Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Amadou Niangaly is active.

Publication


Featured researches published by Amadou Niangaly.


Nature Genetics | 2014

Reappraisal of known malaria resistance loci in a large multicenter study

Kirk A. Rockett; Geraldine M. Clarke; Kathryn Fitzpatrick; Christina Hubbart; Anna Jeffreys; Kate Rowlands; Rachel Craik; Muminatou Jallow; David J. Conway; Kalifa Bojang; Margaret Pinder; Stanley Usen; Fatoumatta Sisay-Joof; Giorgio Sirugo; Ousmane Toure; Mahamadou A. Thera; Salimata Konate; Sibiry Sissoko; Amadou Niangaly; Belco Poudiougou; V. Mangano; Edith C. Bougouma; Sodiomon B. Sirima; David Modiano; Lucas Amenga-Etego; Anita Ghansah; Kwadwo A. Koram; Michael D. Wilson; Anthony Enimil; Jennifer L. Evans

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.


The New England Journal of Medicine | 2011

A Field Trial to Assess a Blood-Stage Malaria Vaccine

Mahamadou A. Thera; Ogobara K. Doumbo; Drissa Coulibaly; Matthew B. Laurens; Amed Ouattara; Abdoulaye K. Kone; Ando Guindo; Karim Traore; Idrissa Traore; Bourema Kouriba; Dapa A. Diallo; Issa Diarra; Modibo Daou; Amagana Dolo; Youssouf Tolo; Mahamadou S Sissoko; Amadou Niangaly; Mady Sissoko; Shannon Takala-Harrison; Kirsten E. Lyke; Yukun Wu; William C. Blackwelder; Olivier Godeaux; Johan Vekemans; Marie-Claude Dubois; W. Ripley Ballou; Joe Cohen; Darby Thompson; Tina Dube; Lorraine Soisson

BACKGROUND Blood-stage malaria vaccines are intended to prevent clinical disease. The malaria vaccine FMP2.1/AS02(A), a recombinant protein based on apical membrane antigen 1 (AMA1) from the 3D7 strain of Plasmodium falciparum, has previously been shown to have immunogenicity and acceptable safety in Malian adults and children. METHODS In a double-blind, randomized trial, we immunized 400 Malian children with either the malaria vaccine or a control (rabies) vaccine and followed them for 6 months. The primary end point was clinical malaria, defined as fever and at least 2500 parasites per cubic millimeter of blood. A secondary end point was clinical malaria caused by parasites with the AMA1 DNA sequence found in the vaccine strain. RESULTS The cumulative incidence of the primary end point was 48.4% in the malaria-vaccine group and 54.4% in the control group; efficacy against the primary end point was 17.4% (hazard ratio for the primary end point, 0.83; 95% confidence interval [CI], 0.63 to 1.09; P=0.18). Efficacy against the first and subsequent episodes of clinical malaria, as defined on the basis of various parasite-density thresholds, was approximately 20%. Efficacy against clinical malaria caused by parasites with AMA1 corresponding to that of the vaccine strain was 64.3% (hazard ratio, 0.36; 95% CI, 0.08 to 0.86; P=0.03). Local reactions and fever after vaccination were more frequent with the malaria vaccine. CONCLUSIONS On the basis of the primary end point, the malaria vaccine did not provide significant protection against clinical malaria, but on the basis of secondary results, it may have strain-specific efficacy. If this finding is confirmed, AMA1 might be useful in a multicomponent malaria vaccine. (Funded by the National Institute of Allergy and Infectious Diseases and others; ClinicalTrials.gov number, NCT00460525.).


