1
|
Ling J, Hryckowian AJ. Re-framing the importance of Group B Streptococcus as a gut-resident pathobiont. Infect Immun 2024; 92:e0047823. [PMID: 38436256 PMCID: PMC11392526 DOI: 10.1128/iai.00478-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2024] Open
Abstract
Streptococcus agalactiae (Group B Streptococcus, GBS) is a Gram-positive bacterial species that causes disease in humans across the lifespan. While antibiotics are used to mitigate GBS infections, it is evident that antibiotics disrupt human microbiomes (which can predispose people to other diseases later in life), and antibiotic resistance in GBS is on the rise. Taken together, these unintended negative impacts of antibiotics highlight the need for precision approaches for minimizing GBS disease. One possible approach involves selectively depleting GBS in its commensal niches before it can cause disease at other body sites or be transmitted to at-risk individuals. One understudied commensal niche of GBS is the adult gastrointestinal (GI) tract, which may predispose colonization at other body sites in individuals at risk for GBS disease. However, a better understanding of the host-, microbiome-, and GBS-determined variables that dictate GBS GI carriage is needed before precise GI decolonization approaches can be developed. In this review, we synthesize current knowledge of the diverse body sites occupied by GBS as a pathogen and as a commensal. We summarize key molecular factors GBS utilizes to colonize different host-associated niches to inform future efforts to study GBS in the GI tract. We also discuss other GI commensals that are pathogenic in other body sites to emphasize the broader utility of precise de-colonization approaches for mitigating infections by GBS and other bacterial pathogens. Finally, we highlight how GBS treatments could be improved with a more holistic understanding of GBS enabled by continued GI-focused study.
Collapse
Affiliation(s)
- Joie Ling
- Department of
Medicine, Division of Gastroenterology and Hepatology, University of
Wisconsin School of Medicine and Public
Health, Madison,
Wisconsin, USA
- Department of Medical
Microbiology and Immunology, University of Wisconsin School of Medicine
and Public Healthon,
Madison, Wisconsin, USA
- Microbiology Doctoral
Training Program, University of
Wisconsin-Madison, Madison,
Wisconsin, USA
| | - Andrew J. Hryckowian
- Department of
Medicine, Division of Gastroenterology and Hepatology, University of
Wisconsin School of Medicine and Public
Health, Madison,
Wisconsin, USA
- Department of Medical
Microbiology and Immunology, University of Wisconsin School of Medicine
and Public Healthon,
Madison, Wisconsin, USA
| |
Collapse
|
2
|
Boscarino G, Romano R, Iotti C, Tegoni F, Perrone S, Esposito S. An Overview of Antibiotic Therapy for Early- and Late-Onset Neonatal Sepsis: Current Strategies and Future Prospects. Antibiotics (Basel) 2024; 13:250. [PMID: 38534685 DOI: 10.3390/antibiotics13030250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 03/04/2024] [Accepted: 03/07/2024] [Indexed: 03/28/2024] Open
Abstract
Neonatal sepsis is a clinical syndrome mainly associated with a bacterial infection leading to severe clinical manifestations that could be associated with fatal sequalae. According to the time of onset, neonatal sepsis is categorized as early- (EOS) or late-onset sepsis (LOS). Despite blood culture being the gold standard for diagnosis, it has several limitations, and early diagnosis is not immediate. Consequently, most infants who start empirical antimicrobial therapy do not have an underlying infection. Despite stewardship programs partially reduced this negative trend, in neonatology, antibiotic overuse still persists, and it is associated with several relevant problems, the first of which is the increase in antimicrobial resistance (AMR). Starting with these considerations, we performed a narrative review to summarize the main findings and the future prospects regarding antibiotics use to treat neonatal sepsis. Because of the impact on morbidity and mortality that EOS and LOS entail, it is essential to start an effective and prompt treatment as soon as possible. The use of targeted antibiotics is peremptory as soon as the pathogen in the culture is detected. Although prompt therapy is essential, it should be better assessed whether, when and how to treat neonates with antibiotics, even those at higher risk. Considering that we are certainly in the worrying era defined as the "post-antibiotic era", it is still essential and urgent to define novel strategies for the development of antibacterial compounds with new targets or mechanisms of action. A future strategy could also be to perform well-designed studies to develop innovative algorithms for improving the etiological diagnosis of infection, allowing for more personalized use of the antibiotics to treat EOS and LOS.
Collapse
Affiliation(s)
- Giovanni Boscarino
- Pediatric Clinic, University Hospital, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| | - Rossana Romano
- Pediatric Clinic, University Hospital, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| | - Carlotta Iotti
- Pediatric Clinic, University Hospital, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| | - Francesca Tegoni
- Pediatric Clinic, University Hospital, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| | - Serafina Perrone
- PNeonatology Unit, University Hospital, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| | - Susanna Esposito
- Pediatric Clinic, University Hospital, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| |
Collapse
|
3
|
Bjerkhaug AU, Ramalingham S, Mboizi R, Le Doare K, Klingenberg C. The immunogenicity and safety of Group B Streptococcal maternal vaccines: A systematic review. Vaccine 2024; 42:84-98. [PMID: 38072754 DOI: 10.1016/j.vaccine.2023.11.056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 11/22/2023] [Accepted: 11/27/2023] [Indexed: 01/01/2024]
Abstract
PURPOSE To systematically review immunogenicity and safety data of maternal group B streptococcal (GBS) vaccines in published clinical trials until July 2023. METHODS EMBASE, MEDLINE, Cochrane Library and clinicaltrial.gov. databases were searched for clinical studies that reported immunogenicity and/or safety of GBS vaccine in non-pregnant adults, pregnant women and infants between 1st of January 1996 to 31st of July 2023. Pairs of reviewers independently selected, data extracted, and assessed the risk of bias of the studies. Discrepancies were resolved by consensus. (PROSPERO CRD42020185213). RESULTS We retrieved 1472 records from the literature search; 20 studies and 6 sub-studies were included, involving 4440 non-pregnant participants and 1325 pregnant women with their newborns. There was a significantly higher IgG Geometric Mean Concentration (GMC) and IgG placental transfer ratios in vaccinated compared to placebo groups, with peak response 4-8 weeks after vaccination. Placental transfer ratio varied from 0.4 to 1.4 across five studies. The different clinical trials used different assays that limited direct comparison. There were no significant differences in the risk of serious adverse events (adjusted OR 0.73; 95 % CI 0.49-1.07), serious adverse events leading to withdrawal (adjusted OR 0.44; 95 % CI 0.13-1.51), and systemic illness or fever (adjusted OR 1.05; 95 % CI 0.26-4.19) between the vaccine and placebo groups. CONCLUSIONS The published clinical trials show significant IgG GMC response in subjects receiving the conjugated capsular polysaccharide and surface subunit protein vaccines compared to placebo. In current clinical trials of experimental GBS maternal vaccines, there have been no observed serious adverse events of special interest directly linked to vaccination.
Collapse
Affiliation(s)
- Aline U Bjerkhaug
- Paediatric Research Group, Faculty of Health Sciences, UiT-The Arctic University of Norway, Tromsø, Norway; Department of Paediatrics and Adolescence Medicine, University Hospital of North Norway, Tromsø, Norway.
| | - Shouwmika Ramalingham
- Paediatric Research Group, Faculty of Health Sciences, UiT-The Arctic University of Norway, Tromsø, Norway
| | - Robert Mboizi
- Makerere University Johns Hopkins University (MU-JHU) Research Collaboration (MUJHU CARE LTD), Kampala, Uganda
| | - Kirsty Le Doare
- Makerere University Johns Hopkins University (MU-JHU) Research Collaboration (MUJHU CARE LTD), Kampala, Uganda; Centre for Neonatal and Paediatric Infection, Maternal and Neonatal Vaccine Immunology Research Group, St Georgés University of London, United Kingdom
| | - Claus Klingenberg
- Paediatric Research Group, Faculty of Health Sciences, UiT-The Arctic University of Norway, Tromsø, Norway; Department of Paediatrics and Adolescence Medicine, University Hospital of North Norway, Tromsø, Norway
| |
Collapse
|
4
|
Carboni F, Cozzi R, Romagnoli G, Tuscano G, Balocchi C, Buffi G, Bodini M, Brettoni C, Giusti F, Marchi S, Brogioni G, Brogioni B, Cinelli P, Cappelli L, Nocciolini C, Senesi S, Facciotti C, Frigimelica E, Fabbrini M, Stranges D, Savino S, Maione D, Adamo R, Wizel B, Margarit I, Romano MR. Proof of concept for a single-dose Group B Streptococcus vaccine based on capsular polysaccharide conjugated to Qβ virus-like particles. NPJ Vaccines 2023; 8:152. [PMID: 37803013 PMCID: PMC10558462 DOI: 10.1038/s41541-023-00744-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 09/15/2023] [Indexed: 10/08/2023] Open
Abstract
A maternal vaccine to protect neonates against Group B Streptococcus invasive infection is an unmet medical need. Such a vaccine should ideally be offered during the third trimester of pregnancy and induce strong immune responses after a single dose to maximize the time for placental transfer of protective antibodies. A key target antigen is the capsular polysaccharide, an anti-phagocytic virulence factor that elicits protective antibodies when conjugated to carrier proteins. The most prevalent polysaccharide serotypes conjugated to tetanus or diphtheria toxoids have been tested in humans as monovalent and multivalent formulations, showing excellent safety profiles and immunogenicity. However, responses were suboptimal in unprimed individuals after a single shot, the ideal schedule for vaccination during the third trimester of pregnancy. In the present study, we obtained and optimized self-assembling virus-like particles conjugated to Group B Streptococcus capsular polysaccharides. The resulting glyco-nanoparticles elicited strong immune responses in mice already after one immunization, providing pre-clinical proof of concept for a single-dose vaccine.
Collapse
|
5
|
Dhar N, Mohamed E, Kirstein F, Williams M, Dorasamy S, van Zyl P, Robertson MJ, Anderson T, Harden LM, Jardine K, Veeraraghavan B, Wilson S, Tippoo P, Madhi SA, Kwatra G. Immune responses against group B Streptococcus monovalent and pentavalent capsular polysaccharide tetanus toxoid conjugate vaccines in Balb/c mice. iScience 2023; 26:107380. [PMID: 37575182 PMCID: PMC10415928 DOI: 10.1016/j.isci.2023.107380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 05/31/2023] [Accepted: 07/07/2023] [Indexed: 08/15/2023] Open
Abstract
Immunization of pregnant women with Group B Streptococcus (GBS) capsular polysaccharide (CPS) conjugate vaccine (CV) could protect young infants against invasive GBS disease. We evaluated the immunogenicity of investigational five GBS monovalent (serotypes Ia, Ib, II, III, and V) CPS-tetanus toxoid (TT)-CV with adjuvant and GBS pentavalent CPS-TT-CV with adjuvant (GBS5-CV-adj) and without adjuvant (GBS5-CV-no-adj), in Balb/c mice. Aluminum phosphate was the adjuvant in the formulations, where included. The homotypic immunoglobulin G (IgG) geometric mean concentration (GMC) and opsonophagocytic activity (OPA) geometric mean titer (GMT) did not differ after the third dose of the GBS5-CV-adj vaccine compared with the monovalent counterparts for all five serotypes. The GBS5-CV-adj induced higher post-vaccination serotype-specific IgG GMCs and OPA GMTs compared to GBS5-CV-no_adj. The GBS5-CV with and without adjuvant should be considered for further development as a potential vaccine for pregnant women to protect their infants against invasive GBS disease.
Collapse
Affiliation(s)
- Nisha Dhar
- South African Medical Research Council Vaccines and Infectious Diseases Analytics Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | | | | | | | | | | | | | | | - Lois M. Harden
- Brain Function Research Group, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Kimberly Jardine
- Wits Research Animal Facility, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | | | | | | | - Shabir A. Madhi
- South African Medical Research Council Vaccines and Infectious Diseases Analytics Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- African Leadership in Vaccinology Expertise, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Gaurav Kwatra
- South African Medical Research Council Vaccines and Infectious Diseases Analytics Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Department of Clinical Microbiology, Christian Medical College, Vellore, India
- African Leadership in Vaccinology Expertise, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
6
|
Gonzalez-Miro M, Pawlowski A, Lehtonen J, Cao D, Larsson S, Darsley M, Kitson G, Fischer PB, Johansson-Lindbom B. Safety and immunogenicity of the group B streptococcus vaccine AlpN in a placebo-controlled double-blind phase 1 trial. iScience 2023; 26:106261. [PMID: 36915681 PMCID: PMC10005905 DOI: 10.1016/j.isci.2023.106261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 11/28/2022] [Accepted: 02/17/2023] [Indexed: 02/25/2023] Open
Abstract
Group B streptococcus (GBS) is a leading cause of life-threatening neonatal infections and subsets of adverse pregnancy outcomes. Essentially all GBS strains possess one allele of the alpha-like protein (Alp) family. A maternal GBS vaccine, consisting of the fused N-terminal domains of the Alps αC and Rib (GBS-NN), was recently demonstrated to be safe and immunogenic in healthy adult women. To enhance antibody responses to all clinically relevant Alps, a second-generation vaccine has been developed (AlpN), also containing the N-terminal domain of Alp1 and the one shared by Alp2 and Alp3. In this study, the safety and immunogenicity of AlpN is assessed in a randomized, double-blind, placebo-controlled, and parallel-group phase I study, involving 60 healthy non-pregnant women. AlpN is well tolerated and elicits similarly robust and persistent antibody responses against all four Alp-N-terminal domains, resulting in enhanced opsonophagocytic killing of all Alp serotypes covered by the vaccine.
Collapse
Affiliation(s)
| | | | - Janne Lehtonen
- Minervax A/S, Ole Maaløes Vej 3, 2200 Copenhagen N, Denmark
| | - Duojia Cao
- Immunology Section, Lund University, BMC D14, Lund, Sweden
| | - Sara Larsson
- Immunology Section, Lund University, BMC D14, Lund, Sweden
| | | | - Geoff Kitson
- Minervax A/S, Ole Maaløes Vej 3, 2200 Copenhagen N, Denmark
| | - Per B Fischer
- Minervax A/S, Ole Maaløes Vej 3, 2200 Copenhagen N, Denmark
| | - Bengt Johansson-Lindbom
- Immunology Section, Lund University, BMC D14, Lund, Sweden.,Minervax A/S, Ole Maaløes Vej 3, 2200 Copenhagen N, Denmark
| |
Collapse
|
7
|
Krauss SR, Barbateskovic M, Klingenberg SL, Djurisic S, Petersen SB, Kenfelt M, Kong DZ, Jakobsen JC, Gluud C. Aluminium adjuvants versus placebo or no intervention in vaccine randomised clinical trials: a systematic review with meta-analysis and Trial Sequential Analysis. BMJ Open 2022; 12:e058795. [PMID: 35738649 PMCID: PMC9226993 DOI: 10.1136/bmjopen-2021-058795] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 05/19/2022] [Indexed: 11/11/2022] Open
Abstract
OBJECTIVES To assess the benefits and harms of aluminium adjuvants versus placebo or no intervention in randomised clinical trials in relation to human vaccine development. DESIGN Systematic review with meta-analysis and trial sequential analysis assessing the certainty of evidence with Grading of Recommendations Assessment, Development and Evaluation (GRADE). DATA SOURCES We searched CENTRAL, MEDLINE, Embase, LILACS, BIOSIS, Science Citation Index Expanded and Conference Proceedings Citation Index-Science until 29 June 2021, and Chinese databases until September 2021. ELIGIBILITY CRITERIA Randomised clinical trials irrespective of type, status and language of publication, with trial participants of any sex, age, ethnicity, diagnosis, comorbidity and country of residence. DATA EXTRACTION AND SYNTHESIS Two independent reviewers extracted data and assessed risk of bias with Cochrane's RoB tool 1. Dichotomous data were analysed as risk ratios (RRs) and continuous data as mean differences. We explored both fixed-effect and random-effects models, with 95% CI. Heterogeneity was quantified with I2 statistic. We GRADE assessed the certainty of the evidence. RESULTS We included 102 randomised clinical trials (26 457 participants). Aluminium adjuvants versus placebo or no intervention may have no effect on serious adverse events (RR 1.18, 95% CI 0.97 to 1.43; very low certainty) and on all-cause mortality (RR 1.02, 95% CI 0.74 to 1.41; very low certainty). No trial reported on quality of life. Aluminium adjuvants versus placebo or no intervention may increase adverse events (RR 1.13, 95% CI 1.07 to 1.20; very low certainty). We found no or little evidence of a difference between aluminium adjuvants versus placebo or no intervention when assessing serology with geometric mean titres or concentrations or participants' seroprotection. CONCLUSIONS Based on evidence at very low certainty, we were unable to identify benefits of aluminium adjuvants, which may be associated with adverse events considered non-serious.
Collapse
Affiliation(s)
- Sara Russo Krauss
- The Copenhagen Trial Unit, Centre for Clinical Intervention Research, The Capital Region, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Marija Barbateskovic
- The Copenhagen Trial Unit, Centre for Clinical Intervention Research, The Capital Region, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Sarah Louise Klingenberg
- The Copenhagen Trial Unit, Centre for Clinical Intervention Research, The Capital Region, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Snezana Djurisic
- The Copenhagen Trial Unit, Centre for Clinical Intervention Research, The Capital Region, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Sesilje Bondo Petersen
- Department of Occupational and Environmental Medicine, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Copenhagen, Denmark
| | | | - De Zhao Kong
- The Evidence-Based Medicine Research Center of Traditional Chinese Medicine, Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning, China
- Department of Evidence-based Chinese Medicine Research Centre, The Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning, China
| | - Janus C Jakobsen
- The Copenhagen Trial Unit, Centre for Clinical Intervention Research, The Capital Region, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
- Department of Regional Health Research, The Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
| | - Christian Gluud
- The Copenhagen Trial Unit, Centre for Clinical Intervention Research, The Capital Region, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
- Department of Regional Health Research, The Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
8
|
Brokaw A, Nguyen S, Quach P, Orvis A, Furuta A, Johansson-Lindbom B, Fischer PB, Rajagopal L. A Recombinant Alpha-Like Protein Subunit Vaccine (GBS-NN) Provides Protection in Murine Models of Group B Streptococcus Infection. J Infect Dis 2022; 226:177-187. [PMID: 35429401 PMCID: PMC9890916 DOI: 10.1093/infdis/jiac148] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 04/11/2022] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Group B Streptococcus (GBS) transmission during pregnancy causes preterm labor, stillbirths, fetal injury, or neonatal infections. Rates of adult infections are also rising. The GBS-NN vaccine, engineered by fusing N-terminal domains of GBS Alpha C and Rib proteins, is safe in healthy, nonpregnant women, but further assessment is needed for use during pregnancy. Here, we tested GBS-NN vaccine efficacy using mouse models that recapitulate human GBS infection outcomes. METHODS Following administration of GBS-NN vaccine or adjuvant, antibody profiles were compared by ELISA. Vaccine efficacy was examined by comparing infection outcomes in GBS-NN vaccinated versus adjuvant controls during systemic and pregnancy-associated infections, and during intranasal infection of neonatal mice following maternal vaccination. RESULTS Vaccinated mice had higher GBS-NN-specific IgG titers versus controls. These antibodies bound alpha C and Rib on GBS clinical isolates. Fewer GBS were recovered from systemically challenged vaccinated mice versus controls. Although vaccination did not eliminate GBS during ascending infection in pregnancy, vaccinated dams experienced fewer in utero fetal deaths. Additionally, maternal vaccination prolonged neonatal survival following intranasal GBS challenge. CONCLUSIONS These findings demonstrate GBS-NN vaccine efficacy in murine systemic and perinatal GBS infections and suggest that maternal vaccination facilitates the transfer of protective antibodies to neonates.
Collapse
Affiliation(s)
- Alyssa Brokaw
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington, USA,Department of Global Health, University of Washington, Seattle, Washington, USA
| | - Shayla Nguyen
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington, USA
| | - Phoenicia Quach
- Present affiliation: Phoenicia Quach, Universal Cells, Seattle 98121, Washington
| | - Austyn Orvis
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington, USA
| | - Anna Furuta
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington, USA,Department of Global Health, University of Washington, Seattle, Washington, USA
| | | | | | - Lakshmi Rajagopal
- Correspondence: L. Rajagopal, PhD, Department of Pediatrics, University of Washington, Seattle Children’s Hospital Research Institute, 307 Westlake Ave N, Seattle, WA 98109 ()
| |
Collapse
|
9
|
Izu A, Kwatra G, Madhi SA, Rigat F. Estimation of invasive Group B Streptococcus disease risk in young infants from case-control serological studies. BMC Med Res Methodol 2022; 22:85. [PMID: 35350991 PMCID: PMC8961496 DOI: 10.1186/s12874-022-01529-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 01/20/2022] [Indexed: 11/12/2022] Open
Abstract
Background Group B Streptococcus (GBS) infections are a major cause of invasive disease (IGbsD) in young infants and cause miscarriage and stillbirths. Immunization of pregnant women against GBS in addition to intrapartum antibiotic prophylaxis could prevent disease. Establishing accurate serological markers of protection against IGbsD could enable use of efficient clinical trial designs for vaccine development and licensure, without needing to undertake efficacy trials in prohibitively large number of mother-infant dyads. The association of maternal naturally acquired serotype-specific anti-capsular antibodies (IgG) against serotype-specific IGbsD in their infants has been studied in case-control studies. The statistical models used so far to estimate IGbsD risk from these case-control studies assumed that the antibody concentrations measured sharing the same disease status are sampled from the same population, not allowing for differences between mothers colonised by GBS and mothers also potentially infected (e.g urinary tract infection or chorioamnionitis) by GBS during pregnancy. This distinction is relevant as infants born from infected mothers with occult medical illness may be exposed to GBS prior to the mother developing antibodies measured in maternal or infant sera. Methods Unsupervised mixture model averaging (MMA) is proposed and applied here to accurately estimate infant IGbsD risk from case-control study data in presence or absence of antibody concentration subgroups potentially associated to maternal GBS carriage or infection. MMA estimators are compared to non-parametric disease risk estimators in simulation studies and by analysis of two published GBS case-control studies. Results MMA provides more accurate relative risk estimates under a broad range of data simulation scenarios and more accurate absolute disease risk estimates when the proportion of IGbsD cases with high antibody levels is not ignorable. MMA estimates of the relative and absolute disease risk curves are more amenable to clinical interpretation compared to non-parametric estimates with no detectable overfitting of the data. Antibody concentration thresholds predictive of protection from infant IGbsD estimated by MMA from maternal and infant sera are consistent with non-parametric estimates. Conclusions MMA is a flexible and robust method for design, accurate analysis and clinical interpretation of case-control studies estimating relative and absolute IGbsD risk from antibody concentrations measured at or after birth.
Collapse
Affiliation(s)
- Alane Izu
- South African Medical Research Council: Vaccines and Infectious Diseases Analytical Research Unit (VIDA), University of the Witwatersrand, Faculty of Health Science, Johannesburg, South Africa. .,Department of Science and Innovation/National Research Foundation South African Research Chair Initiative in Vaccine Preventable Diseases Unit, University of the Witwatersrand, Faculty of Health Science, Johannesburg, South Africa.
| | - Gaurav Kwatra
- South African Medical Research Council: Vaccines and Infectious Diseases Analytical Research Unit (VIDA), University of the Witwatersrand, Faculty of Health Science, Johannesburg, South Africa.,Department of Science and Innovation/National Research Foundation South African Research Chair Initiative in Vaccine Preventable Diseases Unit, University of the Witwatersrand, Faculty of Health Science, Johannesburg, South Africa
| | - Shabir A Madhi
- South African Medical Research Council: Vaccines and Infectious Diseases Analytical Research Unit (VIDA), University of the Witwatersrand, Faculty of Health Science, Johannesburg, South Africa.,Department of Science and Innovation/National Research Foundation South African Research Chair Initiative in Vaccine Preventable Diseases Unit, University of the Witwatersrand, Faculty of Health Science, Johannesburg, South Africa
| | - Fabio Rigat
- Statistics and Decision Sciences, Janssen Pharmaceuticals R & D, High Wycombe, United Kingdom.
| |
Collapse
|
10
|
Stefanetti G, Borriello F, Richichi B, Zanoni I, Lay L. Immunobiology of Carbohydrates: Implications for Novel Vaccine and Adjuvant Design Against Infectious Diseases. Front Cell Infect Microbiol 2022; 11:808005. [PMID: 35118012 PMCID: PMC8803737 DOI: 10.3389/fcimb.2021.808005] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 12/22/2021] [Indexed: 12/14/2022] Open
Abstract
Carbohydrates are ubiquitous molecules expressed on the surface of nearly all living cells, and their interaction with carbohydrate-binding proteins is critical to many immunobiological processes. Carbohydrates are utilized as antigens in many licensed vaccines against bacterial pathogens. More recently, they have also been considered as adjuvants. Interestingly, unlike other types of vaccines, adjuvants have improved immune response to carbohydrate-based vaccine in humans only in a few cases. Furthermore, despite the discovery of many new adjuvants in the last years, aluminum salts, when needed, remain the only authorized adjuvant for carbohydrate-based vaccines. In this review, we highlight historical and recent advances on the use of glycans either as vaccine antigens or adjuvants, and we review the use of currently available adjuvants to improve the efficacy of carbohydrate-based vaccines. A better understanding of the mechanism of carbohydrate interaction with innate and adaptive immune cells will benefit the design of a new generation of glycan-based vaccines and of immunomodulators to fight both longstanding and emerging diseases.
Collapse
Affiliation(s)
- Giuseppe Stefanetti
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA, United States
| | - Francesco Borriello
- Division of Immunology, Harvard Medical School and Boston Children’s Hospital, Boston, MA, United States
| | - Barbara Richichi
- Department of Chemistry “Ugo Schiff”, University of Florence, Florence, Italy
| | - Ivan Zanoni
- Division of Immunology, Division of Gastroenterology, Harvard Medical School and Boston Children’s Hospital, Boston, MA, United States
| | - Luigi Lay
- Department of Chemistry, University of Milan, Milan, Italy
| |
Collapse
|
11
|
Miselli F, Frabboni I, Di Martino M, Zinani I, Buttera M, Insalaco A, Stefanelli F, Lugli L, Berardi A. Transmission of Group B Streptococcus in late-onset neonatal disease: a narrative review of current evidence. Ther Adv Infect Dis 2022; 9:20499361221142732. [PMID: 36569815 PMCID: PMC9780763 DOI: 10.1177/20499361221142732] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 11/16/2022] [Indexed: 12/24/2022] Open
Abstract
Group B streptococcus (GBS) late-onset disease (LOD, occurring from 7 through 89 days of life) is an important cause of sepsis and meningitis in infants. The pathogenesis and modes of transmission of LOD to neonates are yet to be elucidated. Established risk factors for the incidence of LOD include maternal GBS colonisation, young maternal age, preterm birth, HIV exposure and African ethnicity. The mucosal colonisation by GBS may be acquired perinatally or in the postpartum period from maternal or other sources. Growing evidence has demonstrated the predominant role of maternal sources in the transmission of LOD. Intrapartum antibiotic prophylaxis (IAP) to prevent early-onset disease reduces neonatal GBS colonisation during delivery; however, a significant proportion of IAP-exposed neonates born to GBS-carrier mothers acquire the pathogen at mucosal sites in the first weeks of life. GBS-infected breast milk, with or without presence of mastitis, is considered a potential vehicle for transmitting GBS. Furthermore, horizontal transmission is possible from nosocomial and other community sources. Although unfrequently reported, nosocomial transmission of GBS in the neonatal intensive care unit is probably less rare than is usually believed. GBS disease can sometime recur and is usually caused by the same GBS serotype that caused the primary infection. This review aims to discuss the dynamics of transmission of GBS in the neonatal LOD.
