Abstract
Drug-based interventions are at the heart of global efforts to reach elimination as a public health problem (trachoma, soil-transmitted helminthiases, schistosomiasis, lymphatic filariasis) or elimination of transmission (onchocerciasis) for 5 of the most prevalent neglected tropical diseases tackled via the World Health Organization preventive chemotherapy strategy. While for some of these diseases there is optimism that currently available drugs will be sufficient to achieve the proposed elimination goals, for others—particularly onchocerciasis—there is a growing consensus that novel therapeutic options will be needed. Since in this area no high return of investment is possible, minimizing wasted money and resources is essential. Here, we use illustrative results to show how mathematical modeling can guide the drug development pathway, yielding resource-saving and efficiency payoffs, from the refinement of target product profiles and intended context of use to the design of clinical trials.
Keywords: drug development, neglected tropical diseases, mathematical modeling, filariases, onchocerciasis
Modeling is used in the commercial pharmaceutical sector to facilitate drug development. The efficiency payoffs of modeling in the resource-scarce neglected tropical disease (NTD) domain can be particularly valuable. Modeling should be routinely integrated into the NTD drug development pathway.
Mass drug administration (MDA), by which at-risk populations are regularly treated without the need for individual diagnosis, is at the core of the preventive chemotherapy strategy recommended by the World Health Organization (WHO) to eliminate 5 of the most globally prevalent neglected tropical diseases (NTDs): trachoma, soil-transmitted helminthiases (STHs), schistosomiasis, lymphatic filariasis, and onchocerciasis [1]. Four of these are earmarked for elimination as a public health problem (EPHP) in the new WHO 2030 NTD roadmap, while elimination of transmission (EOT) is proposed for onchocerciasis [2].
Therapeutic options for trachoma, the 3 major STHs (roundworm, whipworm, and hookworm), and schistosomiasis are reasonably efficacious and often curative, albeit with some exceptions, such as the inadequacy of benzimidazoles alone for treating whipworm [3]. While the therapeutic arsenal is limited for these NTDs—and therefore potentially vulnerable to emerging drug resistance—there is cautious optimism that if MDA is delivered at adequate frequency, coverage, and duration, then the WHO’s 2030 elimination goals can be met [4–6]. Similarly, although treatments for lymphatic filariasis are not generally considered “curative,” they are effective in strongly suppressing the microfilarial progeny (the stages that are transmitted to mosquito vectors) [7, 8] of adult filariae (macrofilariae) and may have partial anti-macrofilarial effects [9]. These treatments are therefore also considered compatible with EPHP [10].
The EOT goal for onchocerciasis places scrutiny on the efficacy of current treatment options for this filarial disease. A single standard dose (150 µg/kg) of ivermectin—the cornerstone of onchocerciasis MDA—is effective in mediating the killing of the microfilarial skin-dwelling transmission stages (to blackfly vectors) of adult Onchocerca volvulus and in inducing a temporary sterilizing (embryostatic) effect [11] (although suboptimal responses have been documented in Ghana [12]). But ivermectin only has partial killing or “curative” efficacy against adult worms and only after repeated MDA rounds [13]. Mathematical transmission dynamics modeling that integrates these pharmacodynamics indicates that ivermectin MDA alone, particularly if delivered annually, may be insufficient to achieve elimination in highly endemic areas where transmission is intense [14, 15]. In West Africa, before ivermectin MDA for onchocerciasis began in earnest in the late 1980s, transmission in 50–75% of endemic communities was deemed high (hyperendemic) or very high (holoendemic) [16]. Transmission remains ongoing in many such foci despite decades of intervention [17], attesting to the challenge of achieving widespread EOT by 2030.
The registration of moxidectin for the treatment of onchocerciasis in 2018 [18] was a significant milestone in enhancing the prospects of achieving EOT. Moxidectin, like ivermectin, is a macrocyclic lactone but with superior pharmacokinetics and pharmacodynamics, suppressing microfilariae to lower levels and for longer duration [18, 19]. This means that less transmission to vectors is possible between consecutive MDA rounds [18, 20]. But moxidectin will only be part of the solution to achieving widespread elimination of onchocerciasis. Financial and operational feasibility to allow uptake of moxidectin into MDA programs needs to be tackled. There are also safety concerns on the use of macrocyclic lactones in people heavily coinfected with Loa loa [21], another filarial parasite, that occurs in forest areas of central Africa [22]. Tetracycline antibiotics exert macrofilaricidal activity by depleting Wolbachia endosymbionts from O. volvulus and, crucially, are considered a safer alternative in people infected with L. loa (which lacks Wolbachia) [23, 24]. These represent important adjuvant or alternative therapies to the macrocyclic lactones and will likely become increasingly used if shorter treatment courses are found to be as efficacious as those currently available (eg, doxycycline, which requires 4–6 weeks of daily treatment) [25, 26].
For other NTDs, there have been similar recent developments in the therapeutic armory. The strong and sustained suppression of microfilaremia by the combination of ivermectin, diethylcarbamazine (DEC), and albendazole (so-called IDA therapy) presents a significant leap forward for the elimination of lymphatic filariasis [27, 28], albeit cautiously due to safety concerns of treating with DEC in Africa where many people are coinfected with onchocerciasis [28]. For whipworm, various combinations of existing antiparasitic drugs have shown improved efficacy over benzimidazoles alone [29, 30]. These examples all attest to drug development and repurposing being an active and important component of a holistic global strategy to enhance preventive chemotherapy for NTDs [14, 17, 31]. Continued drug development also provides insurance against the specter of emerging drug resistance, a particularly ominous prospect for diseases such as schistosomiasis, for which there is sole reliance on a single treatment [32].
Research and development (R&D) costs associated with drug development are high because of high attrition rates, with many promising molecules failing during preclinical development or in subsequent clinical safety and efficacy testing. The level of investment in R&D for new products for NTDs is nowhere near the level of funding required, and that funding, when available, is rarely allocated in a manner likely to ensure that products successfully move through the pipeline to reach end-users. It is therefore essential that stakeholders, funders, industry, academics, and nongovernmental organizations adopt a cooperative approach and share responsibility to reduce risks and overcome obstacles [33]. Joint efforts are being made to cut the cost of R&D for new drugs for NTDs and increase the attractiveness of this sector to funders and investors. For example, supportive programs by the US Food and Drug Administration (priority review voucher program) [34] and the European Medicines Agency (article 58) aim to increase incentives for companies to engage in NTD drug development.
Not-for-profit organizations (eg, Drugs for Neglected Diseases initiative [DNDi], https://dndi.org; and Medicines Development for Global Health, https://www.medicinesdevelopment.com), supported through impact investment and philanthropic, governmental, and supranational grants and donations—have, over the last decade, started to fill the space left by commercial pharmaceutical companies who have little incentive to invest in new medicines without a profitable market [18, 35]. Indeed, the precedent that pharmaceuticals should be donated for NTDs may further disincentivize investment. For not-for-profit organizations—which naturally operate with fewer resources than their commercial counterparts—optimizing target product profiles (TPPs; see, eg, https://dndi.org/diseases/filaria-river-blindness/target-product-profile/) and the design of clinical trials [36] is essential to maximizing the efficiency of the drug development pathway. Modeling of all types has long been used in the commercial pharmaceutical sector to inform trial design [37]. However, it has only recently been adopted in the preclinical [38] and clinical [36] NTD domain, where resource-saving payoffs can result from better informed decision making and quantitative insights on proposed TPPs.
Since drug discovery and development for NTDs is largely driven by the unmet medical needs identified by the global health community and without the possibility of significant financial returns on investment, efficient use of money and other resources to deliver development programs is essential. To ensure that any development project is able to address the complex requirements of patients and healthcare providers with drug and formulation characteristics suitable for storage and use in tropical climates, the TPP provides a carefully defined framework for describing both the ideal and acceptable attributes of a novel medicine. In particular, the TPP must define the necessary characteristics and associated minimum and desired criteria against which a new therapy will be assessed. This may include relative safety, efficacy, effectiveness, and superiority against existing treatments. For NTDs in the era of elimination, specific challenges need to be tackled, beginning with the absence of a healthy drug discovery process. Further, the TPP should consider not only the individual-level therapeutic benefits but also the capacity for a new drug to facilitate elimination at the population level. But challenges can arise when properties conferring individual-level therapeutic benefits do not completely align with those most compatible with enhancing elimination.
We can illustrate this conundrum by considering the (individual-level) efficacy and (population-level) effectiveness of macrofilaricides (drugs that kill adult filariae) for the treatment of onchocerciasis. Figure 1a shows the modeled dynamics (using the transmission model EPIONCHO-IBM) [39] of microfilarial prevalence elicited by 4 drugs—with distinct pharmacodynamic properties—given annually by MDA in an onchocerciasis-endemic community. The strong microfilaricidal properties of ivermectin elicit immediate but transient declines in prevalence, which in the long term can suppress transmission to the point of elimination (although this is not necessarily the case when simulating more highly endemic populations) [39]. By contrast, macrofilaricides with either direct killing activity (MOM/MAMM in Figure 1a) or indirect depletion of endosymbiotic Wolbachia [26] (AWOL in Figure 1a)—but without substantial microfilaricidal action—elicit only slow reductions in microfilaridermia but can, in the long term, enhance the likelihood of elimination. Therefore, from the population perspective, a TPP may stress the importance of developing a macrofilaricide or long-term sterilizing drug with efficacy that increases the likelihood (probability) of elimination compared with ivermectin. But, from an individual perspective, as microfilariae are associated with the majority of clinical manifestations [40, 41], the slow decline in microfilaridermia elicited by a macrofilaricide without significant microfilaricidal activity may be associated with unacceptably slow therapeutic benefit.
Figure 1.
Projected dynamics, using EPIONCHO-IBM [39], of Onchocerca volvulus microfilarial prevalence. Panel a shows dynamics through 15 years of annual MDA with IVM or using a macrofilaricidal therapy in a mesoendemic setting. The light-blue line indicates MDA with IVM. Blue, orange, and red lines indicate, respectively, MDA with a hypothetical macrofilaricidal-only macrofilaricide (MOM) with 90% efficacy (90% of adult worms killed within 3 months following treatment) [36], a hypothetical macrofilaricidal and microfilaricidal macrofilaricide (MAMM; ie, a macrofilaricide with additional microfilaricidal activity) with 90% efficacy, and an anti-Wolbachia (AWOL) therapy (with 90% efficacy and pharmacodynamics based on those of doxycycline and prophylactic effect against incoming worms during the treatment course) [26]. The (model-predicted) probabilities of elimination (95% confidence intervals as error bars) for this scenario are depicted in panel b: 0% (0–7%), 38% (34–42%), 80% (76–83%), and 79% (75–82%) for IVM, MOM (90%), MAMM (90%), and AWOL, respectively. Panel c shows dynamics through 15 years of MDA with IVM followed by 15 years of IVM alone or in combination with a macrofilaricidal therapy in a hyperendemic setting. The light-blue line indicates MDA with IVM. Blue, orange, and red lines indicate, respectively, MDA with IVM for 15 years followed by the addition of either a MOM (with 90% or 60% efficacy, respectively) or an AWOL therapy. The (model-predicted) probabilities of elimination (95% confidence intervals) in this scenario are shown in panel d: 0% (0–7%), 14% (11–17%), 41% (37–45%), and 64% (60–68%) for IVM, IVM + MOM (60%), IVM + MOM (90%), and IVM + AWOL, respectively. Model projections assumed a hypothetical treatment coverage of 80% with 1% systematic nonadherence. Potential differences in eligibility criteria for different drugs (which would affect treatment coverage) are not considered in the interests of showing a simple vis-à-vis comparison of the impact of different pharmacodynamic properties. Abbreviations: IVM, ivermectin; MDA, mass drug administration.
The examples shown in Figure 1a are deliberately simple to enable vis-à-vis comparison of the effectiveness of drugs with contrasting pharmacodynamics. In reality, it is unlikely that a new macrofilaricide would be used as a replacement to ivermectin. It would more likely be used either to complement ivermectin in epidemiological settings where elimination has proved elusive and requires acceleration or in difficult-to-treat areas (such as where L. loa is highly co-endemic) as part of focused MDA or test-and-treat or not-treat strategies. As an example, Figure 1b illustrates the complementary use of a macrofilaricide with ivermectin in settings with a long (15-year) history of annual MDA intervention. Particularly noteworthy here is that the addition of a macrofilaricide—even with a modest 60% efficacy (assuming 60% of adult worms are killed within 3 months of administration)—results in an increased probability of elimination compared with ongoing ivermectin treatment alone. This example encapsulates the challenge of designing TPPs when faced with competing individual- versus population-level properties. We know from clinical trial simulation that demonstrating the benefit of a macrofilaricide with modest efficacy would likely require prohibitively long follow-up times and/or very large sample sizes [36]. Yet, should a macrofilaricide with 60% efficacy be discarded on this basis when it may be useful in accelerating progress towards elimination in mature intervention programs?
With these insights, one may choose to focus from the outset on comparing against ivermectin, combination therapies with pharmacodynamics similar to those of a a macrofilaricide with microfilaricidal action as shown in Figure 1a. This would restrict regulatory-approved usage to a combination therapy, although use of the macrofilaricide as monotherapy or in alternative combinations could continue to be further explored in clinical trials. Clinical studies supporting such combination use would be restricted to settings that align closely with the intended use scenario (eg, mature MDA programs where elimination has proved elusive and requires a new strategy). However, determining patients' past use of ivermectin is highly challenging and interpreting trial outcomes is complicated by the drug’s cumulative pharmacodynamics on O. volvulus, which accrue with long-term use [13]. Irrespective of the chosen path, transmission dynamics modeling can assist with evaluating the likely effectiveness of a new drug or combination in its proposed context of use and with practical decision making to optimize the design of trials to demonstrate efficacy. Indeed, without modeling, the implications of desired pharmacodynamic properties defined in the TPP may not be fully appreciated.
The use of mathematical modeling to support decision making along the NTD drug development pathway is still in its infancy. Pharmacokinetic-pharmacodynamics modeling has been used to translate preclinical data to humans [38] and transmission modeling has been used to make projections on the long-term epidemiological impact (effectiveness) of new drugs (eg, moxidectin for onchocerciasis) [18, 20] or repurposed combinations (eg, IDA for lymphatic filariasis [27] or benzimidazoles plus ivermectin for whipworm [42]) that have already been demonstrated as safe and efficacious in clinical studies. In onchocerciasis, we have partnered with DNDi to use transmission modeling at earlier stages of clinical development to inform the design of trials [36], but this is yet to become more commonplace for other NTD drug development programs.
Naturally, the usefulness of models is defined by the quality of the underlying assumptions and, as in any practical application, it is crucial to communicate uncertainty in a clear and transparent manner [43]. In supporting drug development, it is essential that modeling acknowledges that the precise pharmacodynamics of a new drug (or indeed an existing comparator) will not be completely understood from the outset and exploration of the impact of key assumptions is paramount. Notwithstanding, qualitative insights remain useful and can help guide design decisions aimed at resolving uncertainties. For example, modeling may indicate (within a range) a minimum macrofilaricidal efficacy below which a new drug would not provide sufficient acceleration towards elimination. Armed with this insight, a clinical trial can be designed to collect data at particular time points that may not be most appropriate for final outcome assessment but will provide early information on likely pharmacodynamics properties (eg, early rates of microfilarial depletion) [36].
The illustrative results presented here aim to show the utility of transmission models when used in a prospective capacity to support decision making along the NTD drug development pathway. Specifically, modeling can help inform TPPs by projecting the likely effectiveness of new drugs, with different proposed pharmacodynamics, used in different intervention and epidemiological contexts. Modeling can also help to identify particular pharmacodynamic properties that may be useful for enhancing elimination—but not necessarily of immediate therapeutic benefit to the individual—and guide the appropriate design of phase 2/3 efficacy trials [36]. The integration of transmission modeling with economic cost-benefit analyses is a further important tool that remains relatively underused in the development of treatments for NTDs [44] but is a crucial component for informing donor investment decisions. Overall, we believe that mathematical transmission modeling should be more routinely adopted to inform NTD drug development decisions.
Notes
Author contributions. M. W. drafted the manuscript; J. I. D. H. performed the modeling; all authors commented for intellectual input and edited and approved the final version.
Acknowledgments. The authors are indebted to Rebecca Smith, PhD, for her comments and feedback on the final submitted version of the manuscript.
Financial support. M. W. and M.-G. B. acknowledge financial support from the Drugs for Neglected Diseases initiative (DNDi; https://www.dndi.org). M. W., J. I. D. H., and M.-G. B. also acknowledge funding from the NTD Modelling Consortium by the Bill and Melinda Gates Foundation (grant number OPP 1184344). M.-G. B. further acknowledges joint center funding (grant number MR/R015600/1) by the Medical Research Council (MRC) and the UK Department for International Development (DFID) under the MRC/DFID Concordat agreement, which is also part of the European and Developing Countries Partnership for Clinical Trials (EDCTP2) program supported by the European Union. P. M. acknowledges support by a Doctoral Training Program studentship of the UK MRC.
Potential conflicts of interest. F. M., S. S., and B. P. work at the organization (DNDi) that partly sponsored this study. However, the study sponsor did not have any influence in the study design, analysis, interpretation of results, the writing of the manuscript, or the decision to submit the paper for publication, other than critical, independent, and scientific contribution. No payment was received from any other funding sources or pharmaceutical company. The corresponding author had final responsibility for decision to submit for publication. All other authors report no potential conflicts. All authors have submitted the ICMJE Form for Disclosure of Potential Conflicts of Interest. Conflicts that the editors consider relevant to the content of the manuscript have been disclosed.
References
- 1.World Health Organization. Crossing the billion. Lymphatic filariasis, onchocerciasis, schistosomiasis, soil-transmitted helminthiases and trachoma. Preventive chemotherapy for neglected tropical diseases. Geneva, Switzerland: World Health Organization, 2017. Available at: https://www.who.int/neglected_diseases/resources/9789240696471/en/. Accessed: 13 April 2021. [Google Scholar]
- 2.World Health Organization. Ending the neglect to attain the Sustainable Development Goals. A road map for neglected tropical diseases 2021–2030. Geneva, Switzerland: World Health Organization, 2020. Available at: https://www.who.int/neglected_diseases/Ending-the-neglect-to-attain-the-SDGs--NTD-Roadmap.pdf?ua=1. Accessed 13 April 2021. [Google Scholar]
- 3.Moser W, Schindler C, Keiser J. Efficacy of recommended drugs against soil transmitted helminths: systematic review and network meta-analysis. BMJ 2017; 358:j4307. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.NTD Modelling Consortium Discussion Group on Soil-Transmitted Helminths. Insights from quantitative analysis and mathematical modelling on the proposed WHO 2030 goals for soil-transmitted helminths. Gates Open Res 2019; 3:1632. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.NTD Modelling Consortium Schistosomiasis Group. Insights from quantitative and mathematical modelling on the proposed WHO 2030 goal for schistosomiasis. Gates Open Res 2019; 3:1517. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.NTD Modelling Consortium Discussion Group on Trachoma. Insights from mathematical modelling and quantitative analysis on the proposed 2030 goals for trachoma. Gates Open Res 2019; 3:1721. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Tisch DJ, Michael E, Kazura JW. Mass chemotherapy options to control lymphatic filariasis: a systematic review. Lancet Infect Dis 2005; 5:514–23. [DOI] [PubMed] [Google Scholar]
- 8.King CL, Suamani J, Sanuku N, et al. A trial of a triple-drug treatment for lymphatic filariasis. N Engl J Med 2018; 379:1801–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Pion SDS, Chesnais CB, Weil GJ, Fischer PU, Missamou F, Boussinesq M. Effect of 3 years of biannual mass drug administration with albendazole on lymphatic filariasis and soil-transmitted helminth infections: a community-based study in Republic of the Congo. Lancet Infect Dis 2017; 17:763–9. [DOI] [PubMed] [Google Scholar]
