Impact case study database
Search and filter
Filter by
- Liverpool School of Tropical Medicine
- 1 - Clinical Medicine
- Submitting institution
- Liverpool School of Tropical Medicine
- Unit of assessment
- 1 - Clinical Medicine
- Summary impact type
- Health
- Is this case study continued from a case study submitted in 2014?
- No
1. Summary of the impact
Cryptococcal meningitis (CM) is an opportunistic fungal infection that is responsible for approximately 15-20% of deaths of people living with HIV-infection.
LSTM has played a leading role in pivotal trials that have evaluated novel approaches for the prevention and management of CM in low-resource settings. We have identified novel biomedical treatment and prevention strategies that are practical, low-cost and effective, which have been incorporated into World Health Organisation (WHO) guidelines and led to changes in clinical practice in Africa. Our research findings stimulated investment in a generic version of a key drug identified in our research (Flucytosine) to increase access for low-resource settings and also catalysed a USD20,000,000 investment from the global health organisation, UNITAID, enabling seven African countries to acquire the supported by the trials. These are being rolled out in the first quarter of 2021, combined with training of health care staff. Together, these improvements to CM management are saving tens of thousands of lives annually in Africa and other low-resource settings.
2. Underpinning research
Despite significant advances in the availability of HIV care and treatment, a large proportion of HIV positive (HIV+) patients in resource-poor settings will present with advanced disease and die from opportunistic infections. CM is the most common such infection. In 2014 there were 225,000 cases globally with 180,000 deaths. In low income countries (LICs), the 12-month mortality of cryptococcal meningitis is approximately 70%, compared to between 20% and 30% in high income countries (HICs). The main causes for this can be summarised as:
Limited ability to detect cases early and prevent development of disease
Limited availability of affordable and appropriate antifungals
Uncertainty about the optimal antifungal regimen
Limited ability of the healthcare system to monitor and manage treatment‐limiting toxicities of the antifungal drugs.
Our research focussed on two approaches for reducing deaths from CM in HIV+ people:
Combination Treatment
Lalloo (Director, LSTM, in collaboration with Oxford University) conceived, gained initial funding and instigated a phase 3 trial in Vietnam (conducted between 2004 and 2010) which compared 4 week amphotericin B monotherapy with 2 weeks combination therapy of amphotericin B and either flucytosine or fluconazole. A combination of intravenous amphotericin B combined with oral flucytosine led to a decrease of over 40% in all-cause mortality when compared with intravenous amphotericin B monotherapy. Combination therapy of amphotericin with fluconazole did not improve patient outcomes [1].
This finding clearly demonstrated the value of combination treatment with flucytosine for the first time, led to changes in WHO policy and changed clinical practice in South East Asia. However, a 2-week course of amphotericin B administered intravenously (i.v) requires regular toxicity monitoring and was prohibitively expensive in Africa, where the majority of CM cases occur. Furthermore, flucytosine was not registered anywhere in Africa and there were no generic formulations available. Instead in most of Africa, CM was routinely treated with 2 weeks of high dose oral fluconazole, which is associated with a high mortality rate (of approximately 70%).
The ACTA trial (Advancing Cryptococcal Meningitis Treatment for Africa 2015-2018; Jaffar, co-designed and co-supervised the trial and co-supervised the statistical analysis and dissemination; Lalloo: co-investigator, supported design and set up of Malawi sites) evaluated practical CM treatment regimens for Africa. It was the largest trial ever conducted in CM, with 9 sites in 5 countries, and compared three treatment strategies as well as the two alternative partner drugs for amphotericin B. The two new treatment strategies, consisting of either i) 1-week of i.v amphotericin B plus oral flucytosine or ii) an all oral combination of flucytosine plus fluconazole, were compared with a 2-week i.v. regime of amphotericin B combination-based treatment. One week of amphotericin B and flucytosine was associated with lower mortality, and 2 weeks of the oral drug combination was non-inferior to the 2 weeks of i.v amphotericin-based regimens. Both the novel 1-week i.v amphotericin B and all oral combination tested in ACTA had mortality rates between 2 and 3-fold lower than observed with previous standard treatment practice in Africa of fluconazole monotherapy [2]. Crucially, we also showed that the 1-week i.v amphotericin B was less costly as well as more effective when compared with the 2 weeks of i.v amphotericin-based regimens (reduction of USD500 per patient) [3]. The combination oral regimen was also highly cost-effective compared with fluconazole monotherapy, with an incremental cost-effectiveness ratio of between USD28 and USD44 per life-year saved (costs adjusted to 2015 USD prices) [4]. Thus, if flucytosine could be made available in Africa, then ACTA provides the evidence of regimens that could substantially reduce HIV-associated mortality in Africa at lower cost.
The potent steroid dexamethasone had commonly been used as an adjuvant in the treatment of CM to reduce brain swelling. However, Lalloo (in collaboration with Oxford University) co-designed and played a key part in running a phase 3 trial in 6 countries, between 2013 and 2014, which showed that dexamethasone did not reduce mortality in CM and was actually deleterious with more adverse events [5].
Detection and pre-emptive oral treatment
Once CM has developed (i.e. the infection has reached the brain), then treatment becomes hugely challenging. Screening for cryptococcal antigen (CrAg) in immunosuppressed patients, combined with pre-emptive antifungal treatment, can prevent many cases of CM as CrAg is detected in the blood weeks to months before symptoms of meningitis appear.
The REMSTART trial, between 2012 and 2015, led by Jaffar whilst at LSHTM, found that screening serum for CrAg combined with an adherence package led to a decrease of 28% (95% confidence intervals 10% - 43%) in all causes of mortality. After joining LSTM in 2015 Jaffar and Niessen led the health economics analysis of REMSTART, showing that the intervention was highly cost-effective with health service care cost per life-saved of USD70 (based on 2017 USD [6]). Jaffar was also a key investigator on a follow-on study, TRIP, funded by EDCTP (European and Developing Countries Clinical Trials Partnership) designed to facilitate the scale up of the approach tested in REMSTART. Working in 18 health facilities in Tanzania, TRIP demonstrated that the intervention could be scaled up in low-resource settings in a cost-effective way, achieving similar efficacy as seen under trial conditions.
Furthermore, the CRYPTO-PRO trial (commenced 2004; Lalloo PI: conceived, designed and supervised trial) demonstrated that use of routine fluconazole prophylaxis prevented cryptococcal disease in patients who could not rapidly access antiretroviral therapy [7].
3. References to the research
Day JN, Chau TTH, Wolbers M, Mai PP, Dung NT, Mai NH, Phu NH, Nghia HD, Phong ND, Thai CQ, Thai LH, Chuong LV, Sinh DX, Duong VA, Hoang TN, Diep PT, Campbell JI, Sieu TPM, Baker SG, Chau NVV, Hien TT, Lalloo DG, Farrar JJ. Combination antifungal therapy for cryptococcal meningitis. N Engl J Med. 2013. DOI: 10.1056/NEJMoa1110404
Molloy SF, Kanyama C, Heyderman RS, Loyse A, Kouanfack C, Chanda D, Mfinanga S, Temfack E, Lakhi S, Lesikari S, Chan AK, Stone N, Kalata N, Karunaharan N, Gaskell K, Peirse M, Ellis J, Chawinga C, Lontsi S, Ndong JG, Bright P, Lupiya D, Chen T, Bradley J, Adams J, van der Horst C, van Oosterhout JJ, Sini V, Mapoure YN, Mwaba P, Bicanic T, Lalloo DG, Wang D, Hosseinipour MC, Lortholary O, Jaffar S, Harrison TS; ACTA Trial Study Team. Antifungal Combinations for Treatment of Cryptococcal Meningitis in Africa. N Engl J Med. 2018. DOI: 10.1056/NEJMoa1710922
Chen T, Mwenge L, Lakhi S, Chanda D, Mwaba P, Molloy SF, Gheorghe A, Griffiths UK, Heyderman RS, Kanyama C, Kouanfack C, Mfinanga S, Chan AK, Temfack E, Kivuyo S, Hosseinipour MC, Lortholary O, Loyse A, Jaffar S, Harrison TS, Niessen LW; ACTA Trial Team. Healthcare Costs and Life-years Gained From Treatments Within the Advancing Cryptococcal Meningitis Treatment for Africa (ACTA) Trial on Cryptococcal Meningitis: A Comparison of Antifungal Induction Strategies in Sub-Saharan Africa. Clin Infect Dis. 2019. DOI: 10.1093/cid/ciy971
Shiri T, Loyse A, Mwenge L, Chen T, Lakhi S, Chanda D, Mwaba P, Molloy SF, Heyderman RS, Kanyama C, Hosseinipour MC, Kouanfack C, Temfack E, Mfinanga S, Kivuyo S, Chan AK, Jarvis JN, Lortholary O, Jaffar S, Niessen LW, Harrison TS. Addition of Flucytosine to Fluconazole for the Treatment of Cryptococcal Meningitis in Africa: A Multicountry Cost-effectiveness Analysis. Clin Infect Dis. 2020. DOI: 10.1093/cid/ciz163
Beardsley J, Wolbers M, Kibengo FM, Ggayi AB, Kamali A, Cuc NT, Binh TQ, Chau NV, Farrar J, Merson L, Phuong L, Thwaites G, Van Kinh N, Thuy PT, Chierakul W, Siriboon S, Thiansukhon E, Onsanit S, Supphamongkholchaikul W, Chan AK, Heyderman R, Mwinjiwa E, van Oosterhout JJ, Imran D, Basri H, Mayxay M, Dance D, Phimmasone P, Rattanavong S, Lalloo DG, Day JN; CryptoDex Investigators. Adjunctive Dexamethasone in HIV-Associated Cryptococcal Meningitis. N Engl J Med. 2016. DOI: 10.1056/NEJMoa1509024
Kimaro GD, Guinness L, Shiri T, Kivuyo S, Chanda D, Bottomley C, Chen T, Kahwa A, Hawkins N, Mwaba P, Mfinanga SG, Harrison TS, Jaffar S, Niessen LW. Cryptococcal Meningitis Screening and Community-based Early Adherence Support in People With Advanced Human Immunodeficiency Virus Infection Starting Antiretroviral Therapy in Tanzania and Zambia: A Cost-effectiveness Analysis. Clin Infect Dis. 2020. DOI: 10.1093/cid/ciz453
Parkes-Ratanshi R, Wakeham K, Levin J, Namusoke D, Whitworth J, Coutinho A, Mugisha NK, Grosskurth H, Kamali A, Lalloo DG; Cryptococcal Trial Team. Primary prophylaxis of cryptococcal disease with fluconazole in HIV-positive Ugandan adults: a double-blind, randomised, placebo-controlled trial. Lancet Infect Dis. 2011. DOI: 10.1016/S1473-3099(11)70245-6
4. Details of the impact
Our studies identified improved and cost-effective solutions to prevent and treat CM in low resource settings. The inclusion into WHO guidelines between 2013 and 2018 led to widespread adoption of these new approaches in most countries when practicable. This benefitted both individuals living with HIV and at risk of the opportunistic infection CM, and resource-poor healthcare systems, particularly in Africa, by improving outcomes and reducing the costs associated with CM treatment.
