Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 55
Filtrer
1.
NPJ Vaccines ; 9(1): 171, 2024 Sep 17.
Article de Anglais | MEDLINE | ID: mdl-39289377

RÉSUMÉ

The relative conservation of the influenza hemagglutinin (HA) stem compared to that of the immunodominant HA head makes the HA stem an attractive target for broadly protective influenza vaccines. Here we report the first-in-human, dose-escalation, open-label trial (NCT04579250) evaluating an unadjuvanted group 2 stabilized stem ferritin nanoparticle vaccine based on the H10 A/Jiangxi-Donghu/346/2013 influenza HA, H10ssF, in healthy adults. Participants received a single 20 mcg dose (n = 3) or two 60 mcg doses 16 weeks apart (n = 22). Vaccination with H10ssF was safe and well tolerated with only mild systemic and local reactogenicity reported. No serious adverse events occurred. Vaccination significantly increased homologous H10 HA stem binding and neutralizing antibodies at 2 weeks after both first and second vaccinations, and these responses remained above baseline at 40 weeks. Heterologous H3 and H7 binding antibodies also significantly increased after each vaccination and remained elevated throughout the study. These data indicate that the group 2 HA stem nanoparticle vaccine is safe and induces stem-directed binding and neutralizing antibodies.

2.
Front Immunol ; 15: 1399960, 2024.
Article de Anglais | MEDLINE | ID: mdl-38873606

RÉSUMÉ

The VH6-1 class of antibodies includes some of the broadest and most potent antibodies that neutralize influenza A virus. Here, we elicit and isolate anti-idiotype antibodies against germline versions of VH6-1 antibodies, use these to sort human leukocytes, and isolate a new VH6-1-class member, antibody L5A7, which potently neutralized diverse group 1 and group 2 influenza A strains. While its heavy chain derived from the canonical IGHV6-1 heavy chain gene used by the class, L5A7 utilized a light chain gene, IGKV1-9, which had not been previously observed in other VH6-1-class antibodies. The cryo-EM structure of L5A7 in complex with Indonesia 2005 hemagglutinin revealed a nearly identical binding mode to other VH6-1-class members. The structure of L5A7 bound to the isolating anti-idiotype antibody, 28H6E11, revealed a shared surface for binding anti-idiotype and hemagglutinin that included two critical L5A7 regions: an FG motif in the third heavy chain-complementary determining region (CDR H3) and the CDR L1 loop. Surprisingly, the chemistries of L5A7 interactions with hemagglutinin and with anti-idiotype were substantially different. Overall, we demonstrate anti-idiotype-based isolation of a broad and potent influenza A virus-neutralizing antibody, revealing that anti-idiotypic selection of antibodies can involve features other than chemical mimicry of the target antigen.


Sujet(s)
Anticorps anti-idiotypiques , Anticorps neutralisants , Anticorps antiviraux , Glycoprotéine hémagglutinine du virus influenza , Virus de la grippe A , Humains , Virus de la grippe A/immunologie , Anticorps antiviraux/immunologie , Anticorps neutralisants/immunologie , Anticorps neutralisants/isolement et purification , Anticorps anti-idiotypiques/immunologie , Anticorps anti-idiotypiques/isolement et purification , Glycoprotéine hémagglutinine du virus influenza/immunologie , Grippe humaine/immunologie , Grippe humaine/virologie , Animaux , Chaines lourdes des immunoglobulines/immunologie , Chaines lourdes des immunoglobulines/composition chimique
3.
Cell Rep ; 43(5): 114171, 2024 May 28.
Article de Anglais | MEDLINE | ID: mdl-38717904

RÉSUMÉ

Influenza A virus subtype H2N2, which caused the 1957 influenza pandemic, remains a global threat. A recent phase 1 clinical trial investigating a ferritin nanoparticle vaccine displaying H2 hemagglutinin (HA) in H2-naive and H2-exposed adults enabled us to perform comprehensive structural and biochemical characterization of immune memory on the breadth and diversity of the polyclonal serum antibody response elicited. We temporally map the epitopes targeted by serum antibodies after vaccine prime and boost, revealing that previous H2 exposure results in higher responses to the variable HA head domain. In contrast, initial responses in H2-naive participants are dominated by antibodies targeting conserved epitopes. We use cryoelectron microscopy and monoclonal B cell isolation to describe the molecular details of cross-reactive antibodies targeting conserved epitopes on the HA head, including the receptor-binding site and a new site of vulnerability deemed the medial junction. Our findings accentuate the impact of pre-existing influenza exposure on serum antibody responses post-vaccination.


