use of this therapy. Evidence for the use of ILE in both human and veterinary medicine is composed primarily from experimental animal data. The use of ILE appears to be a safe therapy for the poisoned animal patient, but is warranted only with certain toxicoses. Adverse events associated with ILE in veterinary medicine are rare and anecdotal. Standard resuscitation protocols should be exhausted before considering this therapy and the potential side effects should be evaluated before administration of ILE as a potential antidote in cases of lipophilic drug toxicoses. Further research is waranted.
Article
2 dogs (dogs 1 and 2) were examined for sudden onset of blindness. Both dogs had mild obtundation and mydriasis in both eyes. It was thought that dog 1 may have ingested ivermectin; dog 2 had been treated with ivermectin for demodectic mange. On initial examination, both dogs had mydriasis and decreased pupillary light reflexes in both eyes. Dog 1 had an absent menace response bilaterally. Fundic examination of both eyes in both dogs revealed regions of multifocal retinal edema and folds with low-lying retinal separation. The electroretinogram was extinguished in dog 1 and attenuated in dog 2. Ivermectin was detected in serum samples from both dogs. Both dogs made a complete clinical recovery following cessation of exposure to ivermectin; electroretinographic findings improved, and retinal edema resolved with some residual chorioretinal scarring. To our knowledge, this is the first report of resolution of retinal edema and electroretinographic changes associated with ivermectin toxicosis in dogs. In dogs that develop blindness suddenly, fundic examination, electroretinography, and assessment of serum ivermectin concentration are diagnostically useful, even if exposure to ivermectin is unknown.