373 results on '"Korber, B."'
Search Results
102. Candidate mosaic proteins for a pan-filoviral cytotoxic T-Cell lymphocyte vaccine
- Author
-
Korber, B [Los Alamos National Laboratory]
- Published
- 2008
103. The mode of delivery and the risk of vertical transmission of human immunodeficiency virus type 1--a meta-analysis of 15 prospective cohort studies.
- Author
-
International Perinatal HIV Group, Andiman, W, Bryson, Y, de Martino, M, Fowler, M, Harris, D, Hutto, C, Korber, B, Kovacs, A, Landesman, S, Lindsay, M, Lapointe, N, Mandelbrot, L, Newell, M-L, Peavy, H, Read, J, Rudin, C, Semprini, A, Simonds, R, and Tuomala, R
- Abstract
Background: To evaluate the relation between elective cesarean section and vertical transmission of human immunodeficiency virus type 1 (HIV-1), we performed a meta-analysis using data on individual patients from 15 prospective cohort studies.Methods: North American and European studies of at least 100 mother-child pairs were included in the meta-analysis. Uniform definitions of modes of delivery were used. Elective cesarean sections were defined as those performed before onset of labor and rupture of membranes. Multivariate logistic-regression analysis was used to adjust for other factors known to be associated with vertical transmission.Results: The primary analysis included data on 8533 mother-child pairs. After adjustment for receipt of antiretroviral therapy, maternal stage of disease, and infant birth weight, the likelihood of vertical transmission of HIV-1 was decreased by approximately 50 percent with elective cesarean section, as compared with other modes of delivery (adjusted odds ratio, 0.43; 95 percent confidence interval, 0.33 to 0.56). The results were similar when the study population was limited to those with rupture of membranes shortly before delivery. The likelihood of transmission was reduced by approximately 87 percent with both elective cesarean section and receipt of antiretroviral therapy during the prenatal, intrapartum, and neonatal periods, as compared with other modes of delivery and the absence of therapy (adjusted odds ratio, 0.13; 95 percent confidence interval, 0.09 to 0.19). Among mother-child pairs receiving antiretroviral therapy during the prenatal, intrapartum, and neonatal periods, rates of vertical transmission were 2.0 percent among the 196 mothers who underwent elective cesarean section and 7.3 percent among the 1255 mothers with other modes of delivery.Conclusions: The results of this meta-analysis suggest that elective cesarean section reduces the risk of transmission of HIV-1 from mother to child independently of the effects of treatment with zidovudine. [ABSTRACT FROM AUTHOR]- Published
- 1999
104. Experimental medicine study with stabilised native-like HIV-1 Env immunogens drives long-term antibody responses, but lacks neutralising breadth.
- Author
-
Pollock KM, Cheeseman HM, McFarlane LR, Day S, Tolazzi M, Turner HL, Joypooranachandran J, Shramko K, Dispinseri S, Mundsperger P, Bontjer I, Lemm NM, Coelho S, Tanaka M, Cole T, Korber B, Katinger D, Sattentau QJ, Ward AB, Scarlatti G, Sanders RW, and Shattock RJ
- Abstract
Background: We report findings from an experimental medicine study of rationally designed prefusion stabilised native-like HIV envelope glycoprotein (Env) immunogens, representative of global circulating strains, delivered by sequential intramuscular injection., Methods: Healthy adult volunteers were enrolled into one of five groups (A to E) each receiving a different schedule of one of two consensus Env immunogens (ConM SOSIP, ConS UFO, either unmodified or stabilised by chemical cross-linking, followed by a boost with two mosaic Env immunogens (Mos3.1 and Mos3.2). All immunogens were co-formulated with liposomal Monophosphoryl-Lipid A (MPLA) adjuvant, and volunteers were followed up for 28 days post final Mosaic booster injection. Participants gave written informed consent to join the study. The study is registered on ClinicalTrials.gov ID NCT03816137., Findings: Fifty-one participants (men n = 23 and women n = 28) aged 18-55 were enrolled. The seroconversion rate against Env was 100% with all participants having measurable anti-Env IgG antibodies after their second injection and throughout the study. Neutralisation was detected against the ConM pseudovirus in sera of those who had received both ConM and ConS immunogens. However, this activity was limited in breadth and was neither boosted nor broadened in those receiving the Mos3.1 and Mos3.2 immunogens. Neutralising antibody function correlated with binding to V1/V3 and V5 epitopes and peaked after the third injection., Interpretation: Rationally designed prefusion-stabilised native-like Env trimers are robustly immunogenic in a prime-boost schedule. When given alone they are insufficient to induce neutralising antibody titres of significant breadth, but they represent potentially valuable polishing immunogens after germline-targeting., Funding: European Aids Vaccine initiative (EAVI2020) received funding from EU Horizon 2020, grant number 681137. Structural studies were supported by the Bill and Melinda Gates Foundation (INV-002916)., Competing Interests: Declaration of interests K.M.P was supported in part by the NIHR Imperial Biomedical Research Centre and by the St Mary’s Development Trust, and has received research funding support from Horizon 2020 and from Trevena Inc, Imperial COVID-19 fund, National Institute for Health Research, and The Sir Joseph Hotung Charitable Settlement outside the submitted work. K.M.P is in receipt of an MRC Clinician Scientist Fellowship award (MR/W024977/1) and a grant from the Chan Zuckerberg Initiative outside the submitted work. K.M.P has received payment or honoraria from CSL Seqirus and Sanofi Pasteur for speaking and as a panellist, and travel support from the Chan Zuckerberg Initiative. K.M.P has participated in data safety monitoring boards for Moderna. K.M.P has a role on the British HIV Association immunisation guidelines writing committee and was on the UK Chief Investigators Group NIHR vaccine research programme. R.W.S and I.B declare grants or contracts from any entity - EU (EAVI2020; grant number 681137). B.K. is a co-inventor on a patent covering modified human immunodeficiency virus type 1 (HIV-1) group M consensus envelope glycoproteins (Mosaic) [US-9844589-B2]. A.B.W declares that this work was supported, in whole or in part, by the Bill & Melinda Gates Foundation INV-002916. All other authors declare no conflict of interests regarding any financial and personal relationships with other people or organisations that could inappropriately influence our work., (Copyright © 2024. Published by Elsevier B.V.)
- Published
- 2025
- Full Text
- View/download PDF
105. Transient glycan-shield reduction induces CD4-binding site broadly neutralizing antibodies in SHIV-infected macaques.
- Author
-
Morris DJ, Gorman J, Zhou T, Lora J, Connell AJ, Li H, Liu W, Roark RS, Campion MS, Carey JW, Habib R, Li Y, Martella CL, Park Y, Singh A, Sowers KJ, Teng IT, Wang S, Chohan N, Ding W, Lauer C, Lewis E, Mason RD, Rando JM, Peyton L, Schramm CA, Wagh K, Korber B, Seaman MS, Douek DC, Haynes BF, Kulp DW, Roederer M, Hahn BH, Kwong PD, and Shaw GM
- Abstract
Broadly neutralizing antibodies (bNAbs) targeting the HIV-1 CD4-binding site (CD4bs) occur infrequently in macaques and humans and have not been reproducibly elicited in any outbred animal model. To address this challenge, we first isolated RHA10, an infection-induced rhesus bNAb with 51% breadth. The cryo-EM structure of RHA10 with HIV-1 envelope (Env) resembled prototypic human CD4bs bNAbs with CDR-H3-dominated binding. Env-antibody co-evolution revealed transient elimination of two Env CD4bs-proximal glycans near the time of RHA10-lineage initiation, and these glycan-deficient Envs bound preferentially to early RHA10 intermediates, suggesting glycan deletions in infecting SHIVs could consistently induce CD4bs bNAbs. To test this, we constructed SHIV.CH505.D3 with CD4bs-proximal glycan deletions. Infection of 10 macaques resulted in accelerated CD4bs bNAb responses in 8, compared with 1 of 115 control macaques. Glycan hole-based immunofocusing coupled to Env-Ab co-evolution can consistently induce broad CD4bs responses in macaques and thus serve as a model for HIV vaccine design., Highlights: Out of 115 wildtype HIV-1 Env bearing SHIV infected macaques, only one macaque (T681) developed CD4bs bNAbsCD4bs bNAbs in macaque T681 recognized Env similarly to previously described CDR-H3 dominated human CD4bs bNAbs and exhibited comparable breadth and potencyTransient CD4bs-proximal glycan deletions in macaque T681 preceded bNAb inductionA novel SHIV with CD4bs-proximal glycan holes and enhanced CD4bs antigenicity immunofocused B cell responses to the CD4bs and elicited cross-clade neutralizing responses in 80% of macaques.
- Published
- 2024
- Full Text
- View/download PDF
106. Safety and antiviral effect of a triple combination of HIV-1 broadly neutralizing antibodies: a phase 1/2a trial.
- Author
-
Julg B, Walker-Sperling VEK, Wagh K, Aid M, Stephenson KE, Zash R, Liu J, Nkolola JP, Hoyt A, Castro M, Serebryannyy L, Yanosick K, Speidel T, Borducchi EN, Murzda T, Maxfield L, Arduino R, McDermott AB, Gama L, Giorgi EE, Koup RA, Seaman MS, Rolle CP, DeJesus E, Li W, Korber B, and Barouch DH
- Subjects
- Humans, Male, Adult, Female, Middle Aged, Broadly Neutralizing Antibodies immunology, Broadly Neutralizing Antibodies therapeutic use, CD4 Lymphocyte Count, Viral Load drug effects, Antibodies, Monoclonal administration & dosage, Antibodies, Monoclonal adverse effects, Antibodies, Monoclonal therapeutic use, Antibodies, Monoclonal immunology, HIV-1 immunology, HIV-1 drug effects, HIV Infections drug therapy, HIV Infections immunology, HIV Infections virology, HIV Antibodies immunology, Antibodies, Neutralizing immunology
- Abstract
Human immunodeficiency virus type 1 (HIV-1)-specific broadly neutralizing monoclonal antibodies (bNAbs) have to date shown transient viral suppression when administered as monotherapy or as a cocktail of two antibodies
1-4 . A combination of three bNAbs provides improved neutralization coverage of global viruses, which may more potently suppress viral escape and rebound5-7 . Here we performed an open-label, two-part study evaluating a single intravenous dose of HIV-1 bNAbs, PGT121, PGDM1400 and VRC07-523LS, in six adults without HIV in part 1 and a multicenter trial of up to six monthly infusions of these three bNAbs in 12 people living with HIV with an antiretroviral therapy (ART) interruption in part 2. The primary endpoints were safety, tolerability and pharmacokinetics, and the secondary endpoints in part 2 were antiviral activity following ART discontinuation, changes in CD4+ T cell counts and development of HIV-1 sequence mutations associated with bNAb resistance. The trial met its prespecified endpoints. The bNAb treatment was generally safe and well tolerated. In part 2, 83% of participants (10 of 12) maintained virologic suppression for the duration of antibody therapy for at least 28 weeks, and 42% of participants (5 of 12) showed virologic suppression for at least 38-44 weeks, despite the decline of serum bNAb concentrations to low or undetectable levels. In exploratory analyses, early viral rebound in two individuals correlated with baseline resistance to PGT121 and PGDM1400, whereas long-term virologic control in five individuals correlated with reduced immune activation, T cell exhaustion and proinflammatory signaling following bNAb therapy. Our data show the potential of a triple bNAb cocktail to suppress HIV-1 in the absence of ART. ClinicalTrials.gov registration: NCT03721510 ., Competing Interests: Competing interests: B.J. is a part-time employee of Leyden Labs, B.V. The other authors declare no competing interests., (© 2024. The Author(s).)- Published
- 2024
- Full Text
- View/download PDF
107. Engineering immunogens that select for specific mutations in HIV broadly neutralizing antibodies.
- Author
-
Henderson R, Anasti K, Manne K, Stalls V, Saunders C, Bililign Y, Williams A, Bubphamala P, Montani M, Kachhap S, Li J, Jaing C, Newman A, Cain DW, Lu X, Venkatayogi S, Berry M, Wagh K, Korber B, Saunders KO, Tian M, Alt F, Wiehe K, Acharya P, Alam SM, and Haynes BF
- Subjects
- Humans, HIV Infections immunology, HIV Infections virology, HIV Infections prevention & control, Broadly Neutralizing Antibodies immunology, Broadly Neutralizing Antibodies genetics, B-Lymphocytes immunology, Antibody Affinity immunology, Protein Engineering methods, HIV-1 immunology, HIV-1 genetics, Mutation, HIV Antibodies immunology, AIDS Vaccines immunology, AIDS Vaccines genetics, env Gene Products, Human Immunodeficiency Virus immunology, env Gene Products, Human Immunodeficiency Virus genetics, Molecular Dynamics Simulation, Antibodies, Neutralizing immunology
- Abstract
Vaccine development targeting rapidly evolving pathogens such as HIV-1 requires induction of broadly neutralizing antibodies (bnAbs) with conserved paratopes and mutations, and in some cases, the same Ig-heavy chains. The current trial-and-error search for immunogen modifications that improve selection for specific bnAb mutations is imprecise. Here, to precisely engineer bnAb boosting immunogens, we use molecular dynamics simulations to examine encounter states that form when antibodies collide with the HIV-1 Envelope (Env). By mapping how bnAbs use encounter states to find their bound states, we identify Env mutations predicted to select for specific antibody mutations in two HIV-1 bnAb B cell lineages. The Env mutations encode antibody affinity gains and select for desired antibody mutations in vivo. These results demonstrate proof-of-concept that Env immunogens can be designed to directly select for specific antibody mutations at residue-level precision by vaccination, thus demonstrating the feasibility of sequential bnAb-inducing HIV-1 vaccine design., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
108. A comprehensive engineering strategy improves potency and manufacturability of a near pan-neutralizing antibody against HIV.
- Author
-
Sajadi MM, Abbasi A, Tehrani ZR, Siska C, Clark R, Chi W, Seaman MS, Mielke D, Wagh K, Liu Q, Jumpa T, Ketchem RR, Nguyen DN, Tolbert WD, Pierce BG, Atkinson B, Deming D, Sprague M, Asakawa A, Ferrer D, Dunn Y, Calvillo S, Yin R, Guest JD, Korber B, Mayer BT, Sato AH, Ouyang X, Foulke S, Habibzadeh P, Karimi M, Aslanabadi A, Hojabri M, Saadat S, Zareidoodeji R, Kędzior M, Pozharski E, Heredia A, Montefiori D, Ferrari G, Pazgier M, Lewis GK, Jardine JG, Lusso P, and DeVico A
- Abstract
Anti-HIV envelope broadly neutralizing antibodies (bnAbs) are alternatives to conventional antiretrovirals with the potential to prevent and treat infection, reduce latent reservoirs, and/or mediate a functional cure. Clinical trials with "first generation" bnAbs used alone or in combination show promising antiviral effects but also highlight that additional engineering of "enhanced" antibodies will be required for optimal clinical utility, while preserving or enhancing cGMP manufacturing capability. Here we report the engineering of an anti-CD4 binding-site (CD4bs) bnAb, N49P9.3, purified from the plasma of an HIV elite-neutralizer. Through a series of rational modifications we produced a variant that demonstrates: enhanced potency; superior antiviral activity in combination with other bnAbs; low polyreactivity; and longer circulating half-life. Additional engineering for manufacturing produced a final variant, eN49P9, with properties conducive to cGMP production. Overall, these efforts demonstrate the feasibility of developing enhanced anti-CD4bs bnAbs with greatly improved antiviral properties as well as potential translational value.
- Published
- 2024
- Full Text
- View/download PDF
109. Immune correlates analysis of the Imbokodo (HVTN 705/HPX2008) efficacy trial of a mosaic HIV-1 vaccine regimen evaluated in Southern African people assigned female sex at birth: a two-phase case-control study.
- Author
-
Kenny A, van Duijn J, Dintwe O, Heptinstall J, Burnham R, Sawant S, Zhang L, Mielke D, Khuzwayo S, Omar FL, Stanfield-Oakley S, Keyes T, Dunn B, Goodman D, Fong Y, Benkeser D, Zou R, Hural J, Hyrien O, Juraska M, Luedtke A, van der Laan L, Giorgi EE, Magaret C, Carpp LN, Pattacini L, van de Kerkhof T, Korber B, Willems W, Fisher LH, Schuitemaker H, Swann E, Kublin JG, Pau MG, Buchbinder S, Tomaka F, Nijs S, Lavreys L, Gelderblom HC, Corey L, Mngadi K, Gray GE, Borducchi E, Hendriks J, Seaton KE, Zolla-Pazner S, Barouch DH, Ferrari G, De Rosa SC, McElrath MJ, Andersen-Nissen E, Stieh DJ, Tomaras GD, and Gilbert PB
- Subjects
- Humans, Female, Case-Control Studies, Male, Adult, Vaccine Efficacy, HIV Antibodies blood, HIV Antibodies immunology, Immunoglobulin G blood, Immunoglobulin G immunology, env Gene Products, Human Immunodeficiency Virus immunology, Africa, Southern, Young Adult, Southern African People, AIDS Vaccines immunology, AIDS Vaccines administration & dosage, HIV Infections immunology, HIV Infections prevention & control, HIV Infections virology, HIV-1 immunology
- Abstract
Background: The HVTN 705 Imbokodo trial of 2636 people without HIV and assigned female sex at birth, conducted in southern Africa, evaluated a heterologous HIV-1 vaccine regimen: mosaic adenovirus 26-based vaccine (Ad26.Mos4.HIV) at Months 0, 3, 6, 12 and alum-adjuvanted clade C gp140 at Months 6, 12. Per-protocol vaccine efficacy (VE) against HIV-1 diagnosis from seven to 24 months was 14.1% (95% CI: -22.0% to 39.5%). Immune correlates analysis was performed for markers selected based on prior evidence in efficacy trials and/or nonhuman primate models., Methods: Humoral and cellular immune response markers at Month 7 were evaluated as immune correlates of risk and of protection in a breakthrough case-control cohort (n = 52 cases, 246 non-cases). Primary markers were IgG binding to vaccine-strain gp140, IgG3 binding to diverse Env antigens (IgG3 Env breadth), IgG3 binding to diverse V1V2 antigens (IgG3 V1V2 breadth), antibody-dependent phagocytosis against the vaccine-strain gp140, Env-specific CD4+ and CD8+ T-cell responses, and multi-epitope functions., Findings: No immune markers were statistically significant correlates of risk. IgG3 V1V2 breadth trended toward an inverse association: hazard ratio 0.70 (95% CI: 0.36 to 1.35; p = 0.29) per 10-fold increase and 0.51 (95% CI: 0.21 to 1.24; p = 0.14) in a Cox model with all primary markers. The VE estimate was 11.8% (95% CI: -17.9% to 34.0%) at all IgG3 V1V2 breadth values below 667 weighted geometric mean net MFI; just above this value, the VE estimate sharply increased to 62.6% (95% CI: -17.9% to 89.6%), and further increased to 80.9% (95% CI: -17.9% to 99.5%) at 1471 MFI, the 95th percentile of the marker distribution. Mediation analysis yielded a VE of 35.7% (95% CI: 15.0% to 51.3%) attributable to the vaccine's impact on this marker., Interpretation: The trend in association of greater IgG3 V1V2 antibody breadth with lower likelihood of HIV acquisition is consistent with the identification of antibodies against V1V2 as immune correlates in three other HIV vaccine efficacy trials and suggests that a greater emphasis should be placed on studying this region in the HIV-1 envelope as a vaccine immunogen., Funding: National Institute of Allergy and Infectious Diseases and Janssen Vaccines & Prevention BV., Competing Interests: Declaration of interests TvdK has a patent application with Johnson & Johnson and has stocks in Johnson & Johnson. BK received internal support for the present manuscript from her employer (Los Alamos National Laboratory). In the past 36 months, she received support for attending meetings and/or travel from NIH NIAID and from the Gates foundation. Her institution (LANL) had a patent on this work, although she did not receive any personal funds through this patent and was not involved with the licensing of the design to Johnson & Johnson. DHB has a patent on the mosaic HIV vaccine, but no royalties. FT was an employee of Janssen/Johnson & Johnson at the time the work was conducted and owns stock in Johnson & Johnson. LL received support from Janssen Infectious Diseases BV, Beerse, Belgium for travel expenses to attend HIV conferences and has stock or stock options in Johnson & Johnson. JvD, MGP, WW, TvdK and JHen are employees of Janssen/Johnson & Johnson and hold stock or stock options in Johnson & Johnson. WW has a patent planned, issued, or pending with Johnson & Johnson. SCDR had contracts in the past 36 months to perform immunogenicity testing for Janssen, Sanofi, and Moderna. HS and DJS were employees of Janssen Vaccines & Prevention BV and had stock and/or stock options in Johnson & Johnson at the time the work was conducted. SN was an employee of Janssen Infectious Diseases BV and had stock and/or stock options in Johnson & Johnson at the time the work was conducted. LP was an employee of Janssen Vaccines & Prevention BV at the time the work was conducted. GDT has received consulting fees for a scientific consulting session. All other authors have no potential competing interests to disclose. Funding for the Imbokodo Study and Correlates Group is the same as listed in “Acknowledgments” for the current work., (Copyright © 2024 The Author(s). Published by Elsevier B.V. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
110. Design, Immunogenicity and Preclinical Efficacy of the ChAdOx1.COVconsv12 Pan-Sarbecovirus T-Cell Vaccine.
- Author
-
Wee EG, Kempster S, Ferguson D, Hall J, Ham C, Morris S, Crook A, Gilbert SC, Korber B, Almond N, and Hanke T
- Abstract
During the COVID-19 pandemic, antibody-based vaccines targeting the SARS-CoV-2 spike glycoprotein were the focus for development because neutralizing antibodies were associated with protection against the SARS-CoV-2 infection pre-clinically and in humans. While deploying these spike-based vaccines saved millions of lives worldwide, it has become clear that the immunological mechanisms of protection against severe disease are multifaceted and involve non-neutralizing antibody components. Here, we describe a novel pan-sarbecovirus T-cell vaccine, ChAdOx1.COVconsv12, designed to complement and broaden the protection of spike vaccines. The vaccine immunogen COVconsv12 employs the two regions in the viral proteome most conserved among sarbecoviruses, which are delivered by replication-deficient vector ChAdOx1. It directs T cells towards epitopes shared among sarbecoviruses including evolving SARS-CoV-2 variants. Here, we show that ChAdOx1.COVconsv12 induced broad T-cell responses in the BALB/c and C57BL/6 mice. In the Syrian hamster challenge model, ChAdOx1.COVconsv12 alone did not protect against the SARS-CoV-2 infection, but when co-administered with 1/50th of the ChAdOx1 nCoV-19 spike vaccine protective dose, faster recovery and lower oral swab viral load were observed. Induction of CD8
+ T cells may decrease COVID-19 severity and extend the T-cell response coverage of variants to match the known (and as yet unknown) members of the β-coronavirus family.- Published
- 2024
- Full Text
- View/download PDF
111. SARS-CoV-2 Omicron XBB lineage spike structures, conformations, antigenicity, and receptor recognition.
- Author
-
Zhang QE, Lindenberger J, Parsons RJ, Thakur B, Parks R, Park CS, Huang X, Sammour S, Janowska K, Spence TN, Edwards RJ, Martin M, Williams WB, Gobeil S, Montefiori DC, Korber B, Saunders KO, Haynes BF, Henderson R, and Acharya P
- Subjects
- Humans, Protein Binding, Immune Evasion, Models, Molecular, Protein Domains, Binding Sites, Spike Glycoprotein, Coronavirus genetics, Spike Glycoprotein, Coronavirus chemistry, Spike Glycoprotein, Coronavirus immunology, Spike Glycoprotein, Coronavirus metabolism, SARS-CoV-2 genetics, SARS-CoV-2 immunology, SARS-CoV-2 metabolism, SARS-CoV-2 chemistry, Cryoelectron Microscopy, COVID-19 virology, COVID-19 immunology, Mutation, Protein Conformation
- Abstract
A recombinant lineage of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) Omicron variant, named XBB, appeared in late 2022 and evolved descendants that successively swept local and global populations. XBB lineage members were noted for their improved immune evasion and transmissibility. Here, we determine cryoelectron microscopy (cryo-EM) structures of XBB.1.5, XBB.1.16, EG.5, and EG.5.1 spike (S) ectodomains to reveal reinforced 3-receptor binding domain (RBD)-down receptor-inaccessible closed states mediated by interprotomer RBD interactions previously observed in BA.1 and BA.2. Improved XBB.1.5 and XBB.1.16 RBD stability compensated for stability loss caused by early Omicron mutations, while the F456L substitution reduced EG.5 RBD stability. S1 subunit mutations had long-range impacts on conformation and epitope presentation in the S2 subunit. Our results reveal continued S protein evolution via simultaneous optimization of multiple parameters, including stability, receptor binding, and immune evasion, and the dramatic effects of relatively few residue substitutions in altering the S protein conformational landscape., Competing Interests: Declaration of interests B.F.H., K.O.S., R.J.E., S.G., and P.A. are named in patents submitted on the SARS-CoV-2 monoclonal antibodies studied in this paper., (Copyright © 2024 Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
112. Vaccine induction of heterologous HIV-1-neutralizing antibody B cell lineages in humans.
- Author
-
Williams WB, Alam SM, Ofek G, Erdmann N, Montefiori DC, Seaman MS, Wagh K, Korber B, Edwards RJ, Mansouri K, Eaton A, Cain DW, Martin M, Hwang J, Arus-Altuz A, Lu X, Cai F, Jamieson N, Parks R, Barr M, Foulger A, Anasti K, Patel P, Sammour S, Parsons RJ, Huang X, Lindenberger J, Fetics S, Janowska K, Niyongabo A, Janus BM, Astavans A, Fox CB, Mohanty I, Evangelous T, Chen Y, Berry M, Kirshner H, Van Itallie E, Saunders KO, Wiehe K, Cohen KW, McElrath MJ, Corey L, Acharya P, Walsh SR, Baden LR, and Haynes BF
- Subjects
- Humans, HIV Infections immunology, HIV Infections virology, Cell Lineage, Liposomes, env Gene Products, Human Immunodeficiency Virus immunology, Mutation, HIV Envelope Protein gp41 immunology, AIDS Vaccines immunology, HIV-1 immunology, Antibodies, Neutralizing immunology, B-Lymphocytes immunology, HIV Antibodies immunology
- Abstract
A critical roadblock to HIV vaccine development is the inability to induce B cell lineages of broadly neutralizing antibodies (bnAbs) in humans. In people living with HIV-1, bnAbs take years to develop. The HVTN 133 clinical trial studied a peptide/liposome immunogen targeting B cell lineages of HIV-1 envelope (Env) membrane-proximal external region (MPER) bnAbs (NCT03934541). Here, we report MPER peptide-liposome induction of polyclonal HIV-1 B cell lineages of mature bnAbs and their precursors, the most potent of which neutralized 15% of global tier 2 HIV-1 strains and 35% of clade B strains with lineage initiation after the second immunization. Neutralization was enhanced by vaccine selection of improbable mutations that increased antibody binding to gp41 and lipids. This study demonstrates proof of concept for rapid vaccine induction of human B cell lineages with heterologous neutralizing activity and selection of antibody improbable mutations and outlines a path for successful HIV-1 vaccine development., Competing Interests: Declaration of interests B.F.H. and S.M.A. have US patents 9402917, 9402893, 9717789, and 10588960 and US patent application 63/540482. B.F.H., S.M.A., and B.K. have US patent 10076567. B.F.H. and K.O.S. have patent applications PCT/US2023/077677 and PCT/US2023/077686. C.B.F. has patents on PEGylated liposomes., (Copyright © 2024 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
113. Waning immunity and IgG4 responses following bivalent mRNA boosting.
- Author
-
Lasrado N, Collier AY, Miller J, Hachmann NP, Liu J, Anand T, A Bondzie E, Fisher JL, Mazurek CR, Patio RC, Rodrigues SL, Rowe M, Surve N, Ty DM, Wu C, Chicz TM, Tong X, Korber B, McNamara RP, and Barouch DH
- Subjects
- Antibodies, Neutralizing, Immunization, RNA, Messenger genetics, mRNA Vaccines, Antibody Formation, Immunoglobulin G
- Abstract
Messenger RNA (mRNA) vaccines were highly effective against the ancestral SARS-CoV-2 strain, but the efficacy of bivalent mRNA boosters against XBB variants was substantially lower. Here, we show limited durability of neutralizing antibody (NAb) responses against XBB variants and isotype switching to immunoglobulin G4 (IgG4) responses following bivalent mRNA boosting. Bivalent mRNA boosting elicited modest XBB.1-, XBB.1.5-, and XBB.1.16-specific NAbs that waned rapidly within 3 months. In contrast, bivalent mRNA boosting induced more robust and sustained NAbs against the ancestral WA1/2020 strain, suggesting immune imprinting. Following bivalent mRNA boosting, serum antibody responses were primarily IgG2 and IgG4 responses with poor Fc functional activity. In contrast, a third monovalent mRNA immunization boosted all isotypes including IgG1 and IgG3 with robust Fc functional activity. These data show substantial immune imprinting for the ancestral spike and isotype switching to IgG4 responses following bivalent mRNA boosting, with important implications for future booster designs and boosting strategies.
- Published
- 2024
- Full Text
- View/download PDF
114. Engineering immunogens that select for specific mutations in HIV broadly neutralizing antibodies.
- Author
-
Henderson R, Anasti K, Manne K, Stalls V, Saunders C, Bililign Y, Williams A, Bubphamala P, Montani M, Kachhap S, Li J, Jaing C, Newman A, Cain D, Lu X, Venkatayogi S, Berry M, Wagh K, Korber B, Saunders KO, Tian M, Alt F, Wiehe K, Acharya P, Alam SM, and Haynes BF
- Abstract
Vaccine development targeting rapidly evolving pathogens such as HIV-1 requires induction of broadly neutralizing antibodies (bnAbs) with conserved paratopes and mutations, and, in some cases, the same Ig-heavy chains. The current trial-and-error search for immunogen modifications that improve selection for specific bnAb mutations is imprecise. To precisely engineer bnAb boosting immunogens, we used molecular dynamics simulations to examine encounter states that form when antibodies collide with the HIV-1 Envelope (Env). By mapping how bnAbs use encounter states to find their bound states, we identified Env mutations that were predicted to select for specific antibody mutations in two HIV-1 bnAb B cell lineages. The Env mutations encoded antibody affinity gains and selected for desired antibody mutations in vivo . These results demonstrate proof-of-concept that Env immunogens can be designed to directly select for specific antibody mutations at residue-level precision by vaccination, thus demonstrating the feasibility of sequential bnAb-inducing HIV-1 vaccine design., Competing Interests: Declaration of Interests The authors declare the following competing interests: A patent application covering HIV-1 Envelope modifications based on this study has been submitted by Duke University. The authors declare no other competing interests.
- Published
- 2023
- Full Text
- View/download PDF
115. Mutational basis of serum cross-neutralization profiles elicited by infection or vaccination with SARS-CoV-2 variants.
- Author
-
Wagh K, Shen X, Theiler J, Girard B, Marshall JC, Montefiori DC, and Korber B
- Abstract
A series of SARS-CoV-2 variants emerged during the pandemic under selection for neutralization resistance. Convalescent and vaccinated sera show consistently different cross-neutralization profiles depending on infecting or vaccine variants. To understand the basis of this heterogeneity, we modeled serum cross-neutralization titers for 165 sera after infection or vaccination with historically prominent lineages tested against 18 variant pseudoviruses. Cross-neutralization profiles were well captured by models incorporating autologous neutralizing titers and combinations of specific shared and differing mutations between the infecting/vaccine variants and pseudoviruses. Infecting/vaccine variant-specific models identified mutations that significantly impacted cross-neutralization and quantified their relative contributions. Unified models that explained cross-neutralization profiles across all infecting and vaccine variants provided accurate predictions of holdout neutralization data comprising untested variants as infecting or vaccine variants, and as test pseudoviruses. Finally, comparative modeling of 2-dose versus 3-dose mRNA-1273 vaccine data revealed that the third dose overcame key resistance mutations to improve neutralization breadth., Highlights: Modeled SARS-CoV-2 cross-neutralization using mutations at key sitesIdentified resistance mutations and quantified relative impactAccurately predicted holdout variant and convalescent/vaccine sera neutralizationShowed that the third dose of mRNA-1273 vaccination overcomes resistance mutations.
- Published
- 2023
- Full Text
- View/download PDF
116. Neutralization profiles of HIV-1 viruses from the VRC01 Antibody Mediated Prevention (AMP) trials.
- Author
-
Mkhize NN, Yssel AEJ, Kaldine H, van Dorsten RT, Woodward Davis AS, Beaume N, Matten D, Lambson B, Modise T, Kgagudi P, York T, Westfall DH, Giorgi EE, Korber B, Anthony C, Mapengo RE, Bekker V, Domin E, Eaton A, Deng W, DeCamp A, Huang Y, Gilbert PB, Gwashu-Nyangiwe A, Thebus R, Ndabambi N, Mielke D, Mgodi N, Karuna S, Edupuganti S, Seaman MS, Corey L, Cohen MS, Hural J, McElrath MJ, Mullins JI, Montefiori D, Moore PL, Williamson C, and Morris L
- Subjects
- Humans, HIV Antibodies, Broadly Neutralizing Antibodies, Antibodies, Neutralizing, Polysaccharides, HIV Infections, HIV-1, HIV Seropositivity
- Abstract
The VRC01 Antibody Mediated Prevention (AMP) efficacy trials conducted between 2016 and 2020 showed for the first time that passively administered broadly neutralizing antibodies (bnAbs) could prevent HIV-1 acquisition against bnAb-sensitive viruses. HIV-1 viruses isolated from AMP participants who acquired infection during the study in the sub-Saharan African (HVTN 703/HPTN 081) and the Americas/European (HVTN 704/HPTN 085) trials represent a panel of currently circulating strains of HIV-1 and offer a unique opportunity to investigate the sensitivity of the virus to broadly neutralizing antibodies (bnAbs) being considered for clinical development. Pseudoviruses were constructed using envelope sequences from 218 individuals. The majority of viruses identified were clade B and C; with clades A, D, F and G and recombinants AC and BF detected at lower frequencies. We tested eight bnAbs in clinical development (VRC01, VRC07-523LS, 3BNC117, CAP256.25, PGDM1400, PGT121, 10-1074 and 10E8v4) for neutralization against all AMP placebo viruses (n = 76). Compared to older clade C viruses (1998-2010), the HVTN703/HPTN081 clade C viruses showed increased resistance to VRC07-523LS and CAP256.25. At a concentration of 1μg/ml (IC80), predictive modeling identified the triple combination of V3/V2-glycan/CD4bs-targeting bnAbs (10-1074/PGDM1400/VRC07-523LS) as the best against clade C viruses and a combination of MPER/V3/CD4bs-targeting bnAbs (10E8v4/10-1074/VRC07-523LS) as the best against clade B viruses, due to low coverage of V2-glycan directed bnAbs against clade B viruses. Overall, the AMP placebo viruses represent a valuable resource for defining the sensitivity of contemporaneous circulating viral strains to bnAbs and highlight the need to update reference panels regularly. Our data also suggests that combining bnAbs in passive immunization trials would improve coverage of global viruses., Competing Interests: The authors have no competing interests to declare., (Copyright: © 2023 Mkhize et al. This is an open access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.)
- Published
- 2023
- Full Text
- View/download PDF
117. Structural basis for breadth development in the HIV-1 V3-glycan targeting DH270 antibody clonal lineage.
- Author
-
Henderson R, Zhou Y, Stalls V, Wiehe K, Saunders KO, Wagh K, Anasti K, Barr M, Parks R, Alam SM, Korber B, Haynes BF, Bartesaghi A, and Acharya P
- Subjects
- Humans, Antibodies, Neutralizing, HIV Antibodies, Polysaccharides, HIV Infections prevention & control, HIV-1 genetics
- Abstract
Antibody affinity maturation enables adaptive immune responses to a wide range of pathogens. In some individuals broadly neutralizing antibodies develop to recognize rapidly mutating pathogens with extensive sequence diversity. Vaccine design for pathogens such as HIV-1 and influenza has therefore focused on recapitulating the natural affinity maturation process. Here, we determine structures of antibodies in complex with HIV-1 Envelope for all observed members and ancestral states of the broadly neutralizing HIV-1 V3-glycan targeting DH270 antibody clonal B cell lineage. These structures track the development of neutralization breadth from the unmutated common ancestor and define affinity maturation at high spatial resolution. By elucidating contacts mediated by key mutations at different stages of antibody development we identified sites on the epitope-paratope interface that are the focus of affinity optimization. Thus, our results identify bottlenecks on the path to natural affinity maturation and reveal solutions for these that will inform immunogen design aimed at eliciting a broadly neutralizing immune response by vaccination., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
118. Efficacy of antivirals and mRNA vaccination against an XBF clinical isolate.
- Author
-
Uraki R, Ito M, Kiso M, Yamayoshi S, Iwatsuki-Horimoto K, Sakai-Tagawa Y, Imai M, Koga M, Yamamoto S, Adachi E, Saito M, Tsutsumi T, Otani A, Kashima Y, Kikuchi T, Theiler J, Yotsuyanagi H, Suzuki Y, Korber B, and Kawaoka Y
- Abstract
Competing Interests: Y.Kawaoka has received unrelated funding support from Daiichi Sankyo Pharmaceutical, Toyama Chemical, Tauns Laboratories, Inc., Shionogi & Co. LTD, Otsuka Pharmaceutical, KM Biologics, Kyoritsu Seiyaku, Shinya Corporation, and Fuji Rebio. T.K. is employed by Nihon Sumo Kyokai. The remaining authors declare that they have no competing interests.
- Published
- 2023
- Full Text
- View/download PDF
119. SARS-CoV-2 variant transition dynamics are associated with vaccination rates, number of co-circulating variants, and convalescent immunity.
- Author
-
Beesley LJ, Moran KR, Wagh K, Castro LA, Theiler J, Yoon H, Fischer W, Hengartner NW, Korber B, and Del Valle SY
- Subjects
- Humans, Pandemics, Retrospective Studies, SARS-CoV-2 genetics, COVID-19 epidemiology, COVID-19 prevention & control
- Abstract
Background: Throughout the COVID-19 pandemic, the SARS-CoV-2 virus has continued to evolve, with new variants outcompeting existing variants and often leading to different dynamics of disease spread., Methods: In this paper, we performed a retrospective analysis using longitudinal sequencing data to characterize differences in the speed, calendar timing, and magnitude of 16 SARS-CoV-2 variant waves/transitions for 230 countries and sub-country regions, between October 2020 and January 2023. We then clustered geographic locations in terms of their variant behavior across several Omicron variants, allowing us to identify groups of locations exhibiting similar variant transitions. Finally, we explored relationships between heterogeneity in these variant waves and time-varying factors, including vaccination status of the population, governmental policy, and the number of variants in simultaneous competition., Findings: This work demonstrates associations between the behavior of an emerging variant and the number of co-circulating variants as well as the demographic context of the population. We also observed an association between high vaccination rates and variant transition dynamics prior to the Mu and Delta variant transitions., Interpretation: These results suggest the behavior of an emergent variant may be sensitive to the immunologic and demographic context of its location. Additionally, this work represents the most comprehensive characterization of variant transitions globally to date., Funding: Laboratory Directed Research and Development (LDRD), Los Alamos National Laboratory., Competing Interests: Declaration of interests Dr. Theiler received a Bill and Melinda Gates Foundation Grant for bioinformatic analysis unrelated to the present work. The authors have no other conflicts of interest to report., (Copyright © 2023 The Author(s). Published by Elsevier B.V. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
120. US National Institutes of Health Prioritization of SARS-CoV-2 Variants.
- Author
-
Turner S, Alisoltani A, Bratt D, Cohen-Lavi L, Dearlove BL, Drosten C, Fischer WM, Fouchier RAM, Gonzalez-Reiche AS, Jaroszewski L, Khalil Z, LeGresley E, Johnson M, Jones TC, Mühlemann B, O'Connor D, Sedova M, Shukla M, Theiler J, Wallace ZS, Yoon H, Zhang Y, van Bakel H, Degrace MM, Ghedin E, Godzik A, Hertz T, Korber B, Lemieux J, Niewiadomska AM, Post DJ, Rolland M, Scheuermann R, and Smith DJ
- Subjects
- United States epidemiology, Humans, National Institutes of Health (U.S.), SARS-CoV-2 genetics, COVID-19 epidemiology
- Abstract
Since late 2020, SARS-CoV-2 variants have regularly emerged with competitive and phenotypic differences from previously circulating strains, sometimes with the potential to escape from immunity produced by prior exposure and infection. The Early Detection group is one of the constituent groups of the US National Institutes of Health National Institute of Allergy and Infectious Diseases SARS-CoV-2 Assessment of Viral Evolution program. The group uses bioinformatic methods to monitor the emergence, spread, and potential phenotypic properties of emerging and circulating strains to identify the most relevant variants for experimental groups within the program to phenotypically characterize. Since April 2021, the group has prioritized variants monthly. Prioritization successes include rapidly identifying most major variants of SARS-CoV-2 and providing experimental groups within the National Institutes of Health program easy access to regularly updated information on the recent evolution and epidemiology of SARS-CoV-2 that can be used to guide phenotypic investigations.
- Published
- 2023
- Full Text
- View/download PDF
121. Author Correction: Strategies for HIV-1 vaccines that induce broadly neutralizing antibodies.
- Author
-
Haynes BF, Wiehe K, Borrow P, Saunders KO, Korber B, Wagh K, McMichael AJ, Kelsoe G, Hahn BH, Alt F, and Shaw GM
- Published
- 2023
- Full Text
- View/download PDF
122. Characterization of SARS-CoV-2 Omicron BA.2.75 clinical isolates.
- Author
-
Uraki R, Iida S, Halfmann PJ, Yamayoshi S, Hirata Y, Iwatsuki-Horimoto K, Kiso M, Ito M, Furusawa Y, Ueki H, Sakai-Tagawa Y, Kuroda M, Maemura T, Kim T, Mine S, Iwamoto N, Li R, Liu Y, Larson D, Fukushi S, Watanabe S, Maeda K, Wang Z, Ohmagari N, Theiler J, Fischer W, Korber B, Imai M, Suzuki T, and Kawaoka Y
- Subjects
- Animals, Cricetinae, SARS-CoV-2, Biological Assay, DNA Replication, India, Mesocricetus, COVID-19
- Abstract
The prevalence of the Omicron subvariant BA.2.75 rapidly increased in India and Nepal during the summer of 2022, and spread globally. However, the virological features of BA.2.75 are largely unknown. Here, we evaluated the replicative ability and pathogenicity of BA.2.75 clinical isolates in Syrian hamsters. Although we found no substantial differences in weight change among hamsters infected with BA.2, BA.5, or BA.2.75, the replicative ability of BA.2.75 in the lungs is higher than that of BA.2 and BA.5. Of note, BA.2.75 causes focal viral pneumonia in hamsters, characterized by patchy inflammation interspersed in alveolar regions, which is not observed in BA.5-infected hamsters. Moreover, in competition assays, BA.2.75 replicates better than BA.5 in the lungs of hamsters. These results suggest that BA.2.75 can cause more severe respiratory disease than BA.5 and BA.2 in a hamster model and should be closely monitored., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
123. Positive-strand RNA viruses-a Keystone Symposia report.
- Author
-
Cable J, Denison MR, Kielian M, Jackson WT, Bartenschlager R, Ahola T, Mukhopadhyay S, Fremont DH, Kuhn RJ, Shannon A, Frazier MN, Yuen KY, Coyne CB, Wolthers KC, Ming GL, Guenther CS, Moshiri J, Best SM, Schoggins JW, Jurado KA, Ebel GD, Schäfer A, Ng LFP, Kikkert M, Sette A, Harris E, Wing PAC, Eggenberger J, Krishnamurthy SR, Mah MG, Meganck RM, Chung D, Maurer-Stroh S, Andino R, Korber B, Perlman S, Shi PY, Bárcena M, Aicher SM, Vu MN, Kenney DJ, Lindenbach BD, Nishida Y, Rénia L, and Williams EP
- Subjects
- Humans, SARS-CoV-2, Positive-Strand RNA Viruses, Antiviral Agents therapeutic use, Pandemics, COVID-19, Zika Virus, Zika Virus Infection epidemiology, Zika Virus Infection prevention & control, Zika Virus Infection drug therapy
- Abstract
Positive-strand RNA viruses have been the cause of several recent outbreaks and epidemics, including the Zika virus epidemic in 2015, the SARS outbreak in 2003, and the ongoing SARS-CoV-2 pandemic. On June 18-22, 2022, researchers focusing on positive-strand RNA viruses met for the Keystone Symposium "Positive-Strand RNA Viruses" to share the latest research in molecular and cell biology, virology, immunology, vaccinology, and antiviral drug development. This report presents concise summaries of the scientific discussions at the symposium., (© 2023 New York Academy of Sciences.)
- Published
- 2023
- Full Text
- View/download PDF
124. Strategies for HIV-1 vaccines that induce broadly neutralizing antibodies.
- Author
-
Haynes BF, Wiehe K, Borrow P, Saunders KO, Korber B, Wagh K, McMichael AJ, Kelsoe G, Hahn BH, Alt F, and Shaw GM
- Subjects
- Humans, Broadly Neutralizing Antibodies, HIV Antibodies, Antibodies, Neutralizing, Antigens, Viral, HIV-1, AIDS Vaccines, HIV Infections
- Abstract
After nearly four decades of research, a safe and effective HIV-1 vaccine remains elusive. There are many reasons why the development of a potent and durable HIV-1 vaccine is challenging, including the extraordinary genetic diversity of HIV-1 and its complex mechanisms of immune evasion. HIV-1 envelope glycoproteins are poorly recognized by the immune system, which means that potent broadly neutralizing antibodies (bnAbs) are only infrequently induced in the setting of HIV-1 infection or through vaccination. Thus, the biology of HIV-1-host interactions necessitates novel strategies for vaccine development to be designed to activate and expand rare bnAb-producing B cell lineages and to select for the acquisition of critical improbable bnAb mutations. Here we discuss strategies for the induction of potent and broad HIV-1 bnAbs and outline the steps that may be necessary for ultimate success., (© 2022. Springer Nature Limited.)
- Published
- 2023
- Full Text
- View/download PDF
125. Substantial Neutralization Escape by SARS-CoV-2 Omicron Variants BQ.1.1 and XBB.1.
- Author
-
Miller J, Hachmann NP, Collier AY, Lasrado N, Mazurek CR, Patio RC, Powers O, Surve N, Theiler J, Korber B, and Barouch DH
- Subjects
- Humans, Antibodies, Neutralizing genetics, Antibodies, Neutralizing immunology, Antibodies, Viral genetics, Antibodies, Viral immunology, COVID-19 genetics, COVID-19 immunology, COVID-19 virology, Immune Evasion genetics, Immune Evasion immunology, SARS-CoV-2 genetics, SARS-CoV-2 immunology
- Published
- 2023
- Full Text
- View/download PDF
126. Waning Immunity Against XBB.1.5 Following Bivalent mRNA Boosters.
- Author
-
Lasrado N, Collier AY, Miller J, Hachmann NP, Liu J, Sciacca M, Wu C, Anand T, Bondzie EA, Fisher JL, Mazurek CR, Patio RC, Powers O, Rodrigues SL, Rowe M, Surve N, Ty DM, Korber B, and Barouch DH
- Abstract
The SARS-CoV-2 Omicron variant has continued to evolve. XBB is a recombinant between two BA.2 sublineages, XBB.1 includes the G252V mutation, and XBB.1.5 includes the G252V and F486P mutations. XBB.1.5 has rapidly increased in frequency and has become the dominant virus in New England. The bivalent mRNA vaccine boosters have been shown to increase neutralizing antibody (NAb) titers to multiple variants, but the durability of these responses remains to be determined. We assessed humoral and cellular immune responses in 30 participants who received the bivalent mRNA boosters and performed assays at baseline prior to boosting, at week 3 after boosting, and at month 3 after boosting. Our data demonstrate that XBB.1.5 substantially escapes NAb responses but not T cell responses after bivalent mRNA boosting. NAb titers to XBB.1 and XBB.1.5 were similar, suggesting that the F486P mutation confers greater transmissibility but not increased immune escape. By month 3, NAb titers to XBB.1 and XBB.1.5 declined essentially to baseline levels prior to boosting, while NAb titers to other variants declined less strikingly., Competing Interests: Conflicts of Interest The authors report no conflicts of interest.
- Published
- 2023
- Full Text
- View/download PDF
127. Network analysis uncovers the communication structure of SARS-CoV-2 spike protein identifying sites for immunogen design.
- Author
-
Manrique PD, Chakraborty S, Henderson R, Edwards RJ, Mansbach R, Nguyen K, Stalls V, Saunders C, Mansouri K, Acharya P, Korber B, and Gnanakaran S
- Abstract
The COVID-19 pandemic, caused by the SARS-CoV-2 virus, has triggered myriad efforts to understand the structure and dynamics of this complex pathogen. The spike glycoprotein of SARS-CoV-2 is a significant target for immunogens as it is the means by which the virus enters human cells, while simultaneously sporting mutations responsible for immune escape. These functional and escape processes are regulated by complex molecular-level interactions. Our study presents quantitative insights on domain and residue contributions to allosteric communication, immune evasion, and local- and global-level control of functions through the derivation of a weighted graph representation from all-atom MD simulations. Focusing on the ancestral form and the D614G-variant, we provide evidence of the utility of our approach by guiding the selection of a mutation that alters the spike's stability. Taken together, the network approach serves as a valuable tool to evaluate communication "hot-spots" in proteins to guide design of stable immunogens., Competing Interests: The authors declare no competing interests., (© 2022.)
- Published
- 2023
- Full Text
- View/download PDF
128. Multiple lineages of monkeypox virus detected in the United States, 2021-2022.
- Author
-
Gigante CM, Korber B, Seabolt MH, Wilkins K, Davidson W, Rao AK, Zhao H, Smith TG, Hughes CM, Minhaj F, Waltenburg MA, Theiler J, Smole S, Gallagher GR, Blythe D, Myers R, Schulte J, Stringer J, Lee P, Mendoza RM, Griffin-Thomas LA, Crain J, Murray J, Atkinson A, Gonzalez AH, Nash J, Batra D, Damon I, McQuiston J, Hutson CL, McCollum AM, and Li Y
- Subjects
- Humans, Nigeria epidemiology, United States epidemiology, Mutation, Evolution, Molecular, Adenosine genetics, Cytidine genetics, Mpox, Monkeypox enzymology, Mpox, Monkeypox virology, Monkeypox virus genetics, Monkeypox virus isolation & purification, APOBEC Deaminases metabolism, Host-Pathogen Interactions, RNA Editing
- Abstract
Monkeypox is a viral zoonotic disease endemic in Central and West Africa. In May 2022, dozens of non-endemic countries reported hundreds of monkeypox cases, most with no epidemiological link to Africa. We identified two lineages of monkeypox virus (MPXV) among two 2021 and seven 2022 US monkeypox cases: the major 2022 outbreak variant called B.1 and a minor contemporaneously sampled variant called A.2. Analyses of mutations among these two variants revealed an extreme preference for GA-to-AA mutations indicative of human APOBEC3 cytosine deaminase activity among Clade IIb MPXV (previously West African, Nigeria) sampled since 2017. Such mutations were not enriched within other MPXV clades. These findings suggest that APOBEC3 editing may be a recurrent and a dominant driver of MPXV evolution within the current outbreak.
- Published
- 2022
- Full Text
- View/download PDF
129. Immunogenicity and protective efficacy of a rhesus adenoviral vaccine targeting conserved COVID-19 replication transcription complex.
- Author
-
Dagotto G, Ventura JD, Martinez DR, Anioke T, Chung BS, Siamatu M, Barrett J, Miller J, Schäfer A, Yu J, Tostanoski LH, Wagh K, Baric RS, Korber B, and Barouch DH
- Abstract
The COVID-19 pandemic marks the third coronavirus pandemic this century (SARS-CoV-1, MERS, SARS-CoV-2), emphasizing the need to identify and evaluate conserved immunogens for a pan-sarbecovirus vaccine. Here we investigate the potential utility of a T-cell vaccine strategy targeting conserved regions of the sarbecovirus proteome. We identified the most conserved regions of the sarbecovirus proteome as portions of the RNA-dependent RNA polymerase (RdRp) and Helicase proteins, both of which are part of the coronavirus replication transcription complex (RTC). Fitness constraints suggest that as SARS-CoV-2 continues to evolve these regions may better preserve cross-reactive potential of T-cell responses than Spike, Nucleocapsid, or Membrane proteins. We sought to determine if vaccine-elicited T-cell responses to the highly conserved regions of the RTC would reduce viral loads following challenge with SARS-CoV-2 in mice using a rhesus adenovirus serotype 52 (RhAd52) vector. The RhAd52.CoV.Consv vaccine generated robust cellular immunity in mice and led to significant reductions in viral loads in the nasal turbinates following challenge with a mouse-adapted SARS-CoV-2. These data suggest the potential utility of T-cell targeting of conserved regions for a pan-sarbecovirus vaccine., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
130. Neutralization titer biomarker for antibody-mediated prevention of HIV-1 acquisition.
- Author
-
Gilbert PB, Huang Y, deCamp AC, Karuna S, Zhang Y, Magaret CA, Giorgi EE, Korber B, Edlefsen PT, Rossenkhan R, Juraska M, Rudnicki E, Kochar N, Huang Y, Carpp LN, Barouch DH, Mkhize NN, Hermanus T, Kgagudi P, Bekker V, Kaldine H, Mapengo RE, Eaton A, Domin E, West C, Feng W, Tang H, Seaton KE, Heptinstall J, Brackett C, Chiong K, Tomaras GD, Andrew P, Mayer BT, Reeves DB, Sobieszczyk ME, Garrett N, Sanchez J, Gay C, Makhema J, Williamson C, Mullins JI, Hural J, Cohen MS, Corey L, Montefiori DC, and Morris L
- Subjects
- Animals, Humans, Antibodies, Neutralizing, Biomarkers, Broadly Neutralizing Antibodies, HIV Antibodies, HIV Infections, HIV-1
- Abstract
The Antibody Mediated Prevention trials showed that the broadly neutralizing antibody (bnAb) VRC01 prevented acquisition of human immunodeficiency virus-1 (HIV-1) sensitive to VRC01. Using AMP trial data, here we show that the predicted serum neutralization 80% inhibitory dilution titer (PT
80 ) biomarker-which quantifies the neutralization potency of antibodies in an individual's serum against an HIV-1 isolate-can be used to predict HIV-1 prevention efficacy. Similar to the results of nonhuman primate studies, an average PT80 of 200 (meaning a bnAb concentration 200-fold higher than that required to reduce infection by 80% in vitro) against a population of probable exposing viruses was estimated to be required for 90% prevention efficacy against acquisition of these viruses. Based on this result, we suggest that the goal of sustained PT80 <200 against 90% of circulating viruses can be achieved by promising bnAb regimens engineered for long half-lives. We propose the PT80 biomarker as a surrogate endpoint for evaluatinon of bnAb regimens, and as a tool for benchmarking candidate bnAb-inducing vaccines., (© 2022. The Author(s).)- Published
- 2022
- Full Text
- View/download PDF
131. Characterization of the SARS-CoV-2 B.1.621 (Mu) variant.
- Author
-
Halfmann PJ, Kuroda M, Armbrust T, Theiler J, Balaram A, Moreno GK, Accola MA, Iwatsuki-Horimoto K, Valdez R, Stoneman E, Braun K, Yamayoshi S, Somsen E, Baczenas JJ, Mitamura K, Hagihara M, Adachi E, Koga M, McLaughlin M, Rehrauer W, Imai M, Yamamoto S, Tsutsumi T, Saito M, Friedrich TC, O'Connor SL, O'Connor DH, Gordon A, Korber B, and Kawaoka Y
- Subjects
- Antibodies, Neutralizing, Antibodies, Viral, Humans, Immunization, Passive, Membrane Glycoproteins genetics, Neutralization Tests, SARS-CoV-2, Spike Glycoprotein, Coronavirus genetics, Vaccination, COVID-19 Serotherapy, COVID-19 therapy, Viral Envelope Proteins genetics
- Abstract
The SARS-CoV-2 B.1.621 (Mu) variant emerged in January 2021 and was categorized as a variant of interest by the World Health Organization in August 2021. This designation prompted us to study the sensitivity of this variant to antibody neutralization. In a live virus neutralization assay with serum samples from individuals vaccinated with the Pfizer/BioNTech or Moderna mRNA vaccines, we measured neutralization antibody titers against B.1.621, an early isolate (spike 614D), and a variant of concern (B.1.351, Beta variant). We observed reduced neutralizing antibody titers against the B.1.621 variant (3.4- to 7-fold reduction, depending on the serum sample and time after the second vaccination) compared to the early isolate and a similar reduction when compared to B.1.351. Likewise, convalescent serum from hamsters previously infected with an early isolate neutralized B.1.621 to a lower degree. Despite this antibody titer reduction, hamsters could not be efficiently rechallenged with the B.1.621 variant, suggesting that the immune response to the first infection is adequate to provide protection against a subsequent infection with the B.1.621 variant.
- Published
- 2022
- Full Text
- View/download PDF
132. Structural diversity of the SARS-CoV-2 Omicron spike.
- Author
-
Gobeil SM, Henderson R, Stalls V, Janowska K, Huang X, May A, Speakman M, Beaudoin E, Manne K, Li D, Parks R, Barr M, Deyton M, Martin M, Mansouri K, Edwards RJ, Eaton A, Montefiori DC, Sempowski GD, Saunders KO, Wiehe K, Williams W, Korber B, Haynes BF, and Acharya P
- Subjects
- Angiotensin-Converting Enzyme 2, Humans, Mutation, Spike Glycoprotein, Coronavirus chemistry, COVID-19, SARS-CoV-2 genetics
- Abstract
Aided by extensive spike protein mutation, the SARS-CoV-2 Omicron variant overtook the previously dominant Delta variant. Spike conformation plays an essential role in SARS-CoV-2 evolution via changes in receptor-binding domain (RBD) and neutralizing antibody epitope presentation, affecting virus transmissibility and immune evasion. Here, we determine cryo-EM structures of the Omicron and Delta spikes to understand the conformational impacts of mutations in each. The Omicron spike structure revealed an unusually tightly packed RBD organization with long range impacts that were not observed in the Delta spike. Binding and crystallography revealed increased flexibility at the functionally critical fusion peptide site in the Omicron spike. These results reveal a highly evolved Omicron spike architecture with possible impacts on its high levels of immune evasion and transmissibility., Competing Interests: Declaration of interests B.F.H., G.D.S., and K.O.S. have patents submitted on the SARS-CoV-2 monoclonal antibodies studied in this paper. R.H., K.O.S., B.F.H., and P.A. have patents submitted on the SARS-CoV-2 rS2d and u1s2q designs. Other authors declare no competing interests., (Copyright © 2022 Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
133. Safety and antiviral activity of triple combination broadly neutralizing monoclonal antibody therapy against HIV-1: a phase 1 clinical trial.
- Author
-
Julg B, Stephenson KE, Wagh K, Tan SC, Zash R, Walsh S, Ansel J, Kanjilal D, Nkolola J, Walker-Sperling VEK, Ophel J, Yanosick K, Borducchi EN, Maxfield L, Abbink P, Peter L, Yates NL, Wesley MS, Hassell T, Gelderblom HC, deCamp A, Mayer BT, Sato A, Gerber MW, Giorgi EE, Gama L, Koup RA, Mascola JR, Monczor A, Lupo S, Rolle CP, Arduino R, DeJesus E, Tomaras GD, Seaman MS, Korber B, and Barouch DH
- Subjects
- Adult, Antibodies, Monoclonal adverse effects, Antibodies, Neutralizing, Antiviral Agents therapeutic use, Broadly Neutralizing Antibodies, HIV Antibodies, Humans, Viremia drug therapy, HIV Infections, HIV Seropositivity, HIV-1
- Abstract
HIV-1 therapy with single or dual broadly neutralizing antibodies (bNAbs) has shown viral escape, indicating that at least a triple bNAb therapy may be needed for robust suppression of viremia. We performed a two-part study consisting of a single-center, randomized, double-blind, dose-escalation, placebo-controlled first-in-human trial of the HIV-1 V2-glycan-specific antibody PGDM1400 alone or in combination with the V3-glycan-specific antibody PGT121 in 24 adults without HIV in part 1, as well as a multi-center, open-label trial of the combination of PGDM1400, PGT121 and the CD4-binding-site antibody VRC07-523LS in five viremic adults living with HIV not on antiretroviral therapy (ART) in part 2 ( NCT03205917 ). The primary endpoints were safety, tolerability and pharmacokinetics for both parts and antiviral activity among viremic adults living with HIV and not on ART for part 2 of the study. The secondary endpoints were changes in CD4
+ T cell counts and development of HIV-1 sequence variations associated with PGDM1400, PGT121 and VRC07-523LS resistance in part 2. Intravenously administered PGDM1400 was safe and well-tolerated at doses up to 30 mg kg-1 and when given in combination with PGT121 and VRC07-523LS. A single intravenous infusion of 20 mg kg-1 of each of the three antibodies reduced plasma HIV RNA levels in viremic individuals by a maximum mean of 2.04 log10 copies per ml; however, viral rebound occurred in all participants within a median of 20 days after nadir. Rebound viruses demonstrated partial to complete resistance to PGDM1400 and PGT121 in vitro, whereas susceptibility to VRC07-523LS was preserved. Viral rebound occurred despite mean VRC07-523LS serum concentrations of 93 µg ml-1 . The trial met the pre-specified endpoints. Our data suggest that future bNAb combinations likely need to achieve broad antiviral activity, while also maintaining high serum concentrations, to mediate viral control., (© 2022. The Author(s).)- Published
- 2022
- Full Text
- View/download PDF
134. Defining the risk of SARS-CoV-2 variants on immune protection.
- Author
-
DeGrace MM, Ghedin E, Frieman MB, Krammer F, Grifoni A, Alisoltani A, Alter G, Amara RR, Baric RS, Barouch DH, Bloom JD, Bloyet LM, Bonenfant G, Boon ACM, Boritz EA, Bratt DL, Bricker TL, Brown L, Buchser WJ, Carreño JM, Cohen-Lavi L, Darling TL, Davis-Gardner ME, Dearlove BL, Di H, Dittmann M, Doria-Rose NA, Douek DC, Drosten C, Edara VV, Ellebedy A, Fabrizio TP, Ferrari G, Fischer WM, Florence WC, Fouchier RAM, Franks J, García-Sastre A, Godzik A, Gonzalez-Reiche AS, Gordon A, Haagmans BL, Halfmann PJ, Ho DD, Holbrook MR, Huang Y, James SL, Jaroszewski L, Jeevan T, Johnson RM, Jones TC, Joshi A, Kawaoka Y, Kercher L, Koopmans MPG, Korber B, Koren E, Koup RA, LeGresley EB, Lemieux JE, Liebeskind MJ, Liu Z, Livingston B, Logue JP, Luo Y, McDermott AB, McElrath MJ, Meliopoulos VA, Menachery VD, Montefiori DC, Mühlemann B, Munster VJ, Munt JE, Nair MS, Netzl A, Niewiadomska AM, O'Dell S, Pekosz A, Perlman S, Pontelli MC, Rockx B, Rolland M, Rothlauf PW, Sacharen S, Scheuermann RH, Schmidt SD, Schotsaert M, Schultz-Cherry S, Seder RA, Sedova M, Sette A, Shabman RS, Shen X, Shi PY, Shukla M, Simon V, Stumpf S, Sullivan NJ, Thackray LB, Theiler J, Thomas PG, Trifkovic S, Türeli S, Turner SA, Vakaki MA, van Bakel H, VanBlargan LA, Vincent LR, Wallace ZS, Wang L, Wang M, Wang P, Wang W, Weaver SC, Webby RJ, Weiss CD, Wentworth DE, Weston SM, Whelan SPJ, Whitener BM, Wilks SH, Xie X, Ying B, Yoon H, Zhou B, Hertz T, Smith DJ, Diamond MS, Post DJ, and Suthar MS
- Subjects
- Animals, Biological Evolution, COVID-19 Vaccines, Humans, National Institute of Allergy and Infectious Diseases (U.S.), Pandemics prevention & control, Pharmacogenomic Variants, United States epidemiology, Virulence, COVID-19, SARS-CoV-2 genetics, SARS-CoV-2 pathogenicity
- Abstract
The global emergence of many severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants jeopardizes the protective antiviral immunity induced after infection or vaccination. To address the public health threat caused by the increasing SARS-CoV-2 genomic diversity, the National Institute of Allergy and Infectious Diseases within the National Institutes of Health established the SARS-CoV-2 Assessment of Viral Evolution (SAVE) programme. This effort was designed to provide a real-time risk assessment of SARS-CoV-2 variants that could potentially affect the transmission, virulence, and resistance to infection- and vaccine-induced immunity. The SAVE programme is a critical data-generating component of the US Government SARS-CoV-2 Interagency Group to assess implications of SARS-CoV-2 variants on diagnostics, vaccines and therapeutics, and for communicating public health risk. Here we describe the coordinated approach used to identify and curate data about emerging variants, their impact on immunity and effects on vaccine protection using animal models. We report the development of reagents, methodologies, models and notable findings facilitated by this collaborative approach and identify future challenges. This programme is a template for the response to rapidly evolving pathogens with pandemic potential by monitoring viral evolution in the human population to identify variants that could reduce the effectiveness of countermeasures., (© 2022. Springer Nature Limited.)
- Published
- 2022
- Full Text
- View/download PDF
135. SARS-CoV-2 Omicron Variant Neutralization after mRNA-1273 Booster Vaccination.
- Author
-
Pajon R, Doria-Rose NA, Shen X, Schmidt SD, O'Dell S, McDanal C, Feng W, Tong J, Eaton A, Maglinao M, Tang H, Manning KE, Edara VV, Lai L, Ellis M, Moore KM, Floyd K, Foster SL, Posavad CM, Atmar RL, Lyke KE, Zhou T, Wang L, Zhang Y, Gaudinski MR, Black WP, Gordon I, Guech M, Ledgerwood JE, Misasi JN, Widge A, Sullivan NJ, Roberts PC, Beigel JH, Korber B, Baden LR, El Sahly H, Chalkias S, Zhou H, Feng J, Girard B, Das R, Aunins A, Edwards DK, Suthar MS, Mascola JR, and Montefiori DC
- Published
- 2022
- Full Text
- View/download PDF
136. mRNA-encoded HIV-1 Env trimer ferritin nanoparticles induce monoclonal antibodies that neutralize heterologous HIV-1 isolates in mice.
- Author
-
Mu Z, Wiehe K, Saunders KO, Henderson R, Cain DW, Parks R, Martik D, Mansouri K, Edwards RJ, Newman A, Lu X, Xia SM, Eaton A, Bonsignori M, Montefiori D, Han Q, Venkatayogi S, Evangelous T, Wang Y, Rountree W, Korber B, Wagh K, Tam Y, Barbosa C, Alam SM, Williams WB, Tian M, Alt FW, Pardi N, Weissman D, and Haynes BF
- Subjects
- Animals, Antibodies, Monoclonal, Antibodies, Neutralizing, COVID-19 Vaccines, Epitopes, Ferritins genetics, HIV Antibodies, Humans, Liposomes, Mice, RNA, Messenger, env Gene Products, Human Immunodeficiency Virus genetics, AIDS Vaccines, COVID-19, HIV-1, Nanoparticles
- Abstract
The success of nucleoside-modified mRNAs in lipid nanoparticles (mRNA-LNP) as COVID-19 vaccines heralded a new era of vaccine development. For HIV-1, multivalent envelope (Env) trimer protein nanoparticles are superior immunogens compared with trimers alone for priming of broadly neutralizing antibody (bnAb) B cell lineages. The successful expression of complex multivalent nanoparticle immunogens with mRNAs has not been demonstrated. Here, we show that mRNAs can encode antigenic Env trimers on ferritin nanoparticles that initiate bnAb precursor B cell expansion and induce serum autologous tier 2 neutralizing activity in bnAb precursor V
H + VL knock-in mice. Next-generation sequencing demonstrates acquisition of critical mutations, and monoclonal antibodies that neutralize heterologous HIV-1 isolates are isolated. Thus, mRNA-LNP can encode complex immunogens and may be of use in design of germline-targeting and sequential boosting immunogens for HIV-1 vaccine development., Competing Interests: Declaration of interests B.F.H., K.O.S., and K.W. have patent applications on some of the concepts and immunogens discussed in this paper., (Copyright © 2022 The Authors. Published by Elsevier Inc. All rights reserved.)- Published
- 2022
- Full Text
- View/download PDF
137. Booster of mRNA-1273 Strengthens SARS-CoV-2 Omicron Neutralization.
- Author
-
Doria-Rose NA, Shen X, Schmidt SD, O'Dell S, McDanal C, Feng W, Tong J, Eaton A, Maglinao M, Tang H, Manning KE, Edara VV, Lai L, Ellis M, Moore K, Floyd K, Foster SL, Atmar RL, Lyke KE, Zhou T, Wang L, Zhang Y, Gaudinski MR, Black WP, Gordon I, Guech M, Ledgerwood JE, Misasi JN, Widge A, Roberts PC, Beigel J, Korber B, Pajon R, Mascola JR, Suthar MS, and Montefiori DC
- Abstract
The Omicron variant of SARS-CoV-2 is raising concerns because of its increased transmissibility and potential for reduced susceptibility to antibody neutralization. To assess the potential risk of this variant to existing vaccines, serum samples from mRNA-1273 vaccine recipients were tested for neutralizing activity against Omicron and compared to neutralization titers against D614G and Beta in live virus and pseudovirus assays. Omicron was 41-84-fold less sensitive to neutralization than D614G and 5.3-7.4-fold less sensitive than Beta when assayed with serum samples obtained 4 weeks after 2 standard inoculations with 100 μg mRNA-1273. A 50 μg boost increased Omicron neutralization titers and may substantially reduce the risk of symptomatic vaccine breakthrough infections.
- Published
- 2021
- Full Text
- View/download PDF
138. Preexisting memory CD4+ T cells contribute to the primary response in an HIV-1 vaccine trial.
- Author
-
Campion SL, Brenna E, Thomson E, Fischer W, Ladell K, McLaren JE, Price DA, Frahm N, McElrath JM, Cohen KW, Maenza JR, Walsh SR, Baden LR, Haynes BF, Korber B, Borrow P, and McMichael AJ
- Subjects
- Adolescent, Adult, Antibodies, Neutralizing immunology, DNA analysis, Double-Blind Method, Epitopes chemistry, Female, HIV Infections immunology, Humans, Immunity, Immunization, Secondary, Male, Middle Aged, Vaccines, DNA immunology, Vaccinia virus immunology, Young Adult, env Gene Products, Human Immunodeficiency Virus immunology, AIDS Vaccines immunology, CD4-Positive T-Lymphocytes immunology, HIV Antibodies immunology, HIV-1 immunology, Immunologic Memory
- Abstract
Naive and memory CD4+ T cells reactive with human immunodeficiency virus type 1 (HIV-1) are detectable in unexposed, unimmunized individuals. The contribution of preexisting CD4+ T cells to a primary immune response was investigated in 20 HIV-1-seronegative volunteers vaccinated with an HIV-1 envelope (Env) plasmid DNA prime and recombinant modified vaccinia virus Ankara (MVA) boost in the HVTN 106 vaccine trial (clinicaltrials.gov NCT02296541). Prevaccination naive or memory CD4+ T cell responses directed against peptide epitopes in Env were identified in 14 individuals. After priming with DNA, 40% (8/20) of the elicited responses matched epitopes detected in the corresponding preimmunization memory repertoires, and clonotypes were shared before and after vaccination in 2 representative volunteers. In contrast, there were no shared epitope specificities between the preimmunization memory compartment and responses detected after boosting with recombinant MVA expressing a heterologous Env. Preexisting memory CD4+ T cells therefore shape the early immune response to vaccination with a previously unencountered HIV-1 antigen.
- Published
- 2021
- Full Text
- View/download PDF
139. Safety, pharmacokinetics and antiviral activity of PGT121, a broadly neutralizing monoclonal antibody against HIV-1: a randomized, placebo-controlled, phase 1 clinical trial.
- Author
-
Stephenson KE, Julg B, Tan CS, Zash R, Walsh SR, Rolle CP, Monczor AN, Lupo S, Gelderblom HC, Ansel JL, Kanjilal DG, Maxfield LF, Nkolola J, Borducchi EN, Abbink P, Liu J, Peter L, Chandrashekar A, Nityanandam R, Lin Z, Setaro A, Sapiente J, Chen Z, Sunner L, Cassidy T, Bennett C, Sato A, Mayer B, Perelson AS, deCamp A, Priddy FH, Wagh K, Giorgi EE, Yates NL, Arduino RC, DeJesus E, Tomaras GD, Seaman MS, Korber B, and Barouch DH
- Subjects
- Adult, Antiretroviral Therapy, Highly Active, Antiviral Agents immunology, Antiviral Agents pharmacokinetics, Broadly Neutralizing Antibodies immunology, CD4-Positive T-Lymphocytes drug effects, CD4-Positive T-Lymphocytes virology, Double-Blind Method, Female, HIV Envelope Protein gp120 antagonists & inhibitors, HIV Envelope Protein gp120 immunology, HIV Infections genetics, HIV Infections pathology, HIV Infections virology, HIV-1 pathogenicity, Humans, Male, Middle Aged, Peptide Fragments antagonists & inhibitors, Peptide Fragments immunology, Placebos, Viral Load drug effects, Viral Load immunology, Young Adult, Antiviral Agents administration & dosage, Broadly Neutralizing Antibodies administration & dosage, HIV Infections drug therapy, HIV-1 drug effects
- Abstract
Human immunodeficiency virus (HIV)-1-specific broadly neutralizing monoclonal antibodies are currently under development to treat and prevent HIV-1 infection. We performed a single-center, randomized, double-blind, dose-escalation, placebo-controlled trial of a single administration of the HIV-1 V3-glycan-specific antibody PGT121 at 3, 10 and 30 mg kg
-1 in HIV-uninfected adults and HIV-infected adults on antiretroviral therapy (ART), as well as a multicenter, open-label trial of one infusion of PGT121 at 30 mg kg-1 in viremic HIV-infected adults not on ART (no. NCT02960581). The primary endpoints were safety and tolerability, pharmacokinetics (PK) and antiviral activity in viremic HIV-infected adults not on ART. The secondary endpoints were changes in anti-PGT121 antibody titers and CD4+ T-cell count, and development of HIV-1 sequence variations associated with PGT121 resistance. Among 48 participants enrolled, no treatment-related serious adverse events, potential immune-mediated diseases or Grade 3 or higher adverse events were reported. The most common reactions among PGT121 recipients were intravenous/injection site tenderness, pain and headache. Absolute and relative CD4+ T-cell counts did not change following PGT121 infusion in HIV-infected participants. Neutralizing anti-drug antibodies were not elicited. PGT121 reduced plasma HIV RNA levels by a median of 1.77 log in viremic participants, with a viral load nadir at a median of 8.5 days. Two individuals with low baseline viral loads experienced ART-free viral suppression for ≥168 days following antibody infusion, and rebound viruses in these individuals demonstrated full or partial PGT121 sensitivity. The trial met the prespecified endpoints. These data suggest that further investigation of the potential of antibody-based therapeutic strategies for long-term suppression of HIV is warranted, including in individuals off ART and with low viral load., (© 2021. The Author(s).)- Published
- 2021
- Full Text
- View/download PDF
140. Effect of natural mutations of SARS-CoV-2 on spike structure, conformation, and antigenicity.
- Author
-
Gobeil SM, Janowska K, McDowell S, Mansouri K, Parks R, Stalls V, Kopp MF, Manne K, Li D, Wiehe K, Saunders KO, Edwards RJ, Korber B, Haynes BF, Henderson R, and Acharya P
- Subjects
- Amino Acid Substitution, Angiotensin-Converting Enzyme 2 metabolism, Animals, Antibodies, Neutralizing immunology, Antibodies, Viral immunology, Antigens, Viral immunology, COVID-19 transmission, COVID-19 veterinary, COVID-19 virology, Cryoelectron Microscopy, Host Adaptation, Humans, Immune Evasion, Mink virology, Models, Molecular, Mutation, Protein Binding, Protein Conformation, Protein Interaction Domains and Motifs, Protein Structure, Quaternary, Protein Subunits chemistry, Receptors, Coronavirus metabolism, SARS-CoV-2 immunology, Spike Glycoprotein, Coronavirus immunology, Spike Glycoprotein, Coronavirus metabolism, SARS-CoV-2 chemistry, SARS-CoV-2 genetics, Spike Glycoprotein, Coronavirus chemistry, Spike Glycoprotein, Coronavirus genetics
- Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants with multiple spike mutations enable increased transmission and antibody resistance. We combined cryo-electron microscopy (cryo-EM), binding, and computational analyses to study variant spikes, including one that was involved in transmission between minks and humans, and others that originated and spread in human populations. All variants showed increased angiotensin-converting enzyme 2 (ACE2) receptor binding and increased propensity for receptor binding domain (RBD)-up states. While adaptation to mink resulted in spike destabilization, the B.1.1.7 (UK) spike balanced stabilizing and destabilizing mutations. A local destabilizing effect of the RBD E484K mutation was implicated in resistance of the B.1.1.28/P.1 (Brazil) and B.1.351 (South Africa) variants to neutralizing antibodies. Our studies revealed allosteric effects of mutations and mechanistic differences that drive either interspecies transmission or escape from antibody neutralization., (Copyright © 2021, American Association for the Advancement of Science.)
- Published
- 2021
- Full Text
- View/download PDF
141. HIV-1 and SARS-CoV-2: Patterns in the evolution of two pandemic pathogens.
- Author
-
Fischer W, Giorgi EE, Chakraborty S, Nguyen K, Bhattacharya T, Theiler J, Goloboff PA, Yoon H, Abfalterer W, Foley BT, Tegally H, San JE, de Oliveira T, Gnanakaran S, and Korber B
- Subjects
- COVID-19 immunology, Evolution, Molecular, Genome, Viral, Humans, Immune Evasion, Mutation, Receptors, Virus genetics, Recombination, Genetic, Selection, Genetic, Spike Glycoprotein, Coronavirus chemistry, Spike Glycoprotein, Coronavirus genetics, Viral Proteins chemistry, Viral Proteins genetics, HIV-1 genetics, Pandemics, SARS-CoV-2 genetics
- Abstract
Humanity is currently facing the challenge of two devastating pandemics caused by two very different RNA viruses: HIV-1, which has been with us for decades, and SARS-CoV-2, which has swept the world in the course of a single year. The same evolutionary strategies that drive HIV-1 evolution are at play in SARS-CoV-2. Single nucleotide mutations, multi-base insertions and deletions, recombination, and variation in surface glycans all generate the variability that, guided by natural selection, enables both HIV-1's extraordinary diversity and SARS-CoV-2's slower pace of mutation accumulation. Even though SARS-CoV-2 diversity is more limited, recently emergent SARS-CoV-2 variants carry Spike mutations that have important phenotypic consequences in terms of both antibody resistance and enhanced infectivity. We review and compare how these mutational patterns manifest in these two distinct viruses to provide the variability that fuels their evolution by natural selection., Competing Interests: Declaration of interests B.K., W.F., J.T., T.B., and S.G. have provisional patents and patents relating to vaccine design to address viral diversity as applied to HIV-1 and/or SARS-CoV-2., (Published by Elsevier Inc.)
- Published
- 2021
- Full Text
- View/download PDF
142. SARS-CoV-2 Variants of Interest and Concern naming scheme conducive for global discourse.
- Author
-
Konings F, Perkins MD, Kuhn JH, Pallen MJ, Alm EJ, Archer BN, Barakat A, Bedford T, Bhiman JN, Caly L, Carter LL, Cullinane A, de Oliveira T, Druce J, El Masry I, Evans R, Gao GF, Gorbalenya AE, Hamblion E, Herring BL, Hodcroft E, Holmes EC, Kakkar M, Khare S, Koopmans MPG, Korber B, Leite J, MacCannell D, Marklewitz M, Maurer-Stroh S, Rico JAM, Munster VJ, Neher R, Munnink BO, Pavlin BI, Peiris M, Poon L, Pybus O, Rambaut A, Resende P, Subissi L, Thiel V, Tong S, van der Werf S, von Gottberg A, Ziebuhr J, and Van Kerkhove MD
- Subjects
- COVID-19 virology, Communication, Global Health, Humans, World Health Organization, SARS-CoV-2 classification, SARS-CoV-2 genetics, Terminology as Topic
- Published
- 2021
- Full Text
- View/download PDF
143. Neutralization of SARS-CoV-2 Variants B.1.429 and B.1.351.
- Author
-
Shen X, Tang H, Pajon R, Smith G, Glenn GM, Shi W, Korber B, and Montefiori DC
- Subjects
- 2019-nCoV Vaccine mRNA-1273, Antibodies, Neutralizing blood, Antibodies, Viral blood, COVID-19 immunology, Cross Reactions immunology, Humans, Microbiological Techniques, Mutation, Neutralization Tests, SARS-CoV-2 genetics, Spike Glycoprotein, Coronavirus genetics, Antibodies, Neutralizing immunology, COVID-19 prevention & control, COVID-19 Vaccines immunology, SARS-CoV-2 immunology
- Published
- 2021
- Full Text
- View/download PDF
144. Structural and genetic convergence of HIV-1 neutralizing antibodies in vaccinated non-human primates.
- Author
-
Cai F, Chen WH, Wu W, Jones JA, Choe M, Gohain N, Shen X, LaBranche C, Eaton A, Sutherland L, Lee EM, Hernandez GE, Wu NR, Scearce R, Seaman MS, Moody MA, Santra S, Wiehe K, Tomaras GD, Wagh K, Korber B, Bonsignori M, Montefiori DC, Haynes BF, de Val N, Joyce MG, and Saunders KO
- Subjects
- Animals, HIV Infections immunology, HIV-1 immunology, Humans, Macaca mulatta, AIDS Vaccines immunology, Antibodies, Neutralizing immunology, HIV Antibodies immunology, env Gene Products, Human Immunodeficiency Virus immunology
- Abstract
A primary goal of HIV-1 vaccine development is the consistent elicitation of protective, neutralizing antibodies. While highly similar neutralizing antibodies (nAbs) have been isolated from multiple HIV-infected individuals, it is unclear whether vaccination can consistently elicit highly similar nAbs in genetically diverse primates. Here, we show in three outbred rhesus macaques that immunization with Env elicits a genotypically and phenotypically conserved nAb response. From these vaccinated macaques, we isolated four antibody lineages that had commonalities in immunoglobulin variable, diversity, and joining gene segment usage. Atomic-level structures of the antigen binding fragments of the two most similar antibodies showed nearly identical paratopes. The Env binding modes of each of the four vaccine-induced nAbs were distinct from previously known monoclonal HIV-1 neutralizing antibodies, but were nearly identical to each other. The similarities of these antibodies show that the immune system in outbred primates can respond to HIV-1 Env vaccination with a similar structural and genotypic solution for recognizing a particular neutralizing epitope. These results support rational vaccine design for HIV-1 that aims to reproducibly elicit, in genetically diverse primates, nAbs with specific paratope structures capable of binding conserved epitopes., Competing Interests: I have read the journal’s policy and the authors of this manuscript have the following competing interests: B.F.H. has filed International Patent Application PCT/US2004/030397 and National Stage Applications directed to CON-S and its use as an immunogen. K.O.S. has filed International Patent Application PCT/US2019/020436 directed to CON-S and its use as an immunogen.
- Published
- 2021
- Full Text
- View/download PDF
145. Exploring the Role of Glycans in the Interaction of SARS-CoV-2 RBD and Human Receptor ACE2.
- Author
-
Nguyen K, Chakraborty S, Mansbach RA, Korber B, and Gnanakaran S
- Subjects
- Amino Acid Sequence, Binding Sites, Glycosylation, Host Microbial Interactions, Humans, Molecular Dynamics Simulation, Mutation, Protein Binding, Protein Conformation, Protein Interaction Domains and Motifs, Virus Attachment, Angiotensin-Converting Enzyme 2 chemistry, COVID-19 virology, Polysaccharides chemistry, SARS-CoV-2 physiology, Spike Glycoprotein, Coronavirus chemistry
- Abstract
COVID-19 is a highly infectious respiratory disease caused by the novel coronavirus SARS-CoV-2. It has become a global pandemic and its frequent mutations may pose new challenges for vaccine design. During viral infection, the Spike RBD of SARS-CoV-2 binds the human host cell receptor ACE2, enabling the virus to enter the host cell. Both the Spike and ACE2 are densely glycosylated, and it is unclear how distinctive glycan types may modulate the interaction of RBD and ACE2. Detailed understanding of these determinants is key for the development of novel therapeutic strategies. To this end, we perform extensive all-atom simulations of the (i) RBD-ACE2 complex without glycans, (ii) RBD-ACE2 with oligomannose MAN9 glycans in ACE2, and (iii) RBD-ACE2 with complex FA2 glycans in ACE2. These simulations identify the key residues at the RBD-ACE2 interface that form contacts with higher probabilities, thus providing a quantitative evaluation that complements recent structural studies. Notably, we find that this RBD-ACE2 contact signature is not altered by the presence of different glycoforms, suggesting that RBD-ACE2 interaction is robust. Applying our simulated results, we illustrate how the recently prevalent N501Y mutation may alter specific interactions with host ACE2 that facilitate the virus-host binding. Furthermore, our simulations reveal how the glycan on Asn90 of ACE2 can play a distinct role in the binding and unbinding of RBD. Finally, an energetics analysis shows that MAN9 glycans on ACE2 decrease RBD-ACE2 affinity, while FA2 glycans lead to enhanced binding of the complex. Together, our results provide a more comprehensive picture of the detailed interplay between virus and human receptor, which is much needed for the discovery of effective treatments that aim at modulating the physical-chemical properties of this virus.
- Published
- 2021
- Full Text
- View/download PDF
146. Effect of epitope variant co-delivery on the depth of CD8 T cell responses induced by HIV-1 conserved mosaic vaccines.
- Author
-
Wee EG, Moyo N, Hannoun Z, Giorgi EE, Korber B, and Hanke T
- Abstract
To stop the HIV-1 pandemic, vaccines must induce responses capable of controlling vast HIV-1 variants circulating in the population as well as those evolved in each individual following transmission. Numerous strategies have been proposed, of which the most promising include focusing responses on the vulnerable sites of HIV-1 displaying the least entropy among global isolates and using algorithms that maximize vaccine match to circulating HIV-1 variants by vaccine cocktails of optimized complementing sequences. In this study, we investigated CD8 T cell responses induced by a bi-valent mosaic of highly conserved HIVconsvX regions delivered by a combination of simian adenovirus ChAdOx1 and poxvirus MVA. We compared partially and fully mono- and bi-valent prime-boost regimens and their ability to elicit T cells recognizing natural epitope variants using an interferon-γ enzyme-linked immunospot (ELISPOT) assay. We used 11 well-defined CD8 T cell epitopes in two mouse haplotypes and, for each epitope, assessed recognition of the two vaccine forms together with the other most frequent epitope variants in the HIV-1 database. We conclude that for the magnitude and depth of epitope recognition, CD8 T cell responses benefitted in most comparisons from the combined bi-valent mosaic and envisage the main advantage of the bi-valent vaccine during its deployment to diverse populations., Competing Interests: T.H. and B.K. are co-inventors on patents PCT/US14/58422 and EP14846993.5. The remaining authors declare no competing interests., (© 2021 OXFORD UNIVERSITY.)
- Published
- 2021
- Full Text
- View/download PDF
147. The SARS-CoV-2 Spike variant D614G favors an open conformational state.
- Author
-
Mansbach RA, Chakraborty S, Nguyen K, Montefiori DC, Korber B, and Gnanakaran S
- Subjects
- Amino Acid Sequence, Angiotensin-Converting Enzyme 2 chemistry, Angiotensin-Converting Enzyme 2 metabolism, Antibodies, Neutralizing immunology, COVID-19 pathology, COVID-19 virology, Glycosylation, Humans, Hydrogen Bonding, Molecular Dynamics Simulation, Mutation, Protein Binding, Protein Structure, Quaternary, Protein Subunits chemistry, Protein Subunits immunology, SARS-CoV-2 immunology, SARS-CoV-2 isolation & purification, Spike Glycoprotein, Coronavirus metabolism, Virus Internalization, SARS-CoV-2 genetics, Spike Glycoprotein, Coronavirus chemistry
- Abstract
The COVID-19 (coronavirus disease 2019) pandemic underwent a rapid transition with the emergence of a dominant viral variant (from the "D-form" to the "G-form") that carried an amino acid substitution D614G in its "Spike" protein. The G-form is more infectious in vitro and is associated with increased viral loads in the upper airways. To gain insight into the molecular-level underpinnings of these characteristics, we used microsecond all-atom simulations. We show that changes in the protein energetics favor a higher population of infection-capable states in the G-form through release of asymmetry present in the D-form inter-protomer interactions. Thus, the increased infectivity of the G-form is likely due to a higher rate of profitable binding encounters with the host receptor. It is also predicted to be more neutralization sensitive owing to enhanced exposure of the receptor binding domain, a key target region for neutralizing antibodies. These results are critical for vaccine design., (Copyright © 2021 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works. Distributed under a Creative Commons Attribution NonCommercial License 4.0 (CC BY-NC).)
- Published
- 2021
- Full Text
- View/download PDF
148. SARS-CoV-2 variant B.1.1.7 is susceptible to neutralizing antibodies elicited by ancestral spike vaccines.
- Author
-
Shen X, Tang H, McDanal C, Wagh K, Fischer W, Theiler J, Yoon H, Li D, Haynes BF, Sanders KO, Gnanakaran S, Hengartner N, Pajon R, Smith G, Glenn GM, Korber B, and Montefiori DC
- Subjects
- 2019-nCoV Vaccine mRNA-1273, Adolescent, Adult, Aged, Aged, 80 and over, Antibodies, Monoclonal immunology, Female, Humans, Male, Middle Aged, Mutation, Neutralization Tests, Spike Glycoprotein, Coronavirus genetics, Spike Glycoprotein, Coronavirus immunology, Young Adult, Antibodies, Neutralizing immunology, Antibodies, Viral immunology, COVID-19 Vaccines immunology, SARS-CoV-2 immunology
- Abstract
All current vaccines for COVID-19 utilize ancestral SARS-CoV-2 spike with the goal of generating protective neutralizing antibodies. The recent emergence and rapid spread of several SARS-CoV-2 variants carrying multiple spike mutations raise concerns about possible immune escape. One variant, first identified in the United Kingdom (B.1.1.7, also called 20I/501Y.V1), contains eight spike mutations with potential to impact antibody therapy, vaccine efficacy, and risk of reinfection. Here, we show that B.1.1.7 remains sensitive to neutralization, albeit at moderately reduced levels (∼sim;2-fold), by serum samples from convalescent individuals and recipients of an mRNA vaccine (mRNA-1273, Moderna) and a protein nanoparticle vaccine (NVX-CoV2373, Novavax). A subset of monoclonal antibodies to the receptor binding domain (RBD) of spike are less effective against the variant, while others are largely unaffected. These findings indicate that variant B.1.1.7 is unlikely to be a major concern for current vaccines or for an increased risk of reinfection., Competing Interests: Declaration of interests R.P. is an employee of Moderna, Inc. G.S. and G.M.G. are employees of Novavax, Inc., (Copyright © 2021 Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
149. Epigraph hemagglutinin vaccine induces broad cross-reactive immunity against swine H3 influenza virus.
- Author
-
Bullard BL, Corder BN, DeBeauchamp J, Rubrum A, Korber B, Webby RJ, and Weaver EA
- Subjects
- Animals, Antibody Formation immunology, Epitopes immunology, Female, Humans, Influenza, Human blood, Influenza, Human immunology, Influenza, Human virology, Lung pathology, Lung virology, Male, Mice, Inbred BALB C, Orthomyxoviridae Infections blood, Orthomyxoviridae Infections immunology, Orthomyxoviridae Infections prevention & control, Orthomyxoviridae Infections virology, Swine, T-Lymphocytes immunology, Vaccination, Weight Loss, Mice, Algorithms, Cross Reactions immunology, Hemagglutinin Glycoproteins, Influenza Virus immunology, Immunity, Influenza A virus immunology, Influenza Vaccines immunology
- Abstract
Influenza A virus infection in swine impacts the agricultural industry in addition to its zoonotic potential. Here, we utilize epigraph, a computational algorithm, to design a universal swine H3 influenza vaccine. The epigraph hemagglutinin proteins are delivered using an Adenovirus type 5 vector and are compared to a wild type hemagglutinin and the commercial inactivated vaccine, FluSure. In mice, epigraph vaccination leads to significant cross-reactive antibody and T-cell responses against a diverse panel of swH3 isolates. Epigraph vaccination also reduces weight loss and lung viral titers in mice after challenge with three divergent swH3 viruses. Vaccination studies in swine, the target species for this vaccine, show stronger levels of cross-reactive antibodies and T-cell responses after immunization with the epigraph vaccine compared to the wild type and FluSure vaccines. In both murine and swine models, epigraph vaccination shows superior cross-reactive immunity that should be further investigated as a universal swH3 vaccine.
- Published
- 2021
- Full Text
- View/download PDF
150. D614G Spike Mutation Increases SARS CoV-2 Susceptibility to Neutralization.
- Author
-
Weissman D, Alameh MG, de Silva T, Collini P, Hornsby H, Brown R, LaBranche CC, Edwards RJ, Sutherland L, Santra S, Mansouri K, Gobeil S, McDanal C, Pardi N, Hengartner N, Lin PJC, Tam Y, Shaw PA, Lewis MG, Boesler C, Şahin U, Acharya P, Haynes BF, Korber B, and Montefiori DC
- Subjects
- Adolescent, Adult, Animals, Antibodies, Monoclonal immunology, Binding Sites, COVID-19 immunology, COVID-19 Vaccines immunology, Female, HEK293 Cells, Humans, Immunization, Passive methods, Macaca mulatta, Male, Mice, Inbred BALB C, Middle Aged, Neutralization Tests, SARS-CoV-2 pathogenicity, Spike Glycoprotein, Coronavirus chemistry, Young Adult, COVID-19 Serotherapy, Mice, COVID-19 therapy, Mutation, SARS-CoV-2 genetics, Spike Glycoprotein, Coronavirus genetics, Spike Glycoprotein, Coronavirus immunology
- Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike protein acquired a D614G mutation early in the pandemic that confers greater infectivity and is now the globally dominant form. To determine whether D614G might also mediate neutralization escape that could compromise vaccine efficacy, sera from spike-immunized mice, nonhuman primates, and humans were evaluated for neutralization of pseudoviruses bearing either D614 or G614 spike. In all cases, the G614 pseudovirus was moderately more susceptible to neutralization. The G614 pseudovirus also was more susceptible to neutralization by receptor-binding domain (RBD) monoclonal antibodies and convalescent sera from people infected with either form of the virus. Negative stain electron microscopy revealed a higher percentage of the 1-RBD "up" conformation in the G614 spike, suggesting increased epitope exposure as a mechanism of enhanced vulnerability to neutralization. Based on these findings, the D614G mutation is not expected to be an obstacle for current vaccine development., Competing Interests: Declaration of Interests In accordance with the University of Pennsylvania policies and procedures and our ethical obligations as researchers, we report that N.P. and D.W. are named on patents that describe the use of nucleoside-modified mRNA as a platform to deliver therapeutic proteins and vaccines. We have disclosed those interests fully to the University of Pennsylvania, and we have in place an approved plan for managing any potential conflicts arising from licensing of our patents., (Copyright © 2020. Published by Elsevier Inc.)
- Published
- 2021
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.