Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters

Country/Region as subject
Publication year range
1.
N Engl J Med ; 388(21): 1942-1955, 2023 May 25.
Article in English | MEDLINE | ID: mdl-37224196

ABSTRACT

BACKGROUND: An effective, affordable, multivalent meningococcal conjugate vaccine is needed to prevent epidemic meningitis in the African meningitis belt. Data on the safety and immunogenicity of NmCV-5, a pentavalent vaccine targeting the A, C, W, Y, and X serogroups, have been limited. METHODS: We conducted a phase 3, noninferiority trial involving healthy 2-to-29-year-olds in Mali and Gambia. Participants were randomly assigned in a 2:1 ratio to receive a single intramuscular dose of NmCV-5 or the quadrivalent vaccine MenACWY-D. Immunogenicity was assessed at day 28. The noninferiority of NmCV-5 to MenACWY-D was assessed on the basis of the difference in the percentage of participants with a seroresponse (defined as prespecified changes in titer; margin, lower limit of the 96% confidence interval [CI] above -10 percentage points) or geometric mean titer (GMT) ratios (margin, lower limit of the 98.98% CI >0.5). Serogroup X responses in the NmCV-5 group were compared with the lowest response among the MenACWY-D serogroups. Safety was also assessed. RESULTS: A total of 1800 participants received NmCV-5 or MenACWY-D. In the NmCV-5 group, the percentage of participants with a seroresponse ranged from 70.5% (95% CI, 67.8 to 73.2) for serogroup A to 98.5% (95% CI, 97.6 to 99.2) for serogroup W; the percentage with a serogroup X response was 97.2% (95% CI, 96.0 to 98.1). The overall difference between the two vaccines in seroresponse for the four shared serogroups ranged from 1.2 percentage points (96% CI, -0.3 to 3.1) for serogroup W to 20.5 percentage points (96% CI, 15.4 to 25.6) for serogroup A. The overall GMT ratios for the four shared serogroups ranged from 1.7 (98.98% CI, 1.5 to 1.9) for serogroup A to 2.8 (98.98% CI, 2.3 to 3.5) for serogroup C. The serogroup X component of the NmCV-5 vaccine generated seroresponses and GMTs that met the prespecified noninferiority criteria. The incidence of systemic adverse events was similar in the two groups (11.1% in the NmCV-5 group and 9.2% in the MenACWY-D group). CONCLUSIONS: For all four serotypes in common with the MenACWY-D vaccine, the NmCV-5 vaccine elicited immune responses that were noninferior to those elicited by the MenACWY-D vaccine. NmCV-5 also elicited immune responses to serogroup X. No safety concerns were evident. (Funded by the U.K. Foreign, Commonwealth, and Development Office and others; ClinicalTrials.gov number, NCT03964012.).


Subject(s)
Epidemics , Health Status , Meningitis , Meningococcal Vaccines , Vaccines, Conjugate , Humans , Gambia/epidemiology , Mali/epidemiology , Vaccines, Conjugate/administration & dosage , Vaccines, Conjugate/adverse effects , Vaccines, Conjugate/therapeutic use , Meningococcal Vaccines/administration & dosage , Meningococcal Vaccines/adverse effects , Meningococcal Vaccines/therapeutic use , Child, Preschool , Child , Adolescent , Young Adult , Adult , Immunogenicity, Vaccine , Injections, Intramuscular , Meningitis/epidemiology , Meningitis/prevention & control , Epidemics/prevention & control
2.
N Engl J Med ; 387(26): 2411-2424, 2022 12 29.
Article in English | MEDLINE | ID: mdl-36516078