Science Translational Medicine | 2009

Extreme Polymorphism in a Vaccine Antigen and Risk of Clinical Malaria: Implications for Vaccine Development

Shannon L. Takala; Drissa Coulibaly; Mahamadou A. Thera; Adrian H. Batchelor; Michael P. Cummings; Ananias A. Escalante; Amed Ouattara; Karim Traore; Amadou Niangaly; Abdoulaye Djimde; Ogobara K. Doumbo; Christopher V. Plowe

Children from Mali who are repeatedly infected with malaria are more likely to get sick if the parasite is highly variable, a possible obstacle to an effective vaccine. From the point of view of the parasite, malaria is a successful disease. Transmitted by the bite of infected mosquitoes, malaria sickens about 400 million people a year. Although drugs, insecticides, and bed nets can help, a vaccine would be the most effective way to fight malaria. But the malaria parasite is coated with ever-changing proteins, and it has proven difficult to develop an effective vaccine against these constantly moving targets. By examining the natural immune response to malaria infection in children from Mali, West Africa, Takala et al. have identified antibody targets in the parasite that can best protect children from infection, a boon in designing a vaccine to combat this quick-change artist. Vaccines against the malaria parasite, Plasmodium falciparum, are directed against proteins on its surface in order to prevent these proteins from initiating the invasion of host cells by the parasite. The main target protein for vaccines currently in development is the highly variable apical membrane antigen–1 (AMA-1). For 3 years, the authors of this study collected P. falciparum organisms that naturally infected 100 children, ages 3 months to 20 years, in a remote rural town. Because each child was repeatedly infected, the authors could determine which characteristics of the infecting parasite determined whether the child was protected from subsequent illness. Sequencing of the parasites’ AMA-1 genes revealed a startling amount of diversity; ~500 separate infections exhibited 214 unique combinations of altered nucleotides. The overall number of resulting amino acid changes in the AMA-1 protein from one infection to another determined whether the child would succumb to sickness with the second infection. That is, the child’s natural immunity from the first infection could only protect against illness in a subsequent infection if AMA-1 from the second infecting parasite was similar to that of the first. This seems like bad news for malaria vaccine development, but additional data suggest a new approach. The authors’ analysis points to a particular region of AMA-1 (domain I c1L) that is highly variable and is responsible for much of the parasite’s ability to escape control by the human immune system. Thus, a vaccine that elicits protective antibodies to the most common variations of this region might be effective. Understanding how natural immunity to P. falciparum can (and cannot) protect against malaria will help to harness our own immune systems for protection against this serious disease. Vaccines directed against the blood stages of Plasmodium falciparum malaria are intended to prevent the parasite from invading and replicating within host cells. No blood-stage malaria vaccine has shown clinical efficacy in humans. Most malaria vaccine antigens are parasite surface proteins that have evolved extensive genetic diversity, and this diversity could allow malaria parasites to escape vaccine-induced immunity. We examined the extent and within-host dynamics of genetic diversity in the blood-stage malaria vaccine antigen apical membrane antigen–1 in a longitudinal study in Mali. Two hundred and fourteen unique apical membrane antigen–1 haplotypes were identified among 506 human infections, and amino acid changes near a putative invasion machinery binding site were strongly associated with the development of clinical symptoms, suggesting that these residues may be important to consider in designing polyvalent apical membrane antigen–1 vaccines and in assessing vaccine efficacy in field trials. This extreme diversity may pose a serious obstacle to an effective polyvalent recombinant subunit apical membrane antigen–1 vaccine.


PLOS ONE | 2008

Safety and immunogenicity of an AMA1 malaria vaccine in Malian children: results of a phase 1 randomized controlled trial.

Mahamadou A. Thera; Ogobara K. Doumbo; Drissa Coulibaly; Dapa A. Diallo; Abdoulaye K. Kone; Ando Guindo; Karim Traore; Alassane Dicko; Issaka Sagara; Mahamadou S Sissoko; Mounirou Baby; Mady Sissoko; Issa Diarra; Amadou Niangaly; Amagana Dolo; Modibo Daou; Sory I. Diawara; D. Gray Heppner; V. Ann Stewart; Evelina Angov; Elke S. Bergmann-Leitner; David E. Lanar; Sheetij Dutta; Lorraine Soisson; Carter Diggs; Amanda Leach; Alex Owusu; Marie-Claude Dubois; Joe Cohen; Jason N. Nixon