Collapse
Affiliation(s)
- Francesca Miselli
- Neonatal Intensive Care Unit, Azienda Ospedaliero-Universitaria Policlinico, Modena, Italy
| | - Ilaria Frabboni
- Pediatric Post-Graduate School, University of Modena e Reggio Emilia, Modena, Italy
| | - Marianna Di Martino
- Pediatric Post-Graduate School, University of Modena e Reggio Emilia, Modena, Italy
| | - Isotta Zinani
- Pediatric Post-Graduate School, University of Modena e Reggio Emilia, Modena, Italy
| | - Martina Buttera
- Pediatric Post-Graduate School, University of Modena e Reggio Emilia, Modena, Italy
| | - Anna Insalaco
- Pediatric Post-Graduate School, University of Modena e Reggio Emilia, Modena, Italy
| | - Francesca Stefanelli
- Pediatric Post-Graduate School, University of Modena e Reggio Emilia, Modena, Italy
| | - Licia Lugli
- Neonatal Intensive Care Unit, Azienda Ospedaliero-Universitaria Policlinico, Modena, Italy
| | - Alberto Berardi
- Neonatal Intensive Care Unit, Policlinico University Hospital, 41124 Modena, Italy
| |
Collapse
|
12
|
Jansen KU, Gruber WC, Simon R, Wassil J, Anderson AS. The impact of human vaccines on bacterial antimicrobial resistance. A review. ENVIRONMENTAL CHEMISTRY LETTERS 2021; 19:4031-4062. [PMID: 34602924 PMCID: PMC8479502 DOI: 10.1007/s10311-021-01274-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 07/09/2021] [Indexed: 05/07/2023]
Abstract
At present, the dramatic rise in antimicrobial resistance (AMR) among important human bacterial pathogens is reaching a state of global crisis threatening a return to the pre-antibiotic era. AMR, already a significant burden on public health and economies, is anticipated to grow even more severe in the coming decades. Several licensed vaccines, targeting both bacterial (Haemophilus influenzae type b, Streptococcus pneumoniae, Salmonella enterica serovar Typhi) and viral (influenza virus, rotavirus) human pathogens, have already proven their anti-AMR benefits by reducing unwarranted antibiotic consumption and antibiotic-resistant bacterial strains and by promoting herd immunity. A number of new investigational vaccines, with a potential to reduce the spread of multidrug-resistant bacterial pathogens, are also in various stages of clinical development. Nevertheless, vaccines as a tool to combat AMR remain underappreciated and unfortunately underutilized. Global mobilization of public health and industry resources is key to maximizing the use of licensed vaccines, and the development of new prophylactic vaccines could have a profound impact on reducing AMR.
Collapse
Affiliation(s)
| | | | - Raphael Simon
- Pfizer Vaccine Research and Development, Pearl River, NY USA
| | - James Wassil
- Pfizer Patient and Health Impact, Collegeville, PA USA
- Present Address: Vaxcyte, 353 Hatch Drive, Foster City, CA 94404 USA
| | | |
Collapse
|
13
|
Safety and immunogenicity of a prototype recombinant alpha-like protein subunit vaccine (GBS-NN) against Group B Streptococcus in a randomised placebo-controlled double-blind phase 1 trial in healthy adult women. Vaccine 2021; 39:4489-4499. [PMID: 34215454 DOI: 10.1016/j.vaccine.2021.06.046] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 05/24/2021] [Accepted: 06/18/2021] [Indexed: 12/14/2022]
Abstract
BACKGROUND Group B Streptococcus (GBS) is the leading cause of life-threatening infections in new-borns and may cause invasive disease, stillbirth and preterm delivery during pregnancy. While no licensed vaccine exists, maternal immunization might protect against neonatal disease and adverse pregnancy outcomes. We assessed the safety and immunogenicity of a prototype vaccine consisting of the fused N-terminal domains of the AlphaC and Rib surface proteins of GBS (GBS-NN). METHODS GBS-NN was tested in a randomised, double-blind, placebo-controlled, parallel group, phase I study, in healthy non-pregnant women. A dose-escalation phase, with two doses, four weeks apart, of 10, 50 or 250 µg, administered with or without aluminium hydroxide, was initially assessed (n = 60). This was followed by a dose-confirmation study, where one dose of 100 µg adjuvanted GBS-NN was compared with two doses of either 50 or 100 µg adjuvanted GBS-NN, again administered with four weeks interval between the doses (n = 180). Safety and immunogenicity were monitored for one year. RESULTS GBS-NN was well tolerated with some, mostly mild, injection site reactions observed. Adjuvant significantly increased antibody concentrations and the response was boosted by a second dose. The IgG GMCs remained strongly elevated during the whole one-year duration of the study. Maximal responses occurred after two 50 µg doses, resulting in IgG GMC of 16.9 µg/ml at the primary immunological endpoint, twelve weeks after the first dose. For this regimen, 100% and 89% of the subjects achieved antibody levels above the arbitrary thresholds of 1 and 4 µg/ml, respectively. The added beneficial effect of a second dose was most pronounced for subjects with pre-existing IgG levels below the median of the entire cohort. CONCLUSION The prototype GBS-NN vaccine was found to be well tolerated and highly immunogenic with an optimal regimen of two doses of 50 µg in the presence of adjuvant. Further development of a maternal vaccine based on the N-terminal domains of the alpha-like protein family of GBS is warranted (NCT02459262).
Collapse
|
14
|
Bedeley E, Gori A, Yeboah-Manu D, Diallo K. Control of Streptococcal Infections: Is a Common Vaccine Target Achievable Against Streptococcus agalactiae and Streptococcus pneumoniae. Front Microbiol 2021; 12:658824. [PMID: 33967998 PMCID: PMC8103614 DOI: 10.3389/fmicb.2021.658824] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 03/30/2021] [Indexed: 12/21/2022] Open
Abstract
Both Streptococcus agalactiae [group B streptococcus (GBS)] and Streptococcus pneumoniae (pneumococcus) remain significant pathogens as they cause life threatening infections mostly in children and the elderly. The control of diseases caused by these pathogens is dependent on antibiotics use and appropriate vaccination. The introduction of the pneumococcal conjugate vaccines (PCVs) against some serotypes has led to reduction in pneumococcal infections, however, the subsequent serotype switching, and replacement has been a serious challenge. On the other hand, no vaccine is yet licensed for use in the control of GBS diseases. In this review, we provide an overview of the history and global disease burden, disease pathophysiology and management, vaccines update, and the biology of both pathogens. Furthermore, we address recent findings regarding structural similarities that could be explored for vaccine targets across both mucosal pathogens. Finally, we conclude by proposing future genomic sequence comparison using the wealth of available sequences from both species and the possibility of identifying more related structural components that could be exploited for pan-pathogen vaccine development.
Collapse
Affiliation(s)
- Edmund Bedeley
- Department of Bacteriology, Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana
| | - Andrea Gori
- NIHR Global Health Research Unit on Mucosal Pathogens, Division of Infection and Immunity, University College London, London, United Kingdom
| | - Dorothy Yeboah-Manu
- Department of Bacteriology, Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana
- West African Centre for Cell Biology of Infectious Pathogens, College of Basic and Applied Sciences, University of Ghana, Accra, Ghana
| | - Kanny Diallo
- West African Centre for Cell Biology of Infectious Pathogens, College of Basic and Applied Sciences, University of Ghana, Accra, Ghana
- Centre Suisse de Recherche Scientifique de Côte d’Ivoire, Abidjan, Côte d’Ivoire
| |
Collapse
|
15
|
Leroux-Roels G, Bebia Z, Maes C, Aerssens A, De Boever F, Grassano L, Buffi G, Margarit I, Karsten A, Cho S, Slobod K, Corsaro B, Henry O. Safety and Immunogenicity of a Second Dose of an Investigational Maternal Trivalent Group B Streptococcus Vaccine in Nonpregnant Women 4-6 Years After a First Dose: Results From a Phase 2 Trial. Clin Infect Dis 2021; 70:2570-2579. [PMID: 31394574 PMCID: PMC7286364 DOI: 10.1093/cid/ciz737] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 08/09/2019] [Indexed: 12/23/2022] Open
Abstract
Background Maternal immunization against group B streptococcus (GBS) could protect infants from invasive GBS disease. Additional doses in subsequent pregnancies may be needed. We evaluated the safety and immunogenicity of a second dose of an investigational trivalent CRM197-glycoconjugate GBS vaccine (targeting serotypes Ia/Ib/III), administered to nonpregnant women 4–6 years postdose 1. Methods Healthy women either previously vaccinated with 1 dose of trivalent GBS vaccine 4–6 years before enrollment (n = 53) or never GBS vaccinated (n = 27) received a single trivalent GBS vaccine injection. Adverse events (AEs) were recorded. Serotype-specific (Ia/Ib/III) anti-GBS antibodies were measured by multiplex immunoassay prevaccination and 30/60 days postvaccination. Results AEs were reported with similar rates after a first or second dose; none were serious. Of previously GBS-vaccinated women, 92%–98% had anti-GBS concentrations that exceeded an arbitrary threshold (8 µg/mL) for each serotype 60 days postdose 2 vs 36%–56% postdose 1 in previously non–GBS-vaccinated women. Of previously GBS-vaccinated women with undetectable baseline (predose 1) anti-GBS levels, 90%–98% reached this threshold postdose 2. For each serotype, anti-GBS geometric mean concentrations (GMCs) 30/60 days postdose 2 in previously GBS-vaccinated women were ≥200-fold higher than baseline GMCs. Among women with undetectable baseline anti-GBS levels, postdose 2 GMCs in previously GBS-vaccinated women exceeded postdose 1 GMCs in previously non–GBS-vaccinated women (≥7-fold). Conclusions A second trivalent GBS vaccine dose administered 4–6 years postdose 1 was immunogenic with a favorable safety profile. Women with undetectable preexisting anti-GBS concentrations may benefit from a sufficiently spaced second vaccine dose. Clinical Trials Registration NCT02690181
Collapse
Affiliation(s)
- Geert Leroux-Roels
- Center for Vaccinology, Ghent University and Ghent University Hospital, Belgium
| | | | - Cathy Maes
- Center for Vaccinology, Ghent University and Ghent University Hospital, Belgium
| | - Annelies Aerssens
- Center for Vaccinology, Ghent University and Ghent University Hospital, Belgium
| | - Fien De Boever
- Center for Vaccinology, Ghent University and Ghent University Hospital, Belgium
| | | | | | | | | | - Stephen Cho
- Novartis, Cambridge, Massachusetts.,GSK, Cambridge, Massachusetts
| | - Karen Slobod
- Novartis, Cambridge, Massachusetts.,GSK, Cambridge, Massachusetts
| | | | | |
Collapse
|
16
|
Swamy GK, Metz TD, Edwards KM, Soper DE, Beigi RH, Campbell JD, Grassano L, Buffi G, Dreisbach A, Margarit I, Karsten A, Henry O, Lattanzi M, Bebia Z. Safety and immunogenicity of an investigational maternal trivalent group B streptococcus vaccine in pregnant women and their infants: Results from a randomized placebo-controlled phase II trial. Vaccine 2020; 38:6930-6940. [PMID: 32883555 DOI: 10.1016/j.vaccine.2020.08.056] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 08/18/2020] [Accepted: 08/20/2020] [Indexed: 01/31/2023]
Abstract
BACKGROUND This study evaluated the safety and immunogenicity of an investigational trivalent group B streptococcus (GBS) vaccine in US pregnant women, transplacental serotype-specific antibody transfer and persistence in infants, and serotype-specific antibodies in breast milk. METHODS This randomized, observer-blind, placebo-controlled trial administered one dose of trivalent GBS vaccine (n = 49) or placebo (n = 26) to healthy pregnant 18-40-year-old women at 240/7-346/7 weeks' gestation. Women were enrolled from March 2014 to August 2015. Safety follow-up continued through postpartum day 180. Primary immunogenicity objectives were to evaluate serotype Ia/Ib/III-specific immunoglobulin G (IgG) levels in sera from women on day 1 (pre-vaccination), day 31, delivery and postpartum days 42 and 90, and from infants at birth (cord blood), days 42 and 90. Antibody transfer ratios (cord blood/maternal sera at delivery) and serotype-specific secretory immunoglobulin A (sIgA) and IgG in breast milk after delivery and on postpartum days 42 and 90 were evaluated. The planned sample size was not based on statistical assumptions for this descriptive study. RESULTS Baseline characteristics were similar between groups. Serious adverse events were reported for 16% of GBS-vaccinated women and 15% of their infants, and 15% of placebo recipients and 12% of their infants; none were fatal or deemed vaccine-related. Serotype-specific IgG geometric mean concentrations (GMCs) were 13-23-fold higher in vaccine vs placebo recipients on day 31 and persisted until postpartum day 90. Median antibody concentrations were substantially higher in women with detectable pre-vaccination antibody concentrations. Antibody transfer ratios in the vaccine group were 0.62-0.82. Infant IgG GMCs and breast milk sIgA GMCs were higher in the vaccine vs the placebo group at all timepoints. CONCLUSIONS Maternal immunization with the trivalent GBS vaccine in US women had a favorable safety profile, elicited antibodies that were transplacentally transferred and persisted in infants for a minimum of 3 months. CLINICAL TRIAL REGISTRATION Clinicaltrials.gov, NCT02046148.
Collapse
Affiliation(s)
- Geeta K Swamy
- Department of Obstetrics and Gynecology, Duke University School of Medicine, Durham, NC, USA.
| | - Torri D Metz
- Department of Obstetrics and Gynecology, University of Colorado Denver, Aurora, CO, USA.
| | - Kathryn M Edwards
- Vanderbilt Vaccine Research Program, Vanderbilt University, Nashville, TN, USA.
| | - David E Soper
- Medical University of South Carolina, Charleston, SC, USA.
| | - Richard H Beigi
- UPMC Magee-Women's Hospital, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| | - James D Campbell
- Center for Vaccine Development, University of Maryland School of Medicine, Baltimore, MD, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Carreras-Abad C, Ramkhelawon L, Heath PT, Le Doare K. A Vaccine Against Group B Streptococcus: Recent Advances. Infect Drug Resist 2020; 13:1263-1272. [PMID: 32425562 PMCID: PMC7196769 DOI: 10.2147/idr.s203454] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Accepted: 03/10/2020] [Indexed: 01/12/2023] Open
Abstract
Group B streptococcus (GBS) causes a high burden of neonatal and infant disease globally. Implementing a vaccine for pregnant women is a promising strategy to prevent neonatal and infant GBS disease and has been identified as a priority by the World Health Organisation (WHO). GBS serotype-specific polysaccharide – protein conjugate vaccines are at advanced stages of development, but a large number of participants would be required to undertake Phase III clinical efficacy trials. Efforts are therefore currently focused on establishing serocorrelates of protection in natural immunity studies as an alternative pathway for licensure of a GBS vaccine, followed by Phase IV studies to evaluate safety and effectiveness. Protein vaccines are in earlier stages of development but are highly promising as they might confer protection irrespective of serotype. Further epidemiological, immunological and health economic studies are required to enable the vaccine to reach its target population as soon as possible.
Collapse
Affiliation(s)
- Clara Carreras-Abad
- Paediatric Infectious Diseases Research Group and Vaccine Institute, Institute for Infection and Immunity, St George's, University of London, London, UK.,Department of Paediatrics, Obstetrics and Gynecology and Preventive Medicine and Public Health, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Laxmee Ramkhelawon
- Paediatric Infectious Diseases Research Group and Vaccine Institute, Institute for Infection and Immunity, St George's, University of London, London, UK
| | - Paul T Heath
- Paediatric Infectious Diseases Research Group and Vaccine Institute, Institute for Infection and Immunity, St George's, University of London, London, UK
| | - Kirsty Le Doare
- Paediatric Infectious Diseases Research Group and Vaccine Institute, Institute for Infection and Immunity, St George's, University of London, London, UK.,Pathogen Immunity Group, Public Health England, Porton Down,UK.,Medical Research Council/Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine, Uganda Research Unit, Entebbe, Uganda
| |
Collapse
|
18
|
Beran J, Leroux-Roels G, Van Damme P, de Hoon J, Vandermeulen C, Al-Ibrahim M, Johnson C, Peterson J, Baker S, Seidl C, Dreisbach A, Karsten A, Corsaro B, Henry O, Lattanzi M, Bebia Z. Safety and immunogenicity of fully liquid and lyophilized formulations of an investigational trivalent group B streptococcus vaccine in healthy non-pregnant women: Results from a randomized comparative phase II trial. Vaccine 2020; 38:3227-3234. [DOI: 10.1016/j.vaccine.2020.02.085] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2019] [Revised: 02/20/2020] [Accepted: 02/29/2020] [Indexed: 01/10/2023]
|
19
|
Maertens K, Orije MRP, Van Damme P, Leuridan E. Vaccination during pregnancy: current and possible future recommendations. Eur J Pediatr 2020; 179:235-242. [PMID: 31912233 PMCID: PMC7222942 DOI: 10.1007/s00431-019-03563-w] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 11/15/2019] [Accepted: 12/23/2019] [Indexed: 11/15/2022]
Abstract
Immunizing pregnant women to protect the mother, fetus and infant from infection has increasingly been used over the last decade. Protection against infectious diseases in neonates is mainly provided by maternal antibodies transferred from mother to infant during pregnancy through transplacental transport or after delivery via breastfeeding. Both the transplacental- and breast milk-derived maternal antibodies function as the primary source of protection against infectious diseases in neonates during the first vulnerable weeks of life. During recent infectious disease outbreaks (influenza, pertussis, Zika…) and for other infectious diseases (CMV, GBS…), pregnant women are increasingly identified as an important target for vaccination. For some of these diseases, vaccines are already on the market, and recommended during pregnancy. For others, vaccines are currently under development; furthermore, some are even specifically designed to be administered during pregnancy.Conclusion: This review article provides an overview on the rationale and main mechanism of the maternal vaccination strategy and gives a summary about the current and possible future recommendations for maternal vaccination.What is Known:• Maternal vaccination has a far-reaching potential in the protection of both women and offspring.• Currently, tetanus, pertussis and influenza vaccination during pregnancy is recommended in some countries. Several new vaccines specifically designed for use in pregnancy are currently under development.What is New:• Review providing a timely overview of the rationale and main mechanisms of the maternal vaccination strategy• Up-to-date summary of the current and possible future recommendations for maternal vaccination.
Collapse
Affiliation(s)
- Kirsten Maertens
- Centre for the Evaluation of Vaccination, Vaccine & Infectious Diseases Institute, University of Antwerp, Universiteitsplein 1, 2610, Wilrijk, Belgium.
| | - Marjolein Rozemarie Paulien Orije
- grid.5284.b0000 0001 0790 3681Centre for the Evaluation of Vaccination, Vaccine & Infectious Diseases Institute, University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium
| | - Pierre Van Damme
- grid.5284.b0000 0001 0790 3681Centre for the Evaluation of Vaccination, Vaccine & Infectious Diseases Institute, University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium
| | - Elke Leuridan
- grid.5284.b0000 0001 0790 3681Centre for the Evaluation of Vaccination, Vaccine & Infectious Diseases Institute, University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium
| |
Collapse
|
20
|
Villagomez AN, Muñoz FM, Peterson RL, Colbert AM, Gladstone M, MacDonald B, Wilson R, Fairlie L, Gerner GJ, Patterson J, Boghossian NS, Burton VJ, Cortés M, Katikaneni LD, Larson JCG, Angulo AS, Joshi J, Nesin M, Padula MA, Kochhar S, Connery AK. Neurodevelopmental delay: Case definition & guidelines for data collection, analysis, and presentation of immunization safety data. Vaccine 2019; 37:7623-7641. [PMID: 31783983 PMCID: PMC6899448 DOI: 10.1016/j.vaccine.2019.05.027] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Accepted: 05/09/2019] [Indexed: 12/15/2022]
Affiliation(s)
- Adrienne N Villagomez
- University of Colorado School of Medicine, Aurora, CO, USA; Children's Hospital of Colorado, Aurora, CO, USA
| | - Flor M Muñoz
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Robin L Peterson
- University of Colorado School of Medicine, Aurora, CO, USA; Children's Hospital of Colorado, Aurora, CO, USA
| | - Alison M Colbert
- University of Colorado School of Medicine, Aurora, CO, USA; Children's Hospital of Colorado, Aurora, CO, USA
| | - Melissa Gladstone
- Department of Women and Children's Health, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | | | - Rebecca Wilson
- University of Colorado School of Medicine, Aurora, CO, USA; Children's Hospital of Colorado, Aurora, CO, USA
| | - Lee Fairlie
- Wits Reproductive Health and HIV Institute, University of the Witwatersrand, Johannesburg, South Africa
| | - Gwendolyn J Gerner
- Kennedy Krieger Institute, Baltimore, MD, USA; Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jackie Patterson
- University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Nansi S Boghossian
- Department of Epidemiology and Biostatistics, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA
| | - Vera Joanna Burton
- Kennedy Krieger Institute, Baltimore, MD, USA; Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | | | - Jennifer C G Larson
- Department of Physical Medicine and Rehabilitation, University of Michigan, Ann Arbor, MI, USA
| | - Abigail S Angulo
- University of Colorado School of Medicine, Aurora, CO, USA; Children's Hospital of Colorado, Aurora, CO, USA
| | - Jyoti Joshi
- Center for Disease Dynamics Economics & Policy, Amity Institute of Public Health, Amity University, India
| | - Mirjana Nesin
- Division of Microbiology and Infectious Diseases, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Michael A Padula
- Children's Hospital of Philadelphia, Department of Pediatrics, Philadelphia, PA, USA
| | - Sonali Kochhar
- Global Healthcare Consulting, India; University of Washington, Seattle, USA; Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Amy K Connery
- University of Colorado School of Medicine, Aurora, CO, USA; Children's Hospital of Colorado, Aurora, CO, USA.
| |
Collapse
|
21
|
Kobayashi M, Schrag SJ, Alderson MR, Madhi SA, Baker CJ, Sobanjo-Ter Meulen A, Kaslow DC, Smith PG, Moorthy VS, Vekemans J. WHO consultation on group B Streptococcus vaccine development: Report from a meeting held on 27-28 April 2016. Vaccine 2019; 37:7307-7314. [PMID: 28017431 PMCID: PMC6892266 DOI: 10.1016/j.vaccine.2016.12.029] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Accepted: 10/17/2016] [Indexed: 11/29/2022]
Abstract
Globally, group B Streptococcus (GBS) remains a leading cause of sepsis and meningitis in infants in the first 90days of life. Intrapartum antibiotic prophylaxis (IAP) for women at increased risk of transmitting GBS to their newborns has been effective in reducing part, but not all, of the GBS disease burden in many high income countries (HICs). In low- and middle-income countries (LMICs), IAP use is low. Immunization of pregnant women with a GBS vaccine represents an alternative strategy to protecting newborns and young infants, through transplacental antibody transfer and potentially by reducing new vaginal colonization. This vaccination strategy was first suggested in the 1970s and several potential GBS vaccines have completed phase I/II clinical trials. During the 2015 WHO Product Development for Vaccines Advisory Committee meeting, GBS was identified as a high priority for the development of a vaccine for maternal immunization because of the major public health burden posed by GBS in LMICs, and the high technical feasibility for successful development. Following this meeting, the first WHO technical consultation on GBS vaccines was held on the 27th and 28th of April 2016, to consider development pathways for such vaccines, focused on their potential role in reducing newborn and young infant deaths and possibly stillbirths in LMICs. Discussion topics included: (1) pathophysiology of disease; (2) current gaps in the knowledge of global disease burden and serotype distribution; (3) vaccine candidates under development; (4) design considerations for phase III trials; and (5) pathways to licensure, policy recommendations and use. Efforts to address gaps identified in each of these areas are needed to establish the public health need for, the development and deployment of, efficacious GBS vaccines. In particular, more work is required to understand the global disease burden of GBS-associated stillbirths, and to develop quality-assured standardized antibody assays to identify correlates of protection.
Collapse
Affiliation(s)
- Miwako Kobayashi
- National Center for Immunization and Respiratory Diseases, Division of Bacterial Diseases, Centers for Disease Control and Prevention, Atlanta, GA 30329-4027, USA; Epidemic Intelligence Service, Centers for Disease Control and Prevention, Atlanta, GA 30333, USA
| | - Stephanie J Schrag
- National Center for Immunization and Respiratory Diseases, Division of Bacterial Diseases, Centers for Disease Control and Prevention, Atlanta, GA 30329-4027, USA
| | - Mark R Alderson
- Center for Vaccine Innovation and Access, PATH, Seattle, WA 98121, USA
| | - Shabir A Madhi
- Department of Science and Technology/National Research Foundation: Vaccine Preventable Diseases, and Medical Research Council: Respiratory and Meningeal Pathogens Research Unit, University of the Witwatersrand, Johannesburg, South Africa
| | - Carol J Baker
- Department of Pediatrics, Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
| | | | - David C Kaslow
- Center for Vaccine Innovation and Access, PATH, Seattle, WA 98121, USA
| | - Peter G Smith
- MRC Tropical Epidemiology Group, London School of Hygiene & Tropical Medicine, London, UK
| | - Vasee S Moorthy
- Initiative for Vaccine Research, World Health Organization, CH-1211 Geneva 27, Switzerland
| | - Johan Vekemans
- Initiative for Vaccine Research, World Health Organization, CH-1211 Geneva 27, Switzerland.
| |
Collapse
|
22
|
Giles ML, Buttery J, Davey MA, Wallace E. Pregnant women's knowledge and attitude to maternal vaccination including group B streptococcus and respiratory syncytial virus vaccines. Vaccine 2019; 37:6743-6749. [PMID: 31540809 DOI: 10.1016/j.vaccine.2019.08.084] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 08/11/2019] [Accepted: 08/30/2019] [Indexed: 01/31/2023]
Abstract
BACKGROUND Maternal immunisation is an important strategy to reduce neonatal mortality and morbidity. New maternal vaccines such as Respiratory Syncytial Virus (RSV) and Group B streptococcus (GBS) are in development and/or clinical trials. However, little is known about pregnant women's knowledge about these diseases. METHODS Women attending antenatal clinics in Melbourne, Australia were invited to complete a questionnaire collecting demographic information, past vaccination history, understanding of risk of GBS and RSV disease in pregnancy and likelihood to accept these theoretical vaccines in the future. FINDINGS 495 women (48% born outside of Australia, from 48 different countries) completed the questionnaire. A large number of women had never heard of GBS (63%) or RSV (83%). Women over 35 years, born in Australia and women who had more than one child were more likely to have heard of GBS or RSV (p < 0.001). Women who had received influenza or pertussis vaccine in pregnancy were more likely to accept a RSV or GBS vaccine (p < 0.001). CONCLUSIONS This study has shown that knowledge of GBS and RSV is poor. However, when provided with information about the two diseases, acceptance of a hypothetical vaccine for both diseases was high. This study highlights the enormous amount of work that needs to be done in educating pregnant women about the seriousness of these two diseases if a future vaccine is ever to be accepted and high coverage achieved among the target population.
Collapse
Affiliation(s)
- Michelle L Giles
- The Ritchie Centre, Department of Obstetrics and Gynaecology, Monash University, Monash Medical Centre, 246 Clayton Road, Clayton, Victoria 3168, Australia; Women's and Children's Health, Monash Health, 246 Clayton Road, Clayton, Victoria 3168, Australia; Monash Immunisation, Monash Health, 246 Clayton Road, Clayton, Victoria 3168, Australia.
| | - Jim Buttery
- School of Public Health and Preventive Medicine and Department of Pediatrics, Monash University, 246 Clayton Road, Clayton, Victoria 3168, Australia; Monash Immunisation, Monash Health, 246 Clayton Road, Clayton, Victoria 3168, Australia
| | - Mary-Ann Davey
- The Ritchie Centre, Department of Obstetrics and Gynaecology, Monash University, Monash Medical Centre, 246 Clayton Road, Clayton, Victoria 3168, Australia
| | - Euan Wallace
- The Ritchie Centre, Department of Obstetrics and Gynaecology, Monash University, Monash Medical Centre, 246 Clayton Road, Clayton, Victoria 3168, Australia
| |
Collapse
|
23
|
Novel Multiplex Immunoassays for Quantification of IgG against Group B Streptococcus Capsular Polysaccharides in Human Sera. mSphere 2019; 4:4/4/e00273-19. [PMID: 31391276 PMCID: PMC6686225 DOI: 10.1128/msphere.00273-19] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Group B Streptococcus (GBS) infections constitute a major cause of invasive disease during the first three months of life and an unmet medical need that could be addressed by maternal vaccination. The GBS capsular polysaccharides (CPSs) have shown promise as vaccine targets in clinical studies. A highly specific serological assay to quantify maternal and neonatal anti-CPS antibody levels will be instrumental for GBS vaccine licensure. Here, we describe the development and comparison of two novel multiplex immunoassays (MIAs) based on the Luminex technology for the quantification of IgG antibodies recognizing the five most frequent GBS capsular variants (Ia, Ib, II, III, and V) out of the ten types identified. The first assay is based on the use of biotinylated CPSs coupled to streptavidin-derivatized magnetic microspheres (Biotin-CPS MIA), while the second is a sandwich assay with plain CPSs coupled to magnetic microspheres coated with polysaccharide-specific mouse monoclonal antibodies (Sandwich MIA). Both assays showed good specificity, linearity, and precision, although the Biotin-CPS MIA presented higher sensitivity and lower complexity than the Sandwich MIA. A panel of human sera representing a wide range of anti-CPS IgG concentrations was tested in parallel by the two assays, which resulted in comparable titers. Our data support the preservation of antigenic epitopes in the biotinylated polysaccharides and the suitability of the Biotin-CPS MIA for the precise determination of GBS anti-CPS IgG concentrations in human sera.IMPORTANCE Group B streptococcal infections can cause death in neonates up to 3 months of age. Intrapartum antibiotic prophylaxis in GBS-colonized mothers has limited early infections but has no impact after the first week of life. The development of a maternal vaccine to address this unmet medical need has been identified as a priority by the World Health Organization, and the GBS CPSs are considered the best antigen targets. However, to date there are no accepted standardized assays to measure immune responses to the investigational vaccines and for establishment of serocorrelates of protection. Here, we describe the performance of two microsphere-based pentaplex immunoassays for the determination of antibodies recognizing the five most frequent GBS serotypes. Our data confirm that an assay based on biotinylated polysaccharides coupled to streptavidin microspheres would be suitable for the intended purpose.
Collapse
|
24
|
Francois Watkins LK, McGee L, Schrag SJ, Beall B, Jain JH, Pondo T, Farley MM, Harrison LH, Zansky SM, Baumbach J, Lynfield R, Snippes Vagnone P, Miller LA, Schaffner W, Thomas AR, Watt JP, Petit S, Langley GE. Epidemiology of Invasive Group B Streptococcal Infections Among Nonpregnant Adults in the United States, 2008-2016. JAMA Intern Med 2019; 179:479-488. [PMID: 30776079 PMCID: PMC6450309 DOI: 10.1001/jamainternmed.2018.7269] [Citation(s) in RCA: 141] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2018] [Accepted: 10/26/2018] [Indexed: 12/17/2022]
Abstract
Importance Group B Streptococcus (GBS) is an important cause of invasive bacterial disease. Previous studies have shown a substantial and increasing burden of GBS infections among nonpregnant adults, particularly older adults and those with underlying medical conditions. Objective To update trends of invasive GBS disease among US adults using population-based surveillance data. Design, Setting, and Participants In this population-based surveillance study, a case was defined as isolation of GBS from a sterile site between January 1, 2008, and December 31, 2016. Demographic and clinical data were abstracted from medical records. Rates were calculated using US Census data. Antimicrobial susceptibility testing and serotyping were performed on a subset of isolates. Case patients were residents of 1 of 10 catchment areas of the Active Bacterial Core surveillance (ABCs) network, representing approximately 11.5% of the US adult population. Patients were included in the study if they were nonpregnant, were 18 years or older, were residents of an ABCs catchment site, and had a positive GBS culture from a normally sterile body site. Main Outcomes and Measures Trends in GBS cases overall and by demographic characteristics (sex, age, and race), underlying clinical conditions of patients, and isolate characteristics are described. Results The ABCs network detected 21 250 patients with invasive GBS among nonpregnant adults from 2008 through 2016. The GBS incidence in this population increased from 8.1 cases per 100 000 population in 2008 to 10.9 in 2016 (P = .002 for trend). There were 3146 cases reported in 2016 (59% male; median age, 64 years; age range, 18-103 years). The GBS incidence was higher among men than women and among blacks than whites and increased with age. Projected to the US population, an estimated 27 729 cases of invasive disease and 1541 deaths occurred in the United States in 2016. Ninety-five percent of cases in 2016 occurred in someone with at least 1 underlying condition, most commonly obesity (53.9%) and diabetes (53.4%). Resistance to clindamycin increased from 37.0% of isolates in 2011 to 43.2% in 2016 (P = .02). Serotypes Ia, Ib, II, III, and V accounted for 86.4% of isolates in 2016; serotype IV increased from 4.7% in 2008 to 11.3% in 2016 (P < .001 for trend). Conclusions and Relevance The public health burden of invasive GBS disease among nonpregnant adults is substantial and continues to increase. Chronic diseases, such as obesity and diabetes, may contribute.
Collapse
Affiliation(s)
- Louise K. Francois Watkins
- Epidemic Intelligence Service Program, Centers for Disease Control and Prevention, Atlanta, Georgia
- Respiratory Diseases Branch, Centers for Disease Control and Prevention, Atlanta, Georgia
- Division of Foodborne, Waterborne, and Environmental Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Lesley McGee
- Respiratory Diseases Branch, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Stephanie J. Schrag
- Respiratory Diseases Branch, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Bernard Beall
- Respiratory Diseases Branch, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Jennifer Hudson Jain
- Respiratory Diseases Branch, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Tracy Pondo
- Respiratory Diseases Branch, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Monica M. Farley
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Lee H. Harrison
- Johns Hopkins Bloomberg School of Public Health, The Johns Hopkins University, Baltimore, Maryland
| | | | | | | | | | - Lisa A. Miller
- Colorado School of Public Health, University of Colorado Denver, Aurora
- Colorado Department of Public Health and Environment, Denver
| | | | | | | | - Susan Petit
- Connecticut Department of Public Health, Hartford
| | - Gayle E. Langley
- Respiratory Diseases Branch, Centers for Disease Control and Prevention, Atlanta, Georgia
- Division of Viral Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia
| |
Collapse
|
25
|
Lin SM, Jang AY, Zhi Y, Gao S, Lim S, Lim JH, Song JY, Sullam PM, Rhee JH, Seo HS. Vaccination With a Latch Peptide Provides Serotype-Independent Protection Against Group B Streptococcus Infection in Mice. J Infect Dis 2019; 217:93-102. [PMID: 29106586 DOI: 10.1093/infdis/jix565] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Accepted: 10/24/2017] [Indexed: 11/14/2022] Open
Abstract
Streptococcus agalactiae (group B streptococcus [GBS]) is a leading cause of invasive diseases in neonates and severe infections in elderly individuals. GBS serine-rich repeat glycoprotein 1 (Srr1) acts as a critical virulence factor by facilitating GBS invasion into the central nervous system through interaction with the fibrinogen Aα chain. This study revealed that srr1 is highly conserved, with 86.7% of GBS clinical isolates expressing the protein. Vaccination of mice with different Srr1 truncated peptides revealed that only Srr1 truncates containing the latch domain protected against GBS meningitis. Furthermore, the latch peptide alone was immunogenic and elicited protective antibodies, which efficiently enhanced antibody-mediated opsonophagocytic killing of GBS by HL60 cells and provided heterogeneous protection against 4 different GBS serogroups. Taken together, these findings indicated that the latch domain of Srr1 may constitute an effective peptide vaccine candidate for GBS.
Collapse
Affiliation(s)
- Shun-Mei Lin
- Biotechnology Division, Korea Atomic Energy Research Institute, Jeongeup.,Brain Korea 21 Program for Leading Universities and Students, Department of Molecular Medicine, Chonnam National University Medical School, Gwangju
| | - A-Yeung Jang
- Biotechnology Division, Korea Atomic Energy Research Institute, Jeongeup.,Department of Biological Sciences, Chonbuk National University, Jeonju
| | - Yong Zhi
- Biotechnology Division, Korea Atomic Energy Research Institute, Jeongeup.,Department of Radiation Biotechnology and Applied Radioisotope Science, University of Science and Technology, Daejeon
| | - Shuang Gao
- Biotechnology Division, Korea Atomic Energy Research Institute, Jeongeup.,Brain Korea 21 Program for Leading Universities and Students, Department of Molecular Medicine, Chonnam National University Medical School, Gwangju
| | - Sangyong Lim
- Biotechnology Division, Korea Atomic Energy Research Institute, Jeongeup.,Department of Radiation Biotechnology and Applied Radioisotope Science, University of Science and Technology, Daejeon
| | - Jae Hyang Lim
- Department of Radiation Biotechnology and Applied Radioisotope Science, University of Science and Technology, Daejeon
| | - Joon Young Song
- Department of Internal Medicine, Korea University College of Medicine, Seoul, Republic of Korea
| | - Paul M Sullam
- Department of Microbiology, College of Medicine, Ewha Womans University, Seoul, Republic of Korea.,Division of Infectious Diseases, Veterans Affairs Medical Center, University of California-San Francisco, San Francisco, California.,Department of Medicine, University of California-San Francisco, San Francisco, California.,Northern California Institute for Research and Education, San Francisco, California
| | - Joon Haeng Rhee
- Brain Korea 21 Program for Leading Universities and Students, Department of Molecular Medicine, Chonnam National University Medical School, Gwangju.,Department of Microbiology, Chonnam National University Medical School, Gwangju.,Clinical Vaccine Research and Development Center, Chonnam National University Medical School, Gwangju
| | - Ho Seong Seo
- Biotechnology Division, Korea Atomic Energy Research Institute, Jeongeup.,Department of Radiation Biotechnology and Applied Radioisotope Science, University of Science and Technology, Daejeon
| |
Collapse
|
26
|
Rothen J, Pothier JF, Foucault F, Blom J, Nanayakkara D, Li C, Ip M, Tanner M, Vogel G, Pflüger V, Daubenberger CA. Subspecies Typing of Streptococcus agalactiae Based on Ribosomal Subunit Protein Mass Variation by MALDI-TOF MS. Front Microbiol 2019; 10:471. [PMID: 30915057 PMCID: PMC6421976 DOI: 10.3389/fmicb.2019.00471] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Accepted: 02/22/2019] [Indexed: 01/08/2023] Open
Abstract
Background: A ribosomal subunit protein (rsp)-based matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOF MS) method was developed for fast subspecies-level typing of Streptococcus agalactiae (Group B Streptococcus, GBS), a major cause of neonatal sepsis and meningitis. Methods: A total of 796 GBS whole genome sequences, covering the genetic diversity of the global GBS population, were used to in silico predict molecular mass variability of 28 rsp and to identify unique rsp mass combinations, termed “rsp-profiles”. The in silico established GBS typing scheme was validated by MALDI-TOF MS analysis of GBS isolates at two independent research sites in Europe and South East Asia. Results: We identified in silico 62 rsp-profiles, with the majority (>80%) of the 796 GBS isolates displaying one of the six rsp-profiles 1–6. These dominant rsp-profiles classify GBS strains in high concordance with the core-genome based phylogenetic clustering. Validation of our approach by in-house MALDI-TOF MS analysis of 248 GBS isolates and external analysis of 8 GBS isolates showed that across different laboratories and MALDI-TOF MS platforms, the 28 rsp were detected reliably in the mass spectra, allowing assignment of clinical isolates to rsp-profiles at high sensitivity (99%) and specificity (97%). Our approach distinguishes the major phylogenetic GBS genotypes, identifies hyper-virulent strains, predicts the probable capsular serotype and surface protein variants and distinguishes between GBS genotypes of human and animal origin. Conclusion: We combine the information depth of whole genome sequences with the highly cost efficient, rapid and robust MALDI-TOF MS approach facilitating high-throughput, inter-laboratory, large-scale GBS epidemiological and clinical studies based on pre-defined rsp-profiles.
Collapse
Affiliation(s)
- Julian Rothen
- Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute (Swiss TPH) Basel, Basel, Switzerland.,University of Basel, Basel, Switzerland
| | - Joël F Pothier
- Research Group for Environmental Genomics and Systems Biology, Institute of Natural Resource Sciences, Zurich University of Applied Sciences (ZHAW), Wädenswil, Switzerland
| | | | - Jochen Blom
- Bioinformatics and Systems Biology, Justus-Liebig-Universität Gießen, Giessen, Germany
| | - Dulmini Nanayakkara
- Department of Microbiology, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Carmen Li
- Department of Microbiology, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Margaret Ip
- Department of Microbiology, The Chinese University of Hong Kong, Shatin, Hong Kong
| | | | | | | | - Claudia A Daubenberger
- Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute (Swiss TPH) Basel, Basel, Switzerland.,University of Basel, Basel, Switzerland
| |
Collapse
|
27
|
Paoletti LC, Kasper DL. Surface Structures of Group B Streptococcus Important in Human Immunity. Microbiol Spectr 2019; 7:10.1128/microbiolspec.gpp3-0001-2017. [PMID: 30873933 PMCID: PMC11590616 DOI: 10.1128/microbiolspec.gpp3-0001-2017] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Indexed: 11/20/2022] Open
Abstract
The surface of the Gram-positive opportunistic pathogen Streptococcus agalactiae, or group B Streptococcus (GBS), harbors several carbohydrate and protein antigens with the potential to be effective vaccines. Capsular polysaccharides of all clinically-relevant GBS serotypes coupled to immunogenic proteins of both GBS and non-GBS origin have undergone extensive testing in animals that led to advanced clinical trials in healthy adult women. In addition, GBS proteins either alone or in combination have been tested in animals; a fusion protein construct has recently advanced to human clinical studies. Given our current understanding of the antigenicity and immunogenicity of the wide array of GBS surface antigens, formulations now exist for the generation of viable vaccines against diseases caused by GBS.
Collapse
Affiliation(s)
- Lawrence C Paoletti
- Division of Immunology, Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115
| | - Dennis L Kasper
- Division of Immunology, Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115
| |
Collapse
|
28
|
Bengtson AM, Sanfilippo AM, Hughes BL, Savitz DA. Maternal immunisation to improve the health of HIV-exposed infants. THE LANCET. INFECTIOUS DISEASES 2018; 19:e120-e131. [PMID: 30529212 DOI: 10.1016/s1473-3099(18)30545-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2018] [Revised: 07/28/2018] [Accepted: 08/21/2018] [Indexed: 12/13/2022]
Abstract
HIV-exposed but uninfected (HEU) infants are at an increased risk of many infectious diseases that can contribute to the high mortality seen among HEU children. Maternal immunisation could be a promising strategy to reduce infections in HEU infants. However, very little research has explored the effect of HIV on the immunogenicity and effectiveness of vaccines given during pregnancy. We review the available evidence on maternal immunisation among women living with HIV (WLWH) for all vaccines recommended, considered, or being investigated for routine or risk-based use during pregnancy. Of the 11 vaccines included, only three have been investigated in WLWH. Available evidence suggests that maternal HIV infection limits the immunogenicity of several vaccines, leaving HEU infants more susceptible to infection during their first few months of life. Whether maternal immunisation reduces the infectious morbidity and mortality associated with infectious diseases in HEU children remains unknown. We conclude the Review by identifying future research priorities.
Collapse
Affiliation(s)
- Angela M Bengtson
- Department of Epidemiology, Brown University School of Public Health, Providence, RI, USA.
| | - Alan M Sanfilippo
- Department of Pathology, Beth Israel Deaconess Medical Center, and Harvard Medical School, Boston, MA, USA
| | - Brenna L Hughes
- Division of Maternal Fetal Medicine, Duke University, Durham, NC, USA
| | - David A Savitz
- Department of Epidemiology, Brown University School of Public Health, Providence, RI, USA
| |
Collapse
|
29
|
Giles ML, Krishnaswamy S, Wallace EM. Maternal immunisation: What have been the gains? Where are the gaps? What does the future hold? F1000Res 2018; 7:F1000 Faculty Rev-1733. [PMID: 30443339 PMCID: PMC6213781 DOI: 10.12688/f1000research.15475.1] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/24/2018] [Indexed: 12/22/2022] Open
Abstract
The vaccination of pregnant women has enormous potential to protect not only mothers from vaccine-preventable diseases but also their infants through the passive acquisition of protective antibodies before they are able to themselves acquire protection through active childhood immunisations. Maternal tetanus programmes have been in place since 1989, and as of March 2018, only 14 countries in the world were still to reach maternal neonatal tetanus elimination status. This has saved hundreds of thousands of lives. Building on this success, influenza- and pertussis-containing vaccines have been recommended for pregnant women and introduced into immunisation programmes, albeit predominantly in resource-rich settings. These have highlighted some important challenges when additional immunisations are introduced into the antenatal context. With new vaccine candidates, such as respiratory syncytial virus (RSV) and group B streptococcus (GBS), on the horizon, it is important that we learn from these experiences, identify the information gaps, and close these to ensure safe and successful implementation of maternal vaccines in the future, particularly in low- and middle-income countries with a high burden of disease.
Collapse
Affiliation(s)
- Michelle L. Giles
- Ritchie Centre, Department of Obstetrics and Gynaecology, Monash University, Melbourne, VIC, Australia
| | - Sushena Krishnaswamy
- Ritchie Centre, Department of Obstetrics and Gynaecology, Monash University, Melbourne, VIC, Australia
- Monash Infectious Diseases, Monash Health, Melbourne, VIC, Australia
| | - Euan M. Wallace
- Ritchie Centre, Department of Obstetrics and Gynaecology, Monash University, Melbourne, VIC, Australia
- Safer Care Victoria, Victorian Government, Melbourne, VIC, Australia
| |
Collapse
|
30
|
Adjuvant-Dependent Enhancement of HIV Env-Specific Antibody Responses in Infant Rhesus Macaques. J Virol 2018; 92:JVI.01051-18. [PMID: 30089691 DOI: 10.1128/jvi.01051-18] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2018] [Accepted: 07/30/2018] [Indexed: 12/16/2022] Open
Abstract
Toward the goal of developing an effective HIV vaccine that can be administered in infancy to protect against postnatal and lifelong sexual HIV transmission risks, the current pilot study was designed to compare the effect of novel adjuvants on the induction of HIV Env-specific antibody responses in infant macaques. Aligning our studies with the adjuvanted proteins evaluated in a prime-boost schedule with ALVAC in the ongoing HVTN (HIV Vaccine Trials Network) 702 efficacy trial, we selected the bivalent clade C Env immunogens gp120 C.1086 and gp120 TV1 in combination with the MF59 adjuvant. However, we hypothesized that the adjuvant system AS01, that is included in the pediatric RTS,S malaria vaccine, would promote Env-specific antibody responses superior to those of the oil-in-water MF59 emulsion adjuvant. In a second study arm, we compared two emulsions, glucopyranosyl lipid adjuvant formulated in a stable emulsion (GLA-SE) and 3M-052-SE, containing Toll-like receptor 4 (TLR4) and TLR7/TLR8 (TLR7/8) ligand, respectively. The latter adjuvant had been previously demonstrated to be especially effective in activating neonatal antigen-presenting cells. Our results demonstrate that different adjuvants drive quantitatively or qualitatively distinct responses to the bivalent Env vaccine. AS01 induced higher Env-specific plasma IgG antibody levels than the antigen in MF59 and promoted improved antibody function in infants, and 3M-052-SE outperformed GLA-SE by inducing the highest breadth and functionality of antibody responses. Thus, distinct adjuvants are likely to be required for maximizing vaccine-elicited immune responses in infants, particularly when immunization in infancy aims to elicit both perinatal and lifelong immunity against challenging pathogens such as HIV.IMPORTANCE Alum remains the adjuvant of choice for pediatric vaccines. Yet the distinct nature of the developing immune system in infants likely requires novel adjuvants targeted specifically at the pediatric population to reach maximal vaccine efficacy with an acceptable safety profile. The current study supports the idea that additional adjuvants for pediatric vaccines should be, and need to be, tested in infants for their potential to enhance immune responses. Using an infant macaque model, our results suggest that both AS01 and 3M-052-SE can significantly improve and better sustain HIV Env-specific antibody responses than alum. Despite the limited number of animals, the results revealed interesting differences that warrant further testing of promising novel adjuvant candidates in larger preclinical and clinical studies to define the mechanisms leading to adjuvant-improved antibody responses and to identify targets for adjuvant and vaccine optimization.
Collapse
|
31
|
Hartley J, Li Y, Kunkel L, Crowcroft NS. The burden of infant group B streptococcal infections in Ontario: Analysis of administrative data to estimate the potential benefits of new vaccines. Hum Vaccin Immunother 2018; 15:193-202. [PMID: 30130440 PMCID: PMC6363068 DOI: 10.1080/21645515.2018.1511666] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Revised: 07/17/2018] [Accepted: 08/05/2018] [Indexed: 01/31/2023] Open
Abstract
Group B streptococcus (GBS) is a leading bacterial cause of neonatal sepsis and meningitis in many countries as well as an important cause of disease in pregnant women. Currently, serotype-specific conjugate vaccines are being developed. We conducted an epidemiological analysis of health administrative data to estimate the burden of infant GBS disease in Ontario, Canada and combined these estimates with literature on serotype distribution to estimate the burden of disease likely to be vaccine-preventable. Between 1st January 2005 and 31st December 2015, 907 of 64320 health care encounters in Ontario in patients under 1 year old had codes specifically identifying GBS as the cause of the disease, of which 717 were under one month of age. In addition, application of epidemiological data to the remaining patients allowed us to estimate a further 2322 cases and among them 1822 were under one month of age. In the same period, 579 confirmed neonatal invasive GBS cases in patients up to one month of age were reported to public health. Depending on serotype distribution, vaccination coverage and early versus late onset disease (0-6 days and 7-90 days of age respectively), the preventable fraction ranged widely. With a vaccine that is 90% effective and 60% immunization coverage, up to 52% of early and late onset disease could be prevented by forthcoming vaccines. GBS is under-reported in Ontario. Uncertainty about the potential impact of vaccine indicates that further analysis and research may be needed to prepare for policy-decision making, including clinical validation studies and an economic evaluation of GBS vaccination in Ontario.
Collapse
Affiliation(s)
- James Hartley
- Applied Immunization Research and Evaluation, Public Health Ontario, Toronto, Ontario, Canada
| | - Ye Li
- Knowledge Services, Public Health Ontario, Toronto, Ontario, Canada
| | - Liz Kunkel
- Applied Immunization Research and Evaluation, Public Health Ontario, Toronto, Ontario, Canada
| | - Natasha S. Crowcroft
- Applied Immunization Research and Evaluation, Public Health Ontario, Toronto, Ontario, Canada
| |
Collapse
|
32
|
Rappuoli R. Glycoconjugate vaccines: Principles and mechanisms. Sci Transl Med 2018; 10:10/456/eaat4615. [DOI: 10.1126/scitranslmed.aat4615] [Citation(s) in RCA: 104] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 08/10/2018] [Indexed: 12/25/2022]
|
33
|
Díaz-Dinamarca DA, Jerias JI, Soto DA, Soto JA, Díaz NV, Leyton YY, Villegas RA, Kalergis AM, Vásquez AE. The Optimisation of the Expression of Recombinant Surface Immunogenic Protein of Group B Streptococcus in Escherichia coli by Response Surface Methodology Improves Humoral Immunity. Mol Biotechnol 2018; 60:215-225. [PMID: 29442290 DOI: 10.1007/s12033-018-0065-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Group B Streptococcus (GBS) is the leading cause of neonatal meningitis and a common pathogen in livestock and aquaculture industries around the world. Conjugate polysaccharide and protein-based vaccines are under development. The surface immunogenic protein (SIP) is a conserved protein in all GBS serotypes and has been shown to be a good target for vaccine development. The expression of recombinant proteins in Escherichia coli cells has been shown to be useful in the development of vaccines, and the protein purification is a factor affecting their immunogenicity. The response surface methodology (RSM) and Box-Behnken design can optimise the performance in the expression of recombinant proteins. However, the biological effect in mice immunised with an immunogenic protein that is optimised by RSM and purified by low-affinity chromatography is unknown. In this study, we used RSM for the optimisation of the expression of the rSIP, and we evaluated the SIP-specific humoral response and the property to decrease the GBS colonisation in the vaginal tract in female mice. It was observed by NI-NTA chromatography that the RSM increases the yield in the expression of rSIP, generating a better purification process. This improvement in rSIP purification suggests a better induction of IgG anti-SIP immune response and a positive effect in the decreased GBS intravaginal colonisation. The RSM applied to optimise the expression of recombinant proteins with immunogenic capacity is an interesting alternative in the evaluation of vaccines in preclinical phase, which could improve their immune response.
Collapse
Affiliation(s)
- Diego A Díaz-Dinamarca
- Sección Biotecnología, Instituto de Salud Pública de Chile, Santiago, Chile.,Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Millenium Institute on Immunology and Immunotherapy, Santiago, Chile
| | - José I Jerias
- Sección Biotecnología, Instituto de Salud Pública de Chile, Santiago, Chile
| | - Daniel A Soto
- Sección Biotecnología, Instituto de Salud Pública de Chile, Santiago, Chile
| | - Jorge A Soto
- Sección Biotecnología, Instituto de Salud Pública de Chile, Santiago, Chile.,Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Millenium Institute on Immunology and Immunotherapy, Santiago, Chile
| | - Natalia V Díaz
- Sección Biotecnología, Instituto de Salud Pública de Chile, Santiago, Chile
| | - Yessica Y Leyton
- Sección Biotecnología, Instituto de Salud Pública de Chile, Santiago, Chile
| | - Rodrigo A Villegas
- Departamento de Asuntos Científicos, Instituto de Salud Pública de Chile, Santiago, Chile
| | - Alexis M Kalergis
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Millenium Institute on Immunology and Immunotherapy, Santiago, Chile.,Departamento de Endocrinología, Facultad de Medicina Pontificia, Universidad Católica de Chile, Santiago, Chile
| | - Abel E Vásquez
- Sección Biotecnología, Instituto de Salud Pública de Chile, Santiago, Chile. .,Facultad de Ciencia, Universidad San Sebastián, Providencia, Santiago, Chile.
| |
Collapse
|
34
|
Dzanibe S, Madhi SA. Systematic review of the clinical development of group B streptococcus serotype-specific capsular polysaccharide-based vaccines. Expert Rev Vaccines 2018; 17:635-651. [PMID: 29961350 DOI: 10.1080/14760584.2018.1496021] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Vaccination against group B Streptococcus (GBS) during pregnancy could provide protection against disease in the mother, fetus, and newborn. Immunity through transplacental acquired antibodies in the newborns could persist through early infancy, reducing the risk of early-onset (<7 days age) and late-onset (7-89 days age) disease. We conducted a systematic review of clinical trials on GBS capsular polysaccharide (CPS) vaccine to assess its safety and immunogenicity in pregnant and nonpregnant adults. AREAS COVERED We searched literature databases PubMed (Medline), Scopus, and the Cochrane library and identified 25 unique records on GBS CPS vaccines with or without conjugant protein. EXPERT COMMENTARY GBS vaccines were well tolerated, with mild local reactogenicity being the main solicited adverse event and no difference in reporting of other serious adverse events compared to placebo recipients. CPS vaccines conjugated to immunogenic proteins induced ≥fourfold increase of serotype-specific antibodies with high longevity (1-2 years); and capable of promoting homotypic GBS opsonophagocytic killing. Feto-maternal transplacental antibody ratio of serotype-specific IgG ranged between 0.49 and 0.81. The clinical relevance of these immunogenicity studies, however, need to be weighed against a correlate of protection against invasive GBS disease in infants, which is yet to be established using a universally accepted standardized assay.
Collapse
Affiliation(s)
- Sonwabile Dzanibe
- a Department of Science and Technology/National Research Foundation: Vaccine Preventable Diseases, Faculty of Health Sciences , University of the Witwatersrand , Johannesburg , South Africa.,b Medical Research Council: Respiratory and Meningeal Pathogens Research Unit, Faculty of Health Sciences , University of the Witwatersrand , Johannesburg , South Africa.,c Division of Immunology , University of Cape Town , Cape Town , South Africa
| | - Shabir A Madhi
- a Department of Science and Technology/National Research Foundation: Vaccine Preventable Diseases, Faculty of Health Sciences , University of the Witwatersrand , Johannesburg , South Africa.,b Medical Research Council: Respiratory and Meningeal Pathogens Research Unit, Faculty of Health Sciences , University of the Witwatersrand , Johannesburg , South Africa
| |
Collapse
|
35
|
Madhi SA, Koen A, Cutland CL, Jose L, Govender N, Wittke F, Olugbosi M, Sobanjo-Ter Meulen A, Baker S, Dull PM, Narasimhan V, Slobod K. Antibody Kinetics and Response to Routine Vaccinations in Infants Born to Women Who Received an Investigational Trivalent Group B Streptococcus Polysaccharide CRM197-Conjugate Vaccine During Pregnancy. Clin Infect Dis 2018; 65:1897-1904. [PMID: 29029127 PMCID: PMC5848233 DOI: 10.1093/cid/cix666] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Accepted: 08/22/2017] [Indexed: 11/27/2022] Open
Abstract
Background Maternal vaccination against group B Streptococcus (GBS) might provide protection against invasive GBS disease in infants. We investigated the kinetics of transplacentally transferred GBS serotype-specific capsular antibodies in the infants and their immune response to diphtheria toxoid and pneumococcal vaccination. Methods This phase 1b/2, observer-blind, single-center study (NCT01193920) enrolled infants born to women previously randomized (1:1:1:1) to receive either GBS vaccine at dosages of 0.5, 2.5, or 5.0 μg of each of 3 CRM197-glycoconjugates (serotypes Ia, Ib, and III), or placebo. Infants received routine immunization: combination diphtheria vaccine (diphtheria-tetanus-acellular pertussis–inactivated poliovirus/Haemophilus influenzae type b vaccine; age 6/10/ 14 weeks) and 13-valent pneumococcal CRM197-conjugate vaccine (PCV13; age 6/14 weeks and 9 months). Antibody levels were assessed at birth, day (D) 43, and D91 for GBS serotypes; 1 month postdose 3 (D127) for diphtheria; and 1 month postprimary (D127) and postbooster (D301) doses for pneumococcal serotypes. Results Of 317 infants enrolled, 295 completed the study. In infants of GBS vaccine recipients, GBS serotype-specific antibody geometric mean concentrations were significantly higher than in the placebo group at all timepoints and predictably decreased to 41%–61% and 26%–76% of birth levels by D43 and D91, respectively. Across all groups, ≥95% of infants were seroprotected against diphtheria at D127 and ≥91% of infants had seroprotective antibody levels against each PCV13 pneumococcal serotype at D301. Conclusions Maternal vaccination with an investigational CRM197-glycoconjugate GBS vaccine elicited higher GBS serotype-specific antibody levels in infants until 90 days of age, compared with a placebo group, and did not affect infant immune responses to diphtheria toxoid and pneumococcal vaccination. Clinical Trials Registration NCT01193920.
Collapse
Affiliation(s)
- Shabir A Madhi
- Medical Research Council: Respiratory and Meningeal Pathogens Research Unit.,Department of Science and Technology/National Research Foundation: Vaccine Preventable Diseases, University of the Witwatersrand.,National Institute for Communicable Diseases, National Health Laboratory Service, Centre for Vaccines and Immunology, Johannesburg, South Africa
| | - Anthonet Koen
- Medical Research Council: Respiratory and Meningeal Pathogens Research Unit.,Department of Science and Technology/National Research Foundation: Vaccine Preventable Diseases, University of the Witwatersrand
| | - Clare L Cutland
- Medical Research Council: Respiratory and Meningeal Pathogens Research Unit.,Department of Science and Technology/National Research Foundation: Vaccine Preventable Diseases, University of the Witwatersrand
| | - Lisa Jose
- Medical Research Council: Respiratory and Meningeal Pathogens Research Unit.,Department of Science and Technology/National Research Foundation: Vaccine Preventable Diseases, University of the Witwatersrand
| | - Niresha Govender
- Medical Research Council: Respiratory and Meningeal Pathogens Research Unit.,Department of Science and Technology/National Research Foundation: Vaccine Preventable Diseases, University of the Witwatersrand
| | | | | | | | - Sherryl Baker
- GSK and Novartis Vaccines Division, Cambridge, Massachusetts
| | - Peter M Dull
- GSK and Novartis Vaccines Division, Cambridge, Massachusetts
| | - Vas Narasimhan
- GSK and Novartis Vaccines Division, Cambridge, Massachusetts
| | - Karen Slobod
- GSK and Novartis Vaccines Division, Cambridge, Massachusetts
| |
Collapse
|
36
|
Abstract
The problem of antimicrobial resistance (AMR) and the associated morbidity and mortality due to antibiotic resistant bacterial pathogens is not new. However, AMR has been increasing at an alarming rate with appearances of diseases caused by bacteria exhibiting resistance to not just one but multiple classes of antibiotics. The World Health Organization (WHO) supported by governments, health ministries and health agencies has formulated global action plans to combat the rise in AMR, supporting a number of proven initiatives such as antimicrobial stewardship, investments in development of new classes of antibiotics, and educational programs designed to eliminate inappropriate antibiotic use. Vaccines as tools to reduce AMR have historically been under-recognized, yet the positive effect in reducing AMR has been well established. For example Haemophilus influenzae type B (Hib) as well as Streptococcus pneumoniae (pneumococcal) conjugate vaccines have impressive track records in not only preventing life threatening diseases caused by these bacteria, but also reducing antibiotic use and AMR. This paper will describe the drivers of antibiotic use and subsequent development of AMR; it will make the case how existing vaccines are already participating in combatting AMR, describe future prospects for the role of new vaccines in development to reduce AMR, and highlight challenges associated with future vaccine development to combat AMR.
Collapse
Affiliation(s)
- Kathrin U Jansen
- a Pfizer Vaccine Research and Development , Pearl River , NY , USA
| | | |
Collapse
|
37
|
Shabayek S, Spellerberg B. Group B Streptococcal Colonization, Molecular Characteristics, and Epidemiology. Front Microbiol 2018; 9:437. [PMID: 29593684 PMCID: PMC5861770 DOI: 10.3389/fmicb.2018.00437] [Citation(s) in RCA: 172] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Accepted: 02/26/2018] [Indexed: 11/13/2022] Open
Abstract
Streptococcus agalactiae or group B streptococcus (GBS) is a leading cause of serious neonatal infections. GBS is an opportunistic commensal constituting a part of the intestinal and vaginal physiologic flora and maternal colonization is the principal route of GBS transmission. GBS is a pathobiont that converts from the asymptomatic mucosal carriage state to a major bacterial pathogen causing severe invasive infections. At present, as many as 10 serotypes (Ia, Ib, and II–IX) are recognized. The aim of the current review is to shed new light on the latest epidemiological data and clonal distribution of GBS in addition to discussing the most important colonization determinants at a molecular level. The distribution and predominance of certain serotypes is susceptible to variations and can change over time. With the availability of multilocus sequence typing scheme (MLST) data, it became clear that GBS strains of certain clonal complexes possess a higher potential to cause invasive disease, while other harbor mainly colonizing strains. Colonization and persistence in different host niches is dependent on the adherence capacity of GBS to host cells and tissues. Bacterial biofilms represent well-known virulence factors with a vital role in persistence and chronic infections. In addition, GBS colonization, persistence, translocation, and invasion of host barriers are largely dependent on their adherence abilities to host cells and extracellular matrix proteins (ECM). Major adhesins mediating GBS interaction with host cells include the fibrinogen-binding proteins (Fbs), the laminin-binding protein (Lmb), the group B streptococcal C5a peptidase (ScpB), the streptococcal fibronectin binding protein A (SfbA), the GBS immunogenic bacterial adhesin (BibA), and the hypervirulent adhesin (HvgA). These adhesins facilitate persistent and intimate contacts between the bacterial cell and the host, while global virulence regulators play a major role in the transition to invasive infections. This review combines for first time epidemiological data with data on adherence and colonization for GBS. Investigating the epidemiology along with understanding the determinants of mucosal colonization and the development of invasive disease at a molecular level is therefore important for the development of strategies to prevent invasive GBS disease worldwide.
Collapse
Affiliation(s)
- Sarah Shabayek
- Department of Microbiology and Immunology, Faculty of Pharmacy, Suez Canal University, Ismailia, Egypt
| | - Barbara Spellerberg
- Institute of Medical Microbiology and Hygiene, University of Ulm, Ulm, Germany
| |
Collapse
|
38
|
Munoz FM. Current Challenges and Achievements in Maternal Immunization Research. Front Immunol 2018; 9:436. [PMID: 29559976 PMCID: PMC5845678 DOI: 10.3389/fimmu.2018.00436] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2017] [Accepted: 02/19/2018] [Indexed: 12/22/2022] Open
Abstract
Maternal immunization has the potential to significantly improve maternal and child health worldwide by reducing maternal and infant morbidity and mortality associated with disease caused by pathogens that are particularly relevant in the perinatal period and in early life, and for which no alternative effective preventive strategies exist. Research on all aspects related to vaccines for administration during pregnancy is ongoing with support of multiple stakeholders and global participation. Substantial progress has been made, and the availability of new vaccines licensed exclusively for use in pregnant women to protect their infants has become an achievable goal. This review provides an update of the current challenges and achievements in maternal immunization research, focusing on recent milestones that advance the field and the prospects to make maternal immunization a feasible and accessible strategy to improve global health.
Collapse
Affiliation(s)
- Flor M. Munoz
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, United States
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
39
|
Lin SM, Zhi Y, Ahn KB, Lim S, Seo HS. Status of group B streptococcal vaccine development. Clin Exp Vaccine Res 2018; 7:76-81. [PMID: 29399583 PMCID: PMC5795048 DOI: 10.7774/cevr.2018.7.1.76] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Revised: 01/08/2018] [Accepted: 01/14/2018] [Indexed: 12/22/2022] Open
Abstract
Streptococcus agalactiae (group B streptococcus, GBS) is a leading causal organism of neonatal invasive diseases and severe infections in the elderly. Despite significant advances in the diagnosis and treatment of GBS infections and improvement in personal hygiene standards, this pathogen is still a global health concern. Thus, an effective vaccine against GBS would augment existing strategies to substantially decrease GBS infection. In 2014, World Health Organization convened the first meeting for consultation on GBS vaccine development, focusing on the GBS maternal immunization program, which was aimed at reducing infections in neonates and young infants worldwide. Here, we review the history of GBS infections, the current vaccine candidates, and the current status of immunogenicity assays used to evaluate the clinical efficacy of GBS vaccines.
Collapse
Affiliation(s)
- Shun Mei Lin
- Research Division for Biotechnology, Korea Atomic Energy Research Institute, Jeongeup, Korea.,Department of Molecular Medicine (BrainKorea21 Plus), Chonnam National University Graduate School, Gwangju, Korea
| | - Yong Zhi
- Research Division for Biotechnology, Korea Atomic Energy Research Institute, Jeongeup, Korea.,Department of Radiation Biotechnology and Applied Radioisotope Science, University of Science and Technology, Daejeon, Korea
| | - Ki Bum Ahn
- Research Division for Biotechnology, Korea Atomic Energy Research Institute, Jeongeup, Korea
| | - Sangyong Lim
- Research Division for Biotechnology, Korea Atomic Energy Research Institute, Jeongeup, Korea.,Department of Radiation Biotechnology and Applied Radioisotope Science, University of Science and Technology, Daejeon, Korea
| | - Ho Seong Seo
- Research Division for Biotechnology, Korea Atomic Energy Research Institute, Jeongeup, Korea.,Department of Radiation Biotechnology and Applied Radioisotope Science, University of Science and Technology, Daejeon, Korea
| |
Collapse
|
40
|
Jansen KU, Knirsch C, Anderson AS. The role of vaccines in preventing bacterial antimicrobial resistance. Nat Med 2018; 24:10-19. [DOI: 10.1038/nm.4465] [Citation(s) in RCA: 147] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Accepted: 12/04/2017] [Indexed: 01/03/2023]
|
41
|
Association between antibodies against group B Streptococcus surface proteins and recto-vaginal colonisation during pregnancy. Sci Rep 2017; 7:16454. [PMID: 29184151 PMCID: PMC5705700 DOI: 10.1038/s41598-017-16757-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Accepted: 11/16/2017] [Indexed: 11/08/2022] Open
Abstract
Group B Streptococcus (GBS) recto-vaginal colonisation in pregnant women is the major risk factor for early-onset invasive GBS disease in their newborns. We aimed to determine the association between serum antibody levels against 11 GBS surface proteins and recto-vaginal acquisition of GBS colonisation during pregnancy. Sera collected from pregnant women at 20-25 weeks and ≥37 weeks of gestation age were measured for IgG titres against GBS surface proteins using a multiplex immunoassay. Women were evaluated for recto-vaginal colonisation every 4-5 weeks. We observed that the likelihood of becoming colonised with GBS during pregnancy was lower in women with IgG titres ≥200 U/mL against gbs0233 (adjusted OR = 0.47 [95% CI: 0.25-0.89], p = 0.021) and ≥85 U/mL for gbs1539 (adjusted OR = 0.44 [95% CI: 0.24-0.82], p = 0.01) when comparing between women who acquired GBS colonisation and those that remained free of GBS colonisation throughout pregnancy. IgG titres (U/mL) specific to BibA and Sip were higher in pregnant women colonised with GBS (380.19 and 223.87, respectively) compared to women with negative GBS cultures (234.42 and 186.21, respectively; p < 0.01) at ≥37 weeks gestation. Antibodies induced by gbs0233 and gbs1539 were associated with a reduced likelihood of recto-vaginal GBS acquisition during pregnancy and warrant further investigation as vaccine targets.
Collapse
|
42
|
Abstract
Group B Streptococcal isolates (n = 235) from the South of Ireland were characterised by serotyping, antimicrobial susceptibility and determination of the phenotypic and genotypic mechanisms of resistance. Resistance to erythromycin and clindamycin was observed in 21·3% and 20·4% of the total population, respectively. The c-MLSB phenotype was the most common phenotype detected (62%), with ermB being the predominant genetic determinant, present in 84% of resistant isolates. The rare L phenotype was observed in 2·9% (n = 7) of isolates, four of which harboured the lsaC gene responsible for clindamycin resistance. Serotypes Ia, III and II were the most common amongst the entire study population (28·1%, 24·7% and 14%, respectively). Four of the seven L phenotype isolates were serotype III and two of these strains were confirmed as the hypervirulent clone, ST-17 and harboured the hvgA gene. This is the first documented case of the L phenotype in Ireland to date and the study findings emphasise the need for continued monitoring of antibiotic resistance and serotype distribution in GBS isolates from Ireland.
Collapse
|
43
|
Cost-effectiveness of a potential group B streptococcal vaccine for pregnant women in the United States. Vaccine 2017; 35:6238-6247. [PMID: 28951085 DOI: 10.1016/j.vaccine.2017.08.085] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 08/07/2017] [Accepted: 08/29/2017] [Indexed: 11/23/2022]
Abstract
BACKGROUND In the U.S., intrapartum antibiotic prophylaxis (IAP) for pregnant women colonized with group B streptococcus (GBS) has reduced GBS disease in the first week of life (early-onset/EOGBS). Nonetheless, GBS remains a leading cause of neonatal sepsis, including 1000 late-onset (LOGBS) cases annually. A maternal vaccine under development could prevent EOGBS and LOGBS. METHODS Using a decision-analytic model, we compared the public health impact, costs, and cost-effectiveness of five strategies to prevent GBS disease in infants: (1) no prevention; (2) currently recommended screening/IAP; (3) maternal GBS immunization; (4) maternal immunization with IAP when indicated for unimmunized women; (5) maternal immunization plus screening/IAP for all women. We modeled a pentavalent vaccine covering serotypes 1a, 1b, II, III, and V, which cause almost all GBS disease. RESULTS In the base case, screening/IAP alone prevents 46% of EOGBS compared to no prevention, at a cost of $70,275 per quality-adjusted life-year (QALY) from a healthcare and $51,249/QALY from a societal perspective (2013 US$). At coverage rates typical of maternal vaccines in the U.S., a pentavalent vaccine alone would not prevent as much disease as screening/IAP until its efficacy approached 90%, but would cost less per QALY. At vaccine efficacy of ≥70%, maternal immunization together with IAP for unimmunized women would prevent more disease than screening/IAP, at a similar cost/QALY. CONCLUSIONS GBS maternal immunization, with IAP as indicated for unvaccinated women, could be an attractive alternative to screening/IAP if a pentavalent vaccine is sufficiently effective. Coverage, typically low for maternal vaccines, is key to the vaccine's public health impact.
Collapse
|
44
|
Abstract
Maternal immunization has undergone a paradigm shift in recent years, as women and healthcare providers accept and recognize the benefits of this strategy not only for the pregnant woman but also for the developing fetus and young infant. This article reviews the evidence for active immunization during pregnancy, with an emphasis on perinatal and infant outcomes. Current recommendations for immunization during pregnancy are presented, with particular focus on the routinely recommended vaccines during pregnancy: influenza and Tdap (tetanus, diphtheria, and pertussis). We discuss future research directions, maternal vaccines in development, and considerations for optimizing and advancing this underutilized strategy.
Collapse
Affiliation(s)
- Kirsten P Perrett
- Vaccine and Immunisation Research Group, Murdoch Childrens Research Institute and Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, VIC, Australia.
- Melbourne Children's Trial Centre, Murdoch Childrens Research Institute, Melbourne, VIC, Australia.
- Department of General Medicine, Royal Children's Hospital, Melbourne, VIC, Australia.
| | - Terry M Nolan
- Vaccine and Immunisation Research Group, Murdoch Childrens Research Institute and Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
45
|
Nielsen M, Sheikh N, Fitzgerald E, Meehan M, LeBlanc D, Eogan M, El-Khuffash A, Drew RJ. Screening for early-onset invasive group B Streptococcal disease in neonates in an Irish hospital (2001-2014): a retrospective audit. Infect Dis (Lond) 2017; 49:466-470. [PMID: 28276804 DOI: 10.1080/23744235.2017.1285045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
Abstract
Group B Streptococcus (GBS) is the most common cause of early-onset neonatal sepsis and meningitis. In babies with no clinical suspicion of infection, who are at risk of early-onset invasive disease based on maternal risk factors, blood cultures are taken to detect bacteraemia. In our institution, lumbar punctures are performed in infants with clinical signs of sepsis but not in infants who are well at the time of screening. Between 2001 and 2014, there were 112,361 live births weighing >500 g, of whom 13,959 (12.4%) infants had a blood culture taken on the first or second day of life, and 1971 (14.1%) of these infants had lumbar punctures on these first two days of life. Fifty-three cases of early-onset GBS disease were identified. Only three patients with invasive GBS disease had no clinical suspicion for sepsis at the time of testing. Thus, the number of blood cultures taken to detect one case of GBS bacteraemia in an infant who is well at the time of testing was 3996.
Collapse
Affiliation(s)
- Maryke Nielsen
- a Department of Microbiology , Rotunda Hospital , Dublin 1 , Ireland
| | - Naveed Sheikh
- b Department of Neonatology , Rotunda Hospital , Dublin 1 , Ireland
| | - Eoin Fitzgerald
- b Department of Neonatology , Rotunda Hospital , Dublin 1 , Ireland
| | - Mary Meehan
- c Irish Meningitis and Sepsis Reference Laboratory , Temple Street Children's University Hospital , Dublin 7 , Ireland
| | - David LeBlanc
- a Department of Microbiology , Rotunda Hospital , Dublin 1 , Ireland
| | - Maeve Eogan
- d Department of Obstetrics , Rotunda Hospital , Dublin 1 , Ireland
| | - Afif El-Khuffash
- b Department of Neonatology , Rotunda Hospital , Dublin 1 , Ireland
| | - Richard J Drew
- a Department of Microbiology , Rotunda Hospital , Dublin 1 , Ireland.,c Irish Meningitis and Sepsis Reference Laboratory , Temple Street Children's University Hospital , Dublin 7 , Ireland.,e Department of Microbiology , Royal College of Surgeons in Ireland , Dublin 2 , Ireland
| |
Collapse
|
46
|
Madhi SA, Dangor Z. Prospects for preventing infant invasive GBS disease through maternal vaccination. Vaccine 2017; 35:4457-4460. [PMID: 28237500 DOI: 10.1016/j.vaccine.2017.02.025] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 02/05/2017] [Accepted: 02/08/2017] [Indexed: 11/26/2022]
Abstract
Group B streptococcus (GBS) is a leading cause of neonatal sepsis, with the highest incidence (1.3 per 1000 live births) reported from Africa. Although the incidence of invasive GBS disease is reportedly low in South Asia, there is disconnect between prevalence of maternal recto-vaginal colonization and the incidence of early-onset disease (EOD). This is possibly due to case-ascertainment biases that omit investigation of newborns dying on day-0 of life, which accounts for >90% of EOD. Furthermore, GBS is associated with approximately 15% of all infection related stillbirths. Vaccination of pregnant women with a serotype-specific polysaccharide epitope vaccine could possibly protect against EOD and late-onset disease (LOD) in their infants through transplacental transfer of serotype-specific capsular antibody. Furthermore, vaccination of pregnant women might also protect against impaired neurodevelopment following GBS associated neonatal sepsis, and fetal loss/stillbirths. Licensure of a GBS vaccine might be feasible based on safety evaluation and a sero-correlate of protection, with vaccine effectiveness subsequently being demonstrated in phase IV studies. A randomized-controlled trial would, however, be best suited as a vaccine-probe to fully characterize the contribution of GBS to neonatal sepsis associated morbidity and mortality and adverse fetal outcomes.
Collapse
Affiliation(s)
- Shabir A Madhi
- Medical Research Council, Respiratory and Meningeal Pathogens Research Unit, Faculty of Health Sciences, University of the Witwatersrand, South Africa; Department of Science and Technology/National Research Foundation, Vaccine Preventable Diseases, Faculty of Health Sciences, University of the Witwatersrand, South Africa.
| | - Ziyaad Dangor
- Medical Research Council, Respiratory and Meningeal Pathogens Research Unit, Faculty of Health Sciences, University of the Witwatersrand, South Africa; Department of Science and Technology/National Research Foundation, Vaccine Preventable Diseases, Faculty of Health Sciences, University of the Witwatersrand, South Africa; Department of Paediatrics, Faculty of Health Sciences, University of the Witwatersrand, South Africa.
| |
Collapse
|
47
|
Serotype Distribution, Population Structure, and Antimicrobial Resistance of Group B Streptococcus Strains Recovered from Colonized Pregnant Women. J Clin Microbiol 2016; 55:412-422. [PMID: 27852675 DOI: 10.1128/jcm.01615-16] [Citation(s) in RCA: 83] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Accepted: 11/12/2016] [Indexed: 11/20/2022] Open
Abstract
Using serotyping, multilocus sequence typing, and whole-genome sequencing (WGS) of selected strains, we studied the population structure of 102 group B Streptococcus (GBS) isolates prospectively sampled in 2014 from vaginal/rectal swabs of healthy pregnant women in metropolitan Toronto, Canada. We also determined the susceptibilities of each of the colonizing isolates to penicillin, erythromycin, clindamycin, tetracycline, and other antimicrobial agents. Overall, we observed a high rate of tetracycline resistance (89%) among colonizing GBS isolates. We found resistance to erythromycin in 36% of the strains, and 33% were constitutively or inducibly resistant to clindamycin. The most frequently identified serotypes were III (25%), Ia (23%), and V (19%). Serotype IV accounted for 6% of the colonizing isolates, a rate consistent with that observed among patients with invasive GBS infections in metropolitan Toronto. The majority of serotype IV isolates belonged to sequence type (ST)459, a tetracycline-, erythromycin-, and clindamycin-resistant ST first identified in Minnesota, which is considered to be the main driver of serotype IV GBS expansion in North America. WGS revealed that ST459 isolates from Canada are clonally related to colonizing and invasive ST459 organisms circulating in regions of the United States. We also used WGS to study recombination in selected colonizing strains from metropolitan Toronto, which revealed multiple episodes of capsular switching. Present and future circulating GBS organisms and their genetic diversity may influence GBS vaccine development.
Collapse
|
48
|
Abstract
INTRODUCTION The success of the vaccines available on the market has significantly increased interest in vaccine development. Areas covered: The main aim of this paper is to discuss the most important vaccines of pediatric interest that are currently being developed. New pneumococcal vaccines and vaccines against group B Streptococcus, Staphylococcus aureus and respiratory syncytial virus are analyzed in detail. Expert commentary: Advances in understanding human immunology, including human monoclonal antibody identification, sequencing technology, and the ability to solve atomic level structures of vaccine targets have provided tools to guide the rational design of future vaccines. It is likely that some of these vaccines will reach the market in the future and will thus partially contribute to the prevention of very severe diseases that significantly affect the morbidity and mortality of children. However, further studies in animals and several clinical trials in children must be performed before new vaccines become licensed.
Collapse
Affiliation(s)
- Susanna Esposito
- a Pediatric Highly Intensive Care Unit, Department of Pathophysiology and Transplantation , Università degli Studi di Milano, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico , Milan , Italy
| | - Nicola Principi
- a Pediatric Highly Intensive Care Unit, Department of Pathophysiology and Transplantation , Università degli Studi di Milano, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico , Milan , Italy
| |
Collapse
|
49
|
Kobayashi M, Vekemans J, Baker CJ, Ratner AJ, Le Doare K, Schrag SJ. Group B Streptococcus vaccine development: present status and future considerations, with emphasis on perspectives for low and middle income countries. F1000Res 2016; 5:2355. [PMID: 27803803 PMCID: PMC5070600 DOI: 10.12688/f1000research.9363.1] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/08/2016] [Indexed: 01/07/2023] Open
Abstract
Globally, group B Streptococcus (GBS) remains the leading cause of sepsis and meningitis in young infants, with its greatest burden in the first 90 days of life. Intrapartum antibiotic prophylaxis (IAP) for women at risk of transmitting GBS to their newborns has been effective in reducing, but not eliminating, the young infant GBS disease burden in many high income countries. However, identification of women at risk and administration of IAP is very difficult in many low and middle income country (LMIC) settings, and is not possible for home deliveries. Immunization of pregnant women with a GBS vaccine represents an alternate pathway to protecting newborns from GBS disease, through the transplacental antibody transfer to the fetus in utero. This approach to prevent GBS disease in young infants is currently under development, and is approaching late stage clinical evaluation. This manuscript includes a review of the natural history of the disease, global disease burden estimates, diagnosis and existing control options in different settings, the biological rationale for a vaccine including previous supportive studies, analysis of current candidates in development, possible correlates of protection and current status of immunogenicity assays. Future potential vaccine development pathways to licensure and use in LMICs, trial design and implementation options are discussed, with the objective to provide a basis for reflection, rather than recommendations.
Collapse
Affiliation(s)
- Miwako Kobayashi
- National Center for Immunization and Respiratory Diseases, Division of Bacterial Diseases, Centers for Disease Control and Prevention, Atlanta, USA
- Epidemic Intelligence Service, Centers for Disease Control and Prevention, Atlanta, USA
| | - Johan Vekemans
- Initiative for Vaccine Research, World Health Organization, Geneva, Switzerland
| | - Carol J. Baker
- Department of Pediatrics, Baylor College of Medicine, Houston, USA
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, USA
- Center for Vaccine Awareness and Research, Texas Children's Hospital, Houston, USA
| | - Adam J. Ratner
- Departments of Pediatrics and Microbiology, New York University School of Medicine, New York, USA
| | - Kirsty Le Doare
- Centre for International Child Health, Imperial College, London, UK
| | - Stephanie J. Schrag
- National Center for Immunization and Respiratory Diseases, Division of Bacterial Diseases, Centers for Disease Control and Prevention, Atlanta, USA
| |
Collapse
|
50
|
Dzanibe S, Adrian PV, Kimaro Mlacha SZ, Madhi SA. Natural acquired group B Streptococcus capsular polysaccharide and surface protein antibodies in HIV-infected and HIV-uninfected children. Vaccine 2016; 34:5217-5224. [PMID: 27663669 DOI: 10.1016/j.vaccine.2016.09.015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Revised: 09/02/2016] [Accepted: 09/14/2016] [Indexed: 01/03/2023]
Abstract
Group B Streptococcus (GBS) is a major cause of invasive disease in young infants and also in older immunocompromised individuals, including HIV-infected persons. We compared naturally acquired antibody titres to GBS polysaccharide and surface protein antigens in HIV-uninfected and HIV-infected children aged 4-7 years. A multiplex Luminex immunoassay was used to measure IgG concentrations against GBS capsular polysaccharides (CPS) for serotypes Ia, Ib, III and V; and also extracellular localizing proteins which included cell-wall anchored proteins: Fibrinogen binding surface Antigen (FbsA), GBS Immunogenic Bacterial Adhesin (BibA), Surface immunogenic protein (Sip), gbs0393, gbs1356, gbs1539, gbs0392; and lipoproteins gbs0233, gbs2106 and Foldase PsrA. HIV-infected children (n=68) had significantly lower IgG GMT compared to HIV-uninfected (n=77) children against CPS of serotype Ib (p=0.012) and V (p=0.0045), and surface proteins Sip (p<0.001) and gbs2106 (p=0.0014). IgG GMT against GBS surface proteins: FbsA, gbs1539, gbs1356, gbs0392, gbs0393 and Foldase PsrA were significantly higher in HIV-infected children (p<0.004). Moreover, amongst HIV infected children, IgG GMT to GBS surface proteins were higher in those with CD4+ lymphocyte counts <500cell/μL compared to those who had CD4+ lymphocyte count ⩾500cell/μL with the exception of Sip. The increased susceptibility to invasive GBS disease in HIV-infected individuals could be due to the lower serotype specific capsular antibody and possibly due to lower antibody to some of the GBS proteins such as Sip and gbs2106.
Collapse
Affiliation(s)
- Sonwabile Dzanibe
- Department of Science and Technology/National Research Foundation: Vaccine Preventable Diseases, University of the Witwatersrand, Johannesburg, South Africa; MRC, Respiratory and Meningeal Pathogens Research Unit, University of the Witwatersrand, Johannesburg, South Africa
| | - Peter V Adrian
- Department of Science and Technology/National Research Foundation: Vaccine Preventable Diseases, University of the Witwatersrand, Johannesburg, South Africa; MRC, Respiratory and Meningeal Pathogens Research Unit, University of the Witwatersrand, Johannesburg, South Africa.
| | - Sheila Z Kimaro Mlacha
- Department of Science and Technology/National Research Foundation: Vaccine Preventable Diseases, University of the Witwatersrand, Johannesburg, South Africa; MRC, Respiratory and Meningeal Pathogens Research Unit, University of the Witwatersrand, Johannesburg, South Africa
| | - Shabir A Madhi
- Department of Science and Technology/National Research Foundation: Vaccine Preventable Diseases, University of the Witwatersrand, Johannesburg, South Africa; MRC, Respiratory and Meningeal Pathogens Research Unit, University of the Witwatersrand, Johannesburg, South Africa; National Institutes for Communicable Diseases, Johannesburg, South Africa
| |
Collapse
|