61 results on '"Borducchi EN"'
Search Results
2. 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
3. Decline of Mpox Antibody Responses After Modified Vaccinia Ankara-Bavarian Nordic Vaccination.
- Author
-
Collier AY, McMahan K, Jacob-Dolan C, Liu J, Borducchi EN, Moss B, and Barouch DH
- Published
- 2024
- Full Text
- View/download PDF
4. Rapid Decline of Mpox Antibody Responses Following MVA-BN Vaccination.
- Author
-
Collier AY, McMahan K, Jacob-Dolan C, Liu J, Borducchi EN, Moss B, and Barouch DH
- Abstract
The replication-incompetent modified vaccinia Ankara-Bavarian Nordic vaccine (MVA-BN; Jynneos) was deployed during the 2022 clade IIb mpox outbreak. On August 14, 2024, the World Health Organization declared the mpox clade Ib outbreak in the Democratic Republic of the Congo a public health emergency of international concern, which has raised the question about the durability of vaccine immunity after MVA-BN vaccination. In this study, we show that the MVA-BN vaccine generated mpox serum antibody responses that largely waned after 6-12 months., Competing Interests: Conflicts of Interest The authors report no conflicts of interest.
- Published
- 2024
- Full Text
- View/download PDF
5. Ad26.M.Env ZIKV vaccine protects pregnant rhesus macaques and fetuses against Zika virus infection.
- Author
-
Martinot AJ, Cox F, Abbink P, Hecht JL, Bronson R, Borducchi EN, Rinaldi WJ, Ferguson MJ, De La Barrera RA, Zahn R, van der Fits L, and Barouch DH
- Abstract
At the start of the Zika virus (ZIKV) epidemic in 2015, ZIKV spread across South and Central America, and reached parts of the southern United States placing pregnant women at risk for fetal microcephaly, fetal loss, and other adverse pregnancy outcomes associated with congenital ZIKA syndrome (CZS). For this reason, testing of a safe and efficacious ZIKV vaccine remains a global health priority. Here we report that a single immunization with Ad26.M.Env ZIKV vaccine, when administered prior to conception, fully protects pregnant rhesus macaques from ZIKV viral RNA in blood and tissues with no adverse effects in dams and fetuses. Furthermore, vaccination prevents ZIKV distribution to fetal tissues including the brain. ZIKV associated neuropathology was absent in offspring of Ad26.M.Env vaccinated dams, although pathology was limited in fetuses from non-immunized, challenged dams. Vaccine efficacy is associated with induction of ZIKV neutralizing antibodies in pregnant rhesus macaques. These data suggest the feasibility of vaccine prevention of CZS in humans., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
6. Immunogenicity of 2 therapeutic mosaic HIV-1 vaccine strategies in individuals with HIV-1 on antiretroviral therapy.
- Author
-
Julg B, Stephenson KE, Tomaka F, Walsh SR, Sabrina Tan C, Lavreys L, Sarnecki M, Ansel JL, Kanjilal DG, Jaegle K, Speidel T, Nkolola JP, Borducchi EN, Braams E, Pattacini L, Burgess E, Ilan S, Bartsch Y, Yanosick KE, Seaman MS, Stieh DJ, van Duijn J, Willems W, Robb ML, Michael NL, Walker BD, Pau MG, Schuitemaker H, and Barouch DH
- Abstract
Mosaic HIV-1 vaccines have been shown to elicit robust humoral and cellular immune responses in people living with HIV-1 (PLWH), that had started antiretroviral therapy (ART) during acute infection. We evaluated the safety and immunogenicity of 2 mosaic vaccine regimens in virologically suppressed individuals that had initiated ART during the chronic phase of infection, exemplifying the majority of PLWH. In this double-blind, placebo-controlled phase 1 trial (IPCAVD013/HTX1002) 25 ART-suppressed PLWH were randomized to receive Ad26.Mos4.HIV/MVA-Mosaic (Ad26/MVA) (n = 10) or Ad26.Mos4.HIV/Ad26.Mos4.HIV plus adjuvanted gp140 protein (Ad26/Ad26+gp140) (n = 9) or placebo (n = 6). Primary endpoints included safety and tolerability and secondary endpoints included HIV-specific binding and neutralizing antibody titers and HIV-specific T cell responses. Both vaccine regimens were well tolerated with pain/tenderness at the injection site and fatigue, myalgia/chills and headache as the most commonly reported solicited local and grade 3 systemic adverse events, respectively. In the Ad26/Ad26+gp140 group, Env-specific IFN-γ T cell responses showed a median 12-fold increase while responses to Gag and Pol increased 1.8 and 2.4-fold, respectively. The breadth of T cell responses to individual peptide subpools increased from 11.0 pre-vaccination to 26.0 in the Ad26/Ad26+gp140 group and from 10.0 to 14.5 in the Ad26/MVA group. Ad26/Ad26+gp140 vaccination increased binding antibody titers against vaccine-matched clade C Env 5.5-fold as well as augmented neutralizing antibody titers against Clade C pseudovirus by 7.2-fold. Both vaccine regimens were immunogenic, while the addition of the protein boost resulted in additional T cell and augmented binding and neutralizing antibody titers. These data suggest that the Ad26/Ad26+gp140 regimen should be tested further., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
7. Immunogenicity of BA.5 Bivalent mRNA Vaccine Boosters.
- Author
-
Collier AY, Miller J, Hachmann NP, McMahan K, Liu J, Bondzie EA, Gallup L, Rowe M, Schonberg E, Thai S, Barrett J, Borducchi EN, Bouffard E, Jacob-Dolan C, Mazurek CR, Mutoni A, Powers O, Sciacca M, Surve N, VanWyk H, Wu C, and Barouch DH
- Subjects
- Humans, Antibodies, Neutralizing, Antibodies, Viral, Vaccines, Combined immunology, Vaccines, Combined therapeutic use, Vaccines, Synthetic immunology, Vaccines, Synthetic therapeutic use, mRNA Vaccines, Immunogenicity, Vaccine immunology, COVID-19 genetics, COVID-19 immunology, COVID-19 prevention & control, COVID-19 virology
- Published
- 2023
- Full Text
- View/download PDF
8. Immunogenicity of the BA.5 Bivalent mRNA Vaccine Boosters.
- Author
-
Collier AY, Miller J, Hachmann NP, McMahan K, Liu J, Apraku Bondzie E, Gallup L, Rowe M, Schonberg E, Thai S, Barrett J, Borducchi EN, Bouffard E, Jacob-Dolan C, Mazurek CR, Mutoni A, Powers O, Sciacca M, Surve N, VanWyk H, Wu C, and Barouch DH
- Abstract
Waning immunity following mRNA vaccination and the emergence of SARS-CoV-2 variants has led to reduced mRNA vaccine efficacy against both symptomatic infection and severe disease. Bivalent mRNA boosters expressing the Omicron BA.5 and ancestral WA1/2020 Spike proteins have been developed and approved, because BA.5 is currently the dominant SARS-CoV-2 variant and substantially evades neutralizing antibodies (NAbs). Our data show that BA.5 NAb titers were comparable following monovalent and bivalent mRNA boosters.
- Published
- 2022
- Full Text
- View/download PDF
9. Therapeutic efficacy of combined active and passive immunization in ART-suppressed, SHIV-infected rhesus macaques.
- Author
-
Walker-Sperling VEK, Mercado NB, Chandrashekar A, Borducchi EN, Liu J, Nkolola JP, Lewis M, Murry JP, Yang Y, Geleziunas R, Robb ML, Michael NL, Pau MG, Wegmann F, Schuitemaker H, Fray EJ, Kumar MR, Siliciano JD, Siliciano RF, and Barouch DH
- Subjects
- Animals, Immunization, Passive, Macaca mulatta, Toll-Like Receptor 7, Viral Load, HIV Infections drug therapy, HIV-1, Simian Acquired Immunodeficiency Syndrome, Simian Immunodeficiency Virus
- Abstract
The latent viral reservoir is the critical barrier for developing an HIV-1 cure. Previous studies have shown that therapeutic vaccination or broadly neutralizing antibody (bNAb) administration, together with a Toll-like receptor 7 (TLR7) agonist, enhanced virologic control or delayed viral rebound, respectively, following discontinuation of antiretroviral therapy (ART) in SIV- or SHIV-infected rhesus macaques. Here we show that the combination of active and passive immunization with vesatolimod may lead to higher rates of post-ART virologic control compared to either approach alone. Therapeutic Ad26/MVA vaccination and PGT121 administration together with TLR7 stimulation with vesatolimod resulted in 70% post-ART virologic control in SHIV-SF162P3-infected rhesus macaques. These data suggest the potential of combining active and passive immunization targeting different immunologic mechanisms as an HIV-1 cure strategy., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
10. 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
11. Therapeutic efficacy of an Ad26/MVA vaccine with SIV gp140 protein and vesatolimod in ART-suppressed rhesus macaques.
- Author
-
Ventura JD, Nkolola JP, Chandrashekar A, Borducchi EN, Liu J, Mercado NB, Hope DL, Giffin VM, McMahan K, Geleziunas R, Murry JP, Yang Y, Lewis MG, Pau MG, Wegmann F, Schuitemaker H, Fray EJ, Kumar MR, Siliciano JD, Siliciano RF, Robb ML, Michael NL, and Barouch DH
- Abstract
Developing an intervention that results in virologic control following discontinuation of antiretroviral therapy (ART) is a major objective of HIV-1 cure research. In this study, we investigated the therapeutic efficacy of a vaccine consisting of adenovirus serotype 26 (Ad26) and modified vaccinia Ankara (MVA) with or without an SIV Envelope (Env) gp140 protein with alum adjuvant in combination with the TLR7 agonist vesatolimod (GS-9620) in 36 ART-suppressed, SIVmac251-infected rhesus macaques. Ad26/MVA therapeutic vaccination led to robust humoral and cellular immune responses, and the Env protein boost increased antibody responses. Following discontinuation of ART, virologic control was observed in 5/12 animals in each vaccine group, compared with 0/12 animals in the sham control group. These data demonstrate therapeutic efficacy of Ad26/MVA vaccination with vesatolimod but no clear additional benefit of adding an Env protein boost. SIV-specific cellular immune responses correlated with virologic control. Our findings show partial efficacy of therapeutic vaccination following ART discontinuation in SIV-infected rhesus macaques., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
12. Defining the determinants of protection against SARS-CoV-2 infection and viral control in a dose-down Ad26.CoV2.S vaccine study in nonhuman primates.
- Author
-
Zhu DY, Gorman MJ, Yuan D, Yu J, Mercado NB, McMahan K, Borducchi EN, Lifton M, Liu J, Nampanya F, Patel S, Peter L, Tostanoski LH, Pessaint L, Van Ry A, Finneyfrock B, Velasco J, Teow E, Brown R, Cook A, Andersen H, Lewis MG, Lauffenburger DA, Barouch DH, and Alter G
- Subjects
- Ad26COVS1, Animals, Antibodies, Neutralizing, Antibodies, Viral, COVID-19 Vaccines, Humans, Primates, Receptors, Fc, SARS-CoV-2, Spike Glycoprotein, Coronavirus, COVID-19 prevention & control
- Abstract
Despite the rapid creation of Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) vaccines, the precise correlates of immunity against severe Coronavirus Disease 2019 (COVID-19) are still unknown. Neutralizing antibodies represent a robust surrogate of protection in early Phase III studies, but vaccines provide protection prior to the evolution of neutralization, vaccines provide protection against variants that evade neutralization, and vaccines continue to provide protection against disease severity in the setting of waning neutralizing titers. Thus, in this study, using an Ad26.CoV2.S dose-down approach in nonhuman primates (NHPs), the role of neutralization, Fc effector function, and T-cell immunity were collectively probed against infection as well as against viral control. While dosing-down minimally impacted neutralizing and binding antibody titers, Fc receptor binding and functional antibody levels were induced in a highly dose-dependent manner. Neutralizing antibody and Fc receptor binding titers, but minimally T cells, were linked to the prevention of transmission. Conversely, Fc receptor binding/function and T cells were linked to antiviral control, with a minimal role for neutralization. These data point to dichotomous roles of neutralization and T-cell function in protection against transmission and disease severity and a continuous role for Fc effector function as a correlate of immunity key to halting and controlling SARS-CoV-2 and emerging variants., Competing Interests: I have read the journal’s policy and the authors of this manuscript have the following competing interests: GA is the founder of SeromYx Systems Inc. D.H.B. is a co-inventor on provisional vaccine patent (63/121,482; 63/133,969; 63/135,182).
- Published
- 2022
- Full Text
- View/download PDF
13. HIV envelope antibodies and TLR7 agonist partially prevent viral rebound in chronically SHIV-infected monkeys.
- Author
-
Moldt B, Chandrashekar A, Borducchi EN, Nkolola JP, Stephenson H, Nagel M, Hung M, Goldsmith J, Pace CS, Carr B, Thomsen ND, Blair WS, Geleziunas R, and Barouch DH
- Subjects
- Animals, Anti-Retroviral Agents pharmacology, Anti-Retroviral Agents therapeutic use, Broadly Neutralizing Antibodies, HIV Antibodies therapeutic use, Immunoglobulin G, Macaca mulatta, Toll-Like Receptor 7 agonists, Viral Load, HIV Infections, HIV-1, Simian Acquired Immunodeficiency Syndrome, Simian Immunodeficiency Virus
- Abstract
A key challenge for the development of a cure to HIV-1 infection is the persistent viral reservoir established during early infection. Previous studies using Toll-like receptor 7 (TLR7) agonists and broadly neutralizing antibodies (bNAbs) have shown delay or prevention of viral rebound following antiretroviral therapy (ART) discontinuation in simian-human immunodeficiency virus (SHIV)-infected rhesus macaques. In these prior studies, ART was initiated early during acute infection, which limited the size and diversity of the viral reservoir. Here we evaluated in SHIV-infected rhesus macaques that did not initiate ART until 1 year into chronic infection whether the TLR7 agonist vesatolimod in combination with the bNAb PGT121, formatted either as a human IgG1, an effector enhanced IgG1, or an anti-CD3 bispecific antibody, would delay or prevent viral rebound following ART discontinuation. We found that all 3 antibody formats in combination with vesatolimod were able to prevent viral rebound following ART discontinuation in a subset of animals. These data indicate that a TLR7 agonist combined with antibodies may be a promising strategy to achieve long-term ART-free HIV remission in humans., Competing Interests: I have read the journal’s policy and the authors of this manuscript have the following competing interests: B.M., H.S., M.N., M.H., J.G., C.P., B.C., N.D.T., W.S.B., and R.G. are employees of Gilead Sciences. R.G. is a co-inventor on U.S. Patent No. 11,116,774, Modulators of Toll-like receptors for the treatment of HIV. Vesatolimod is currently in clinical development at Gilead. All other authors declare no competing interests.
- Published
- 2022
- Full Text
- View/download PDF
14. 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
15. Immunogenicity of Ad26.COV2.S vaccine against SARS-CoV-2 variants in humans.
- Author
-
Alter G, Yu J, Liu J, Chandrashekar A, Borducchi EN, Tostanoski LH, McMahan K, Jacob-Dolan C, Martinez DR, Chang A, Anioke T, Lifton M, Nkolola J, Stephenson KE, Atyeo C, Shin S, Fields P, Kaplan I, Robins H, Amanat F, Krammer F, Baric RS, Le Gars M, Sadoff J, de Groot AM, Heerwegh D, Struyf F, Douoguih M, van Hoof J, Schuitemaker H, and Barouch DH
- Subjects
- Ad26COVS1, Adolescent, Adult, Antibodies, Neutralizing immunology, Antibodies, Viral immunology, COVID-19 prevention & control, COVID-19 Vaccines administration & dosage, Humans, Immunity, Cellular, Immunity, Humoral, Middle Aged, SARS-CoV-2 genetics, Spike Glycoprotein, Coronavirus chemistry, Spike Glycoprotein, Coronavirus genetics, Spike Glycoprotein, Coronavirus immunology, Young Adult, COVID-19 immunology, COVID-19 virology, COVID-19 Vaccines immunology, SARS-CoV-2 immunology
- Abstract
The Ad26.COV2.S vaccine
1-3 has demonstrated clinical efficacy against symptomatic COVID-19, including against the B.1.351 variant that is partially resistant to neutralizing antibodies1 . However, the immunogenicity of this vaccine in humans against SARS-CoV-2 variants of concern remains unclear. Here we report humoral and cellular immune responses from 20 Ad26.COV2.S vaccinated individuals from the COV1001 phase I-IIa clinical trial2 against the original SARS-CoV-2 strain WA1/2020 as well as against the B.1.1.7, CAL.20C, P.1 and B.1.351 variants of concern. Ad26.COV2.S induced median pseudovirus neutralizing antibody titres that were 5.0-fold and 3.3-fold lower against the B.1.351 and P.1 variants, respectively, as compared with WA1/2020 on day 71 after vaccination. Median binding antibody titres were 2.9-fold and 2.7-fold lower against the B.1.351 and P.1 variants, respectively, as compared with WA1/2020. Antibody-dependent cellular phagocytosis, complement deposition and natural killer cell activation responses were largely preserved against the B.1.351 variant. CD8 and CD4 T cell responses, including central and effector memory responses, were comparable among the WA1/2020, B.1.1.7, B.1.351, P.1 and CAL.20C variants. These data show that neutralizing antibody responses induced by Ad26.COV2.S were reduced against the B.1.351 and P.1 variants, but functional non-neutralizing antibody responses and T cell responses were largely preserved against SARS-CoV-2 variants. These findings have implications for vaccine protection against SARS-CoV-2 variants of concern., (© 2021. The Author(s).)- Published