Background The objective was to evaluate the safety, reactogenicity and immunogenicity of the AMA-1-based blood-stage malaria vaccine FMP2.1/AS02A in adults exposed to seasonal malaria. Methodology/Principal Findings A phase 1 double blind randomized controlled dose escalation trial was conducted in Bandiagara, Mali, West Africa, a rural town with intense seasonal transmission of Plasmodium falciparum malaria. The malaria vaccine FMP2.1/AS02A is a recombinant protein (FMP2.1) based on apical membrane antigen-1 (AMA-1) from the 3D7 clone of P. falciparum, adjuvanted with AS02A. The comparator vaccine was a cell-culture rabies virus vaccine (RabAvert). Sixty healthy, malaria-experienced adults aged 18–55 y were recruited into 2 cohorts and randomized to receive either a half dose or full dose of the malaria vaccine (FMP2.1 25 µg/AS02A 0.25 mL or FMP2.1 50 µg/AS02A 0.5 mL) or rabies vaccine given in 3 doses at 0, 1 and 2 mo, and were followed for 1 y. Solicited symptoms were assessed for 7 d and unsolicited symptoms for 30 d after each vaccination. Serious adverse events were assessed throughout the study. Titers of anti-AMA-1 antibodies were measured by ELISA and P. falciparum growth inhibition assays were performed on sera collected at pre- and post-vaccination time points. Transient local pain and swelling were common and more frequent in both malaria vaccine dosage groups than in the comparator group. Anti-AMA-1 antibodies increased significantly in both malaria vaccine groups, peaking at nearly 5-fold and more than 6-fold higher than baseline in the half-dose and full-dose groups, respectively. Conclusion/Significance The FMP2.1/AS02A vaccine had a good safety profile, was well-tolerated, and was highly immunogenic in malaria-exposed adults. This malaria vaccine is being evaluated in Phase 1 and 2 trials in children at this site. Trial Registration ClinicalTrials.gov NCT00308061


The Journal of Infectious Diseases | 2013

Molecular Basis of Allele-Specific Efficacy of a Blood-Stage Malaria Vaccine: Vaccine Development Implications

Amed Ouattara; Shannon Takala-Harrison; Mahamadou A. Thera; Drissa Coulibaly; Amadou Niangaly; Renion Saye; Youssouf Tolo; Sheetij Dutta; D. Gray Heppner; Lorraine Soisson; Carter Diggs; Johan Vekemans; Joe Cohen; William C. Blackwelder; Tina Dube; Matthew B. Laurens; Ogobara K. Doumbo; Christopher V. Plowe

The disappointing efficacy of blood-stage malaria vaccines may be explained in part by allele-specific immune responses that are directed against polymorphic epitopes on blood-stage antigens. FMP2.1/AS02(A), a blood-stage candidate vaccine based on apical membrane antigen 1 (AMA1) from the 3D7 strain of Plasmodium falciparum, had allele-specific efficacy against clinical malaria in a phase II trial in Malian children. We assessed the cross-protective efficacy of the malaria vaccine and inferred which polymorphic amino acid positions in AMA1 were the targets of protective allele-specific immune responses. FMP2.1/AS02(A) had the highest efficacy against AMA1 alleles that were identical to the 3D7 vaccine-type allele at 8 highly polymorphic amino acid positions in the cluster 1 loop (c1L) but differed from 3D7 elsewhere in the molecule. Comparison of the incidence of vaccine-type alleles before and after vaccination in the malaria vaccine and control groups and examination of the patterns of allele change at polymorphic positions in consecutive malaria episodes suggest that the highly polymorphic amino acid position 197 in c1L was the most critical determinant of allele-specific efficacy. These results indicate that a multivalent AMA1 vaccine with broad efficacy could include only a limited set of key alleles of this extremely polymorphic antigen.


The Journal of Infectious Diseases | 2010

Early interferon-gamma response against Plasmodium falciparum correlates with interethnic differences in susceptibility to parasitemia between sympatric Fulani and Dogon in Mali.

Matthew McCall; Joost Hopman; Modibo Daou; Boubacar Maiga; Victor Dara; Ivo Ploemen; Krystelle Nganou-Makamdop; Amadou Niangaly; Youssouf Tolo; Charles Arama; J. Teun Bousema; Jos W. M. van der Meer; Andre van der Ven; Marita Troye-Blomberg; Amagana Dolo; Ogobara K. Doumbo; Robert W. Sauerwein

INTRODUCTION Interethnic differences in susceptibility to malaria provide a unique opportunity to explore immunological correlates of protection. The Fulani of Sahelian Africa are known for their reduced susceptibility to Plasmodium falciparum, compared with surrounding tribes, yet the immunology underlying this is still poorly understood. METHODS AND RESULTS Here, we show that mononuclear cells from Fulani elicit >10-fold stronger interferon (IFN)-gamma production following a 24-h in vitro coincubation with asexual parasites than cells from sympatric Dogon. This response appears to be specific for P. falciparum among a panel of other human pathogens and is independent of the lower number of regulatory T cell counts present in Fulani. IFN-gamma responses in both tribes were inversely correlated with peripheral parasite density as quantified by nucleic acid sequenced-based amplification, but responses of Fulani remained significantly stronger than those of Dogon after adjustment for concurrent parasitemia, suggesting that hard-wired immunological differences underlie the observed protection. CONCLUSIONS These results underscore the value of early IFN-gamma responses to P. falciparum as a correlate of anti-parasite immunity, not only in this setting but also in the wider context of malaria, and support the development of malaria vaccines aimed at inducing such responses.


Malaria Journal | 2010

Lack of allele-specific efficacy of a bivalent AMA1 malaria vaccine

Amed Ouattara; Jianbing Mu; Shannon Takala-Harrison; Renion Saye; Issaka Sagara; Alassane Dicko; Amadou Niangaly; Junhui Duan; Ruth D. Ellis; Louis H. Miller; Xin-Zhuan Su; Christopher V. Plowe; Ogobara K. Doumbo

BackgroundExtensive genetic diversity in vaccine antigens may contribute to the lack of efficacy of blood stage malaria vaccines. Apical membrane antigen-1 (AMA1) is a leading blood stage malaria vaccine candidate with extreme diversity, potentially limiting its efficacy against infection and disease caused by Plasmodium falciparum parasites with diverse forms of AMA1.MethodsThree hundred Malian children participated in a Phase 2 clinical trial of a bivalent malaria vaccine that found no protective efficacy. The vaccine consists of recombinant AMA1 based on the 3D7 and FVO strains of P. falciparum adjuvanted with aluminum hydroxide (AMA1-C1). The gene encoding AMA1 was sequenced from P. falciparum infections experienced before and after immunization with the study vaccine or a control vaccine. Sequences of ama1 from infections in the malaria vaccine and control groups were compared with regard to similarity to the vaccine antigens using several measures of genetic diversity. Time to infection with parasites carrying AMA1 haplotypes similar to the vaccine strains with respect to immunologically important polymorphisms and the risk of infection with vaccine strain haplotypes were compared.ResultsBased on 62 polymorphic AMA1 residues, 186 unique ama1 haplotypes were identified among 315 ama1 sequences that were included in the analysis. Eight infections had ama1 sequences identical to 3D7 while none were identical to FVO. Several measures of genetic diversity showed that ama1 sequences in the malaria vaccine and control groups were comparable both at baseline and during follow up period. Pre- and post-immunization ama1 sequences in both groups all had a similar degree of genetic distance from FVO and 3D7 ama1. No differences were found in the time of first clinical episode or risk of infection with an AMA1 haplotype similar to 3D7 or FVO with respect to a limited set of immunologically important polymorphisms found in the cluster 1 loop of domain I of AMA1.ConclusionThis Phase 2 trial of a bivalent AMA1 malaria vaccine found no evidence of vaccine selection or strain-specific efficacy, suggesting that the extreme genetic diversity of AMA1 did not account for failure of the vaccine to provide protection.


