Skip to main content
Top
Published in: Malaria Journal 1/2018

Open Access 01-12-2018 | Research

Differential susceptibilities of Anopheles albimanus and Anopheles stephensi mosquitoes to ivermectin

Authors: Staci M. Dreyer, Kelsey J. Morin, Jefferson A. Vaughan

Published in: Malaria Journal | Issue 1/2018

Login to get access

Abstract

Background

Vector control is a crucial element of anti-malaria campaigns and works best when there is a thorough knowledge of the biology and behaviour of the Anopheles vector species responsible for transmitting malaria within a given locale. With the push to eradicate malaria stronger than ever, there is a growing need to develop and deploy control strategies that exploit the behavioural attributes of local vector species. This is especially true in regions where the vectors are exophagic (i.e., prefer to bite outdoors), exophilic (i.e., prefer to remain outdoors), and zoophagic (i.e., as likely to feed on non-humans as humans). One promising strategy targeting vectors with these behavioural traits is the administration of avermectin-based endectocides, such as ivermectin, to humans and livestock. When ingested in a blood meal, ivermectin has been shown to reduce mosquito survivorship and fecundity in a number of Anopheles species. In this study, the relative toxicity of ivermectin was compared between two zoophagic, exophilic malaria vectors—Anopheles albimanus and Anopheles stephensi.

Results

Toxicity of ivermectin was assessed using membrane feedings, intrathoracic injections, and mosquito feedings on treated mice. When ingested in a blood meal, ivermectin was much less toxic to An. albimanus (4-day oral LC50 = 1468 ng/ml) than to An. stephensi (4-day oral LC50 = 7 ng/ml). However when injected into the haemocoel of An. albimanus, ivermectin was much more toxic (3-day parenteral LC50 = 188 ng/ml). Because the molecular targets of ivermectin (i.e., glutamate-gated chloride channels) reside outside the midgut in nerves and muscles, this suggests that ingested ivermectin was not readily absorbed across the midgut of An. albimanus. In contrast, ivermectin was considerably more toxic to An. stephensi when ingested (4-day oral LC50 = 7 ng/ml) than when injected (3-day parenteral LC50 = 49 ng/ml). This suggests that metabolic by-products from the digestion of ivermectin may play a role in the oral toxicity of ivermectin to An. stephensi. Blood meal digestion and subsequent oviposition rates were significantly hindered in both species by ingested ivermectin but only at concentrations at or above their respective oral LC50 concentrations. To test mosquitocidal activity of ivermectin in a live host system, two groups of three mice each received subcutaneous injections of either ivermectin (600 µg/kg BW) or saline (control). One day after injection, the ivermectin-treated mice (n = 3) exhibited significant mosquitocidal activity against both An. stephensi (85% mortality vs 0% in control-fed) and, to a lesser degree, An. albimanus (44% mortality vs 11% in control-fed). At 3 days, the mosquitocidal activity of ivermectin-treated mice waned and was effective only against An. stephensi (31% mortality vs 3% in control-fed).