ABSTRACT

BACKGROUND: Questions remain concerning the rapidity of immune responses and the durability and safety of vaccines used to prevent Zaire Ebola virus disease. METHODS: We conducted two randomized, placebo-controlled trials - one involving adults and one involving children - to evaluate the safety and immune responses of three vaccine regimens against Zaire Ebola virus disease: Ad26.ZEBOV followed by MVA-BN-Filo 56 days later (the Ad26-MVA group), rVSVΔG-ZEBOV-GP followed by placebo 56 days later (the rVSV group), and rVSVΔG-ZEBOV-GP followed by rVSVΔG-ZEBOV-GP 56 days later (the rVSV-booster group). The primary end point was antibody response at 12 months, defined as having both a 12-month antibody concentration of at least 200 enzyme-linked immunosorbent assay units (EU) per milliliter and an increase from baseline in the antibody concentration by at least a factor of 4. RESULTS: A total of 1400 adults and 1401 children underwent randomization. Among both adults and children, the incidence of injection-site reactions and symptoms (e.g., feverishness and headache) was higher in the week after receipt of the primary and second or booster vaccinations than after receipt of placebo but not at later time points. These events were largely low-grade. At month 12, a total of 41% of adults (titer, 401 EU per milliliter) and 78% of children (titer, 828 EU per milliliter) had a response in the Ad26-MVA group; 76% (titer, 992 EU per milliliter) and 87% (titer, 1415 EU per milliliter), respectively, had a response in the rVSV group; 81% (titer, 1037 EU per milliliter) and 93% (titer, 1745 EU per milliliter), respectively, had a response in the rVSV-booster group; and 3% (titer, 93 EU per milliliter) and 4% (titer, 67 EU per milliliter), respectively, had a response in the placebo group (P<0.001 for all comparisons of vaccine with placebo). In both adults and children, antibody responses with vaccine differed from those with placebo beginning on day 14. CONCLUSIONS: No safety concerns were identified in this trial. With all three vaccine regimens, immune responses were seen from day 14 through month 12. (Funded by the National Institutes of Health and others; PREVAC ClinicalTrials.gov number, NCT02876328; EudraCT numbers, 2017-001798-18 and 2017-001798-18/3rd; and Pan African Clinical Trials Registry number, PACTR201712002760250.).


Subject(s)
Ebola Vaccines , Ebolavirus , Hemorrhagic Fever, Ebola , Adult , Child , Humans , Antibodies, Viral , Democratic Republic of the Congo , Ebola Vaccines/therapeutic use , Hemorrhagic Fever, Ebola/prevention & control
3.
J Viral Hepat ; 28(12): 1683-1689, 2021 12.
Article in English | MEDLINE | ID: mdl-34467609

ABSTRACT

Hepatitis B virus (HBV) infection is the tenth leading cause of death worldwide. Mother-to-child transmission of HBV occurring mainly at delivery remains one of the most common routes of infection in developing countries. One of the main challenges concerning HBV in Africa is to implement a prevention policy aiming at interrupting the cycle of pseudo-vertical transmission of this infection. The aim of this study was to assess the implication of certain bacterial and viral factors in mother-to-child transmission of HBV. This prospective study was conducted on 165 pregnant women carriers of HBV surface antigen (HBsAg) and their 169 newborns who attended care at the Gynecology Department of the University Hospital of Cocody. Serological, molecular, and bacteriological analyses were performed on blood samples and vaginal secretions. Mean viral load (VL) was 4.5 ± 1.3 log10  IU/ml, while mean HBsAg titres were 3.5 ± 0.9 log10  IU/ml. HBV DNA was found in vaginal secretions in 13.3% of mothers and in the blood of 10.3% of the newborns. Six bacterial species were identified in the vaginal discharge of pregnant women during labour before delivery. Staphylococcus aureus and Enterococcus faecalis were the most frequent species found in 23.0% and 13.9% of cases. Mothers positive for vaginal HBV DNA displayed higher plasma HBV DNA loads than negative mothers (6.2 ± 1.6 log10  IU/ml vs. 4.3 ± 1.0 log10  IU/ml, p < .0001). In conclusion, our study showed that presence of HBV DNA in vaginal secretions and the presence of S. aureus could play a role in mother-to-child transmission of HBV. HBV DNA detection in vaginal discharge represents a promising biomarker to identify newborns at risk of perinatal persistent infection.