Sujet(s)
Anticorps antiviraux , Mémoire immunologique , Sous-type H2N2 du virus de la grippe A , Vaccins antigrippaux , Vaccination , Humains , Anticorps antiviraux/immunologie , Vaccins antigrippaux/immunologie , Sous-type H2N2 du virus de la grippe A/immunologie , Grippe humaine/immunologie , Grippe humaine/prévention et contrôle , Production d'anticorps/immunologie , Glycoprotéine hémagglutinine du virus influenza/immunologie , Épitopes/immunologie , Adulte , Lymphocytes B/immunologie
4.
Immunity ; 57(3): 574-586.e7, 2024 Mar 12.
Article de Anglais | MEDLINE | ID: mdl-38430907

RÉSUMÉ

Continuously evolving influenza viruses cause seasonal epidemics and pose global pandemic threats. Although viral neuraminidase (NA) is an effective drug and vaccine target, our understanding of the NA antigenic landscape still remains incomplete. Here, we describe NA-specific human antibodies that target the underside of the NA globular head domain, inhibit viral propagation of a wide range of human H3N2, swine-origin variant H3N2, and H2N2 viruses, and confer both pre- and post-exposure protection against lethal H3N2 infection in mice. Cryo-EM structures of two such antibodies in complex with NA reveal non-overlapping epitopes covering the underside of the NA head. These sites are highly conserved among N2 NAs yet inaccessible unless the NA head tilts or dissociates. Our findings help guide the development of effective countermeasures against ever-changing influenza viruses by identifying hidden conserved sites of vulnerability on the NA underside.


Sujet(s)
Vaccins antigrippaux , Grippe humaine , Infections à Orthomyxoviridae , Humains , Animaux , Souris , Suidae , Protéines virales/génétique , Sialidase , Sous-type H3N2 du virus de la grippe A , Anticorps monoclonaux , Anticorps antiviraux
5.
Sci Immunol ; 9(93): eadj9534, 2024 03 22.
Article de Anglais | MEDLINE | ID: mdl-38517951

RÉSUMÉ

Antigenic drift, the gradual accumulation of amino acid substitutions in the influenza virus hemagglutinin (HA) receptor protein, enables viral immune evasion. Antibodies (Abs) specific for the drift-resistant HA stem region are a promising universal influenza vaccine target. Although anti-stem Abs are not believed to block viral attachment, here we show that complement component 1q (C1q), a 460-kilodalton protein with six Ab Fc-binding domains, confers attachment inhibition to anti-stem Abs and enhances their fusion and neuraminidase inhibition. As a result, virus neutralization activity in vitro is boosted up to 30-fold, and in vivo protection from influenza PR8 infection in mice is enhanced. These effects reflect increased steric hindrance and not increased Ab avidity. C1q greatly expands the anti-stem Ab viral escape repertoire to include residues throughout the HA, some of which cause antigenic alterations in the globular region or modulate HA receptor avidity. We also show that C1q enhances the neutralization activity of non-receptor binding domain anti-SARS-CoV-2 spike Abs, an effect dependent on spike density on the virion surface. These findings demonstrate that C1q can greatly expand Ab function and thereby contribute to viral evolution and immune escape.


Sujet(s)
Vaccins antigrippaux , Grippe humaine , Souris , Animaux , Humains , Hémagglutinines , Complément C1q , Attachement viral , Glycoprotéine hémagglutinine du virus influenza , Anticorps antiviraux
6.
Sci Transl Med ; 16(728): eadd5960, 2024 01 03.
Article de Anglais | MEDLINE | ID: mdl-38170789

RÉSUMÉ

Durable humoral immunity is mediated by long-lived plasma cells (LLPCs) that reside in the bone marrow. It remains unclear whether severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike protein vaccination is able to elicit and maintain LLPCs. Here, we describe a sensitive method to identify and isolate antigen-specific LLPCs by tethering antibodies secreted by these cells onto the cell surface. Using this method, we found that two doses of adjuvanted SARS-CoV-2 spike protein vaccination are able to induce spike protein-specific LLPC reservoirs enriched for receptor binding domain specificities in the bone marrow of nonhuman primates that are detectable for several months after vaccination. Immunoglobulin gene sequencing confirmed that several of these LLPCs were clones of memory B cells elicited 2 weeks after boost that had undergone further somatic hypermutation. Many of the antibodies secreted by these LLPCs also exhibited improved neutralization and cross-reactivity compared with earlier time points. These findings establish our method as a means to sensitively and reliably detect rare antigen-specific LLPCs and demonstrate that adjuvanted SARS-CoV-2 spike protein vaccination establishes spike protein-specific LLPC reservoirs.


Sujet(s)
COVID-19 , Glycoprotéine de spicule des coronavirus , Animaux , Humains , Plasmocytes/métabolisme , Anticorps antiviraux , SARS-CoV-2 , COVID-19/prévention et contrôle , Vaccination , Adjuvants immunologiques , Primates , Anticorps neutralisants
7.
Cell Rep ; 42(12): 113450, 2023 12 26.
Article de Anglais | MEDLINE | ID: mdl-38019653

RÉSUMÉ

HIV gp120 engineered outer domain germline-targeting version 8 (eOD-GT8) was designed specifically to engage naive B cell precursors of VRC01-class antibodies. However, the frequency and affinity of naive B cell precursors able to recognize eOD-GT8 have been evaluated only in U.S. populations. HIV infection is disproportionally concentrated in sub-Saharan Africa, so we seek to characterize naive B cells able to recognize eOD-GT8 in sub-Saharan cohorts. We demonstrate that people from sub-Saharan Africa have a higher or equivalent frequency of naive B cells able to engage eOD-GT8 compared with people from the U.S. Genetically, the higher frequency of eOD-GT8-positive cells is accompanied by a higher level of naive B cells with gene signatures characteristic of the VRC01 class, as well as other CD4bs-directed antibodies. Our study demonstrates that vaccination with eOD-GT8 in sub-Saharan Africa could be successful at expanding and establishing a pool of CD4bs-directed memory B cells from naive precursors.


Sujet(s)
Infections à VIH , VIH-1 (Virus de l'Immunodéficience Humaine de type 1) , Humains , Anticorps neutralisants , Anticorps anti-VIH , Précurseurs lymphoïdes B , Protéine d'enveloppe gp120 du VIH
8.
bioRxiv ; 2023 Aug 24.
Article de Anglais | MEDLINE | ID: mdl-37781590

RÉSUMÉ

Influenza A virus subtype H2N2, which caused the 1957 influenza pandemic, remains a global threat. A recent phase I clinical trial investigating a ferritin nanoparticle displaying H2 hemagglutinin in H2-naïve and H2-exposed adults. Therefore, we could perform comprehensive structural and biochemical characterization of immune memory on the breadth and diversity of the polyclonal serum antibody response elicited after H2 vaccination. We temporally map the epitopes targeted by serum antibodies after first and second vaccinations and show previous H2 exposure results in higher responses to the variable head domain of hemagglutinin while initial responses in H2-naïve participants are dominated by antibodies targeting conserved epitopes. We use cryo-EM and monoclonal B cell isolation to describe the molecular details of cross-reactive antibodies targeting conserved epitopes on the hemagglutinin head including the receptor binding site and a new site of vulnerability deemed the medial junction. Our findings accentuate the impact of pre-existing influenza exposure on serum antibody responses.

