Abstract
Ivermectin is a broad-spectrum antiparasitic agent that interferes with glutamate-gated chloride channels found in invertebrates but not in vertebrate species. Mass drug administration (MDA) with ivermectin-based regimes has been a mainstay of elimination efforts targeting onchocerciasis and lymphatic filariasis for more than 3 decades. More recently, interest in the use of ivermectin to control other neglected tropical diseases (NTDs) such as soil-transmitted helminths and scabies has grown. Interest has been further stimulated by the fact that ivermectin displays endectocidal efficacy against various Anopheles species capable of transmitting malaria. Therefore there is growing interest in using ivermectin MDA as a tool that might aid in the control of both malaria and several NTDs. In this review we outline the evidence base to date on these emerging indications for ivermectin MDA with reference to clinical and public health data and discuss the rationale for evaluating the range of impacts of a malaria ivermectin MDA on other NTDs.
Keywords: ivermectin, neglected tropical diseases, malaria, mass drug administration, soil-transmitted helminths
Introduction
Ivermectin is a macrocyclic lactone compound and part of the avermectin family. Avermectins were discovered by Satoshi Omura and William C. Campbell in Japan in the 1970s, during analysis of Streptomyces avermitilis compounds, and they subsequently discovered ivermectin. In 2015, both scientists received the Nobel Prize in Physiology or Medicine for their discovery.1 Since its introduction, the drug's utility has seen its use extended in veterinary medicine and animal husbandry to treat endo- and ectoparasites.2–4
Ivermectin is a mainstay in the success of the control and elimination of Onchocerca volvulus, the causative agent of river blindness. It has been extensively used by the African Programme for Onchocerciasis Control, the Expanded Special Project for the Elimination of Neglected Tropical Diseases in Africa and the Onchocerciasis Elimination Program of the Americas. Ivermectin is also known to affect a variety of invertebrate species.5–7 Due to its broad application, it is considered an endectocide, a drug affecting several ecto- and endoparasites, and its use has steadily expanded in the years since its discovery. In recent years ivermectin has been successfully applied on a larger scale against several pathogens/parasites, including scabies mites (Sarcoptes scabiei), lice (Pediculus humanus sp.) and helminths such as Strongyloides stercoralis8–11 and there is growing interest in its use as a mosquitocidal agent as part of malaria control.
We aimed to summarise data on the use of oral ivermectin in non-immuncompromised patients across a range of emerging indications. We highlight key data on the rationale, dosage considerations and existing evidence supporting the use of ivermectin for each new indication. The pharmacology and mode of action of ivermectin have been extensively reviewed elsewhere12–16 and we therefore primarily limit this literature review to factors of direct relevance to its extended use. However, a short summary of the mode of action and pharmacology will be given for completeness. Finally, this literature review is restricted to multicellular parasites, excluding suggested but unproven applications in oncology17 or virology,18,19 including severe acute respiratory syndrome coronavirus 2.
Mode of action
In invertebrates, ivermectin interferes with glutamate-gated chloride channels (GluCls), which are not expressed in vertebrates. GluCls play a role in several processes in invertebrates and their inhibition affects motility, feeding and reproduction.15,20 These effects are shown at nanomolar concentrations. At higher concentrations, ivermectin interacts with a variety of receptors such as γ-aminobutyric acid, glycine, histamine and nicotinic acetylcholine receptors, which are expressed in both invertebrates and vertebrates.20
Vertebrates, including humans, express P-glycoprotein (P-gp), also known as multidrug resistance protein 1 (MDR1), in their blood–brain barrier, which functions as a transport efflux pump of ivermectin out of the central nervous system.16,21 The combination of its receptor specificity and the existence of P-gp is thought to be the major factor behind the safety and side-effect profile of ivermectin. Notably, some species, such as certain dog or horse breeds, do not possess the gene encoding P-gp, and recently a human case was found.22 Therefore, in specific animal species, the use of ivermectin, especially at high dosages, can lead to drowsiness, coma and death,23,24 clearly demonstrating the protective role of P-gp in humans.16
Safety considerations
Ivermectin has an extremely well-established safety profile, with billions of doses being administered since the inception of the Mectizan Donation Programme by Merck in 1987 for onchocerciasis and filariasis control.25 Pharmacokinetic dosing studies have suggested that doses of ivermectin up to six times the recommended dose as well as repeated daily or monthly doses26–32 are well tolerated. There is a well-established risk associated with the use of ivermectin in Loa loa (a filariform parasite) endemic areas. In this setting, ivermectin can lead to a rapid die-off of large numbers of Loa loa microfilaria in the central nervous system, leading to a potentially fatal encephalopathy.33,34
Currently, due to a lack of safety data, ivermectin should not be given to pregnant women35, however, inadvertent use in control programmes has occurred regularly.36 The majority of data currently are based on observed teratogenicity from animal models using P-gp-deficient mice37 or very high doses in rats and rabbits with 10–50 and 7–30 times the human equivalent, respectively.38–40 The relevance of these animal data to humans is therefore questionable and better data are needed. Currently children whose weight or height is <15 kg or <90 cm are also not recommended to receive ivermectin. The basis for these restrictions is the unproven concept of an immature ‘leaky’ blood–brain barrier, for which there is no scientific support.41–43 In contrast to theoretical concerns, there is an increasing accumulation of real-world data showing safety among young children.44–50
Malaria
Malaria control measures over the past 2 decades have resulted in a significant reduction in morbidity and mortality, driven by a combination of long-lasting insecticidal nets, indoor residual spraying, artemisinin-based combination therapy and rapid diagnostic tests.51 However, the emergence of drug and insecticide resistance, and changes in vector behaviour, such as increased outdoor biting and resting behaviour, is threatening this progress.52–54 Over the past decade, interest has emerged in the use of ivermectin as an additional tool for the control of malaria.55,56
Anopheles mosquitoes predominantly express GluCls in organs and tissues responsible for their sensory and motor function.14 The same channels exist in the culicine nervous system; however, ivermectin appears to be unable to penetrate into the haemocoel and only exerts an effect at levels 10 times greater than shown for Anopheles sp. Its effect on culicine species such as Aedes and Culex is therefore greatly reduced57,58 unless the drug is injected directly into the haemocoel.59
Several historical studies have explored the use of ivermectin and its impact on mosquito control,60–62 but significant interest for malaria vector control has re-emerged recently.63 These studies use different methods to assess ivermectin's effect. Specifically, membrane feeding assays (MFAs) involve feeding mosquitos on donated blood, either from donors who have taken oral ivermectin or on blood spiked with ivermectin. Direct feeding assays (DFAs) involve feeding mosquitos on volunteers treated with ivermectin. Different Anopheles species, such as Anopheles gambiae (MFA, DFA), Anopheles arabiensis (MFA), Anopheles aquasalis (MFA, DFA), Anopheles minimus (DFA), Anopheles campestris (DFA), Anopheles sawadwongporni (DFA), Anopheles dirus (MFA), Anopheles darlingi (MFA), Anopheles farauti (DFA), and Anopheles stephensi (human MFA, mouse DFA), have all shown high mortality after ingesting blood containing ivermectin levels comparable to those reached in humans after an oral dose of 200, 400 and 600 μg/kg body weight.58,64–69 The IVERMAL trial found no difference in ivermectin mosquitocidal toxicity between MFAs and DFAs against A. gambiae using placebo (n=23), 300 μg/kg/d (n=24) or 600 μg/kg/d (n=22).70 Although DFAs showed higher mosquitocidal toxicity than MFAs in a trial by Sampaio et al.,64 the number of participants was small (n=6).
Pharmacokinetic considerations limit the effectiveness of a single standard dose of ivermectin of 200 μg/kg for malaria control programmes. The half-life of 18 h means that these dosing regimens only generate a mosquitocidal effect lasting for about 5–6 d,71 which is inadequate for malaria control. Furthermore, vectors from outside the treated areas, especially in open systems on larger landmasses, will quickly repopulate these losses. To improve the pharmacokinetic profile, and hence the duration of its endectocidal effect, alternative dosages have been suggested: a single dose of 400 μg/kg or three consecutive daily doses of 300 μg/kg.72 The latter regime was investigated in the IVERMAL trial conducted in Kenya and was given once a month for three consecutive months in human volunteers. The treatment had a good safety profile and the mosquitocidal effect lasted for up to 28 d.73
In the Repeat Ivermectin Mass Drug Administrations for Control of Malaria: a Pilot Safety and Efficacy Study (RIMDAMAL) conducted in Burkina Faso, villages were randomly assigned to ivermectin (150–200 μg/kg) and albendazole (400 mg) at baseline in both arms followed by the same single doses of ivermectin every 3 weeks over 18 weeks in the intervention arm or no treatment in the control arm. The study aimed to evaluate the effect on the cumulative incidence of uncomplicated malaria. The results showed evidence of a reduction in incidence in children <5 y of age,74 although the statistical methods for analysis have been disputed.75,76
The results of these relatively small trials have led to the planning of larger trials. The 300 μg/kg/d for 3 d treatment schedule is now being evaluated in ongoing or planned cluster randomized trials: the MASSIV trial (NCT03576313) in Gambia,77 the Adjunctive Ivermectin Mass Drug Administration for Malaria Control (MATAMAL) trial in the Bijagos Islands, in Guinea Bissau (NCT04844905) and RIMDAMAL II in Burkina Faso (NCT03967054). The BOHEMIA trial is currently planned to be conducted in Tanzania and Mozambique, in which ivermectin will be administered to both livestock and humans. Another trial is planned in Thailand using ivermectin in rubber plantation workers, but it has not yet started.
Potential veterinary application of ivermectin as part of malaria MDA
Several Anopheles species, such as A. arabiensis and A. farauti, exhibit both anthropophagy and zoophagy, particularly for peridomestic animals such as cattle and pigs.78,79 These alternative feeding sources can therefore sustain the mosquito population and complicate control efforts.80 Treating livestock therefore offers a possible addition for vector control for malaria transmission and has been shown to be feasible in field studies in Belize, Burkina Faso and Tanzania.81–83 Veterinary applications of ivermectin allow for higher and repeated dosing than are possible in humans, as well as application of potential long-lasting formulations.84–86
Similarly, Glossina palpalis and Glossina morsitans, the vectors for Trypanosoma gambiense and Trypanosoma rhodesiense, West and East African sleeping sickness, respectively, take their blood meal from humans, wild animals and livestock alike. Field studies have shown these species exhibit similar susceptibility to ivermectin as Anopheles mosquitos. This included dose-dependent reduced lifespan and fecundity.87–89 Similar data from animal models exist for some triatomine bugs (Triatoma infestans and Rhodnius neglectus), vectors of Trypanosoma cruzi, the causative agent of Chagas disease.90
This ‘One Health’ approach could offer additional advantages by treating animals for endoparasites and ectoparasites, improving the health and economic value of domestic animals,91 while also providing vector control for malaria and other diseases. The use of ivermectin in animals is restricted by public health policies, such as the withdrawal times for slaughter or milking,92 which could make this strategy technically challenging.93 Another important aspect is the effect of ivermectin in livestock on dung degradation and non-target fauna, which could cause environmental concerns94–98 and needs to be addressed.
Soil-transmitted helminths (STHs)
STHs are among the most prevalent parasitic infections in humans both in tropical and subtropical regions of the globe99,100 and are associated with broad health impacts including anaemia, stunting and delays in cognitive development.101
MDA with benzimidazol derivatives (albendazole and mebendazole) is recommended to reduce the STH burden in a community,102 because these drugs have a significantly greater efficacy compared with ivermectin in most STH species.103,104 Data on the effect of ivermectin on hookworms show a variable reduction of 0–33%,105,106 with the most successful application being two doses of 200 μg/kg 10 d apart reported from Brazil.8 In comparison, both Ascaris lumbricoides and Strongyloides stercoralis respond well to a single standard ivermectin dose of 200 μg/kg each, with field studies finding cure rates of 98–100% and 83–96%,107,108 respectively. Reports on Trichuris trichiura are mixed, ranging from 11% in Tanzania to 84% in Peru.8,103,105,109,110 The reasons for these geographical differences in susceptibility are not yet well understood but could be due to different species.111 Other nematodes such as Ancylostoma braziliense, Ancylostoma caninum and Uncinaria stenocephala are primarily zoonotic diseases that cause cutaneous larva migrans (CLM) syndrome in humans. Depending on the clinical presentation, one to two standard doses of ivermectin have been used and have been shown to resolve the lesions in 81–100% of cases.112,113
Currently there are no published data evaluating the impact of higher-dose multiple treatment regimes, as utilised for malaria control, on STHs. Ongoing malaria MDA provides an additional opportunity to investigate these potential synergistic impacts.
Filarial worms
Filarial infections were the first human disease targeted for control using ivermectin. Widespread roll-out of ivermectin MDA has produced a significant impact on filarial disease–related morbidity, including blindness and severe pruritus caused by O. volvulus and lymphatic obstruction and secondary bacterial skin disease caused by Wuchereria bancrofti, Brugia malayi and Brugia timori.114–116
Ivermectin as a single dose administered annually at 150–200 μg/kg for onchocerciasis will reduce the microfilarial load by 99% after 1–2 months and administered over 16–18 y interrupts transmission and leads to elimination.117,118 Recent data have shown that a sterilizing effect on adult onchocercal filaria can be achieved with administration every 3 months over 3 y.119
In lymphatic filariasis (LF), caused by W. bancrofti, B. malayi and B. timori, ivermectin (200 μg/kg) lacks activity against the adult filaria responsible for the pathology and it is therefore used in combination with either albendazole or diethylcarbamazine citrate (DEC) or as a triple combination of all three outside onchocerciasis areas.120–122 The latter combination of ivermectin, DEC and albendazole has shown superior efficacy compared with the dual combination120,122–124 and is now recommended by the World Health Organization for use in many LF-endemic regions.
Ivermectin is used with caution in Loa loa-endemic areas with a surveillance system for early detection and management of post-treatment severe adverse events, as it results in rapid killing of microfilaria (mf),125 which can cause acute encephalitis, leading to disability and even death.33,34,126 For other common filarial parasites such as Mansonella streptocerca and Mansonella ozzardi, ivermectin treatment with 150 μg/kg and 150–200 μg/kg, respectively, leads to a reduction of microfilaria and possibly some impact on macrofilaria.127–130Mansonella perstans was shown not to be affected by a standard single dose of ivermectin,131–134 with reports of repeated doses being potentially more successful.32,135 Importantly, ivermectin does not appear to affect the vector of these filaria, Culicoides sp.136,137
Food-borne nematodes
For food-borne nematodes such as Gnathostoma sp., the recommended daily dosage is 200 μg/kg for 2–3 d.138,139 Caution is advised in infections of the central nervous system, as treatment could cause deleterious inflammation. For trichinellosis, ivermectin was effective in rat and mouse models against the free-living stage in the gut but was ineffective against the encysted stage of the parasite.140,141
Other nematodes
Enterobius vermicularis, colloquially known as pinworm/threadworm, is a common cosmopolitan parasite primarily causing anal pruritus and in rare cases appendicitis. It has been successfully treated with a single dose of ivermectin (200 μg/kg), with a study from Peru reporting cure rates of 89% 3 d after treatment and 78% after 30 d,109 but a study from China showed a lower cure rate of 52.9%.105
Ectoparasites
Scabies is a globally occurring skin disease caused by the scabies mite (Sarcoptes scabiei var. hominis) that is especially common in poor and crowded communities in tropical and subtropical areas142 and causes both significant morbidity and mortality through its downstream sequelae.143,144
There is limited pharmacodynamic data available on the use of ivermectin for scabies, although an animal model in pigs is available.145 Doses ≤150 μg/kg have lower efficacy,146 and even at standard doses of 200 μg/kg, increased survival times have been found in vitro over the last decade.147 The use of a higher dose and repeated administration may improve the cure rate.143
Several large-scale trials have demonstrated significant reductions in the prevalence of scabies following MDA with ivermectin. The Skin Health Intervention Fiji Trial was a three-arm randomised trial in which communities were randomized to standard of care, MDA with topical permethrin or MDA with ivermectin. MDA was superior to other treatment options, with a relative reduction in prevalence of 94% for ivermectin, 62% for permethrin and 49% for standard of care.9 The Azithromycin Ivermectin Mass Drug Administration trial on the Solomon Islands, a prospective single-arm, before and after community intervention trial using ivermectin and azithromycin in combination and permethrin 5% for pregnant and breastfeeding women and children weighing <12.5 kg, showed an 88% relative reduction of baseline scabies prevalence after 12 months.148 Similar results have been reported from studies in Australia using ivermectin MDA for scabies control in remote aboriginal communities10 and Brazil using ivermectin as a community intervention for several susceptible parasites.8,149
Success of ivermectin-based MDA for scabies control is dependent on treating individuals with a contraindication to ivermectin. Currently this is through topical permethrin treatment, but increasing safety data on ivermectin in these populations, especially for children <5 y of age, may increase the proportion of the population who can be treated with ivermectin.
Humans are host to three species of closely related lice: Pediculus humanus capitis, Pediculus humanus corporis and Phtirus pubis. Of these, only the body louse P. humanus corporis commonly acts as a vector of potentially life-threatening infectious diseases. However, recent data have shown the potential for head lice to also transmit similar pathogens,150 are a cause of bacterial pyoderma of the scalp151 and even cause iron deficiency in heavy infestations.152 All three of these species cause pruritus and hence morbidity.153,154
In a cluster randomized trial including centres in the UK, Ireland, France and Israel, a dose of 400 μg/kg/d 1 week apart resulted in a 97.1% reduction of head lice on day 15.155 Another randomized household-level trial in Brazil using 200 μg/kg/d twice 10 d apart led to 16% in the intervention arm being louse free compared with 4% in the control arm at 60 d post-intervention.156 Several non-randomized studies from Egypt and Mexico using 200 μg/kg/d showed cure rates of 92.5–97% after a second dose 8 d later if the first dose failed.157–159 A study in the Solomon Islands using MDA with a dose of 200 μg/kg/d on days 0 and 7 resulted in a 89% reduction of head lice at day 14 post-MDA160 and a study in Thailand using the same schedule showed a 95% reduction at 14 d post-MDA.161
A study from Senegal using 400 μg/kg/d resulted in a 77.4% reduction in the ivermectin arm compared with 32.3% in the d-phenothrin shampoo arm at day 15. However, 7.4% of the children showed treatment failure to ivermectin162 and there was some evidence of potential ivermectin resistance in head lice. Additional molecular analysis confirmed a genetic mutation of the GluCl receptor, the primary target of ivermectin in arthropods.163
Data on ivermectin for the treatment of body lice and pubic lice are scarce and mainly from smaller case series or cohort studies. These data appear to show a significant reduction in prevalence.164,165 In this context, a potential ivermectin resistance pathway has been described outside of the GluCl receptor, called complexin, a synaptic exocytosis and neurotransmitter release regulator protein.166 Aside from resistance, reintroduction and re-infestation is a common problem in all three species of lice even after successful MDA.160,164,167
Data from Brazil on the treatment of Tunga penetrans with a standard dose of ivermectin did not show efficacy, although it may be dependent on seasonality and the timing of the application.149,168 In myiasis, which is common in tropical communities and can cause significant morbidity, ivermectin has been successfully used to facilitate extraction of larvae.169,170
There are only experimental blood feeding data from human studies using ivermectin to treat Cimex lectularius and Cimex hemipterus, the cause of bed bugs, a global nuisance. These data show some impact, but real-world data are unavailable.171–173 Ivermectin has also been used with variable success for the treatment of Demodex mites, which are associated with a variety of inflammatory skin diseases, including acne, rosacea, blepharitis and peri-oral dermatitis,174–176 but larger randomized studies are needed to show specific efficacy of ivermectin.
Table 1.
Endoparasites | Potential impact of ivermectin MDA | Ivermectin dose (individual treatment) | Ivermectin MDA schedule for control | Reduction at recommended dose (%)a | References |
---|---|---|---|---|---|
Ascaris lumbricoides | Yes | 200 μg/kg, once | 98–100% | 8, 103, 106 | |
Necator americanus | Unclear | Not recommended, two doses of 200 μg/kg 10 d apart | 0–33% single dose of 20 μg/kg, 68% two doses of 200 μg/kg 10 d apart | 8, 103, 105, 106 | |
Ancylostoma duodenale | Unclear | Not recommendedb | b | b | |
Strongyloides stercoralis c | Yes | 200 μg/kg once or multiple several days apart (day 1, 2, 15 and 16) | 83–96% | 8, 103, 106, 107, 108, 109 | |
Trichuris trichiura d | Yes | 200 μg/kg for 3 de, 200 μg/kg twice 10 days apart | 11–88%c; 81.7–84% 200 μg/kg twice 10 d apart | 8, 103, 105, 109, 110 | |
Enterobius vermicularis | Yes | 200 μg/kg once, plus repeat after 14 d | 52.6–89% | 105, 109 | |
Onchocerca volvulus | Yes | 150–200 μg/kg biannually or annually | 99% reduction in microfilaria after 1–2 months; transmission interruption and elimination after 16–18 y | 117–119 | |
Loa loa | Yes | Not recommended | 125 | ||
Wuchereria bancrofti | Yes | Ivermectin monotherapy not recommended | 200 μg/kg annually in combination with a second drug or as triple therapy | 94% reduction in microfilaria using IDA | 120–124 |
Brugia malayi | Yes | see W. bancrofti | |||
Brugia timori | Yes | see W. bancrofti | |||
Mansonella perstans | Unclear | 200–600 μg/kg once, not recommended | 400 μg/kg once then 800 μg/kg annually for 3 y or 400 μg/kg twice then 800 μg/kg every 3 months for 3 y20 | No effect short term; MDA 85–97% reduction | 131–135 |
Mansonella streptocerca | Yes | 150 μg/kg once | 55–60% reduction in microfilariaf | 127, 128 | |
Mansonella ozzardi | Yes | 150–200 μg/kg once | 94–100% reduction in microfilaria | 128–130 | |
Gnathostoma sp. | Yes | 200 μg/kg for 2 d | 76–100% | 138, 139 | |
Trichinella spiralis | Mixed | 200 μg/kg once, not recommended | No effect on encysted form; 80–90% in free living formsg | 140, 141 | |
Ancylostoma braziliense, Ancylostoma canium, Uncinaria stenocephala h | Yes | 200 μg/kg, 1–2 doses depending on the clinical picture | 81–100% | 112, 113 |
aCure rate if not otherwise indicated.
bPossibly a similar situation as N. americanus; no speciation conducted.
cIn immunocompetent patients.
d T. trichiura may consist of several species explaining the geographically different rates in reduction after treatment.
eUnknown.
fPotential effect on macrofiliaria similar to O. volvulus.
gOnly animal model data available.
hAll responsible for CLM.
Table 2.
Ectoparasites (excluding Anopheles) | Potential impact of ivermectin MDA | Ivermectin dose (individual treatment) | Ivermectin MDA schedule for control | Reduction at recommended dose (%)a | Parasite mortality (%) after n days | References |
---|---|---|---|---|---|---|
Sarcoptes scabiei var. hominis (scabies) | Yes | 200 μg/kg/day, 2 weeks apart or a single dose | 200 μg/kg/d 1–2 weeks apart | 83–100% at 12 monthsb | 8–10, 146–149 | |
Pediculus humanus capitis (head louse) | Yes | 200–400 μg/kg/d 1 week apart | 77.4–97.1% for 400 μg/kg/d, 89.1–95% for 200 μg/kg/d | 154–162 | ||
Pediculus humanus corporis (body louse) | Yes | 200 μg/kg on day 0, 7 and 14 | 78% | 164 | ||
Phtirus pubis (pubic louse) | Yes | 200 μg/kg/d 1–2 weeks apart | 100% | 165 | ||
Cimex lectularius (common bedbug) | Yes | 200 μg/kg once | 67% after 20 d; blood meal 3 h after oral ivermectin: moulting reduced to 0% at 20 d in the same groupc | 171–173 | ||
Cimex hemipterus (tropical bedbug) | Uncleard | Unclear | Unclear | Uncleare | d | |
Demodex sp. | Likely | 200 μg/kg | Unclear | 174–176 | ||
Tunga penetrans | No | 200 μg/kg | 149, 168 | |||
Myiasis (botfly larva) | Unclearf | 200 μg/kg | Unclear | 169, 170d |
aCure rate if not otherwise indicated.
Topical treatment for children <15 kg.
cWithout molting sexual maturity does not occur.
dCircumstantial observation.
eExpected to be similar to C. lectularius.
fRecommended only in conjunction with surgery.
Conclusions
Ivermectin has been the mainstay of onchocerciasis and LF control programmes worldwide. Within the last decade, ivermectin has shown considerable promise for use in a broader range of diseases, in particular for malaria, scabies and as an adjunct for STH control. These diseases have highly overlapping distributions, suggesting that in some circumstances MDA for malaria may also result in additional health and economic benefits through ‘off-target’ effects.
Ongoing and planned malaria control trials utilising ivermectin MDA provide opportunities to explore these potential synergies (Box 1). Incorporating STH and scabies endpoints into these trials should be strongly considered to more fully capture the potential health impacts of these programmes. On the other hand, current onchocerciasis, LF, STH and scabies dosing schedules are unlikely to have significant impacts on mosquito populations or malaria transmission. A key question is whether the platforms can be coordinated alongside newer malaria control efforts to accelerate progress. The expansion of ivermectin use requires careful consideration of the development of resistance in both on- and off-target organisms. Potential environmental problems could also arise from its use in animals for malaria vector control or its impact on non-target insect species.94,96
Box 1.
After >30 y as the mainstay for control and elimination programmes for onchocerciasis and LF there is increasing evidence for a range of expanded indications including scabies and malaria control.
Extended use of ivermectin MDA for malaria vector control has the potential to impact several co-endemic parasites by reducing their burden of disease.
There is a need for exploration of reliable affordable generic supply of ivermectin to support expanded applications for which donations are currently unlikely.
Safety data on use in, at present, excluded populations such as pregnant or breastfeeding women and younger children (<5 y of age) is needed.
In summary, as we enter the decade of the Sustainable Development Goals, it appears the role of ivermectin may be expanding not contracting. Data emerging from recently completed, ongoing and future well-designed clinical trials using ivermectin MDA for malaria control in varied settings, as mentioned in the malaria section, will answer key programmatic questions about its future role in disease control programmes worldwide.
Acknowledgments
None.
Contributor Information
Christian Kositz, Clinical Research Department, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, Keppel Street, WC1E 7HT, London, UK.
John Bradley, MRC International Statistics and Epidemiology Group, Faculty of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine, Keppel Street, WC1E 7HT, London, UK.
Harry Hutchins, Clinical Research Department, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, Keppel Street, WC1E 7HT, London, UK.
Anna Last, Clinical Research Department, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, Keppel Street, WC1E 7HT, London, UK; Hospital for Tropical Diseases, Mortimer Market Capper Street, WC1E 6JB, London, UK.
Umberto D'Alessandro, Disease Control and Elimination, Medical Research Council Unit Gambia at London School of Hygiene and Tropical Medicine, Atlantic Boulevard, Fajara, The Gambia.
Michael Marks, Clinical Research Department, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, Keppel Street, WC1E 7HT, London, UK; Hospital for Tropical Diseases, Mortimer Market Capper Street, WC1E 6JB, London, UK.
Authors’ contributions
CK, JB and MM were responsible for study conceptualization. JB, HH, AL, UD and MM were responsible for review and editing of the manuscript. CK was responsible for original draft preparation. CK, JB, HH, AL, UD and MM were responsible for analysis and interpretation of data and read and approved the final version.
Funding
CK was supported by a Wellcome Trust Clinical Research Fellowship (217357/Z/19/Z). This research was funded in whole or in part by the Wellcome Trust (217357/Z/19/Z). For the purpose of Open Access, the author has applied a CC BY public copyright licence to any author accepted manuscript version arising from this submission.
Competing interests
None declared.
Ethical approval
Not required.
Data availability
No new data were generated or analysed in support of this research.
References
- 1. Omura S, Crump A. The life and times of ivermectin—a success story. Nat Rev Microbiol. 2004;2:(12):984–9. [DOI] [PubMed] [Google Scholar]
- 2. McArthur MJ, Reinemeyer CR. Herding the U.S. cattle industry toward a paradigm shift in parasite control. Vet Parasitol. 2014;204(1–2):34–43. [DOI] [PubMed] [Google Scholar]
- 3. Burgess CGS, Bartley Y, Redman Eet al. A survey of the trichostrongylid nematode species present on UK sheep farms and associated anthelmintic control practices. Vet Parasitol. 2012;189(2–4):299–307. [DOI] [PubMed] [Google Scholar]
- 4. Chabala JC, Mrozik H, Tolman RLet al. Ivermectin, a new broad-spectrum antiparasitic agent. J Med Chem. 1980;23(10):1134–6. [DOI] [PubMed] [Google Scholar]
- 5. Glaziou P, Cartel JL, Alzieu Pet al. Comparison of ivermectin and benzyl benzoate for treatment of scabies. Trop Med Parasitol. 1993;44(4):331–2. [PubMed] [Google Scholar]
- 6. Wilson ML. Avermectins in arthropod vector management—prospects and pitfalls. Parasitol Today. 1993;9(3):83–7. [DOI] [PubMed] [Google Scholar]
- 7. Whitworth JAG, Morgan D, Maude GHet al. A field study of the effect of ivermectin on intestinal helminths in man. Trans R Soc Trop Med Hyg. 1991;85(2):232–4. [DOI] [PubMed] [Google Scholar]
- 8. Heukelbach J, Wilcke T, Winter Bet al. Efficacy of ivermectin in a patient population concomitantly infected with intestinal helminths and ectoparasites. Arzneimittelforschung. 2011;54(7):416–21. [DOI] [PubMed] [Google Scholar]
- 9. Romani L, Whitfeld MJ, Koroivueta Jet al. Mass drug administration for scabies control in a population with endemic disease. N Engl J Med. 2015;373(24):2305–13. [DOI] [PubMed] [Google Scholar]
- 10. Kearns TM, Speare R, Cheng ACet al. Impact of an ivermectin mass drug administration on scabies prevalence in a remote Australian aboriginal community. PLoS Negl Trop Dis. 2015;9(10):e0004151. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Marks M, Gwyn S, Toloka Het al. Impact of community treatment with ivermectin for the control of scabies on the prevalence of antibodies to Strongyloides stercoralis in children. Clin Infect Dis. 2020;71(12):3226–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Chaccour C, Hammann F, Rabinovich NR.. Ivermectin to reduce malaria transmission I. Pharmacokinetic and pharmacodynamic considerations regarding efficacy and safety. Malar J. 2017;16:161. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. Omura S. Mode of action of avermectin. In: Macrolide antibiotics: chemistry, biology and practice. New York: Academic Press, 2002:571–6. [Google Scholar]
- 14. Meyers JI, Gray M, Kuklinski Wet al. Characterization of the target of ivermectin, the glutamate-gated chloride channel, from Anopheles gambiae. J Exp Biol. 2015;218(10):1478–86. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Laing R, Gillan V, Devaney E. Ivermectin – old drug, new tricks? Trends Parasitol. 2017;33(6):463–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Edwards G. Ivermectin: does P-glycoprotein play a role in neurotoxicity? Filaria J. 2003;2(Suppl 1):S8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Juarez M, Schcolnik-Cabrera A, Dueñas-Gonzalez A. The multitargeted drug ivermectin: from an antiparasitic agent to a repositioned cancer drug. Am J Cancer Res. 2018;8(2):317–31. [PMC free article] [PubMed] [Google Scholar]
- 18. Mastrangelo E, Pezzullo M, De Burghgraeve Tet al. Ivermectin is a potent inhibitor of flavivirus replication specifically targeting NS3 helicase activity: new prospects for an old drug. J Antimicrob Chemother. 2012;67(8):1884–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Heidary F, Gharebaghi R. Ivermectin: a systematic review from antiviral effects to COVID-19 complementary regimen. J Antibiot (Tokyo). 2020;73(9):593–602. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. Wolstenholme AJ, Rogers AT. Glutamate-gated chloride channels and the mode of action of the avermectin/milbemycin anthelmintics. Parasitology. 2006;131(Suppl 1):S85. [DOI] [PubMed] [Google Scholar]
- 21. Schinkel AH, Smit JJ, van Tellingen Oet al. Disruption of the mouse mdr1a P-glycoprotein gene leads to a deficiency in the blood-brain barrier and to increased sensitivity to drugs. Cell. 1994;77(4):491–502. [DOI] [PubMed] [Google Scholar]
- 22. Baudou E, Lespine A, Durrieu Get al. Serious ivermectin toxicity and human ABCB1 nonsense mutations. N Engl J Med. 2020;383(8):787–9. [DOI] [PubMed] [Google Scholar]
- 23. Mealey KL, Bentjen SA, Gay JMet al. Ivermectin sensitivity in collies is associated with a deletion mutation of the mdr1 gene. Pharmacogenetics. 2001;11(8):727–33. [DOI] [PubMed] [Google Scholar]
- 24. Dowling P. Pharmacogenetics: it's not just about ivermectin in collies. Can Vet J. 2006;47(12):1165–8. [PMC free article] [PubMed] [Google Scholar]
- 25. Mectizan Donation Program. History of the program. Available from: https://mectizan.org/what/history/ [accessed 8 January 2021]. [Google Scholar]
- 26. Guzzo CA, Furtek CI, Porras AGet al. Safety, tolerability, and pharmacokinetics of escalating high doses of ivermectin in healthy adult subjects. J Clin Pharmacol. 2002;42(10):1122–33. [DOI] [PubMed] [Google Scholar]
- 27. Smit MR, Ochomo EO, Aljayyoussi Get al. Safety and mosquitocidal efficacy of high-dose ivermectin when co-administered with dihydroartemisinin-piperaquine in Kenyan adults with uncomplicated malaria (IVERMAL): a randomised, double-blind, placebo-controlled trial. Lancet Infect Dis. 2018;18(6):615–26. [DOI] [PubMed] [Google Scholar]
- 28. Smit MR, Ochomo EO, Waterhouse Det al. Pharmacokinetics-pharmacodynamics of high-dose ivermectin with dihydroartemisinin-piperaquine on mosquitocidal activity and QT-prolongation (IVERMAL). Clin Pharmacol Ther. 2019;105(2):388–401. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29. Kamgno J, Gardon J, Gardon-Wendel Net al. Adverse systemic reactions to treatment of onchocerciasis with ivermectin at normal and high doses given annually or three-monthly. Trans R Soc Trop Med Hyg. 2004;98(8):496–504. [DOI] [PubMed] [Google Scholar]
- 30. Awadzi K, Attah SK, Addy ETet al. The effects of high-dose ivermectin regimens on Onchocerca volvulus in onchocerciasis patients. Trans R Soc Trop Med Hyg. 1999;93(2):189–94. [DOI] [PubMed] [Google Scholar]
- 31. Awadzi K, Opoku NO, Addy ETet al. The chemotherapy of onchocerciasis. XIX: the clinical and laboratory tolerance of high dose ivermectin. Trop Med Parasitol. 1995;46(2):131–7. [PubMed] [Google Scholar]
- 32. Gardon J, Kamgno J, Gardon-Wendel Net al. Efficacy of repeated doses of ivermectin against Mansonella perstans. Trans R Soc Trop Med Hyg. 2002;96(3):325–6. [DOI] [PubMed] [Google Scholar]
- 33. Kamgno J, Boussinesq M, Labrousse Fet al. Encephalopathy after ivermectin treatment in a patient infected with Loa loa and Plasmodium spp. Am J Trop Med Hyg. 2008;78(4):546–51. [PubMed] [Google Scholar]
- 34. Boussinesq M, Gardon J, Gardon-Wendel Net al. Clinical picture, epidemiology and outcome of Loa-associated serious adverse events related to mass ivermectin treatment of onchocerciasis in Cameroon. Filaria J. 2003;2(Suppl 1):S4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Nicolas P, Maia MF, Bassat Qet al. Safety of oral ivermectin during pregnancy: a systematic review and meta-analysis. Lancet Glob Health. 2020;8(1):e92–100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Gyapong JO, Chinbuah MA, Gyapong M. Inadvertent exposure of pregnant women to ivermectin and albendazole during mass drug administration for lymphatic filariasis. Trop Med Int Health. 2003;8(12):1093–101. [DOI] [PubMed] [Google Scholar]
- 37. Lankas GR, Wise LD, Cartwright MEet al. Placental P-glycoprotein deficiency enhances susceptibility to chemically induced birth defects in mice. Reprod Toxicol. 1998;12(4):457–63. [DOI] [PubMed] [Google Scholar]
- 38. Merck & Co. Stromectrol (Ivermectin) . Package insert. Available from: https://www.accessdata.fda.gov/drugsatfda_docs/label/2009/050742s026lbl.pdf [accessed 3 January 2020]. [Google Scholar]
- 39.US Food and Drug Administration Center for Drug Evaluation and Research. Approval package for Mectizan. Available from: https://www.accessdata.fda.gov/drugsatfda_docs/nda/96/050742ap.pdf [accessed 25 June 2021]. [Google Scholar]
- 40. Campbell WC (ed.). Ivermectin and abamectin. New York: Springer, 1989. Available from: 10.1007/978-1-4612-3626-9 [accessed 16 February 2021]. [DOI] [Google Scholar]
- 41. Saunders NR, Dreifuss J-J, Dziegielewska KMet al. The rights and wrongs of blood-brain barrier permeability studies: a walk through 100 years of history. Front Neurosci. 2014;8:404. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42. Ek CJ, Dziegielewska KM, Habgood MDet al. Barriers in the developing brain and neurotoxicology. NeuroToxicology. 2012;33(3):586–604. [DOI] [PubMed] [Google Scholar]
- 43. Lam J, Baello S, Iqbal Met al. The ontogeny of P-glycoprotein in the developing human blood–brain barrier: implication for opioid toxicity in neonates. Pediatr Res. 2015;78(4):417–21. [DOI] [PubMed] [Google Scholar]
- 44. Levy M, Martin L, Bursztejn A-Cet al. Ivermectin safety in infants and children under 15 kg treated for scabies: a multicentric observational study. Br J Dermatol. 2020;182(4):1003–6. [DOI] [PubMed] [Google Scholar]
- 45. Colebunders R, Wafula ST, Hotterbeekx Aet al. Ivermectin use in children below 15 kg: potential benefits for onchocerciasis and scabies elimination programmes. Br J Dermatol. 2020;182(4):1064. [DOI] [PubMed] [Google Scholar]
- 46. Morris-Jones R. Oral ivermectin for infants and children under 15 kg appears to be a safe and effective treatment for scabies. Br J Dermatol. 2020;182(4):835–6. [DOI] [PubMed] [Google Scholar]
- 47. Wilkins AL, Steer AC, Cranswick Net al. Question 1: is it safe to use ivermectin in children less than five years of age and weighing less than 15 kg? Arch Dis Child. 2018;103(5):514–9. [DOI] [PubMed] [Google Scholar]
- 48. Chosidow A, Gendrel D.. Tolérance de l'ivermectine orale chez l'enfant. Arch Pédiatrie. 2016;23(2):204–9. [DOI] [PubMed] [Google Scholar]
- 49. Brussee JM, Schulz JD, Coulibaly JTet al. Ivermectin dosing strategy to achieve equivalent exposure coverage in children and adults. Clin Pharmacol Ther. 2019;106(3):661–7. [DOI] [PubMed] [Google Scholar]
- 50. Jittamala P, Monteiro W, Smit MRet al. A systematic review and an individual patient data meta-analysis of ivermectin use in children weighing less than fifteen kilograms: is it time to reconsider the current contraindication? PLoS Negl Trop Dis. 2021;15(3):e0009144. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51. World Health Organization . World malaria report 2019. Geneva: World Health Organization, 2019. [Google Scholar]
- 52. Thomsen EK, Koimbu G, Pulford Jet al. Mosquito behavior change after distribution of bednets results in decreased protection against malaria exposure. J Infect Dis. 2017;215(5):790–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53. Charlwood JD, Kessy E, Yohannes Ket al. Studies on the resting behaviour and host choice of Anopheles gambiae and An. arabiensis from Muleba, Tanzania: resting in Anopheles gambiae s.l. Med Vet Entomol. 2018;32(3):263–70. [DOI] [PubMed] [Google Scholar]
- 54. World Health Organization . Insecticide resistance. Available from: http://www.who.int/malaria/areas/vector_control/insecticide_resistance/en/ [accessed 18 January 2021]. [Google Scholar]
- 55. Alout H, Foy BD. Ivermectin: a complimentary weapon against the spread of malaria? Expert Rev Anti Infect Ther. 2017;15(3):231–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56. Chaccour CJ, Kobylinski KC, Bassat Qet al. Ivermectin to reduce malaria transmission: a research agenda for a promising new tool for elimination. Malar J. 2013;12:153. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57. Deus KM, Saavedra-Rodriguez K, Butters MPet al. The effect of ivermectin in seven strains of Aedes aegypti (Diptera: Culicidae) including a genetically diverse laboratory strain and three permethrin resistant strains. J Med Entomol. 2012;49(2):356–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58. Kobylinski KC, Deus KM, Butters MPet al. The effect of oral anthelmintics on the survivorship and re-feeding frequency of anthropophilic mosquito disease vectors. Acta Trop. 2010;116(2):119–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59. Meyers JI, Gray M, Foy BD. Mosquitocidal properties of IgG targeting the glutamate-gated chloride channel in three mosquito disease vectors (Diptera: Culicidae). J Exp Biol. 2015;218(10):1487–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60. Bockarie MJ, Hii JL, Alexander NDet al. Mass treatment with ivermectin for filariasis control in Papua New Guinea: impact on mosquito survival. Med Vet Entomol. 1999;13(2):120–3. [DOI] [PubMed] [Google Scholar]
- 61. Simonsen PE, Pedersen EM, Rwegoshora RTet al. Lymphatic filariasis control in Tanzania: effect of repeated mass drug administration with ivermectin and albendazole on infection and transmission. PLoS Negl Trop Dis. 2010;4(6):e696. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62. Gardner K, Meisch MV, Meek CLet al. Effects of ivermectin in canine blood on Anopheles quadrimaculatus, Aedes albopictus and Culex salinarius. J Am Mosq Control Assoc. 1993;9(4):400–2. [PubMed] [Google Scholar]
- 63. Chaccour C, Lines J, Whitty CJM. Effect of ivermectin on Anopheles gambiae mosquitoes fed on humans: the potential of oral insecticides in malaria control. J Infect Dis. 2010;202(1):113–6. [DOI] [PubMed] [Google Scholar]
- 64. Sampaio VS, Beltrán TP, Kobylinski KCet 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65. Kobylinski KC, Escobedo-Vargas KS, López-Sifuentes VMet al. Ivermectin susceptibility, sporontocidal effect, and inhibition of time to re-feed in the Amazonian malaria vector Anopheles darlingi. Malar J. 2017;16:474. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66. Kobylinski KC, Ubalee R, Ponlawat Aet al. Ivermectin susceptibility and sporontocidal effect in Greater Mekong subregion Anopheles. Malar J. 2017;16:280. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67. Dreyer SM, Morin KJ, Vaughan JA.. Differential susceptibilities of Anopheles albimanus and Anopheles stephensi mosquitoes to ivermectin. Malar J. 2018;17:148. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68. Kositz C, Talina J, Diau Jet al. Incidental mosquitocidal effect of an ivermectin mass drug administration on Anopheles farauti conducted for scabies control in the Solomon Islands. Trans R Soc Trop Med Hyg. 2017;111(3):97–101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69. Sylla M, Kobylinski KC, Gray Met 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70. Smit MR, Ochomo EO, Aljayyoussi Get al. Human direct skin feeding versus membrane feeding to assess the mosquitocidal efficacy of high-dose ivermectin (IVERMAL trial). Clin Infect Dis. 2019;69(7):1112–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71. Chaccour CJ, Rabinovich NR, Slater Het al. Establishment of the ivermectin research for malaria elimination network: updating the research agenda. Malar J. 2015;14:243. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72. Slater HC, Foy BD, Kobylinski Ket al. Ivermectin as a novel complementary malaria control tool to reduce incidence and prevalence: a modelling study. Lancet Infect Dis. 2020;20(4):498–508. [DOI] [PubMed] [Google Scholar]
- 73. Smit MR, Ochomo E, Aljayyoussi Get al. Efficacy and safety of high-dose ivermectin for reducing malaria transmission (IVERMAL): protocol for a double-blind, randomized, placebo-controlled, dose-finding trial in western Kenya. JMIR Res Protoc. 2016;5(4):e213. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74. Foy BD, Alout H, Seaman JAet al. Efficacy and risk of harms of repeat ivermectin mass drug administrations for control of malaria (RIMDAMAL): a cluster-randomised trial. Lancet. 2019;393(10180):1517–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75. Bradley J, Moulton LH, Hayes R. Analysis of the RIMDAMAL trial. Lancet. 2019;394(10203):1005–6. [DOI] [PubMed] [Google Scholar]
- 76. Foy BD, Rao S, Parikh Set al. Analysis of the RIMDAMAL trial – authors’ reply. Lancet. 2019;394 (10203):1006–7. [DOI] [PubMed] [Google Scholar]
- 77. Dabira ED, Soumare HM, Lindsay SWet al. Mass drug administration with high-dose ivermectin and dihydroartemisinin-piperaquine for malaria elimination in an area of low transmission with high coverage of malaria control interventions: protocol for the MASSIV cluster randomized clinical trial. JMIR Res Protoc. 2020;9(11):e20904. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78. Pasay CJ, Yakob L, Meredith HRet al. Treatment of pigs with endectocides as a complementary tool for combating malaria transmission by Anopheles farauti (s.s.) in Papua New Guinea. Parasit Vectors. 2019;12:124. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79. Fornadel CM, Norris LC, Glass GEet al. Analysis of Anopheles arabiensis blood feeding behavior in southern Zambia during the two years after introduction of insecticide-treated bed nets. Am J Trop Med Hyg. 2010;83(4):848–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80. Waite JL, Swain S, Lynch PAet al. Increasing the potential for malaria elimination by targeting zoophilic vectors. Sci Rep. 2017;7:40551. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81. Dreyer SM, Leiva D, Magaña Met al. Fipronil and ivermectin treatment of cattle reduced the survival and ovarian development of field-collected Anopheles albimanus in a pilot trial conducted in northern Belize. Malar J. 2019;18:296. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82. Pooda HS, Rayaisse J-B, de Sale Hien DFet al. Administration of ivermectin to peridomestic cattle: a promising approach to target the residual transmission of human malaria. Malar J. 2015;14:496. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83. Lyimo IN, Kessy ST, Mbina KFet al. 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84. Chaccour C, Abizanda G, Irigoyen Áet al. Pilot study of a slow-release ivermectin formulation for malaria control in a pig model. Antimicrob Agents Chemother. 2017;61(3):e02104–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85. Lifschitz A, Virkel G, Ballent Met al. Ivermectin (3.15%) long-acting formulations in cattle: absorption pattern and pharmacokinetic considerations. Vet Parasitol. 2007;147(3–4):303–10. [DOI] [PubMed] [Google Scholar]
- 86. Lifschitz A, Pis A, Alvarez Let al. Bioequivalence of ivermectin formulations in pigs and cattle. J Vet Pharmacol Ther. 1999;22(1):27–34. [DOI] [PubMed] [Google Scholar]
- 87. Pooda SH, Mouline K, De Meeûs Tet al. Decrease in survival and fecundity of Glossina palpalis gambiensis vanderplank 1949 (Diptera: Glossinidae) fed on cattle treated with single doses of ivermectin. Parasit Vectors. 2013;6:165. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88. Langley PA, Roe JM. Ivermectin as a possible control agent for the tsetse fly, Glossina morsitans. Entomol Exp Appl. 1984;36(2):137–43. [Google Scholar]
- 89. van den Abbeele J, D'Haeseleer F, Goossens M. Efficacy of ivermectin on the reproductive biology of Glossina palpalis palpalis (Rob.-Desv.) (Glossinidae: Diptera). Ann Soc Belg Med Trop. 1986;66:167–72. [PubMed] [Google Scholar]
- 90. Dias JCP, Schofield CJ, Machado EMet al. Ticks, ivermectin, and experimental Chagas disease. Mem Inst Oswaldo Cruz. 2005;100(8):829–32. [DOI] [PubMed] [Google Scholar]
- 91. Chaccour C, Killeen GF. Mind the gap: residual malaria transmission, veterinary endectocides and livestock as targets for malaria vector control. Malar J. 2016;15:24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92. Ivermectin Roadmappers, Billingsley P, Binka Fet al. A roadmap for the development of ivermectin as a complementary malaria vector control tool. Am J Trop Med Hyg. 2020;102(2 Suppl):3–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93. Chaccour C. Veterinary endectocides for malaria control and elimination: prospects and challenges. Phil Trans R Soc Lond B Biol Sci. 2021;376(1818):20190810. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94. Mancini L, Lacchetti I, Chiudioni Fet al. Need for a sustainable use of medicinal products: environmental impacts of ivermectin. Ann Ist Super Sanita. 2020;56:492–6. [DOI] [PubMed] [Google Scholar]
- 95. Liebig M, Fernandez ÁA, Blübaum-Gronau Eet al. Environmental risk assessment of ivermectin: a case study. Integr Environ Assess Manag. 2010;6(Suppl):567–87. [DOI] [PubMed] [Google Scholar]
- 96. Ishikawa I, Iwasa M.. Toxicological effect of ivermectin on the survival, reproduction, and feeding activity of four species of dung beetles (Coleoptera: Scarabaeidae and Geotrupidae) in Japan. Bull Entomol Res. 2020;110(1):106–14. [DOI] [PubMed] [Google Scholar]
- 97. Pecenka JR, Lundgren JG.. Effects of herd management and the use of ivermectin on dung arthropod communities in grasslands. Basic Appl Ecol. 2019;40:19–29. [Google Scholar]
- 98. Bloom RA, Matheson JC.. Environmental assessment of avermectins by the US Food and Drug Administration. Vet Parasitol. 1993;48(1–4):281–94. [DOI] [PubMed] [Google Scholar]
- 99. Singer R, Xu TH, Herrera LNSet al. Prevalence of intestinal parasites in a low-income Texas community. Am J Trop Med Hyg. 2020;102(6):1386–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100. McKenna ML, McAtee S, Bryan PEet al. Human intestinal parasite burden and poor sanitation in rural Alabama. Am J Trop Med Hyg. 2017;97(5):1623–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101. Hotez PJ, Molyneux DH, Fenwick Aet al. Control of neglected tropical diseases. N Engl J Med. 2007;357(10):1018–27. [DOI] [PubMed] [Google Scholar]
- 102. Guideline: preventive chemotherapy to control soil-transmitted helminth infections in at-risk population groups. Geneva: World Health Organization, 2017. Available from: http://www.ncbi.nlm.nih.gov/books/NBK487927/ [accessed 25 June 2021]. [PubMed] [Google Scholar]
- 103. Marti H, Haji HJ, Savioli Let al. A comparative trial of a single-dose ivermectin versus three days of albendazole for treatment of Strongyloides stercoralis and other soil-transmitted helminth infections in children. Am J Trop Med Hyg. 1996;55(5):477–81. [DOI] [PubMed] [Google Scholar]
- 104. Campbell WC. Ivermectin as an antiparasitic agent for use in humans. Annu Rev Microbiol. 1991;45:445–74. [DOI] [PubMed] [Google Scholar]
- 105. Wen L-Y, Yan X-L, Sun F-Het al. A randomized, double-blind, multicenter clinical trial on the efficacy of ivermectin against intestinal nematode infections in China. Acta Trop. 2008;106(3):190–4. [DOI] [PubMed] [Google Scholar]
- 106. Freedman DO, Zierdt WS, Lujan Aet al. The efficacy of ivermectin in the chemotherapy of gastrointestinal helminthiasis in humans. J Infect Dis. 1989;159(6):1151–3. [DOI] [PubMed] [Google Scholar]
- 107. Buonfrate D, Salas-Coronas J, Muñoz Jet al. Multiple-dose versus single-dose ivermectin for Strongyloides stercoralis infection (Strong Treat 1 to 4): a multicentre, open-label, phase 3, randomised controlled superiority trial. Lancet Infect Dis. 2019;19(11):1181–90. [DOI] [PubMed] [Google Scholar]
- 108. Suputtamongkol Y, Premasathian N, Bhumimuang Ket al. Efficacy and safety of single and double doses of ivermectin versus 7-day high dose albendazole for chronic strongyloidiasis. PLoS Negl Trop Dis. 2011;5(5):e1044. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109. Naquira C, Jimenez G, Guerra JGet al. Ivermectin for human strongyloidiasis and other intestinal helminths. Am J Trop Med Hyg. 1989;40(3):304–9. [DOI] [PubMed] [Google Scholar]
- 110. Wimmersberger D, Coulibaly JT, Schulz JDet al. Efficacy and safety of ivermectin against Trichuris trichiura in preschool-aged and school-aged children: a randomized controlled dose-finding trial. Clin Infect Dis. 2018;67(8):1247–55. [DOI] [PubMed] [Google Scholar]
- 111. Betson M, Søe MJ, Nejsum P. Human trichuriasis: whipworm genetics, phylogeny, transmission and future research directions. Curr Trop Med Rep. 2015;2(4):209–17. [Google Scholar]
- 112. Vanhaecke C, Perignon A, Monsel Get al. The efficacy of single dose ivermectin in the treatment of hookworm related cutaneous larva migrans varies depending on the clinical presentation. J Eur Acad Dermatol Venereol. 2014;28(5):655–7. [DOI] [PubMed] [Google Scholar]
- 113. Caumes E. Treatment of cutaneous larva migrans. Clin Infect Dis. 2000;30(5):811–4. [DOI] [PubMed] [Google Scholar]
- 114. Ottesen EA, Hooper PJ, Bradley Met al. The global programme to eliminate lymphatic filariasis: health impact after 8 years. PLoS Negl Trop Dis. 2008;2(10):e317. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115. World Health Organization . Progress Report 2000–2009 and Strategic Plan 2010–2020 of the Global Programme to Eliminate Lymphatic Filariasis. Geneva: World Health Organization, 2010. [Google Scholar]
- 116. World Health Organization . Onchocerciasis. Geneva: World Health Organization. Available from: https://www.who.int/news-room/fact-sheets/detail/onchocerciasis [accessed 1 May 2020]. [Google Scholar]
- 117. Traore MO, Sarr MD, Badji Aet al. Proof-of-principle of onchocerciasis elimination with ivermectin treatment in endemic foci in Africa: final results of a study in Mali and Senegal. PLoS Negl Trop Dis. 2012;6(9):e1825. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118. Basáñez M-G, Pion SD, Boakes Eet al. Effect of single-dose ivermectin on Onchocerca volvulus: a systematic review and meta-analysis. Lancet Infect Dis. 2008;8(5):310–22. [DOI] [PubMed] [Google Scholar]
- 119. Walker M, Pion SDS, Fang Het al. Macrofilaricidal efficacy of repeated doses of ivermectin for the treatment of river blindness. Clin Infect Dis. 2017;65(12):2026–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120. Bjerum CM, Ouattara AF, Aboulaye Met al. Efficacy and safety of a single dose of ivermectin, diethylcarbamazine, and albendazole for treatment of lymphatic filariasis in Côte d'Ivoire: an open-label randomized controlled trial. Clin Infect Dis. 2020;71(7):e68–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121. Horton J, Witt C, Ottesen EAet al. An analysis of the safety of the single dose, two drug regimens used in programmes to eliminate lymphatic filariasis. Parasitology. 2000;121(Suppl 1):S147–60. [DOI] [PubMed] [Google Scholar]
- 122. Thomsen EK, Sanuku N, Baea Met al. Efficacy, safety, and pharmacokinetics of coadministered diethylcarbamazine, albendazole, and ivermectin for treatment of bancroftian filariasis. Clin Infect Dis. 2016;62(3):334–41. [DOI] [PubMed] [Google Scholar]
- 123. Dubray CL, Sircar AD, de Rochars VMBet al. Safety and efficacy of co-administered diethylcarbamazine, albendazole and ivermectin during mass drug administration for lymphatic filariasis in Haiti: results from a two-armed, open-label, cluster-randomized, community study. PLoS Negl Trop Dis. 2020;14(6):e0008298. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124. Hardy M, Samuela J, Kama Met al. The safety of combined triple drug therapy with ivermectin, diethylcarbamazine and albendazole in the neglected tropical diseases co-endemic setting of Fiji: a cluster randomised trial. PLoS Negl Trop Dis. 2020;14(3):e0008106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125. Pion SD, Tchatchueng-Mbougua JB, Chesnais CBet al. Effect of a single standard dose (150–200 μg/kg) of ivermectin on Loa loa microfilaremia: systematic review and meta-analysis. Open Forum Infect Dis. 2019;6(4):ofz019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126. Chippaux J-P, Boussinesq M, Gardon Jet al. Severe adverse reaction risks during mass treatment with ivermectin in loiasis-endemic areas. Parasitol Today. 1996;12(11):448–50. [DOI] [PubMed] [Google Scholar]
- 127. Fischer P, Tukesiga E, Büttner DW. Long-term suppression of Mansonella streptocerca microfilariae after treatment with ivermectin. J Infect Dis. 1999;180(4):1403–5. [DOI] [PubMed] [Google Scholar]
- 128. Ta-Tang T-H, Crainey JL, Post RJet al. Mansonellosis: current perspectives. Res Rep Trop Med. 2018;9:9–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129. de Almeida Basano S, de Souza Almeida Aranha Camargo J, Fontes Get al. Phase III clinical trial to evaluate ivermectin in the reduction of Mansonella ozzardi infection in the Brazilian Amazon. Am J Trop Med Hyg. 2018;98(3):786–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130. Medeiros JF, Vera LJS, Crispim Pet al. Sustained clearance of Mansonella ozzardi infection after treatment with ivermectin in the Brazilian Amazon. Am J Trop Med Hyg. 2014;90(6):1170–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131. Richard-Lenoble D, Kombila M, Rupp EAet al. Ivermectin in loiasis and concomitant O. volvulus and M. perstans infections. Am J Trop Med Hyg. 1988;39(5):480–3. [DOI] [PubMed] [Google Scholar]
- 132. Bregani ER, Rovellini A, Mbaïdoum Net al. Comparison of different anthelminthic drug regimens against Mansonella perstans filariasis. Trans R Soc Trop Med Hyg. 2006;100(5):458–63. [DOI] [PubMed] [Google Scholar]
- 133. van den Enden E, van Gompel A, van der Stuyft Pet al. Treatment failure of a single high dose of ivermectin for Mansonella perstans filariasis. Trans R Soc Trop Med Hyg. 1993;87(1):90. [DOI] [PubMed] [Google Scholar]
- 134. Asio SM, Simonsen PE, Onapa AW. A randomised, double-blind field trial of ivermectin alone and in combination with albendazole for the treatment of Mansonella perstans infections in Uganda. Trans R Soc Trop Med Hyg. 2009;103(3):274–9. [DOI] [PubMed] [Google Scholar]
- 135. Kyelem D, Sanou S, Boatin Bet al. Impact of long-term ivermectin (Mectizan®) on Wuchereria bancrofti and Mansonella perstans infections in Burkina Faso: strategic and policy implications. Ann Trop Med Parasitol. 2003;97(8):827–38. [DOI] [PubMed] [Google Scholar]
- 136. Reeves WK, Nol P, Miller MMet al. Effects of ivermectin on the susceptibility of Culicoides sonorensis (Diptera: Ceratopogonidae) to bluetongue and epizootic hemorrhagic disease viruses. J Vector Ecol. 2009;34(1):161–3. [DOI] [PubMed] [Google Scholar]
- 137. Murchie AK, Thompson GM, Clawson Set al. Field evaluation of deltamethrin and ivermectin applications to cattle on culicoides host alighting, blood-feeding, and emergence. Viruses. 2019;11(8):731. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138. Herman JS, Chiodini PL. Gnathostomiasis, another emerging imported disease. Clin Microbiol Rev. 2009;22(3):484–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139. Kraivichian K, Nuchprayoon S, Sitichalernchai Pet al. Treatment of cutaneous gnathostomiasis with ivermectin. Am J Trop Med Hyg. 2004;71(5):623–8. [PubMed] [Google Scholar]
