- Systematic Review
- Open access
- Published:
Antifilarial treatment strategies: a systematic review and network meta-analysis
BMC Infectious Diseases volume 25, Article number: 712 (2025)
Abstract
Background
The World Health Organization (WHO) prescribes mass drug administration (MDA) to eradicate lymphatic filariasis within endemic populations. The WHO endorsed using ivermectin with diethylcarbamazine and albendazole (IDA) for MDA in specific settings devoid of onchocerciasis or loiasis. Still, the utilization of IDA in sub-Saharan Africa is restricted due to the potential of diethylcarbamazine to induce severe adverse ocular events in individuals with onchocerciasis.
Aim
We aim to investigate all documented combinations of antifilarial drugs available in the literature using a network meta-analysis (NWM) design, focusing specifically on the treatment of Lymphatic Filariasis (LF).
Methods
A meticulous search was conducted across four electronic databases to identify pertinent studies. Subsequently, a frequentist NWM was executed. Risk ratios (RRs) served as the effect size metric for categorical outcomes, each with a 95% confidence interval (CI).
Results
Our study encompassed 45 studies, including 61,369 patients. At six months, multiple doses of diethylcarbamazine plus albendazole (multiple DA) regimens demonstrated superior efficacy in reducing microfilaremia compared to a single intake of DA, diethylcarbamazine, ivermectin, and albendazole with RR and CI as follows: 0.37 [0.19; 0.72], 0.35 [0.17; 0.69], 0.30 [0.14; 0.64], and 0.28 [0.13; 0.57]. The combination of ivermectin plus albendazole (IA) also showed significant efficacy against the use of each of these drugs alone, with RR: 0.74 [0.57; 0.96] for ivermectin and 0.69 [0.53; 0.89] for albendazole, while diethylcarbamazine combined with albendazole showed substantial superiority over albendazole alone or placebo: RR = 0.09 [0.02; 0.36] and 0.08 [0.02; 0.34], respectively. By the twelfth month, diethylcarbamazine, followed by albendazole, ranked superior to IDA and DA: 0.12 [0.02; 0.89] and 0.11 [0.01; 0.79], respectively. At 24 months, no significant differences were found among the assessed drugs in reducing microfilaremia. The comparisons revealed no significant differences between the drug combinations we studied regarding safety and adverse events.
Conclusion
Multiple doses of the DA regimen showed superior efficacy in reducing microfilaremia compared to combinations involving IA, diethylcarbamazine, ivermectin, and albendazole at six and twelve months. However, by the twenty-four-month, no significant differences were found. Safety profiles among interventions were generally comparable, with no specific drug showing superiority in adverse events.
Introduction
Lymphatic Filariasis (LF) is a parasitic infection transmitted by mosquitoes, causing lymphatic issues like hydroceles and elephantiasis. The WHO’s Global Programme to Eliminate LF (GPELF) has distributed over 9 billion doses of medication between 2000 and 2021 through mass drug administration (MDA). Despite progress in reducing the risk of LF transmission and the overall burden of LF, MDA is still recommended for about 885 million people in 45 countries to combat the disease [1]. Previously, GPELF recommended tailored mass drug administration: ivermectin plus albendazole (IA) where onchocerciasis exists, albendazole twice yearly in LF and Loa loa co-endemic areas, and diethylcarbamazine (DEC) plus albendazole (DA) elsewhere for combating LF [1].
Following clinical trials conducted in Papua New Guinea and Coˆte d’Ivoire, which demonstrated the superior efficacy of the combination of ivermectin plus DEC plus albendazole (IDA) compared to two-drug combinations [2,3,4], subsequent large-scale multinational safety trials involving over 26,000 participants revealed no escalation in treatment-emergent adverse events with IDA in contrast to DA [5]. As a result, the WHO endorsed using IDA for MDA in specific settings devoid of onchocerciasis or loiasis [6]. Nevertheless, the utilization of IDA in sub-Saharan Africa is restricted due to the potential of DEC to induce severe adverse ocular events in individuals with onchocerciasis. Hence, there persists a critical requirement for a safe and more efficacious treatment regimen applicable for MDA within LF elimination initiatives operating in onchocerciasis co-endemic regions.
In 2018, the US Food and Drug Administration authorized the utilization of moxidectin for the treatment of onchocerciasis [7]. Moxidectin, classified as a macrocyclic lactone akin to Ivermectin (IVM), exhibits increased lipophilicity, a larger volume of distribution, and an extended half-life. Studies conducted on onchocerciasis in Liberia, the Democratic Republic of Congo, and Ghana revealed moxidectin to outperform IVM in the clearance of microfilaremia among individuals with onchocerciasis while maintaining a treatment-emergent adverse events profile similar to that of ivermectin [7]. Moreover, antibiotics such as doxycycline, targeting the Wolbachia endosymbiont within the parasite, have introduced novel prospects for macrofilaricidal therapy [8, 9]. These antibiotics have also paved the way for abbreviated combined therapy approaches [10, 11]. Our study aims to investigate all documented combinations of antifilarial drugs available in the literature using a network meta-analysis (NWM) design. This approach is intended to evaluate the efficacy and safety of these combinations relative to each other.
Methods
The study’s design adhered to the guidelines outlined in the Cochrane Handbook for Systematic Reviews of Interventions and followed the recommendations outlined in the Preferred Reporting Items for Systematic Reviews and Meta-analyses (PRISMA) extension for network meta-analysis [12, 13]. We chose NMA because it allows for the simultaneous comparison of multiple interventions by integrating both direct and indirect evidence across a network of studies. This method offers several advantages over traditional meta-analysis, including the ability to compare multiple treatments comprehensively, yielding more precise estimates, ranking interventions to identify the most effective ones, and incorporating indirect comparisons to utilize more data.
Literature search
A detailed search was systematically conducted using multiple electronic databases, including Web of Science, MEDLINE via PubMed, Scopus, and Cochrane CENTRAL, covering the period from their inception until 29 October 2023. The search strategy involved a combination of terms related to filariasis and antifilarial drugs. The full search strategies and the number of publications retrieved from each database are provided in Supplementary Table 1.
Eligibility criteria
We included randomized controlled trials (RCTs) that compared various drug interventions such as DEC, ivermectin, albendazole, doxycycline, and moxidectin in every feasible combination and dosage documented in the literature (Table 1). These interventions were administered to individuals confirmed to have LF or to communities residing in areas where LF was known to be endemic. We included studies with any of the following endpoints: (1) measuring indicative of transmission potential (microfilariae prevalence); our primary endpoint; (2) studies assessed markers associated with adult worm infection, encompassing antigenemia prevalence; (3) studies compile data related to adverse events linked to the interventions.