Influence upon treatment guidelines and clinical practice.
The Vietnam trial demonstrating clear benefit from combination therapy with amphotericin and flucytosine in terms of survival rates, and showing that both drugs could be safely administered in settings where therapeutic drug monitoring is not available, informed policy and clinical practice in many countries (including Germany, USA, Australia and Indonesia) and has underpinned WHO recommendations for combination therapy since 2013 [1,2].
Although most countries in Africa typically follow WHO guidance, this new treatment regime was not widely adopted in this continent due to resource constraints. The ACTA trial evaluated practical comparatively low-cost interventions for Africa including a new, shorter treatment regime which proved to be highly cost effective. It led to an immediate change in WHO guidelines, published simultaneously alongside the paper in 2018 recommending this regimen as the gold standard for CM management [2]. The advice to change to the ACTA regimens was graded as “strong recommendation, moderate-certainty evidence”. The dexamethasone study demonstrating that use of corticosteroids was harmful in HIV-related CM was also cited as high-quality evidence against the use of dexamethasone in the 2018 WHO guidelines [2] and immediately influenced clinical practice in many countries, being adopted into US IDSA guidelines in the same year for example. The findings of all these trials are included in the on-line decision-making resource ‘UpToDate’ used by 1,900,000 doctors worldwide [3].
The WHO had been recommending CrAg screening and it changed its guidelines to “strong recommendation” following the REMSTART trial [4]. However, uptake of this recommendation has been very limited outside of South Africa (which has a well-funded health system), because the REMSTART package was a complex intervention involving both patient empowerment and CrAg screening and needed substantial infrastructure and resources to scale-up. The TRIP study adapted the REMSTART package and demonstrated that a) it can be scaled up in a low-resource setting and b) that in a real-life setting, the death rate is as low as observed under trial conditions in the REMSTART trial. Tanzania, with one of the weakest infrastructures for their HIV programmes, has used the evidence generated to scale up screening for CrAg in over 200 health facilities so far (over 1,000,000 people with HIV-infection in the country) and by the end of 2021 coverage is expected to cover the entire country.
The CRYPTO-PRO trial demonstrated the value of fluconazole prophylaxis in certain populations and was one of only two studies contributing to “high quality evidence” for recommendations in the 2017 WHO advanced HIV guidelines [4] and 2018 WHO Cryptococcal guidelines [2] recommending that fluconazole prophylaxis should be used in patients with advanced HIV that could not immediately access antiretroviral therapy.
Implementation of revised guidelines for CM treatment and diagnosis.
As a result of the ACTA trial findings and WHO recommendations that followed, in January 2019 UNITAID announced USD20,000,000 funding to support 7 African countries to scale the WHO recommendation on advanced HIV disease (which REMSTART and TRIP had informed) and scale up management of CM (which ACTA had informed) [5]. The African countries targeted by UNITAID were Tanzania, Nigeria, South Africa, Botswana, Malawi, Lesotho, and Uganda and they have used the funds to support training of health care staff and acquire commodities including CrAg tests and treatments (flucytosine and amphotericin). The US Centers for Disease Control has also invested in supporting governments to scale up CrAg screening in these countries and approximately 21 other countries in Africa, who are at varying levels of scale-up. On the basis of our research, it is evident that this rapid adoption of improved screening and treatment strategies has been key to saving tens of thousands of lives in the continent with the highest burden of CM.
Increased availability of treatment
Prior to the ACTA trial, flucytosine was not registered in Africa and no generic formulation existed. The results from this trial incentivised the pharmaceutical industry to manufacture flucytosine and the WHO approved the first generic version of this drug, manufactured by Mylan, in 2018 [6]. Other pharmaceutical companies started working on generic flucytosine in 2019 including Lupin Pharmaceuticals (India), Strides Pharma (India), Macleods Pharmaceuticals (India). This will ensure, for the first time, that CM management will become widely available in many more centres. In those centres unable to provide the gold standard of i.v. amphotericin and flucytosine, an effective oral combination regimen of fluconazole and flucytosine can be used (this is far superior than using fluconazole alone, as was common practice prior to the ACTA trial).
5. Sources to corroborate the impact
Influence upon treatment guidelines and clinical practice.
Treatment guidelines
2019 Indonesian guidelines for the treatment of HIV: PEDOMAN NASIONAL PELAYANAN KEDOKTERAN TATA LAKSANA HIV http://siha.depkes.go.id/portal/files_upload/PNPK_HIV_Kop_Garuda__1_.pdf
Australian Consensus guidelines for the treatment of yeast infections in the haematology, oncology and intensive care setting 2014. DOI: 10.1111/imj.12597
German 2016: CNS infections in patients with hematological disorders (including allogeneic stem-cell transplantation)—Guidelines of the Infectious Diseases Working Party (AGIHO) of the German Society of Hematology and Medical Oncology (DGHO). DOI: 10.1093/annonc/mdw155
US department of Health and Human Services Guidelines for the Prevention and Treatment of Opportunistic Infections in Adults and Adolescents with HIV. http://aidsinfo.nih.gov/contentfiles/lvguidelines/adult_oi.pdf]
World Health Organisation Guidelines For The Diagnosis, Prevention And Management Of Cryptococcal Disease In HIV-Infected Adults, Adolescents And Children March 2018. https://www.who.int/hiv/pub/guidelines/cryptococcal-disease/en/
UpToDate Treatment of Cryptococcal Meningitis: https://www.uptodate.com/contents/cryptococcus-neoformans-treatment-of-meningoencephalitis-and-disseminated-infection-in-hiv-seronegative-patients?search=cryptococcus&source=search_result&selectedTitle=3~128&usage_type=default&display_rank=3#H3349804803
Guidelines for managing advanced HIV disease and rapid initiation of antiretroviral therapy, July 2017. Geneva: World Health Organization. https://www.who.int/hiv/pub/guidelines/advanced-HIV-disease/en/
Implementation of revised guidelines for CM diagnosis and treatment.
- Information on UNITAID’s investment is available here: https://unitaid.org/news-blog/targeting-opportunistic-infections-to-cut-hiv-related-deaths/#en
Increased availability of treatment
- Prequalification of Flucytosine ( https://extranet.who.int/prequal/medicine/3628)
- Submitting institution
- Liverpool School of Tropical Medicine
- Unit of assessment
- 1 - Clinical Medicine
- Summary impact type
- Health
- Is this case study continued from a case study submitted in 2014?
- No
1. Summary of the impact
Insecticide Treated Nets (ITNs) are the main malaria prevention tool in Africa, but their efficacy is being eroded by pyrethroid resistance in the mosquito vectors. LSTM’s research determining the underlying molecular mechanisms led to new classes of ITNs that can control resistant mosquito populations. Our research and advocacy led to the founding of the Product Development Partnership IVCC and has generated the evidence leading to deployment of new classes of ITNs. By 2020, 13 of the 23 malaria endemic countries in Africa included these new classes of nets in their national distribution campaign, protecting over 35,000,000 individuals. A large-scale trial in Uganda found that use of this new net class reduced malaria prevalence by 27%.
2. Underpinning research
Since 2000 the global burden of malaria, especially in Africa, has reduced. Clinical incidence of malaria across the continent decreased by over 50% between 2000 and 2018, and annual deaths decreased from 596,000 to 405,000. The decline has been largely attributed to the distribution of over 2,000,000,000 pyrethroid insecticide-treated bednets (ITNs) protecting at-risk populations from bites from malaria mosquitoes that typically feed indoors and at night. However, the success of ITNs is threatened by the rapid increase in pyrethroid resistance in mosquitoes. All ITNs contain pyrethroids, and resistance is putting the entire global fight to reduce, and ultimately eliminate, malaria at risk. Historically, malaria resurges rapidly if effective interventions are not maintained at an adequate level of coverage and effectiveness. Reversing this trend is difficult and imposes major economic and public health burdens on the disease endemic countries.
LSTM has pioneered research into the causes, consequences and rapid spread of pyrethroid resistance in African malaria vectors and provided evidence-based solutions to combat the problem.
Mosquito genetics and behaviour
Understanding the genetic basis of resistance in mosquitoes has driven the development of strategies to lessen its impact. Since 2003, LSTM researchers (Donnelly, Paine, Lycett, Ranson, Weetman, Wondji) used a variety of genomic and biochemical approaches to identify the key genetic changes associated with pyrethroid resistance. We found the most potent mechanism present in African malaria vectors to be increased rates of insecticide detoxification, caused by elevated production of a small number of cytochrome P450 enzymes that are very efficient at metabolizing this insecticide class [1]. We developed in vitro and in vivo platforms for screening new insecticides for potential cross-resistance, which have been widely adopted by agrochemical companies [2]. Novel video techniques (developed by McCall) to observe and measure mosquito behaviour at bed nets demonstrated that the vast majority of activity occurs at and above the roof of the net [3], a seminal discovery that had major impacts for bednet design; for example, in trials in Liverpool and Burkina Faso, McCall has shown that small net panels, or barriers, attached outside the top of a bed net, improve the net’s performance, while using less insecticide.
Impact of resistance on malaria control
LSTM has been monitoring and mapping insecticide resistance spread in Africa for over 20 years and has supported World Health Organization (WHO) in the generation, analysis and dissemination of this data via lead author contributions to the Global Plan for Insecticide Resistance in Malaria Vectors (2012) (Coleman, Ranson and Hemingway) and the Global Report on Insecticide Resistance in Malaria Vectors; 2010-2016 (2018) (Coleman). We have developed new bioassay approaches for pyrethroids (and new insecticides with novel modes of action) and developed a comprehensive suite of molecular markers to track the spread of resistance through the application of genomic technologies (Ranson, Weetman, Wondji, Donnelly). These tools have enabled the impact of resistance on malaria transmission to be directly assessed [4] and are being incorporated into WHO guidelines.
Evaluation of new vector control tools to combat insecticide resistance
The Liverpool Insect Testing Establishment (LITE), established by Ranson and housed within LSTM’s Vector Biology Department, has developed a screening pipeline to evaluate new insecticides and formulations against fully characterized resistant mosquito colonies [2]. All the major insecticide and ITN manufacturers have utilized LITE’s services to assess performance of new insecticides and formulated products against our mosquito colonies, and many of these industry partners have also enlisted LSTM’s expertise for field trials. For example, an LSTM-led consortium conducted the first clinical trial of a dual insecticide bednet (Olyset Duo®) in Burkina Faso, which demonstrated a significant public health benefit of these nets in areas of high pyrethroid resistance (decrease of 12% in malaria incidence, p=0.04) [5]. Hemingway and Donnelly led the evaluation of the first operational deployment of PBO-pyrethroid nets in Uganda; 12 months post deployment, PBO-pyrethroid nets reduced malaria infection prevalence by 27% (p=0.0001) and malaria mosquito density in houses by 87% (p<0.0001) relative to conventional nets [6].
3. References to the research
Mitchell SN, Stevenson BJ, Müller P, Wilding CS, Egyir-Yawson A, Field SG, Hemingway J, Paine MJ, Ranson H, Donnelly MJ. Identification and validation of a gene causing cross-resistance between insecticide classes in Anopheles gambiae from Ghana. Proc Natl Acad Sci U S A. 2012. DOI: 10.1073/pnas.1203452109
Lees RS, Ismail HM, Logan RAE, Malone D, Davies R, Anthousi A, Adolfi A, Lycett GJ, Paine MJ. New insecticide screening platforms indicate that Mitochondrial Complex I inhibitors are susceptible to cross-resistance by mosquito P450s that metabolise pyrethroids. Sci Rep. 2020. DOI: 10.1038/s41598-020-73267-x
Parker JE, Angarita-Jaimes N, Abe M, Towers CE, Towers D, McCall PJ. Infrared video tracking of Anopheles gambiae at insecticide-treated bed nets reveals rapid decisive impact after brief localised net contact. Sci Rep. 2015. DOI: 10.1038/srep13392
Barnes KG, Weedall GD, Ndula M, Irving H, Mzihalowa T, Hemingway J, Wondji CS. Genomic Footprints of Selective Sweeps from Metabolic Resistance to Pyrethroids in African Malaria Vectors Are Driven by Scale up of Insecticide-Based Vector Control. PLoS Genet. 2017. DOI: 10.1371/journal.pgen.1006539
Tiono AB, Ouédraogo A, Ouattara D, Bougouma EC, Coulibaly S, Diarra A, Faragher B, Guelbeogo MW, Grisales N, Ouédraogo IN, Ouédraogo ZA, Pinder M, Sanon S, Smith T, Vanobberghen F, Sagnon N, Ranson H, Lindsay SW. Efficacy of Olyset Duo, a bednet containing pyriproxyfen and permethrin, versus a permethrin-only net against clinical malaria in an area with highly pyrethroid-resistant vectors in rural Burkina Faso: a cluster-randomised controlled trial. Lancet. 2018. DOI: 10.1016/S0140-6736(18)31711-2
Staedke SG, Gonahasa S, Dorsey G, Kamya MR, Maiteki-Sebuguzi C, Lynd A, Katureebe A, Kyohere M, Mutungi P, Kigozi SP, Opigo J, Hemingway J, Donnelly MJ. Effect of long-lasting insecticidal nets with and without piperonyl butoxide on malaria indicators in Uganda (LLINEUP): a pragmatic, cluster-randomised trial embedded in a national LLIN distribution campaign. Lancet. 2020. DOI: 10.1016/S0140-6736(20)30214-2
4. Details of the impact
The beneficiaries of two decades of research at LSTM on the biology and behaviour of malaria mosquitoes include: (1) bednet manufacturers, who have translated our research into new ‘resistance-breaking’ nets that now make up a significant share of total net sales; (2) communities in malaria-endemic regions of Africa, who are now at reduced risk of malaria; (3) global policy makers and implementers, who have a strengthened evidence base on which to select malaria-prevention tools; and (4) national malaria control programmes, who have an improved toolset to mitigate against insecticide resistance.
Impact on bednet manufacturers, agrochemical companies and the communities their products protect
LSTM’s research into the mechanisms of insecticide resistance has directly led to the development of new ITNs. Our evidence that blocking P450-based metabolism with the synergist piperonyl butoxide (PBO) can restore pyrethroid efficacy in malaria mosquitoes led to manufacturers developing a new class of nets that contain pyrethroids plus PBO, with the aim of maintaining ITN efficacy even in the face of pyrethroid resistance. Four manufacturers (Vestergaard (Switzerland), Sumitomo Chemical Co., Ltd (Japan), AKA Polymers (India) and Moon Netting (Pakistan)) now produce PBO-pyrethroid nets. These have all been pre-qualified by WHO (a necessary step before most major donors will procure a new type of net). Over 43,000,000 PBO-pyrethroid nets were distributed in Africa in 2020, protecting over 95,000,000 people. Since 2020, 27 of the 40 African national ITN distribution programmes included PBO-pyrethroid nets, with PBO-nets constituting 21% of the market share of nets delivered to Africa for malaria prevention in 2020 [1].
The discovery that malaria vector mosquitoes preferentially contact the top of a net when seeking a bloodmeal led one major net manufacturer (Vestergaard) to incorporate PBO only into the roof panel in their market leading PermaNet 3.0 ®. Since 2017, LSTM have been working with Vestergaard to develop novel bednet prototypes exploiting this aspect of mosquito behaviour. Limiting insecticides to small external net barriers improves the net’s performance, while using less insecticide thereby reducing manufacturers costs, and limiting the risk of adverse effects caused by insecticide exposure [2, 3].
Our clinical trial of the pyriproxyfen-pyrethroid net, Olyset Duo®, demonstrated the potential of insect sterilizing agents to reduce malaria transmission when combined with pyrethroid insecticides in bednets. Although Olyset Duo® is not currently being deployed, the positive results of our trial of ITNs with this mode of action led to a very similar competitor product (Royal Guard, DCT USA that was listed by WHO in 2019) being deployed in Mozambique and Nigeria since 2020 (1,000,000 nets distributed in 2020) [1, 4].
Hemingway established the Innovative Vector Control Consortium (IVCC) in 2005 at LSTM. IVCC is the leading global agency for the development of new insecticides and vector control tools. The consortium has re-formulated two classes of agricultural insecticide that have been in use for indoor residual spraying, a very effective alternative to ITNs for malaria control, since 2014. Rotation of these chemistries has enabled the implementation of resistance management strategies, and these new formulations have been deployed in indoor residual spraying in 30 malaria endemic countries, averting over 4,800,000 malaria cases between 2016 and 2019 [5]. In addition, the work of the Liverpool Insect Testing Establishment (LITE), plus the suite of tools developed by LSTM including the P450 in vitro screen and transgenic mosquito lines, has streamlined the insecticide development pathway, terminating investment in chemistries exhibiting potential resistance liabilities and accelerating the most promising candidates. For example, the multi-national agrochemical company Syngenta (Switzerland) used the LSTM testing pipeline as a critical differentiator to drive candidate selection and investment decisions [6].
Impact on global policy makers, implementation agencies and national malaria programmes
LSTM led the evaluation of PBO-pyrethroid classes of nets pre and post deployment. Our meta analyses of the performance of PBO-pyrethroid versus standard ITNs on mosquito populations, which Ranson presented at Evidence Review Groups on PBO-pyrethroid nets at WHO in 2015 and 2017, led to policy recommendations on use of this net class (WHO 2015, 2017) [7]. We conducted one of only two clinical trials that have demonstrated significant improved public health benefit of PBO-pyrethroid nets in reducing malaria transmission compared to standard ITNs; the scale of this trial was unprecedented and enabled 10,700,000 Ugandans to benefit from the increased protection afforded by this new net class (reduction of 27% in malaria prevalence) whilst the evaluation was ongoing [8].
LSTM vector biologists play a key role in advising WHO. Donnelly was the lead entomologist on the WHO study on the impact of insecticide resistance on malaria control (between 2012 and 2016) which helped raise the profile of the public health threat posed by pyrethroid resistance. We have been members of the WHO Vector Control Advisory Group (VCAG) since its inception in 2014 (Hemingway, Ranson) advising WHO on trial designs, and evaluating results from trials prior to policy recommendations. Wondji is a member of the WHO pre-qualification team of independent assessors who review data packages submitted by ITN manufacturers to determine whether these meet accepted WHO standards [9]
LSTM has made a major contribution to the development of WHO guidelines on resistance monitoring and management, both from the outputs of our research which has informed best practice, and via our participation in WHO committees. For example, Ranson and Donnelly contributed to the 2016 WHO guidelines on insecticide resistance monitoring and Coleman, Hemingway and Ranson were major contributors to the 2012 WHO Global Report on Insecticide Resistance Management in malaria vectors [10]. Via long-term partnerships with African Ministries of Health, we have greatly strengthened capacity in resistance monitoring and supported the interpretation of national data to inform the sub national deployment of different insecticide-based strategies (e.g. Zambia and Equatorial Guinea Insecticide Resistance Management Plans and policy briefs for Malawi [11]).
5. Sources to corroborate the impact
Impact on bednet manufacturers, agrochemical companies and the communities their products protect
Database on net procurements for 2020: https://allianceformalariaprevention.com/net-mapping-project/
Letter from CEO of Vestergaard Frandsen confirming impact of mosquito behaviour study on net design
Murray GPD, Lissenden N, Jones J, Voloshin V, Toé KH, Sherrard-Smith E, Foster GM, Churcher TS, Parker JEA, Towers CE, N'Falé S, Guelbeogo WM, Ranson H, Towers D, McCall PJ. Barrier bednets target malaria vectors and expand the range of usable insecticides. Nat Microbiol. 2020. DOI: 10.1038/s41564-019-0607-2
https://www.ivcc.com/market-access/new-nets-project/ (Evidence Base for New Dual-AI Nets download).