Subject(s)
Hepatitis B , Pregnancy Complications, Infectious , DNA, Viral , Female , Hepatitis B/epidemiology , Hepatitis B Surface Antigens , Hepatitis B e Antigens , Hepatitis B virus/genetics , Humans , Infant, Newborn , Infectious Disease Transmission, Vertical , Pregnancy , Prospective Studies , Staphylococcus aureus , Viral Load
4.
Clin Infect Dis ; 70(1): 59-66, 2020 01 01.
Article in English | MEDLINE | ID: mdl-30810160

ABSTRACT

BACKGROUND: Few studies describe the respiratory syncytial virus (RSV) burden in African populations, and most have utilized hospital-based surveillance. In Mali, no community-based studies exist of the incidence or epidemiology of RSV infection. This study provides the first estimates of RSV incidence in Mali. METHODS: In a cohort of infants enrolled in a clinical trial of maternal influenza vaccination, we estimate incidence of RSV-associated febrile illness in the first 6 months of life and identify risk factors for RSV infection and progression to severe disease. Infants (N = 1871) were followed from birth to 6 months of age and visited weekly to detect pneumonia and influenza-like illness. Baseline covariates were explored as risk factors for RSV febrile illness and RSV pneumonia or hospitalization. RESULTS: Incidence of RSV illness was estimated at 536.8 per 1000 person-years, and 86% (131/153) of RSV illness episodes were positive for RSV-B. RSV illness was most frequent in the fifth month of life and associated with having older mothers and with lower parity. The incidence of RSV-associated hospitalizations was 45.6 per 1000 person-years. Among infants with RSV illness, males were more likely to be hospitalized. The incidence of RSV pneumonia was 29 cases per 1000 person-years. CONCLUSIONS: In the first 6 months of life, Malian infants have a high incidence of RSV illness, primarily caused by RSV-B. Prevention of early RSV will require passive protection via maternal immunization in pregnancy. Mali is the first country where RSV-B has been identified as the dominant subtype, with potential implications for vaccine development.


Subject(s)
Respiratory Syncytial Virus Infections , Respiratory Syncytial Virus, Human , Female , Hospitalization , Humans , Incidence , Infant , Infant, Newborn , Male , Mali/epidemiology , Pregnancy , Respiratory Syncytial Virus Infections/epidemiology , Risk Factors
5.
J Infect Dis ; 218(4): 606-613, 2018 07 13.
Article in English | MEDLINE | ID: mdl-29659924

ABSTRACT

Background: Rotavirus vaccines given to infants are safe and efficacious. A booster dose of rotavirus vaccine could extend protection into the second year of life in low-resource countries. Methods: We conducted an open-label, individual-randomized trial in Bamako, Mali. We assigned 600 infants aged 9-11 months to receive measles vaccine (MV), yellow fever vaccine (YFV), and meningococcal A conjugate vaccine (MenAV) with or without pentavalent rotavirus vaccine (PRV). We assessed the noninferiority (defined as a difference of ≤10%) of seroconversion and seroresponse rates to MV, YFV, and MenAV. We compared the seroresponse to PRV. Results: Seroconversion to MV occurred in 255 of 261 PRV recipients (97.7%) and 246 of 252 control infants (97.6%; difference, 0.1% [95% confidence interval {CI}, -4.0%-4.2%]). Seroresponse to YFV occurred in 48.1% of PRV recipients (141 of 293), compared with 52.2% of controls (153 of 293; difference, -4.1% [95% CI, -12.2%-4.0%]). A 4-fold rise in meningococcus A bactericidal titer was observed in 273 of 292 PRV recipients (93.5%) and 276 of 293 controls (94.2%; difference, -0.7% [95% CI, -5.2%-3.8%]). Rises in geometric mean concentrations of immunoglobulin A and immunoglobulin G antibodies to rotavirus were higher among PRV recipients (118 [95% CI, 91-154] and 364 [95% CI, 294-450], respectively), compared with controls (68 [95% CI, 50-92] and 153 [95% CI, 114-207], respectively). Conclusions: PRV did not interfere with MV and MenAV; this study could not rule out interference with YFV. PRV increased serum rotavirus antibody levels. Clinical Trials Registration: NCT02286895.


Subject(s)
Immunization, Secondary , Measles Vaccine/administration & dosage , Meningococcal Vaccines/administration & dosage , Rotavirus Infections/prevention & control , Rotavirus Vaccines/administration & dosage , Yellow Fever Vaccine/administration & dosage , Antibodies, Bacterial/blood , Antibodies, Viral/analysis , Antibodies, Viral/blood , Drug Interactions , Female , Humans , Immunoglobulin A/analysis , Immunoglobulin G/blood , Infant , Male , Mali , Measles Vaccine/immunology , Meningococcal Vaccines/immunology , Rotavirus Vaccines/immunology , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/immunology , Vaccines, Conjugate/administration & dosage , Vaccines, Conjugate/immunology , Yellow Fever Vaccine/immunology
6.
Lancet ; 389(10068): 505-518, 2017 02 04.
Article in English | MEDLINE | ID: mdl-28017403

ABSTRACT

BACKGROUND: rVSV-ZEBOV is a recombinant, replication competent vesicular stomatitis virus-based candidate vaccine expressing a surface glycoprotein of Zaire Ebolavirus. We tested the effect of rVSV-ZEBOV in preventing Ebola virus disease in contacts and contacts of contacts of recently confirmed cases in Guinea, west Africa. METHODS: We did an open-label, cluster-randomised ring vaccination trial (Ebola ça Suffit!) in the communities of Conakry and eight surrounding prefectures in the Basse-Guinée region of Guinea, and in Tomkolili and Bombali in Sierra Leone. We assessed the efficacy of a single intramuscular dose of rVSV-ZEBOV (2×107 plaque-forming units administered in the deltoid muscle) in the prevention of laboratory confirmed Ebola virus disease. After confirmation of a case of Ebola virus disease, we definitively enumerated on a list a ring (cluster) of all their contacts and contacts of contacts including named contacts and contacts of contacts who were absent at the time of the trial team visit. The list was archived, then we randomly assigned clusters (1:1) to either immediate vaccination or delayed vaccination (21 days later) of all eligible individuals (eg, those aged ≥18 years and not pregnant, breastfeeding, or severely ill). An independent statistician generated the assignment sequence using block randomisation with randomly varying blocks, stratified by location (urban vs rural) and size of rings (≤20 individuals vs >20 individuals). Ebola response teams and laboratory workers were unaware of assignments. After a recommendation by an independent data and safety monitoring board, randomisation was stopped and immediate vaccination was also offered to children aged 6-17 years and all identified rings. The prespecified primary outcome was a laboratory confirmed case of Ebola virus disease with onset 10 days or more from randomisation. The primary analysis compared the incidence of Ebola virus disease in eligible and vaccinated individuals assigned to immediate vaccination versus eligible contacts and contacts of contacts assigned to delayed vaccination. This trial is registered with the Pan African Clinical Trials Registry, number PACTR201503001057193. FINDINGS: In the randomised part of the trial we identified 4539 contacts and contacts of contacts in 51 clusters randomly assigned to immediate vaccination (of whom 3232 were eligible, 2151 consented, and 2119 were immediately vaccinated) and 4557 contacts and contacts of contacts in 47 clusters randomly assigned to delayed vaccination (of whom 3096 were eligible, 2539 consented, and 2041 were vaccinated 21 days after randomisation). No cases of Ebola virus disease occurred 10 days or more after randomisation among randomly assigned contacts and contacts of contacts vaccinated in immediate clusters versus 16 cases (7 clusters affected) among all eligible individuals in delayed clusters. Vaccine efficacy was 100% (95% CI 68·9-100·0, p=0·0045), and the calculated intraclass correlation coefficient was 0·035. Additionally, we defined 19 non-randomised clusters in which we enumerated 2745 contacts and contacts of contacts, 2006 of whom were eligible and 1677 were immediately vaccinated, including 194 children. The evidence from all 117 clusters showed that no cases of Ebola virus disease occurred 10 days or more after randomisation among all immediately vaccinated contacts and contacts of contacts versus 23 cases (11 clusters affected) among all eligible contacts and contacts of contacts in delayed plus all eligible contacts and contacts of contacts never vaccinated in immediate clusters. The estimated vaccine efficacy here was 100% (95% CI 79·3-100·0, p=0·0033). 52% of contacts and contacts of contacts assigned to immediate vaccination and in non-randomised clusters received the vaccine immediately; vaccination protected both vaccinated and unvaccinated people in those clusters. 5837 individuals in total received the vaccine (5643 adults and 194 children), and all vaccinees were followed up for 84 days. 3149 (53·9%) of 5837 individuals reported at least one adverse event in the 14 days after vaccination; these were typically mild (87·5% of all 7211 adverse events). Headache (1832 [25·4%]), fatigue (1361 [18·9%]), and muscle pain (942 [13·1%]) were the most commonly reported adverse events in this period across all age groups. 80 serious adverse events were identified, of which two were judged to be related to vaccination (one febrile reaction and one anaphylaxis) and one possibly related (influenza-like illness); all three recovered without sequelae. INTERPRETATION: The results add weight to the interim assessment that rVSV-ZEBOV offers substantial protection against Ebola virus disease, with no cases among vaccinated individuals from day 10 after vaccination in both randomised and non-randomised clusters. FUNDING: WHO, UK Wellcome Trust, the UK Government through the Department of International Development, Médecins Sans Frontières, Norwegian Ministry of Foreign Affairs (through the Research Council of Norway's GLOBVAC programme), and the Canadian Government (through the Public Health Agency of Canada, Canadian Institutes of Health Research, International Development Research Centre and Department of Foreign Affairs, Trade and Development).


Subject(s)
Ebola Vaccines , Hemorrhagic Fever, Ebola/prevention & control , Adolescent , Adult , Child , Cluster Analysis , Contact Tracing , Ebolavirus , Female , Guinea , Hemorrhagic Fever, Ebola/diagnosis , Hemorrhagic Fever, Ebola/transmission , Humans , Male , Membrane Glycoproteins , Middle Aged , Treatment Outcome , Vesiculovirus , Young Adult
7.
Lancet ; 386(9996): 857-66, 2015 Aug 29.
Article in English | MEDLINE | ID: mdl-26248676

ABSTRACT

BACKGROUND: A recombinant, replication-competent vesicular stomatitis virus-based vaccine expressing a surface glycoprotein of Zaire Ebolavirus (rVSV-ZEBOV) is a promising Ebola vaccine candidate. We report the results of an interim analysis of a trial of rVSV-ZEBOV in Guinea, west Africa. METHODS: For this open-label, cluster-randomised ring vaccination trial, suspected cases of Ebola virus disease in Basse-Guinée (Guinea, west Africa) were independently ascertained by Ebola response teams as part of a national surveillance system. After laboratory confirmation of a new case, clusters of all contacts and contacts of contacts were defined and randomly allocated 1:1 to immediate vaccination or delayed (21 days later) vaccination with rVSV-ZEBOV (one dose of 2 × 10(7) plaque-forming units, administered intramuscularly in the deltoid muscle). Adults (age ≥18 years) who were not pregnant or breastfeeding were eligible for vaccination. Block randomisation was used, with randomly varying blocks, stratified by location (urban vs rural) and size of rings (≤20 vs >20 individuals). The study is open label and masking of participants and field teams to the time of vaccination is not possible, but Ebola response teams and laboratory workers were unaware of allocation to immediate or delayed vaccination. Taking into account the incubation period of the virus of about 10 days, the prespecified primary outcome was laboratory-confirmed Ebola virus disease with onset of symptoms at least 10 days after randomisation. The primary analysis was per protocol and compared the incidence of Ebola virus disease in eligible and vaccinated individuals in immediate vaccination clusters with the incidence in eligible individuals in delayed vaccination clusters. This trial is registered with the Pan African Clinical Trials Registry, number PACTR201503001057193. FINDINGS: Between April 1, 2015, and July 20, 2015, 90 clusters, with a total population of 7651 people were included in the planned interim analysis. 48 of these clusters (4123 people) were randomly assigned to immediate vaccination with rVSV-ZEBOV, and 42 clusters (3528 people) were randomly assigned to delayed vaccination with rVSV-ZEBOV. In the immediate vaccination group, there were no cases of Ebola virus disease with symptom onset at least 10 days after randomisation, whereas in the delayed vaccination group there were 16 cases of Ebola virus disease from seven clusters, showing a vaccine efficacy of 100% (95% CI 74·7-100·0; p=0·0036). No new cases of Ebola virus disease were diagnosed in vaccinees from the immediate or delayed groups from 6 days post-vaccination. At the cluster level, with the inclusion of all eligible adults, vaccine effectiveness was 75·1% (95% CI -7·1 to 94·2; p=0·1791), and 76·3% (95% CI -15·5 to 95·1; p=0·3351) with the inclusion of everyone (eligible or not eligible for vaccination). 43 serious adverse events were reported; one serious adverse event was judged to be causally related to vaccination (a febrile episode in a vaccinated participant, which resolved without sequelae). Assessment of serious adverse events is ongoing. INTERPRETATION: The results of this interim analysis indicate that rVSV-ZEBOV might be highly efficacious and safe in preventing Ebola virus disease, and is most likely effective at the population level when delivered during an Ebola virus disease outbreak via a ring vaccination strategy. FUNDING: WHO, with support from the Wellcome Trust (UK); Médecins Sans Frontières; the Norwegian Ministry of Foreign Affairs through the Research Council of Norway; and the Canadian Government through the Public Health Agency of Canada, Canadian Institutes of Health Research, International Development Research Centre, and Department of Foreign Affairs, Trade and Development.


Subject(s)
Ebola Vaccines , Hemorrhagic Fever, Ebola/prevention & control , Adult , Ebolavirus/immunology , Female , Genetic Vectors , Guinea/epidemiology , Hemorrhagic Fever, Ebola/epidemiology , Humans , Incidence , Kaplan-Meier Estimate , Male , Membrane Glycoproteins/metabolism , Middle Aged , Vaccination/methods , Vesiculovirus/metabolism , Young Adult
8.
Clin Infect Dis ; 61 Suppl 5: S507-13, 2015 Nov 15.
Article in English | MEDLINE | ID: mdl-26553682

ABSTRACT

BACKGROUND: A safe, affordable, and highly immunogenic meningococcal A conjugate vaccine (PsA-TT, MenAfriVac) was developed to control epidemic group A meningitis in Africa. Documentation of the safety specifications of the PsA-TT vaccine was warranted, with sufficient exposure to detect potential rare vaccine-related adverse reactions. METHODS: This phase 3, double-blind, randomized, active controlled clinical study was designed to evaluate the safety--primarily vaccine-related serious adverse events (SAEs)--up to 3 months after administration of a single dose of the PsA-TT vaccine to subjects aged 1-29 years in Mali. Safety outcomes were also compared to those following a single dose of a licensed meningococcal ACWY polysaccharide vaccine (PsACWY). RESULTS: No vaccine-related SAEs occurred during the 3 months of follow-up of 4004 subjects vaccinated with a single dose of PsA-TT. When compared to PsACWY (1996 subjects), tenderness at the injection site appeared to be more frequent in the PsA-TT group. However, rates of local induration, systemic reactions, adverse events (AEs), and SAEs were similar in both groups, and unsolicited AEs and SAEs were all unrelated to the study vaccines. CONCLUSIONS: The study confirmed on a large scale the excellent safety profile of a single dose of PsA-TT when administered to its entire target population of 1-29 years of age. CLINICAL TRIALS REGISTRATION: PACTR ATMR201003000191317.


Subject(s)
Drug-Related Side Effects and Adverse Reactions/epidemiology , Drug-Related Side Effects and Adverse Reactions/pathology , Meningococcal Vaccines/administration & dosage , Meningococcal Vaccines/adverse effects , Adolescent , Adult , Child , Child, Preschool , Double-Blind Method , Female , Humans , Infant , Male , Mali/epidemiology , Meningitis, Meningococcal/epidemiology , Meningitis, Meningococcal/microbiology , Meningitis, Meningococcal/prevention & control , Neisseria meningitidis, Serogroup A/immunology , Young Adult
9.
Clin Infect Dis ; 61 Suppl 5: S514-20, 2015 Nov 15.
Article in English | MEDLINE | ID: mdl-26553683

ABSTRACT

BACKGROUND: Following mass vaccination campaigns in the African meningitis belt with group A meningococcal conjugate vaccine, MenAfriVac (PsA-TT), disease due to group A meningococci has nearly disappeared. Antibody persistence in healthy African toddlers was investigated. METHODS: African children vaccinated at 12-23 months of age with PsA-TT were followed for evaluation of antibody persistence up to 5 years after primary vaccination. Antibody persistence was evaluated by measuring group A serum bactericidal antibody (SBA) with rabbit complement and by a group A-specific IgG enzyme-linked immunosorbent assay (ELISA). RESULTS: Group A antibodies measured by SBA and ELISA were shown to decline in the year following vaccination and plateaued at levels significantly above baseline for up to 5 years following primary vaccination. CONCLUSIONS: A single dose of PsA-TT induces long-term sustained levels of group A meningococcal antibodies for up to 5 years after vaccination. CLINICAL TRIALS REGISTRATION: ISRTCN78147026.


Subject(s)
Antibodies, Bacterial/blood , Blood Bactericidal Activity , Meningococcal Vaccines/administration & dosage , Meningococcal Vaccines/immunology , Africa , Animals , Complement System Proteins , Enzyme-Linked Immunosorbent Assay , Female , Follow-Up Studies , Humans , Immunoglobulin G/blood , Infant , Male , Rabbits , Time Factors
10.
J Infect Dev Ctries ; 18(5): 679-686, 2024 May 30.
Article in English | MEDLINE | ID: mdl-38865396

ABSTRACT

INTRODUCTION: Few studies on SARS-CoV-2 seroprevalence in Ivory Coast have been conducted since the first case was reported on March 11, 2020. The main objective of this study was to observe and better understand the circulation of SARS-CoV-2 in Abidjan. METHODOLOGY: This prospective study collected data concerning age, sex, vaccination status, municipality of origin, monthly income, frequency of malaria, and frequency of diarrheal diseases in persons who gave their informed consent. Venous blood samples were taken to test for anti-SARS-COV-2 antibodies (IgM and IgG) using the VIDAS automated system. RESULTS: A total of 1504 patients were recruited and tested for antibodies directed against SARS-COV-2. Data analysis revealed the presence of anti-SARS-CoV-2 in 53.5% of participants, 17.3% of the participants had a previous symptomatic infection, 31.4% had an asymptomatic infection, and 44.8% were never in contact with the virus. Seroprevalence of SARS-CoV-2 was higher in Marcory (70.9%) and Cocody (61.1%) than in the remaining townships (48.4%). Township rates of serologically confirmed SARS-CoV-2 infection correlated with poverty index (p = 0.025), children < 5 years' proportion in the township (p = 0.026), and levels of malaria (p = 0.034). CONCLUSIONS: In the city of Abidjan, COVID-19 is strongly modulated by poverty, the proportion of babies and toddlers in the community, or exposure to malaria.


Subject(s)
Antibodies, Viral , COVID-19 , SARS-CoV-2 , Humans , Cote d'Ivoire/epidemiology , COVID-19/epidemiology , Prospective Studies , Male , Female , Seroepidemiologic Studies , SARS-CoV-2/immunology , Adult , Middle Aged , Adolescent , Young Adult , Antibodies, Viral/blood , Child , Child, Preschool , Infant , Aged , Immunoglobulin G/blood , Immunoglobulin M/blood
SELECTION OF CITATIONS
SEARCH DETAIL