We excluded non-English articles, animal studies, abstracts without full text available, and non-published data. Non-English articles were excluded due to potential translation challenges and resource limitations.
Data extraction
For the data extraction process, we use offline data extraction sheets; we collect information regarding study characteristics and outcome data from each included study. The extracted data illustrated the following aspects: study design, geographic location, study arms, population characteristics: number and age of participants in each arm, gender distribution, treatment description, and follow-up duration; as regards outcomes of interest, it was microfilaraemia prevalence, markers of adult worm infection and safety outcomes presented in the form of adverse events.
Risk of bias assessment
Our retrieved RCTs were assessed for interventional studies using the Cochrane Risk of Bias Assessment Tool 1 (ROB1) [14]. This tool includes the following domains: selection, performance, detection, attrition, reporting, and other possible sources of bias. The authors’ judgment was categorized as “high,” “low,” and “unclear” risk of bias. A third assessor was consulted in case of discrepancy.
Data analysis
We conducted a frequentist network meta-analysis (NWM) utilizing aggregate data to derive network estimates for the outcomes under scrutiny. In quantifying dichotomous outcomes, we employed risk ratios (RRs) as the effect size, accompanied by a 95% confidence interval (CI). A significance level of p < 0.05 was adopted as the threshold for statistical significance. Statistical heterogeneity was also assessed among the pooled studies using the I2 statistic and the chi-squared test. A p-value less than 0.1 was interpreted as indicative of heterogeneity, while an I2 value equal to or exceeding 50% indicated high heterogeneity. All statistical analyses were done using R software [15].
Results
Literature search results
Our literature search process across the distinct databases yielded a total of 1538 studies. Subsequently, after eliminating duplicate entries, 1485 records were evaluated based on titles and abstracts. Title and abstract screening yields 76 relevant articles for full-text assessment. Based on our performed inclusion criteria, only 45 studies were included in our systematic review; of them, 31 studies were eligible for NWM [2,3,4,5, 10, 11, 16,17,18,19,20,21,22,23,24,25,26,27,28,29,30,31,32,33,34,35,36,37,38,39,40,41,42,43,44,45,46,47,48,49,50,51,52,53,54]. Supplementary Fig. 1 represents the PRISMA flow diagram for selecting eligible studies.
Characteristics of the included studies
Our 45 RCTs were conducted mainly in India, the USA, Papua New Guinea, and 15 other countries around the world, encompassing a total of 61,369 patients. In our study, patients were subjected to ten different treatment regimens, with a total of 31,471 patients being given the IDA regimen, 5,716 were treated with IA, 989 were given albendazole, and 928 were given Placebo. Demographic characteristics of patients and a summary of the included studies are provided in Supplementary Table 2.
Risk of bias evaluation
According to the Cochrane ROB1 tool, our 45 RCTs were ranked from fair to poor quality. However, in terms of each domain, six studies represented high risk regarding selection biases, and 15 reported low risk in performance bias; only eight showed unclear detection bias, while twenty-two studies were judged as being either unclear or at high risk of attrition bias, collectively most of our included studies had a high risk regarding performance and attrition biases. However, most studies represented low risk in the domain of reporting bias. Supplementary Fig. 2 visually depicts the risk of bias summary according to the Cochrane ROB 1 tool.
Outcomes
Primary outcomes
Measures of transmission potential
Microfilaremia was assessed at six, twelve, and twenty-four months after treatment, at six months. Our pooled RR showed that multiple DA regimen was significantly superior in reducing microfilaraemia compared to the following drugs, Dox, IA, DA, DEC &Dox, DEC, IVM, ALB, and placebo with RRs as follows: 0.42, 0.40, 0.37, 0.35, 0.35, 0.30, 0.28 and 0.26 respectively. Also, IA showed significant efficacy in microfilariemia reduction compared to IVM, ALB, and placebo with 0.74, 0.69, and 0.64, respectively. However, DEC then ALB administration is significantly better than ALB alone or placebo with RRs 0.09 and 0.08 respectively; the same showed for IDA when compared to ALB or placebo with 0.13 and 0.12 respectively. Heterogeneity was significant across the pooled studies at the three-time points with chi2p < 0.0001. Figure 1 illustrates this outcome’s forest plot, net league, and plot.
However, at 12 months of follow-up DEC then, ALB ranked superior to IDA, DA, “ALB and Dox,” DEC, IA, Placebo, IVM, Dox, and ALB with 0.12, 0.11, 0.05, 0.10, 0.03, 0.03, 0.03, 0.03 and 0.03 respectively, similarly multiple DA was significantly better than IDA, DA, DEC, IA, Placebo, IVM, Dox, and ALB. The pooled studies at 12 months showed heterogeneity with chi2p < 0.0006 (Fig. 2).
On the other hand, our NWM at 24 months revealed No significant difference between any of these drugs, multiple DA, IDA, IA, ALB, DA, and DEC, regarding this outcome. (Fig. 3).
Secondary outcomes
Markers of adult worm infection
In terms of adult worm infection markers, they were assessed at two-time points (six and twelve months) by measuring the prevalence of antigenemia. Based on antigenemia prevalence at six months, no statistically significant differences were shown between DA and these drugs ALB, Placebo, and DEC. Similarly, comparison between ALB, Placebo, and DEC exhibited differences; our pooled studies were homogenous with chi2p = 0.16 (Fig. 4).
Regarding antigenemia assessment at 12 months, our network model did not significantly prioritize any of the following drugs over each other: “DEC then ALB”, “DEC and Dox”, IDA, IA, DA, IVM, ALB, DEC, and Placebo. “DEC then ALB” compared to IDA and “DEC and Dox” showed RRs 1.04 [0.75; 1.43] and 0.86 [0.65; 1.15], respectively. Homogeneity was shown among the pooled studies with chi2p = 0.65 (Fig. 5).
Safety and adverse events
In terms of total adverse events, IDA showed no significant difference when compared to the following drugs “DEC then ALB”, IA, “DA with Mox” and “ALB with Mox”. Also, “ALB with Dox” versus “DEC and Dox”, IVM, DEC, DA, IDA, “DEC then ALB”, IA, “DA with Mox” and “ALB with Mox” alike all of them are insignificant. In the same manner, when we compared DA versus IDA, “DEC then ALB”, IA, “DA with Mox” and “ALB with Mox” we found the following RRs respectively: 0.95 [0.80; 1.14], 0.92 [0.53; 1.60], 0.93 [0.57; 1.50], 0.84 [0.46; 1.53] and 0.77 [0.43; 1.38]. Similarly, our pooled NWM analysis illustrated no significant difference among any of these treatment regimens ALB, Dox, “ALB and Dox”, “DEC and Dox”, IVM, DEC, DA, IDA, “DEC then ALB”, IA, “DA with Mox”, “ALB and Mox” and Placebo. (Fig. 6).
When comparing serious adverse events, our NWM showed no significant difference favoring any of these drugs, IDA, DEC, and DA. With pooled RRs, when IDA compared to DEC and DA, it is as follows: 0.56 and 0.53, respectively. Our pooled studies for serious adverse events exhibit homogeneity with a chi2p value = 0.15 (Fig. 7).
Discussion
Our study assessed microfilaremia reduction over six, twelve, and twenty-four months following various drug regimens. At six months, multiple doses of the DA regimen demonstrated superior efficacy in reducing microfilaremia compared to Dox, IA, DA, DEC & Dox, DEC, IVM, ALB, and placebo. IA also showed significant efficacy against IVM, ALB, and placebo, while DEC combined with ALB showcased substantial superiority over ALB alone or placebo. By the twelfth month, DEC, followed by ALB, ranked superior to several regimens, including IDA, DA, and various combinations involving ALB and Dox. Still, multiple doses of DA remained notably better than several drug combinations. At the twenty-four-month mark, no significant differences were found among the assessed drugs in reducing microfilaremia. In terms of adult worm infection markers, there were no significant differences between most studied interventions. Regarding safety and adverse events, the comparisons revealed no significant differences between several drug combinations, including IDA against DEC, then ALB, IA, and various combinations involving DA and ALB or Mox. Similarly, no significant differences were found between DA, IDA, DEC, and other combinations. The analysis of serious adverse events did not favor any specific drug over others, with no significant differences observed between IDA, DEC, and DA.
The primary approach employed by the GPELF involves implementing community-wide MDA across populations deemed at risk. The objective is to halt the transmission of the disease and mitigate morbidity resulting from infection. Prophylactic medication is deemed essential in regions, such as provinces, districts, or smaller units within a country, where the prevalence of infection within the total population reaches 1% or higher. Preventive chemotherapy aims to disrupt disease transmission by consistently diminishing community microfilariae levels to a critical threshold or entirely eradicating the microfilariae [55, 56]. GPELF advocates for annual administration of a single-dose regimen comprising two drugs (albendazole combined with either DEC or ivermectin [56]. This regimen is recommended for a minimum duration of five years, aligning with the reproductive lifespan of the adult worm. The goal is to attain a coverage rate of at least 65% among the at-risk population to prevent disease transmission effectively [17]. The WHO has advocated for the utilization of an annual triple-drug therapy involving ivermectin, DEC, and albendazole (known as IDA) in specific settings instead of the previously recommended two-drug therapy consisting of albendazole and DEC [6]. In the absence of treatment, it is believed that the overall prevalence rates of microfilariae remain relatively constant over time within endemic communities due to the cycle of reinfection and the continuous production of microfilariae by new adult worms [57].
DEC has been a longstanding treatment for filariasis for over 50 years. Initially, the recommended regimen for DEC was 6 mg/kg daily for 12 days [56]. Subsequently, clinical and community trials determined that single doses administered at varying intervals—weekly, monthly, semi-annually, and annually—proved to be equally effective [16, 58]. Substantial evidence from ultrasound and clinical observations indicates that DEC can eliminate certain adult worms following single doses [56, 59].
Ivermectin is a treatment for onchocerciasis caused by the filarial worm Onchocerca volvulus. It has also demonstrated effectiveness in community control programs targeting LF [60, 61]. In regions where both onchocerciasis and LF coexist, ivermectin is preferred over DEC due to the risk of ocular damage associated with DEC administration in individuals with onchocerciasis. However, ivermectin is not recognized for its macrofilaricidal activity. Ultrasound studies have indicated that even at high doses over six months, ivermectin does not eliminate adult worms [17].
In regions of Central and West Africa where both LF and Loa loa, the eye worm causing loiasis, coexist, treatment with ivermectin or DEC can lead to serious adverse events when Loa loa microfilariae densities are high, surpassing 30,000 mf/mL [62, 63]. In these areas, the recommendation is to administer albendazole alone twice a year, combined with vector control, if ivermectin has not been previously distributed for either onchocerciasis or LF [6, 64]. However, in cases where individuals with onchocerciasis have high Loa loa densities, ivermectin treatment can also lead to SAEs. Despite this risk, treatment with ivermectin is still recommended for meso- and high-endemic areas of onchocerciasis, employing one of three strategies to manage complications should they arise [65].
Albendazole, widely used since the late 1980s against intestinal parasites, shows promise in LF control [66]. Addiss et al. suggested high doses might sterilize or eliminate Wuchereria bancrofti adult worms, yet uncertainty lingers over its impact when combined with DEC or ivermectin [67]. Its role in MDA programs extends beyond filariasis control, aiding in managing other helminth infections [68]. However, it’s cautioned that albendazole alone isn’t recommended for filariasis treatment [69].
Macfarlane et al. metanalysis included 13 trials involving 8,713 participants [56]. Results for microfilariae prevalence showed no significant difference when albendazole was used alone or combined with another drug. Ultrasound detection of adult worm prevalence at 12 months indicated minimal difference when albendazole was combined with another drug. Adverse events reported by participants did not significantly differ with albendazole use. Overall, evidence suggested limited or negligible effects of albendazole on various filariasis indicators, with uncertainties in some outcomes due to low-certainty evidence [56].
A previous trial involving a seven-day coadministration of DEC and Albendazole was inconclusive regarding the swift clearance of microfilariae within a 90-day timeframe [19]. Turner et al. noted that a three-week course of doxycycline treatment, combined with standard anti-filarial therapy, proves more efficacious in eliciting prolonged amicrofilaremia than standard treatment administered alone [24]. Nevertheless, De Britto et al. reported a significant decrease in microfilaria count 30 days after treatment, regardless of whether a single dose or combination therapy was administered [10]. Field trials have demonstrated that a four-week course of doxycycline completely eradicates adult worm nests from scrotal lymphatics in 100% of infected individuals during an 18-month follow-up period [70]. Additionally, a three-week doxycycline therapy combined with a single dose of DEC at four months post-treatment achieved complete clearance of microfilariae, adult worm nests, and a reversal of lymphatic pathology at 12 months post-treatment [71].
While the Multiple DA regimen demonstrated superior efficacy in reducing microfilaremia, it is important to consider the logistical implications of administering multiple doses of diethylcarbamazine. This approach requires careful planning and coordination to ensure adherence and effective implementation, which may pose challenges in large-scale mass drug administration (MDA) programs. In contrast, the current approach of administering the drug for LF elimination at the same time is more straightforward and may be easier to implement in endemic regions.
Supali et al. study revealed that the IDA regimen exhibited a faster clearance of microfilariae from the bloodstream than the DA regimen. Specifically, 25 out of 28 and 8 out of 27 subjects achieved complete microfilariae clearance within 24 h after treatment with IDA and DA, respectively [23]. These superior results in microfilariae clearance observed with IDA align closely with findings from similar clinical trials conducted in Papua New Guinea to treat W. bancrofti infection [2, 22]. Contrarily, a separate clinical trial for W. bancrofti infection in Cote d’Ivoire reported an 89% clearance of microfilariae six months after IDA treatment, reducing to 71% clearance after one year [4]. Consequently, the outcomes of B. timori infection in Indonesia closely resemble the results observed in the W. bancrofti trials in Papua New Guinea rather than those from trials conducted in Cote d’Ivoir.
Adverse effects stemming from antifilarial drugs can be significant, although fatalities are rare, often hindering individuals from initiating or completing treatment. The most severe adverse reactions seem to arise from a host immunologic response triggered by the rapid elimination of microfilariae, associated with the release of inflammatory Wolbachia lipoproteins [72]. Such adverse effects encompass fever, headache, malaise, muscle pain, and the presence of blood in urine. Additionally, localized effects may manifest as localized pain, tender nodules, lymphadenitis, and lymphangitis [56]. The safety profile of IDA has been established through small randomized trials [22, 73] and, more recently, in a comprehensive multi-site community safety trial, including Fiji [5].
Previous investigation revealed a substantial positive correlation between markers indicating filarial infection and the frequency of adverse events (AEs) [74]. This association was particularly pronounced among participants exhibiting microfilaremia identified by the 60 µl smear. Notably, a similar pattern was observed in participants who tested positive for circulating filarial antigen without detectable microfilaremia. Given that most adverse events are linked to the death of microfilariae [74], it is plausible that these participants might have harbored low-density microfilaremia undetectable by smear or had microfilariae not actively circulating at the time of testing. The uncertainty regarding the presence of microfilariae could have been mitigated if we had utilized the more sensitive membrane filtration method with 1 ml venous blood. However, this approach was not feasible for a study involving a large number of participants [75].
Our study represents the introductory NWM encompassing a comprehensive array of interventions and regimens for diverse filariasis treatments. Our systematic review incorporated a total of 45 studies, encompassing 61,369 patients evaluated at three distinct time points. The extensive inclusion of these studies renders our evidence robust, potentially serving as a cornerstone for guiding future clinical practices and formulating guidelines concerning filariasis treatment.
However, our study was not free of limitations. First, most of our studies had unclear or even high risk of bias. Second, although we included many comparison arms, many arms were less representative than others. Third, the effectiveness of a single dose of the triple-drug regimen in clearing microfilariae a year after treatment has shown varying results across different regions. In Papua New Guinea and Haiti, it achieved clearance rates between 94% and 97%. However, the clearance rate in India was 84%, 78% in Côte d’Ivoire, and 63% in Fiji [4, 18, 20,21,22]. These differences could stem from factors such as the varying susceptibility of adult worms to the drugs, differences in how the drugs are absorbed and metabolized in individuals, as well as variables like compliance with taking the drugs as prescribed and the possibility of reinfection. These factors collectively contribute to the observed variability in the measured effectiveness of the treatment. Fourth, in Papua New Guinea, LF is transmitted by anopheline mosquitoes, considered less efficient vectors than culicine or Aedes mosquitoes. Due to this difference in vector efficiency, areas where transmission is through Culex or Aedes mosquitoes might require more rounds of mass drug administration with the triple-drug regimen to eliminate LF [76]. This need for additional treatment rounds is based on the understanding that more competent vectors can sustain transmission more effectively, potentially requiring increased intervention efforts to interrupt the disease’s spread and achieve elimination in those regions. The observed variations mentioned earlier warrant further investigations to stratify these variables and assess their potential impact on treatment regimens. Additional studies could provide insights into how these factors influence the effectiveness and outcomes of different treatment protocols.
Our study contributes to the World Health Organization’s global LF control efforts by providing evidence on the most effective drug regimens for reducing microfilaremia. This is directly aligned with WHO’s goal of eliminating LF as a public health problem by ensuring safe and effective treatment regimens for mass drug administration programs.
Conclusion
Multiple doses of DA regimen exhibited heightened efficacy in diminishing microfilaremia in contrast to combinations involving IA, diethylcarbamazine, ivermectin, and albendazole at six and twelve months. Nevertheless, as the study progressed to the twenty-four-month mark, no significant differences were observed. Safety evaluations across interventions generally revealed comparable profiles, with no specific drug displaying superiority in inducing adverse events. However, the earlier discussed limitations underscore the necessity for further investigations to stratify these variables and evaluate their potential impact on treatment protocols. Subsequent studies could provide valuable insights into how these factors influence the effectiveness and outcomes of distinct treatment regimens.
Data availability
All data generated or analyzed during this study are included in this published article.
References
World Health Organization. Global programme to eliminate lymphatic filariasis: progress report 2020.
Thomsen EK, Sanuku N, Baea M, Satofan S, Maki E, Lombore B, et al. Efficacy, safety, and pharmacokinetics of coadministered diethylcarbamazine, Albendazole, and Ivermectin for treatment of Bancroftian filariasis. Clin Infect Dis. 2016;62:334–41. https://doiorg.publicaciones.saludcastillayleon.es/10.1093/cid/civ882.