Conclusions

Ivermectin was not uniformly toxic to both Anopheles species. Previous studies indicate that ivermectin is a good choice of endectocide to use against malaria vectors in southeast Asia and Africa. However, these data suggest that ivermectin may not be the optimal endectocide to use in Central America or the Caribbean where An. albimanus is a major malaria vector species. If endectocides are to be used to help eradicate malaria, then additional efficacy data will be needed to define the activity of specific endectocides against the major malaria vector species of the world.
Literature
1.
go back to reference World Health Organization. World malaria report 2017. Geneva: World Health Organization; 2017. p. 1–196. World Health Organization. World malaria report 2017. Geneva: World Health Organization; 2017. p. 1–196.
2.
go back to reference Molineaux L, Gramiccia G. The Garki project. Geneva: World Health Organization; 1980. p. 311. Molineaux L, Gramiccia G. The Garki project. Geneva: World Health Organization; 1980. p. 311.
3.
go back to reference Graves PM, Brabin BJ, Charlwood JD, Burkot TR, Cattani JA, Ginny M, et al. Reduction in incidence and prevalence of Plasmodium falciparum in under-5-year-old children by permethrin impregnation of mosquito nets. Bull World Health Organ. 1987;65:869–77.PubMedPubMedCentral Graves PM, Brabin BJ, Charlwood JD, Burkot TR, Cattani JA, Ginny M, et al. Reduction in incidence and prevalence of Plasmodium falciparum in under-5-year-old children by permethrin impregnation of mosquito nets. Bull World Health Organ. 1987;65:869–77.PubMedPubMedCentral
4.
go back to reference Lindblade KA, Mwandama D, Mzilahowa T, Steinhardt L, Gimnig J, Shah M, et al. A cohort study of the effectiveness of insecticide-treated bed nets to prevent malaria in an area of moderate pyrethroid resistance, Malawi. Malar J. 2015;14:31.CrossRefPubMedPubMedCentral Lindblade KA, Mwandama D, Mzilahowa T, Steinhardt L, Gimnig J, Shah M, et al. A cohort study of the effectiveness of insecticide-treated bed nets to prevent malaria in an area of moderate pyrethroid resistance, Malawi. Malar J. 2015;14:31.CrossRefPubMedPubMedCentral
5.
go back to reference Bruce-Chwatt LJ, Garrett-Jones C, Weitz B. Ten years’ study (1955–64) of host selection by anopheline mosquitos. Bull World Health Organ. 1966;35:405–39.PubMedPubMedCentral Bruce-Chwatt LJ, Garrett-Jones C, Weitz B. Ten years’ study (1955–64) of host selection by anopheline mosquitos. Bull World Health Organ. 1966;35:405–39.PubMedPubMedCentral
6.
go back to reference Campbell WC, Fisher MH, Stapley EO, Albers-Schönberg G, Jacob TA. Ivermectin: a potent new antiparasitic agent. Science. 1983;221:823–8.CrossRefPubMed Campbell WC, Fisher MH, Stapley EO, Albers-Schönberg G, Jacob TA. Ivermectin: a potent new antiparasitic agent. Science. 1983;221:823–8.CrossRefPubMed
7.
go back to reference Crump A, Omura S. Ivermectin, “Wonder drug” from Japan: the human use perspective. PJA Series B. 2011;87:13–28. Crump A, Omura S. Ivermectin, “Wonder drug” from Japan: the human use perspective. PJA Series B. 2011;87:13–28.
8.
go back to reference Crump A, Morel CM, Omura S. The onchocerciasis chronicle: from the beginning to the end? Trends Parasitol. 2012;28:280–8.CrossRefPubMed Crump A, Morel CM, Omura S. The onchocerciasis chronicle: from the beginning to the end? Trends Parasitol. 2012;28:280–8.CrossRefPubMed
9.