- 10.NTD Modelling Consortium Lymphatic Filariasis Group. The roadmap towards elimination of lymphatic filariasis by 2030: insights from quantitative and mathematical modelling. Gates Open Res 2019; 3:1538. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Basáñez MG, Pion SDS, Boakes E, Filipe JAN, Churcher TS, Boussinesq M. Effect of single-dose ivermectin on Onchocerca volvulus: a systematic review and meta-analysis. Lancet Infect Dis 2008; 8:310–22. [DOI] [PubMed] [Google Scholar]
- 12.Frempong KK, Walker M, Cheke RA, et al. Does increasing treatment frequency address suboptimal responses to ivermectin for the control and elimination of river blindness? Clin Infect Dis 2016; 62:1338–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Walker M, Pion SDS, Fang H, et al. Macrofilaricidal efficacy of repeated doses of ivermectin for the treatment of river blindness. Clin Infect Dis 2017; 65:2026–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Verver S, Walker M, Kim YE, 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; 66:267–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.NTD Modelling Consortium Onchocerciasis Group. The World Health Organization 2030 goals for onchocerciasis: insights and perspectives from mathematical modelling. Gates Open Res 2019; 3:1545. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.O’Hanlon SJ, Slater HC, Cheke RA, et al. Model-based geostatistical mapping of the prevalence of Onchocerca volvulus in West Africa. PLoS Negl Trop Dis 2016; 10:e0004328. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Dadzie Y, Amazigo UV, Boatin BA, Sékétéli A. Is onchocerciasis elimination in Africa feasible by 2025: a perspective based on lessons learnt from the African control programmes. Infect Dis Poverty 2018; 7:63. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Milton P, Hamley JID, Walker M, Basáñez MG. Moxidectin: an oral treatment for human onchocerciasis. Expert Rev Anti Infect Ther 2020; 18:1067–81. [DOI] [PubMed] [Google Scholar]
- 19.Opoku NO, Bakajika DK, Kanza EM, 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Turner HC, Walker M, Attah SK, et al. The potential impact of moxidectin on onchocerciasis elimination in Africa: an economic evaluation based on the Phase II clinical trial data. Parasit Vectors 2015; 8:167. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Boussinesq M. A new powerful drug to combat river blindness. Lancet 2018; 392:1170–2. [DOI] [PubMed] [Google Scholar]
- 22.Zouré HG, Wanji S, Noma M, et al. The geographic distribution of Loa loa in Africa: results of large-scale implementation of the Rapid Assessment Procedure for Loiasis (RAPLOA). PLoS Negl Trop Dis 2011; 5:e1210. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Turner JD, Tendongfor N, Esum M, et al. Macrofilaricidal activity after doxycycline only treatment of Onchocerca volvulus in an area of Loa loa co-endemicity: a randomized controlled trial. PLoS Negl Trop Dis 2010; 4:e660. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Wanji S, Tendongfor N, Nji T, et al. Community-directed delivery of doxycycline for the treatment of onchocerciasis in areas of co-endemicity with loiasis in Cameroon. Parasit Vectors 2009; 2:39. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Taylor MJ, von Geldern TW, Ford L, et al. Preclinical development of an oral anti-Wolbachia macrolide drug for the treatment of lymphatic filariasis and onchocerciasis. Sci Transl Med 2019; 11: eaau2086. [DOI] [PubMed] [Google Scholar]
- 26.Walker M, Specht S, Churcher TS, Hoerauf A, Taylor MJ, Basáñez MG. Therapeutic efficacy and macrofilaricidal activity of doxycycline for the treatment of river blindness. Clin Infect Dis 2015; 60:1199–207. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Irvine MA, Stolk WA, Smith ME, et al. Effectiveness of a triple-drug regimen for global elimination of lymphatic filariasis: a modelling study. Lancet Infect Dis 2017; 17:451–8. [DOI] [PubMed] [Google Scholar]