9.
Sci Transl Med ; 15(692): eade4976, 2023 04 19.
Article de Anglais | MEDLINE | ID: mdl-37075126

RÉSUMÉ

Current yearly seasonal influenza vaccines primarily induce an antibody response directed against the immunodominant but continually diversifying hemagglutinin (HA) head region. These antibody responses provide protection against the vaccinating strain but little cross-protection against other influenza strains or subtypes. To focus the immune response on subdominant but more conserved epitopes on the HA stem that might protect against a broad range of influenza strains, we developed a stabilized H1 stem immunogen lacking the immunodominant head displayed on a ferritin nanoparticle (H1ssF). Here, we evaluated the B cell response to H1ssF in healthy adults ages 18 to 70 in a phase 1 clinical trial (NCT03814720). We observed both a strong plasmablast response and sustained elicitation of cross-reactive HA stem-specific memory B cells after vaccination with H1ssF in individuals of all ages. The B cell response was focused on two conserved epitopes on the H1 stem, with a highly restricted immunoglobulin repertoire unique to each epitope. On average, two-thirds of the B cell and serological antibody response recognized a central epitope on the H1 stem and exhibited broad neutralization across group 1 influenza virus subtypes. The remaining third recognized an epitope near the viral membrane anchor and was largely limited to H1 strains. Together, we demonstrate that an H1 HA immunogen lacking the immunodominant HA head produces a robust and broadly neutralizing HA stem-directed B cell response.


Sujet(s)
Vaccins antigrippaux , Grippe humaine , Adolescent , Adulte , Sujet âgé , Humains , Adulte d'âge moyen , Jeune adulte , Anticorps neutralisants , Anticorps antiviraux , Épitopes , Glycoprotéine hémagglutinine du virus influenza , Hémagglutinines
10.
Sci Transl Med ; 15(692): eade4790, 2023 04 19.
Article de Anglais | MEDLINE | ID: mdl-37075129

RÉSUMÉ

Influenza vaccines could be improved by platforms inducing cross-reactive immunity. Immunodominance of the influenza hemagglutinin (HA) head in currently licensed vaccines impedes induction of cross-reactive neutralizing stem-directed antibodies. A vaccine without the variable HA head domain has the potential to focus the immune response on the conserved HA stem. This first-in-human dose-escalation open-label phase 1 clinical trial (NCT03814720) tested an HA stabilized stem ferritin nanoparticle vaccine (H1ssF) based on the H1 HA stem of A/New Caledonia/20/1999. Fifty-two healthy adults aged 18 to 70 years old enrolled to receive either 20 µg of H1ssF once (n = 5) or 60 µg of H1ssF twice (n = 47) with a prime-boost interval of 16 weeks. Thirty-five (74%) 60-µg dose participants received the boost, whereas 11 (23%) boost vaccinations were missed because of public health restrictions in the early stages of the COVID-19 pandemic. The primary objective of this trial was to evaluate the safety and tolerability of H1ssF, and the secondary objective was to evaluate antibody responses after vaccination. H1ssF was safe and well tolerated, with mild solicited local and systemic reactogenicity. The most common symptoms included pain or tenderness at the injection site (n = 10, 19%), headache (n = 10, 19%), and malaise (n = 6, 12%). We found that H1ssF elicited cross-reactive neutralizing antibodies against the conserved HA stem of group 1 influenza viruses, despite previous H1 subtype head-specific immunity. These responses were durable, with neutralizing antibodies observed more than 1 year after vaccination. Our results support this platform as a step forward in the development of a universal influenza vaccine.


Sujet(s)
COVID-19 , Vaccins antigrippaux , Grippe humaine , Adolescent , Adulte , Sujet âgé , Humains , Adulte d'âge moyen , Jeune adulte , Anticorps neutralisants , Anticorps antiviraux , Anticorps neutralisants à large spectre , Glycoprotéine hémagglutinine du virus influenza , Hémagglutinines , Pandémies
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE