44 results on '"Bougouma EC"'
Search Results
2. Insights into malaria susceptibility using genome-wide data on 17,000 individuals from Africa, Asia and Oceania
- Author
-
Band, G, Le, QS, Clarke, GM, Kivinen, K, Hubbart, C, Jeffreys, AE, Rowlands, K, Leffler, EM, Jallow, M, Conway, DJ, Sisay-Joof, F, Sirugo, G, d'Alessandro, U, Toure, OB, Thera, MA, Konate, S, Sissoko, S, Mangano, VD, Bougouma, EC, Sirima, SB, Amenga-Etego, LN, Ghansah, AK, Hodgson, AVO, Wilson, MD, Enimil, A, Ansong, D, Evans, J, Ademola, SA, Apinjoh, TO, Ndila, CM, Manjurano, A, Drakeley, C, Reyburn, H, Nguyen, HP, Nguyen, TNQ, Cao, QT, Tran, TH, Teo, YY, Manning, L, Laman, M, Michon, P, Karunajeewa, H, Siba, P, Allen, S, Allen, A, Bahlo, M, Davis, TME, Simpson, V, Shelton, J, Spencer, CCA, Busby, GBJ, Kerasidou, A, Drury, E, Stalker, J, Dilthey, A, Mentzer, AJ, McVean, G, Bojang, KA, Doumbo, O, Modiano, D, Koram, KA, Agbenyega, T, Amodu, OK, Achidi, E, Williams, TN, Marsh, K, Riley, EM, Molyneux, M, Taylor, T, Dunstan, SJ, Farrar, J, Mueller, I, Rockett, KA, Kwiatkowski, DP, Band, G, Le, QS, Clarke, GM, Kivinen, K, Hubbart, C, Jeffreys, AE, Rowlands, K, Leffler, EM, Jallow, M, Conway, DJ, Sisay-Joof, F, Sirugo, G, d'Alessandro, U, Toure, OB, Thera, MA, Konate, S, Sissoko, S, Mangano, VD, Bougouma, EC, Sirima, SB, Amenga-Etego, LN, Ghansah, AK, Hodgson, AVO, Wilson, MD, Enimil, A, Ansong, D, Evans, J, Ademola, SA, Apinjoh, TO, Ndila, CM, Manjurano, A, Drakeley, C, Reyburn, H, Nguyen, HP, Nguyen, TNQ, Cao, QT, Tran, TH, Teo, YY, Manning, L, Laman, M, Michon, P, Karunajeewa, H, Siba, P, Allen, S, Allen, A, Bahlo, M, Davis, TME, Simpson, V, Shelton, J, Spencer, CCA, Busby, GBJ, Kerasidou, A, Drury, E, Stalker, J, Dilthey, A, Mentzer, AJ, McVean, G, Bojang, KA, Doumbo, O, Modiano, D, Koram, KA, Agbenyega, T, Amodu, OK, Achidi, E, Williams, TN, Marsh, K, Riley, EM, Molyneux, M, Taylor, T, Dunstan, SJ, Farrar, J, Mueller, I, Rockett, KA, and Kwiatkowski, DP
- Abstract
The human genetic factors that affect resistance to infectious disease are poorly understood. Here we report a genome-wide association study in 17,000 severe malaria cases and population controls from 11 countries, informed by sequencing of family trios and by direct typing of candidate loci in an additional 15,000 samples. We identify five replicable associations with genome-wide levels of evidence including a newly implicated variant on chromosome 6. Jointly, these variants account for around one-tenth of the heritability of severe malaria, which we estimate as ~23% using genome-wide genotypes. We interrogate available functional data and discover an erythroid-specific transcription start site underlying the known association in ATP2B4, but are unable to identify a likely causal mechanism at the chromosome 6 locus. Previously reported HLA associations do not replicate in these samples. This large dataset will provide a foundation for further research on thegenetic determinants of malaria resistance in diverse populations.
- Published
- 2019
3. Characterisation of the opposing effects of G6PD deficiency on cerebral malaria and severe malarial anaemia
- Author
-
Clarke, GM, Rockett, K, Kivinen, K, Hubbart, C, Jeffreys, AE, Rowlands, K, Jallow, M, Conway, DJ, Bojang, KA, Pinder, M, Usen, S, Sisay-Joof, F, Sirugo, G, Toure, O, Thera, MA, Konate, S, Sissoko, S, Niangaly, A, Poudiougou, B, Mangano, VD, Bougouma, EC, Sirima, SB, Modioano, D, Amenga-Etego, LN, Ghansah, A, Koram, KA, Wilson, MD, Enimil, A, Evans, J, Amodu, OK, Olaniyan, S, Apinjoh, T, Mugri, R, Ndi, A, Ndila, CM, Uyoga, S, Macharia, A, Peshu, N, Williams, TN, Manjurano, A, Sepulveda, N, Clark, TG, Riley, E, Drakeley, C, Reyburn, H, Nyirongo, V, Kachala, D, Molyneux, M, Dunstan, SJ, Nguyen, HP, Nguyen, NQ, Cao, QT, Tran, TH, Manning, L, Laman, M, Siba, P, Karunajeewa, H, Allen, S, Allen, A, Davis, TME, Michon, P, Mueller, I, Molloy, SF, Campino, S, Kerasidou, A, Cornelius, VJ, Hart, L, Shah, SS, Band, G, Spencer, CCA, Agbenyega, T, Achidi, E, Doumbo, OK, Farrar, J, Marsh, K, Taylor, T, Kwaitkowski, DP, Clarke, GM, Rockett, K, Kivinen, K, Hubbart, C, Jeffreys, AE, Rowlands, K, Jallow, M, Conway, DJ, Bojang, KA, Pinder, M, Usen, S, Sisay-Joof, F, Sirugo, G, Toure, O, Thera, MA, Konate, S, Sissoko, S, Niangaly, A, Poudiougou, B, Mangano, VD, Bougouma, EC, Sirima, SB, Modioano, D, Amenga-Etego, LN, Ghansah, A, Koram, KA, Wilson, MD, Enimil, A, Evans, J, Amodu, OK, Olaniyan, S, Apinjoh, T, Mugri, R, Ndi, A, Ndila, CM, Uyoga, S, Macharia, A, Peshu, N, Williams, TN, Manjurano, A, Sepulveda, N, Clark, TG, Riley, E, Drakeley, C, Reyburn, H, Nyirongo, V, Kachala, D, Molyneux, M, Dunstan, SJ, Nguyen, HP, Nguyen, NQ, Cao, QT, Tran, TH, Manning, L, Laman, M, Siba, P, Karunajeewa, H, Allen, S, Allen, A, Davis, TME, Michon, P, Mueller, I, Molloy, SF, Campino, S, Kerasidou, A, Cornelius, VJ, Hart, L, Shah, SS, Band, G, Spencer, CCA, Agbenyega, T, Achidi, E, Doumbo, OK, Farrar, J, Marsh, K, Taylor, T, and Kwaitkowski, DP
- Abstract
Glucose-6-phosphate dehydrogenase (G6PD) deficiency is believed to confer protection against Plasmodium falciparum malaria, but the precise nature of the protective effecthas proved difficult to define as G6PD deficiency has multiple allelic variants with different effects in males and females, and it has heterogeneous effects on the clinical outcome of P. falciparum infection. Here we report an analysis of multiple allelic forms of G6PD deficiency in a large multi-centre case-control study of severe malaria, using the WHO classification of G6PD mutations to estimate each individual's level of enzyme activity from their genotype. Aggregated across all genotypes, we find that increasing levels of G6PD deficiency are associated with decreasing risk of cerebral malaria, but with increased risk of severe malarial anaemia. Models of balancing selection based on these findings indicate that an evolutionary trade-off between different clinical outcomes of P. falciparum infection could have been a major cause of the high levels of G6PD polymorphism seen in human populations.
- Published
- 2017
4. Investigating the genetic basis of the Fulani’s lower susceptibility to malaria: the role of autoimmunity loci
- Author
-
Mangano, Valentina, Bougouma, Ec, Verra, Federica, Sepulveda, N, Rockett, Ka, Corran, Ph, Kabore, Y, Diarra, A, Nebie, I, Kwiatkowski, Dp, Sirima, Bs, Modiano, David, and The MalariaGEN Consortium
- Published
- 2012
5. Investigating the evolutionary link between malaria and autoimmunity: a large scale immunogenetic study in two West African populations
- Author
-
Mangano, Valentina, Bougouma, Ec, Verra, Federica, Rockett, Ka, Corran, Ph, Silva, N, Kabore, Y, Diarra, A, Nebie, I, Kwiatkowski, Dp, Sirima, Bs, Modiano, D., and The MalariaGEN Consortium
- Published
- 2012
6. Investigating the genetic basis of the lower susceptibility to malaria shown by the Fulani of West-Africa
- Author
-
Mangano, Valentina, Bougouma, Ec, Verra, Federica, Sepulveda, N, Rockett, Ka, Corran, Ph, Kabore, Y, Diarra, A, Nebie, I, Kwiatkowski, Dp, Sirima, Bs, Modiano, David, and The MalariaGEN Consortium
- Published
- 2012
7. Exploring the evolutionary link between immune responses to Plasmodium falciparum and autoimmunity: a large-scale epidemiological study from Burkina Faso
- Author
-
Mangano, Valentina, Bougouma, Ec, Verra, Federica, Corran, Ph, Rockett, Ka, Silva, N, Avellino, Pamela, Bartoloni, A, Kabore, Y, Diarra, A, Nebie, I, Riley, Em, Kwiatkowski, Dp, Sirima, Bs, Modiano, David, and The MalariaGEN Consortium
- Published
- 2010
8. Association of gene polymorphisms on the susceptibility to malaria in three ethnic groups living in stable and seasonal malaria transmission area of Burkina Faso
- Author
-
Bougouma, Ec, Mangano, Valentina, Kabore, Y, Verra, Federica, Diarra, A, Tiono, A, Ouedraogo, A, Soulama, I, Nebie, I, Rockett, Ka, Kwiatkowski, Dk, Modiano, David, Sirima, Bs, and The MalariaGEN Consortium
- Published
- 2010
9. Reappraisal of known malaria resistance loci in a large multicenter study
- Author
-
Rockett, KA, Clarke, GM, Fitzpatrick, K, Hubbart, C, Jeffreys, AE, Rowlands, K, Craik, R, Jallow, M, Conway, DJ, Bojang, KA, Pinder, M, Usen, S, Sisay-Joof, F, Sirugo, G, Toure, O, Thera, MA, Konate, S, Sissoko, S, Niangaly, A, Poudiougou, B, Mangano, VD, Bougouma, EC, Sirima, SB, Modiano, D, Amenga-Etego, LN, Ghansah, A, Koram, KA, Wilson, MD, Enimil, A, Evans, J, Amodu, O, Olaniyan, S, Apinjoh, T, Mugri, R, Ndi, A, Ndila, CM, Uyoga, S, Macharia, A, Peshu, N, Williams, TN, Manjurano, A, Riley, E, Drakeley, C, Reyburn, H, Nyirongo, V, Kachala, D, Molyneux, M, Dunstan, SJ, Nguyen, HP, Nguyen, TNQ, Cao, QT, Tran, TH, Manning, L, Laman, M, Siba, P, Karunajeewa, H, Allen, S, Allen, A, Davis, TME, Michon, P, Mueller, I, Green, A, Molloy, S, Johnson, KJ, Kerasidou, A, Cornelius, V, Hart, L, Vanderwal, A, SanJoaquin, M, Band, G, Le, SQ, Pirinen, M, Sepulveda, N, Spencer, CCA, Clark, TG, Agbenyega, T, Achidi, E, Doumbo, O, Farrar, J, Marsh, K, Taylor, T, Kwiatkowski, DP, Rockett, KA, Clarke, GM, Fitzpatrick, K, Hubbart, C, Jeffreys, AE, Rowlands, K, Craik, R, Jallow, M, Conway, DJ, Bojang, KA, Pinder, M, Usen, S, Sisay-Joof, F, Sirugo, G, Toure, O, Thera, MA, Konate, S, Sissoko, S, Niangaly, A, Poudiougou, B, Mangano, VD, Bougouma, EC, Sirima, SB, Modiano, D, Amenga-Etego, LN, Ghansah, A, Koram, KA, Wilson, MD, Enimil, A, Evans, J, Amodu, O, Olaniyan, S, Apinjoh, T, Mugri, R, Ndi, A, Ndila, CM, Uyoga, S, Macharia, A, Peshu, N, Williams, TN, Manjurano, A, Riley, E, Drakeley, C, Reyburn, H, Nyirongo, V, Kachala, D, Molyneux, M, Dunstan, SJ, Nguyen, HP, Nguyen, TNQ, Cao, QT, Tran, TH, Manning, L, Laman, M, Siba, P, Karunajeewa, H, Allen, S, Allen, A, Davis, TME, Michon, P, Mueller, I, Green, A, Molloy, S, Johnson, KJ, Kerasidou, A, Cornelius, V, Hart, L, Vanderwal, A, SanJoaquin, M, Band, G, Le, SQ, Pirinen, M, Sepulveda, N, Spencer, CCA, Clark, TG, Agbenyega, T, Achidi, E, Doumbo, O, Farrar, J, Marsh, K, Taylor, T, and Kwiatkowski, DP
- Abstract
Many human genetic associations with resistance to malaria have been reported, but few have been reliably replicated. We collected data on 11,890 cases of severe malaria due to Plasmodium falciparum and 17,441 controls from 12 locations in Africa, Asia and Oceania. We tested 55 SNPs in 27 loci previously reported to associate with severe malaria. There was evidence of association at P < 1 × 10(-4) with the HBB, ABO, ATP2B4, G6PD and CD40LG loci, but previously reported associations at 22 other loci did not replicate in the multicenter analysis. The large sample size made it possible to identify authentic genetic effects that are heterogeneous across populations or phenotypes, with a striking example being the main African form of G6PD deficiency, which reduced the risk of cerebral malaria but increased the risk of severe malarial anemia. The finding that G6PD deficiency has opposing effects on different fatal complications of P. falciparum infection indicates that the evolutionary origins of this common human genetic disorder are more complex than previously supposed.
- Published
- 2014
10. Low-high season variation in Plasmodium falciparum erythrocyte binding antigen 175 (eba-175) allelic forms in malaria endemic area of Burkina Faso.
- Author
-
Soulama I, Bougouma EC, Diarra A, Nebie I, Sirima SB, Soulama, Issiaka, Bougouma, Edith C, Diarra, Amidou, Nebie, Issa, and Sirima, Sodiomon B
- Abstract
Objective: To assess the impact of seasonal variation on the distribution of the eba-175 allelic forms in the area where malaria transmission is markedly seasonal.Methods: Blood samples were collected from 291 and 239 children under 5 years of age during the low and the high malaria transmission season, respectively, in four villages named Dawelgué, Kounda, Tanghin and Watenga of Saponé Health District, then screened for eba 175 F- and C- alleles by nested PCR analysis.Results: F- alleles were more prevalent than C-alleles in the low [0.66 vs. 0.34 (P < 0.0001)] and high transmission season [0.67 vs. 0.33 (P < 0.0001)]. No significant seasonal variation was observed in the distribution of the two alleles. However, according to Sewall Wright rules, the population pairwise F(ST) values, between Dawelgué and Tanghin during the low transmission season (F(ST_) value = 0.10415, P-value = 0.0090 and during the high season (F(ST_)value = 0.08244, P-value < 0.00001), between Tanghin and Watenga during the low season (F(ST) value = 0.07414, P-value = 0.009) indicated a moderate but statistically significant genetic differentiation.Conclusion: Although there was a moderate but significant genetic differentiation between some study villages at different times of the year, this study result in the seasonal stability of eba-175 allele's distribution in the study area. [ABSTRACT FROM AUTHOR]- Published
- 2010
- Full Text
- View/download PDF
11. Admixture into and within sub-Saharan Africa
- Author
-
Angeliki Kerasidou, J O'Brien, Aaron Vanderwal, Christina Hubbart, Alistair Miles, Catherine L. Moyes, A Nyika, Abier Elzein, J Shelton, Spencer Cca., Anthony Enimil, A Diss, C Hughes, Lucas Amenga-Etego, E Somaskantharajah, Ogobara K. Doumbo, Jacob Almagro Garcia, Valentina D. Mangano, E Drury, Edith Bougama, Angie Green, Busby Gbj., Geraldine M. Clarke, Dominic P. Kwiatkowski, Jiannis Ragoussis, Alphaxard Manjurano, Bronwyn MacInnis, Tobias O. Apinjoh, D Mead, Gareth Maslen, George B.J. Busby, Kirk A. Rockett, Dushyanth Jyothi, C Potter, C Malangone, Muminatou Jallow, I Ragoussis, Ellen M. Leffler, J Rogers, J Stalker, Quang Si Le, J Rodford, D Barnwell, Alieu Mendy, J deVries, Anna E. Jeffreys, Carolyne M. Ndila, E Hilton, Vysaul Nyirongo, The Wellcome Trust Centre for Human Genetics [Oxford], University of Oxford [Oxford], The Wellcome Trust Sanger Institute [Cambridge], Medical Research Council Unit The Gambia (MRC), Centre National de Recherche et de Formation sur le Paludisme [Ouagadougou, Burkina Faso] (CNRFP), Università degli Studi di Roma 'La Sapienza' = Sapienza University [Rome], Navrongo Health Research Centre [Navrongo, Ghana] (NHRC), Komfo Anokye Teaching Hospital, University of Buéa, KEMRI-Wellcome Trust Research Programme (KWTRP), London School of Hygiene and Tropical Medicine (LSHTM), University of Malawi, University of Bamako [Mali], Institut Pasteur de Dakar, Réseau International des Instituts Pasteur (RIIP), Wellcome Trust, Medical Research Council, Foundation for the National Institutes of Health, Malaria Genomics Epidemiology Network : Vanderwal A, Elzein A, Nyika A, Mendy A, Miles A, Diss A, Kerasidou A, Green A, Jeffreys AE, MacInnis B, Hughes C, Moyes C, Spencer CC, Hubbart C, Malangone C, Potter C, Mead D, Barnwell D, Kwiatkowski DP, Jyothi D, Drury E, Somaskantharajah E, Hilton E, Leffler E, Maslen G, Band G, Busby G, Clarke GM, Ragoussis I, Garcia JA, Rogers J, deVries J, Shelton J, Ragoussis J, Stalker J, Rodford J, O'Brien J, Evans J, Rowlands K, Cook K, Fitzpatrick K, Kivinen K, Small K, Johnson KJ, Rockett KA, Hart L, Manske M, McCreight M, Stevens M, Pirinen M, Hennsman M, Parker M, SanJoaquin M, Seplúveda N, Cook O, Miotto O, Deloukas P, Craik R, Wrigley R, Watson R, Pearson R, Hutton R, Oyola S, Auburn S, Shah S, Le SQ, Molloy S, Bull S, Campino S, Clark TG, Ruano-Rubio V, Cornelius V, Teo YY, Corran P, Silva ND, Risley P, Doyle A, Evans J, Horstmann R, Plowe C, Duffy P, Carucci D, Gottleib M, Tall A, Ly AB, Dolo A, Sakuntabhai A, Puijalon O, Bah A, Camara A, Sadiq A, Khan AA, Jobarteh A, Mendy A, Ebonyi A, Danso B, Taal B, Casals-Pascual C, Conway DJ, Onykwelu E, Sisay-Joof F, Sirugo G, Kanyi H, Njie H, Obu H, Saine H, Sambou I, Abubakar I, Njie J, Fullah J, Jaiteh J, Bojang KA, Jammeh K, Sabally-Ceesay K, Manneh L, Camara L, Yamoah L, Njie M, Njie M, Pinder M, Jallow M, Aiyegbo M, Jasseh M, Keita ML, Saidy-Khan M, Jallow M, Ceesay N, Rasheed O, Ceesay PL, Esangbedo P, Cole-Ceesay R, Olaosebikan R, Correa S, Njie S, Usen S, Dibba Y, Barry A, Djimdé A, Sall AH, Abathina A, Niangaly A, Dembele A, Poudiougou B, Diarra E, Bamba K, Thera MA, Doumbo O, Toure O, Konate S, Sissoko S, Diakite M, Konate AT, Modiano D, Bougouma EC, Bancone G, Ouedraogo IN, Simpore J, Sirima SB, Mangano VD, Troye-Blomberg M, Oduro AR, Hodgson AV, Ghansah A, Nkrumah F, Atuguba F, Koram KA, Amenga-Etego LN, Wilson MD, Ansah NA, Mensah N, Ansah PA, Anyorigiya T, Asoala V, Rogers WO, Akoto AO, Ofori AO, Enimil A, Ansong D, Sambian D, Asafo-Agyei E, Sylverken J, Antwi S, Agbenyega T, Orimadegun AE, Amodu FA, Oni O, Omotade OO, Amodu O, Olaniyan S, Ndi A, Yafi C, Achidi EA, Mbunwe E, Anchang-Kimbi J, Mugri R, Besingi R, Apinjoh TO, Titanji V, Elhassan A, Hussein A, Mohamed H, Elhassan I, Ibrahim M, Kokwaro G, Oluoch T, Macharia A, Ndila CM, Newton C, Opi DH, Kamuya D, Bauni E, Marsh K, Peshu N, Molyneux S, Uyoga S, Williams TN, Marsh V, Manjurano A, Nadjm B, Maxwell C, Drakeley C, Riley E, Mtei F, Mtove G, Wangai H, Reyburn H, Joseph S, Ishengoma D, Lemnge M, Mutabingwa T, Makani J, Cox S, Phiri A, Munthali A, Kachala D, Njiragoma L, Molyneux ME, Moore M, Ntunthama N, Pensulo P, Taylor T, Nyirongo V, Carter R, Fernando D, Karunaweera N, Dewasurendra R, Suriyaphol P, Singhasivanon P, Simmons CP, Thai CQ, Sinh DX, Farrar J, Chuong LV, Phu NH, Hieu NT, Hoang Mai NT, Ngoc Quyen NT, Day N, Dunstan SJ, O'Riordan SE, Hong Chau TT, Hien TT, Allen A, Lin E, Karunajeewa H, Mueller I, Reeder J, Manning L, Laman M, Michon P, Siba P, Allen S, Davis TM., Commission of the European Communities, and Wellcome Trust
- Subjects
0301 basic medicine ,Population genetics ,Gene flow ,0302 clinical medicine ,MESH: Genetic Variation ,Biology (General) ,African Continental Ancestry Group ,media_common ,Genetics ,0303 health sciences ,education.field_of_study ,Human migration ,General Neuroscience ,030305 genetics & heredity ,General Medicine ,[SDV.BIBS]Life Sciences [q-bio]/Quantitative Methods [q-bio.QM] ,Geography ,Genomics and Evolutionary Biology ,MESH: Human Migration ,[SDV.MP.VIR]Life Sciences [q-bio]/Microbiology and Parasitology/Virology ,Medicine ,admixture ,gene-flow ,Research Article ,Gene Flow ,QH301-705.5 ,Science ,media_common.quotation_subject ,Human Migration ,Population ,Black People ,Genomics ,Biology ,africa ,chromosome painting ,evolutionary biology ,genomics ,human ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,Genetic variation ,Humans ,[SDV.MP.PAR]Life Sciences [q-bio]/Microbiology and Parasitology/Parasitology ,MESH: Africa South of the Sahara ,Allele ,education ,Africa South of the Sahara ,MESH: Gene Flow ,MESH: Genome, Human ,030304 developmental biology ,Genetic diversity ,MESH: Humans ,[SDV.GEN.GPO]Life Sciences [q-bio]/Genetics/Populations and Evolution [q-bio.PE] ,General Immunology and Microbiology ,business.industry ,Genome, Human ,Haplotype ,Genetic Variation ,MESH: Haplotypes ,030104 developmental biology ,Genetic epidemiology ,Haplotypes ,[SDV.GEN.GH]Life Sciences [q-bio]/Genetics/Human genetics ,Agriculture ,Evolutionary biology ,Africa ,[SDV.SPEE]Life Sciences [q-bio]/Santé publique et épidémiologie ,MESH: African Continental Ancestry Group ,[INFO.INFO-BI]Computer Science [cs]/Bioinformatics [q-bio.QM] ,business ,030217 neurology & neurosurgery ,Demography ,Diversity (politics) - Abstract
Similarity between two individuals in the combination of genetic markers along their chromosomes indicates shared ancestry and can be used to identify historical connections between different population groups due to admixture. We use a genome-wide, haplotype-based, analysis to characterise the structure of genetic diversity and gene-flow in a collection of 48 sub-Saharan African groups. We show that coastal populations experienced an influx of Eurasian haplotypes over the last 7000 years, and that Eastern and Southern Niger-Congo speaking groups share ancestry with Central West Africans as a result of recent population expansions. In fact, most sub-Saharan populations share ancestry with groups from outside of their current geographic region as a result of gene-flow within the last 4000 years. Our in-depth analysis provides insight into haplotype sharing across different ethno-linguistic groups and the recent movement of alleles into new environments, both of which are relevant to studies of genetic epidemiology. DOI: http://dx.doi.org/10.7554/eLife.15266.001, eLife digest Our genomes contain a record of historical events. This is because when groups of people are separated for generations, the DNA sequence in the two groups’ genomes will change in different ways. Looking at the differences in the genomes of people from the same population can help researchers to understand and reconstruct the historical interactions that brought their ancestors together. The mixing of two populations that were previously separate is known as admixture. Africa as a continent has few written records of its history. This means that it is somewhat unknown which important movements of people in the past generated the populations found in modern-day Africa. Busby et al. have now attempted to use DNA to look into this and reconstruct the last 4000 years of genetic history in African populations. As has been shown in other regions of the world, the new analysis showed that all African populations are the result of historical admixture events. However, Busby et al. could characterize these events to unprecedented level of detail. For example, multiple ethnic groups from The Gambia and Mali all show signs of sharing the same set of ancestors from West Africa, Europe and Asia who mixed around 2000 years ago. Evidence of a migration of people from Central West Africa, known as the Bantu expansion, could also be detected, and was shown to carry genes to the south and east. An important next step will be to now look at the consequences of the observed gene-flow, and ask if it has contributed to spreading beneficial, or detrimental, mutations around Africa. DOI: http://dx.doi.org/10.7554/eLife.15266.002
- Published
- 2016
- Full Text
- View/download PDF
12. Evaluating artesunate monotherapy and dihydroartemisinin-piperaquine as potential antimalarial options for prevaccination radical cures during future malaria vaccine field efficacy trials.
- Author
-
Ouédraogo A, Ouattara D, Ouattara SM, Diarra A, Badoum ES, Hema A, Ouédraogo AZ, Hien D, Bougouma EC, Nébié I, Bocquet V, Vaillant M, Tiono AB, and Sirima SB
- Subjects
- Humans, Child, Preschool, Child, Male, Female, Infant, Cohort Studies, Treatment Outcome, Polymerase Chain Reaction, Piperazines, Artemisinins therapeutic use, Artemisinins administration & dosage, Antimalarials therapeutic use, Antimalarials administration & dosage, Quinolines administration & dosage, Quinolines therapeutic use, Malaria, Falciparum prevention & control, Artesunate therapeutic use, Malaria Vaccines administration & dosage, Malaria Vaccines immunology, Plasmodium falciparum drug effects, Plasmodium falciparum immunology
- Abstract
Background: In malaria vaccine clinical trials, immune responses after vaccination may be compromised due to immunosuppression caused by concurrent Plasmodium falciparum infection. This has a direct effect on the protective efficacy of the vaccine being evaluated. Therefore, parasite clearance prior to vaccination is being considered. Drugs with good safety and efficacy profiles and a short posttreatment prophylaxis period should be used. Two antimalarial drugs, artesunate (AS) as monotherapy and dihydroartemisinin-piperaquine (DHAPQ), have been evaluated in order to identify the most suitable option for use in future trials., Methods: A cohort of children aged 1.5-12 years living in the Banfora Health District area was recruited. They were randomly assigned to receive supervised curative doses of AS monotherapy for 7 days or DHAPQ for 3 days. A polymerase chain reaction (PCR) was performed 21 days after treatment to confirm clearance of infection, and only those with a negative PCR were included in the study cohort for a 6-month longitudinal follow-up. Cohort children were actively visited fortnightly to collect blood samples for P. falciparum detection via microscopy and PCR. Passive surveillance was also conducted at the local health facility to record incident malaria episodes that occurred between two active visits., Results: A total of 513 children were treated. Among these patients, 458 (89.3%) were free of P. falciparum malaria infection on day 21: 87.3% (226/259) in the AS group vs 91.3% (232/254) in the DHAPQ group (p = 0.053). The mean time to first malaria infection by microscopy was 154.9 (2.9) days in the DHAPQ arm and 129.0 (3.9) days in the AS arm (p < 0.01). The incidence rates of clinical malaria episodes during the follow-up period were 0.507 (0.369-0.645) and 0.293 (0.190-0.397) in the AS and DHAPQ arms, respectively (p < 0.05)., Conclusions: These findings suggest that although both drugs are effective in clearing P. falciparum infections, AS is likely to cause no more than minimal interference with the evaluation of vaccine efficacy endpoints and could, therefore, be considered for use., Trial Registration: NCT04601714., Competing Interests: Declarations. Ethics approval and consent to participate: The study was approved by the National Ethics Committee of the Ministry of Health (under deliberation No. 2020-6-101 dated 10/06/2020). Consent for publication: Not applicable. Competing interests: The authors declare no competing interests., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
13. A randomized first-in-human phase I trial of differentially adjuvanted Pfs48/45 malaria vaccines in Burkinabé adults.
- Author
-
Tiono AB, Plieskatt JL, Ouedraogo A, Soulama BI, Miura K, Bougouma EC, Naghizadeh M, Barry A, Yaro JBB, Ezinmegnon S, Henry N, Ofori EA, Adu B, Singh SK, Konkobo A, Lövgren Bengtsson K, Diarra A, Carnrot C, Reimer JM, Ouedraogo A, Tienta M, Long CA, Ouedraogo IN, Sagara I, Sirima SB, and Theisen M
- Subjects
- Adult, Humans, Plasmodium falciparum, Protozoan Proteins, Adjuvants, Immunologic, Antigens, Protozoan, Aluminum Hydroxide, Antibodies, Protozoan, Malaria Vaccines, Malaria, Malaria, Falciparum
- Abstract
BACKGROUNDMalaria transmission-blocking vaccines aim to interrupt the transmission of malaria from one person to another.METHODSThe candidates R0.6C and ProC6C share the 6C domain of the Plasmodium falciparum sexual-stage antigen Pfs48/45. R0.6C utilizes the glutamate-rich protein (GLURP) as a carrier, and ProC6C includes a second domain (Pfs230-Pro) and a short 36-amino acid circumsporozoite protein (CSP) sequence. Healthy adults (n = 125) from a malaria-endemic area of Burkina Faso were immunized with 3 intramuscular injections, 4 weeks apart, of 30 μg or 100 μg R0.6C or ProC6C each adsorbed to Alhydrogel (AlOH) adjuvant alone or in combination with Matrix-M (15 μg or 50 μg, respectively). The allocation was random and double-blind for this phase I trial.RESULTSThe vaccines were safe and well tolerated with no vaccine-related serious adverse events. A total of 7 adverse events, mild to moderate in intensity and considered possibly related to the study vaccines, were recorded. Vaccine-specific antibodies were highest in volunteers immunized with 100 μg ProC6C-AlOH with Matrix-M, and 13 of 20 (65%) individuals in the group showed greater than 80% transmission-reducing activity (TRA) when evaluated in the standard membrane feeding assay at 15 mg/mL IgG. In contrast, R0.6C induced sporadic TRA.CONCLUSIONAll formulations were safe and well tolerated in a malaria-endemic area of Africa in healthy adults. The ProC6C-AlOH/Matrix-M vaccine elicited the highest levels of functional antibodies, meriting further investigation.TRIAL REGISTRATIONPan-African Clinical Trials Registry (https://pactr.samrc.ac.za) PACTR202201848463189.FUNDINGThe study was funded by the European and Developing Countries Clinical Trials Partnership (grant RIA2018SV-2311).
- Published
- 2024
- Full Text
- View/download PDF
14. Persistence of Anti-SE36 Antibodies Induced by the Malaria Vaccine Candidate BK-SE36/CpG in 5-10-Year-Old Burkinabe Children Naturally Exposed to Malaria.
- Author
-
Nebie I, Palacpac NMQ, Bougouma EC, Diarra A, Ouédraogo A, D'Alessio F, Houard S, Tiono AB, Cousens S, Horii T, and Sirima SB
- Abstract
Information on the dynamics and decline/persistence of antibody titres is important in vaccine development. A recent vaccine trial in malaria-exposed, healthy African adults and children living in a malaria hyperendemic and seasonal area (Ouagadougou, Burkina Faso) was the first study in which BK-SE36/CpG was administered to different age groups. In 5- to 10-year-old children, the risk of malaria infection was markedly lower in the BK-SE36/CpG arm compared to the control arm. We report here data on antibody titres measured in this age-group after the high malaria transmission season of 2021 (three years after the first vaccine dose was administered). At Year 3, 83% of children had detectable anti-SE36 total IgG antibodies. Geometric mean antibody titres and the proportion of children with detectable anti-SE36 antibodies were markedly higher in the BK-SE36/CpG arm than the control (rabies) arm. The information obtained in this study will guide investigators on future vaccine/booster schedules for this promising blood-stage malaria vaccine candidate.
- Published
- 2024
- Full Text
- View/download PDF
15. Safety and immunogenicity of BK-SE36/CpG malaria vaccine in healthy Burkinabe adults and children: a phase 1b randomised, controlled, double-blinded, age de-escalation trial.
- Author
-
Ouédraogo A, Bougouma EC, Palacpac NMQ, Houard S, Nebie I, Sawadogo J, Berges GD, Soulama I, Diarra A, Hien D, Ouedraogo AZ, Konaté AT, Kouanda S, Myoui A, Ezoe S, Ishii KJ, Sato T, D'Alessio F, Leroy O, Tiono AB, Cousens S, Horii T, and Sirima SB
- Subjects
- Male, Humans, Adult, Child, Infant, Child, Preschool, Young Adult, Middle Aged, Double-Blind Method, Peptides, Malaria Vaccines, Malaria, Falciparum prevention & control, Malaria prevention & control
- Abstract
Background: BK-SE36/CpG is a recombinant blood-stage malaria vaccine candidate based on the N-terminal Plasmodium falciparum serine repeat antigen5 (SE36), adsorbed to aluminium hydroxide gel and reconstituted, prior to administration, with synthetic oligodeoxynucleotides bearing CpG motifs. In healthy Japanese adult males, BK-SE36/CpG was well tolerated. This study assessed its safety and immunogenicity in healthy malaria-exposed African adults and children., Methods: A double-blind, randomised, controlled, age de-escalating clinical trial was conducted in an urban area of Ouagadougou, Burkina Faso. Healthy participants (n=135) aged 21-45 years (Cohort 1), 5-10 years (Cohort 2) and 12-24 months (Cohort 3) were randomised to receive three vaccine doses (Day 0, 28 and 112) of BK-SE36/CpG or rabies vaccine by intramuscular injection., Results: One hundred thirty-four of 135 (99.2%) subjects received all three scheduled vaccine doses. Vaccinations were well tolerated with no related Grade 3 (severe) adverse events (AEs). Pain/limitation of limb movement, headache in adults and fever in younger children (all mild to moderate in intensity) were the most frequently observed local and systemic AEs. Eighty-three of BK-SE36/CpG (91%) recipients and 37 of control subjects (84%) had Grade 1/2 events within 28 days post vaccination. Events considered by the investigator to be vaccine related were experienced by 38% and 14% of subjects in BK-SE36/CpG and control arms, respectively. Throughout the trial, six Grade 3 events (in 4 subjects), not related to vaccination, were recorded in the BK-SE36/CpG arm: 5 events (in 3 subjects) within 28 days of vaccination. All serious adverse events (SAEs) (n=5) were due to severe malaria (52-226 days post vaccination) and not related to vaccination. In all cohorts, BK-SE36/CpG arm had higher antibody titres after Dose 3 than after Dose 2. Younger cohorts had stronger immune responses (12-24-month-old > 5-10 years-old > 21-45 years-old). Sera predominantly reacted to peptides that lie in intrinsically unstructured regions of SE36. In the control arm, there were no marked fold changes in antibody titres and participants' sera reacted poorly to all peptides spanning SE36., Conclusion: BK-SE36/CpG was well-tolerated and immunogenic. These results pave the way for further proof-of-concept studies to demonstrate vaccine efficacy., Clinical Trial Registration: https://pactr.samrc.ac.za/TrialDisplay.aspx?TrialID=1921, PACTR201701001921166., Competing Interests: TH is the inventor of BK-SE36; TH and KI are inventors of BK-SE36/CpG. NP and TH are supported by a research fund from Nobelpharma Co., Ltd NPC, the clinical trial sponsor. TS is an employee and SE is medical adviser of NPC. These involvements did not influence the design of the study, the collection, analyses, access to and interpretation of data, and the writing of this manuscript. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest. The author(s) declared that they were an editorial board member of Frontiers, at the time of submission. This had no impact on the peer review process and the final decision., (Copyright © 2023 Ouédraogo, Bougouma, Palacpac, Houard, Nebie, Sawadogo, Berges, Soulama, Diarra, Hien, Ouedraogo, Konaté, Kouanda, Myoui, Ezoe, Ishii, Sato, D’Alessio, Leroy, Tiono, Cousens, Horii and Sirima.)
- Published
- 2023
- Full Text
- View/download PDF
16. Plasmodium falciparum infection coinciding with the malaria vaccine candidate BK-SE36 administration interferes with the immune responses in Burkinabe children.
- Author
-
Tiono AB, Palacpac NMQ, Bougouma EC, Nebie I, Ouédraogo A, Houard S, Arisue N, D'Alessio F, Horii T, and Sirima SB
- Subjects
- Humans, Child, Plasmodium falciparum, Antigens, Protozoan, Vaccination adverse effects, Immunoglobulin G, Immunity, Malaria Vaccines, Malaria, Falciparum prevention & control, Malaria prevention & control
- Abstract
Background: A vaccine targeting the erythrocyte stages of Plasmodium falciparum could play a role in preventing clinical disease. BK-SE36 is a promising malaria vaccine candidate that has shown a good safety profile and immunological responses during field evaluations. It was observed that repeated natural infections could result in immune tolerance against SE36 molecule., Methods: The primary trial was conducted to assess the safety and immunogenicity of the BK-SE36 in two cohorts of children aged 25-60 months (Cohort 1) and 12-24 months (Cohort 2). Immunization was at full dose (1.0 mL) administered at 0, 1, and 6 months. Blood samples were collected before each vaccination for immunological assessments and detection of Plasmodium falciparum infection by microscopy. Blood samples were further collected one month post each vaccination to evaluate immunogenicity., Results: Of seventy-two (72) subjects that have received BK-SE36 vaccination, 71 had available blood smears during vaccination days. One month post Dose 2, the geometric mean of SE36 antibodies was 263.2 (95% CI: 178.9-387.1) in uninfected individuals compared to 77.1 (95% CI: 47.3-125.7) in infected participants. The same trend was observed one-month post booster dose. Participants uninfected at the time of booster vaccination had significantly higher GMTs compared to those who were infected (424.1 (95% CI: 301.9-595.8) vs . 92.8 (95% CI: 34.9-246.6), p = 0.002. There was a 14.3 (95% CI: 9.7-21.1) and 2.4 (95% CI: 1.3-4.4) fold-change, respectively, in uninfected and infected participants between one-month post Dose 2 and booster. The difference was statistically significant ( p < 0.001)., Conclusion: Concomitant infection by P. falciparum during BK-SE36 vaccine candidate administration is associated with reduced humoral responses. However, it is to be noted that the BK-SE36 primary trial was not designed to investigate the influence of concomitant infection on vaccine-induced immune response and should be interpreted cautiously., Trial Registration: WHO ICTRP, PACTR201411000934120., Competing Interests: TH is the inventor of BK-SE36 and all rights have now been turned over to Nobelpharma Co., Ltd (NPC), Tokyo, Japan. NP served as a contract researcher for NPC, Apr - Sept 2017. SH and FD received support from NPC for salaries, travel, and CRO cost for clinical monitoring. EB also received support from NPC for salaries during the long-term follow-up. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2023 Tiono, Palacpac, Bougouma, Nebie, Ouédraogo, Houard, Arisue, D’Alessio, Horii and Sirima.)
- Published
- 2023
- Full Text
- View/download PDF
17. African-specific polymorphisms in Plasmodium falciparum serine repeat antigen 5 in Uganda and Burkina Faso clinical samples do not interfere with antibody response to BK-SE36 vaccination.
