21
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      The impact of clinical research activities on communities in rural Africa: the development of the Clinical Research Unit of Nanoro (CRUN) in Burkina Faso

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Background

          The opportunities for developing new drugs and vaccines for malaria control look brighter now than ten years ago. However, there are few places in sub-Saharan Africa with the necessary infrastructure and expertise to support such research in compliance to international standards of clinical research (ICH-GCP). The Clinical Research Unit of Nanoro (CRUN) was founded in 2008 to provide a much-needed GCP-compliant clinical trial platform for an imminent large-scale Phase 3 malaria vaccine trial. A dynamic approach was used that entailed developing the required infrastructure and human resources, while engaging local communities in the process as key stakeholders. This provided a better understanding and ownership of the research activities by the local population.

          Case description

          Within five years (2008–2013), the CRUN set up a fully and well-equipped GCP-compliant clinical trial research facility, which enabled to attract 25 grants. The research team grew from ten health workers prior to 2008 to 254 in 2013. A Health and Demographic Surveillance System (HDSS), which covers a total population of about 60,000 people in 24 villages was set up in the district. The local community contributed to the development of the facility through the leadership of the king and the mayor of Nanoro. As a result of their active advocacy, the government extended the national electrical grid to the new research center, and later to the entire village. This produced a positive impact on the community’s quality of life. The quality of health care improved substantially, due to the creation of more elaborate clinical laboratory services and the acquisition of state-of-the-art equipment.

          Conclusion

          Involving the community in the key steps of establishing the centre provided the foundation for what was to become the CRUN success story. This experience demonstrates that when clinical trials research sites are carefully developed and implemented, they can have a positive and powerful impact on local communities in resource-poor settings, well beyond the task of generating expected study data.

          Related collections

          Most cited references11

          • Record: found
          • Abstract: found
          • Article: not found

          From malaria control to eradication: The WHO perspective.

          Efforts to control malaria have been boosted in the past few years with increased international funding and greater political commitment. Consequently, the reported malaria burden is being reduced in a number of countries throughout the world, including in some countries in tropical Africa where the burden of malaria is greatest. These achievements have raised new hopes of eradicating malaria. This paper summarizes the outcomes of a World Health Organization's expert meeting on the feasibility of such a goal. Given the hindsight and experience of the Global Malaria Eradication Programme of the 1950s and 1960s, and current knowledge of the effectiveness of antimalarial tools and interventions, it would be feasible to effectively control malaria in all parts of the world and greatly reduce the enormous morbidity and mortality of malaria. It would also be entirely feasible to eliminate malaria from countries and regions where the intensity of transmission is low to moderate, and where health systems are strong. Elimination of malaria requires a re-orientation of control activity, moving away from a population-based coverage of interventions, to one based on a programme of effective surveillance and response. Sustained efforts will be required to prevent the resurgence of malaria from where it is eliminated. Eliminating malaria from countries where the intensity of transmission is high and stable such as in tropical Africa will require more potent tools and stronger health systems than are available today. When such countries have effectively reduced the burden of malaria, the achievements will need to be consolidated before a programme re-orientation towards malaria elimination is contemplated. Malaria control and elimination are under the constant threat of the parasite and vector mosquito developing resistance to medicines and insecticides, which are the cornerstones of current antimalarial interventions. The prospects of malaria eradication, therefore, rest heavily on the outcomes of research and development for new and improved tools. Malaria control and elimination are complementary objectives in the global fight against malaria.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: found
            Is Open Access

            Dihydroartemisinin-Piperaquine and Artemether-Lumefantrine for Treating Uncomplicated Malaria in African Children: A Randomised, Non-Inferiority Trial

            Background Artemisinin combination therapies (ACTs) are currently the preferred option for treating uncomplicated malaria. Dihydroartemisinin-piperaquine (DHA-PQP) is a promising fixed-dose ACT with limited information on its safety and efficacy in African children. Methodology/Principal Findings The non-inferiority of DHA-PQP versus artemether-lumefantrine (AL) in children 6–59 months old with uncomplicated P. falciparum malaria was tested in five African countries (Burkina Faso, Kenya, Mozambique, Uganda and Zambia). Patients were randomised (2∶1) to receive either DHA-PQP or AL. Non-inferiority was assessed using a margin of −5% for the lower limit of the one-sided 97.5% confidence interval on the treatment difference (DHA-PQP vs. AL) of the day 28 polymerase chain reaction (PCR) corrected cure rate. Efficacy analysis was performed in several populations, and two of them are presented here: intention-to-treat (ITT) and enlarged per-protocol (ePP). 1553 children were randomised, 1039 receiving DHA-PQP and 514 AL. The PCR-corrected day 28 cure rate was 90.4% (ITT) and 94.7% (ePP) in the DHA-PQP group, and 90.0% (ITT) and 95.3% (ePP) in the AL group. The lower limits of the one-sided 97.5% CI of the difference between the two treatments were −2.80% and −2.96%, in the ITT and ePP populations, respectively. In the ITT population, the Kaplan-Meier estimate of the proportion of new infections up to Day 42 was 13.55% (95% CI: 11.35%–15.76%) for DHA-PQP vs 24.00% (95% CI: 20.11%–27.88%) for AL (p<0.0001). Conclusions/Significance DHA-PQP is as efficacious as AL in treating uncomplicated malaria in African children from different endemicity settings, and shows a comparable safety profile. The occurrence of new infections within the 42-day follow up was significantly lower in the DHA-PQP group, indicating a longer post-treatment prophylactic effect. Trial Registration Controlled-trials.com ISRCTN16263443
              Bookmark
              • Record: found
              • Abstract: found
              • Article: found
              Is Open Access

              A Head-to-Head Comparison of Four Artemisinin-Based Combinations for Treating Uncomplicated Malaria in African Children: A Randomized Trial

              (2011)
              Introduction The burden of malaria has declined substantially in several areas of sub-Saharan Africa, particularly in the past 3–5 y [1]. Such a change has been attributed to a combination of factors [2], including large scale indoor residual spraying campaigns [3],[4], massive distribution of insecticide-treated bed nets [5], and the introduction of artemisinin-based combination treatments (ACTs) [6],[7]. The scale-up of the interventions has been possible thanks to the availability of more funding, especially from the Global Fund to Fight AIDS, Tuberculosis and Malaria [8], that has allowed an increasing number of countries to include ACTs in their national treatment guidelines as first and, in some cases, second-line treatments, and to the massive scale-up implementation of treatment programs [9]. In addition, increased funding for research, often through effective public–private partnerships [10], has resulted in the availability of several ACTs [11]. However, data to guide individual countries in choosing the most appropriate ACTs are limited. The World Health Organization (WHO), which recently produced revised guidelines for the treatment of malaria, states that the choice of ACT in a country or region should be based on the level of resistance to the medicine partnered to the artemisinin derivative in the combination [12]. However, up-to-date treatment efficacy data for the partner medicine to the artemisinin derivative are scarce. The WHO recommends five ACTs, namely artemether-lumefantrine (AL), amodiaquine-artesunate (ASAQ), mefloquine-artesunate, sulfadoxine-pyrimethamine-artesunate, and, most recently included, dihydroartemisinin-piperaquine (DHAPQ) [12]. Each of these combinations may have different advantages and disadvantages that vary according to a number of factors, including malaria endemicity, safety, tolerability, dosing, post-treatment prophylactic effect, resistance to the partner drug of the prevailing parasites in the area, and price. Accordingly, we carried out a head-to-head comparison of the safety and efficacy of several ACTs, with the aim of providing the information necessary to make an informed choice for the formulation of relevant national antimalarial treatment policies. The ACTs tested included three of those recommended by the WHO, namely AL, ASAQ, DHAPQ, and one that was under development, chlorproguanil-dapsone-artesunate (CD+A). AL was the first co-formulated ACT to become available and, together with ASAQ, is the most common ACT used in sub-Saharan Africa. DHAPQ has been recently submitted for registration to the European Medicines Agency under the orphan drug legislation [13] following two phase III trials that were carried out in Africa [14] and Asia [15]. DHAPQ is currently used as a recommended treatment only in Asia [16], and a formulation approved by a stringent drug regulatory authority, such as the European Medicine Agency, or pre-qualified by the WHO is not yet available. At the time our trial started, combining chlorproguanil-dapsone (Lapdap, GlaxoSmithKline) with artesunate (CD+A) was considered to be a promising combination. Lapdap was on the market until 2008, when the producer withdrew it following the results of several studies showing that it caused significant reductions in hemoglobin (Hb) levels in patients with glucose-6-phosphate dehydrogenase deficiency [17]. Therefore, the CD+A arm was stopped (because the drug was no longer in development owing to concerns over safety), and our trial continued with the other three ACTs under evaluation. Methods Study Design, Sites, and Concealment of Patient Allocation Between 9 July 2007 and 19 June 2009, a randomized, open-label, multicenter, non-inferiority clinical trial was carried out at 12 sites located in seven African countries (Nanoro, Burkina Faso; Fougamou and Lambaréné, Gabon; Afokang and Pamol, Nigeria; Mashesha and Rukara, Rwanda; Jinja, Tororo, and Mbarara, Uganda; Ndola, Zambia; and Manhiça, Mozambique). See protocol (Text S1) and amendments (Texts S3–S5), and CONSORT checklist (Text S2). Each site compared three of the four ACTs under investigation, ASAQ, DHAPQ, AL, or CD+A. The decision of which treatments to test at a given site was made by considering the current first-line treatments, the known antimalarial resistance profile, and local malaria endemicity (Table 1). Patients were individually randomized according to a 1∶1∶1 scheme, with six sites testing ASAQ versus DHAPQ versus AL, four testing DHAPQ versus CD+A versus AL, and two testing ASAQ versus CD+A versus DHAPQ (Table 1). A randomization list was produced for each recruiting site by the National Institute for Health Research Medicines for Children Research Network Clinical Trials Unit, University of Liverpool, UK, with each treatment allocation concealed in opaque sealed envelopes that were opened only after the patient's recruitment. 10.1371/journal.pmed.1001119.t001 Table 1 Study treatment to be tested by country. Country Sites Transmission (Entomological Inoculation Rate) Percent with Chloroquine Resistance Percent with Sulfadoxine-Pyrimethamine Resistance Study Treatments Burkina Faso Nanoro Seasonal, high (50–60)[46] 24 [46] 4 [46] ASAQ DHAPQ AL Gabon Fougamou, Lambaréné Perennial, high (50) 100 [47] 23 [48] ASAQ DHAPQ AL Nigeria Afokang, Pamol Perennial, high 45 [49] 30 [49] ASAQ DHAPQ AL Zambia Ndola Seasonal, mesoendemic High 19 (in adults) [50] ASAQ DHAPQ AL Rwanda Rukara Seasonal, high 40 [51] 36 [51] DHAPQ CD+A AL Rwanda Mashesha Seasonal, high 50 [51] 12 [51] DHAPQ CD+A AL Uganda Jinja Perennial, low (6) [34] 28 [52],[53] 49 [52],[53] DHAPQ CD+A AL Uganda Tororo Perennial (>563) [34] 45 [52],[53] 9–15 [52],[53] DHAPQ CD+A AL Mozambique Manhiça Perennial, mesoendemic [54] 78 [55] 22 [55] ASAQ CD+A DHAPQ Uganda Mbarara Mesoendemic 81 [56] 25 [56] ASAQ CD+A DHAPQ Entomological inoculation rate is infective bites/person/year. Children 6–59 mo old (12–59 mo old at sites where CD+A was used) attending the health facilities with suspected uncomplicated malaria were included in the study if they fulfilled all the following inclusion criteria: body weight >5 kg, microscopically confirmed Plasmodium falciparum mono-infection with asexual parasite densities between 2,000 and 200,000/µl, fever (axillary temperature ≥37.5°C) or history of fever in the preceding 24 h, and Hb ≥7.0 g/dl. Patients were not recruited if they met at least one of the following exclusion criteria: participation in any other investigational drug study during the previous 30 d; known hypersensitivity to the study drugs; severe malaria [18] or other danger signs, e.g., not able to drink or breast-feed, vomiting (more than twice in 24 h), recent history of convulsions (more than once in 24 h), unconscious state, or unable to sit or stand; severe malnutrition (weight for height 200,000/µl at day 0, no fever or history of fever and parasite density 90% and the trial aimed at showing non-inferiority at a 10% difference threshold. Non-inferiority was demonstrated for the three pair-wise comparisons involving DHAPQ, ASAQ, and AL at most sites, with a few exceptions where the individual sites' 95% CI cross the non-inferiority limit. In west Africa, ASAQ continues to have excellent efficacy, comparable to that of AL [27],[28], while in east Africa doubts about its use have been expressed [29]. Indeed, in a study carried out in Kampala, Uganda, ASAQ had a lower efficacy than AL, a result confirmed in subsequent years [30]. The superior efficacy of AL compared to ASAQ was explained by the presence of amodiaquine resistance in east Africa that may render this ACT increasingly less efficacious, similar to what has been observed for sulfadoxine-pyrimetamine-artesunate