Tavul L, Laman M, Howard C, Kotty B, Samuel A, Bjerum C, et al. Safety and efficacy of mass drug administration with a single-dose triple-drug regimen of albendazole + diethylcarbamazine + ivermectin for lymphatic filariasis in Papua new Guinea: an open-label, cluster-randomised trial. PLoS Negl Trop Dis. 2022;16:e0010096. https://doiorg.publicaciones.saludcastillayleon.es/10.1371/journal.pntd.0010096.
Bjerum CM, Ouattara AF, Aboulaye M, Kouadio O, Marius VK, Andersen BJ, et 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:e68–75. https://doiorg.publicaciones.saludcastillayleon.es/10.1093/cid/ciz1050.
Weil GJ, Bogus J, Christian M, Dubray C, Djuardi Y, Fischer PU, et al. The safety of double- and triple-drug community mass drug administration for lymphatic filariasis: A multicenter, open-label, cluster-randomized study. PLoS Med. 2019;16:e1002839. https://doiorg.publicaciones.saludcastillayleon.es/10.1371/journal.pmed.1002839.
WHO, Guideline. Alternative mass drug administration regimens to eliminate lymphatic filariasis. Geneva: World Health Organization; 2017.
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. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/S0140-6736(17)32844-1.
Taylor MJ, Makunde WH, McGarry HF, Turner JD, Mand S, Hoerauf A. Macrofilaricidal activity after Doxycycline treatment of Wuchereria Bancrofti: a double-blind, randomised placebo-controlled trial. Lancet. 2005;365:2116–21. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/S0140-6736(05)66591-9.
Specht S, Mand S, Marfo-Debrekyei Y, Debrah AY, Konadu P, Adjei O, et al. Efficacy of 2- and 4-week rifampicin treatment on the Wolbachia of onchocerca volvulus. Parasitol Res. 2008;103:1303–9. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s00436-008-1133-y.
Pani S, Subramanyam Reddy G, Das L, Vanamail P, Hoti S, Ramesh J, et al. Tolerability and efficacy of single dose Albendazole, diethylcarbamazine citrate (DEC) or co-administration of albendazole with DEC in the clearance of Wuchereria bancrofti in asymptomatic microfilaraemic volunteers in Pondicherry, South India: a hospital-based study. Filar J. 2002;1:1. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/1475-2883-1-1.
Bockarie MJ, Tavul L, Ibam I, Kastens W, Hazlett F, Tisch DJ, et al. Efficacy of single-dose diethylcarbamazine compared with diethylcarbamazine combined with albendazole against Wuchereria bancrofti infection in Papua new Guinea. Am J Trop Med Hyg. 2007;76:62–6.
Page MJ, McKenzie JE, Bossuyt PM, Boutron I, Hoffmann TC, Mulrow CD, et al. The PRISMA 2020 statement: an updated guideline for reporting systematic reviews. BMJ. 2021;n71. https://doiorg.publicaciones.saludcastillayleon.es/10.1136/bmj.n71.
Higgins JPT, Thomas J, Chandler J, Cumpston M, Li T, Page MJ, Welch VA, editors. Cochrane handbook for systematic reviews of interventions. 2nd ed. Chichester (UK): Wiley; 2019.
Higgins JPT, Altman DG, Gotzsche PC, Juni P, Moher D, Oxman AD, et al. The Cochrane collaboration’s tool for assessing risk of bias in randomised trials. BMJ. 2011;343:d5928–5928. https://doiorg.publicaciones.saludcastillayleon.es/10.1136/bmj.d5928.
Butts CT. [internet]etwork. Classes for relational data [Internet]. The Statnet Project; 2020.
Andrade LD, Medeiros Z, Pires ML, Pimentel A, Rocha A, Figueredo-Silva J, et al. Comparative efficacy of three different diethylcarbamazine regimens in lymphatic filariasis. Trans R Soc Trop Med Hyg. 1995;89:319–21. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/0035-9203(95)90561-8.
Dreyer G, Addiss D, Noroes J, Amaral F, Rocha A, Coutinho A. Ultrasonographic assessment of the adulticidal efficacy of repeat high-dose Ivermectin in Bancroftian filariasis. Trop Med Int Health. 1996;1:427–32. https://doiorg.publicaciones.saludcastillayleon.es/10.1046/j.1365-3156.1996.d01-79.x.
Dubray CL, Sircar AD, Beau De Rochars VM, Bogus J, Direny AN, Ernest JR, et 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:e0008298. https://doiorg.publicaciones.saludcastillayleon.es/10.1371/journal.pntd.0008298.
El Setouhy M, Ramzy RMR, Ahmed ES, Kandil AM, Hussain O, Farid HA, et al. A randomized clinical trial comparing single- and multi-dose combination therapy with diethylcarbamazine and albendazole for treatment of Bancroftian filariasis. Am J Trop Med Hyg. 2004;70:191–6.
Hardy M, Samuela J, Kama M, Tuicakau M, Romani L, Whitfeld MJ, et 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:e0008106. https://doiorg.publicaciones.saludcastillayleon.es/10.1371/journal.pntd.0008106.
Jambulingam P, Kuttiatt VS, Krishnamoorthy K, Subramanian S, Srividya A, Raju HKK, et al. An open label, block randomized, community study of the safety and efficacy of co-administered Ivermectin, diethylcarbamazine plus albendazole vs. diethylcarbamazine plus albendazole for lymphatic filariasis in India. PLoS Negl Trop Dis. 2021;15:e0009069. https://doiorg.publicaciones.saludcastillayleon.es/10.1371/journal.pntd.0009069.
King CL, Suamani J, Sanuku N, Cheng Y-C, Satofan S, Mancuso B, et al. A trial of a Triple-Drug treatment for lymphatic filariasis. N Engl J Med. 2018;379:1801–10. https://doiorg.publicaciones.saludcastillayleon.es/10.1056/NEJMoa1706854.
Supali T, Djuardi Y, Christian M, Iskandar E, Alfian R, Maylasari R, et al. An open label, randomized clinical trial to compare the tolerability and efficacy of Ivermectin plus diethylcarbamazine and albendazole vs. diethylcarbamazine plus albendazole for treatment of Brugian filariasis in Indonesia. PLoS Negl Trop Dis. 2021;15:e0009294. https://doiorg.publicaciones.saludcastillayleon.es/10.1371/journal.pntd.0009294.
Turner JD, Mand S, Debrah AY, Muehlfeld J, Pfarr K, McGarry HF, et al. A randomized, Double-Blind clinical trial of a 3-Week course of Doxycycline plus albendazole and Ivermectin for the treatment of Wuchereria bancrofti infection. Clin Infect Dis. 2006;42:1081–9. https://doiorg.publicaciones.saludcastillayleon.es/10.1086/501351.
Addiss DG, Beach MJ, Streit TG, Lutwick S, LeConte FH, Lafontant JG, et al. Randomised placebo-controlled comparison of Ivermectin and albendazole alone and in combination for Wuchereria bancrofti microfilaraemia in Haitian children. Lancet. 1997;350:480–4. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/S0140-6736(97)02231-9.
Beach MJ, Addiss DG, Roberts JM, Lammie PJ, Streit TG, Prospere R. Assessment of combined Ivermectin and albendazole for treatment of intestinal helminth and Wuchereria bancrofti infections in Haitian schoolchildren. Am J Trop Med Hyg. 1999;60:479–86. https://doiorg.publicaciones.saludcastillayleon.es/10.4269/ajtmh.1999.60.479.
Bernhard P, Magnussen P, Lemnge MM. A randomized, double-blind, placebo-controlled study with diethylcarbamazine for the treatment of hydrocoele in an area of Tanzania endemic for lymphatic filariasis. Trans R Soc Trop Med Hyg. 2001;95:534–6. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/S0035-9203(01)90031-8.
Bjerum CM, Koudou BG, Ouattara AF, Lew D, Goss CW, Gabo PT, et al. Safety and tolerability of moxidectin and Ivermectin combination treatments for lymphatic filariasis in Côte D’Ivoire: A randomized controlled superiority study. PLoS Negl Trop Dis. 2023;17:e0011633. https://doiorg.publicaciones.saludcastillayleon.es/10.1371/journal.pntd.0011633.
Britto D. Enhanced efficacy of sequential administration of albendazole for the clearance of Wuchereria bancrofti infection: Double blind RCT n.d.
Dahoma. A randomized community trial on safety and eJicacy of co-administration of albendazole and ivermectin on lymphatic filariasis and its secondary eJects on geohelminths in Zanzibar. 2020.
Dembele B, Coulibaly YI, Dolo H, Konate S, Coulibaly SY, Sanogo D, et al. Use of High-Dose, Twice‐Yearly albendazole and Ivermectin to suppress Wuchereria bancrofti microfilarial levels. CLIN INFECT DIS. 2010;51:1229–35. https://doiorg.publicaciones.saludcastillayleon.es/10.1086/657063.
Dreyer G, Addiss D, Abiel S. Direct assessment in vivo of the efficacy of combined single-dose Ivermectin and diethylcarbamazine against adult Wuchereria bancroti 1998.
Dreyer G, Addiss D, Williamson J, Norões J. Efficacy of co-administered diethylcarbamazine and albendazole against adult Wuchereria bancrofti. Trans R Soc Trop Med Hyg. 2006;100:1118–25. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.trstmh.2006.04.006.
Dunyo SK, Nkrumah FK, Simonsen PE. A randomized double-blind placebo-controlled field trial of Ivermectin and albendazole alone and in combination for the treatment of lymphatic filariasis in Ghana. Trans R Soc Trop Med Hyg. 2000;94:205–11. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/S0035-9203(00)90278-5.
Gayen P, Nayak A, Saini P, Mukherjee N, Maitra S, Sarkar P, et al. A double-blind controlled field trial of Doxycycline and albendazole in combination for the treatment of Bancroftian filariasis in India. Acta Trop. 2013;125:150–6. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.actatropica.2012.10.011.
Glaziou P, Moulia-Pelat N. Double-blind controlled trial of a single dose of the combination ivermectin 400 pg/kg plus diethylcarbamazine 6 mg/kg for the treatment of bancroftian filariasis: results at six months 1994.
Helmy H, El Setouhy M, Kandil AM, Farid HA, Ahmed ES, Weil GJ, A RANDOMIZED CLINICAL TRIAL COMPARING SINGLE- AND MULTI-DOSE COMBINATION THERAPY WITH DIETHYLCARBAMAZINE AND ALBENDAZOLE FOR TREATMENT OF BANCROFTIAN FILARIASIS, et al. Am J Trop Med Hyg. 2004;70:191–6. https://doiorg.publicaciones.saludcastillayleon.es/10.4269/ajtmh.2004.70.191.
Ismail MM, Jayakody RL, Weil GJ, Fernando D, De Silva MSG, De Silva GAC, et al. Long-term efficacy of single-dose combinations of Albendazole, Ivermectin and diethylcarbamazine for the treatment of Bancroftian filariasis. Trans R Soc Trop Med Hyg. 2001;95:332–5. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/S0035-9203(01)90257-3.
Kamgno J, Nguipdop-Djomo P, Gounoue R, Téjiokem M, Kuesel AC. Effect of two or six doses 800 mg of albendazole every two months on Loa loa microfilaraemia: A double blind, randomized, Placebo-Controlled trial. PLoS Negl Trop Dis. 2016;10:e0004492. https://doiorg.publicaciones.saludcastillayleon.es/10.1371/journal.pntd.0004492.
Kar SK, Dwibedi B, Kerketa AS, Maharana A, Panda SS, Mohanty PC, et al. A randomized controlled trial of increased dose and frequency of albendazole with standard dose DEC for treatment of Wuchereria bancrofti microfilaremics in Odisha, India. PLoS Negl Trop Dis. 2015;9:e0003583. https://doiorg.publicaciones.saludcastillayleon.es/10.1371/journal.pntd.0003583.
Klion AD, Massougbodji A, Horton J, Ekoué S, Lanmasso T, Ahouissou N-L, et al. Albendazole in human loiasis: results of a Double-Blind, Placebo-Controlled trial. J Infect Dis. 1993;168:202–6. https://doiorg.publicaciones.saludcastillayleon.es/10.1093/infdis/168.1.202.
Kshirsagar NA, Gogtay NJ, Garg BS, Deshmukh PR, Rajgor DD, Kadam VS, et al. Efficacy and tolerability of treatment with single doses of diethylcarbamazine (DEC) and DEC plus albendazole (ABZ) for three consecutive years in lymphatic filariasis: a field study in India. Parasitol Res. 2017;116:2683–94. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s00436-017-5577-9.
Lafontant J, Furness BW, Desire D, Beach MJ, Haser JK, Lammie PJ, TOLERANCE AND EFFICACY OF COMBINED DIETHYLCARBAMAZINE AND ALBENDAZOLE FOR TREATMENT OF WUCHERERIA BANCROFTI AND INTESTINAL HELMINTH INFECTIONS IN HAITIAN CHILDREN, et al. Am J Trop Med Hyg. 2005;73:115–21. https://doiorg.publicaciones.saludcastillayleon.es/10.4269/ajtmh.2005.73.115.