go back to reference Rebollo MP, Bockarie MJ. Toward the elimination of lymphatic filariasis by 2020: treatment update and impact assessment for the endgame. Expert Rev Anti Infect Ther. 2013;11:723–31.CrossRefPubMed Rebollo MP, Bockarie MJ. Toward the elimination of lymphatic filariasis by 2020: treatment update and impact assessment for the endgame. Expert Rev Anti Infect Ther. 2013;11:723–31.CrossRefPubMed
10.
go back to reference Canga AG, Prieto AMS, Liebana MJD, Martinez NG, Vega MS, Vieitez JJG. The pharmacokinetics and interactions of ivermectin in humans—a mini-review. AAPS J. 2008;10:42–6.CrossRef Canga AG, Prieto AMS, Liebana MJD, Martinez NG, Vega MS, Vieitez JJG. The pharmacokinetics and interactions of ivermectin in humans—a mini-review. AAPS J. 2008;10:42–6.CrossRef
11.
go back to reference Fritz ML, Siegert PY, Walker ED, Bayoh MN, Vulule JR, Miller JR. Toxicity of bloodmeals from ivermectin-treated cattle to Anopheles gambiae s.1. Ann Trop Med Parasitol. 2009;103:539–47.CrossRefPubMed Fritz ML, Siegert PY, Walker ED, Bayoh MN, Vulule JR, Miller JR. Toxicity of bloodmeals from ivermectin-treated cattle to Anopheles gambiae s.1. Ann Trop Med Parasitol. 2009;103:539–47.CrossRefPubMed
12.
go back to reference Kobylinski KC, Deus KM, Butters MP, Hongyu T, Gray M, Marques da Silva I, et al. The effect of oral anthelmintics on the survivorship and re-feeding frequency of anthropophilic mosquito disease vectors. Acta Trop. 2010;116:119–26.CrossRefPubMedPubMedCentral Kobylinski KC, Deus KM, Butters MP, Hongyu T, Gray M, Marques da Silva I, et al. The effect of oral anthelmintics on the survivorship and re-feeding frequency of anthropophilic mosquito disease vectors. Acta Trop. 2010;116:119–26.CrossRefPubMedPubMedCentral
13.
go back to reference Butters MP, Kobylinski KC, Deus KM, Marques da Silva I, Gray M, Sylla M, et al. Comparative evaluation of systemic drugs for their effects against Anopheles gambiae. Acta Trop. 2012;121:34–43.CrossRefPubMed Butters MP, Kobylinski KC, Deus KM, Marques da Silva I, Gray M, Sylla M, et al. Comparative evaluation of systemic drugs for their effects against Anopheles gambiae. Acta Trop. 2012;121:34–43.CrossRefPubMed
14.
go back to reference Fritz ML, Walker ED, Miller JR. Lethal and sublethal effects of avermectin/milbemycin parasiticides on the African malaria vector, Anopheles arabiensis. J Med Entomol. 2012;49:326–31.CrossRefPubMed Fritz ML, Walker ED, Miller JR. Lethal and sublethal effects of avermectin/milbemycin parasiticides on the African malaria vector, Anopheles arabiensis. J Med Entomol. 2012;49:326–31.CrossRefPubMed
15.
go back to reference Kobylinski KC, Ubalee R, Ponlawat A, Nitatsukprasert C, Phasomkulsolsil S, Wattanakul T, et al. Ivermectin susceptibility and sporontocidal effect in Greater Mekong Subregion Anopheles. Malar J. 2017;16:280.CrossRefPubMedPubMedCentral Kobylinski KC, Ubalee R, Ponlawat A, Nitatsukprasert C, Phasomkulsolsil S, Wattanakul T, et al. Ivermectin susceptibility and sporontocidal effect in Greater Mekong Subregion Anopheles. Malar J. 2017;16:280.CrossRefPubMedPubMedCentral
16.
go back to reference Sampaio VS, Beltrán TP, Kobylinski KC, Melo GC, Lima JBP, Silva SGM, et al. Filling gaps on ivermectin knowledge: effects on the survival and reproduction of Anopheles aquasalis, a Latin American malaria vector. Malar J. 2016;15:491.CrossRefPubMedPubMedCentral Sampaio VS, Beltrán TP, Kobylinski KC, Melo GC, Lima JBP, Silva SGM, et al. Filling gaps on ivermectin knowledge: effects on the survival and reproduction of Anopheles aquasalis, a Latin American malaria vector. Malar J. 2016;15:491.CrossRefPubMedPubMedCentral