Lancet Infectious Diseases | 2017

Safety and efficacy of PfSPZ Vaccine against Plasmodium falciparum via direct venous inoculation in healthy malaria-exposed adults in Mali: a randomised, double-blind phase 1 trial

Mahamadou S Sissoko; Sara A. Healy; Abdoulaye Katile; Freda Omaswa; Irfan Zaidi; Erin E. Gabriel; Bourama Kamaté; Yacouba Samake; Merepen A. Guindo; Amagana Dolo; Amadou Niangaly; Karamoko Niaré; Amatigue Zeguime; Kourane Sissoko; Hama Diallo; Ismaila Thera; Kelly Ding; Michael P. Fay; Elise O'Connell; Thomas B. Nutman; Sharon Wong-Madden; Tooba Murshedkar; Adam Ruben; Minglin Li; Yonas Abebe; Anita Manoj; Anusha Gunasekera; Sumana Chakravarty; B. Kim Lee Sim; Peter F. Billingsley

BACKGROUND Plasmodium falciparum sporozite (PfSPZ) Vaccine is a metabolically active, non-replicating, whole malaria sporozoite vaccine that has been reported to be safe and protective against P falciparum controlled human malaria infection in malaria-naive individuals. We aimed to assess the safety and protective efficacy of PfSPZ Vaccine against naturally acquired P falciparum in malaria-experienced adults in Mali. METHODS After an open-label dose-escalation study in a pilot safety cohort, we did a double-blind, randomised, placebo-controlled trial based in Donéguébougou and surrounding villages in Mali. We recruited 18-35-year-old healthy adults who were randomly assigned (1:1) in a double-blind manner, with stratification by village and block randomisation, to receive either five doses of 2·7 × 105 PfSPZ or normal saline at days 0, 28, 56, 84, and 140 during the dry season (January to July inclusive). Participants and investigators were masked to group assignments, which were unmasked at the final study visit, 6 months after receipt of the last vaccination. Participants received combined artemether and lumefantrine (four tablets, each containing 20 mg artemether and 120 mg lumefantrine, given twice per day over 3 days for a total of six doses) to eliminate P falciparum before the first and last vaccinations. We collected blood smears every 2 weeks and during any illness for 24 weeks after the fifth vaccination. The primary outcome was the safety and tolerability of the vaccine, assessed as local and systemic reactogenicity and adverse events. The sample size was calculated for the exploratory efficacy endpoint of time to first P falciparum infection beginning 28 days after the fifth vaccination. The safety analysis included all participants who received at least one dose of investigational product, whereas the efficacy analyses included only participants who received all five vaccinations. This trial is registered at ClinicalTrials.gov, number NCT01988636. FINDINGS Between Jan 18 and Feb 24, 2014, we enrolled 93 participants into the main study cohort with 46 participants assigned PfSPZ Vaccine and 47 assigned placebo, all of whom were evaluable for safety. We detected no significant differences in local or systemic adverse events or laboratory abnormalities between the PfSPZ Vaccine and placebo groups, and only grade 1 (mild) local or systemic adverse events occurred in both groups. The most common solicited systemic adverse event in the vaccine and placebo groups was headache (three [7%] people in the vaccine group vs four [9%] in the placebo group) followed by fatigue (one [2%] person in the placebo group), fever (one [2%] person in the placebo group), and myalgia (one [2%] person in each group). The exploratory efficacy analysis included 41 participants from the vaccine group and 40 from the placebo group. Of these participants, 37 (93%) from the placebo group and 27 (66%) from the vaccine group developed P falciparum infection. The hazard ratio for vaccine efficacy was 0·517 (95% CI 0·313-0·856) by time-to-infection analysis (log-rank p=0·01), and 0·712 (0·528-0·918) by proportional analysis (p=0·006). INTERPRETATION PfSPZ Vaccine was well tolerated and safe. PfSPZ Vaccine showed significant protection in African adults against P falciparum infection throughout an entire malaria season. FUNDING US National Institutes of Health Intramural Research Program, Sanaria.