Laman M, Tavul L, Karl S, Kotty B, Kerry Z, Kumai S, et al. Mass drug administration of Ivermectin, diethylcarbamazine, plus albendazole compared with diethylcarbamazine plus albendazole for reduction of lymphatic filariasis endemicity in Papua new Guinea: a cluster-randomised trial. Lancet Infect Dis. 2022;22:1200–9. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/S1473-3099(22)00026-3.
Ottesen EA, Vijayasree M, Kumaraswami V. A Controlled trial of Ivermectin and Diethylcarbamaezine in lymphatic Filariasis. 1990.
Ouattara AF, Bjerum CM, Aboulaye M, Kouadio O, Marius VK, Andersen B, et al. Semiannual treatment of albendazole alone is efficacious for treatment of lymphatic filariasis: A randomized Open-label trial in Cote D’Ivoire. Clin Infect Dis. 2022;74:2200–8. https://doiorg.publicaciones.saludcastillayleon.es/10.1093/cid/ciab194.
Rizzo JÂ, Belo C, Lins R, Dreyer G. Children and adolescents infected with Wuchereria bancrofti in greater Recife, Brazil: a randomized, year-long clinical trial of single treatments with diethylcarbamazine or diethylcarbamazine–albendazole. Annals Trop Med Parasitol. 2007;101:423–33. https://doiorg.publicaciones.saludcastillayleon.es/10.1179/136485907X176517.
Shenoy RK, Kumaraswami V, Rajan K, Thankom S, Jalajakumari. A comparative study of the efficacy and tolerability of single and split doses of Ivermectin and diethylcarbamazine in periodic Brugian filariasis. Annals Trop Med Parasitol. 1993;87:459–67. https://doiorg.publicaciones.saludcastillayleon.es/10.1080/00034983.1993.11812796.
Simonsen PE, Magesa SM, Dunyo SK, Malecela-Lazaro MN, Michael E. The effect of single dose Ivermectin alone or in combination with albendazole on Wuchereria bancrofti infection in primary school children in Tanzania. Trans R Soc Trop Med Hyg. 2004;98:462–72. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.trstmh.2003.12.005.
Supali T, Djuardi Y, Pfarr KM, Wibowo H, Taylor MJ, Hoerauf A, et al. Doxycycline treatment of Brugia Malayi–Infected persons reduces microfilaremia and adverse reactions after diethylcarbamazine and albendazole treatment. CLIN INFECT DIS. 2008;46:1385–93. https://doiorg.publicaciones.saludcastillayleon.es/10.1086/586753.
Tafatatha TT, Ngwira BM, Taegtmeyer M, Phiri AJ, Wilson TP, Banda LG, et al. Randomised controlled clinical trial of increased dose and frequency of albendazole and Ivermectin on Wuchereria bancrofti microfilarial clearance in Northern Malawi. Trans R Soc Trop Med Hyg. 2015;109:393–9. https://doiorg.publicaciones.saludcastillayleon.es/10.1093/trstmh/trv027.
Wamae CN, Njenga SM, Ngugi BM, Mbui J, Njaanake HK. Evaluation of effectiveness of Diethylcarbamazine/albendazole combination in reduction of Wuchereria bancrofti infection using multiple infection parameters. Acta Trop. 2011;120:S33–8. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.actatropica.2010.09.009.
Yongyuth P, Koyadun S, BNr NJ, Sampuch A, Bhumiratana A. Efficacy of a Single-Dose Treatment with 300 mg Diethylcarbamazine and a Combination of 400 mg Albendazole in Reduction of Wuchereria bancrofti Antigenemia and Concomitant Geohelminths in Myanmar Migrants in Southern Thailand 2006;89.
Zoleko-Manego R, Kreuzmair R, Veletzky L, Ndzebe-Ndoumba W, Ekoka Mbassi D, Okwu DG, et al. Efficacy, safety, and tolerability of albendazole and Ivermectin based regimens for the treatment of microfilaraemic loiasis in adult patients in Gabon: A randomized controlled assessor blinded clinical trial. PLoS Negl Trop Dis. 2023;17:e0011584. https://doiorg.publicaciones.saludcastillayleon.es/10.1371/journal.pntd.0011584.
Ottesen EA. Lymphatic Filariasis: Treatment, Control and Elimination. Advances in Parasitology, vol. 61, Elsevier; 2006, pp. 395–441. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/S0065-308X(05)61010-X
Macfarlane CL, Budhathoki SS, Johnson S, Richardson M, Garner P. Albendazole alone or in combination with microfilaricidal drugs for lymphatic filariasis. Cochrane Database Syst Reviews 2019;2019. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/14651858.CD003753.pub4
Meyrowitsch DW, Simonsen PE, Makunde WH. A 16-year follow-up study on Bancroftian filariasis in three communities of north-eastern Tanzania. Annals Trop Med Parasitol. 1995;89:665–75. https://doiorg.publicaciones.saludcastillayleon.es/10.1080/00034983.1995.11813000.
Simonsen PE, Makunde WH, Meyrowitsch DW, Magnussen P. Selective diethylcarbamazine chemotherapy for control of Bancroftian filariasis in two communities of Tanzania: compared efficacy of a standard dose treatment and two Semi-Annual single dose treatments. Am J Trop Med Hyg. 1995;53:267–72. https://doiorg.publicaciones.saludcastillayleon.es/10.4269/ajtmh.1995.53.267.
Norões J, Dreyer G, Santos A, Mendes VG, Medeiros Z, Addiss D. Assessment of the efficacy of diethylcarbamazine on adult Wuchereria bancrofti in vivo. Trans R Soc Trop Med Hyg. 1997;91:78–81. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/S0035-9203(97)90405-3.
Andrade LD, Medeiros Z, Moura I, Ottesen EA, Rizzo JA, Rocha A, et al. Ivermectin treatment of Bancroftian filariasis in Recife, Brazil. Am J Trop Med Hyg. 1994;50:339–48. https://doiorg.publicaciones.saludcastillayleon.es/10.4269/ajtmh.1994.50.339.
Cao W, Van Der Ploeg CPB, Plaisier AP, Van Der Sluijs IJS, Habbema JDF. Ivermectin for the chemotherapy of Bancroftian filariasis: a meta-analysis of the effect of single treatment. Trop Med Int Health. 1997;2:393–403. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/j.1365-3156.1997.tb00157.x.
Boussinesq And M, Gardon J. Prevalences of Loa loa microfilaraemia throughout the area endemic for the infection. Ann Trop Med Parasitol. 1997;91:573–90. https://doiorg.publicaciones.saludcastillayleon.es/10.1080/00034989760671.
Gardon J, Gardon-Wendel N, Demanga-Ngangue, Kamgno J, Chippaux J-P, Boussinesq M. Serious reactions after mass treatment of onchocerciasis with Ivermectin in an area endemic for Loa loa infection. Lancet. 1997;350:18–22. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/S0140-6736(96)11094-1.
WHO. Provisional Strategy For Interrupting Lymphatic Filariasis Transmission In Loiasis-Endemic Countries 2012.
Mectizan® Expert Committee. The Technical Consultative Committee. Recommendations for the Treatment of Onchocerciasis with Mectizan® in Areas Co-endemic for Onchocerciasis and Loiasis. 2004.
Ottesen EA, Ismail MM, Horton J. The role of albendazole in programmes to eliminate lymphatic filariasis. Parasitol Today. 1999;15:382–6. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/s0169-4758(99)01486-6.
Addiss D, Gamble CL, Garner P, Gelband H, Ejere HO, Critchley JA, et al. Albendazole for lymphatic filariasis. Cochrane Database Syst Reviews. 2005. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/14651858.CD003753.pub3.
Shenoy RK, Bockarie MJ. Lymphatic filariasis in children: clinical features, infection burdens and future prospects for elimination. Parasitology. 2011;138:1559–68. https://doiorg.publicaciones.saludcastillayleon.es/10.1017/S003118201100117X.
Horton J. The development of albendazole for lymphatic filariasis. Annals Trop Med Parasitol. 2009;103:33–40. https://doiorg.publicaciones.saludcastillayleon.es/10.1179/000349809X12502035776595.
Debrah AY, Mand S, Marfo-Debrekyei Y, Batsa L, Albers A, Specht S, et al. Macrofilaricidal activity in Wuchereria bancrofti after 2 weeks treatment with a combination of rifampicin plus Doxycycline. J Parasitol Res. 2011;2011:1–9. https://doiorg.publicaciones.saludcastillayleon.es/10.1155/2011/201617.
Mand S, Pfarr K, Sahoo PK, Satapathy AK, Specht S, Klarmann U, et al. Macrofilaricidal activity and amelioration of lymphatic pathology in Bancroftian filariasis after 3 weeks of Doxycycline followed by single-dose diethylcarbamazine. Am J Trop Med Hyg. 2009;81:702–11. https://doiorg.publicaciones.saludcastillayleon.es/10.4269/ajtmh.2009.09-0155.
Turner JD, Langley RS, Johnston KL, Gentil K, Ford L, Wu B, et al. Wolbachia lipoprotein stimulates innate and adaptive immunity through Toll-like receptors 2 and 6 to induce disease manifestations of filariasis. J Biol Chem. 2009;284:22364–78. https://doiorg.publicaciones.saludcastillayleon.es/10.1074/jbc.M901528200.
Edi C, Bjerum CM, Ouattara AF, Chhonker YS, Penali LK, Méité A, et al. Pharmacokinetics, safety, and efficacy of a single co-administered dose of diethylcarbamazine, albendazole and Ivermectin in adults with and without Wuchereria bancrofti infection in Côte D’Ivoire. PLoS Negl Trop Dis. 2019;13:e0007325. https://doiorg.publicaciones.saludcastillayleon.es/10.1371/journal.pntd.0007325.
Budge PJ, Herbert C, Andersen BJ, Weil GJ. Adverse events following single dose treatment of lymphatic filariasis: observations from a review of the literature. PLoS Negl Trop Dis. 2018;12:e0006454. https://doiorg.publicaciones.saludcastillayleon.es/10.1371/journal.pntd.0006454.
World Health Organization. Control of lymphatic filariasis: a manual for health personnel. World Health Organization; 1987.
Bockarie MJ, Pedersen EM, White GB. Role of vector control in the global program to eliminate lymphatic filariasis. Annu Rev Entomol. 2009;54:469–87. https://doiorg.publicaciones.saludcastillayleon.es/10.1146/annurev.ento.54.110807.090626.
Acknowledgements
None.
Funding
This scientific paper is derived from a research grant funded by the Research, Development, and Innovation Authority (RDIA) - Kingdom of Saudi Arabia - with grant number (12982-iau-2023-TAU-R-3-1-HW-).
Author information
Authors and Affiliations
Contributions
The conceptualization was performed by MA and ES. Data curation was performed by MA, ES, SS, MA, HE, and ME. Formal analysis was conducted by MA, SS, MA, and ES. Quality assessment and risk of bias evaluation were performed by SS, MA, ES, and AM. Methodological guidance was provided by HE and HE. Project administration was managed by MA. Resources were acquired by MA. Software management was handled by SS. Validation of analytical methods was carried out by HE, HE, SS, MA, and ES. Data visualization and interpretation were led by MA. Writing of the original draft was done by MA, ES, and MA. Writing– review and editing was contributed by MA, ES, SS, MA, HE, and ME. Funding acquisition was handled by HE. All authors reviewed the manuscript.
Corresponding author
Ethics declarations
Ethics approval and consent to participate
Not applicable.
Consent for publication
Not applicable.
Competing interests
The authors declare no competing interests.
Clinical trial number
Not applicable.
Additional information
Publisher’s note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Electronic supplementary material
Below is the link to the electronic supplementary material.

12879_2025_11105_MOESM1_ESM.jpg
Supplementary Material 1: Supplementary fig.1: PRISMA flow diagram for the selection process of eligible studies in our systematic review and NWA

12879_2025_11105_MOESM2_ESM.png
Supplementary Material 2: Supplementar fig.2: Visually depicted the risk of bias summary according to the Cochrane ROB 1 tool
Rights and permissions
Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. 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-nc-nd/4.0/.
About this article
Cite this article
Albadrani, M.S., Molla, A., Elbadawy, H.M. et al. Antifilarial treatment strategies: a systematic review and network meta-analysis. BMC Infect Dis 25, 712 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12879-025-11105-z
Received:
Accepted:
Published:
DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12879-025-11105-z