- 140. Soliman GA, Taher ES, Mahmoud MA. Therapeutic efficacy of dormectin, ivermectin and levamisole against different stages of Trichinella spiralis in rats. Turk J Parasitol. 2011;35(2):86–91. [DOI] [PubMed] [Google Scholar]
- 141. Campbell WC, Blair LS, Lotti VJ. Efficacy of avermectins against Trichinella spiralis in mice. J Helminthol. 1979;53(3):254–6. [DOI] [PubMed] [Google Scholar]
- 142. Hay RJ, Steer AC, Engelman Det al. Scabies in the developing world—its prevalence, complications, and management. Clin Microbiol Infect. 2012;18(4):313–23. [DOI] [PubMed] [Google Scholar]
- 143. Lawrence G, Leafasia J, Sheridan Jet al. Control of scabies, skin sores and haematuria in children in the Solomon Islands: another role for ivermectin. Bull World Health Org. 2005;83:34–42. [PMC free article] [PubMed] [Google Scholar]
- 144. Chung S-D, Wang K-H, Huang C-Cet al. Scabies increased the risk of chronic kidney disease: a 5-year follow-up study. J Eur Acad Dermatol Venereol. 2014;28(3):286–92. [DOI] [PubMed] [Google Scholar]
- 145. Mounsey K, Ho M-F, Kelly Aet al. A tractable experimental model for study of human and animal scabies. PLoS Negl Trop Dis. 2010;4(7):e756. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146. Chouela EN, Abeldaño AM, Pellerano Get al. Equivalent therapeutic efficacy and safety of ivermectin and lindane in the treatment of human scabies. Arch Dermatol. 1999;135(6):651–5. [DOI] [PubMed] [Google Scholar]
- 147. Mounsey KE, Holt DC, McCarthy JSet al. Longitudinal evidence of increasing in vitro tolerance of scabies mites to ivermectin in scabies endemic communities. Arch Dermatol. 2009;145(7):840–1. [DOI] [PubMed] [Google Scholar]
- 148. Romani L, Marks M, Sokana Oet al. Efficacy of mass drug administration with ivermectin for control of scabies and impetigo, with coadministration of azithromycin: a single-arm community intervention trial. Lancet Infect Dis. 2019;19(5):510–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149. Heukelbach J, Winter B, Wilcke Tet al. Selective mass treatment with ivermectin to control intestinal helminthiases and parasitic skin diseases in a severely affected population. Bull World Health Org. 2004;82:563–71. [PMC free article] [PubMed] [Google Scholar]
- 150. Amanzougaghene N, Fenollar F, Raoult Det al. Where are we with human lice? A review of the current state of knowledge. Front Cell Infect Microbiol. 2019;9:474. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151. Burgess IF. Human lice and their management. Adv Parasitol. 1995;36:271–342. [DOI] [PubMed] [Google Scholar]
- 152. Burke S, Mir P. Pediculosis causing iron deficiency anaemia in school children. Arch Dis Child. 2011;96(10):989. [DOI] [PubMed] [Google Scholar]
- 153. Heukelbach J, Wilcke T, Winter Bet al. Epidemiology and morbidity of scabies and pediculosis capitis in resource-poor communities in Brazil. Br J Dermatol. 2005;153(1):150–6. [DOI] [PubMed] [Google Scholar]
- 154. Coates SJ, Thomas C, Chosidow Oet al. Ectoparasites. J Am Acad Dermatol. 2020;82(3):551–69. [DOI] [PubMed] [Google Scholar]
- 155. Chosidow O, Giraudeau B, Cottrell Jet al. Oral ivermectin versus malathion lotion for difficult-to-treat head lice. N Engl J Med. 2010;362(10):896–905. [DOI] [PubMed] [Google Scholar]
- 156. Pilger D, Heukelbach J, Khakban Aet al. Household-wide ivermectin treatment for head lice in an impoverished community: randomized observer-blinded controlled trial. Bull World Health Org. 2010;88(2):90–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157. Ahmad HM, Abdel-Azim ES, Abdel-Aziz RT. Assessment of topical versus oral ivermectin as a treatment for head lice: topical versus oral ivermectin for head lice. Dermatol Ther. 2014;27(5):307–10. [DOI] [PubMed] [Google Scholar]
- 158. Ameen M, Arenas R, Villanueva-Reyes Jet al. Oral ivermectin for treatment of pediculosis capitis. Pediatr Infect Dis J. 2010;29(11):991–3. [PubMed] [Google Scholar]
- 159. Nofal A. Oral ivermectin for head lice: a comparison with 0.5% topical malathion lotion: oral ivermectin vs. malathion lotion for head lice. JDDG J Dtsch Dermatol Ges. 2010;8:985–8. [DOI] [PubMed] [Google Scholar]
- 160. Coscione S, Esau T, Kekeubata Eet al. Impact of ivermectin administered for scabies treatment on the prevalence of head lice in Atoifi, Solomon Islands. PLoS Negl Trop Dis. 2018;12(9):e0006825. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161. Singhasivanon O, Lawpoolsri S, Mungthin Met al. Prevalence and alternative treatment of head lice infestation in rural Thailand: a community-based study. Korean J Parasitol. 2019;57(5):499–504. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162. Leulmi H, Diatta G, Sokhna Cet al. Assessment of oral ivermectin versus shampoo in the treatment of pediculosis (head lice infestation) in rural areas of Sine-Saloum, Senegal. Int J Antimicrob Agents. 2016;48(6):627–32. [DOI] [PubMed] [Google Scholar]
- 163. Amanzougaghene N, Fenollar F, Diatta Get al. Mutations in GluCl associated with field ivermectin-resistant head lice from Senegal. Int J Antimicrob Agents. 2018;52(5):593–8. [DOI] [PubMed] [Google Scholar]
- 164. Foucault C, Ranque S, Badiaga Set al. Oral ivermectin in the treatment of body lice. J Infect Dis. 2006;193(3):474–6. [DOI] [PubMed] [Google Scholar]
- 165. Salavastru CM, Chosidow O, Janier Met al. European guideline for the management of pediculosis pubis. J Eur Acad Dermatol Venereol. 2017;31(9):1425–8. [DOI] [PubMed] [Google Scholar]
- 166. Amanzougaghene N, Fenollar F, Nappez Cet al. Complexin in ivermectin resistance in body lice. PLoS Genet. 2018;14(8):e1007569. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167. Munirathinam A, Sunish IP, Rajendran Ret al. Impact of ivermectin drug combinations on Pediculus humanus capitis infestation in primary schoolchildren of south Indian rural villages. Int J Dermatol. 2009;48(11):1201–5. [DOI] [PubMed] [Google Scholar]
- 168. Heukelbach J, Franck S, Feldmeier H. Therapy of tungiasis: a double-blinded randomized controlled trial with oral ivermectin. Mem Inst Oswaldo Cruz. 2004;99(8):873–6. [DOI] [PubMed] [Google Scholar]
- 169. Shinohara EH, Martini MZ, de Oliveira Neto HGet al. Oral myiasis treated with ivermectin: case report. Braz Dent J. 2004;15(1):79–81. [DOI] [PubMed] [Google Scholar]
- 170. Osorio J, Moncada L, Molano Aet al. Role of ivermectin in the treatment of severe orbital myiasis due to Cochliomyia hominivorax. Clin Infect Dis. 2006;43(6):e57–9. [DOI] [PubMed] [Google Scholar]
- 171. Sheele JM, Anderson JF, Tran TDet al. Ivermectin causes Cimex lectularius (bedbug) morbidity and mortality. J Emerg Med. 2013;45(3):433–40. [DOI] [PubMed] [Google Scholar]
- 172. Baraka GT, Nyundo BA, Thomas Aet al. Susceptibility status of bedbugs (Hemiptera: Cimicidae) against pyrethroid and organophosphate insecticides in Dar es Salaam, Tanzania. J Med Entomol. 2020;57(2):524–8. [DOI] [PubMed] [Google Scholar]
- 173. Dang K, Doggett SL, Veera Singham Get al. Insecticide resistance and resistance mechanisms in bed bugs, Cimex spp. (Hemiptera: Cimicidae). Parasit Vectors. 2017;10:318. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174. Brown M, Hernández-Martín A, Clement Aet al. Severe demodex folliculorum–associated oculocutaneous rosacea in a girl successfully treated with ivermectin. JAMA Dermatol. 2014;150(1):61–3. [DOI] [PubMed] [Google Scholar]
- 175. Salem DA-B, El-Shazly A, Nabih Net al. Evaluation of the efficacy of oral ivermectin in comparison with ivermectin-metronidazole combined therapy in the treatment of ocular and skin lesions of Demodex folliculorum. Int J Infect Dis. 2013;17(5):e343–7. [DOI] [PubMed] [Google Scholar]
- 176. Chen W, Plewig G. Human demodicosis: revisit and a proposed classification. Br J Dermatol. 2014;170(6):1219–25. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Data Availability Statement
No new data were generated or analysed in support of this research.