17.
go back to reference Kobylinski KC, Escobedo-Vargas KS, López-Sifuentes VM, Durand S, Smith ES, Baldeviano GC, et al. Ivermectin susceptibility, sporontocidal effect, and inhibition of time to re-feed in the Amazonian malaria vector Anopheles darlingi. Malar J. 2017;16:479.CrossRef Kobylinski KC, Escobedo-Vargas KS, López-Sifuentes VM, Durand S, Smith ES, Baldeviano GC, et al. Ivermectin susceptibility, sporontocidal effect, and inhibition of time to re-feed in the Amazonian malaria vector Anopheles darlingi. Malar J. 2017;16:479.CrossRef
18.
go back to reference Sylla M, Kobylinski KC, Gray M, Chapman PL, Sarr MD, Rasgon JL, et al. Mass drug administration of ivermectin in south-eastern Senegal reduces the survivorship of wild-caught, blood fed malaria vectors. Malar J. 2010;9:365.CrossRefPubMedPubMedCentral Sylla M, Kobylinski KC, Gray M, Chapman PL, Sarr MD, Rasgon JL, et al. Mass drug administration of ivermectin in south-eastern Senegal reduces the survivorship of wild-caught, blood fed malaria vectors. Malar J. 2010;9:365.CrossRefPubMedPubMedCentral
19.
go back to reference Alout H, Krajacich BJ, Meyers JI, Grubaugh ND, Brackney DE, Kobylinski KC, et al. Evaluation of ivermectin mass drug administration for malaria transmission control across different West African environments. Malar J. 2014;13:417.CrossRefPubMedPubMedCentral Alout H, Krajacich BJ, Meyers JI, Grubaugh ND, Brackney DE, Kobylinski KC, et al. Evaluation of ivermectin mass drug administration for malaria transmission control across different West African environments. Malar J. 2014;13:417.CrossRefPubMedPubMedCentral
20.
go back to reference Saul A. Zooprophylaxis or zoopotentiation: the outcome of introducing animals on vector transmission is highly dependent on the mosquito mortality while searching. Malar J. 2003;2:32.CrossRefPubMedPubMedCentral Saul A. Zooprophylaxis or zoopotentiation: the outcome of introducing animals on vector transmission is highly dependent on the mosquito mortality while searching. Malar J. 2003;2:32.CrossRefPubMedPubMedCentral
21.
go back to reference Franco AO, Gomes MGM, Rowland M, Coleman PG, Davies CR. Controlling malaria using livestock-based interventions: a one-health approach. PLoS ONE. 2014;9:e101699.CrossRefPubMedPubMedCentral Franco AO, Gomes MGM, Rowland M, Coleman PG, Davies CR. Controlling malaria using livestock-based interventions: a one-health approach. PLoS ONE. 2014;9:e101699.CrossRefPubMedPubMedCentral
22.
go back to reference Zimmerman RH. Ecology of malaria vectors in the Americas and future direction. Mem Inst Oswaldo Cruz. 1992;87(3):371–83.CrossRefPubMed Zimmerman RH. Ecology of malaria vectors in the Americas and future direction. Mem Inst Oswaldo Cruz. 1992;87(3):371–83.CrossRefPubMed
23.
go back to reference Sinka ME, Bangs MJ, Manguin S, Rubio-Palis Y, Chareonviriyaphap T, Coetzee M, et al. A global map of dominant malaria vectors. Parasit Vectors. 2012;5:69.CrossRefPubMedPubMedCentral Sinka ME, Bangs MJ, Manguin S, Rubio-Palis Y, Chareonviriyaphap T, Coetzee M, et al. A global map of dominant malaria vectors. Parasit Vectors. 2012;5:69.CrossRefPubMedPubMedCentral
24.
go back to reference Zilberstein Y, Ayali A. The role of the frontal ganglion in locust feeding and moulting related behaviours. J Exp Biol. 2002;205:2833–41.PubMed Zilberstein Y, Ayali A. The role of the frontal ganglion in locust feeding and moulting related behaviours. J Exp Biol. 2002;205:2833–41.PubMed
25.
go back to reference Detinova TS, Beklemishev WN, Bertram DS. Age-grouping methods in Diptera of medical importance. World Heal Organ Monogr Ser. 1962;47:1–213. Detinova TS, Beklemishev WN, Bertram DS. Age-grouping methods in Diptera of medical importance. World Heal Organ Monogr Ser. 1962;47:1–213.
26.
go back to reference Abbott WS. A method of computing the effectiveness of an insecticide. J Econ Entomol. 1925;18:265–7.CrossRef Abbott WS. A method of computing the effectiveness of an insecticide. J Econ Entomol. 1925;18:265–7.CrossRef
27.
go back to reference Meyers JI, Gray M, Kuklinski W, Johnson LB, Snow CD, Black WC 4th, et al. Characterization of the target of ivermectin, the glutamate-gated chloride channel, from Anopheles gambiae. J Exp Biol. 2015;218:1478–86.CrossRefPubMedPubMedCentral Meyers JI, Gray M, Kuklinski W, Johnson LB, Snow CD, Black WC 4th, et al. Characterization of the target of ivermectin, the glutamate-gated chloride channel, from Anopheles gambiae. J Exp Biol. 2015;218:1478–86.CrossRefPubMedPubMedCentral
28.
go back to reference Toutain PL, Upson DW, Terhune TN, McKenzie ME. Comparative pharmacokinetics of doramectin and ivermectin in cattle. Vet Parasitol. 1997;72:3–8.CrossRefPubMed Toutain PL, Upson DW, Terhune TN, McKenzie ME. Comparative pharmacokinetics of doramectin and ivermectin in cattle. Vet Parasitol. 1997;72:3–8.CrossRefPubMed
29.
go back to reference Lanusse C, Lifschitz A, Virkel G, Alvarez L, Sanchez S, Sutra JF, et al. Comparative plasma disposition kinetics of ivermectin, moxidectin and doramectin in cattle. J Vet Pharmacol Ther. 1997;20:91–9.CrossRefPubMed Lanusse C, Lifschitz A, Virkel G, Alvarez L, Sanchez S, Sutra JF, et al. Comparative plasma disposition kinetics of ivermectin, moxidectin and doramectin in cattle. J Vet Pharmacol Ther. 1997;20:91–9.CrossRefPubMed
30.
go back to reference Ndong TB, Kane Y, Ba MA, Sane I, Sutra JF, Alvinerie M. Pharmacokinetics of ivermectin in zebu Gobra (Bos indicus). Vet Parasitol. 2005;128:169–73.CrossRefPubMed Ndong TB, Kane Y, Ba MA, Sane I, Sutra JF, Alvinerie M. Pharmacokinetics of ivermectin in zebu Gobra (Bos indicus). Vet Parasitol. 2005;128:169–73.CrossRefPubMed
31.
go back to reference Lyimo IN, Kessy ST, Mbina KF, Daraja AA, Mnyone LL. Ivermectin-treated cattle reduces blood digestion, egg production and survival of a free-living population of Anopheles arabiensis under semi-field condition in south-eastern Tanzania. Malar J. 2017;16:239.CrossRefPubMedPubMedCentral Lyimo IN, Kessy ST, Mbina KF, Daraja AA, Mnyone LL. Ivermectin-treated cattle reduces blood digestion, egg production and survival of a free-living population of Anopheles arabiensis under semi-field condition in south-eastern Tanzania. Malar J. 2017;16:239.CrossRefPubMedPubMedCentral
32.
go back to reference Naz S, Maqbool A, Ahmad M, Anjum AA. Efficacy of ivermectin for control of zoophilic malaria vectors in Pakistan. Pak J Zool. 2013;45:1585–91. Naz S, Maqbool A, Ahmad M, Anjum AA. Efficacy of ivermectin for control of zoophilic malaria vectors in Pakistan. Pak J Zool. 2013;45:1585–91.
Metadata
Title
Differential susceptibilities of Anopheles albimanus and Anopheles stephensi mosquitoes to ivermectin
Authors
Staci M. Dreyer
Kelsey J. Morin
Jefferson A. Vaughan
Publication date
01-12-2018
Publisher
BioMed Central
Published in
Malaria Journal / Issue 1/2018
Electronic ISSN: 1475-2875
DOI
https://doi.org/10.1186/s12936-018-2296-3

Other articles of this Issue 1/2018

Malaria Journal 1/2018 Go to the issue