Data available on NGENIRs website: https://www.ivcc.com/market-access/ngenirs/ (see NGENIRS Evidence, NGENIRS project overview)
Letter from Syngenta, confirming changes in lead candidate after evaluation using LSTM testing pipeline.
Impact on global policy makers, implementation agencies and national malaria programmes
Conditions for deployment of mosquito nets treated with a pyrethroid and piperonyl butoxide: WHO reference number: HTM/GMP/2017.17
Staedke SG, Gonahasa S, Dorsey G, Kamya MR, Maiteki-Sebuguzi C, Lynd A, Katureebe A, Kyohere M, Mutungi P, Kigozi SP, Opigo J, Hemingway J, Donnelly MJ. Effect of long-lasting insecticidal nets with and without piperonyl butoxide on malaria indicators in Uganda (LLINEUP): a pragmatic, cluster-randomised trial embedded in a national LLIN distribution campaign. Lancet. 2020. DOI: 10.1016/S0140-6736(20)30214-2
Letter from WHO Global Malaria Programme, acknowledging LSTM’s contribution to WHOs work on ITNs
Global Malaria Programme, WHO, Test procedures for insecticide resistance monitoring in malaria vector mosquitoes 2016 (2nd edition) https://www.who.int/malaria/publications/atoz/9789241511575/en/; Global Malaria Programme, WHO, Global Report on Insecticide Resistance Management in malaria vectors 2012 https://www.who.int/malaria/publications/atoz/gpirm/en/
Zambia: Chanda E, Thomsen EK, Musapa M, Kamuliwo M, Brogdon WG, Norris DE, Masaninga F, Wirtz R, Sikaala CH, Muleba M, Craig A, Govere JM, Ranson H, Hemingway J, Seyoum A, Macdonald MB, Coleman M. An Operational Framework for Insecticide Resistance Management Planning. Emerg Infect Dis. 2016. DOI: 10.3201/eid2205.150984. Equatorial Guinea: Hemingway J, Vontas J, Poupardin R, Raman J, Lines J, Schwabe C, Matias A, Kleinschmidt I. Country-level operational implementation of the Global Plan for Insecticide Resistance Management. Proc Natl Acad Sci U S A. 2013. DOI: 10.1073/pnas.1307656110. Malawi: https://www.piivec.org/resources/evidence-to-inform-how-new-bed-nets-can-be-used-to-prevent-malaria-in-malawi
- Submitting institution
- Liverpool School of Tropical Medicine
- Unit of assessment
- 1 - Clinical Medicine
- Summary impact type
- Health
- Is this case study continued from a case study submitted in 2014?
- No
1. Summary of the impact
Snakebite has historically been perhaps the most under-researched, under-resourced high mortality and high morbidity neglected tropical disease. The research and advocacy activities of Liverpool School of Tropical Medicine (LSTM) have impacted three key areas:
defining the burden of and consequences of the problem, permitting increased funder and policymaker focus on snakebite - this directly led to the World Health Organization (WHO) formally classifying snakebite as a priority Neglected Tropical Disease in 2017 and, in 2019, establishing a strategy (co-written by LSTM scientists) to halve the global snakebite mortality and morbidity by 2030;
design and implementation of a model intervention program, ensuring the delivery of significantly improved snakebite therapies (antivenoms) to countries in West Africa – this resulted in new, cost-effective, life-saving treatments;
securing the supply of quality antivenoms by developing a ‘prequalification’ programme for ensuring the quality of snakebite therapeutics for Africa; this in turn has resulted in direct commercial impact for antivenom manufacturers.
2. Underpinning research
Snakebite is a medical emergency and the world’s most lethal neglected tropical disease. Snakebite predominately affects poor rural populations of the tropics and results in approximately 138,000 deaths and approximately 400,000 disabilities per annum. Snakebite has long been neglected by funders and policy makers, which has resulted in only limited investment and funding. Studies led by LSTM have revealed the true burden of envenoming and have led to more efficacious, safer and affordable snakebite therapies.
Impacts have been achieved in collaboration with the University of Kelaniya in Sri Lanka, the Nigerian Federal Ministry of Health and Bayero University of Kano, Nigeria and, more recently, from the 96+ researchers that LSTM has gathered for snakebite clinical and public health research in Nigeria, Kenya, eSwatini and India.
Quantifying the burden of disease
LSTM-led research has been central to the international effort to increase awareness of the substantial disease burden posed by snakebite worldwide. Lalloo coordinated the highly influential publication estimating the global burden of snakebite, snakebite envenomings and snakebite deaths based on literature analysis and modelling [1]. Harrison and Lalloo next demonstrated that snakebite is a disease of rural tropical poverty by evidencing clear associations between snakebite mortality and key economic and socioeconomic indices of poverty [2]. Lalloo’s later publication [3] was the first to demonstrate that snakebite also causes severe chronic psychological morbidity.
Laboratory and Clinical Evaluation of Antivenoms
The EchiTAb Study Group was established by LSTM and partners in 2006 and led by Theakston and Harrison as a response to the international crisis in the supply of effective antivenom for African snakebite victims, first highlighted by Theakston [4]. This collaboration of scientists, physicians, antivenom manufacturers and Nigerian Federal Ministry of Health representatives was funded by GBP1,920,000 from the Nigerian Federal Ministry of Health (between 2006 and 2012). The EchiTAb study group focussed on mitigating snakebite mortality and morbidity in West Africa. Medically-important snakes were exported from Nigeria and housed at LSTM where research on the venoms was performed. LSTM designed and delivered venom mixtures for antivenom production by manufacturers in UK, Costa Rica, Egypt, Colombia and Mexico. LSTM then preclinically tested the efficacy of the resulting 5 antivenoms. Of these new antivenoms, 3 were submitted to (i) phase I clinical dose-finding and safety studies (resulting in the deletion of 1 product with an unacceptable safety profile) and (ii) a randomised controlled double-blind non-inferiority study of the remaining 2 antivenoms in Nigeria conducted between 2005 and 2007. This resulted in demonstrable efficacy and safety for both products [5], and remains the largest clinical trial undertaken on snakebite to date. Harrison and Casewell also demonstrated preclinical efficacy of these products against other snake species to which the antivenoms were not originally designed, thereby advocating for their broader clinical use across sub-Saharan Africa [6].
The initiation, funding, coordination, development, testing and delivery of the EchiTAb antivenoms exemplifies LSTM’s ‘bench to bedside’ research philosophy and capability. Other studies have been key in improving patient outcomes related to how antivenoms are used. Lalloo co-designed and ran a major clinical trial in partnership with the University of Kelaniya demonstrating that severe, potentially life-threatening, antivenom reactions could be decreased by 25% using adrenaline (epinephrine) prophylaxis [7].
3. References to the research
Kasturiratne A, Wickremasinghe AR, de Silva N, Gunawardena NK, Pathmeswaran A, Premaratna R, Savioli L, Lalloo DG, de Silva HJ. The global burden of snakebite: a literature analysis and modelling based on regional estimates of envenoming and deaths. PLoS Med. 2008. DOI: 10.1371/journal.pmed.0050218
Harrison RA, Hargreaves A, Wagstaff SC, Faragher B, Lalloo DG. Snake envenoming: a disease of poverty. PLoS Negl Trop Dis. 2009. DOI: 10.1371/journal.pntd.0000569
Williams SS, Wijesinghe CA, Jayamanne SF, Buckley NA, Dawson AH, Lalloo DG, de Silva HJ. Delayed psychological morbidity associated with snakebite envenoming. PLoS Negl Trop Dis. 2011. DOI: 10.1371/journal.pntd.0001255
Theakston RD, Warrell DA. Crisis in snake antivenom supply for Africa. Lancet. 2000. DOI: 10.1016/s0140-6736(05)74319-1
Abubakar IS, Abubakar SB, Habib AG, Nasidi A, Durfa N, Yusuf PO, Larnyang S, Garnvwa J, Sokomba E, Salako L, Theakston RD, Juszczak E, Alder N, Warrell DA; Nigeria-UK EchiTab Study Group. Randomised controlled double-blind non-inferiority trial of two antivenoms for saw-scaled or carpet viper (Echis ocellatus) envenoming in Nigeria. PLoS Negl Trop Dis. 2010. DOI: 10.1371/journal.pntd.0000767
Casewell NR, Cook DA, Wagstaff SC, Nasidi A, Durfa N, Wüster W, Harrison RA. Pre-clinical assays predict pan-African Echis viper efficacy for a species-specific antivenom. LoS Negl Trop Dis. 2010. DOI: 10.1371/journal.pntd.0000851
de Silva HA, Pathmeswaran A, Ranasinha CD, Jayamanne S, Samarakoon SB, Hittharage A, Kalupahana R, Ratnatilaka GA, Uluwatthage W, Aronson JK, Armitage JM, Lalloo DG, de Silva HJ. Low-dose adrenaline, promethazine, and hydrocortisone in the prevention of acute adverse reactions to antivenom following snakebite: a randomised, double-blind, placebo-controlled trial. PLoS Med. 2011. DOI: 10.1371/journal.pmed.1000435
4. Details of the impact
Positioning snakebite as a priority neglected tropical disease
LSTM has been central to the international effort to increase awareness of the substantial disease burden posed by snakebite that culminated in the WHO recommendation that snakebite be listed as a priority neglected tropical disease. We organised highly influential international meetings that raised the global profile of snakebite, including the Wellcome-funded ‘ Mechanisms to reverse the public health neglect of snakebite victims’ 2015 meeting chaired by Harrison [1], and the 2016 Kofi Annan Foundation meeting, also organized and chaired by Harrison, in which Mr Kofi Annan declared snakebite as “ the biggest public health crisis you’ve never heard of ” [2]. In 2018, Casewell co-organised the Dutch Government funded “ Snakebite: from science to society” international meeting that brought over 300 individuals (scientists, clinicians, pharmaceutical companies, public health practitioners, charities, policy makers, governments and funders) together to discuss solutions for tropical snakebite [3]. These meetings were instrumental in effecting change in international health policy. Initiatives resulting from this series of meetings include, WHO’s 2018 listing of snakebite as a priority NTD (see below), the 2019 WHO strategy to halve snakebite mortality and morbidity by 2030 co-authored by Harrison and Lalloo [4] and Wellcome’s 2019 investment of GBP80,000,000 in snakebite research.
The 2017 WHO recommendation that snakebite be listed as a priority neglected tropical disease. was ratified by the World Health Assembly in 2018 [4] and has galvanised action from several governments. For example, the Kenya Ministry of Health NTD department has established a Snakebite Task Force, which includes a LSTM-research collaborator (Dr George Omondi) in its membership, and written national guidelines [5], that LSTM and its Kenya partners are delivering to hospitals and communities across Kenya. While awaiting full impact assessments, we have already observed increased hospital admission of snakebite victims (Kitui County) and demand for antivenom from Ministries of Health (Kitui & Baringo Counties).
LSTM has led many other advocacy activities that have improved global recognition of the public health burden posed by snakebite including advising and participating in numerous documentaries with a global reach between 2013 and 2019 for the BBC, BBC World, Discovery Channel, China Global Television Network, Natural History Museum and Royal Society for Tropical Medicine and Hygiene [6]. In particular, Harrison had extensive input into the development of a documentary with the Lillian Lincoln Foundation entitled “ Minutes to Die” [6] which highlighted the plights of victims and the scientists seeking to redress the issue.
In 2018, LSTM was a main partner with the Royal Society of Tropical Medicine & Hygiene in the development of the inaugural International Snakebite Awareness Day. This initiative included participation from the Wellcome Trust, the Global Snakebite Initiative, Medecins Sans Frontiers, the Kofi Annan Foundation and Health Action International. Casewell and Harrison filmed a short documentary film on snakebite and contributed to the recording of BBC World visual and audio documentaries on snakebite in Kenya, both of which were used in the inaugural event in London [6]. Casewell’s co-written opinion piece for the general public on the BBC News website in 2019 “Why are so many people still dying from snake bites?” [6] has been viewed over 500,000 times.
Improving snakebite therapies
The EchiTAb Study Group activities resulted in the development of two new, efficacious, safe and cost-effective antivenoms that received National Marketing Authorisations from the Nigerian National Agency for Food and Drug Administration and Control (NAFDAC), in 2005 [7]. A two year evaluation on the impact of the introduction of these antivenoms into Nigeria, conducted between 2009 and 2010, found snakebite mortality decreased from 35-45% in the absence of antivenom to 1.52% after the venom was introduced [8]. In addition, the dose of antivenom required with the new therapeutics was decreased from 6 vials to between 1 and 3 vials, compared with previous products [8], thereby also resulting in a decrease of at least 50% to the cost of treatment to impoverished victims and the health system. Our in-country partners indicate that these pre-REF 2021 efficacy, cost-effectiveness and mortality-reduction figures have remained constant thereafter and the EchiTAb antivenoms remain the standard of care in Nigeria and other countries (see below).
Our findings that adrenaline prophylaxis can reduce severe reactions to antivenom led to the routine recommendation of adrenaline prophylaxis in WHO guidelines in 2016 [9]; adrenaline is included as a strong recommendation in almost all national snakebite guidelines. The routine use of adrenaline prophylaxis has been reported to be associated with reduced mortality from snakebite; some clinicians avoided antivenom use because of their fear of reactions and being able to prevent or manage reactions using adrenaline increased the use of life-saving antivenom [10].
Enabling access to antivenom
As a result of our advocacy on the need for quality assurance processes, WHO have established an ‘antivenom prequalification’ process, by which independent risk benefit analyses following robust laboratory assessment of available antivenoms are performed to ensure the quality of snakebite therapeutics for Africa [11]. The EchiTAb Study Group-developed antivenom EchiTAbG, manufactured by MicroPharm Limited, was the first product to receive WHO-approval via this process in 2019.
The EchiTAb Study Group project also resulted in commercial and reputational benefits to the antivenom manufacturers. Thus, the SME, MicroPharm Limited (UK) benefitted from (i) 5 years investment in their production of EchiTAbG, (ii) ownership of a brand that is now the standard snakebite treatment throughout Nigeria and neighbouring countries, and (iii) an Africa-antivenom production reputation that resulted in its selection to sustain the production/delivery of FavAfrique from Sanofi (2019/20). Similarly, based upon the success of its antivenom (EchiTAb-Plus) in Nigeria, the Costa Rican antivenom manufacturer, Instituto Clodomiro Picado (ICP), is now distributing EchiTAb-Plus to many other countries, including Burkina Faso, Mali, Ghana, Central African Republic and irregularly to other West African countries. While no formal epidemiological studies have quantified the impact this antivenom has had in these countries, the similar pattern of snake envenoming and well documented challenges with poor quality or non-availability of antivenom make it highly likely that there has been a decrease in mortality of at least 33%, as observed in north-east Nigeria [8]. The EchiTAb Study Group project has since become recognised as a model for north-south and south-south collaboration for delivery of significantly improved snakebite therapies to tropical countries in greatest need [12]. Indeed, using the ‘EchiTAb’ model, ICP has established similar new partnerships in Sri Lanka (~2014-2018) and Papua New Guinea (~2012-2016) to deliver new life-saving antivenom products to market in these resource-poor, high snakebite burden, areas [12].
5. Sources to corroborate the impact
Positioning snakebite as a priority neglected tropical disease
Publication: Harrison RA, Gutiérrez JM. Priority Actions and Progress to Substantially and Sustainably Reduce the Mortality, Morbidity and Socioeconomic Burden of Tropical Snakebite. Toxins (Basel). 2016. DOI: 10.3390/toxins8120351
Meeting report: Kofi Annan Foundation meeting on snakebite https://www.kofiannanfoundation.org/combatting-hunger/public-health-snakebite/
Meeting report: Snakebite: from science to society. 2018
Strategy: World Health Organization. (2019) Snakebite envenoming: A strategy for prevention and control (Lalloo and Harrisson co-authors). https://apps.who.int/iris/bitstream/handle/10665/324838/9789241515641-eng.pdf
Kenyan national guidelines, https://kma.co.ke/Documents/Snakebite%20Envenoming%20in%20Kenya.pdf
Documentaries: BBC World and Lillian Lincoln
https://www.youtube.com/watch?v=TKoQrXcDafc&fbclid=IwAR2Ob4kBATotAs03B5XpeDa4UeACqra5K3581ZynpAGaQIyG5XCb8E7Xnpg&app=desktop and https://www.youtube.com/channel/UCrLPYxMWPLVAqCbho6Oy70Q and https://www.bbc.co.uk/news/world-45332002
Improving snakebite therapies
- Website: Nigeria Drugs and Devices website for EchiTAbG antivenom
https://rxnigeria.com/en/items?task=view&id=2639 and for EchiTAb-Plus antivenom
- Publication: Ademola-Majekodunmi FO, Oyediran FO, Abubakar SB. Incidence of snakebites in Kaltungo, Gombe State and the efficacy of a new highly purified monovalent antivenom in treating snakebite patients from January 2009 to December 2010. Bull Soc Pathol Exot. 2012. DOI: 10.1007/s13149-012-0232-2
1. Guidelines for the management of snakebites, 2nd edition. https://www.who.int/snakebites/resources/9789290225300/en/
1. Statement on how use of adrenaline (epinephrine) prophylaxis has led to increased use of anti-venom in India. Bawaskar HS, Bawaskar PH. Snakebite envenoming. Lancet. 2019. DOI: 10.1016/S0140-6736(18)32745-4
Enabling access to antivenom
WHO Assessment and Listing of Antivenoms: https://www.who.int/medicines/news/snake_antivenoms_assessment_listing/en/
Examples of Partnerships following the EchiTab model:
Papua New Guinea: Gutiérrez JM. Understanding and confronting snakebite envenoming: The harvest of cooperation. Toxicon. 2016. DOI: 10.1016/j.toxicon.2015.11.013
Sri Lanka: Sri Lanka's antivenom leap forward https://www.aljazeera.com/indepth/features/2017/02/sri-lanka-anti-venom-leap-170205103054069.html
- Submitting institution
- Liverpool School of Tropical Medicine
- Unit of assessment
- 1 - Clinical Medicine
- Summary impact type
- Health
- Is this case study continued from a case study submitted in 2014?
- No
1. Summary of the impact
Malaria in pregnancy is a leading cause of adverse pregnancy outcomes. Research led by LSTM contributed directly to improved World Health Organisation (WHO) and endemic country policies and practices for the treatment and prevention of malaria in pregnancy in different transmission and drug resistance strata in sub-Saharan Africa and Asia-Pacific. Specifically, ministries of health in 36 African nations are now implementing a more effective malaria prevention strategy improving the outcome of approximately 32,000,000 pregnancies at risk annually and thereby the lives of mothers and their infants. Furthermore, following our studies on the safety of artemisinin-based combination therapy, regulatory authorities and WHO pre-qualification have updated drug labels for use in pregnancy.
2. Underpinning research
Between 2007 and 2017, ter Kuile (Head) and Hill (Project Manager) led the MiP (Malaria in Pregnancy) Consortium, a global network of 40 research institutions, that aimed to evaluate new strategies for the control of malaria in pregnancy to inform policy and improve the lives of mothers and their infants.
WHO recommends intermittent preventive treatment (IPTp) with sulphadoxine-pyrimethamine (SP) for the prevention of malaria in pregnancy in Africa. IPTp comprises a treatment dose of an antimalarial given presumptively at each scheduled antenatal care (ANC) visit, alongside case management and use of insecticide-treated nets in malaria-endemic countries. These strategies have come under threat due to increasing drug resistance and suboptimal coverage of these interventions.
Treatment (global)
Since 2006, WHO has recommended 3-day regimens with artemisinin-based combination therapies (ACTs) as first-line treatment for uncomplicated malaria in the 2nd and 3rd trimesters of pregnancy. Due to lack of safety data, ACTs are not recommended for 1st trimester treatment unless no other suitable antimalarials are available. Treatment recommended in the first trimester involves 7-day quinine regimens, which are badly tolerated and associated with poor adherence and therefore high rates of treatment failure. In a systematic review of health provider practices and access to case management of malaria during pregnancy (2014), Hill found widespread substandard practices for the case-management of pregnant women with the continued use of obsolete antimalarials in the 2nd and 3rd trimester that are no longer recommended by WHO and use of ACTs in the 1st trimester [1]. This led Dellicour and ter Kuile to undertake the largest individual participant data meta-analysis on the safety of artemisinin antimalarials in the 1st trimester involving 717 well-documented artemisinin and 947 quinine exposures from Asia and Africa. This showed that, contrary to data from animal models, artemisinin exposure in early pregnancy does not increase the risk of pregnancy loss or congenital anomalies compared to quinine and can be more effective at reducing pregnancy loss [2] as low adherence to the 7-day quinine regime results in a 4-fold higher risk of treatment failure.
Prevention (Africa)
Until 2012, IPTp with SP comprised at least two doses given in the 2nd and 3rd trimester. ter Kuile’s 2013 meta-analysis of IPTp trials showed that three or more doses of SP was well tolerated and safe and far more effective than the 2-dose regimen resulting in 49% (95% CI 32-62) and 20% (6-31) greater reductions in placental malaria and low birth weight, respectively [3]. However, the IPTp strategy is under threat due to increasing resistance to SP. ter Kuile and US Center for Disease Control colleagues showed that the efficacy of SP to clear existing malaria infections or prevent new ones is severely compromised in areas with high SP resistance. In a 2019 meta-analysis involving approximately 100,000 births, van Eijk and ter Kuile showed that alternative strategies are urgently needed in areas where over 37% of parasites carry the highly resistant sextuple-mutant Pfdhps-A581G-containing genotype [4].
In two trials of potential alternatives to IPTp-SP coordinated by ter Kuile (between 2011 and 2016), intermittent screening and treatment strategies (where only women testing positive for malaria are treated) was found not to be a suitable alternative at current levels of rapid diagnostic test (RDT) sensitivity [5,6]. By contrast, IPTp with the antimalarial dihydroartemisinin-piperaquine resulted in a decrease of 68% (44-82) in malaria infections during pregnancy, and a 59% (1-83) reduction in spontaneous miscarriage or stillbirth compared to IPTp-SP. This was the first of eight IPTp trials with dihydroartemisinin-piperaquine which is now considered the most promising alternative to replace SP for IPTp in high SP resistance areas [5]. By November 2020, three trials had been completed (the first of which was led by LSTM), and a further 5 are ongoing (the largest of which is led by LSTM). Results are expected in Q4 2021.
Prevention (Asia)
In the first trial of its kind in Asia, Ahmed and ter Kuile showed that malaria infection decreased by 41% (17-58) with monthly IPTp with dihydroartemisinin-piperaquine and is a promising alternative to the current policy of screening pregnant women for malaria at their first antenatal care visit in areas of moderate-to-high transmission in the Asia-Pacific region [6].
3. References to the research
Hill J, D'Mello-Guyett L, Hoyt J, van Eijk AM, ter Kuile FO, Webster J. Women's access and provider practices for the case management of malaria during pregnancy: a systematic review and meta-analysis. PLoS Med. 2014. DOI: 10.1371/journal.pmed.1001688
Dellicour S, Sevene E, McGready R, Tinto H, Mosha D, Manyando C, Rulisa S, Desai M, Ouma P, Oneko M, Vala A, Rupérez M, Macete E, Menéndez C, Nakanabo-Diallo S, Kazienga A, Valéa I, Calip G, Augusto O, Genton B, Njunju EM, Moore KA, d'Alessandro U, Nosten F, ter Kuile F, Stergachis A. First-trimester artemisinin derivatives and quinine treatments and the risk of adverse pregnancy outcomes in Africa and Asia: A meta-analysis of observational studies. PLoS Med. 2017. DOI: 10.1371/journal.pmed.1002290
Kayentao K, Garner P, van Eijk AM, Naidoo I, Roper C, Mulokozi A, MacArthur JR, Luntamo M, Ashorn P, Doumbo OK, ter Kuile FO. Intermittent preventive therapy for malaria during pregnancy using 2 vs 3 or more doses of sulfadoxine-pyrimethamine and risk of low birth weight in Africa: systematic review and meta-analysis. JAMA. 2013. DOI: 10.1001/jama.2012.216231
van Eijk AM, Larsen DA, Kayentao K, Koshy G, Slaughter DEC, Roper C, Okell LC, Desai M, Gutman J, Khairallah C, Rogerson SJ, Hopkins Sibley C, Meshnick SR, Taylor SM, ter Kuile FO. Effect of Plasmodium falciparum sulfadoxine-pyrimethamine resistance on the effectiveness of intermittent preventive therapy for malaria in pregnancy in Africa: a systematic review and meta-analysis. Lancet Infect Dis. 2019. DOI: 10.1016/S1473-3099(18)30732-1
Desai M, Gutman J, L'lanziva A, Otieno K, Juma E, Kariuki S, Ouma P, Were V, Laserson K, Katana A, Williamson J, ter Kuile FO. Intermittent screening and treatment or intermittent preventive treatment with dihydroartemisinin-piperaquine versus intermittent preventive treatment with sulfadoxine-pyrimethamine for the control of malaria during pregnancy in western Kenya: an open-label, three-group, randomised controlled superiority trial. Lancet. 2015. DOI: 10.1016/S0140-6736(15)00310-4
Ahmed R, Poespoprodjo JR, Syafruddin D, Khairallah C, Pace C, Lukito T, Maratina SS, Asih PBS, Santana-Morales MA, Adams ER, Unwin VT, Williams CT, Chen T, Smedley J, Wang D, Faragher B, Price RN, ter Kuile FO. Efficacy and safety of intermittent preventive treatment and intermittent screening and treatment versus single screening and treatment with dihydroartemisinin-piperaquine for the control of malaria in pregnancy in Indonesia: a cluster-randomised, open-label, superiority trial. Lancet Infect Dis. 2019. DOI: https://doi.org/10.1016/S1473-3099(19)30156-2
4. Details of the impact
Prof ter Kuile and Dr Hill applied a systematic approach to ensure the translation of research into policy by working with WHO to convene four consecutive Evidence Review Group meetings on malaria in pregnancy (between 2012 and 2017), which reviewed results from research led by LSTM and other partners and made recommendations both to WHO’s Malaria Policy Advisory Committee (MPAC) [1] and regulators (see below). Dissemination and technical support activities targeting policymakers and practitioners were undertaken to support policy uptake in endemic countries, improving pregnancy outcomes of millions of women in malaria-endemic countries.
Update to drug labels of ACTs for treatment of malaria in pregnancy
The safety studies in 1st trimester pregnancy described above resulted in label changes by the United States Food and Drug Administration (FDA, August 2019), the Coordination Group for Mutual Recognition and Decentralised Procedures at the European Medicines Agency (September 2020), and the WHO pre-qualification team (March 2018) for the use of the ACT artemisinin-lumefantrine in pregnancy [2-4]. The label change requested by the FDA was to lift the restriction on the use of Coartem® in pregnancy, including in the 1st trimester, based on the results of the 2017 meta-analysis. These label changes affect the use of these drugs in pregnancy by clinical practitioners and enable national malaria programmes to improve the quality of case management in the 1st trimester by shifting to the more effective 3-day ACT regimens from the current 7-day quinine regimen, which is poorly tolerated and badly adhered to, leading to adverse pregnancy outcomes.
The results were also reviewed by WHO’s MPAC in 2015 (6) which recommended a change from quinine to artemisinin combination therapies (ACTs) as a first-line therapeutic option for uncomplicated malaria in the 1st trimester, which was subsequently endorsed by WHO’s Technical Expert Group on Malaria Chemotherapy in December 2017 [1]. As a result of the label changes by the Stringent Regulatory Authorities (SRAs), WHO is proceeding and has requested our group to update the meta-analysis with any new data in preparation for the development of an update to the WHO treatment guidelines in 2021. Independent of WHO, Indonesia was the first country (September 2019) to change its national policy for treatment of malaria in 1st trimester to the ACT, dihydroartemisinin-piperaquine [7], which was already the first line therapy for treatment in all population groups, including in the 2nd and 3rd trimester.
WHO’s updated policy on prevention of malaria in pregnancy in sub-Saharan Africa
IPTp with SP: Increasing resistance to SP in parts of Africa led malaria-endemic countries to place pressure on WHO to provide further guidance on whether to continue using IPTp with SP. The LSTM-led studies on the continued effectiveness of IPTp-SP on reducing low birth weight, even in areas with relatively high SP resistance (likely due to the non-malarial effects of SP), led WHO in September 2013 to recommend continued implementation of IPTp-SP in all endemic areas until alternative drugs become available [5].
IPTp with DP: Our research showing the efficacy of dihydroartemisinin-piperaquine, for use in areas with very high SP resistance, and related studies of its acceptability and feasibility presented to WHO’s MPAC in 2015 [6], led WHO to recommend further studies with IPTp-DP to provide definitive evidence for consideration of IPTp-DP for policy in 2021 [8].
Implementation of revised guidelines on prevention of malaria in pregnancy in Africa
Results from our key meta-analysis comparing 3-or-more doses vs the standard 2-dose regimen of IPTp with SP led WHO to revise its guidelines in 2012, and updated in 2014, from 2 doses to monthly doses of SP [9]. The IMPPACT project (between 2016 and 2019), led by Hill, supported the uptake of evidence from the MiP Consortium in African country-level policies and guidelines. In collaboration with the Roll Back Malaria (RBM) Partnership, and the West Africa Health Organisation (WAHO), the MiP Consortium results were disseminated to 21 African countries [10a]. The WAHO meeting in 2017 involving National Malaria Control Programme Managers, M&E Officers, and Reproductive Health Managers from the 15 Economic Community of West African States (ECOWAS) member States, WHO, UNICEF, RBM, research organisations and the private sector, took place in the context of the preparation of country Global Fund applications between 2018 and 2020 [10b]. Countries reached a regional consensus on the implementation of malaria control activities, including the monthly IPTp-SP policy, articulated in the ECOWAS Regional Strategic Plan for Malaria Control and Elimination. WHO’s 2019 World Malaria Report reported that the updated policy had been adopted in 36 endemic countries in Africa [11]. As a result, annually, 32,000,000 pregnancies in Africa continue to benefit from this life-saving strategy for pregnant women annually.
National prevention policy uptake in Asia-Pacific
Results from the prevention trial with dihydroartemisinin-piperaquine (DP) and nested acceptability, feasibility and cost effectiveness studies in Indonesia were discussed with more than 110 ministries of health representatives from 18 countries in the Asia/Pacific region at a dissemination meeting in collaboration with WHO-WPRO and WHO-SEARO regional offices in 2017 [10c]. In 2019, as a direct result of these studies, the Indonesian Ministry of Health requested support from Hill and ter Kuile to evaluate the pilot implementation of IPTp-DP in Papua-Indonesia, and an LSTM-led study funded by the Medical Research Council (MRC) is ongoing.
5. Sources to corroborate the impact
Statement letter from the office of the Director of the Global Malaria Programme at the WHO, confirming WHO ERG and TEG meeting recommendations.
WHO prequalification medicines: MA122 - Artemether/Lumefantrine - 80mg/480mg - Tablet - Cipla Ltd - India. Part 4 - WHO-PQ recommended summary of product characteristics* March 2018 https://extranet.who.int/prequal/sites/default/files/MA122part4v1.pdf
FDA HIGHLIGHTS OF PRESCRIBING INFORMATION. COARTEM® (artemether and lumefantrine) tablets, for oral use. Initial U.S. Approval: 2009/Revised 8/2019 https://www.accessdata.fda.gov/drugsatfda_docs/label/2019/022268s021lbl.pdf
EMA approved and regulated prescribing and patient information for licensed medicines: Riamet 20 mg/120 mg Tablets. https://www.medicines.org.uk/emc/product/1628#CONTRAINDICATIONS
Malaria Policy Advisory Committee Meeting 11-13 September 2013, WHO Evidence Review Group on (IPT) of malaria in pregnancy: Draft Recommendations on Intermittent Preventive Treatment in Pregnancy (IPTp). http://www.who.int/malaria/mpac/mpac_sep13_erg_ipt_malaria_pregnancy_report.pdf
WHO Malaria Policy Advisory Committee (2016). "Malaria Policy Advisory Committee to the WHO: conclusions and recommendations of the eighth biannual meeting (September 2015)." Malar J 15(1): 117. http://www.ncbi.nlm.nih.gov/pubmed/26911803
Minister of Health, Republic of Indonesia. National Guidelines for Medical Services Malaria Management, documenting policy for treatment with DP of malaria in all trimesters of pregnancy. No. HK.01.07/Menkes / 556/2019. (Page 27)
World Health Organization. Intermittent screening and treatment in pregnancy and the safety of ACTs in the first trimester, November 2015, recommendations (WHO/HTM/GMP/2015.9). 2015. http://www.who.int/malaria/publications/atoz/istp-and-act-in-pregnancy.pdf?ua=1
WHO policy brief for the implementation of intermittent preventive treatment of malaria in pregnancy using sulfadoxine-pyrimethamine (IPTp-SP). WHO/HTM/GMP/2014.4. April 2013 (Revised January 2014). https://www.who.int/malaria/publications/atoz/iptp\-sp\-updated\-policy\-brief\-24jan2014.pdf?ua=1
(a) IMPPACT regional meeting report, Nairobi 2016 Research on the treatment and prevention of malaria in pregnancy in sub-Saharan Africa: East Africa Regional Meeting, 11-12 July 2016 - Fairview Hotel - Nairobi, Kenya. https://www.mip-consortium.org/sites/mip/files/upload/FinalMeetingReportPDF.pdf (b) IMPPACT regional meeting report, Togo 2017. Malaria in Pregnancy Consortium session on malaria in pregnancy (MiP) at the ECOWAS National Control Malaria Managers’ Review Meeting organized by the West Africa Health Organization (WAHO), 4-7th April 2017 in Lomé, Togo. https://www.mip-consortium.org/sites/mip/files/upload/West%20Africa%20Research%20Meeting%20Report%20v4_Publisher_0.pdf (c) APMEN Vivax Working Group Meeting report 9-11 October 2017, Bali. http://apmen.org/apmen/Working%20Groups/Vivax%20Working%20Group/APMEN%20MIP%20report%20%20Day%202%20VxWG%20meeting.pdf
World Malaria Report 2019 p.50 https://www.who.int/publications/i/item/9789241565721"
- Submitting institution
- Liverpool School of Tropical Medicine
- Unit of assessment
- 1 - Clinical Medicine
- Summary impact type
- Health
- Is this case study continued from a case study submitted in 2014?
- No
1. Summary of the impact
Gambian Human African trypanosomiasis (g-HAT), commonly called sleeping sickness, is a fatal disease caused by trypanosomes transmitted by tsetse flies.
World Health Organization (WHO) leads a global programme to eliminate g-HAT as a public health problem by 2020. International research led by the Liverpool School of Tropical Medicine (LSTM) produced ‘Tiny Targets’, a simple method of tsetse control. By 2020, Tiny Targets protected approximately 1,800,000 people in the 5 countries where most (90%) cases of g-HAT occur. Vector control has contributed to WHO achieving its goal of reducing the number of cases of gHAT reported globally to less than 2,000 cases/year by 2020; between 2017 and 2019, the number of new cases reported annually ranged between 1,409 and 864 compared to 10,466 and 2,110 for the previous decade (between 2007 and 2016).
2. Underpinning research
Most (more than 95%) cases of Human African Trypanosomiasis (HAT) occur in Central and West Africa and are caused by Trypanosoma brucei gambiense (Gambian HAT, g-HAT) transmitted by riverine species of tsetse. There are no vaccines or prophylactic drugs effective against g-HAT and disease management has relied almost exclusively on case detection and treatment. Achieving high coverage (more than 70% cases detected) of the population is difficult in the remote settings where g-HAT occurs. Between 30% and 90% of cases are never detected, and without treatment, patients inevitably die. In addition, the standard treatment for advanced (second-stage) disease, nifurtimox-eflornithine combination therapy (NECT), is complex to administer (e.g. 14 intravenous infusions over a fortnight) in the remote settings where g-HAT commonly occurs. Vector control offers the only means of protecting people from infection, but the standard methods of control were not cost-effective for the vectors of T. b. gambiense. Accordingly, Lehane initiated an international programme to develop new, low-cost methods of tsetse control, as part of a global effort, led by WHO, to eliminate g-HAT as a public health problem by 2020 and achieve complete interruption of transmission by 2030.
Six academic staff from LSTM led a multinational team of vector biologists (LSTM, University of Greenwich, Institut de Recherche pour le Développement (IRD), France) and chemical ecologists (Rothamsted Research). The team worked in partnership with scientists from some of the most affected countries (Burkina Faso (Centre international de recherche-développement sur l'elevage en zone subhumide (CIRDES)), Cote d’Ivoire (Institut Pierre Richer(IPR)), Kenya (International Centre of Insect Physiology and Ecology (ICIPE)) and Democratic Republic of Congo (Labovet)), to develop cost-effective methods of controlling tsetse vectors of pathogenic trypanosomes in sub-Saharan Africa. We first carried out field-based analyses of the behavioural responses of riverine tsetse to the visual and olfactory cues produced by hosts (between 2007 and 2011). LSTM led the research programme (PI= Lehane) and LSTM vector biologists worked in Africa with national researchers from each country.
Our work led to the discovery that riverine tsetse are highly responsive to small (25cm2) blue-coloured targets. These initial findings applied to all the most important vectors of T. b. gambiense [1], suggesting that deploying small insecticide-treated targets in the riverine habitats where tsetse concentrate could provide a cost-effective means of vector control. Working with an industrial partner (Vestergaard), we developed ‘Tiny Targets’, small (25cm x 25cm) panels of blue polyester flanked by a panel of insecticide-impregnated netting. Results from field trials of Tiny Targets conducted in Kenya, Uganda and Guinea between 2012 and 2015 showed that the targets dramatically reduced densities of tsetse by 70% to more than 90% [2,3,4], and the incidence of g-HAT by more than 90% [3, 4] For example, in the Mandoul focus of Chad, Tiny Targets were estimated to have contributed 70.4% (95% CI: 51–95%) of the reduction in reported cases between 2014 (90 cases) and 2015 (47 cases) [4]. Economic analyses of the trials conducted in Uganda [5] and Chad [6] showed that the cost of tsetse control was reduced to less than USD100/km2, representing a more than 80% reduction in costs compared to standard methods of tsetse control. More than 80% of g-HAT cases occur in Democratic Republic of Congo (DRC), and epidemiological models of g-HAT in high-endemicity foci suggested that the existing strategy of detecting and treating cases only would delay achieving the 2030 goal by 91- 206 years (2121 and 2236) compared to the year 2024 if Tiny Targets were also used [7]. The development of a cost-effective and logistically simple method of tsetse control led to a shift in the strategies of national programmes and the WHO for eliminating g-HAT, with vector control becoming a named tool that should be combined with case detection and treatment.
3. References to the research
Esterhuizen J, Rayaisse JB, Tirados I, Mpiana S, Solano P, Vale GA , Lehane MJ, Torr SJ. Improving the cost-effectiveness of visual devices for the control of riverine tsetse flies, the major vectors of human African trypanosomiasis. PLoS Negl Trop Dis. 2011. DOI: 10.1371/journal.pntd.0001257
Tirados I, Esterhuizen J, Kovacic V, Mangwiro TN, Vale GA, Hastings I, Solano P, Lehane MJ, Torr SJ. Tsetse Control and Gambian Sleeping Sickness; Implications for Control Strategy. PLoS Negl Trop Dis. 2015. DOI: 10.1371/journal.pntd.0003822
Courtin F, Camara M, Rayaisse JB, Kagbadouno M, Dama E, Camara O, Traoré IS, Rouamba J, Peylhard M, Somda MB, Leno M, Lehane MJ, Torr SJ, Solano P, Jamonneau V, Bucheton B. Reducing Human-Tsetse Contact Significantly Enhances the Efficacy of Sleeping Sickness Active Screening Campaigns: A Promising Result in the Context of Elimination. PLoS Negl Trop Dis. 2015. DOI: 10.1371/journal.pntd.0003727
Mahamat MH, Peka M, Rayaisse JB, Rock KS, Toko MA, Darnas J, Brahim GM, Alkatib AB, Yoni W, Tirados I, Courtin F, Brand SPC, Nersy C, Alfaroukh IO, Torr SJ, Lehane MJ, Solano P. Adding tsetse control to medical activities contributes to decreasing transmission of sleeping sickness in the Mandoul focus (Chad). PLoS Negl Trop Dis. 2017. DOI: 10.1371/journal.pntd.0005792
Shaw AP, Tirados I, Mangwiro CT, Esterhuizen J, Lehane MJ, Torr SJ, Kovacic V. Costs of using "tiny targets" to control Glossina fuscipes fuscipes, a vector of gambiense sleeping sickness in Arua District of Uganda. PLoS Negl Trop Dis. 2015. DOI: 10.1371/journal.pntd.0003624
Rayaisse JB, Courtin F, Mahamat MH, Chérif M, Yoni W, Gadjibet NMO, Peka M, Solano P, Torr SJ, Shaw APM. Delivering 'tiny targets' in a remote region of southern Chad: a cost analysis of tsetse control in the Mandoul sleeping sickness focus. Parasit Vectors. 2020. DOI: 10.1186/s13071-020-04286-w
Rock KS, Torr SJ, Lumbala C, Keeling MJ. Predicting the Impact of Intervention Strategies for Sleeping Sickness in Two High-Endemicity Health Zones of the Democratic Republic of Congo. PLoS Negl Trop Dis. 2017. DOI: 10.1371/journal.pntd.0005162
4. Details of the impact
Tiny Targets protected approximately 1,800,000 people in the 5 countries where 89.6% (34653 of the total 38668 cases) of all g-HAT cases were reported in the last decade (between 2010 and 2019) [1]. Evidence of the impact of Tiny Targets on g-HAT across a range of epidemiological settings led to tsetse control being included in national and global strategies to eliminate g-HAT as a public health problem.
Impact on populations at risk of g-HAT
Following successful trials conducted between 2011 and 2013 (reference 3 & 4 in Section 3), use of Tiny Targets was scaled up in in Uganda and Guinea to cover all g-HAT foci by 2020. Tiny Targets were also adopted by national HAT control programmes in Chad (2014), DRC (2015) and Côte d’Ivoire (2016). The largest deployment was in DRC where the Tryp-Elim programme, funded by the Bill & Melinda Gates Foundation (BMGF) supported the annual deployment of 43,000 Tiny Targets and by 2019 approximately 300,000 people across 5,300km2 were protected from g-HAT. A second BMGF-funded programme (“Trypa-NO!”) has supported the scale-up of Tiny Targets in additional countries and, since 2017, a total of approximately 1,500,000 people have been protected in Guinea (1,900km2, 200,000 people protected), Chad (960km2, 80,000 people), Uganda (3,900km2, 1,139,000 people) and Ivory Coast (250km2, 170,000 people) [2].
The scale-up of Tiny Targets has had a dramatic impact on disease incidence. In addition to our own data from Chad (reference 4 in Section 2), others have shown that targets reduced the annual incidence of g-HAT in the Boffa focus in Guinea by over 90% (14/1279=1.09% without vector control vs 2/2777=0.07% with vector control) [3]. Continued deployment of Tiny Targets in the Mandoul and Maro foci of Chad has contributed to a decrease from 95 (2014) to 16 (2019) in the total number of cases reported nationally. The development of a tsetse control method that is affordable and practicable for local communities (Tiny Targets can be distributed using bikes and dugout canoes rather than lorries) has strengthened the resilience of control programmes. In Guinea, the Ebola crisis interrupted programmes to screen-and-treat populations for g-HAT and in the Boffa focus, prevalence of g-HAT increased in areas where Tiny Targets were absent but remained low in areas where the local community deployed Tiny Targets [3]. Similarly, deployment of Tiny Targets continued in all countries during the Covid-19 pandemic.
Impact on national g-HAT strategies
Economic [4] and epidemiological models [5] indicated that the addition of tsetse control using Tiny Targets to the standard screen-and-treat strategies accelerates progress towards the WHO elimination goals. For instance, analyses of progress towards elimination goals in DRC suggested that 37 of 43 Health Zones in DRC health zones will require vector control to meet the 2030 elimination goal [5]. Since starting with a pilot project in 2015, deploying Tiny Targets in Yasa Bonga Health Zone, which demonstrated >85% reduction in tsetse numbers [6], activities have now expanded to six further Health Zones (Masi Manimba, Kikongo, Bandundu, Kwamouth, Bulungu and Bolobo) as part of the national strategy to meet the 2030 elimination goal. Theoretical and empirical evidence of the impact of Tiny Targets led the national programmes of Chad (PNLTHA), Cote d’Ivoire (IPR), DRC (PNLTHA), Guinea (PNLTHA) and Uganda (COCTU) to adopt Tiny Targets as part of their national strategies to control g-HAT. Consequently, use of Tiny Targets has increased from 10,000 deployed in 2013 to 100,000 deployed annually in 2020. Over the same period, the aggregate area over which Tiny Targets were deployed increased from approximately 750km2 to 13,000km2 to protect approximately 1,800,000 people. Recognising its philanthropic and ethical responsibilities, Vestergaard, the manufacturer of Tiny Targets, announced that they will donate all Tiny Targets free from 2020 onwards [7].
Global policy
Our empirical and theoretical evidence combined with outreach and engagement activities with normative bodies led to a 2014 WHO recommendation that tsetse control should be an integral part of the global programme to eliminate g-HAT [8]. Policy makers were engaged through regular visits by LSTM researchers to the headquarters of WHO (Geneva), African Union (Addis Ababa) and national ministries of health, as well as presentations at international meetings organised by WHO [7], the AU’s Pan-African Tsetse and Trypanosomiasis Eradication Campaign (PATTEC) and International Scientific Council for Trypanosomiasis Research and Control (ISCTRC), meetings with the leaders of national programmes [9] and engagement with print and broadcast media [10].
the inclusion of vector control tools has ensured that the WHO’s program to eliminate g-HAT as a public health problem by 2020 is on track with, for example, the specific goal of less than 2,000 new cases reported annually being achieved from 2017 (1,409 cases) onwards (2018, 953 cases; 2019, 864 cases). These low numbers contrast with the decade between 2005 and 2014, prior to the inclusion of vector control, when the number of new cases reported globally ranged between 3,679 (2014) and 15,624 (2005) annually [1].
5. Sources to corroborate the impact
WHO data showing decline in reported cases of HAT and relative importance of DRC, Chad and Guinea for global burden. http://apps.who.int/neglected_diseases/ntddata/hat/hat.html
Manuscript describing scale up of Tiny Targets Ndung'u JM, Boulangé A, Picado A, Mugenyi A, Mortensen A, Hope A, Mollo BG, Bucheton B, Wamboga C, Waiswa C, Kaba D, Matovu E, Courtin F, Garrod G, Gimonneau G, Bingham GV, Hassane HM, Tirados I, Saldanha I, Kabore J, Rayaisse JB, Bart JM, Lingley J, Esterhuizen J, Longbottom J, Pulford J, Kouakou L, Sanogo L, Cunningham L, Camara M, Koffi M, Stanton M, Lehane M, Kagbadouno MS, Camara O, Bessell P, Mallaye P, Solano P, Selby R, Dunkley S, Torr S, Biéler S, Lejon V, Jamonneau V, Yoni W, Katz Z. Trypa-NO! contributes to the elimination of gambiense human African trypanosomiasis by combining tsetse control with "screen, diagnose and treat" using innovative tools and strategies. PLoS Negl Trop Dis. 2020. DOI: 10.1371/journal.pntd.0008738
Tiny Targets controlling HAT during the Ebola crisis in Guinea. Kagbadouno, M.S., Camara, O., Camara, M., Ilboudo, H., Camara, M.L., Rayaisse, J.-B., Diaby, A., Taore, B., Leno, M., Courtin, F., Jamonneau, V., Solano, P., Bucheton, B., 2018. Ebola outbreak brings to light an unforeseen impact of tsetse control on sleeping sickness transmission in Guinea. bioRxiv, 202762. https://www.biorxiv.org/content/10.1101/202762v1.full.pdf
Economics models showing the cost-effectiveness of tsetse control using Tiny Targets. Sutherland CS, Stone CM, Steinmann P, Tanner M, Tediosi F. Seeing beyond 2020: an economic evaluation of contemporary and emerging strategies for elimination of Trypanosoma brucei gambiense. Lancet Glob Health. 2017. DOI: 10.1016/S2214-109X(16)30237-6
Epidemiological models showing predicted contribution of vector control to efforts against HAT in DRC. Huang C-I, Crump RE, Brown P, Spencer SEF, Mwamba Miaka E, Shampa C, et al. Shrinking the gHAT map: identifying target regions for enhanced control of gambiense human African trypanosomiasis in the Democratic Republic of Congo. medRxiv. https://www.medrxiv.org/content/10.1101/2020.07.03.20145847v1
Reduction in testse fly numbers following implementation of Tiny Targets. Tirados I, Hope A, Selby R, Mpembele F, Miaka EM, Boelaert M, Lehane MJ, Torr SJ, Stanton MC. Impact of tiny targets on Glossina fuscipes quanzensis, the primary vector of human African trypanosomiasis in the Democratic Republic of the Congo. PLoS Negl Trop Dis. 2020. DOI: 10.1371/journal.pntd.0008270
WHO website reporting donation of Tiny Targets by Vestergaard https://www.who.int/neglected_diseases/news/Global_resolve_to_end_NTDs_amid_unprecedented_progress/en/ and Letter from Vestergaard
WHO meeting showing support for vector control as part of elimination programme and specific mention of Tiny Targets. Barrett MP. The elimination of human African trypanosomiasis is in sight: Report from the third WHO stakeholders meeting on elimination of gambiense human African trypanosomiasis. PLoS Negl Trop Dis. 2018. https://doi.org/10.1371/journal.pntd.0006925
Government document reporting national strategy to control HAT in DRC using screen-and-treat integrated with use of Tiny Targets to control tsetse.
HAT Republique Democratique du Congo, Ministere de la Sante Programme National de Lutte contre la Trypanosomiase Humaine Africaine (PNLTHA-RDC). Declaration de la Politique de Lutte Contre la Trypanosomiases Humaine Africaines en Republique Democratique du Congo (December 2015)
- BBC website reporting use of Tiny Targets in Uganda
Tangled up in blue: A sticky end to sleeping sickness. BBC reports use of Tiny Targets in Uganda. Broadcast 28/6/2015. https://www.bbc.co.uk/news/av/health-33274658/tangled-up-in-blue-a-sticky-end-to-sleeping-sickness