in east Africa [29]. In Tanzania, AL was significantly better than ASAQ, but this was an effectiveness study, i.e., the treatment administration was not supervised, and ASAQ was not co-formulated, two important factors that may have influenced the treatment outcome [31]. In our study, although ASAQ was tested mainly in west Africa, it also had excellent efficacy at sites located in eastern and southern Africa, namely Mbarara (Uganda), Ndola (Zambia), and Manhiça (Mozambique). However, the choice of the ACTs to test at the individual sites was influenced by their known drug resistance profile, i.e., ASAQ was not tested in sites with known high amodiaquine resistance. Therefore, although ASAQ is a possible option for some countries in east Africa, it should be not be deployed where amodiaquine resistance is known to be high. For the PCR-unadjusted efficacy at days 28 and 63, none of the pair-wise comparisons could show non-inferiority. Instead, DHAPQ was the best treatment, followed by ASAQ, AL, and then CD+A, which was consistently inferior to the three other ACTs. Considering that most of the recurrent infections were due to new infections and that the risk of re-infection depends on the activity of the non-artemisinin component, this result is largely expected. Indeed, piperaquine has the longer elimination half-life (about 23–28 d), followed by amodiaquine (3 wk), lumefantrine (3.2 d) [32], chlorproguanil (35 h), and dapsone (27 h) [33]. The length and efficacy of the post-treatment prophylaxis may also be influenced by the transmission intensity. In Tororo (Uganda), a site with very high entomological inoculation rates (>500 infective bites/person/year) [34], the difference in the cumulative risk of recurrent malaria between AL and DHAPQ decreased when the follow-up period was extended to 63 d, suggesting that piperaquine's long elimination half-life could do little against the overwhelming risk of recurrent malaria [35]. Nevertheless, in our study and at the same site, the risk of recurrent infections at day 63, though still extremely high, was lower in the DHAPQ than in the AL arm. When considering the forest plot comparing AL and DHAPQ, the OR tended to be lower at sites with the highest transmission intensity, suggesting that piperaquine's longer post-treatment prophylaxis still had an effect despite the high transmission. It should also be noted that places with an intensity of transmission as high as or higher than that of Tororo are not common, particularly in the current context of decreasing malaria burden in sub-Saharan Africa [1]. Though delaying the occurrence of a second clinical attack, via a long post-treatment prophylactic effect, represents an advantage at the individual level, the increased risk of selecting resistant parasites among the new infections should be considered. Such risk occurs during a specific period, the “window of selection,” whose opening and duration is proportional to the drug terminal elimination half-life [36]. Therefore, according to this model, such window would be shorter for lumefantrine (3–5 wk) than for piperaquine. Though this should not be a deterrent for the large-scale deployment of DHAPQ, setting up a reliable early warning system for the detection of resistance, possibly by both in vivo and in vitro tests, would be essential. In children treated with AL, gametocyte prevalence during follow-up and gametocyte carriage time were significantly lower than in children treated with either DHAPQ or ASAQ. The difference remained significant for gametocyte carriage time even when excluding patients with gametocytes at enrollment. Higher gametocyte carriage after treatment with DHAPQ, when compared to either AL or mefloquine-artesunate, has already been reported in some [14],[35],[37]–[39] but not all trials [40]. Similarly, gametocyte carriage was higher after treatment with ASAQ than with AL in some [29],[41] but not all studies [27]. The meaning of such differences in terms of transmission potential is unclear. Compared to molecular methods, microscopy detects only a small fraction of gametocyte carriers, both in individuals with asymptomatic infections [42] and in patients treated with an antimalarial [39]. Children with microscopically detectable gametocytes are more likely to be infectious but those with sub-microscopic gametocytes can also transmit, albeit less efficiently [42]. The gametocyte prevalence as determined by molecular methods has been observed to be higher in patients treated with DHAPQ than with AL [39]. However, about 60% of children treated with AL and without microscopically detectable gametocytes were infectious to mosquitoes, with little difference between treatments, though the probability of a mosquito becoming infected was significantly lower for the ACT (AL and sulfadoxine-pyrimetamine-artesunate) than for monotherapy or non-ACT combinations [43]. This indicates the difficulty of determining the transmission potential on the basis of the gametocyte carriage time as determined by microscopy, so that the differences in gametocyte carriage observed in our trial may not necessarily relate to a significantly different transmission potential. Considering that ACTs reduce the production of gametocytes by both decreasing the asexual reservoir and destroying a substantial proportion of immature, developing gametocytes, still sequestered in the microvasculature [44], and that parasite clearance was similar in the four study arms, the difference observed between the three ACTs may relate to their ability to clear almost mature forms not released in the blood stream yet. Hematological recovery up to day 28 post-treatment was similar for all ACTs tested except for CD+A, for which this was significantly slower, with a more marked Hb decrease up to day 7, confirming previous results [25]. In addition, in the CD+A group, anemia was diagnosed more frequently as AE, providing additional evidence for the higher risk of anemia for this treatment. Aside from anemia risk related to CD+A, all regimens were well tolerated. In conclusion, this is, to our knowledge, the largest head-to-head comparison of most of the currently available ACTs for falciparum malaria in sub-Saharan Africa. CD+A was suspended partway through the trial, leaving AL, ASAQ, and DHAPQ under investigation. These three ACTs showed excellent efficacy, up to day 63 post-treatment, but the risk of recurrent infections was significantly lower, even in areas of high transmission, for DHAPQ, followed by ASAQ, and then AL. Although the gametocyte carriage rate differed between regimens, with those treated with AL having the lowest carriage rate and those treated with ASAQ having the highest carriage rate, the meaning of these different carriage rates with relation to transmission potential is unclear. The possibility of adding a single dose of primaquine to any of these three ACTs, with the objective of further reducing gametocyte carriage, should be explored [41],[45]. AL and/or ASAQ are already included in the antimalarial drug policies of many sub-Saharan African countries. This study confirms that DHAPQ is a valid third option for the treatment of uncomplicated P. falciparum malaria, as its efficacy is excellent and comparable to the other ACTs, while its long post-treatment prophylaxis could be an additional advantage. Supporting Information Text S1 Study protocol. (PDF) Click here for additional data file. Text S2 CONSORT checklist. (PDF) Click here for additional data file. Text S3 Protocol amendment 1. (PDF) Click here for additional data file. Text S4 Protocol amendment 2. (PDF) Click here for additional data file. Text S5 Protocol amendment 3. (PDF) Click here for additional data file.
                Bookmark

                Author and article information

                Contributors
                Journal
                Malar J
                Malar. J
                Malaria Journal
                BioMed Central
                1475-2875
                2014
                22 March 2014
                : 13
                : 113
                Affiliations
                [1 ]Unité de Recherche Paludisme et Maladies Tropicales Négligées, Centre Muraz, Bobo-Dioulasso, Burkina Faso
                [2 ]Institut de Recherche en Sciences de la Santé/Direction Régionale (IRSS/DRO), Bobo-Dioulasso, Burkina Faso
                [3 ]Clinical Research Unit of Nanoro (IRSS-CRUN), Nanoro, Burkina Faso
                [4 ]Institut Supérieur des Sciences de la Santé (INSSA), Bobo-Dioulasso, Burkina Faso
                [5 ]INDEPTH-Network, Accra, Ghana
                Article
                1475-2875-13-113
                10.1186/1475-2875-13-113
                3994337
                24655351
                3c72591d-35f3-4d96-805d-34488fed98f9
                Copyright © 2014 Tinto et al.; licensee BioMed Central Ltd.

                This is an Open Access article distributed under the terms of the Creative Commons Attribution License ( http://creativecommons.org/licenses/by/2.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly credited. The Creative Commons Public Domain Dedication waiver ( http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.

                History
                : 5 December 2013
                : 10 March 2014
                Categories
                Case Study

                Infectious disease & Microbiology
                Infectious disease & Microbiology

                Comments

                Comment on this article

                scite_

                Similar content166

                Cited by15

                Most referenced authors452