- 28.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Clarke NE, Doi SAR, Wangdi K, Chen Y, Clements ACA, Nery SV. Efficacy of anthelminthic drugs and drug combinations against soil-transmitted helminths: a systematic review and network meta-analysis. Clin Infect Dis 2019; 68:96–105. [DOI] [PubMed] [Google Scholar]
- 30.Keller L, Palmeirim MS, Ame SM, et al. Efficacy and safety of ascending dosages of moxidectin and moxidectin-albendazole against Trichuris trichiura in adolescents: a randomized controlled trial. Clin Infect Dis 2020; 70:1193–201. [DOI] [PubMed] [Google Scholar]
- 31.Kuesel AC. Research for new drugs for elimination of onchocerciasis in Africa. Int J Parasitol Drugs Drug Resist 2016; 6:272–86. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Crellen T, Walker M, Lamberton PH, et al. Reduced efficacy of praziquantel against Schistosoma mansoni is associated with multiple rounds of mass drug administration. Clin Infect Dis 2016; 63:1151–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Uniting to Combat Neglected Tropical Diseases. Ten achievements of the London Declaration in Neglected Tropical Diseases. Available at: https://unitingtocombatntds.org/news/ten-achievements-of-the-london-declaration-on-neglected-tropical-diseases/. Accessed 13 April 2021.
- 34.US Food and Drug Administration. Tropical Disease Priority Review Voucher Program. Available at: https://www.fda.gov/about-fda/center-drug-evaluation-and-research-cder/tropical-disease-priority-review-voucher-program. Accessed 13 April 2021.
- 35.Pedrique B, Strub-Wourgaft N, Some C, et al. The drug and vaccine landscape for neglected diseases (2000-11): a systematic assessment. Lancet Glob Health 2013; 1:e371–9. [DOI] [PubMed] [Google Scholar]
- 36.Walker M, Hamley JID, Milton P, Monnot F, Pedrique B, Basáñez MG. Designing antifilarial drug trials using clinical trial simulators. Nat Commun 2020; 11:2685. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Kimko H, Pinheiro J. Model-based clinical drug development in the past, present and future: a commentary. Br J Clin Pharmacol 2015; 79:108–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Aljayyoussi G, Tyrer HE, Ford L, et al. Short-course, high-dose rifampicin achieves Wolbachia depletion predictive of curative outcomes in preclinical models of lymphatic filariasis and onchocerciasis. Sci Rep 2017; 7:210. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Hamley JID, Milton P, Walker M, Basáñez MG. Modelling exposure heterogeneity and density dependence in onchocerciasis using a novel individual-based transmission model, EPIONCHO-IBM: implications for elimination and data needs. PLoS Negl Trop Dis 2019; 13:e0007557. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Little MP, Basáñez MG, Breitling LP, Boatin BA, Alley ES. Incidence of blindness during the Onchocerciasis Control Programme in western Africa, 1971–2002. J Infect Dis 2004; 189:1932–41. [DOI] [PubMed] [Google Scholar]
- 41.Murdoch ME, Asuzu MC, Hagan M, et al. Onchocerciasis: the clinical and epidemiological burden of skin disease in Africa. Ann Trop Med Parasitol 2002; 96:283–96. [DOI] [PubMed] [Google Scholar]
- 42.Turner HC, Truscott JE, Bettis AA, Hollingsworth TD, Brooker SJ, Anderson RM. Analysis of the population-level impact of co-administering ivermectin with albendazole or mebendazole for the control and elimination of Trichuris trichiura. Parasite Epidemiol Control 2016; 1:177–87. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Behrend MR, Basáñez MG, Hamley JID, et al. ; NTD Modelling Consortium . Modelling for policy: the five principles of the Neglected Tropical Diseases Modelling Consortium. PLoS Negl Trop Dis 2020; 14:e0008033. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Turner HC, Walker M, French MD, Blake IM, Churcher TS, Basáñez MG. Neglected tools for neglected diseases: mathematical models in economic evaluations. Trends Parasitol 2014; 30:562–70. [DOI] [PubMed] [Google Scholar]