PLOS ONE | 2012

Candidate Polymorphisms and Severe Malaria in a Malian Population

Ousmane Toure; Salimata Konate; Sibiri Sissoko; Amadou Niangaly; Abdoulaye Barry; Abdourahmane Sall; Elisabeth Diarra; Belco Poudiougou; Nuno Sepúlveda; Susana Campino; Kirk A. Rockett; Taane G. Clark; Mahamadou A. Thera; Ogobara K. Doumbo

Malaria is a major health burden in sub-Saharan African countries, including Mali. The disease is complex, with multiple genetic determinants influencing the observed variation in response to infection, progression, and severity. We assess the influence of sixty-four candidate loci, including the sickle cell polymorphism (HbS), on severe malaria in a case-control study consisting of over 900 individuals from Bamako, Mali. We confirm the known protective effects of the blood group O and the HbS AS genotype on life-threatening malaria. In addition, our analysis revealed a marginal susceptibility effect for the CD40 ligand (CD40L)+220C allele. The lack of statistical evidence for other candidates may demonstrate the need for large-scale genome-wide association studies in malaria to discover new polymorphisms. It also demonstrates the need for establishing the region-specific repertoire of functional variation in important genes, including the glucose-6-phosphatase deficiency gene, before embarking on focused genotyping.


PLOS ONE | 2013

Extended Safety, Immunogenicity and Efficacy of a Blood-Stage Malaria Vaccine in Malian Children: 24-Month Follow-Up of a Randomized, Double-Blinded Phase 2 Trial

Matthew B. Laurens; Mahamadou A. Thera; Drissa Coulibaly; Amed Ouattara; Abdoulaye K. Kone; Ando Guindo; Karim Traore; Idrissa Traore; Bourema Kouriba; Dapa A. Diallo; Issa Diarra; Modibo Daou; Amagana Dolo; Youssouf Tolo; Mahamadou S Sissoko; Amadou Niangaly; Mady Sissoko; Shannon Takala-Harrison; Kirsten E. Lyke; Yukun Wu; William C. Blackwelder; Olivier Godeaux; Johan Vekemans; Marie-Claude Dubois; W. Ripley Ballou; Joe Cohen; Tina Dube; Lorraine Soisson; Carter Diggs; Brent House

Background The FMP2.1/AS02A candidate malaria vaccine was tested in a Phase 2 study in Mali. Based on results from the first eight months of follow-up, the vaccine appeared well-tolerated and immunogenic. It had no significant efficacy based on the primary endpoint, clinical malaria, but marginal efficacy against clinical malaria in secondary analyses, and high allele-specific efficacy. Extended follow-up was conducted to evaluate extended safety, immunogenicity and efficacy. Methods A randomized, double-blinded trial of safety, immunogenicity and efficacy of the candidate Plasmodium falciparum apical membrane antigen 1 (AMA1) vaccine FMP2.1/AS02A was conducted in Bandiagara, Mali. Children aged 1–6 years were randomized in a 1∶1 ratio to receive FMP2.1/AS02A or control rabies vaccine on days 0, 30 and 60. Using active and passive surveillance, clinical malaria and adverse events as well as antibodies against P. falciparum AMA1 were monitored for 24 months after the first vaccination, spanning two malaria seasons. Findings 400 children were enrolled. Serious adverse events occurred in nine participants in the FMP2.1/AS02A group and three in the control group; none was considered related to study vaccination. After two years, anti-AMA1 immune responses remained significantly higher in the FMP2.1/AS02A group than in the control group. For the entire 24-month follow-up period, vaccine efficacy was 7.6% (p = 0.51) against first clinical malaria episodes and 9.9% (p = 0.19) against all malaria episodes. For the final 16-month follow-up period, vaccine efficacy was 0.9% (p = 0.98) against all malaria episodes. Allele-specific efficacy seen in the first malaria season did not extend into the second season of follow-up. Interpretation Allele-specific vaccine efficacy was not sustained in the second malaria season, despite continued high levels of anti-AMA1 antibodies. This study presents an opportunity to evaluate correlates of partial protection against clinical malaria that waned during the second malaria season. Trial Registration Clinicaltrials.gov NCT00460525 NCT00460525

Collaboration


Dive into the Amadou Niangaly's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Bourema Kouriba

University of the Sciences

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge