Skip to main content

Onchocerca volvulus microfilariae in the anterior chambers of the eye and ocular adverse events after a single dose of 8 mg moxidectin or 150 µg/kg ivermectin: results of a randomized double-blind Phase 3 trial in the Democratic Republic of the Congo, Ghana and Liberia

Abstract

Background

After ivermectin became available, diethylcarbamazine (DEC) use was discontinued because of severe adverse reactions, including ocular reactions, in individuals with high Onchocerca volvulus microfilaridermia (microfilariae/mg skin, SmfD). Assuming long-term ivermectin use led to < 5 SmfD with little or no eye involvement, DEC + ivermectin + albendazole treatment a few months after ivermectin was proposed. In 2018, the US FDA approved moxidectin for treatment of O. volvulus infection. The Phase 3 study evaluated SmfD, microfilariae in the anterior chamber (mfAC) and adverse events (AEs) in ivermectin-naïve individuals with ≥ 10 SmfD after 8 mg moxidectin (n = 978) or 150 µg/kg ivermectin (n = 494) treatment.

Methods

We analyzed the data from 1463 participants with both eyes evaluated using six (0, 1–5, 6–10, 11–20, 21–40, > 40) mfAC and three pre-treatment (< 20, 20 to < 50, ≥ 50) and post-treatment (0, > 0–5, > 5) SmfD categories. A linear mixed model evaluated factors and covariates impacting mfAC levels. Ocular AEs were summarized by type and start post-treatment. Logistic models evaluated factors and covariates impacting the risk for ocular AEs.

Results

Moxidectin and ivermectin had the same effect on mfAC levels. These increased from pre-treatment to Day 4 and Month 1 in 20% and 16% of participants, respectively. Six and 12 months post-treatment, mfAC were detected in ≈5% and ≈3% of participants, respectively. Ocular Mazzotti reactions occurred in 12.4% of moxidectin- and 10.2% of ivermectin-treated participants without difference in type or severity. The risk for ≥ 1 ocular Mazzotti reaction increased for women (OR 1.537, 95% CI 1.096–2.157) and with mfAC levels pre- and 4 days post-treatment (OR 0: > 10 mfAC 2.704, 95% CI 1.27–5.749 and 1.619, 95% CI 0.80–3.280, respectively).

Conclusions

The impact of SmfD and mfAC levels before and early after treatment on ocular AEs needs to be better understood before making decisions on the risk-benefit of strategies including DEC. Such decisions should take into account interindividual variability in SmfD, mfAC levels and treatment response and risks to even a small percentage of individuals.

Graphical Abstract

Background

Onchocerciasis is a vector-borne parasitic disease caused by the parasite Onchocerca volvulus, transmitted by Simulium black flies. The morbidity, due primarily to immunological reactions of the body to the dead microfilariae in the skin and the eyes, and resulting socio-economic effects, have motivated large-scale control and elimination programmes in sub-Saharan Africa, Yemen and the Americas [1,2,3,4,5,6,7]. Today, control and elimination programmes are based on mass drug administration of ivermectin (MDAi).

In 2003, an international consultation concluded that parasite elimination through MDAi was feasible in the six Central and South American countries of the Americas (total population at risk around 0.56 million), but not across the vast and partly hyperendemic areas in sub-Saharan Africa [8], requiring research for new drugs or drug combinations [9, 10]. Since then, onchocerciasis has been eliminated in the Americas in all but the large endemic area crossing the Venezuela-Brazil border. This was achieved through between 23 and 36 rounds of biannual MDAi complemented with quarterly MDAi in around 300 communities [7, 11,12,13,14,15]. Data obtained in Africa since 2003 have shown that long-term MDAi, implemented as community-directed treatment (CDTI), may have eliminated transmission or may be close to doing so in many areas [14,15,16,17,18]. These data motivated the objective to eliminate O. volvulus transmission in some African countries by 2020 and in 80% of endemic countries by 2025 [19]. These targets have recently been revised to achieve WHO-verified interruption of parasite transmission in 12 countries worldwide and stop MDAi in at least one focus in 34 countries by 2030 [20].

Expert consultations and data reviews by the African Programme for Onchocerciasis Control (APOC, 1995–2015) concluded that alternative treatment strategies (ATS) are required for onchocerciasis elimination in many areas in Africa [19]. Potential ATS identified include those based on moxidectin which was approved in 2018 for treatment of O. volvulus infected individuals ≥ 12 years of age by the US Food and Drug Administration (FDA) [21,22,23,24,25,26,27,28,29,30]. Discovery, development and implementation research for other ATS [31] is also ongoing including for new or repurposed drugs [10, 32,33,34,35,36], for effective, affordable and sustainable complementary vector control strategies [37, 38] and for approaches to safe use of ivermectin in loiasis co-endemic areas (drugs for safe reduction of Loa loa microfilaraemia [39,40,41] and ‘test-and-not-treat’ or ‘test-and-treat’ strategies [42,43,44,45,46]). Furthermore, research for a vaccine is continuing [47].

The triple drug combination ivermectin, albendazole and diethylcarbamazine (IDA) was recommended in 2017 by WHO as an alternative MDA regimen for elimination of lymphatic filariasis in specific areas [48]. Treatment with IDA following treatment with ivermectin (referred to as ‘pretreat and treat’ strategy) has been proposed as an ATS for onchocerciasis-endemic areas with long-term CDTI to accelerate onchocerciasis elimination if a single IDA dose sterilizes adult O. volvulus worms and eliminates microfilariae from the skin permanently [49].

Oral diethylcarbamazine (DEC) was used for onchocerciasis treatment before ivermectin became available. Treatment schedules differed with total doses ranging from 100 to 6000 mg and the number of administrations ranging from a single dose to daily doses for up to 2 weeks. WHO recommended a 0.5 mg/kg or 1.0 mg/kg starting dose for 1–2 days and a 2.0 mg/kg maintenance dose twice a day for 5–7 days for a total dose of approximately 30 mg/kg for adults [50]. While DEC has low intrinsic toxicity [51, 52], oral DEC treatment of O. volvulus-infected individuals can result in very severe Mazzotti reactions, the signs and symptoms of the immunological reaction of the body to dead and dying microfilariae. These include worsening of onchocercal eye disease and new ocular lesions and anaphylactic reactions, which resulted in the suggestion to start DEC treatment in the hospital since the susceptibility of an individual is not predictable [50, 53,54,55,56,57,58]. The severity of the Mazzotti reactions increases with the microfilariae levels in individuals treated with the same oral DEC dose and increases with the DEC dose when individuals with similar microfilariae burden are compared. These observations led to the conclusion that the severity of the Mazzotti reaction after DEC treatment is a function of the number of microfilariae present before treatment and killed [51, 59]. In 1995, the WHO Expert Committee on Onchocerciasis Control recommended that DEC should no longer be used for treatment of onchocerciasis and, if used for treatment of lymphatic filariasis in patients with onchocerciasis, ivermectin should be administered first and DEC only given after skin and ocular mf have been cleared [58]. The proposed ‘pretreat and treat’ strategy to use IDA in onchocerciasis-endemic areas after long-term MDA with ivermectin plus albendazole or long-term CDTI [49] is based on the assumption that long-term MDAi will have reduced O. volvulus skin microfilariae density (mf/mg skin, SmfD) to ‘usually less than 5 mf/mg skin’. This would reduce the risk of severe Mazzotti reactions, and this risk would be further reduced by treating only those individuals with IDA who took the ivermectin pretreatment [49]. Given data on the percentage of the population that participates in MDAi and long-standing challenge to optimize that percentage, the percentage of individuals with SmfD above the ‘usually less than 5 mf/mg skin’ warrants consideration as do challenges in monitoring participation in MDAi [60,61,62,63,64,65,66,67]. To our knowledge, levels of skin mf and ocular mf below which DEC can be administered without risk of severe Mazzotti reactions/ worsening of onchocercal eye disease and/or new ocular lesions have not been established. Furthermore, the number of individuals included in published studies is small, and we consider it insufficient to establish ‘safe’ maximum skin and ocular mf levels, in particular given inter-individual variability in the occurrence, type and severity of adverse reactions after DEC (as well as ivermectin and moxidectin) treatment. Consequently, more data are needed to determine the risk factors for the type of adverse reactions to oral DEC that could result in an unacceptable benefit-risk ratio of MDA including DEC to inform deliberations about including DEC treatment in onchocerciasis elimination strategies.

A clinical study in Ghana compared the safety and efficacy of single (n = 52) and three daily doses (n = 51) of IDA (150 µg/kg ivermectin, 6 mg/kg DEC, 400 mg albendazole) with the safety and efficacy of ivermectin plus albendazole (IA, 150 µg/kg ivermectin, 400 mg albendazole, n = 52) [68]. Individuals qualifying for enrolment had to meet the following criteria: (i) be among the 231 individuals who participated in a previous trial (https://www.clinicaltrials.gov/ct2/show/NCT03517462) that examined the safety and efficacy of a single dose of ivermectin for reducing skin and ocular microfilariae levels [69], (ii) had received two single doses of 150 µg/kg ivermectin > 1 year apart, the last within 1–28 weeks before IDA or IA treatment and (iii) had ≤ 3 mf/mg skin and ≤ 5 motile mf in the anterior chamber in either eye and no mf detected in the posterior segment of either eye [68]. The study found that the percentage of fertile female worms was lower (p = 0.004) after a single or three daily IDA doses [40/261 (15.3%) and 34/281 (12.1%), respectively] than after a single IA dose [41/180 (22.8%)]. Given the study size and inclusion criteria chosen to minimize the risk of ocular reactions, the study could not provide significant data on the safety of the proposed ‘pre-treat and treat’ MDA strategy.

We have previously reported results of our randomized, double-blind Phase 3 trial comparing the efficacy and safety of a single oral dose of 8 mg moxidectin (n = 978) and 150 µg/kg ivermectin (n = 494) in ivermectin-naïve individuals with ≥ 10 mf/mg skin with or without ocular involvement. The data showed that 1, 6, 12 and 18 months after treatment, SmfD was significantly lower among moxidectin- than ivermectin-treated participants [22, 30]. In contrast, there was no statistically significant difference in the level of live microfilariae in the anterior chamber (mfAC) 12 months after treatment among participants with > 10 mfAC pre-treatment [30].

We are here reporting the number of mfAC before treatment, 4 days and 1, 6, 12 and 18 months after treatment and their relationship to pre- and post-treatment SmfD as well as the ocular adverse events (AEs) observed within 6 months post-treatment. Our primary objective is to contribute to the evidence base available for review by WHO and countries for a decision on whether to include a ‘pretreat and treat’ strategy in control and elimination guidelines and policies for onchocerciasis and/or lymphatic filariasis in onchocerciasis co-endemic areas. While all of our participants had a pre-treatment SmfD at least twice the ‘usually less than 5 mg/skin’, the experience with severe and serious adverse reactions to ivermectin in Loa loa co-endemic areas has shown that decisions on control and elimination strategies need to consider risks for even a very small percentage of individuals [46, 70,71,72,73]. This need drives our emphasis on graphical presentation of individual participant data and presentation of the distribution of participants by mfAC level, which facilitate better appreciation of inter-individual differences than the standard statistical measures of variability or statistical models. Our secondary objective is to contribute to the evidence base for review by WHO and countries for decisions on whether to include moxidectin in guidelines and policies for onchocerciasis elimination.

Methods

A detailed description of trial conduct and methods and a Consort Flow chart have been published previously [30].

Trial registration

The study was registered on 14 November 2008 in Clinicaltrials.gov (ID: NCT00790998).

Regulatory Agency and Ethics Committee approval and participant consent

As previously reported [22, 30], the protocol, information documents for potential participants, participant consent and assent forms and study conduct were approved by the Ghana Food and Drugs Authority and the Ghana Health Service Ethics Review Committee, the Liberia Ministry of Health and Social Welfare and the Ethics Committee of the Liberia Institute for Biomedical Research, the Ministère de la Santé Publique of the Democratic Republic of the Congo (DRC) and the Ethics Committee of the Ecole de la Santé Publique Université de Kinshasa in DRC and the WHO Ethics Review Committee.

Participants documented their informed consent or assent with parental consent to study participation through signature or thumbprint in the presence of a literate witness in their villages. This included consent to publication of summaries of the results. Given lack of consent to sharing individual participant data, many summary tables and figures are included in the manuscript or in Additional file 1.

Overview of study conduct

This study was conducted between April 2009 and May 2012. It enrolled individuals ≥ 12 years old with at least ≥ 10 O. volvulus microfilariae/mg skin in four onchocerciasis-endemic areas where CDTI had not yet been initiated: Nord Kivu Province (current Zones de Santé Kalunguta and Mabalako) in DRC, Ituri province in DRC (Zone de Santé Logo in Northern Ituri, subsequently referred to as Nord-Ituri), Lofa County in Liberia (subsequently referred to as Lofa) and the Kpasa subdistrict within the Nkwanta North health district in Ghana (subsequently referred to as Nkwanta). Details on the location of the villages where participants were recruited and the prevalence of infection as determined during screening for the study have been provided previously [22].

Following randomization stratified by sex and ‘level of infection’ (< 20 mf/mg skin vs. ≥ 20 mf/mg skin), 978 participants received a single oral dose of 8 mg moxidectin and 494 participants a single oral dose of 150 µg/kg ivermectin. To ensure double blinding, each participant received four identical-looking capsules containing 2 mg moxidectin tablets, 3 mg ivermectin tablets or placebo, as required by treatment allocation and weight. The capsules for each participant had been prepared by a pharmacist not involved in participant evaluation. Participants swallowed the capsules under observation by a study team member.

Participants stayed in the research center from screening to Day 6 (± 1 Day) after treatment for follow-up examinations and daily evaluation for AEs. ‘Outpatient’ follow-up was conducted 14 days and 1, 3, 6 and 12 months after treatment (subsequently referred to as Day 14, Month 1, 3, 6 and 12, respectively). The study was initiated with 18 months post-treatment as the last follow-up timepoint. After WHO became the sole study sponsor in July 2011, resource limitations required a protocol amendment eliminating the Month 18 follow-up. This resulted in 96% of moxidectin-treated and 97% of ivermectin-treated participants with Month 12 data but only 78% of participants in both treatment arms with Month 18 data [30].

Measurement of skin microfilariae densities

Four skin snips (one snip from each iliac crest and calf) were taken pre-treatment and at Months 1, 6, 12 and 18 and SmfD determined as described previously [21, 30].

Ocular examinations

Ocular examinations (detailed history and questioning for symptoms, visual acuity, color vision, visual field examination with Frequency Doubling Technology (FDT) perimetry, ocular mobility, pupillary reflex examination, external ocular structures examination, anterior segment examination with a Haag Streit 900 slit-lamp, intraocular pressure and dilated fundus examination by direct and indirect ophthalmoscopy) were conducted pre-treatment as well as 3 or 4 days (subsequently referred to as Day 4 data) and at Months 1, 6, 12 and 18. mfAC and microfilariae in the cornea (mfCOR) were counted in both eyes with 10 × or 16 × magnification after participants had been sitting with their head down for at least 5 min. If considered necessary by the ophthalmologist, the head-down position for at least 5 min was repeated before counting of the microfilariae in the second eye.

Grading of ocular AEs

Any new and clinically significant abnormality or worsening of conditions identified pre-treatment were recorded as an AE. Details of AE recording have been provided previously [30]. In brief, AEs were recorded with start and stop date and their severity graded according to the Onchocerciasis Chemotherapy Research Center (OCRC) grading scale. The scale had been developed to quantify signs and symptoms of O. volvulus infection caused by the immunological reaction to dying and dead microfilariae reaching the end of their natural lifespan, and their aggravation following treatment, i.e. Mazzotti reactions [74, 75]. The scale was expanded to include other types of AEs for the moxidectin Phase 2 [21, 29] and Phase 3 study [22, 30] and has been provided previously [30]. OCRC grading criteria for Mazzotti reactions differ substantially from commonly used grading criteria for similar AE and generally reflect grade for grade much less severe symptoms. For example, the National Cancer Institute criteria include need for medical intervention for grade 2 sometimes, grade 3 frequently and grade 4 nearly always. In contrast, most OCRC criteria grade 4 Mazzotti reactions require no intervention [30]. All AEs were characterized by the investigators in terms of relationship to study drug and Mazzotti reaction. Before unblinding, all AEs were reviewed centrally by one author (NOO) for characterization as Mazzotti reactions. The outcome of that central review forms the basis of the Mazzotti reactions reported here. All AE verbatims were coded using the Medical Dictionary for Regulatory Activities (MedDRA version 13.1). Mazzotti reactions were additionally coded using a Mazzotti reaction-specific dictionary since MedDRA codes the same reaction by body system, which compromises treatment comparisons [30]. Coding using the Mazzotti reaction-specific dictionary is the basis for the Mazzotti reactions reported in the manuscript. Presentation across both Mazzotti reactions and other ocular AEs in Additional file 1 is based on MedDRA coding.

Statistical analysis

Given the focus on changes in mfAC, only the data for the 1463/1472 (99.4%) individuals treated who had both eyes evaluated throughout the study are included (Table 1). All mfAC values in the text, tables and figures are the sum of the mfAC detected across both eyes.

Table 1 Participants with both eyes evaluated by treatment arm, pre-treatment mfAC category and timepoint

In recognition of the fact that as the number of microfilariae in the eyes increases beyond 10 the accuracy of the counts decreases, individual counts are presented in Figures for illustrative purposes while descriptive statistics are based on the following six mfAC categories: undetected (0), 1–5, 6–10, 11–20, 21–40, > 40. Three pre-treatment SmfD categories (10 to < 20, 20 to < 50 and ≥ 50), referred to as ‘intensity of infection’ (IoI), and three post-treatment SmfD categories (0, > 0–5, > 5) were defined.

A linear mixed model was used to evaluate the marginal means of the number of mfAC pre-treatment and on Day 4, Months 1, 6, 12 and 18. The model included treatment and sex as factors, age as adjusting variable and SmfD at baseline as covariate. Study area was included as a random effect. Treatment, sex and IoI adjusted marginal means and 95% confidence intervals (CI) by treatment were extracted from the model. A first series of models for each timepoint was run with the raw mfAC values to calculate the arithmetic adjusted marginal means. A second series of models for each timepoint was run with the log transformed mfAC values to calculate the geometric adjusted marginal means.

A linear mixed model was used to evaluate the effect of both SmfD and mfAC at the timepoint of the outcome and previous timepoints on the number of mfAC pre-treatment, Day 4, and Months 1, 6, 12 and 18. The model included treatment and sex as factors and age as adjusting variable. Study area was included as a random effect.

Generalized linear mixed models with a logit link were used to evaluate covariates and factors impacting the probability of having at least one ocular Mazzotti reaction within the 1st month after treatment and of having at least one ocular AE (whether considered Mazzotti reaction or not) within 6 months after treatment. mfAC levels were included as categorical variable (0, 1–5, 6–10 and ≥ 11) and SmfD levels as defined above. Only four mfAC level categories were considered because of the low number of individuals with mfAC ≥ 11 (n = 135 and n = 76 in the moxidectin and ivermectin treatment arm, respectively). The initial model for ocular Mazzotti reactions included treatment, age, sex, SmfD pre-treatment, mfAC levels pre-treatment and on Day 4 as fixed effects and study area as random effect. The final model included treatment, sex and mfAC levels pre-treatment. The initial model for any ocular AE during the first 6 months after treatment included treatment, age, sex, SmfD pre-treatment and at Month 1, as well as mfAC pre-treatment, on Day 4 and Month 1 as fixed effects and study area as random effect. The final model included treatment, age, sex and mfAC level at Month 1.

Analyses were conducted with SAS version 9.4 (SAS Institute, Cary, NC, USA). Figures were generated in Excel 365.

Results

Consort diagram and number of participants with both eyes evaluated on Day 4 and Months 1, 6, 12 and 18

The study CONSORT flow diagram has been reported previously [30].

Table 1 shows the number of individuals with data on mfAC obtained from both eyes at Day 4 and Month 1, 6, 12 and 18 by pre-treatment mfAC category and treatment arm. The maximum number of mfAC was 102 in the moxidectin and 92 in the ivermectin treatment arm. Additional file 1: Table S1 shows the number of participants by pre-treatment SmfD, mfAC category and sex.

Number of live microfilariae in the anterior chambers on Day 4 and Months 1, 6, 12 and 18

Figures 1 and 2 show the mfAC levels on Day 4 and Month 1, 6, 12 and 18 relative to pre-treatment levels for each participant and illustrate the inter-individual variability in the mfAC level change after treatment.

Fig. 1
figure 1

mfAC 4 days, 1, 6, 12 and 18 months post-treatment relative to pre-treatment mfAC among participants with < 10 mfAC pre-treatment. x-axis mfAC levels pre-treatment, y-axis mfAC levels on Day 4 (D4 1st column), Month 1 (M1 2nd column), Month 6 (M6, 3rd column), Month 12 (M12 4th column) and Month 18 (M18, 5th column) post-treatment with ivermectin (IVM, upper row, orange symbols) or moxidectin (Moxi, lower row, green symbols). Data from participants from Nord Kivu (∆), Nord Ituri (), Lofa County (□) and Nkwanta district (). mfAC live microfilariae in the anterior chambers

Fig. 2
figure 2

mfAC 4 days, 1, 6, 12 and 18 months post-treatment relative to pre-treatment mfAC among participants with ≥ 10 mfAC pre-treatment. x-axis mfAC levels pre-treatment, y-axis mfAC levels on Day 4 (D4 1st column), Month 1 (M1 2nd column), Month 6 (M6, 3rd column), Month 12 (M12 4th column) and Month 18 (M18, 5th column) post-treatment with ivermectin (IVM, upper row, orange symbols) or moxidectin (Moxi, lower row, green symbols). Data from participants from Nord Kivu (∆), Nord Ituri (), Lofa County (□) and Nkwanta district (). mfAC live microfilariae in the anterior chambers

Figure 3 shows the arithmetic means and the linear model derived adjusted geometric mean mfAC pre-treatment and on Day 4 and Month 1, 6, 12 and 18.

Fig. 3
figure 3

Arithmetic mean (A) and linear model derived adjusted geometric mean (B) mfAC pre-treatment (month 0), 4 days, 1, 6, 12 and 18 months post-treatment in the moxidectin and ivermectin treatment arm. Orange triangle: data in the ivermectin treatment arm, green circles: data in the moxidectin treatment arm. Mean: arithmetic mean; GM: geometric mean; CI: confidence interval; mfAC, live microfilariae in the anterior chambers. The inserts provide an enlargement of the data obtained pre-treatment (Month 0) and 4 days and 1 month after treatment

Increases in the number of mfAC detected sufficient to change the mfAC category occurred from pre-treatment to Day 4 and from pre-treatment to Month 1, respectively, in 21% and 16% in the moxidectin treatment arm and 19% and 15% in the ivermectin treatment arm. Furthermore, in 11% of participants in both treatment arms mfAC levels increased from Day 4 to Month 1 (Table 2). This resulted in maximum mfAC levels of up to 23 and 26 on Day 4 and of up to 26 and 24 at Month 1 among participants with undetectable mfAC levels pre-treatment in the moxidectin and ivermectin treatment arm, respectively (Additional file 1: Table S2). The mean, standard deviation, minimum and maximum mfAC levels pre-treatment, on Day 4 and at Month 1 for all participants with mfAC increases from pre-treatment to Day 4 or Month 1 and from Day 4 to Month 1 are provided in Additional file 1: Table S2.

Table 2 Number (%) of participants with mfAC number increases from pre-treatment to Day 4 or Month 1 and from Day 4 to Month 1 resulting in mfAC levels in a higher mfAC category

Additional file 1: Table S3 shows the distribution of participants by mfAC category at Day 4 and Month 1 relative to their distribution pre-treatment and on Day 4 resulting from increases in mfAC levels in some participants and decreases or changes not affecting the mfAC category in others. Across all participants, 54.9% and 71.5% of moxidectin-treated and 54.1% and 71.1% of ivermectin-treated participants had no mfAC detected on Day 4 and Month 1, respectively. mfAC levels > 10 were detected on Day 4 and Month 1 in 13.1% and 4.9% of moxidectin-treated and 15.9% and 5.3% of ivermectin-treated participants, respectively.

At Month 6, 12 and 18, the percentage of participants with 0 mfAC and 1–5 mfAC detected was 94.8% and 4.3%, 98.1% and 1.5%, and 98.7% and 1.2%, respectively, in the moxidectin treatment arm and 95.5% and 4.1%, 95.6% and 3.6%, and 95.8% and 3.1% in the ivermectin treatment arm. With two exceptions, the remaining participants had mfAC levels in the 6–10 or 11–20 mfAC category. Maximum mfAC levels at Month 6, 12 and 18 were 20, 12 and 6 in the moxidectin treatment arm and 11, 12 and 19 in the ivermectin treatment arm, respectively (Table 3). One participant from Nord Ituri treated with ivermectin had mfAC levels of 56, 120, 100, 49, 54 and 77 pre-treatment and on Day 4, Month 1, 6, 12 and 18, respectively, with SmfD levels of 53, 53, 32, 49 and 72 pre-treatment and at Month 1, 6, 12 and 18, respectively, qualifying for suboptimal microfilariae response and suboptimal response [22]. The Mazzotti reactions the participant experienced starting 1 day after ivermectin administration (dizziness, headache, facial oedema) suggest that pharmacodynamically sufficient ivermectin was absorbed even though the participant was recorded as having grade 1 diarrhoea from the day before to 2 days after ivermectin administration. One participant from Nkwanta treated with moxidectin had mfAC levels of 100, 100, 45, 0 and 22 pre-treatment and on Day 4, Month 1, 6 and 12, respectively, with SmfD levels of 44, 0, 0 and 0.4 pre-treatment and at Month 1, 6 and 12, respectively. That participant did not have a Month 18 evaluation because of the protocol amendment abolishing the Month 18 visit.

Table 3 Number (%) of participants by mfAC category at Months 6, 12 and 18 by pre-treatment mfAC category and treatment arm

Pre- and post-treatment mfAC levels relative to pre-treatment SmfD (IoI)

Additional file 1: Figs. S1 and S2 show for participants with < 10 mfAC and ≥ 10 mfAC pre-treatment, respectively, the mfAC counts before and on Days 4 and Month 1, 6, 12 and 18 by IoI and illustrate the inter-individual variability in pre-treatment as well as post-treatment mfAC relative to IoI.

Table 4 provides the corresponding distribution of participants in the defined pre-treatment mfAC categories by IoI. The data indicate that as the IoI increases the percentage of individuals with undetected mfAC decreases. Across both treatment arms, this percentage was 70.3%, 63.3% and 45.6% for participants with IoI ≤ 20 SmfD, 20– < 50 SmfD and ≥ 50 SmfD, respectively. The percentage with mfAC pre-treatment ≥ 40 was 1.9%, 1.6% and 2.7%, respectively. Additional file 1: Table S2 provides these percentages for all mfAC categories. Table 5 presents the distribution by mfAC categories at Day 4 and Month 1 for the participants with an increase in mfAC levels from pre-treatment to Day 4 and Month 1 and from Day 4 to Month 1 that resulted in a higher mfAC category by pre-treatment SmfD category. The data suggest that a higher pre-treatment SmfD is associated with a higher percentage of individuals who experienced a transient increase in mfAC from pre-treatment to Day 4 and Month 1.

Table 4 Number (%) of participants by mfAC category pre-treatment and on Day 4 and Month 1, 6, 12 and 18 by intensity of infection
Table 5 Number (%) of participants by mfAC category on Day 4 and Month 1 and pre-treatment IoI for participants with mfAC number increases from pre-treatment to Day 4 or Month 1 and from Day 4 to Month 1 resulting in mfAC levels in a higher mfAC category

Post-treatment mfAC levels relative to post-treatment SmfD

Table 6 shows the distribution of participants in each treatment arm by mfAC category and SmfD category at Month 6, 12 and 18. At Month 6, 12 and 18 after ivermectin treatment, mfAC were undetectable in all of the 54, 24 and 15 individuals with undetectable SmfD, respectively, while mfAC were undetectable in 91.5%, 92.9% and 94.1% of the 94, 240 and 237 individuals with > 5 SmfD, respectively. At Month 6, 12 and 18 after moxidectin treatment, the percentage of participants with undetectable mfAC was 94.9%, 99.5% and 99.5% among the 830, 434 and 211 participants with undetectable SmfD, respectively. No moxidectin-treated participant had > 5 SmfD at Month 6. At Month 12 and 18 after moxidectin treatment, mfAC were not detected in 90.5% and 95.9% of the 74 and 170 individuals with > 5 SmfD, respectively. Additional file 1: Figs. S3 and S4 show the data for each individual participant for those with < 10 mfAC and those with ≥ 10 mfAC pre-treatment, respectively. They illustrate not only the inter-individual variability in the change in mfAC relative to the change in SmfD but also that mfAC reduction can be slower than SmfD reduction [30].

Table 6 Number (%) of participants by mfAC category at Month 6, 12 and 18 by post-treatment SmfD and treatment

Participant characteristics impacting mfAC levels before and after treatment

As previously reported for mfAC levels at Month 12 in participants with at least 10 mfAC [30], there was no statistically significant difference at any timepoint between the mfAC levels in the moxidectin and the ivermectin treatment arm. Age and sex were not a significant covariate and factor, respectively. Table 7 provides an overview of the impact on the mfAC levels at a particular timepoint of the other covariates (SmfD before or at that timepoint and prior mfAC levels) emerging from the linear model. For all post-treatment timepoints, the mfAC levels were statistically significantly impacted by at least two of these covariates. Additional file 1: Table S4 shows the full output.

Table 7 Impact of SmfD and mfAC levels up to the relevant timepoint on mfAC levels pre-treatment and at different timepoints post-treatment

Microfilariae in the cornea

Live microfilariae in the cornea were detected pre-treatment in the majority of participants from Lofa (count range 3–42) but in only few participants in the other study areas. Similarly, dead microfilariae in the cornea were more frequently seen in participants from Lofa (count range 2–25) than in participants from the other study areas (count range 0–18). The reason for this difference is unknown. At Month 6, 12 and 18, live microfilariae in the cornea were detected only in five (count range 1–9), zero and one (count = 2) participants from Lofa, respectively. Dead microfilariae in the cornea at Month 6, 12 and 18 were found in one (count = 6), zero and zero participants from Nord Kivu, two (counts = 2), zero and one (count = 1) participants from Nord-Ituri, 18 (count range 1–29), two (count = 1) and one (count = 2) participant from Lofa and in zero, one (count = 1) and zero participant from Nkwanta, respectively.

Ocular Mazzotti reactions and other ocular AEs

As reference for the ocular Mazzotti reactions and other post-treatment ocular adverse events, Additional file 1: Table S5 provides the ocular medical history the investigators considered relevant and entered into the data base by study area.

As previously reported across all participants treated [30], a higher percentage of participants treated with moxidectin (121/973, 12.4%) than with ivermectin (50/490, 10.2%) had at least one ocular Mazzotti reaction, but the type of reactions did not differ (Table 8). Among those with ocular Mazzotti reactions, a higher percentage of moxidectin treated (26/121, 21.5%) than ivermectin treated (6/50, 12%) participants had more than one ocular Mazzotti reaction, but there was no trend towards higher severity of the ocular Mazzotti reactions after moxidectin than ivermectin treatment (Table 9). The vast majority of Mazzotti reactions occurred between the day of treatment up to the 2nd day after treatment with a trend towards earlier start after moxidectin than after ivermectin treatment (Table 10), a trend also seen for non-ocular Mazzotti reactions.

Table 8 Participants with ocular Mazzotti reactions starting within 1 Month of treatment among participants with both eyes evaluated by treatment
Table 9 Participants with ocular Mazzotti reactions starting within 1 Month of treatment by severity and treatment among participants with both eyes evaluated
Table 10 Participants with ocular Mazzotti reactions starting within 1 Month of treatment by start day and treatment among participants with both eyes evaluated

The logistic model identified women as at higher risk for ocular Mazzotti reactions (OR 1.537, 95% CI 1.096–2.157, p = 0.0128) and detected pre-treatment mfAC levels as impacting the risk for ocular Mazzotti reactions (OR for 0 mfAC: > 10 mfAC 2.704 with 95% CI 1.272–5.749, p = 0.0098, p = 0.0608 for analysis across all mfAC level categories). The OR for treatment with moxidectin: ivermectin was 1:0.746 (95% CI 0.519–1.072, p = 0.1127). Additional file 1: Table S6 shows the output of the final model.

At least one ocular AE (i.e. considered Mazzotti reaction or not) was recorded for 223/973 (22.9%) and 82/490 (16.7%) of participants to Month 1 and for 92/973 (9.5%) and 43/490 (8.8%) of participants from Month 1 to Month 6 after treatment with moxidectin and ivermectin, respectively. An overview of participant incidence and total number of ocular AEs is provided in Additional file 1: Tables S7 and S8. Grade 3 ocular AEs not considered a Mazzotti reaction in the moxidectin treatment arm were eye pruritus in three participants (starting 64, 86 and 159 days after treatment, respectively), eye pain starting 173 days after treatment and cyclitis starting 174 days after treatment. One ivermectin-treated participant experienced a grade 3 ocular AE not considered a Mazzotti reaction: increased lacrimation starting 10 days after treatment. The logistic model showed a higher risk of at least one ocular AE within 6 months of treatment for women than men (p = 0.015, OR 1.404 with 95% CI 1.068–1.845) and a significant impact of mfAC levels at Month 1 (p = 0.0005 across all mfAC levels evaluated, OR 0 mfAC: > 10 mfAC 2.918 with 95% CI 1.737–4.9 and p < 0.0001). The OR for moxidectin:ivermectin was 1:0.709 (95% CI 0.536–0.939, p = 0.0165). Additional file 1: Table S9 shows the output of the final model.

Discussion

In our study population from CDTI-naïve areas, selected to have at least 10 mf/mg skin, around 60% of participants had undetectable mfAC levels pre-treatment. Approximately 19% and 6% had mfAC levels between 1–5 and 6–10, respectively, and around 2% had > 40 mfAC across both eyes (Table 1). Pre-treatment SmfD significantly impacted pre-treatment mfAC levels (Table 7). Given that pre-treatment mfAC levels of > 10 mfAC increased the risk for at least one ocular Mazzotti reaction (Additional file 1, Table S6), it is noteworthy that this statistical significance occurs on the background of inter-individual variability in SmfD vs. mfAC levels (Additional file 1: Figs. S1 and S2). The levels of mfAC at a particular post-treatment timepoint were statistically significantly impacted by SmfD at that and/or previous timepoints as well as by mfAC levels at one or more previous timepoints (Table 7) but not by the treatment arm (Additional file 1: Table S4). We do not know what drives mfAC levels relative to SmfD levels to explain why SmfD are significantly lower at Months 1, 6, 12 and 18 after moxidectin than ivermectin treatment [21, 22, 30], but there is no treatment difference for mfAC levels.

Higher mfAC levels on Day 4 and Month 1 than pre-treatment were identified in 21% and 16% of moxidectin-treated and 19% and 15% of ivermectin-treated participants, respectively. This increase was not restricted to individuals with low mfAC levels pre-treatment (Tables 2, 5). A transient increase in mfAC levels in some individuals early after ivermectin as well as after DEC treatment has been shown previously, including during the clinical studies that supported regulatory approval of ivermectin [76,77,78,79,80,81,82,83,84,85,86,87]. The peak mfAC increase around Day 4 after treatment determined in these studies informed mfAC measurement on Day 3 or 4 in the moxidectin Phase 2 and 3 studies [21, 30]. An mfAC increase was not mentioned and cannot be deduced from the data provided in other publications of the early ivermectin studies [88,89,90]. Additional file 1: Table S10 provides an overview of the data reported. The way mfAC data are quantified in the publications (frequency of detectable mfAC levels, geometric means, descriptive text) makes it impossible to determine the frequency and extent of the mfAC increases for comparison with our data.

The lack of evaluation of mfAC changes early after treatment in subsequent studies of the efficacy and safety of ivermectin may be due to the fact that once the concern that ivermectin may have a similar risk of severe ocular reactions as DEC had been alleviated, quantifying changes in the number of ocular microfilariae early after ivermectin treatment was not considered important anymore. In the context of the proposed introduction of DEC as part of a 'pretreat and treat’ strategy into onchocerciasis elimination strategies [49], attention to mfAC levels before and increases early after treatment with DEC will become important in studies of the safety of the 'pretreat and treat’ strategy until a better understanding of the relationship between ocular mfAC before and early after treatment and the ocular safety of treatment with DEC has been achieved. In our study, pre-treatment mfAC levels > 10 significantly increased the risk of having at least one ocular Mazzotti reaction [occurring primarily within the first few days after treatment (Table 10)] and mfAC levels > 10 at Month 1 significantly increased the risk of at least one ocular AE within the first 6 months after treatment relative to 0 mfAC at these timepoints (Additional file 1: Tables S6, S9). In light of the fact that in our study women had a higher risk of having at least one ocular Mazzotti reaction and at least one ocular AE of any type than men (Additional file 1: Tables S6, S9), sex disaggregated analyses of the safety data of further studies of the ocular safety of ivermectin, DEC or moxidectin are indicated.

Within the context of the proposed introduction of DEC into onchocerciasis elimination strategies [49], a study of the tolerability and effect of ivermectin on skin and ocular microfilariae was recently conducted in primarily ivermectin-naïve individuals from hypoendemic villages in the Nkwanta North district of Ghana (pre-treatment SmfD 3 to 86.3 mf/mg). The study included mfAC measurement pre-treatment (64/231 with mfAC, mfAC range 1–150) and on Day 7, Month 3 and Month 6 post-treatment. The study identified a higher number of individuals with mfAC on Day 7 (73/231, mfAC range 1–82). The presentation of the data (frequency of detectable mfAC levels, geometric means, ranges and graphical presentation of individual participant data excluding the Day 7 data) [69] limits comparison with our data but the findings presented are consistent with our data and those of the early ivermectin studies. The lack of more detailed presentation of the mfAC data by Opoku et al. may be due to the fact that they attributed the mfAC increases to insensitivity of a single slit-lamp examination for detecting mfAC when counts are low, day to day variation in mfAC and easier detection of mfAC after they have been paralyzed by ivermectin [69]. The motility of mfAC during the first days after ivermectin treatment was found to be abnormal (transient immobility for 0.5–1 s, movement in extended rather than coiled configuration, motion in which mfAC bent only in the middle suggesting spasm) [88, 91] but paralysis facilitating counting has to our knowledge not been described previously. We observed in our study that mfAC were more mobile and difficult to count 3–4 days after treatment than at any other time in the study, resulting in frequent requests to participants to take a head-down position again for at least 5 min before counting of the mfAC in the second eye.

While we do not consider the increase in mfAC we observed early after treatment in around 20% of participants as due to methodological issues, we do recognize the limitations of the accuracy of mfAC counts. It is thus important not to overinterpret ‘0 mf counted’ as indicating that there are no mfAC in the eyes or a small change in mfAC counted as representing a real increase or decrease. However, we do think that the data available support the hypothesis that a ‘mobilization of microfilariae’ into the anterior chamber can occur. This term was used in some of the publications of the early ivermectin studies (e.g. [77, 78]). Mobilisation of microfilariae after DEC and ivermectin treatment into the urine and blood has been described previously as has appearance of microfilariae in other body fluids after DEC treatment [77, 78, 92,93,94,95,96]. We also observed mobilisation of microfilariae into blood and urine in some participants in the moxidectin Phase 2 study (unpublished data, summary statistics provided in Additional file 1: Table S11).

At Month 6 and 12, no mfAC were detected, respectively, in 95% and 98% of participants who had received a single dose of moxidectin and in 95% and 96% of participants who had received a single dose of ivermectin. At these timepoints, > 10 mfAC were detected in 0.2% and 0.2% of moxidectin- and in 0.4% and 0.2% of ivermectin-treated participants, respectively (Tables 3, 4). The above referenced study by Opoku and co-workers [69] identified mfAC 6 months after ivermectin treatment in a comparable percentage of participants [12/212 (5.7%), mfAC range 1–35], including ‘low mfAC counts’ in three participants who had no mfAC detected pre-treatment (but two had mfAC detected on Day 7). In our study, covariates impacting Month 6 mfAC levels significantly were Month 1 SmfD and Day 4 and Month 1 mfAC levels. Covariates with significant impact on Month 12 mfAC levels were SmfD pre-treatment and at Month 12 as well as mfAC levels at pre-treatment, Month 1 and 6 (Table 7). Repeat dose studies will allow us to determine whether the covariates which impacted mfAC levels in our study are also impacting mfAC levels after multiple treatments. Data analysis not only for inter-individual but also intra-individual variability in changes in SmfD and mfAC levels is needed to determine whether individuals show the same pattern of post-treatment changes in SmfD and mfAC levels after each treatment or not. Depending on the outcome of large-scale studies of the safety of the ‘pretreat and treat’ strategy, a non-random response pattern will have implications for assessing the risk of this strategy. As recently pointed out [97], a non-random response pattern also has implications for the prospects of achieving onchocerciasis elimination with ivermectin (conferring an additional advantage to moxidectin-based strategies). This will affect the comparative benefit-risk assessment of MDAi vs. different ATS required for a decision on whether or not to include a ‘pre-treat and treat’ strategy in onchocerciasis elimination guidelines and policies.

Conclusions

Among our ivermectin-naïve study participants from onchocerciasis meso- or hyperendemic villages with at least 10 mf/mg skin, there was no statistically significant difference in the effect of a single dose of 8 mg moxidectin or 150 µg/kg ivermectin on mfAC levels on Day 4 or Month 1, 6, 12 or 18. Ocular Mazzotti reactions occurred in 12.4% of moxidectin and 10.2% of ivermectin treated participants without a difference in the type of these reactions or their severity between the treatment arms. The extent to which findings on the efficacy and safety of ivermectin, moxidectin and DEC in ivermectin-naïve individuals are relevant for decisions on including DEC into strategies for elimination of onchocerciasis in areas with a long history of MDAi needs to be determined. Assuming the benefit of a ‘pre-treat (with ivermectin) and treat (with IDA)’ strategy for elimination of O. volvulus transmission compared to strategies without DEC is demonstrated, additional studies are required to support an assessment of the risks of this strategy. Our results, as well as the results of the early ivermectin studies and past experience with DEC treatment (Additional file 1: Table S10), suggest that safety studies should be designed to improve understanding of (i) the role of SmfD and mfAC levels pre-treatment and of mobilisation of microfilariae into the anterior chambers early after treatment on the safety of DEC and (ii) inter-individual susceptibility to adverse reactions to DEC treatment, ideally including the determinants. Safety study design should also consider the percentage of the population which participates irregularly in MDAi (see e.g. [60, 61, 67, 98, 99]) and the effect of irregular MDAi participation on SmfD as well as the potential for higher risk of women than men for post-treatment AEs. Decisions on the risk-benefit ratio of strategies including DEC should interindividual variability in skin and/or ocular microfilariae levels, inter- and intra-individual variability in response to treatment, interindividual variability in frequency, type and severity of adverse reactions as well as possible differences between onchocerciasis-endemic areas [22]. The decisions should carefully weigh any projected population-based benefits vs. the risks to even a small percentage of individuals. If the benefit of the proposed ‘pretreat and treat’ strategy compared to MDAi or to other ATS is demonstrated, knowledge of the determinants of adverse reactions to DEC treatment in even a very small percentage of individuals could pave the way to ATS which exclude individuals at risk from DEC treatment, as was done for the small percentage of individuals with high Loa loa microfilaraemia at risk of severe and serious reactions to ivermectin [31, 42,43,44,45,46, 100, 101].

Copyright

World Health Organization 2023. This article is published under the CC BY 3.0 IGO license (https://creativecommons.org/licenses/by/3.0/igo) which permits use, sharing, adaptation, distribution, and reproduction in any medium or format, as long as the original work is properly cited, appropriate credit is given to the IGO, a link is provided to the Creative Commons licence, and any changes made are indicated. In any use of this article, there should be no suggestion that WHO endorses any specific organization, products, or services. The use of the WHO logo is not permitted. This notice should be preserved along with the article’s original URL.

Availability of data and materials

Participants consented to publication of summaries of the results, not to sharing of their individual data. Consequently, the Sponsor (WHO) and the authors do not have the participants’ permission to make individual participant data publicly available. Individuals wanting to analyze the data should contact the Sponsor (tdr@who.int) and Medicines Development for Global Health to which WHO has licensed the data (via mark. sullivant@medicinesdevelopment.com or https:// www.medicinesdevelopment.com/contact). Requests should include the objectives, data analysis plan and plans to obtain applicable Ethics Committee approvals and involve the investigators (co-authors on this manuscript) and commitment to not share the data with anybody else.

Abbreviations

AE:

Adverse event

CDTI:

Community directed treatment with ivermectin

CI:

95% Confidence interval

Day 4:

Day 4 after treatment

DEC:

Diethylcarbamazine

IA:

Ivermectin, albendazole

IDA:

Ivermectin, diethylcarbamazine and albendazole

IoI:

Intensity of infection (pre-treatment skin microfilariae density)

Month 1, 6, 12, 18:

Month 1, 6, 12, 18 after treatment

MDAi:

Mass drug administration of ivermectin

MedDRA:

Medical Dictionary for Regulatory Activities

mf:

Microfilaria

mfAC:

Live microfilariae in the anterior chambers

mfCOR:

Microfilariae in the cornea

OCRC:

Onchocerciasis Chemotherapy Research Center (Hohoe, Ghana)

OR:

Odds ratio

PreTx:

Pre-treatment

SmfD:

Skin microfilariae density (mf/mg skin)

References

  1. World Health Organization. Onchocerciasis and its Control. Report of a WHO Expert Committee on Onchocerciasis Control: World Health Organization; 1995. p. 1995.

    Google Scholar 

  2. Boatin B. The Onchocerciasis Control Programme in West Africa (OCP). Ann Trop Med Parasitol. 2008;102:13–7.

    Article  PubMed  Google Scholar 

  3. Sauerbrey M. The Onchocerciasis Elimination Program for the Americas (OEPA). Ann Trop Med Parasitol. 2008;102:25–9.

    Article  PubMed  Google Scholar 

  4. Fobi G, Yameogo L, Noma M, Aholou Y, Koroma JB, Zoure HM, et al. Managing the Fight against Onchocerciasis in Africa: APOC Experience. PLoS Negl Trop Dis. 2015;9:e0003542.

    Article  PubMed  PubMed Central  Google Scholar 

  5. Remme JHF, Boatin B, Boussinesq M. Helminthic Diseases: Onchocerciasis and Loiasis. In: Quah SR, Cockerham WC, editors. The International Encyclopedia of Public Health, vol. 3. 2nd ed. Oxford: Elsevier; 2017. p. 576–87.

    Chapter  Google Scholar 

  6. Al-Kubati AS, Mackenzie CD, Boakye D, Al-Qubati Y, Al-Samie AR, Awad IE, et al. Onchocerciasis in Yemen: moving forward towards an elimination program. Int Health. 2018;10:89–96.

    Article  Google Scholar 

  7. World Health Organization. Elimination of human onchocerciasis: progress report, 2020 - Elimination de l’onchocercose humaine: rapport de situation, 2020. Wkly Epidemiol Rec. 2021;96:557–67.

    Google Scholar 

  8. Dadzie Y, Neira M, Hopkins D. Final report of the Conference on the eradicability of Onchocerciasis. Filaria J. 2003;2(1):2.

  9. Kuesel AC. Research for new drugs for elimination of onchocerciasis in Africa. Int J Parasitol Drugs Drug Resist. 2016;6:272–86.

    Article  PubMed  PubMed Central  Google Scholar 

  10. Pfarr KM, Krome AK, Al-Obaidi I, Batchelor H, Vaillant M, Hoerauf A, et al. The pipeline for drugs for control and elimination of neglected tropical diseases: 1. Anti-infective drugs for regulatory registration. Parasit Vectors. 2023;16:82.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Center for Disease Control and Prevention (CDC). Progress toward elimination of onchocerciasis in the Americas - 1993–2012. MMWR Morb Mortal Wkly Rep. 2013;62(20):405–8.

  12. Sauerbrey M, Rakers LJ, Richards FO. Progress toward elimination of onchocerciasis in the Americas. Int Health. 2018;10:i71–8.

    Article  PubMed  Google Scholar 

  13. World Health Organization. Elimination of human onchocerciasis: progress report, 2018–2019. Wkly Epidemiol Rec. 2019;94:513–23.

    Google Scholar 

  14. World Health Organization = Organisation mondiale de la S. Progress in eliminating onchocerciasis in the WHO Region of the Americas: Advances towards interrupting the transmission of onchocerciasis from the latest preliminary serological assessments conducted in parts of the Yanomami Focus Area, 2018–2022 – Progrès vers l’élimination de l’onchocercose dans la Région OMS des Amériques: récentes analyses sérologiques préliminaires dans certaines parties de la zone Yanomami, indiquant des progrès vers l’interruption de la transmission, 2018–2022. Weekly Epidemiological Record = Relevé épidémiologique hebdomadaire. 2023;98(39):453–7.

  15. World Health Organization = Organisation mondiale de la S. Elimination of human onchocerciasis: progress report, 2022–2023 – Élimination de l’onchocercose humaine: rapport de situation, 2022–2023. Weekly Epidemiological Record = Relevé épidémiologique hebdomadaire. 2023;98(45):572–82.

  16. Diawara L, Traore MO, Badji A, Bissan Y, Doumbia K, Goita SF, et al. Feasibility of onchocerciasis elimination with ivermectin treatment in endemic foci in Africa: first evidence from studies in Mali and Senegal. PLoS Negl Trop Dis. 2009;3:e497.

    Article  PubMed  PubMed Central  Google Scholar 

  17. Traore MO, Sarr MD, Badji A, Bissan Y, Diawara L, Doumbia K, et 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:e1825.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Tekle AH, Zoure HG, Noma M, Boussinesq M, Coffeng LE, Stolk WA, et al. Progress towards onchocerciasis elimination in the participating countries of the African Programme for Onchocerciasis Control: epidemiological evaluation results. Infect Dis Poverty. 2016;5:66–91.

    Article  PubMed  PubMed Central  Google Scholar 

  19. World Health Organization, African Programme for Onchocerciasis Control (APOC). Report of the consultative meetings on strategic options and alternative treatment strategies for accelerating onchocerciasis elimination in Africa. Ouagadougou; 2015 12/23/2015.

  20. World Health Organization. Ending the neglect to attain the sustainable development goals: a road map for neglected tropical diseases 2021–2030. Geneva; 2020 2020.

  21. Awadzi K, Opoku NO, Attah SK, Lazdins-Helds J, Kuesel AC. A Randomized, Single-Ascending-Dose, Ivermectin-Controlled, Double-Blind Study of Moxidectin in Onchocerca volvulus Infection. PLoS Negl Trop Dis. 2014;8:e2953.

    Article  PubMed  PubMed Central  Google Scholar 

  22. Bakajika D, Kanza EM, Opoku NO, Howard HM, Mambandu GL, Nyathirombo A, et al. Effect of a single dose of 8 mg moxidectin or 150 mug/kg ivermectin on O. volvulus skin microfilariae in a randomized trial: Differences between areas in the Democratic Republic of the Congo, Liberia and Ghana and impact of intensity of infection. PLoS Negl Trop Dis. 2022;16:e0010079.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Cotreau MM, Warren S, Ryan JL, Fleckenstein L, Vanapalli SR, Brown KR, et al. The antiparasitic moxidectin: safety, tolerability, and pharmacokinetics in humans. J Clin Pharmacol. 2003;43:1108–15.

    Article  CAS  PubMed  Google Scholar 

  24. Kinrade SA, Mason JW, Sanabria CR, Rayner CR, Bullock JM, Stanworth SH, et al. Evaluation of the Cardiac Safety of Long-Acting Endectocide Moxidectin in a Randomized Concentration-QT Study. Clin Transl Sci. 2018;89:9.

    Google Scholar 

  25. Korth-Bradley JM, Parks V, Chalon S, Gourley I, Matschke K, Cailleux K, et al. The effect of a high-fat breakfast on the pharmacokinetics of moxidectin in healthy male subjects: a randomized phase I trial. Am J Trop Med Hyg. 2012;86:122–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Korth-Bradley JM, Parks V, Chalon S, Gourley I, Matschke K, Gossart S, et al. Excretion of moxidectin into breast milk and pharmacokinetics in healthy lactating women. Antimicrob Agents Chemother. 2011;55:5200–4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Korth-Bradley JM, Parks V, Fio W, Chalon S, Gourley I, Matschke K, et al. Effect of moxidectin on CYP3A4 activity as evaluated by oral midazolam pharmacokinetics in healthy subjects. Clinical Pharmacology in Drug Development. 2014;3:151–7.

    Article  CAS  PubMed  Google Scholar 

  28. Opoku NO, Bakajika DK, Kanza E, Howard H, Attah SK, Tchatchu J-PL, et al. Efficacy of a single oral dose of 8 mg Moxidectin vs 150 ug/kg ivermectin in onchocerca volvulus infection: results of a randomized, double-blind single dose phase 3 study in areas without mass treatment with ivermectin in Liberia, Ghana and DRC. Trop Med Int Health. 2013;18 (1):58

  29. Tan B, Opoku N, Attah SK, Awadzi K, Kuesel AC, Lazdins-Helds J, et al. Pharmacokinetics of oral moxidectin in individuals with Onchocerca volvulus infection. PLoS Negl Trop Dis. 2022;16:e0010005.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Opoku NO, Bakajika DK, Kanza EM, Howard H, Mambandu GL, Nyathirombo A, et al. Single dose moxidectin versus ivermectin for Onchocerca volvulus infection in Ghana, Liberia, and the Democratic Republic of the Congo: a randomised, controlled, double-blind phase 3 trial. Lancet. 2018;392:1207–16.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Boussinesq M, Fobi G, Kuesel AC. Alternative treatment strategies to accelerate the elimination of onchocerciasis. International Health. 2018;10 (suppl_1):i40-i8.

  32. Krucken J, Holden-Dye L, Keiser J, Prichard RK, Townson S, Makepeace BL, et al. Development of emodepside as a possible adulticidal treatment for human onchocerciasis-The fruit of a successful industrial-academic collaboration. PLoS Pathog. 2021;17:e1009682.

    Article  PubMed  PubMed Central  Google Scholar 

  33. Ngwewondo A, Scandale I, Specht S. Onchocerciasis drug development: from preclinical models to humans. Parasitol Res. 2021;120:3939–64.

    Article  PubMed  PubMed Central  Google Scholar 

  34. Johnston KL, Hong WD, Turner JD, O'Neill PM, Ward SA, Taylor MJ. Anti-Wolbachia drugs for filariasis. Trends Parasitol. 2021.

  35. Ehrens A, Hoerauf A, Hubner MP. Current perspective of new anti-Wolbachial and direct-acting macrofilaricidal drugs as treatment strategies for human filariasis. GMS Infect Dis. 2022;10:02.

    Google Scholar 

  36. Pfarr KM, Krome AK, Al-Obaidi I, Batchelor H, Vaillant M, Hoerauf A, et al. The pipeline for drugs for control and elimination of neglected tropical diseases: 2. Oral anti-infective drugs and drug combinations for off-label use. Parasit Vectors. 2023;16:394.

    Article  PubMed  PubMed Central  Google Scholar 

  37. Verver S, Walker M, Kim YE, Fobi G, Tekle AH, Zoure HGM, et al. How Can Onchocerciasis Elimination in Africa Be Accelerated? Modeling the Impact of Increased Ivermectin Treatment Frequency and Complementary Vector Control. Clin Infect Dis. 2018;66:S267–74.

    Article  PubMed  PubMed Central  Google Scholar 

  38. Lakwo T, Oguttu D, Ukety T, Post R, Bakajika D. Onchocerciasis elimination: progress and challenges. Res Rep Trop Med. 2020;11:81–95.

    PubMed  PubMed Central  Google Scholar 

  39. Campillo JT, Bikita P, Hemilembolo M, Louya F, Missamou F, Pion SDS, et al. Safety and efficacy of levamisole in loiasis: a randomized, placebo-controlled, double-blind clinical trial. Clin Infect Dis. 2021;75:19.

    Article  PubMed Central  Google Scholar 

  40. Campillo JT, Boussinesq M, Bertout S, Faillie JL, Chesnais CB. Serious adverse reactions associated with ivermectin: A systematic pharmacovigilance study in sub-Saharan Africa and in the rest of the World. PLoS Negl Trop Dis. 2021;15:e0009354.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Campillo JT, Eiden C, Boussinesq M, Pion SDS, Faillie JL, Chesnais CB. Adverse reactions with levamisole vary according to its indications and misuse: A systematic pharmacovigilance study. Br J Clin Pharmacol. 2022;88:1094–106.

    Article  CAS  PubMed  Google Scholar 

  42. Kamgno J, Nana-Djeunga HC, Pion SD, Chesnais CB, Klion AD, Mackenzie CD, et al. Operationalization of the test and not treat strategy to accelerate the elimination of onchocerciasis and lymphatic filariasis in Central Africa. Int Health. 2018;10:i49–53.

    Article  PubMed  PubMed Central  Google Scholar 

  43. Kamgno J, Pion SD, Chesnais CB, Bakalar MH, D’Ambrosio MV, Mackenzie CD, et al. A test-and-not-treat strategy for onchocerciasis in Loa loa-endemic areas. N Engl J Med. 2017;377:2044–52.

    Article  PubMed  PubMed Central  Google Scholar 

  44. Wanji S, Nji TM, Hamill L, Dean L, Ozano K, Njouendou AJ, et al. Implementation of test-and-treat with doxycycline and temephos ground larviciding as alternative strategies for accelerating onchocerciasis elimination in an area of loiasis co-endemicity: the COUNTDOWN consortium multi-disciplinary study protocol. Parasit Vectors. 2019;12:574.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Blok DJ, Kamgno J, Pion SD, Nana-Djeunga HC, Niamsi-Emalio Y, Chesnais CB, et al. Feasibility of onchocerciasis elimination using a "test-and-not-treat" strategy in Loa loa co-endemic areas. Clin Infect Dis. 2021;72:e1047–55.

    Article  PubMed  Google Scholar 

  46. Chesnais CB, Pion SD, Boulle C, Gardon J, Gardon-Wendel N, Fokom-Domgue J, et al. Individual risk of post-ivermectin serious adverse events in subjects infected with Loa loa. EClinicalMedicine. 2020;28:100582.

    Article  PubMed  PubMed Central  Google Scholar 

  47. Abraham D, Graham-Brown J, Carter D, Gray SA, Hess JA, Makepeace BL, et al. Development of a recombinant vaccine against human onchocerciasis. Expert Rev Vaccines. 2021;78:1–12.

    Google Scholar 

  48. World Health Organization. Guideline: alternative mass drug administration regimens to eliminate lymphatic filariasis. Geneva: World Health Organization; 2017. p. 2017.

    Google Scholar 

  49. Fischer PU, King CL, Jacobson JA, Weil GJ. Potential value of triple drug therapy with ivermectin, diethylcarbamazine, and albendazole (IDA) to accelerate elimination of lymphatic filariasis and onchocerciasis in Africa. PLoS Negl Trop Dis. 2017;11:e0005163.

    Article  PubMed  PubMed Central  Google Scholar 

  50. WHO Expert Committee on Onchocerciasis, World Health Organization,. WHO Expert Committee on Onchocerciasis [meeting held in Geneva from 21 to 29 April 1986]: third report. World Health Organization Technical Report Series, no 752: World Health Organization Geneva; 1987.

  51. Awadzi K, Gilles HM. Diethylcarbamazine in the treatment of patients with onchocerciasis. Br J Clin Pharmacol. 1992;34:281–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. WHO Expert Committee on Onchocerciasis, World Health Organization,. WHO Expert Committee on Onchocerciasis [meeting held in Geneva from 29 June to 5 July 1965]: second report. 1966.

  53. Bryceson AD, Warrell DA, Pope HM. Dangerous reactions to treatment of onchocerciasis with diethylcarbamazine. Br Med J. 1977;1:742–4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Bird AC, El-Sheikh H, Anderson J, Fuglsang H. Changes in visual function and in the posterior segment of the eye during treatment of onchocerciasis with diethylcarbamazine citrate. Br J Ophthalmol. 1980;64:191–200.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Bird AC, El-Sheikh H, Anderson J, Fuglsang H. Visual loss during oral diethylcarbamazine treatment for onchocerciasis. Lancet. 1979;2:46.

    Article  CAS  PubMed  Google Scholar 

  56. Taylor HR. Recent developments in the treatment of onchocerciasis. Bull World Health Organ. 1984;62:509–15.

    CAS  PubMed  PubMed Central  Google Scholar 

  57. Duke BOL, Thylefors B, Rougemont A. Current views on the treatment of onchocerciasis with diethylcarbamazine citrate and suramin. Geneva: World Health Organization; 1981 1981. Contract No.: WHO/ONCHO/81.156.

  58. WHO Expert Committee on Onchocerciasis Control. Geneva Switzerland), World Health Organization, Onchocerciasis and its control: report of a WHO Expert Committee on Onchocerciasis Control. Geneva: World Health Organization; 1993. p. 1995.

    Google Scholar 

  59. Francis H, Awadzi K, Ottesen EA. The Mazzotti reaction following treatment of onchocerciasis with diethylcarbamazine: clinical severity as a function of infection intensity. Am J Trop Med Hyg. 1985;34:529–36.

    Article  CAS  PubMed  Google Scholar 

  60. Brieger WR, Okeibunor JC, Abiose AO, Ndyomugyenyi R, Kisoka W, Wanji S, et al. Feasibility of measuring compliance to annual ivermectin treatment in the African Programme for Onchocerciasis Control. Trop Med Int Health. 2007;12:260–8.

    Article  PubMed  Google Scholar 

  61. Brieger WR, Okeibunor JC, Abiose AO, Wanji S, Elhassan E, Ndyomugyenyi R, et al. Compliance with eight years of annual ivermectin treatment of onchocerciasis in Cameroon and Nigeria. Parasit Vectors. 2011;4:152.

    Article  PubMed  PubMed Central  Google Scholar 

  62. Brieger WR, Otusanya SA, Oke GA, Oshiname FO, Adeniyi JD. Factors associated with coverage in community-directed treatment with ivermectin for onchocerciasis control in Oyo State. Nigeria Trop Med Int Health. 2002;7:11–8.

    Article  PubMed  Google Scholar 

  63. Kifle B, Nigatu M. Compliance to a Five-Year Biannual Ivermectin Treatment for Onchocerciasis Elimination and Its Determinants among Adults in the Bench Maji Zone, Southwest Ethiopia: A Community-Based Cross-Sectional Study. J Parasitol Res. 2021;2021:8866639.

    Article  PubMed  PubMed Central  Google Scholar 

  64. Brieger WR, Okeibunor JC, Abiose AO, Ndyomugyenyi R, Wanji S, Elhassan E, et al. Characteristics of persons who complied with and failed to comply with annual ivermectin treatment. Trop Med Int Health. 2012;17:920–30.

    Article  PubMed  Google Scholar 

  65. Yirga D, Deribe K, Woldemichael K, Wondafrash M, Kassahun W. Factors associated with compliance with community directed treatment with ivermectin for onchocerciasis control in Southwestern Ethiopia. Parasit Vectors. 2010;3:48.

    Article  PubMed  PubMed Central  Google Scholar 

  66. Mushi V. Implementation Challenges of Community Directed Treatment with Ivermectin Program for Control of Onchocerciasis in Ulanga. Tanzania East Afr Health Res J. 2021;5:123–8.

    Article  PubMed  Google Scholar 

  67. Osue HO. Field-based evidence of single and few doses of annual ivermectin treatment efficacy in eliminating skin microfilaria load after a decade of intervention. Ethiop J Health Sci. 2017;27:129–38.

    Article  PubMed  PubMed Central  Google Scholar 

  68. Opoku NO, Doe F, Dubben B, Fetcho N, Fischer K, Fischer PU, et al. A randomized, open-label study of the tolerability and efficacy of one or three daily doses of ivermectin plus diethylcarbamazine and albendazole (IDA) versus one dose of ivermectin plus albendazole (IA) for treatment of onchocerciasis. PLoS Negl Trop Dis. 2023;17:e0011365.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Opoku NO, Gyasi ME, Doe F, Lew D, Hong AR, Chithenga S, et al. A Reevaluation of the tolerability and effects of single-dose ivermectin treatment on onchocerca volvulus microfilariae in the skin and eyes in Eastern Ghana. Am J Trop Med Hyg. 2021;106:740–5.

    Article  PubMed  PubMed Central  Google Scholar 

  70. Gardon J, Gardon-Wendel N, Demanga N, Kamgno J, Chippaux JP, Boussinesq M. Serious reactions after mass treatment of onchocerciasis with ivermectin in an area endemic for Loa loa infection. Lancet. 1997;350:18–22.

    Article  CAS  PubMed  Google Scholar 

  71. Twum-Danso NA, Meredith SE. Variation in incidence of serious adverse events after onchocerciasis treatment with ivermectin in areas of Cameroon co-endemic for loiasis. Trop Med Int Health. 2003;8:820–31.

    Article  PubMed  Google Scholar 

  72. Twum-Danso NA. Serious adverse events following treatment with ivermectin for onchocerciasis control: a review of reported cases. Filaria J. 2003;2:S3.

    Article  PubMed  PubMed Central  Google Scholar 

  73. Mectizan Expert Committee and APOC Technical Consultative Committee. Recommendations for the treatment of onchocerciasis with Mectizan in areas co-endemic for onchocerciasis and Loiasis2004 12/04/2023. https://mectizan.org/wp-content/uploads/2018/06/englishmectccloarecs-june04.pdf.

  74. Awadzi K. The chemotherapy of onchocerciasis II Quantitation of the clinical reaction to microfilaricides. Ann Trop Med Parasitol. 1980;74:189–97.

    Article  CAS  PubMed  Google Scholar 

  75. Hero M, Bird AC, Awadzi K. Quantification of the ocular reactions to microfilaricides in the chemotherapy of onchocerciasis. Eye (Lond). 1992;6:93–6.

    Article  PubMed  Google Scholar 

  76. Taylor HR, Greene BM. Ocular changes with oral and transepidermal diethylcarbamazine therapy of onchocerciasis. Br J Ophthalmol. 1981;65:494–502.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Awadzi K, Dadzie KY, Schulz-Key H, Haddock DR, Gilles HM, Aziz MA. Ivermectin in onchocerciasis. Lancet. 1984;2:921.

    Article  CAS  PubMed  Google Scholar 

  78. Awadzi K, Dadzie KY, Shulz-Key H, Haddock DR, Gilles HM, Aziz MA. The chemotherapy of onchocerciasis X An assessment of four single dose treatment regimes of MK-933 (ivermectin) in human onchocerciasis. Ann Trop Med Parasitol. 1985;79:63–78.

    Article  CAS  PubMed  Google Scholar 

  79. Coulaud JP, Lariviere M, Aziz MA, Gervais MC, Gaxotte P, Deluol AM, et al. Ivermectin in onchocerciasis. Lancet. 1984;2:526–7.

    Article  CAS  PubMed  Google Scholar 

  80. Greene BM, Taylor HR, Cupp EW, Murphy RP, White AT, Aziz MA, et al. Comparison of ivermectin and diethylcarbamazine in the treatment of onchocerciasis. N Engl J Med. 1985;313:133–8.

    Article  CAS  PubMed  Google Scholar 

  81. Taylor HR, Murphy RP, Newland HS, White AT, D’Anna SA, Keyvan-Larijani E, et al. Treatment of onchocerciasis. The ocular effects of ivermectin and diethylcarbamazine. Arch Ophthalmol. 1986;104:863–70.

    Article  CAS  PubMed  Google Scholar 

  82. Lariviere M, Vingtain P, Aziz M, Beauvais B, Weimann D, Derouin F, et al. Double-blind study of ivermectin and diethylcarbamazine in African onchocerciasis patients with ocular involvement. Lancet. 1985;2:174–7.

    Article  CAS  PubMed  Google Scholar 

  83. Awadzi K, Dadzie KY, Schulz-Key H, Gilles HM, Fulford AJ, Aziz MA. The chemotherapy of onchocerciasis. XI. A double-blind comparative study of ivermectin, diethylcarbamazine and placebo in human onchocerciasis in northern Ghana. Ann Trop Med Parasitol. 1986;80:433–42.

    Article  CAS  PubMed  Google Scholar 

  84. Dadzie KY, Bird AC, Awadzi K, Schulz-Key H, Gilles HM, Aziz MA. Ocular findings in a double-blind study of ivermectin versus diethylcarbamazine versus placebo in the treatment of onchocerciasis. Br J Ophthalmol. 1987;71:78–85.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Diallo S, Aziz MA, Lariviere M, Diallo JS, Op-Mar I, N’Dir O, et al. A double-blind comparison of the efficacy and safety of ivermectin and diethylcarbamazine in a placebo controlled study of Senegalese patients with onchocerciasis. Trans R Soc Trop Med Hyg. 1986;80:927–34.

    Article  CAS  PubMed  Google Scholar 

  86. Dadzie KY, Awadzi K, Bird AC, Schulz-Key H. Ophthalmological results from a placebo controlled comparative 3-dose ivermectin study in the treatment of onchocerciasis. Trop Med Parasitol. 1989;40:355–60.

    CAS  PubMed  Google Scholar 

  87. Lariviere M, Beauvais B, Aziz M, Garin JF, Abeloos J, Derouin F, et al. Etude en Cote-d'Ivoire (1985–1987) de l'éfficacité et de la tolérance de l'ivermectin (Mectizan) dans l'onchocercose humaine. I Etude comparative en double insu de 220 sujets onchocerquiens traité par dose unique orale de 100, 150 ou 200 mcg/kg. [A study in the Ivory Coast (1985–1987) of the efficacy and tolerance of ivermectin (Mectizan) in human onchocerciasis. I. A comparative double-blind study of 220 patients with onchocerciasis treated with a single oral dose of 100, 150 or 200 mcg/kg]. Bull Soc Pathol Exot Filiales. 1989;82(1):35–47.

  88. Newland HS, White AT, Greene BM, D’Anna SA, Keyvan-Larijani E, Aziz MA, et al. Effect of single-dose ivermectin therapy on human Onchocerca volvulus infection with onchocercal ocular involvement. Br J Ophthalmol. 1988;72:561–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Albiez EJ, Newland HS, White AT, Kaiser A, Greene BM, Taylor HR, et al. Chemotherapy of onchocerciasis with high doses of diethylcarbamazine or a single dose of ivermectin: microfilaria levels and side effects. Trop Med Parasitol. 1988;39:19–24.

    CAS  PubMed  Google Scholar 

  90. Taylor HR, Semba RD, Newland HS, Keyvan-Larijani E, White A, Dukuly Z, et al. Ivermectin treatment of patients with severe ocular onchocerciasis. Am J Trop Med Hyg. 1989;40:494–500.

    Article  CAS  PubMed  Google Scholar 

  91. Soboslay PT, Newland HS, White AT, Erttmann KD, Albiez EJ, Taylor HR, et al. Ivermectin effect on microfilariae of Onchocerca volvulus after a single oral dose in humans. Trop Med Parasitol. 1987;38:8–10.

    CAS  PubMed  Google Scholar 

  92. Duke BO, Moore PJ, Vincelette J. Factors influencing the passage of Onchocerca volvulus microfilariae into the urine. Tropenmed Parasitol. 1975;26:449–68.

    CAS  PubMed  Google Scholar 

  93. Duke BO, Vincelette J, Moore PJ. The populatin dynamics of Onchocerca volvulus microfilariae during treatment with suramin and diethylcarbamazine. Tropenmed Parasitol. 1976;27:133–44.

    CAS  PubMed  Google Scholar 

  94. Duke BO, Vincelette J, Moore PJ. Microfilariae in the cerebrospinal fluid, and neurological complications, during treatment of onchocerciasis with diethylcarbamazine. Tropenmed Parasitol. 1976;27:123–32.

    CAS  PubMed  Google Scholar 

  95. Basset D, Bouree P, Basset A, Lariviere M. Effects of diethylcarbamazine and ivermectin on the mobilization of microfilariae of Onchocerca volvulus. Pathol Biol (Paris). 1989;37:668–72.

    CAS  PubMed  Google Scholar 

  96. Fuglsang H, Anderson J. Microfilariae of Onchocerca volvulus in blood and urine before, during, and after treatment with diethylcarbamazine. J Helminthol. 1974;48:93–7.

    Article  CAS  PubMed  Google Scholar 

  97. Kura K, Milton P, Hamley JID, Walker M, Bakajika DK, Kanza EM, et al. Can mass drug administration of moxidectin accelerate onchocerciasis elimination in Africa? Philos Trans R Soc Lond B Biol Sci. 1887;2023:20220277.

    Google Scholar 

  98. Newell ED. Effect of mass treatments with ivermectin, with only partial compliance, on prevalence and intensity of O. volvulus infection in adults and in untreated 4 and 5 year-old children in Burundi. Trop Med Int Health. 1997;2:912–6.

    Article  CAS  PubMed  Google Scholar 

  99. Wanji S, Kengne-Ouafo JA, Esum ME, Chounna PW, Adzemye BF, Eyong JE, et al. Relationship between oral declaration on adherence to ivermectin treatment and parasitological indicators of onchocerciasis in an area of persistent transmission despite a decade of mass drug administration in Cameroon. Parasit Vectors. 2015;8:667.

    Article  PubMed  PubMed Central  Google Scholar 

  100. Pion SD, Nana-Djeunga H, Niamsi-Emalio Y, Chesnais CB, Deleglise H, Mackenzie C, et al. Implications for annual retesting after a test-and-not-treat strategy for onchocerciasis elimination in areas co-endemic with Loa loa infection: an observational cohort study. Lancet Infect Dis. 2020;20:102–9.

    Article  PubMed  PubMed Central  Google Scholar 

  101. Eyebe S, Sabbagh A, Pion SD, Nana-Djeunga HC, Kamgno J, Boussinesq M, et al. Familial aggregation and heritability of Loa loa microfilaremia. Clin Infect Dis. 2018;66:751–7.

    Article  PubMed  Google Scholar 

Download references

Acknowledgements

We acknowledge the contribution to data collection in Liberia of Mr. Mawolo Kpawor, who died in October 2016. We acknowledge Dr Fatorma Bolay who directed study center creation and preparation, community engagement, parasitologist training and management of the Liberian study site (see [30]). He died in March 2021. We also gratefully acknowledge Dr Safari L Masembe, a co-investigator in the study team in Nord Kivu. He passed away on 13 January 2022. We furthermore acknowledge the contribution of Jean-Pierre Lotsima who contributed to the data from Ituri. He passed way in March 2023. For others whose contribution to the study we want to acknowledge, see [30].

We are particularly grateful to all study participants for their co-operation.

The authors alone are responsible for the views expressed which do not necessarily represent the views, decisions or policies of the institutions with which the authors are affiliated.

Funding

WHO/TDR funded this study, utilizing contributions from the WHO African Programme for Onchocerciasis Control (APOC), 6.3 million $US from Wyeth and following its acquisition by Pfizer, Pfizer, and WHO/TDR donor countries. Wyeth provided drug for this study and contributed to the study protocol. Wyeth prepared the submissions to the Ministries of Health and provided data management services until July 3, 2011. Pfizer was not further involved in this study in any way, including data verification or analysis and has not commented on this manuscript.

Author information

Authors and Affiliations

Authors

Contributions

NOO, DKB, EMK and HH were the principal investigators responsible for site preparation, community engagement, participant recruitment, study implementation, data review and data query resolution in Ghana, Nord Ituri and Nord-Kivu (DRC), and Liberia, respectively. EMK, AN and JPL did the ophthalmological examinations in Nord-Kivu, Nord Ituri and Liberia, respectively. GLM, MMN, DRU, FN, KKK, KK, KMP, SKA, GO, SA and MM were co-investigators responsible for recruiting, examining and following up participants and collecting data. MV was responsible for data management after WHO became sole study sponsor, co-designed the statistical analysis and conducted the statistical analyses. CMH participated in site training and study initiation, managed conduct, supervised study monitoring and close-out, and supported investigators and MV in data management after TDR became sole study sponsor. ACK was involved in study design and protocol finalization with input from the late Dr K Awadzi, TDR staff, TDR external advisors and advice from the European Medicines Agency. ACK managed research center creation in Liberia and the Democratic Republic of the Congo, study preparation and set up, did staff training and study initiation, managed study conduct, co-designed the statistical analysis, reviewed the data analyses, drafted the manuscript and finalized it with co-author input. All authors reviewed the manuscript and approved the submitted version.

Corresponding author

Correspondence to Annette C. Kuesel.

Ethics declarations

Ethics approval and consent to participate

As previously reported [22, 30], the protocol, information documents for potential participants, participant consent and assent forms and study conduct were approved by the Ghana Food and Drugs Authority and the Ghana Health Service Ethics Review Committee, the Liberia Ministry of Health and Social Welfare and the Ethics Committee of the Liberia Institute for Biomedical Research, the Ministère de la Santé Publique of DRC and the Ethics Committee of the Ecole de la Santé Publique Université de Kinshasa in DRC and the WHO Ethics Review Committee.

Participants documented their consent or assent with parental consent to study participation through signature or thumbprint in the presence of a literate witness in their villages.

Competing interests

ACK and CMH are staff of WHO which funded the work of all co-authors on the study whose data are analysed here through its department UNICEF/UNDP/World Bank/WHO Special Programme for Research and Training in Tropical Diseases (TDR). ACK retired from WHO in March 2023.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Annette C. Kuesel retired since 1 March 2023.

Supplementary Information

13071_2023_6087_MOESM1_ESM.pdf

Additional file 1: Fig S1. mfAC pre-treatment, 4 days, 1, 6, 12 and 18 months post-treatment among participants with < 10 mfAC pre-treatment by pre-treatment SmfD. Fig S2. mfAC pre-treatment, 4 days, 1, 6, 12 and 18 months post-treatment among participants with ≥ 10 mfAC pre-treatment by pre-treatment SmfD. Fig S3. mfAC 1, 6, 12 and 18 months post-treatment among participants with < 10 mfAC pre-treatment by post-treatment SmfD. Fig S4. mfAC 1, 6, 12 and 18 months post-treatment among participants with ≥ 10 mfAC pre-treatment by post-treatment SmfD. Table S1. Participants with both eyes evaluated at each timepoint by pre-treatment SmfD and mfAC category and sex. Table S2. Mean (SD), minimum and maximum mfAC levels among participants with increases from pre-treatment to Day 4 or Month 1 and from Day 4 to Month 1 resulting in mfAC levels in a higher mfAC category. Table S3. Number (%) of participants by mfAC category on Day 4 or Month 1 after treatment by pre-treatment and Day 4 mfAC category and treatment arm. Table S4. Output linear model for mfAC. Table S5. Ocular medical history of study participants by study area. Table S6. Output of final logistic model of the factors impacting the risk to have at least one ocular Mazzotti reaction. Table S7. Number (%) of participants with ocular adverse events starting within 1 and between month 1 and end of 6 months after treatment by ocular AE based on MedDRA coding. Table S8. Number of ocular adverse events starting within 1 and between month 1 and end of 6 months after treatment by ocular AE based on MedDRA coding. Table S9. Output of final logistic model of the factors and covariates impacting the risk to have at least one ocular adverse event. Table S10. Literature data on mfAC number change early after treatment with diethylcarbamazine, ivermectin and suramin. Table S11. Microfilariae in the blood and urine before and after treatment (data from the moxidectin Phase 2 study, across all participants and for participants with > 20mf/mg skin).

Rights and permissions

Open Access This article is licensed under the terms of the Creative Commons Attribution 3.0 IGO License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the WHO, provide a link to the Creative Commons licence and indicate if changes were made.

The use of the WHO’s name, except in reference to the article, and the use of the WHO’s logo, is not authorized as part of this licence. The link provided below includes additional terms and conditions of the licence.

The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder.To view a copy of this licence, visit http://creativecommons.org/licenses/by/3.0/igo/

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Kanza, E.M., Nyathirombo, A., Larbelee, J.P. et al. Onchocerca volvulus microfilariae in the anterior chambers of the eye and ocular adverse events after a single dose of 8 mg moxidectin or 150 µg/kg ivermectin: results of a randomized double-blind Phase 3 trial in the Democratic Republic of the Congo, Ghana and Liberia. Parasites Vectors 17, 137 (2024). https://doi.org/10.1186/s13071-023-06087-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13071-023-06087-3

Keywords