- Author
-
Arisue N, Palacpac NMQ, Ntege EH, Yeka A, Balikagala B, Kanoi BN, Bougouma EC, Tiono AB, Nebie I, Diarra A, Houard S, D'Alessio F, Leroy O, Sirima SB, Egwang TG, and Horii T
- Subjects
- Humans, Antibody Formation, Antigens, Protozoan genetics, Burkina Faso, Cross-Sectional Studies, Plasmodium falciparum genetics, Uganda, Vaccination, Clinical Trials, Phase I as Topic, Malaria Vaccines genetics, Malaria, Falciparum prevention & control
- Abstract
BK-SE36, based on Plasmodium falciparum serine repeat antigen 5 (SERA5), is a blood-stage malaria vaccine candidate currently being evaluated in clinical trials. Phase 1 trials in Uganda and Burkina Faso have demonstrated promising safety and immunogenicity profiles. However, the genetic diversity of sera5 in Africa and the role of allele/variant-specific immunity remain a major concern. Here, sequence analyses were done on 226 strains collected from the two clinical trial/follow-up studies and 88 strains from two cross-sectional studies in Africa. Compared to other highly polymorphic vaccine candidate antigens, polymorphisms in sera5 were largely confined to the repeat regions of the gene. Results also confirmed a SERA5 consensus sequence with African-specific polymorphisms. Mismatches with the vaccine-type SE36 (BK-SE36) in the octamer repeat, serine repeat, and flanking regions, and single-nucleotide polymorphisms in non-repeat regions could compromise vaccine response and efficacy. However, the haplotype diversity of SERA5 was similar between vaccinated and control participants. There was no marked bias or difference in the patterns of distribution of the SE36 haplotype and no statistically significant genetic differentiation among parasites infecting BK-SE36 vaccinees and controls. Results indicate that BK-SE36 does not elicit an allele-specific immune response., Competing Interests: TH is the inventor of BK-SE36 and all rights have now been turned over to NPC. NP, EN, AY, BB, BK, and TE received remuneration from BIKEN for the Ugandan trial and follow-up. For the Burkina Faso clinical trial/follow-up study, the following received some support from NPC: EB, AD, SH, FD, OL, and NP. TH and NP are currently supported by a research fund from NPC. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2022 Arisue, Palacpac, Ntege, Yeka, Balikagala, Kanoi, Bougouma, Tiono, Nebie, Diarra, Houard, D’Alessio, Leroy, Sirima, Egwang and Horii.)
- Published
- 2022
- Full Text
- View/download PDF
18. A randomized controlled trial showing safety and efficacy of a whole sporozoite vaccine against endemic malaria.
- Author
-
Sirima SB, Ouédraogo A, Tiono AB, Kaboré JM, Bougouma EC, Ouattara MS, Kargougou D, Diarra A, Henry N, Ouédraogo IN, Billingsley PF, Manoj A, Abebe Y, Kc N, Ruben A, Richie TL, James ER, Joshi S, Shrestha B, Strauss K, Lyke KE, Plowe CV, Potter GE, Cox C, Jones W, Sim BKL, Hoffman SL, and Laurens MB
- Subjects
- Humans, Animals, Sporozoites, Vaccines
- Abstract
A highly effective malaria vaccine remains elusive despite decades of research. Plasmodium falciparum sporozoite vaccine (PfSPZ Vaccine), a metabolically active, nonreplicating, whole parasite vaccine demonstrated safety and vaccine efficacy (VE) against endemic P. falciparum for 6 months in Malian adults receiving a five-dose regimen. Safety, immunogenicity, and VE of a three-dose regimen were assessed in adults in Balonghin, Burkina Faso in a two-component study: an open-label dose escalation trial with 32 participants followed by a double-blind, randomized, placebo-controlled trial (RCT) with 80 participants randomized to receive three doses of 2.7 × 10
6 PfSPZ ( N = 39) or normal saline ( N = 41) just before malaria season. To clear parasitemia, artesunate monotherapy was administered before first and last vaccinations. Thick blood smear microscopy was performed on samples collected during illness and every 4 weeks for 72 weeks after last vaccinations, including two 6-month malaria transmission seasons. Safety outcomes were assessed in all 80 participants who received at least one dose and VE for 79 participants who received three vaccinations. Myalgia was the only symptom that differed between groups. VE (1 - risk ratio; primary VE endpoint) was 38% at 6 months ( P = 0.017) and 15% at 18 months (0.078). VE (1 - hazard ratio) was 48% and 46% at 6 and 18 months ( P = 0.061 and 0.018). Two weeks after the last dose, antibodies to P. falciparum circumsporozoite protein and PfSPZ were higher in protected versus unprotected vaccinees. A three-dose regimen of PfSPZ Vaccine demonstrated safety and efficacy against malaria infection in malaria-experienced adults.- Published
- 2022
- Full Text
- View/download PDF
19. Safety and immunogenicity of BK-SE36 in a blinded, randomized, controlled, age de-escalating phase Ib clinical trial in Burkinabe children.
- Author
-
Bougouma EC, Palacpac NMQ, Tiono AB, Nebie I, Ouédraogo A, Houard S, Yagi M, Coulibaly SA, Diarra A, Tougan T, Ouedraogo AZ, Soulama I, Arisue N, Yaro JB, D'Alessio F, Leroy O, Cousens S, Horii T, and Sirima SB
- Subjects
- Aluminum, Antigens, Protozoan, Child, Child, Preschool, Humans, Plasmodium falciparum, Malaria Vaccines, Malaria, Falciparum prevention & control
- Abstract
Background: A blood-stage vaccine targeting the erythrocytic-stages of the malaria parasite Plasmodium falciparum could play a role to protect against clinical disease. Antibodies against the P. falciparum serine repeat antigen 5 (SE47 and SE36 domains) correlate well with the absence of clinical symptoms in sero-epidemiological studies. A previous phase Ib trial of the recombinant SE36 antigen formulated with aluminum hydroxyl gel (BK-SE36) was promising. This is the first time the vaccine candidate was evaluated in young children below 5 years using two vaccination routes., Methods: Safety and immunogenicity of BK-SE36 was assessed in a double-blind, randomized, controlled, age de-escalating phase Ib trial. Fifty-four Burkinabe children in each age cohort, 25-60 or 12-24 months, were randomized in a 1:1:1 ratio to receive three doses of BK-SE36 either by intramuscular (BK IM) or subcutaneous (BK SC) route on Day 0, Week 4, and 26; or the control vaccine, Synflorix
® via IM route on Day 0, Week 26 (and physiological saline on Week 4). Safety data and samples for immunogenicity analyses were collected at various time-points., Results: Of 108 subjects, 104 subjects (96.3%) (Cohort 1: 94.4%; Cohort 2: 98.1%) received all three scheduled vaccine doses. Local reactions, mostly mild or of moderate severity, occurred in 99 subjects (91.7%). The proportion of subjects that received three doses without experiencing Grade 3 adverse events was similar across BK-SE36 vaccines and control arms (Cohort 1: 100%, 89%, and 89%; and Cohort 2: 83%, 82%, and 83% for BK IM, BK SC, and control, respectively). BK-SE36 vaccine was immunogenic, inducing more than 2-fold change in antibody titers from pre-vaccination, with no difference between the two vaccination routes. Titers waned before the third dose but in both cohorts titers were boosted 6 months after the first vaccination. The younger cohort had 2-fold and 4-fold higher geometric mean titers compared to the 25- to 60-month-old cohort after 2 and 3 doses of BK-SE36, respectively., Conclusion: BK-SE36 was well tolerated and immunogenic using either intramuscular or subcutaneous routes, with higher immune response in the younger cohort., Clinical Trial Registration: https://pactr.samrc.ac.za/TrialDisplay.aspx?TrialID=934, identifier PACTR201411000934120., Competing Interests: TH is the inventor of BK-SE36 and all rights have now been turned over to NPC. NP served as contract researcher for NPC, Apr - Sept 2017. SH, FD and OL received support from NPC for salaries, travel and CRO cost for clinical monitoring. EB, SaC, AD, AZO, JY also received support from NPC for salaries during the long term follow-up. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2022 Bougouma, Palacpac, Tiono, Nebie, Ouédraogo, Houard, Yagi, Coulibaly, Diarra, Tougan, Ouedraogo, Soulama, Arisue, Yaro, D’Alessio, Leroy, Cousens, Horii and Sirima.)- Published
- 2022
- Full Text
- View/download PDF
20. Safety and immunogenicity of Vi-typhoid conjugate vaccine co-administration with routine 9-month vaccination in Burkina Faso: A randomized controlled phase 2 trial.
- Author
-
Sirima SB, Ouedraogo A, Barry N, Siribie M, Tiono A, Nébié I, Konaté A, Berges GD, Diarra A, Ouedraogo M, Bougouma EC, Soulama I, Hema A, Datta S, Liang Y, Rotrosen ET, Tracy JK, Jamka LP, Oshinsky JJ, Pasetti MF, Neuzil KM, and Laurens MB
- Subjects
- Burkina Faso, Double-Blind Method, Female, Humans, Infant, Male, Measles Vaccine administration & dosage, Polysaccharides, Bacterial administration & dosage, Polysaccharides, Bacterial immunology, Rubella Vaccine administration & dosage, Typhoid-Paratyphoid Vaccines administration & dosage, Typhoid-Paratyphoid Vaccines immunology, Vaccines, Conjugate adverse effects, Vaccines, Conjugate immunology, Yellow Fever Vaccine administration & dosage, Polysaccharides, Bacterial adverse effects, Typhoid-Paratyphoid Vaccines adverse effects
- Abstract
Objectives: In 2017, the World Health Organisation (WHO) pre-qualified a single-dose typhoid conjugate vaccine (TCV) and identified TCV co-administration studies as a research priority. Accordingly, we tested co-administration of Typbar TCV® (Bharat Biotech International) with measles-rubella (MR) and yellow fever (YF) vaccines., Methods: We conducted a randomized, double-blind, and controlled, phase 2 trial in Ouagadougou, Burkina Faso. Healthy children aged 9-11 months were randomized 1:1 to receive TCV (Group 1) or control vaccine (inactivated polio vaccine (IPV), Group 2). Vaccines were administered intramuscularly with routine MR and YF vaccines. Safety was assessed by (1) local and systemic reactions on days 0, 3, and 7; (2) unsolicited adverse events within 28 days; and (3) serious adverse events (SAEs) within six months after immunization., Results: We enrolled, randomized, and vaccinated 100 eligible children (49 Group 1 and 51 Group 2). Safety outcomes occurred with similar frequency in both groups: local/solicited reactions (Group 1: 1/49, Group 2: 3/50), systemic/solicited reactions (Group 1: 4/49, Group 2: 9/50), unsolicited adverse events (Group 1: 26/49, Group 2: 33/51), and SAEs (Group 1: 2/49, Group 2: 3/51). TCV conferred robust immunogenicity without interference with MR or YF vaccines., Conclusion: TCV can be safely co-administered with MR and YF vaccines to children at the 9-month vaccination visit., Competing Interests: Declaration of Competing Interest The authors report no declarations of interest., (Copyright © 2021 The Author(s). Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
21. G6PD Polymorphisms and Hemolysis After Antimalarial Treatment With Low Single-Dose Primaquine: A Pooled Analysis of Six African Clinical Trials.
- Author
-
Sepúlveda N, Grignard L, Curry J, Mahey L, Bastiaens GJH, Tiono AB, Okebe J, Coulibaly SA, Gonçalves BP, Affara M, Ouédraogo A, Bougouma EC, Sanou GS, Nébié I, Lanke K, Sirima SB, Dicko A, d'Alessandro U, Clark TG, Campino S, Chen I, Eziefula AC, Gosling R, Bousema T, and Drakeley C
- Abstract
Primaquine (PQ) is an antimalarial drug with the potential to reduce malaria transmission due to its capacity to clear mature Plasmodium falciparum gametocytes in the human host. However, the large-scale roll-out of PQ has to be counterbalanced by the additional risk of drug-induced hemolysis in individuals suffering from Glucose-6-phospate dehydrogenase (G6PD) deficiency, a genetic condition determined by polymorphisms on the X-linked G6PD gene. Most studies on G6PD deficiency and PQ-associated hemolysis focused on the G6PD A- variant, a combination of the two single nucleotide changes G202A (rs1050828) and A376G (rs1050829), although other polymorphisms may play a role. In this study, we tested the association of 20 G6PD single nucleotide polymorphisms (SNPs) with hemolysis measured seven days after low single dose of PQ given at the dose of 0.1 mg/kg to 0.75 mg/kg in 957 individuals from 6 previously published clinical trials investigating the safety and efficacy of this drug spanning five African countries. After adjusting for inter-study effects, age, gender, baseline hemoglobin level, PQ dose, and parasitemia at screening, our analysis showed putative association signals from the common G6PD mutation, A376G [-log
10 ( p -value) = 2.44] and two less-known SNPs, rs2230037 [-log10 ( p -value] = 2.60), and rs28470352 [-log10 ( p -value) = 2.15]; A376G and rs2230037 were in very strong linkage disequilibrium with each other ( R2 = 0.978). However, when the effects of these SNPs were included in the same regression model, the subsequent associations were in the borderline of statistical significance. In conclusion, whilst a role for the A- variant is well established, we did not observe an important additional role for other G6PD polymorphisms in determining post-treatment hemolysis in individuals treated with low single-dose PQ., Competing Interests: JC and LM were employed by the company LGC Genomics. The remaining authors declare that the research was conducted in the absence of any commercial and financial relationships that could be construed as a potential conflict of interest., (Copyright © 2021 Sepúlveda, Grignard, Curry, Mahey, Bastiaens, Tiono, Okebe, Coulibaly, Gonçalves, Affara, Ouédraogo, Bougouma, Sanou, Nébié, Lanke, Sirima, Dicko, d’Alessandro, Clark, Campino, Chen, Eziefula, Gosling, Bousema and Drakeley.)- Published
- 2021
- Full Text
- View/download PDF
22. Bead-based assays to simultaneously detect multiple human inherited blood disorders associated with malaria.
- Author
-
Grignard L, Mair C, Curry J, Mahey L, Bastiaens GJH, Tiono AB, Okebe J, Coulibaly SA, Gonçalves BP, Affara M, Ouédraogo A, Bougouma EC, Sanou GS, Nébié I, Lanke KHW, Sirima SB, d'Alessandro U, Clark TG, Campino S, Bousema T, and Drakeley C
- Subjects
- Adolescent, Adult, Burkina Faso, Child, Glucosephosphate Dehydrogenase genetics, Hemoglobin C genetics, Hemoglobin, Sickle genetics, Humans, Malaria complications, Male, Middle Aged, Young Adult, Anemia, Sickle Cell diagnosis, Genotyping Techniques methods, Glucosephosphate Dehydrogenase Deficiency diagnosis, Hemoglobin C Disease diagnosis, Polymorphism, Single Nucleotide
- Abstract
Background: Glucose-6-phosphate dehydrogenase deficiency (G6PDd), haemoglobin C (HbC) and S (HbS) are inherited blood disorders (IBD) common in populations in malaria endemic areas. All are associated to some degree with protection against clinical malaria whilst additionally G6PDd is associated with haemolysis following treatment with 8-aminoquinolines. Measuring the prevalence of these inherited blood disorders in affected populations can improve understanding of disease epidemiology. Current methodologies in epidemiological studies commonly rely on individual target amplification and visualization; here a method is presented to simultaneously detect the polymorphisms and that can be expanded to include other single nucleotide polymorphisms (SNPs) of interest., Methods: Human DNA from whole blood samples was amplified in a novel, multiplex PCR reaction and extended with SNP-specific probes in an allele specific primer extension (ASPE) to simultaneously detect four epidemiologically important human markers including G6PD SNPs (G202A and A376G) and common haemoglobin mutations (HbS and HbC). The products were hybridized to magnetic beads and the median fluorescence intensity (MFI) was read on MAGPIX
® (Luminex corp.). Genotyping data was compared to phenotypical data generated by flow cytometry and to established genotyping methods., Results: Seventy-five samples from Burkina Faso (n = 75/78, 96.2%) and 58 samples from The Gambia (n = 58/61, 95.1%) had a G6PD and a HBB genotype successfully assigned by the bead-based assay. Flow cytometry data available for n = 61 samples further supported the concordance between % G6PD normal/deficient cells and genotype., Conclusions: The bead based assay compares well to alternative measures of genotyping and phenotyping for G6PD. The screening is high throughput, adaptable to inclusion of multiple targets of interest and easily standardized.- Published
- 2019
- Full Text
- View/download PDF
23. First field efficacy trial of the ChAd63 MVA ME-TRAP vectored malaria vaccine candidate in 5-17 months old infants and children.
- Author
-
Tiono AB, Nébié I, Anagnostou N, Coulibaly AS, Bowyer G, Lam E, Bougouma EC, Ouedraogo A, Yaro JBB, Barry A, Roberts R, Rampling T, Bliss C, Hodgson S, Lawrie A, Ouedraogo A, Imoukhuede EB, Ewer KJ, Viebig NK, Diarra A, Leroy O, Bejon P, Hill AVS, and Sirima SB
- Subjects
- Adenoviruses, Simian genetics, Double-Blind Method, Enzyme-Linked Immunosorbent Assay, Female, Humans, Infant, Kaplan-Meier Estimate, Kenya, Leukocytes, Mononuclear immunology, Malaria immunology, Malaria prevention & control, Male, Plasmodium falciparum immunology, Plasmodium falciparum pathogenicity, T-Lymphocytes metabolism, Vaccinia virus genetics, Malaria Vaccines therapeutic use
- Abstract
Background: Heterologous prime boost immunization with chimpanzee adenovirus 63 (ChAd63) and Modified Vaccinia Virus Ankara (MVA) vectored vaccines is a strategy previously shown to provide substantial protective efficacy against P. falciparum infection in United Kingdom adult Phase IIa sporozoite challenge studies (approximately 20-25% sterile protection with similar numbers showing clear delay in time to patency), and greater point efficacy in a trial in Kenyan adults., Methodology: We conducted the first Phase IIb clinical trial assessing the safety, immunogenicity and efficacy of ChAd63 MVA ME-TRAP in 700 healthy malaria exposed children aged 5-17 months in a highly endemic malaria transmission area of Burkina Faso., Results: ChAd63 MVA ME-TRAP was shown to be safe and immunogenic but induced only moderate T cell responses (median 326 SFU/106 PBMC (95% CI 290-387)) many fold lower than in previous trials. No significant efficacy was observed against clinical malaria during the follow up period, with efficacy against the primary endpoint estimate by proportional analysis being 13.8% (95%CI -42.4 to 47.9) at sixth month post MVA ME-TRAP and 3.1% (95%CI -15.0 to 18.3; p = 0.72) by Cox regression., Conclusions: This study has confirmed ChAd63 MVA ME-TRAP is a safe and immunogenic vaccine regimen in children and infants with prior exposure to malaria. But no significant protective efficacy was observed in this very highly malaria-endemic setting., Trial Registration: ClinicalTrials.gov NCT01635647. Pactr.org PACTR201208000404131., Competing Interests: AVSH is a named inventor on patent applications and issued patents relating to malaria vectored vaccines and immunization regimes. This does not alter the author’s adherence to all the PLOS ONE policies on sharing data and materials.
- Published
- 2018
- Full Text
- View/download PDF
24. 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.
- Author
-
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, and Lindsay SW
- Subjects
- Animals, Anopheles, Burkina Faso epidemiology, Child, Preschool, Female, Humans, Infant, Insect Vectors, Malaria, Falciparum epidemiology, Male, Insecticide-Treated Bednets, Insecticides, Malaria, Falciparum prevention & control, Permethrin, Pyridines
- Abstract
Background: Substantial reductions in malaria incidence in sub-Saharan Africa have been achieved with massive deployment of long-lasting insecticidal nets (LLINs), but pyrethroid resistance threatens control. Burkina Faso is an area with intense malaria transmission and highly pyrethroid-resistant vectors. We assessed the effectiveness of bednets containing permethrin, a pyrethroid, and pyriproxyfen, an insect growth regulator, versus permethrin-only (standard) LLINs against clinical malaria in children younger than 5 years in Banfora, Burkina Faso., Methods: In this two-group, step-wedge, cluster-randomised, controlled, superiority trial, standard LLINs were incrementally replaced with LLINs treated with permethrin plus pyriproxyfen (PPF) in 40 rural clusters in Burkina Faso. In each cluster, 50 children (aged 6 months to 5 years) were followed up by passive case detection for clinical malaria. Cross-sectional surveys were done at the start and the end of the transmission seasons in 2014 and 2015. We did monthly collections from indoor light traps to estimate vector densities. Primary endpoints were the incidence of clinical malaria, measured by passive case detection, and the entomological inoculation rate. Analyses were adjusted for clustering and for month and health centre. This trial is registered as ISRCTN21853394., Findings: 1980 children were enrolled in the cohort in 2014 and 2157 in 2015. At the end of the study, more than 99% of children slept under a bednet. The incidence of clinical malaria was 2·0 episodes per child-year in the standard LLIN group and 1·5 episodes per child-year in the PPF-treated LLIN group (incidence rate ratio 0·88 [95% CI 0·77-0·99; p=0·04]). The entomological inoculation rate was 85 (95% CI 63-108) infective bites per transmission season in the standard LLIN group versus 42 (32-52) infective bites per transmission season in the PPF-treated LLIN group (rate ratio 0·49, 95% CI 0·32-0·66; p<0·0001)., Interpretation: PPF-treated LLINs provide greater protection against clinical malaria than do standard LLINs and could be used as an alternative to standard LLINs in areas with intense transmission of Plasmodium falciparum malaria and highly pyrethroid-resistant vectors., Funding: EU Seventh Framework Programme., (Copyright © 2018 Elsevier Ltd. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
25. Safety of single low-dose primaquine in glucose-6-phosphate dehydrogenase deficient falciparum-infected African males: Two open-label, randomized, safety trials.
- Author
-
Bastiaens GJH, Tiono AB, Okebe J, Pett HE, Coulibaly SA, Gonçalves BP, Affara M, Ouédraogo A, Bougouma EC, Sanou GS, Nébié I, Bradley J, Lanke KHW, Niemi M, Sirima SB, d'Alessandro U, Bousema T, and Drakeley C
- Subjects
- Adult, Antimalarials adverse effects, Burkina Faso, Humans, Male, Primaquine adverse effects, Young Adult, Antimalarials administration & dosage, Glucosephosphate Dehydrogenase genetics, Malaria, Falciparum drug therapy, Primaquine administration & dosage
- Abstract
Background: Primaquine (PQ) actively clears mature Plasmodium falciparum gametocytes but in glucose-6-phosphate dehydrogenase deficient (G6PDd) individuals can cause hemolysis. We assessed the safety of low-dose PQ in combination with artemether-lumefantrine (AL) or dihydroartemisinin-piperaquine (DP) in G6PDd African males with asymptomatic P. falciparum malaria., Methods and Findings: In Burkina Faso, G6PDd adult males were randomized to treatment with AL alone (n = 10) or with PQ at 0.25 (n = 20) or 0.40 mg/kg (n = 20) dosage; G6PD-normal males received AL plus 0.25 (n = 10) or 0.40 mg/kg (n = 10) PQ. In The Gambia, G6PDd adult males and boys received DP alone (n = 10) or with 0.25 mg/kg PQ (n = 20); G6PD-normal males received DP plus 0.25 (n = 10) or 0.40 mg/kg (n = 10) PQ. The primary study endpoint was change in hemoglobin concentration during the 28-day follow-up. Cytochrome P-450 isoenzyme 2D6 (CYP2D6) metabolizer status, gametocyte carriage, haptoglobin, lactate dehydrogenase levels and reticulocyte counts were also determined. In Burkina Faso, the mean maximum absolute change in hemoglobin was -2.13 g/dL (95% confidence interval [CI], -2.78, -1.49) in G6PDd individuals randomized to 0.25 PQ mg/kg and -2.29 g/dL (95% CI, -2.79, -1.79) in those receiving 0.40 PQ mg/kg. In The Gambia, the mean maximum absolute change in hemoglobin concentration was -1.83 g/dL (95% CI, -2.19, -1.47) in G6PDd individuals receiving 0.25 PQ mg/kg. After adjustment for baseline concentrations, hemoglobin reductions in G6PDd individuals in Burkina Faso were more pronounced compared to those in G6PD-normal individuals receiving the same PQ doses (P = 0.062 and P = 0.022, respectively). Hemoglobin levels normalized during follow-up. Abnormal haptoglobin and lactate dehydrogenase levels provided additional evidence of mild transient hemolysis post-PQ., Conclusions: Single low-dose PQ in combination with AL and DP was associated with mild and transient reductions in hemoglobin. None of the study participants developed moderate or severe anemia; there were no severe adverse events. This indicates that single low-dose PQ is safe in G6PDd African males when used with artemisinin-based combination therapy., Trial Registration: Clinicaltrials.gov NCT02174900 Clinicaltrials.gov NCT02654730.
- Published
- 2018
- Full Text
- View/download PDF
26. Resistance to malaria through structural variation of red blood cell invasion receptors.
- Author
-
Leffler EM, Band G, Busby GBJ, Kivinen K, Le QS, Clarke GM, Bojang KA, Conway DJ, Jallow M, Sisay-Joof F, Bougouma EC, Mangano VD, Modiano D, Sirima SB, Achidi E, Apinjoh TO, Marsh K, Ndila CM, Peshu N, Williams TN, Drakeley C, Manjurano A, Reyburn H, Riley E, Kachala D, Molyneux M, Nyirongo V, Taylor T, Thornton N, Tilley L, Grimsley S, Drury E, Stalker J, Cornelius V, Hubbart C, Jeffreys AE, Rowlands K, Rockett KA, Spencer CCA, and Kwiatkowski DP
- Subjects
- Adult, Africa South of the Sahara, Child, DNA Copy Number Variations genetics, Gene Frequency, Genome, Human genetics, Humans, Protein Structure, Secondary, Receptors, Cell Surface chemistry, Receptors, Cell Surface genetics, Disease Resistance genetics, Erythrocytes parasitology, Glycophorins chemistry, Glycophorins genetics, Glycophorins metabolism, Host-Parasite Interactions genetics, Malaria, Falciparum genetics, Models, Molecular
- Abstract
The malaria parasite Plasmodium falciparum invades human red blood cells by a series of interactions between host and parasite surface proteins. By analyzing genome sequence data from human populations, including 1269 individuals from sub-Saharan Africa, we identify a diverse array of large copy-number variants affecting the host invasion receptor genes GYPA and GYPB We find that a nearby association with severe malaria is explained by a complex structural rearrangement involving the loss of GYPB and gain of two GYPB-A hybrid genes, which encode a serologically distinct blood group antigen known as Dantu. This variant reduces the risk of severe malaria by 40% and has recently increased in frequency in parts of Kenya, yet it appears to be absent from west Africa. These findings link structural variation of red blood cell invasion receptors with natural resistance to severe malaria., (Copyright © 2017, American Association for the Advancement of Science.)
- Published
- 2017
- Full Text
- View/download PDF
27. Characterisation of the opposing effects of G6PD deficiency on cerebral malaria and severe malarial anaemia.
- Author
-
Clarke GM, Rockett K, Kivinen K, Hubbart C, Jeffreys AE, Rowlands K, Jallow M, Conway DJ, Bojang KA, Pinder M, Usen S, Sisay-Joof F, Sirugo G, Toure O, Thera MA, Konate S, Sissoko S, Niangaly A, Poudiougou B, Mangano VD, Bougouma EC, Sirima SB, Modiano D, Amenga-Etego LN, Ghansah A, Koram KA, Wilson MD, Enimil A, Evans J, Amodu OK, Olaniyan S, Apinjoh T, Mugri R, Ndi A, Ndila CM, Uyoga S, Macharia A, Peshu N, Williams TN, Manjurano A, Sepúlveda N, Clark TG, Riley E, Drakeley C, Reyburn H, Nyirongo V, Kachala D, Molyneux M, Dunstan SJ, Phu NH, Quyen NN, Thai CQ, Hien TT, Manning L, Laman M, Siba P, Karunajeewa H, Allen S, Allen A, Davis TM, Michon P, Mueller I, Molloy SF, Campino S, Kerasidou A, Cornelius VJ, Hart L, Shah SS, Band G, Spencer CC, Agbenyega T, Achidi E, Doumbo OK, Farrar J, Marsh K, Taylor T, and Kwiatkowski DP
- Subjects
- Alleles, Anemia pathology, Case-Control Studies, Glucosephosphate Dehydrogenase genetics, Humans, Malaria, Cerebral pathology, Malaria, Falciparum pathology, Risk Assessment, Anemia epidemiology, Glucosephosphate Dehydrogenase Deficiency complications, Malaria, Cerebral epidemiology, Malaria, Falciparum epidemiology
- Abstract
Glucose-6-phosphate dehydrogenase (G6PD) deficiency is believed to confer protection against Plasmodium falciparum malaria, but the precise nature of the protective effecthas proved difficult to define as G6PD deficiency has multiple allelic variants with different effects in males and females, and it has heterogeneous effects on the clinical outcome of P. falciparum infection. Here we report an analysis of multiple allelic forms of G6PD deficiency in a large multi-centre case-control study of severe malaria, using the WHO classification of G6PD mutations to estimate each individual's level of enzyme activity from their genotype. Aggregated across all genotypes, we find that increasing levels of G6PD deficiency are associated with decreasing risk of cerebral malaria, but with increased risk of severe malarial anaemia. Models of balancing selection based on these findings indicate that an evolutionary trade-off between different clinical outcomes of P. falciparum infection could have been a major cause of the high levels of G6PD polymorphism seen in human populations., Competing Interests: JF: Director of the Wellcome Trust, one of the three founding funders of eLife. The other authors declare that no competing interests exist.
- Published
- 2017
- Full Text
- View/download PDF
28. Single low dose primaquine to reduce gametocyte carriage and Plasmodium falciparum transmission after artemether-lumefantrine in children with asymptomatic infection: a randomised, double-blind, placebo-controlled trial.
- Author
-
Gonçalves BP, Tiono AB, Ouédraogo A, Guelbéogo WM, Bradley J, Nebie I, Siaka D, Lanke K, Eziefula AC, Diarra A, Pett H, Bougouma EC, Sirima SB, Drakeley C, and Bousema T
- Subjects
- Artemether, Asymptomatic Infections, Child, Child, Preschool, Double-Blind Method, Drug Therapy, Combination, Female, Humans, Infant, Lumefantrine, Malaria, Falciparum epidemiology, Male, Plasmodium falciparum, Prevalence, Antimalarials administration & dosage, Artemisinins administration & dosage, Ethanolamines administration & dosage, Fluorenes administration & dosage, Malaria, Falciparum drug therapy, Primaquine administration & dosage
- Abstract
Background: A single low dose (0.25 mg/kg) of primaquine is recommended as a gametocytocide in combination with artemisinin-based combination therapies for Plasmodium falciparum but its effect on post-treatment gametocyte circulation and infectiousness to mosquitoes has not been quantified., Methods: In this randomised, double-blind, placebo-controlled trial, 360 asymptomatic parasitaemic children aged 2-15 years were enrolled and assigned to receive: artemether-lumefantrine (AL) and a dose of placebo; AL and a 0.25 mg/kg primaquine dose; or AL and a 0.40 mg/kg primaquine dose. On days 0, 2, 3, 7, 10 and 14, gametocytes were detected and quantified by microscopy, Pfs25 mRNA quantitative nucleic acid sequence based amplification (QT-NASBA), and quantitative reverse-transcriptase PCR (qRT-PCR). For a subset of participants, pre- and post-treatment infectiousness was assessed by mosquito feeding assays on days -1, 3, 7, 10 and 14., Results: Both primaquine arms had lower gametocyte prevalences after day 3 compared to the placebo arm, regardless of gametocyte detection method. The mean (95% confidence interval) number of days to gametocyte clearance in children with patent gametocytes on day 0 (N = 150) was 19.7 (14.6 - 24.8), 7.7 (6.3 - 9.1) and 8.2 (6.7 - 9.6) for the AL-placebo, the 0.25 mg/kg primaquine dose and the 0.40 mg/kg primaquine dose arms, respectively. While 38.0% (30/79) of selected gametocytaemic individuals were infectious before treatment, only 1/251 participant, from the AL-placebo group, infected mosquitoes after treatment., Conclusions: We observed similar gametocyte clearance rates after 0.25 and 0.40 mg/kg primaquine doses. Infectivity to mosquitoes after AL was very low and absent in primaquine arms. CLINICALTRIALS., Gov Registration: NCT01935882.
- Published
- 2016
- Full Text
- View/download PDF
29. Novel Insights Into the Protective Role of Hemoglobin S and C Against Plasmodium falciparum Parasitemia.
- Author
-
Mangano VD, Kabore Y, Bougouma EC, Verra F, Sepulveda N, Bisseye C, Santolamazza F, Avellino P, Tiono AB, Diarra A, Nebie I, Rockett KA, Sirima SB, and Modiano D
- Subjects
- Adolescent, Burkina Faso epidemiology, Child, Child, Preschool, Cross-Sectional Studies, Female, Genetic Predisposition to Disease, Genotype, Heterozygote, Humans, Malaria, Falciparum epidemiology, Male, Odds Ratio, Risk Factors, Sickle Cell Trait blood, Sickle Cell Trait epidemiology, Sickle Cell Trait genetics, Young Adult, Hemoglobin C, Hemoglobin, Sickle, Malaria, Falciparum blood, Parasitemia, Plasmodium falciparum
- Abstract
Although hemoglobin S (HbS) and hemoglobin C (HbC) are well known to protect against severe Plasmodium falciparum malaria, conclusive evidence on their role against infection has not yet been obtained. Here we show, in 2 populations from Burkina Faso (2007-2008), that HbS is associated with a 70% reduction of harboring P. falciparum parasitemia at the heterozygous state (odds ratio [OR] for AS vs AA, 0.27; 95% confidence interval [CI], .11-.66; P = .004). There is no evidence of protection for HbC in the heterozygous state (OR for AC vs AA, 1.49; 95% CI, .69-3.21; P = .31), whereas protection even higher than that observed with AS is observed in the homozygous and double heterozygous states (OR for CC + SC vs AA, 0.04; 95% CI, .01-.29; P = .002). The abnormal display of parasite-adhesive molecules on the surface of HbS and HbC infected erythrocytes, disrupting the pathogenic process of sequestration, might displace the parasite from the deep to the peripheral circulation, promoting its elimination at the spleen level., (© The Author 2015. Published by Oxford University Press on behalf of the Infectious Diseases Society of America.)
- Published
- 2015
- Full Text
- View/download PDF
30. Efficacy and safety of the mosquitocidal drug ivermectin to prevent malaria transmission after treatment: a double-blind, randomized, clinical trial.
- Author
-
Ouédraogo AL, Bastiaens GJ, Tiono AB, Guelbéogo WM, Kobylinski KC, Ouédraogo A, Barry A, Bougouma EC, Nebie I, Ouattara MS, Lanke KH, Fleckenstein L, Sauerwein RW, Slater HC, Churcher TS, Sirima SB, Drakeley C, and Bousema T
- Subjects
- Animals, Antimalarials therapeutic use, Artemether, Lumefantrine Drug Combination, Artemisinins therapeutic use, Double-Blind Method, Drug Combinations, Ethanolamines therapeutic use, Female, Fluorenes therapeutic use, Humans, Malaria, Falciparum drug therapy, Male, Culicidae, Insecticides therapeutic use, Ivermectin therapeutic use, Malaria, Falciparum prevention & control, Malaria, Falciparum transmission
- Abstract
Background: Artemisinin combination therapy effectively clears asexual malaria parasites and immature gametocytes but does not prevent posttreatment malaria transmission. Ivermectin (IVM) may reduce malaria transmission by killing mosquitoes that take blood meals from IVM-treated humans., Methods: In this double-blind, placebo-controlled trial, 120 asymptomatic Plasmodium falciparum parasite carriers were randomized to receive artemether-lumefantrine (AL) plus placebo or AL plus a single or repeated dose (200 µg/kg) of ivermectin (AL-IVM1 and AL-IVM2, respectively). Mosquito membrane feeding was performed 1, 3, and 7 days after initiation of treatment to determine Anopheles gambiae and Anopheles funestus survival and infection rates., Results: The AL-IVM combination was well tolerated. IVM resulted in a 4- to 7-fold increased mortality in mosquitoes feeding 1 day after IVM (P < .001). Day 7 IVM plasma levels were positively associated with body mass index (r = 0.57, P < .001) and were higher in female participants (P = .003), for whom An. gambiae mosquito mortality was increased until 7 days after a single dose of IVM (hazard rate ratio, 1.34 [95% confidence interval, 1.07-1.69]; P = .012). Although we found no evidence that IVM reduced Plasmodium infection rates among surviving mosquitoes, the mosquitocidal effect of AL-IVM1 and AL-IVM2 resulted in 27% and 35% reductions, respectively, in estimated malaria transmission potential during the first week after initiation of treatment., Conclusions: We conclude that IVM can be safely given in combination with AL and can reduce the likelihood of malaria transmission by reducing the life span of feeding mosquitoes., Clinical Trials Registration: NCT0160325., (© The Author 2014. Published by Oxford University Press on behalf of the Infectious Diseases Society of America. All rights reserved. For Permissions, please e-mail: journals.permissions@oup.com.)
- Published
- 2015
- Full Text
- View/download PDF
31. Is Fc gamma receptor IIA (FcγRIIA) polymorphism associated with clinical malaria and Plasmodium falciparum specific antibody levels in children from Burkina Faso?
- Author
-
Cherif MK, Sanou GS, Bougouma EC, Diarra A, Ouédraogo A, Dolo A, Troye-Blomberg M, Cavanagh DR, Theisen M, Modiano D, Sirima SB, and Nebié I
- Subjects
- Burkina Faso epidemiology, Child, Child, Preschool, Cohort Studies, Cross-Sectional Studies, Disease Susceptibility, Female, Humans, Malaria, Falciparum epidemiology, Malaria, Falciparum immunology, Male, Antibodies, Protozoan blood, Malaria, Falciparum genetics, Plasmodium falciparum immunology, Polymorphism, Genetic, Receptors, IgG genetics
- Abstract
In the present study, the influences of FcγRIIA polymorphism on susceptibility to malaria and antibody responses to Plasmodium falciparum antigens were analyzed in children. We recruited 96 healthy children between 3 and 10 years at the beginning of the high transmission season and we followed up for 5 months through the high transmission season to assess the parasitological, immunological and genetic endpoints in relation to clinical malaria status. There was a similar distribution of homozygous and heterozygous individuals carrying the FcγRIIA-131R/R and FcγRIIA-131R/H allele, whereas the number of FcγRIIA-131H/H homozygous individuals was lower. P. falciparum infection frequency was not associated with the FcγRIIa-131R/H polymorphism. Only IgG antibody responses to GLURP R0 showed a significant association between antibody levels and FcγRIIA polymorphism (p=0.02). IgG levels to MSP2a were significantly higher in children who did not experience any clinical malaria episode compared to those who experienced at least one malaria episode (p=0.019). Cytophilic and non-cytophylic IgG subclass levels were higher in children without malaria than those who experienced at least one malaria episode. This difference was statistically significant for IgG1 to MSP3 (p=0.003) and to MSP2a (p=0.006); IgG3 to MSP2a (p=0.007) and to GLURP R0 (p=0.044); IgG2 to MSP2b (p=0.007) and IgG4 to MSP3 (p=0.051) and to MSP2a (p=0.049). In this study, homozygous carriers of the FcγRIIA-131R/R allele had higher malaria-specific antibody levels compare to the heterozygous carriers FcγRIIA-131R/H alleles and to homozygous carriers of FcγRIIA-131H/H alleles. The pre-existing antibodies responses were related to a reduced subsequent risk of clinical malaria., (Copyright © 2014 Elsevier B.V. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
32. Clinical Variation of Plasmodium falciparum eba-175, ama-1, and msp-3 Genotypes in Young Children Living in a Seasonally High Malaria Transmission Setting in Burkina Faso.
- Author
-
Soulama I, Sermé SS, Bougouma EC, Diarra A, Tiono AB, Ouedraogo A, Konate AT, Nebie I, and Sirima SB
- Abstract
The association between P. falciparum eba-175, ama-1, and msp-3 polymorphism in the pathogenicity of malaria disease was investigated. We therefore compared the prevalence of different alleles between symptomatic and asymptomatic malarial children under five years of age living in Burkina Faso. Blood filter papers were collected during the 2008 malaria transmission season from 228 symptomatic and 199 asymptomatic children under five years of age. All patients were living in the rural area of Saponé at about 50 km from Ouagadougou, the capital city of Burkina Faso. P. falciparum parasite DNA was extracted using QIAGEN kits and the alleles diversity was assessed by a nested PCR. PCR products were then digested by restriction enzymes based on already described polymorphic regions of the eba-175, ama-1, and msp-3 genes. The individual alleles eba-175_FCR3 and msp-3_K1 frequencies were statistically higher (p < 0.0001) in the asymptomatic group compared to the symptomatic ones. No statistically significant difference was noted in the prevalence of ama-1-3D7, ama-1-K1, and ama-1-HB3 genotypes between the two groups (p > 0.05). The comparative analysis of P. falciparum genotypes indicated that the polymorphism in eba-175 and msp-3 genotypes varied between asymptomatic and symptomatic clinical groups and may contribute to the pathogenesis of malaria.
- Published
- 2015
- Full Text
- View/download PDF
33. A phase 1b randomized, controlled, double-blinded dosage-escalation trial to evaluate the safety, reactogenicity and immunogenicity of an adenovirus type 35 based circumsporozoite malaria vaccine in Burkinabe healthy adults 18 to 45 years of age.
- Author
-
Ouédraogo A, Tiono AB, Kargougou D, Yaro JB, Ouédraogo E, Kaboré Y, Kangoye D, Bougouma EC, Gansane A, Henri N, Diarra A, Sanon S, Soulama I, Konate AT, Watson NL, Brown V, Hendriks J, Pau MG, Versteege I, Wiesken E, Sadoff J, Nebie I, and Sirima SB
- Subjects
- Adolescent, Adult, Burkina Faso, Dose-Response Relationship, Immunologic, Double-Blind Method, Female, Humans, Malaria, Falciparum genetics, Male, Middle Aged, Plasmodium falciparum genetics, Protozoan Proteins genetics, Adenoviridae, Immunization, Secondary, Malaria, Falciparum immunology, Malaria, Falciparum prevention & control, Plasmodium falciparum immunology, Protozoan Proteins immunology
- Abstract
Background: Ad35.CS.01 is a pre-erythrocytic malaria candidate vaccine. It is a codon optimized nucleotide sequence representing the P. falciparum circumsporozoite (CS) surface antigen inserted in a replication deficient Adenovirus 35 backbone. A Phase 1a trial has been conducted in the USA in naïve adults and showed that the vaccine was safe. The aim of this study is to assess the safety and immunogenicity of ascending dosages in sub Saharan Africa., Methods: A double blind, randomized, controlled, dose escalation, phase Ib trial was conducted in a rural area of Balonghin, the Saponé health district (Burkina Faso). Forty-eight healthy adults aged 18-45 years were randomized into 4 cohorts of 12 to receive three vaccine doses (day 0, 28 and 84) of 10(9), 10(10), 5X10(10), 10(11) vp of Ad35.CS.01 or normal saline by intra muscular injection. Subjects were monitored carefully during the 14 days following each vaccination for non serious adverse events. Severe and serious adverse events were collected throughout the participant study duration (12 months from the first vaccination). Humoral and cellular immune responses were measured on study days 0, 28, 56, 84, 112 and 140., Results: Of the forty-eight subjects enrolled, forty-four (91.7%) received all three scheduled vaccine doses. Local reactions, all of mild severity, occurred in thirteen (27.1%) subjects. Severe (grade 3) laboratory abnormalities occurred in five (10.4%) subjects. One serious adverse event was reported and attributed to infection judged unrelated to vaccine. The vaccine induced both antibody titers and CD8 T cells producing IFNγ and TNFα with specificity to CS while eliciting modest neutralizing antibody responses against Ad35., Conclusion: Study vaccine Ad35.CS.01 at four different dose levels was well-tolerated and modestly immunogenic in this population. These results suggest that Ad35.CS.01 should be further investigated for preliminary efficacy in human challenge models and as part of heterologous prime-boost vaccination strategies., Trial Registration: ClinicalTrials.gov NCT01018459 http://clinicaltrials.gov/ct2/show/NCT01018459.
- Published
- 2013
- Full Text
- View/download PDF
34. Malaria morbidity in high and seasonal malaria transmission area of Burkina Faso.
- Author
-
Ouédraogo A, Tiono AB, Diarra A, Sanon S, Yaro JB, Ouedraogo E, Bougouma EC, Soulama I, Gansané A, Ouedraogo A, Konate AT, Nebie I, Watson NL, Sanza M, Dube TJ, and Sirima SB
- Subjects
- Burkina Faso epidemiology, Child, Preschool, Cohort Studies, Cross-Sectional Studies, Endemic Diseases, Female, Humans, Incidence, Infant, Infant, Newborn, Malaria parasitology, Malaria transmission, Malaria, Falciparum epidemiology, Male, Morbidity, Parasite Load, Parasitemia epidemiology, Parasitemia parasitology, Prevalence, Prospective Studies, Seasons, Malaria epidemiology
- Abstract
Background: Malariometric parameters are often primary endpoints of efficacy trials of malaria vaccine candidates. This study aims to describe the epidemiology of malaria prior to the conduct of a series of drug and vaccine trials in a rural area of Burkina Faso., Methods: Malaria incidence was prospectively evaluated over one year follow-up among two cohorts of children aged 0-5 years living in the Saponé health district. The parents of 1089 children comprising a passive case detection cohort were encouraged to seek care from the local health clinic at any time their child felt sick. Among this cohort, 555 children were randomly selected for inclusion in an active surveillance sub-cohort evaluated for clinical malaria during twice weekly home visits. Malaria prevalence was evaluated by cross-sectional survey during the low and high transmission seasons., Results: Number of episodes per child ranged from 0 to 6 per year. Cumulative incidence was 67.4% in the passive and 86.2% in the active cohort and was highest among children 0-1 years. Clinical malaria prevalence was 9.8% in the low and 13.0% in the high season (p>0.05). Median days to first malaria episode ranged from 187 (95% CI 180-193) among children 0-1 years to 228 (95% CI 212, 242) among children 4-5 years. The alternative parasite thresholds for the malaria case definition that achieved optimal sensitivity and specificity (70-80%) were 3150 parasites/µl in the high and 1350 parasites/µl in the low season., Conclusion: Clinical malaria burden was highest among the youngest age group children, who may represent the most appropriate target population for malaria vaccine candidate development. The pyrogenic threshold of parasitaemia varied markedly by season, suggesting a value for alternative parasitaemia levels in the malaria case defintion. Regional epidemiology of malaria described, Sapone area field centers are positioned for future conduct of malaria vaccine trials.
- Published
- 2013
- Full Text
- View/download PDF
35. FcγRIIa polymorphism and anti-malaria-specific IgG and IgG subclass responses in populations differing in susceptibility to malaria in Burkina Faso.
- Author
-
Cherif MK, Sanou GS, Maiga B, Israelsson E, Ouédraogo AL, Bougouma EC, Diarra A, Ouédraogo A, Ouattara AS, Troye-Blomberg M, Dolo A, Cavanagh DR, Theisen M, Modiano D, Sirima SB, and Nebié I
- Subjects
- Adult, Burkina Faso, Enzyme-Linked Immunosorbent Assay, Female, Genotype, Humans, Immunoglobulin G genetics, Malaria, Falciparum immunology, Male, Polymerase Chain Reaction, Genetic Predisposition to Disease genetics, Immunoglobulin G immunology, Malaria, Falciparum ethnology, Malaria, Falciparum genetics, Polymorphism, Single Nucleotide genetics, Receptors, IgG genetics
- Abstract
FcγRIIa is known to be polymorphic; and certain variants are associated with different susceptibilities to malaria. Studies involving the Fulani ethnic group reported an ethnic difference in FcγRIIa-R131H genotype frequencies between the Fulani and other sympatric groups. No previous studies have addressed these questions in Burkina Faso. This study aimed to assess the influence of FcγRIIa-R131H polymorphism on anti-falciparum malaria IgG and IgG subclass responses in the Fulani and the Mossi ethnic groups living in Burkina Faso. Healthy adults more than 20 years old belonging to the Mossi or the Fulani ethnic groups were enrolled for the assessment of selected parasitological, immunological and genetic variables in relation to their susceptibility to malaria. The prevalence of the Plasmodium falciparum infection frequency was relatively low in the Fulani ethnic group compared to the Mossi ethnic group. For all tested antigens, the Fulani had higher antibody levels than the Mossi group. In both ethnic groups, a similar distribution of FcγRIIa R131H polymorphism was found. Individuals with the R allele of FcγRIIa had higher antibody levels than those with the H allele. This study confirmed that malaria infection affected less the Fulani group than the Mossi group. FcγRIIa-R131H allele distribution is similar in both ethnic groups, and higher antibody levels are associated with the FcγRIIa R allele compared to the H allele., (© 2012 The Authors. Scandinavian Journal of Immunology © 2012 Blackwell Publishing Ltd.)
- Published
- 2012
- Full Text
- View/download PDF
36. Haemoglobin variants and Plasmodium falciparum malaria in children under five years of age living in a high and seasonal malaria transmission area of Burkina Faso.
- Author
-
Bougouma EC, Tiono AB, Ouédraogo A, Soulama I, Diarra A, Yaro JB, Ouédraogo E, Sanon S, Konaté AT, Nébié I, Watson NL, Sanza M, Dube TJ, and Sirima SB
- Subjects
- Body Temperature, Burkina Faso epidemiology, Child, Preschool, Cross-Sectional Studies, Female, Genotype, Hemoglobins classification, Humans, Incidence, Infant, Infant, Newborn, Malaria, Falciparum diagnosis, Male, Parasitemia diagnosis, Parasitemia epidemiology, Plasmodium falciparum isolation & purification, Prevalence, Genetic Predisposition to Disease, Hemoglobins genetics, Malaria, Falciparum epidemiology, Malaria, Falciparum genetics
- Abstract
Background: Genetic factors play a key role in determining resistance/susceptibility to infectious disease. Susceptibility of the human host to malaria infection has been reported to be influenced by genetic factors, which could be confounders if not taken into account in the assessment of the efficacy of interventions against malaria. This study aimed to assess the relationship between haemoglobin genotypes and malaria in children under five years in a site being characterized for future malaria vaccine trials., Methods: The study population consisted of 452 children living in four rural villages. Hb genotype was determined at enrolment. Clinical malaria incidence was evaluated over a one-year period using combined active and passive surveillance. Prevalence of infection was evaluated via bi-annual cross-sectional surveys. At each follow-up visit, children received a brief clinical examination and thick and thin blood films were prepared for malaria diagnosis. A clinical malaria was defined as Plasmodium falciparum parasitaemia >2,500 parasites/μl and axillary temperature ≥37.5°C or reported fever over the previous 24 hours., Results: Frequencies of Hb genotypes were 73.2% AA; 15.0% AC; 8.2% AS; 2.2% CC; 1.1% CS and 0.2% SS. Prevalence of infection at enrolment ranged from 61.9%-54.1% among AA, AC and AS children. After one year follow-up, clinical malaria incidence (95% CI) (episodes per person-year) was 1.9 (1.7-2.0) in AA, 1.6 (1.4-2.1) in AC, and 1.7 (1.4-2.0) in AS children. AC genotype was associated with lower incidence of clinical malaria relative to AA genotype among children aged 1-2 years [rate ratio (95% CI) 0.66 (0.42-1.05)] and 2-3 years [rate ratio (95% CI) 0.37 (0.18-0.75)]; an association of opposite direction was however apparent among children aged 3-4 years. AS genotype was associated with lower incidence of clinical malaria relative to AA genotype among children aged 2-3 years [rate ratio (95% CI) 0.63 (0.40-1.01)]., Conclusions: In this cohort of children, AC or AS genotype was associated with lower risk of clinical malaria relative to AA genotype only among children aged one to three years. It would be advisable for clinical studies of malaria in endemic regions to consider haemoglobin gene differences as a potentially important confounder, particularly among younger children.
- Published
- 2012
- Full Text
- View/download PDF
37. Transplacental Transmission of Plasmodium falciparum in a Highly Malaria Endemic Area of Burkina Faso.
- Author
-
Ouédraogo A, Tiono AB, Diarra A, Bougouma EC, Nébié I, Konaté AT, and Sirima SB
- Abstract
Malaria congenital infection constitutes a major risk in malaria endemic areas. In this study, we report the prevalence of transplacental malaria in Burkina Faso. In labour and delivery units, thick and thin blood films were made from maternal, placental, and umbilical cord blood to determine malaria infection. A total of 1,309 mother/baby pairs were recruited. Eighteen cord blood samples (1.4%) contained malaria parasites (Plasmodium falciparum). Out of the 369 (28.2%) women with peripheral positive parasitemia, 211 (57.2%) had placental malaria and 14 (3.8%) had malaria parasites in their umbilical cord blood. The umbilical cord parasitemia levels were statistically associated with the presence of maternal peripheral parasitemia (OR = 9.24, P ≪ 0.001), placental parasitemia (OR = 10.74, P ≪ 0.001), high-density peripheral parasitemia (OR = 9.62, P ≪ 0.001), and high-density placental parasitemia (OR = 4.91, P = 0.03). In Burkina Faso, the mother-to-child transmission rate of malaria appears to be low.
- Published
- 2012
- Full Text
- View/download PDF
38. Genetic diversity of polymorphic vaccine candidate antigens (apical membrane antigen-1, merozoite surface protein-3, and erythrocyte binding antigen-175) in Plasmodium falciparum isolates from western and central Africa.
- Author
-
Soulama I, Bigoga JD, Ndiaye M, Bougouma EC, Quagraine J, Casimiro PN, Stedman TT, and Sirima SB
- Subjects
- Africa, Central, Africa, Western, Alleles, Antigens, Protozoan immunology, DNA, Protozoan genetics, Genetic Variation genetics, Heterozygote, Humans, Malaria, Falciparum genetics, Malaria, Falciparum parasitology, Malaria, Falciparum prevention & control, Plasmodium falciparum immunology, Polymerase Chain Reaction, Protozoan Proteins immunology, Antigens, Protozoan genetics, Malaria Vaccines genetics, Plasmodium falciparum genetics, Protozoan Proteins genetics
- Abstract
The malaria vaccine candidate antigens erythrocyte binding antigen 175 (EBA-175), merozoite surface protein 3 (MSP-3), and apical membrane antigen (AMA-1) from Plasmodium falciparum isolates from countries in central and west Africa were assessed for allelic diversity. Samples were collected on filter paper from 600 P. falciparum-infected symptomatic patients in Cameroon, Republic of Congo, Burkina Faso, Ghana, and Senegal and screened for class-specific amplification fragments. Genetic diversity, assessed by mean heterozygosity, was comparable among countries. We detected a clinical increase in eba 175 F-allele frequency from west to east across the study region. No statistical difference in msp-3 allele distribution between countries was observed. The ama-1 3D7 alleles were present at a lower frequency in central Africa than in West Africa. We also detected little to no genetic differentiation among sampling locations. This finding indicates that, at least at the level of resolution offered by restriction fragment length polymorphism analysis, these antigens showed remarkable genetic homogeneity throughout the region sampled, perhaps caused by balancing selection to maintain a diverse array of antigen haplotyes.
- Published
- 2011
- Full Text
- View/download PDF
39. Safety and immunogenicity of the malaria vaccine candidate MSP3 long synthetic peptide in 12-24 months-old Burkinabe children.
- Author
-
Sirima SB, Tiono AB, Ouédraogo A, Diarra A, Ouédraogo AL, Yaro JB, Ouédraogo E, Gansané A, Bougouma EC, Konaté AT, Kaboré Y, Traoré A, Chilengi R, Soulama I, Luty AJ, Druilhe P, Cousens S, and Nébié I
- Subjects
- Burkina Faso, Child, Preschool, Dose-Response Relationship, Drug, Double-Blind Method, Humans, Infant, Merozoites immunology, Peptides chemistry, Research Design, Time Factors, Treatment Outcome, Antigens, Protozoan genetics, Antigens, Protozoan metabolism, Malaria prevention & control, Malaria Vaccines therapeutic use, Plasmodium falciparum metabolism, Protozoan Proteins genetics, Protozoan Proteins metabolism
- Abstract
Background: A Phase Ia trial in European volunteers of the candidate vaccine merozoite surface protein 3 (MSP3), a Plasmodium falciparum blood stage membrane, showed that it induces biologically active antibodies able to achieve parasite killing in vitro, while a phase Ib trial in semi-immune adult volunteers in Burkina Faso confirmed that the vaccine was safe. The aim of this study was to assess the safety and immunogenicity of this vaccine candidate in children aged 12-24 months living in malaria endemic area of Burkina Faso., Methods: The study was a double-blind, randomized, controlled, dose escalation phase Ib trial, designed to assess the safety, reactogenicity and immunogenicity of three doses of either 15 or 30 microg of MSP3-LSP adsorbed on aluminum hydroxide in 45 children 12 to 24 months of age randomized into three equal groups. Each group received 3 vaccine doses (on days 0, 28 and 56) of either 15 microg of MSP3-LSP, 30 microg of MSP3-LSP or of the Engerix B hepatitis B vaccine. Children were visited at home daily for the 6 days following each vaccination to solicit symptoms which might be related to vaccination. Serious adverse events occurring during the study period (1 year) were recorded. Antibody responses to MSP3-LSP were measured on days 0, 28, 56 and 84., Results: All 45 enrolled children received three MSP3 vaccine doses. No serious adverse events were reported. Most of the adverse events reported were mild to moderate in severity. The only reported local symptoms with grade 3 severity were swelling and induration, with an apparently dose related response. All grade 3 adverse events resolved without any sequelae. Both MSP3 doses regimens were able to elicit high levels of anti-MSP3 specific IgG1 and IgG3 antibodies in the volunteers with very little or no increase in IgG2, IgG4 and IgM classes: i.e. vaccination induced predominantly the isotypes involved in the monocyte-dependent mechanism of P. falciparum parasite-killing., Conclusion: Our results support the promise of MSP3-LSP as a malaria vaccine candidate, both in terms of tolerability and of immunogenicity. Further assessment of the efficacy of this vaccine is recommended., Trial Registration: ClinicalTrials.gov NCT00452088.
- Published
- 2009
- Full Text
- View/download PDF
40. Placental malaria and low birth weight in pregnant women living in a rural area of Burkina Faso following the use of three preventive treatment regimens.
- Author
-
Tiono AB, Ouedraogo A, Bougouma EC, Diarra A, Konaté AT, Nébié I, and Sirima SB
- Subjects
- Adult, Animals, Burkina Faso, Chloroquine therapeutic use, Drug Combinations, Female, Fetal Blood parasitology, Humans, Infant, Newborn, Placenta parasitology, Plasmodium falciparum isolation & purification, Pregnancy, Pyrimethamine therapeutic use, Rural Population, Sulfadoxine therapeutic use, Treatment Outcome, Young Adult, Antimalarials therapeutic use, Chemoprevention methods, Infant, Low Birth Weight, Malaria, Falciparum complications, Malaria, Falciparum prevention & control, Placenta Diseases parasitology, Placenta Diseases prevention & control, Pregnancy Complications, Infectious prevention & control
- Abstract
Background: The weekly chemoprophylaxis of malaria during pregnancy with chloroquine (CQ) has become problematic with the increasing resistance of Plasmodium falciparum to this drug. There was a need to test the benefits of new strategies over the classical chemoprophylaxis. This study was conducted to provide data to the National Malarial Control Programme for an evidence-based policy change decision making process. It compares the efficacy of two IPT regimens, using chloroquine (CQ) or sulphadoxine/pyrimethamine (SP), with the classical chemoprophylaxis regimen using CQ in reducing the adverse outcomes of malaria infection, for the mother and the foetus., Methods: Pregnant women attending the first antenatal care visit were randomly assigned to one of the three treatment regimens. They were subsequently followed up till delivery. Maternal, placental and cord blood samples were obtained upon delivery to check for P. falciparum infection., Results: A total of 648 pregnant women were enrolled in the study. Delivery outcome were available for 423 of them. Peripheral maternal P. falciparum infection at delivery was found in 25.8% of the women. The proportion of women with maternal infection was significantly lower in the IPTp/SP group than in the CQ group (P << 0.000). The prevalence of placental malaria was 18.8% in the CWC/CQ group; 15.9% in the IPTp/CQ group and 10.6% in the IPTp/SP group. The incidence of LBW (weight < 2,500 g) was significantly higher among infants of mothers in the CWC/CQ group (23.9%) as compared with those of mothers in the IPTp/CQ (15.6%) and IPTp/SP (11.6%) groups (p = 0.02), Conclusion: Intermittent preventive treatment with SP has shown clear superiority in reducing adverse outcomes at delivery, as compared with intermittent preventive treatment with CQ and classical chemoprophylaxis with CQ.
- Published
- 2009
- Full Text
- View/download PDF
41. Plasmodium falciparum genotypes diversity in symptomatic malaria of children living in an urban and a rural setting in Burkina Faso.
- Author
-
Soulama I, Nébié I, Ouédraogo A, Gansane A, Diarra A, Tiono AB, Bougouma EC, Konaté AT, Kabré GB, Taylor WR, and Sirima SB
- Subjects
- Animals, Antigens, Protozoan genetics, Burkina Faso epidemiology, Child, Preschool, Female, Genotype, Humans, Infant, Malaria, Falciparum epidemiology, Male, Merozoite Surface Protein 1 genetics, Plasmodium falciparum isolation & purification, Polymerase Chain Reaction methods, Protozoan Proteins genetics, Rural Population, Urban Population, Genetic Variation, Malaria, Falciparum parasitology, Plasmodium falciparum classification, Plasmodium falciparum genetics
- Abstract
Background: The clinical presentation of malaria, considered as the result of a complex interaction between parasite and human genetics, is described to be different between rural and urban areas. The analysis of the Plasmodium falciparum genetic diversity in children with uncomplicated malaria, living in these two different areas, may help to understand the effect of urbanization on the distribution of P. falciparum genotypes., Methods: Isolates collected from 75 and 89 children with uncomplicated malaria infection living in a rural and an urban area of Burkina Faso, respectively, were analysed by a nested PCR amplification of msp1 and msp2 genes to compare P. falciparum diversity., Results: The K1 allelic family was widespread in children living in the two sites, compared to other msp1 allelic families (frequency >90%). The MAD 20 allelic family of msp1 was more prevalent (p = 0.0001) in the urban (85.3%) than the rural area (63.2%). In the urban area, the 3D7 alleles of msp2 were more prevalent compared to FC27 alleles, with a high frequency for the 3D7 300bp allele (>30%). The multiplicity of infection was in the range of one to six in the urban area and of one to seven in the rural area. There was no difference in the frequency of multiple infections (p = 0.6): 96.0% (95% C.I: 91.6-100) in urban versus 93.1% (95%C.I: 87.6-98.6) in rural areas. The complexity of infection increased with age [p = 0.04 (rural area), p = 0.06 (urban area)]., Conclusion: Urban-rural area differences were observed in some allelic families (MAD20, FC27, 3D7), suggesting a probable impact of urbanization on genetic variability of P. falciparum. This should be taken into account in the implementation of malaria control measures.
- Published
- 2009
- Full Text
- View/download PDF
42. [Comparative impact of three malaria preventive regimens during pregnancy on maternal anemia due to malaria in Burkina Faso].
- Author
-
Ouédraogo A, Bougouma EC, Diarra A, Konaté AT, Nébié I, Tiono AB, and Sirima SB
- Subjects
- Adult, Anemia epidemiology, Burkina Faso, Chloroquine therapeutic use, Drug Therapy, Combination, Female, Humans, Pregnancy, Prevalence, Pyrimethamine therapeutic use, Rural Population, Sulfadoxine therapeutic use, Anemia chemically induced, Antimalarials therapeutic use, Malaria prevention & control, Pregnancy Complications, Infectious prevention & control
- Abstract
Objectives: The main objective of this study was to compare the efficacy of three regimens of malaria prevention during pregnancy for the reduction of anemia between the first and third antenatal consultations. The first treatment arm was the classical weekly chemoprophylaxis with chloroquine; the other two were the intermittent preventive treatment using either three doses of chloroquine or sulfadoxine-pyrimethamine., Design: We conducted an open, randomized, three-arm study in a rural district of Burkina Faso. A cohort was constituted by 648 pregnant women of any parity., Results: The hemoglobin gain was more significant with the intermittent preventive treatment using sulfadoxine-pyrimethamine compared to the other treatment arms. The hemoglobin increased from 10.3g/dl (at the first antenatal consultation) to 11.4 g/dl (at the third antenatal consultation). In the three arms of treatment, the chemoprophylaxis reduced the prevalence of moderate anemia and severe anemia. The reduction of moderate anemia was more substantial in the sulfadoxine-pyrimethamine arm (65.6 to 36.7%) at second antenatal consultation (p=0.069) and third antenatal consultation (p=0.014). Conversely, in the two chloroquine arms, there was no significant reduction either at second antenatal consultation (p=0.72) or third antenatal consultation (p=0.55). The prevalence of peripheral parasitemia decreased in all treatment groups. However, it was significantly higher in the sulfadoxine-pyrimethamine group (44.3%)., Conclusions: Intermittent preventive treatment with three doses of sulfadoxine-pyrimethamine is a more effective strategy to prevent maternal anemia during pregnancy in Burkina Faso.
- Published
- 2008
- Full Text
- View/download PDF
43. Humoral responses to Plasmodium falciparum blood-stage antigens and association with incidence of clinical malaria in children living in an area of seasonal malaria transmission in Burkina Faso, West Africa.
- Author
-
Nebie I, Diarra A, Ouedraogo A, Soulama I, Bougouma EC, Tiono AB, Konate AT, Chilengi R, Theisen M, Dodoo D, Remarque E, Bosomprah S, Milligan P, and Sirima SB
- Subjects
- Adolescent, Animals, Burkina Faso epidemiology, Child, Child, Preschool, Humans, Immunoglobulin G blood, Immunoglobulin Isotypes blood, Immunoglobulin M blood, Incidence, Infant, Infant, Newborn, Antibodies, Protozoan blood, Antigens, Protozoan immunology, Malaria, Falciparum epidemiology, Malaria, Falciparum immunology, Plasmodium falciparum immunology
- Abstract
There is longstanding evidence that immunoglobulin G (IgG) has a role in protection against clinical malaria, and human antibodies of the cytophilic subclasses are thought to be particularly critical in this respect. In this cohort study, 286 Burkinabè children 6 months to 15 years old were kept under malaria surveillance in order to assess the protective role of antibody responses against four antigens which are currently being evaluated as vaccine candidates: apical membrane antigen 1 (AMA1), merozoite surface protein 1-19 (MSP1-19), MSP3, and glutamate-rich protein (GLURP). Total IgG, IgM, and IgG subclass responses were measured just before the malaria transmission season. The incidence of malaria was 2.4 episodes per child year of risk. After adjusting for the confounding effects of age, the level of total IgG to GLURP was strongly associated with reduced malaria incidence (incidence rate ratio associated with a doubling of total IgG, 0.79; 95% confidence interval, 0.66 to 0.94; P = 0.009.); there was a borderline statistically significant association between the level of total IgG to MSP3 and malaria incidence and no evidence of an association for total IgG to AMA1 and to MSP1-19. Of the IgG subclass responses studied, only IgG3 and IgG4 against GLURP and IgG1 against AMA1 were associated with reduced risk of clinical malaria. There was no evidence of an interaction between responses to AMA1 and baseline parasitemia in their effects on malaria incidence. Currently included in malaria vaccine formulations for clinical trials in humans, these blood-stage antigens, AMA1 and GLURP, offer good prospects for malaria vaccine development.
- Published
- 2008
- Full Text
- View/download PDF
44. Malaria prevention during pregnancy: assessing the disease burden one year after implementing a program of intermittent preventive treatment in Koupela District, Burkina Faso.
- Author
-
Sirima SB, Cotte AH, Konaté A, Moran AC, Asamoa K, Bougouma EC, Diarra A, Ouédraogo A, Parise ME, and Newman RD
- Subjects
- Adolescent, Adult, Bedding and Linens, Burkina Faso, Drug Combinations, Female, Humans, Infant, Low Birth Weight physiology, Infant, Newborn, Insecticides administration & dosage, Malaria epidemiology, Middle Aged, National Health Programs standards, Parasitemia drug therapy, Parasitemia epidemiology, Placenta parasitology, Pregnancy, Pregnancy Complications, Parasitic drug therapy, Pregnancy Complications, Parasitic epidemiology, Antimalarials administration & dosage, Malaria drug therapy, Malaria prevention & control, Parasitemia prevention & control, Pregnancy Complications, Parasitic prevention & control, Pyrimethamine administration & dosage, Sulfadoxine administration & dosage
- Abstract
The World Health Organization recommends that pregnant women in malaria-endemic areas receive >or= 2 doses of intermittent preventive treatment with sulfadoxine-pyrimethamine (IPTp/SP) in the second and third trimesters of pregnancy to prevent maternal anemia, placental parasitemia, and low birth weight (LBW). In 2001, a program evaluation in Koupéla District, Burkina Faso demonstrated that despite widespread use of chloroquine chemoprophylaxis, the burden of malaria during pregnancy remained high. In 2003, the Burkina Faso Ministry of Health piloted a program of IPTp/SP (three doses) and accelerated distribution of insecticide-treated nets (ITN) to pregnant women in Koupéla District. In 2004, a follow-up program evaluation was conducted. Coverage with >or= 1 doses of IPTp/SP was high among women attending antenatal clinics (ANCs) (96.2%) and delivery units (DUs) (93.5%); ITN ownership was moderately high (ANC = 53.9%, DU = 61.6%). In multivariate analysis, >or= 1 dose of IPTp/SP was associated with a significant reduction in the prevalence of peripheral parasitemia at ANCs (risk ratio [RR] = 0.49, P = 0.008), >or= 2 doses of IPTp/SP were associated with a reduction in the prevalence of placental parasitemia (RR = 0.56, P = 0.02), and three doses of IPTp/SP were associated with a reduced risk of LBW (RR = 0.51, P = 0.04). The proportions of women at ANCs with peripheral parasitemia and anemia were significantly lower in 2004 than in 2001 (RR = 0.53, P = 0.001 and RR = 0.78, P = 0.003, respectively). The proportions of women at DUs with peripheral and placental parasitemia were also significantly lower in 2004 than in 2001 (RR = 0.66, P < 0.0001 and RR = 0.71, P = 0.0002, respectively). These data suggest that a package of IPTp/SP and ITNs is effective in reducing the burden of malaria during pregnancy in Burkina Faso.
- Published
- 2006
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.