- 2021
- Full Text
- View/download PDF
16. Low-dose Ad26.COV2.S protection against SARS-CoV-2 challenge in rhesus macaques.
- Author
-
He X, Chandrashekar A, Zahn R, Wegmann F, Yu J, Mercado NB, McMahan K, Martinot AJ, Piedra-Mora C, Beecy S, Ducat S, Chamanza R, Huber SR, van Heerden M, van der Fits L, Borducchi EN, Lifton M, Liu J, Nampanya F, Patel S, Peter L, Tostanoski LH, Pessaint L, Van Ry A, Finneyfrock B, Velasco J, Teow E, Brown R, Cook A, Andersen H, Lewis MG, Schuitemaker H, and Barouch DH
- Subjects
- Animals, Antibodies, Neutralizing immunology, Antibodies, Viral immunology, B-Lymphocytes immunology, Female, Immunogenicity, Vaccine immunology, Immunologic Memory immunology, Macaca mulatta, Male, Spike Glycoprotein, Coronavirus immunology, Vaccination methods, Adenoviridae immunology, COVID-19 immunology, COVID-19 Vaccines immunology, SARS-CoV-2 immunology, Viral Vaccines immunology
- Abstract
We previously reported that a single immunization with an adenovirus serotype 26 (Ad26)-vector-based vaccine expressing an optimized SARS-CoV-2 spike (Ad26.COV2.S) protected rhesus macaques against SARS-CoV-2 challenge. To evaluate reduced doses of Ad26.COV2.S, 30 rhesus macaques were immunized once with 1 × 10
11 , 5 × 1010 , 1.125 × 1010 , or 2 × 109 viral particles (vp) Ad26.COV2.S or sham and were challenged with SARS-CoV-2. Vaccine doses as low as 2 × 109 vp provided robust protection in bronchoalveolar lavage, whereas doses of 1.125 × 1010 vp were required for protection in nasal swabs. Activated memory B cells and binding or neutralizing antibody titers following vaccination correlated with protective efficacy. At suboptimal vaccine doses, viral breakthrough was observed but did not show enhancement of disease. These data demonstrate that a single immunization with relatively low dose of Ad26.COV2.S effectively protected against SARS-CoV-2 challenge in rhesus macaques, although a higher vaccine dose may be required for protection in the upper respiratory tract., Competing Interests: Declaration of interests D.H.B., R.Z., F.W., and H.S are co-inventors on provisional vaccine patents (63/121,482; 63/133,969; 63/135,182). R.Z., F.W., S.R.H., M.v.H., L.v.d.F., and H.S. are employees of Janssen Vaccines & Prevention BV and may hold stock in Johnson & Johnson., (Copyright © 2021 Elsevier Inc. All rights reserved.)- Published
- 2021
- Full Text
- View/download PDF
17. Immunogenicity of COVID-19 mRNA Vaccines in Pregnant and Lactating Women.
- Author
-
Collier AY, McMahan K, Yu J, Tostanoski LH, Aguayo R, Ansel J, Chandrashekar A, Patel S, Apraku Bondzie E, Sellers D, Barrett J, Sanborn O, Wan H, Chang A, Anioke T, Nkolola J, Bradshaw C, Jacob-Dolan C, Feldman J, Gebre M, Borducchi EN, Liu J, Schmidt AG, Suscovich T, Linde C, Alter G, Hacker MR, and Barouch DH
- Subjects
- 2019-nCoV Vaccine mRNA-1273, Adult, Antibodies, Neutralizing blood, BNT162 Vaccine, Cross Reactions immunology, Female, Humans, Immunoassay, Lactation, Leukocytes, Mononuclear physiology, Middle Aged, Pregnancy immunology, Prospective Studies, T-Lymphocytes immunology, Vaccines, Synthetic immunology, Young Adult, mRNA Vaccines, Antibodies, Viral blood, COVID-19 immunology, COVID-19 Vaccines immunology, Fetal Blood immunology, Immunogenicity, Vaccine, Milk, Human immunology, SARS-CoV-2 immunology
- Abstract
Importance: Pregnant women are at increased risk of morbidity and mortality from COVID-19 but have been excluded from the phase 3 COVID-19 vaccine trials. Data on vaccine safety and immunogenicity in these populations are therefore limited., Objective: To evaluate the immunogenicity of COVID-19 messenger RNA (mRNA) vaccines in pregnant and lactating women, including against emerging SARS-CoV-2 variants of concern., Design, Setting, and Participants: An exploratory, descriptive, prospective cohort study enrolled 103 women who received a COVID-19 vaccine from December 2020 through March 2021 and 28 women who had confirmed SARS-CoV-2 infection from April 2020 through March 2021 (the last follow-up date was March 26, 2021). This study enrolled 30 pregnant, 16 lactating, and 57 neither pregnant nor lactating women who received either the mRNA-1273 (Moderna) or BNT162b2 (Pfizer-BioNTech) COVID-19 vaccines and 22 pregnant and 6 nonpregnant unvaccinated women with SARS-CoV-2 infection., Main Outcomes and Measures: SARS-CoV-2 receptor binding domain binding, neutralizing, and functional nonneutralizing antibody responses from pregnant, lactating, and nonpregnant women were assessed following vaccination. Spike-specific T-cell responses were evaluated using IFN-γ enzyme-linked immunospot and multiparameter intracellular cytokine-staining assays. Humoral and cellular immune responses were determined against the original SARS-CoV-2 USA-WA1/2020 strain as well as against the B.1.1.7 and B.1.351 variants., Results: This study enrolled 103 women aged 18 to 45 years (66% non-Hispanic White) who received a COVID-19 mRNA vaccine. After the second vaccine dose, fever was reported in 4 pregnant women (14%; SD, 6%), 7 lactating women (44%; SD, 12%), and 27 nonpregnant women (52%; SD, 7%). Binding, neutralizing, and functional nonneutralizing antibody responses as well as CD4 and CD8 T-cell responses were present in pregnant, lactating, and nonpregnant women following vaccination. Binding and neutralizing antibodies were also observed in infant cord blood and breast milk. Binding and neutralizing antibody titers against the SARS-CoV-2 B.1.1.7 and B.1.351 variants of concern were reduced, but T-cell responses were preserved against viral variants., Conclusion and Relevance: In this exploratory analysis of a convenience sample, receipt of a COVID-19 mRNA vaccine was immunogenic in pregnant women, and vaccine-elicited antibodies were transported to infant cord blood and breast milk. Pregnant and nonpregnant women who were vaccinated developed cross-reactive antibody responses and T-cell responses against SARS-CoV-2 variants of concern.
- Published
- 2021
- Full Text
- View/download PDF
18. Immunogenicity of the Ad26.COV2.S Vaccine for COVID-19.
- Author
-
Stephenson KE, Le Gars M, Sadoff J, de Groot AM, Heerwegh D, Truyers C, Atyeo C, Loos C, Chandrashekar A, McMahan K, Tostanoski LH, Yu J, Gebre MS, Jacob-Dolan C, Li Z, Patel S, Peter L, Liu J, Borducchi EN, Nkolola JP, Souza M, Tan CS, Zash R, Julg B, Nathavitharana RR, Shapiro RL, Azim AA, Alonso CD, Jaegle K, Ansel JL, Kanjilal DG, Guiney CJ, Bradshaw C, Tyler A, Makoni T, Yanosick KE, Seaman MS, Lauffenburger DA, Alter G, Struyf F, Douoguih M, Van Hoof J, Schuitemaker H, and Barouch DH
- Subjects
- Adult, COVID-19 immunology, COVID-19 Vaccines administration & dosage, Double-Blind Method, Female, Humans, Immunity, Humoral, Male, Middle Aged, Vaccine Potency, Young Adult, Antibodies, Neutralizing blood, Antibodies, Viral blood, COVID-19 prevention & control, COVID-19 Vaccines immunology, Immunity, Cellular, Immunogenicity, Vaccine
- Abstract
Importance: Control of the global COVID-19 pandemic will require the development and deployment of safe and effective vaccines., Objective: To evaluate the immunogenicity of the Ad26.COV2.S vaccine (Janssen/Johnson & Johnson) in humans, including the kinetics, magnitude, and phenotype of SARS-CoV-2 spike-specific humoral and cellular immune responses., Design, Setting, and Participants: Twenty-five participants were enrolled from July 29, 2020, to August 7, 2020, and the follow-up for this day 71 interim analysis was completed on October 3, 2020; follow-up to assess durability will continue for 2 years. This study was conducted at a single clinical site in Boston, Massachusetts, as part of a randomized, double-blind, placebo-controlled phase 1 clinical trial of Ad26.COV2.S., Interventions: Participants were randomized to receive 1 or 2 intramuscular injections with 5 × 1010 viral particles or 1 × 1011 viral particles of Ad26.COV2.S vaccine or placebo administered on day 1 and day 57 (5 participants in each group)., Main Outcomes and Measures: Humoral immune responses included binding and neutralizing antibody responses at multiple time points following immunization. Cellular immune responses included immunospot-based and intracellular cytokine staining assays to measure T-cell responses., Results: Twenty-five participants were randomized (median age, 42; age range, 22-52; 52% women, 44% male, 4% undifferentiated), and all completed the trial through the day 71 interim end point. Binding and neutralizing antibodies emerged rapidly by day 8 after initial immunization in 90% and 25% of vaccine recipients, respectively. By day 57, binding and neutralizing antibodies were detected in 100% of vaccine recipients after a single immunization. On day 71, the geometric mean titers of spike-specific binding antibodies were 2432 to 5729 and the geometric mean titers of neutralizing antibodies were 242 to 449 in the vaccinated groups. A variety of antibody subclasses, Fc receptor binding properties, and antiviral functions were induced. CD4+ and CD8+ T-cell responses were induced., Conclusion and Relevance: In this phase 1 study, a single immunization with Ad26.COV2.S induced rapid binding and neutralization antibody responses as well as cellular immune responses. Two phase 3 clinical trials are currently underway to determine the efficacy of the Ad26.COV2.S vaccine., Trial Registration: ClinicalTrials.gov Identifier: NCT04436276.
- Published
- 2021
- Full Text
- View/download PDF
19. Persistence of viral RNA in lymph nodes in ART-suppressed SIV/SHIV-infected Rhesus Macaques.
- Author
-
Cadena AM, Ventura JD, Abbink P, Borducchi EN, Tuyishime H, Mercado NB, Walker-Sperling V, Siamatu M, Liu PT, Chandrashekar A, Nkolola JP, McMahan K, Kordana N, Hamza V, Bondzie EA, Fray E, Kumar M, Fischinger S, Shin SA, Lewis MG, Siliciano RF, Alter G, and Barouch DH
- Subjects
- Animals, CD8-Positive T-Lymphocytes, DNA, Viral, Disease Models, Animal, Female, HIV Infections drug therapy, HIV-1 genetics, Lymph Nodes immunology, Lymphoid Tissue virology, Macaca mulatta, Phylogeny, Simian Acquired Immunodeficiency Syndrome drug therapy, Simian Acquired Immunodeficiency Syndrome immunology, Simian Immunodeficiency Virus drug effects, Simian Immunodeficiency Virus genetics, Viral Load, Virus Replication, Anti-Retroviral Agents therapeutic use, HIV Infections virology, Lymph Nodes virology, RNA, Viral genetics, Simian Acquired Immunodeficiency Syndrome virology, Simian Immunodeficiency Virus immunology
- Abstract
The establishment of a long-lived viral reservoir is the key obstacle for achieving an HIV-1 cure. However, the anatomic, virologic, and immunologic features of the viral reservoir in tissues during antiretroviral therapy (ART) remain poorly understood. Here we present a comprehensive necroscopic analysis of the SIV/SHIV viral reservoir in multiple lymphoid and non-lymphoid tissues from SIV/SHIV-infected rhesus macaques suppressed with ART for one year. Viral DNA is observed broadly in multiple tissues and is comparable in animals that had initiated ART at week 1 or week 52 of infection. In contrast, viral RNA is restricted primarily to lymph nodes. Ongoing viral RNA transcription is not the result of unsuppressed viral replication, as single-genome amplification and subsequent phylogenetic analysis do not show evidence of viral evolution. Gag-specific CD8+ T cell responses are predominantly observed in secondary lymphoid organs in animals chronically infected prior to ART and these responses are dominated by CD69+ populations. Overall, we observe that the viral reservoir in rhesus macaques is widely distributed across multiple tissue sites and that lymphoid tissues act as a site of persistent viral RNA transcription under conditions of long-term ART suppression.
- Published
- 2021
- Full Text
- View/download PDF
20. Origin of rebound virus in chronically SIV-infected Rhesus monkeys following treatment discontinuation.
- Author
-
Liu PT, Keele BF, Abbink P, Mercado NB, Liu J, Bondzie EA, Chandrashekar A, Borducchi EN, Hesselgesser J, Mish M, Chin G, Bekerman E, Geleziunas R, and Barouch DH
- Subjects
- Animals, CD4-Positive T-Lymphocytes virology, Female, HIV Infections drug therapy, HIV Infections virology, HIV-1 drug effects, HIV-1 genetics, HIV-1 physiology, Humans, Macaca mulatta, Male, Patient Dropouts, Simian Acquired Immunodeficiency Syndrome virology, Simian Immunodeficiency Virus drug effects, Simian Immunodeficiency Virus genetics, Viral Load drug effects, Virus Latency drug effects, Virus Replication drug effects, Anti-Retroviral Agents administration & dosage, Simian Acquired Immunodeficiency Syndrome drug therapy, Simian Immunodeficiency Virus physiology
- Abstract
Viral rebound following antiretroviral therapy (ART) discontinuation in HIV-1-infected individuals is believed to originate from a small pool of CD4+ T cells harboring replication-competent provirus. However, the origin and nature of the rebound virus has remained unclear. Recent studies have suggested that rebound virus does not originate directly from individual latent proviruses but rather from recombination events involving multiple proviruses. Here we evaluate the origin of rebound virus in 16 ART-suppressed, chronically SIV-infected rhesus monkeys following ART discontinuation. We sequence viral RNA and viral DNA in these animals prior to ART initiation, during ART suppression, and following viral rebound, and we compare rebound viral RNA after ART discontinuation with near full-length viral DNA from peripheral blood and lymph node mononuclear cells (PBMC and LNMC) during ART suppression. Sequences of initial rebound viruses closely match viral DNA sequences in PBMC and LNMC during ART suppression. Recombinant viruses are rare in the initial rebound virus populations but arise quickly within 2-4 weeks after viral rebound. These data suggest that intact proviral DNA in PBMC and LNMC during ART suppression is likely the direct origin of viral rebound in chronically SIV-infected rhesus monkeys following ART discontinuation.
- Published
- 2020
- Full Text
- View/download PDF
21. Passive Transfer of Vaccine-Elicited Antibodies Protects against SIV in Rhesus Macaques.
- Author
-
Alter G, Yu WH, Chandrashekar A, Borducchi EN, Ghneim K, Sharma A, Nedellec R, McKenney KR, Linde C, Broge T, Suscovich TJ, Linnekin T, Abbink P, Mercado NB, Nkolola JP, McMahan K, Bondzie EA, Hamza V, Peter L, Kordana N, Mahrokhian S, Seaman MS, Li W, Lewis MG, Lauffenburger DA, Hangartner L, Sekaly RP, and Barouch DH
- Subjects
- AIDS Vaccines immunology, Animals, Antibodies, Neutralizing immunology, Antibodies, Viral immunology, Antibody Formation immunology, Gene Products, env immunology, Gene Products, gag immunology, Gene Products, pol immunology, HIV-1 immunology, Immunoglobulin G immunology, Macaca mulatta immunology, Simian Acquired Immunodeficiency Syndrome immunology, Immunization, Passive methods, SAIDS Vaccines immunology, Simian Immunodeficiency Virus immunology
- Abstract
Several HIV-1 and SIV vaccine candidates have shown partial protection against viral challenges in rhesus macaques. However, the protective efficacy of vaccine-elicited polyclonal antibodies has not previously been demonstrated in adoptive transfer studies in nonhuman primates. In this study, we show that passive transfer of purified antibodies from vaccinated macaques can protect naive animals against SIVmac251 challenges. We vaccinated 30 rhesus macaques with Ad26-SIV Env/Gag/Pol and SIV Env gp140 protein vaccines and assessed the induction of antibody responses and a putative protective signature. This signature included multiple antibody functions and correlated with upregulation of interferon pathways in vaccinated animals. Adoptive transfer of purified immunoglobulin G (IgG) from the vaccinated animals with the most robust protective signatures provided partial protection against SIVmac251 challenges in naive recipient rhesus macaques. These data demonstrate the protective efficacy of purified vaccine-elicited antiviral antibodies in this model, even in the absence of virus neutralization., Competing Interests: Declaration of Interests D.H.B. is a co-inventor on related HIV-1 vaccine patents. The authors otherwise declare no competing financial interests., (Copyright © 2020 Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
22. Sustained maternal antibody and cellular immune responses in pregnant women infected with Zika virus and mother to infant transfer of Zika-specific antibodies.
- Author
-
Collier AY, Borducchi EN, Chandrashekar A, Moseley E, Peter L, Teodoro NS, Nkolola J, Abbink P, and Barouch DH
- Subjects
- Adult, Cohort Studies, Female, Humans, Immunity, Cellular, Immunity, Humoral, Immunity, Maternally-Acquired, Infectious Disease Transmission, Vertical, Maternal Exposure, Maternal-Fetal Exchange, Pregnancy, Prospective Studies, Zika Virus Infection transmission, Antibodies, Viral metabolism, Zika Virus physiology, Zika Virus Infection immunology
- Abstract
Problem: Evaluation of Zika virus (ZIKV)-specific humoral and cellular immune response in pregnant women exposed to ZIKV., Method of Study: In this observational, prospective cohort study, we recruited pregnant women presenting for prenatal ultrasound for ZIKV exposure at a single academic teaching hospital in Boston, MA from November 2016 to December 2018. We collected blood, urine, and cervicovaginal swabs antepartum, intrapartum, and postpartum; and cord blood and placenta at delivery. We used experimental assays to calculate quantitative viral loads, ZIKV-specific immunoglobulin titers, and ZIKV-specific T-cell responses., Results: We enrolled 22 participants, three of which had serologic-confirmed ZIKV infection. No participants demonstrated sustained ZIKV shedding. ZIKV-specific IgG/IgM antibody was sustained throughout pregnancy and postpartum. ZIKV envelope and capsid-specific T-cell responses were also observed, albeit inconsistent. No newborns in this cohort had congenital Zika syndrome. Infant cord blood of infected mothers exhibited ZIKV-specific IgG, but not IgM antibodies., Conclusion: We detected a robust, prolonged maternal humoral immune response to ZIKV during pregnancy and postpartum. We also demonstrated evidence for efficient transplacental antibody transfer from mother to infant at birth, supporting the importance of neonatal passive immunity to ZIKV. Maternal T-cell responses were less consistent among pregnant women infected with ZIKV., (© 2020 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd.)
- Published
- 2020
- Full Text
- View/download PDF
23. Safety and immunogenicity of a Zika purified inactivated virus vaccine given via standard, accelerated, or shortened schedules: a single-centre, double-blind, sequential-group, randomised, placebo-controlled, phase 1 trial.
- Author
-
Stephenson KE, Tan CS, Walsh SR, Hale A, Ansel JL, Kanjilal DG, Jaegle K, Peter L, Borducchi EN, Nkolola JP, Makoni T, Fogel R, Bradshaw C, Tyler A, Moseley E, Chandrashekar A, Yanosick KE, Seaman MS, Eckels KH, De La Barrera RA, Thompson J, Dawson P, Thomas SJ, Michael NL, Modjarrad K, and Barouch DH
- Subjects
- Adolescent, Adult, Female, Humans, Immunization Schedule, Male, Middle Aged, Vaccines, Inactivated administration & dosage, Vaccines, Inactivated adverse effects, Viral Vaccines administration & dosage, Viral Vaccines adverse effects, Young Adult, Immunogenicity, Vaccine, Vaccines, Inactivated immunology, Viral Vaccines immunology, Zika Virus immunology, Zika Virus Infection prevention & control
- Abstract
Background: The development of an effective vaccine against Zika virus remains a public health priority. A Zika purified inactivated virus (ZPIV) vaccine candidate has been shown to protect animals against Zika virus challenge and to be well tolerated and immunogenic in humans up to 8 weeks of follow-up. We aimed to assess the safety and immunogenicity of ZPIV in humans up to 52 weeks of follow-up when given via standard or accelerated vaccination schedules., Methods: We did a single-centre, double-blind, randomised controlled, phase 1 trial in healthy adults aged 18-50 years with no known history of flavivirus vaccination or infection at Beth Israel Deaconess Medical Center in Boston, MA, USA. Participants were sequentially enrolled into one of three groups: ZPIV given at weeks 0 and 4 (standard regimen), weeks 0 and 2 (accelerated regimen), or week 0 alone (single-dose regimen). Within each group, participants were randomly assigned using a computer-generated randomisation schedule to receive an intramuscular injection of 5 μg ZPIV or saline placebo, in a ratio of 5:1. The sponsor, clinical staff, investigators, participants, and laboratory personnel were masked to treatment assignment. The primary endpoint was safety up to day 364 after final dose administration, and secondary endpoints were proportion of participants with positive humoral immune responses (50% microneutralisation titre [MN
50 ] ≥100) and geometric mean MN50 at observed peak response (ie, the highest neutralising antibody level observed for an individual participant across all timepoints) and week 28. All participants who received at least one dose of ZPIV or placebo were included in the safety population; the analysis of immunogenicity at observed peak included all participants who received at least one dose of ZPIV or placebo and had any adverse events or immunogenicity data after dosing. The week 28 immunogenicity analysis population consisted of all participants who received ZPIV or placebo and had immunogenicity data available at week 28. This trial is registered with ClinicalTrials.gov, NCT02937233., Findings: Between Dec 8, 2016, and May 17, 2017, 12 participants were enrolled into each group and then randomly assigned to vaccine (n=10) or placebo (n=2). There were no serious or grade 3 treatment-related adverse events. The most common reactions among the 30 participants who received the vaccine were injection-site pain (24 [80%]), fatigue (16 [53%]), and headache (14 [46%]). A positive response at observed peak titre was detected in all participants who received ZPIV via the standard regimen, in eight (80%) of ten participants who received ZPIV via the accelerated regimen, and in none of the ten participants who received ZPIV via the single-dose regimen. The geometric mean of all individual participants' observed peak values was 1153·9 (95% CI 455·2-2925·2) in the standard regimen group, 517·7 (142·9-1875·6) in the accelerated regimen group, and 6·3 (3·7-10·8) in the single-dose regimen group. At week 28, a positive response was observed in one (13%) of eight participants who received ZPIV via the standard regimen and in no participant who received ZPIV via the accelerated (n=7) or single-dose (n=10) regimens. The geomteric mean titre (GMT) at this timepoint was 13·9 (95% CI 3·5-55·1) in the standard regimen group and 6·9 (4·0-11·9) in the accelerated regimen group; antibody titres were undetectable at 28 weeks in participants who received ZPIV via the single-dose regimen. For all vaccine schedules, GMTs peaked 2 weeks after the final vaccination and declined to less than 100 by study week 16. There was no difference in observed peak GMTs between the standard 4-week and the accelerated 2-week boosting regimens (p=0·4494)., Interpretation: ZPIV was safe and well tolerated in humans up to 52 weeks of follow-up. ZPIV immunogenicity required two doses and was not durable. Additional studies of ZPIV to optimise dosing schedules are ongoing., Funding: The Henry M Jackson Foundation for the Advancement of Military Medicine., (Copyright © 2020 Elsevier Ltd. All rights reserved.)- Published
- 2020
- Full Text
- View/download PDF
24. Comparison of shortened mosaic HIV-1 vaccine schedules: a randomised, double-blind, placebo-controlled phase 1 trial (IPCAVD010/HPX1002) and a preclinical study in rhesus monkeys (NHP 17-22).
- Author
-
Stephenson KE, Wegmann F, Tomaka F, Walsh SR, Tan CS, Lavreys L, Ansel JL, Kanjilal DG, Jaegle K, Nkolola JP, Peter L, Fogel R, Bradshaw C, Tyler A, Makoni T, Howe L, Quijada D, Chandrashekar A, Bondzie EA, Borducchi EN, Yanosick KE, Hendriks J, Nijs S, Truyers C, Tolboom J, Zahn RC, Seaman MS, Alter G, Stieh DJ, Pau MG, Schuitemaker H, and Barouch DH
- Subjects
- AIDS Vaccines adverse effects, AIDS Vaccines immunology, Adjuvants, Immunologic adverse effects, Adjuvants, Immunologic chemistry, Adult, Animals, Double-Blind Method, Female, HIV Antibodies metabolism, HIV Infections immunology, HIV-1 immunology, Humans, Immunization Schedule, Injections, Intramuscular, Male, Middle Aged, Time Factors, Young Adult, env Gene Products, Human Immunodeficiency Virus adverse effects, env Gene Products, Human Immunodeficiency Virus immunology, AIDS Vaccines administration & dosage, Adjuvants, Immunologic administration & dosage, HIV Infections prevention & control, Macaca mulatta immunology, env Gene Products, Human Immunodeficiency Virus administration & dosage
- Abstract
Background: Current efficacy studies of a mosaic HIV-1 prophylactic vaccine require four vaccination visits over one year, which is a complex regimen that could prove challenging for vaccine delivery at the community level, both for recipients and clinics. In this study, we evaluated the safety, tolerability, and immunogenicity of shorter, simpler regimens of trivalent Ad26.Mos.HIV expressing mosaic HIV-1 Env/Gag/Pol antigens combined with aluminium phosphate-adjuvanted clade C gp140 protein., Methods: We did this randomised, double-blind, placebo-controlled phase 1 trial (IPCAVD010/HPX1002) at Beth Israel Deaconess Medical Center in Boston, MA, USA. We included healthy, HIV-uninfected participants (aged 18-50 years) who were considered at low risk for HIV infection and had not received any vaccines in the 14 days before study commencement. We randomly assigned participants via a computer-generated randomisation schedule and interactive web response system to one of three study groups (1:1:1) testing different regimens of trivalent Ad26.Mos.HIV (5 × 10
10 viral particles per 0·5 mL) combined with 250 μg adjuvanted clade C gp140 protein. They were then assigned to treatment or placebo subgroups (5:1) within each of the three main groups. Participants and investigators were masked to treatment allocation until the end of the follow-up period. Group 1 received Ad26.Mos.HIV alone at weeks 0 and 12 and Ad26.Mos.HIV plus adjuvanted gp140 at weeks 24 and 48. Group 2 received Ad26.Mos.HIV plus adjuvanted gp140 at weeks 0, 12, and 24. Group 3 received Ad26.Mos.HIV alone at week 0 and Ad26.Mos.HIV plus adjuvanted gp140 at weeks 8 and 24. Participants in the control group received 0·5 mL of 0·9% saline. All study interventions were administered intramuscularly. The primary endpoints were Env-specific binding antibody responses at weeks 28, 52, and 72 and safety and tolerability of the vaccine regimens for 28 days after the injection. All participants who received at least one vaccine dose or placebo were included in the safety analysis; immunogenicity was analysed using the per-protocol population. The IPCAVD010/HPX1002 trial is registered with ClinicalTrials.gov, NCT02685020. We also did a parallel preclinical study in rhesus monkeys to test the protective efficacy of the shortened group 3 regimen., Findings: Between March 7, 2016, and Aug 19, 2016, we randomly assigned 36 participants to receive at least one dose of study vaccine or placebo, ten to each vaccine group and two to the corresponding placebo group. 30 (83%) participants completed the full study, and six (17%) discontinued it prematurely because of loss to follow-up, withdrawal of consent, investigator decision, and an unrelated death from a motor vehicle accident. The two shortened regimens elicited comparable antibody titres against autologous clade C Env at peak immunity to the longer, 12-month regimen: geometric mean titre (GMT) 41 007 (95% CI 17 959-93 636) for group 2 and 49 243 (29 346-82 630) for group 3 at week 28 compared with 44 590 (19 345-102 781) for group 1 at week 52). Antibody responses remained increased (GMT >5000) in groups 2 and 3 at week 52 but were highest in group 1 at week 72. Antibody-dependent cellular phagocytosis, Env-specific IgG3, tier 1A neutralising activity, and broad cellular immune responses were detected in all groups. All vaccine regimens were well tolerated. Mild-to-moderate pain or tenderness at the injection site was the most commonly reported solicited local adverse event, reported by 28 vaccine recipients (93%) and two placebo recipients (33%). Grade 3 solicited systemic adverse events were reported by eight (27%) vaccine recipients and no placebo recipients; the most commonly reported grade 3 systemic symptoms were fatigue, myalgia, and chills. The shortened group 3 regimen induced comparable peak immune responses in 30 rhesus monkeys as in humans and resulted in an 83% (95% CI 38·7-95, p=0·004 log-rank test) reduction in per-exposure acquisition risk after six intrarectal challenges with SHIV-SF162P3 at week 54, more than 6 months after final vaccination., Interpretation: Short, 6-month regimens of a mosaic HIV-1 prophylactic vaccine elicited robust HIV-specific immune responses that were similar to responses elicited by a longer, 12-month schedule. Preclinical data showed partial protective efficacy of one of the short vaccine regimens in rhesus monkeys. Further clinical studies are required to test the suitability of the shortened vaccine regimens in humans. Such shortened regimens would be valuable to increase vaccine delivery at the community level, particularly in resource-limited settings., Funding: Ragon Institute (Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard University; Cambridge, MA, USA) and Janssen Vaccines & Prevention (Leiden, Netherlands)., (Copyright © 2020 Elsevier Ltd. All rights reserved.)- Published
- 2020
- Full Text
- View/download PDF
25. Differential Outcomes following Optimization of Simian-Human Immunodeficiency Viruses from Clades AE, B, and C.
- Author
-
Tartaglia LJ, Gupte S, Pastores KC, Trott S, Abbink P, Mercado NB, Li Z, Liu PT, Borducchi EN, Chandrashekar A, Bondzie EA, Hamza V, Kordana N, Mahrokhian S, Lavine CL, Seaman MS, Li H, Shaw GM, and Barouch DH
- Subjects
- Animals, Antibodies, Neutralizing, CD4-Positive T-Lymphocytes, Female, Gene Products, env genetics, HIV Infections virology, HIV-1 genetics, HIV-1 immunology, Humans, Macaca mulatta, Male, Mutation, Sequence Analysis, Simian Acquired Immunodeficiency Syndrome virology, Viral Load, Virus Replication, Simian Acquired Immunodeficiency Syndrome immunology, Simian Immunodeficiency Virus genetics, Simian Immunodeficiency Virus immunology
- Abstract
Simian-human immunodeficiency virus (SHIV) infection of rhesus monkeys is an important preclinical model for human immunodeficiency virus type 1 (HIV-1) vaccines, therapeutics, and cure strategies. SHIVs have been optimized by incorporating HIV-1 Env residue 375 mutations that mimic the bulky or hydrophobic residues typically found in simian immunodeficiency virus (SIV) Env to improve rhesus CD4 binding. We applied this strategy to three SHIV challenge stocks (SHIV-SF162p3, SHIV-AE16, and SHIV-325c) and observed three distinct outcomes. We constructed six Env375 variants (M, H, W, Y, F, and S) for each SHIV, and we performed a pool competition study in rhesus monkeys to define the optimal variant for each SHIV prior to generating large-scale challenge stocks. We identified SHIV-SF162p3S/wild type, SHIV-AE16W, and SHIV-325cH as the optimal variants. SHIV-SF162p3S could not be improved, as it already contained the optimal Env375 residue. SHIV-AE16W exhibited a similar replicative capacity to the parental SHIV-AE16 stock. In contrast, SHIV-325cH demonstrated a 2.6-log higher peak and 1.6-log higher setpoint viral loads than the parental SHIV-325c stock. These data demonstrate the diversity of potential outcomes following Env375 modification in SHIVs. Moreover, the clade C SHIV-325cH challenge stock may prove useful for evaluating prophylactic or therapeutic interventions against clade C HIV-1. IMPORTANCE We sought to enhance the infectivity of three SHIV stocks by optimization of a key residue in human immunodeficiency virus type 1 (HIV-1) Env (Env375). We developed the following three new simian-human immunodeficiency virus (SHIV) stocks: SHIV-SF162p3S/wild type, SHIV-AE16W, and SHIV-325cH. SHIV-SF162p3S could not be optimized, SHIV-AE16W proved comparable to the parental virus, and SHIV-325cH demonstrated markedly enhanced replicative capacity compared with the parental virus., (Copyright © 2020 Tartaglia et al.)
- Published
- 2020
- Full Text
- View/download PDF
26. Adenovirus Vector-Based Vaccines Confer Maternal-Fetal Protection against Zika Virus Challenge in Pregnant IFN-αβR -/- Mice.
- Author
-
Larocca RA, Mendes EA, Abbink P, Peterson RL, Martinot AJ, Iampietro MJ, Kang ZH, Aid M, Kirilova M, Jacob-Dolan C, Tostanoski L, Borducchi EN, De La Barrera RA, and Barouch DH
- Subjects
- Adenoviridae genetics, Adenoviridae immunology, Animals, Antibodies, Neutralizing immunology, Antibodies, Viral blood, Antibodies, Viral immunology, Cell Line, Chlorocebus aethiops, Disease Models, Animal, Female, Male, Mice, Mice, Inbred BALB C, Mice, Inbred C57BL, Mice, Knockout, Pregnancy, Vaccination, Vero Cells, Zika Virus Infection immunology, Antibodies, Neutralizing blood, Immunity, Maternally-Acquired immunology, Receptor, Interferon alpha-beta genetics, Viral Vaccines immunology, Zika Virus immunology, Zika Virus Infection prevention & control
- Abstract
Maternal infection with Zika virus (ZIKV) can lead to microcephaly and other congenital abnormalities of the fetus. Although ZIKV vaccines that prevent or reduce viremia in non-pregnant mice have been described, a maternal vaccine that provides complete fetal protection would be desirable. Here, we show that adenovirus (Ad) vector-based ZIKV vaccines induce potent neutralizing antibodies that confer robust maternal and fetal protection against ZIKV challenge in pregnant, highly susceptible IFN-αβR
-/- mice. Moreover, passive transfer of maternal antibodies from vaccinated dams protected pups against post-natal ZIKV challenge. These data suggest that Ad-based ZIKV vaccines may be able to provide protection in pregnant females against fetal ZIKV transmission in utero as well as in infants against ZIKV infection after birth., (Copyright © 2019 Elsevier Inc. All rights reserved.)- Published
- 2019
- Full Text
- View/download PDF
27. Lack of therapeutic efficacy of an antibody to α 4 β 7 in SIVmac251-infected rhesus macaques.
- Author
-
Abbink P, Mercado NB, Nkolola JP, Peterson RL, Tuyishime H, McMahan K, Moseley ET, Borducchi EN, Chandrashekar A, Bondzie EA, Agarwal A, Belli AJ, Reimann KA, Keele BF, Geleziunas R, Lewis MG, and Barouch DH
- Subjects
- Animals, Anti-Retroviral Agents therapeutic use, Antibodies, Monoclonal administration & dosage, Antibodies, Viral administration & dosage, Codon, Terminator, DNA, Viral blood, HIV Infections therapy, Macaca mulatta, Simian Acquired Immunodeficiency Syndrome blood, Simian Acquired Immunodeficiency Syndrome drug therapy, Simian Immunodeficiency Virus physiology, Viral Load, Viral Regulatory and Accessory Proteins genetics, Viral Regulatory and Accessory Proteins immunology, Antibodies, Monoclonal therapeutic use, Antibodies, Viral therapeutic use, Integrin alpha4 immunology, Integrin beta Chains immunology, Simian Acquired Immunodeficiency Syndrome therapy, Simian Acquired Immunodeficiency Syndrome virology, Simian Immunodeficiency Virus genetics, Simian Immunodeficiency Virus immunology
- Abstract
Sustained virologic control of human immunodeficiency virus type 1 (HIV-1) infection after discontinuation of antiretroviral therapy (ART) is a major goal of the HIV-1 cure field. A recent study reported that administration of an antibody against α
4 β7 induced durable virologic control after ART discontinuation in 100% of rhesus macaques infected with an attenuated strain of simian immunodeficiency virus (SIV) containing a stop codon in nef We performed similar studies in 50 rhesus macaques infected with wild-type, pathogenic SIVmac251. In animals that initiated ART during either acute or chronic infection, anti-α4 β7 antibody infusion had no detectable effect on the viral reservoir or viral rebound after ART discontinuation. These data demonstrate that anti-α4 β7 antibody administration did not provide therapeutic efficacy in the model of pathogenic SIVmac251 infection of rhesus macaques., (Copyright © 2019 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works.)- Published
- 2019
- Full Text
- View/download PDF
28. Publisher Correction: Antibody and TLR7 agonist delay viral rebound in SHIV-infected monkeys.
- Author
-
Borducchi EN, Liu J, Nkolola JP, Cadena AM, Yu WH, Fischinger S, Broge T, Abbink P, Mercado NB, Chandrashekar A, Jetton D, Peter L, McMahan K, Moseley ET, Bekerman E, Hesselgesser J, Li W, Lewis MG, Alter G, Geleziunas R, and Barouch DH
- Abstract
In Fig. 4b of this Article, the x-axis labels 'PGT121' and 'GS-9620' were inadvertently swapped in both graphs. In Fig. 5a, b, 'TLR7' should have been 'GS-9620'. These figures have been corrected online.
- Published
- 2018
- Full Text
- View/download PDF
29. Antibody and TLR7 agonist delay viral rebound in SHIV-infected monkeys.
- Author
-
Borducchi EN, Liu J, Nkolola JP, Cadena AM, Yu WH, Fischinger S, Broge T, Abbink P, Mercado NB, Chandrashekar A, Jetton D, Peter L, McMahan K, Moseley ET, Bekerman E, Hesselgesser J, Li W, Lewis MG, Alter G, Geleziunas R, and Barouch DH
- Subjects
- Adoptive Transfer, Animals, Anti-HIV Agents administration & dosage, Anti-HIV Agents therapeutic use, Antibodies, Neutralizing immunology, CD8 Antigens deficiency, CD8 Antigens immunology, DNA, Viral analysis, Female, HIV Antibodies immunology, HIV-1 genetics, Humans, Immunity, Cellular drug effects, Immunity, Cellular immunology, Immunity, Innate drug effects, Immunity, Innate immunology, Macaca mulatta immunology, Macaca mulatta virology, Male, Pteridines pharmacology, Simian Acquired Immunodeficiency Syndrome drug therapy, Simian Acquired Immunodeficiency Syndrome immunology, Simian Immunodeficiency Virus genetics, Toll-Like Receptor 7 immunology, Viral Load, Antibodies, Viral immunology, HIV-1 drug effects, HIV-1 immunology, Simian Acquired Immunodeficiency Syndrome therapy, Simian Immunodeficiency Virus drug effects, Simian Immunodeficiency Virus immunology, Toll-Like Receptor 7 agonists
- Abstract
The latent viral reservoir is the critical barrier for the development of a cure for HIV-1 infection. Previous studies have shown direct antiviral activity of potent HIV-1 Env-specific broadly neutralizing antibodies (bNAbs) administered when antiretroviral therapy (ART) was discontinued, but it remains unclear whether bNAbs can target the viral reservoir during ART. Here we show that administration of the V3 glycan-dependent bNAb PGT121 together with the Toll-like receptor 7 (TLR7) agonist vesatolimod (GS-9620) during ART delayed viral rebound following discontinuation of ART in simian-human immunodeficiency virus (SHIV)-SF162P3-infected rhesus monkeys in which ART was initiated during early acute infection. Moreover, in the subset of monkeys that were treated with both PGT121 and GS-9620 and that did not show viral rebound after discontinuation of ART, adoptive transfer studies and CD8-depletion studies also did not reveal virus. These data demonstrate the potential of bNAb administration together with innate immune stimulation as a possible strategy for targeting the viral reservoir.
- Published
- 2018
- Full Text
- View/download PDF
30. Evaluation of a mosaic HIV-1 vaccine in a multicentre, randomised, double-blind, placebo-controlled, phase 1/2a clinical trial (APPROACH) and in rhesus monkeys (NHP 13-19).
- Author
-
Barouch DH, Tomaka FL, Wegmann F, Stieh DJ, Alter G, Robb ML, Michael NL, Peter L, Nkolola JP, Borducchi EN, Chandrashekar A, Jetton D, Stephenson KE, Li W, Korber B, Tomaras GD, Montefiori DC, Gray G, Frahm N, McElrath MJ, Baden L, Johnson J, Hutter J, Swann E, Karita E, Kibuuka H, Mpendo J, Garrett N, Mngadi K, Chinyenze K, Priddy F, Lazarus E, Laher F, Nitayapan S, Pitisuttithum P, Bart S, Campbell T, Feldman R, Lucksinger G, Borremans C, Callewaert K, Roten R, Sadoff J, Scheppler L, Weijtens M, Feddes-de Boer K, van Manen D, Vreugdenhil J, Zahn R, Lavreys L, Nijs S, Tolboom J, Hendriks J, Euler Z, Pau MG, and Schuitemaker H
- Subjects
- AIDS Vaccines adverse effects, Abdominal Pain etiology, Adenoviridae, Adolescent, Adult, Animals, Aspartate Aminotransferases analysis, Back Pain etiology, Diarrhea etiology, Dizziness etiology, Dose-Response Relationship, Drug, Double-Blind Method, Fatigue etiology, Genetic Vectors, Healthy Volunteers, Humans, Immunity, Cellular, Immunity, Humoral, Macaca mulatta, Middle Aged, Young Adult, AIDS Vaccines administration & dosage, HIV-1 immunology
- Abstract
Background: More than 1·8 million new cases of HIV-1 infection were diagnosed worldwide in 2016. No licensed prophylactic HIV-1 vaccine exists. A major limitation to date has been the lack of direct comparability between clinical trials and preclinical studies. We aimed to evaluate mosaic adenovirus serotype 26 (Ad26)-based HIV-1 vaccine candidates in parallel studies in humans and rhesus monkeys to define the optimal vaccine regimen to advance into clinical efficacy trials., Methods: We conducted a multicentre, randomised, double-blind, placebo-controlled phase 1/2a trial (APPROACH). Participants were recruited from 12 clinics in east Africa, South Africa, Thailand, and the USA. We included healthy, HIV-1-uninfected participants (aged 18-50 years) who were considered at low risk for HIV-1 infection. We randomly assigned participants to one of eight study groups, stratified by region. Participants and investigators were blinded to the treatment allocation throughout the study. We primed participants at weeks 0 and 12 with Ad26.Mos.HIV (5 × 10
10 viral particles per 0·5 mL) expressing mosaic HIV-1 envelope (Env)/Gag/Pol antigens and gave boosters at weeks 24 and 48 with Ad26.Mos.HIV or modified vaccinia Ankara (MVA; 108 plaque-forming units per 0·5 mL) vectors with or without high-dose (250 μg) or low-dose (50 μg) aluminium adjuvanted clade C Env gp140 protein. Those in the control group received 0·9% saline. All study interventions were administered intramuscularly. Primary endpoints were safety and tolerability of the vaccine regimens and Env-specific binding antibody responses at week 28. Safety and immunogenicity were also assessed at week 52. All participants who received at least one vaccine dose or placebo were included in the safety analysis; immunogenicity was analysed using the per-protocol population. We also did a parallel study in rhesus monkeys (NHP 13-19) to assess the immunogenicity and protective efficacy of these vaccine regimens against a series of six repetitive, heterologous, intrarectal challenges with a rhesus peripheral blood mononuclear cell-derived challenge stock of simian-human immunodeficiency virus (SHIV-SF162P3). The APPROACH trial is registered with ClinicalTrials.gov, number NCT02315703., Findings: Between Feb 24, 2015, and Oct 16, 2015, we randomly assigned 393 participants to receive at least one dose of study vaccine or placebo in the APPROACH trial. All vaccine regimens demonstrated favourable safety and tolerability. The most commonly reported solicited local adverse event was mild-to-moderate pain at the injection site (varying from 69% to 88% between the different active groups vs 49% in the placebo group). Five (1%) of 393 participants reported at least one grade 3 adverse event considered related to the vaccines: abdominal pain and diarrhoea (in the same participant), increased aspartate aminotransferase, postural dizziness, back pain, and malaise. The mosaic Ad26/Ad26 plus high-dose gp140 boost vaccine was the most immunogenic in humans; it elicited Env-specific binding antibody responses (100%) and antibody-dependent cellular phagocytosis responses (80%) at week 52, and T-cell responses at week 50 (83%). We also randomly assigned 72 rhesus monkeys to receive one of five different vaccine regimens or placebo in the NHP 13-19 study. Ad26/Ad26 plus gp140 boost induced similar magnitude, durability, and phenotype of immune responses in rhesus monkeys as compared with humans and afforded 67% protection against acquisition of SHIV-SF162P3 infection (two-sided Fisher's exact test p=0·007). Env-specific ELISA and enzyme-linked immunospot assay responses were the principal immune correlates of protection against SHIV challenge in monkeys., Interpretation: The mosaic Ad26/Ad26 plus gp140 HIV-1 vaccine induced comparable and robust immune responses in humans and rhesus monkeys, and it provided significant protection against repetitive heterologous SHIV challenges in rhesus monkeys. This vaccine concept is currently being evaluated in a phase 2b clinical efficacy study in sub-Saharan Africa (NCT03060629)., Funding: Janssen Vaccines & Prevention BV, National Institutes of Health, Ragon Institute of MGH, MIT and Harvard, Henry M Jackson Foundation for the Advancement of Military Medicine, US Department of Defense, and International AIDS Vaccine Initiative., (Copyright © 2018 Elsevier Ltd. All rights reserved.)- Published
- 2018
- Full Text
- View/download PDF
31. Similar Epitope Specificities of IgG and IgA Antibodies Elicited by Ad26 Vector Prime, Env Protein Boost Immunizations in Rhesus Monkeys.
- Author
-
Kang ZH, Bricault CA, Borducchi EN, Stephenson KE, Seaman MS, Pau M, Schuitemaker H, van Manen D, Wegmann F, and Barouch DH
- Subjects
- Animals, Epitopes genetics, HIV Envelope Protein gp160 genetics, HIV-1 genetics, Macaca mulatta, Adenoviridae, Epitopes immunology, Genetic Vectors, HIV Antibodies immunology, HIV Envelope Protein gp160 immunology, HIV-1 immunology, Immunization, Secondary, Immunoglobulin A immunology, Immunoglobulin G immunology
- Abstract
Vaccine-elicited immunoglobulin G (IgG) has been shown to be important for protection against simian-human immunodeficiency virus (SHIV) infection in rhesus monkeys. However, it remains unclear whether vaccine-elicited IgA responses are beneficial or detrimental for protection. In this study, we evaluated the kinetics, magnitude, breadth, and linear epitope specificities of vaccine-elicited IgG and IgA responses in serum and mucosal secretions following intramuscular immunization with adenovirus 26 (Ad26) prime, Env protein boost vaccination regimens. The systemic and mucosal antibody responses exhibited kinetics similar to those of the serum antibody responses but lower titers than the serum antibody responses. Moreover, the IgG and IgA responses were correlated, both in terms of the magnitude of the responses and in terms of the antibody specificities against linear human immunodeficiency virus type 1 (HIV-1) Env, Gag, and Pol epitopes. These data suggest that IgG and IgA responses are highly coordinated in both peripheral blood and mucosal compartments following Ad26/Env vaccination in rhesus monkeys. IMPORTANCE Vaccine-elicited IgG responses are important for protection against simian-human immunodeficiency virus (SHIV) infection in nonhuman primates. However, much less is known about the role and function of IgA, despite it being the predominant antibody in mucosal sites. There is debate as to whether HIV-1-specific IgA responses are beneficial or detrimental, since serum anti-Env IgA titers were shown to be inversely correlated with protection in the RV144 clinical trial. We thus assessed vaccine-elicited IgG and IgA antibody responses in peripheral blood and mucosal secretions following vaccination with the Ad26/Env vaccine., (Copyright © 2018 Kang et al.)
- Published
- 2018
- Full Text
- View/download PDF
32. First-in-Human Randomized, Controlled Trial of Mosaic HIV-1 Immunogens Delivered via a Modified Vaccinia Ankara Vector.
- Author
-
Baden LR, Walsh SR, Seaman MS, Cohen YZ, Johnson JA, Licona JH, Filter RD, Kleinjan JA, Gothing JA, Jennings J, Peter L, Nkolola J, Abbink P, Borducchi EN, Kirilova M, Stephenson KE, Pegu P, Eller MA, Trinh HV, Rao M, Ake JA, Sarnecki M, Nijs S, Callewaert K, Schuitemaker H, Hendriks J, Pau MG, Tomaka F, Korber BT, Alter G, Dolin R, Earl PL, Moss B, Michael NL, Robb ML, and Barouch DH
- Subjects
- AIDS Vaccines adverse effects, AIDS Vaccines genetics, Adult, Double-Blind Method, Drug Carriers, Drug-Related Side Effects and Adverse Reactions epidemiology, Drug-Related Side Effects and Adverse Reactions pathology, Female, Genetic Vectors, Humans, Immunity, Cellular, Immunity, Humoral, Longitudinal Studies, Male, Middle Aged, Placebos administration & dosage, Treatment Outcome, Vaccines, Subunit administration & dosage, Vaccines, Subunit adverse effects, Vaccines, Subunit genetics, Vaccines, Subunit immunology, Vaccines, Synthetic administration & dosage, Vaccines, Synthetic adverse effects, Vaccines, Synthetic genetics, Vaccines, Synthetic immunology, Vaccinia virus genetics, Young Adult, AIDS Vaccines administration & dosage, AIDS Vaccines immunology, HIV Infections prevention & control, HIV-1 immunology
- Abstract
Background: Mosaic immunogens are bioinformatically engineered human immunodeficiency virus type 1 (HIV-1) sequences designed to elicit clade-independent coverage against globally circulating HIV-1 strains., Methods: This phase 1, double-blinded, randomized, placebo-controlled trial enrolled healthy HIV-uninfected adults who received 2 doses of a modified vaccinia Ankara (MVA)-vectored HIV-1 bivalent mosaic immunogen vaccine or placebo on days 0 and 84. Two groups were enrolled: those who were HIV-1 vaccine naive (n = 15) and those who had received an HIV-1 vaccine (Ad26.ENVA.01) 4-6 years earlier (n = 10). We performed prespecified blinded cellular and humoral immunogenicity analyses at days 0, 14, 28, 84, 98, 112, 168, 270, and 365., Results: All 50 planned vaccinations were administered. Vaccination was safe and generally well tolerated. No vaccine-related serious adverse events occurred. Both cellular and humoral cross-clade immune responses were elicited after 1 or 2 vaccinations in all participants in the HIV-1 vaccine-naive group. Env-specific responses were induced after a single immunization in nearly all subjects who had previously received the prototype Ad26.ENVA.01 vaccine., Conclusions: No safety concerns were identified, and multiclade HIV-1-specific immune responses were elicited., Clinical Trials Registration: NCT02218125.
- Published
- 2018
- Full Text
- View/download PDF
33. Therapeutic and protective efficacy of a dengue antibody against Zika infection in rhesus monkeys.
- Author
-
Abbink P, Larocca RA, Dejnirattisai W, Peterson R, Nkolola JP, Borducchi EN, Supasa P, Mongkolsapaya J, Screaton GR, and Barouch DH
- Subjects
- Animals, Female, Macaca mulatta, Male, Mice, Inbred BALB C, Antibodies, Viral therapeutic use, Dengue immunology, Zika Virus physiology, Zika Virus Infection prevention & control, Zika Virus Infection virology
- Abstract
Strategies to treat Zika virus (ZIKV) infection in dengue virus (DENV)-endemic areas are urgently needed. Here we show that a DENV-specific antibody against the E-dimer epitope (EDE) potently cross-neutralizes ZIKV and provides robust therapeutic efficacy as well as prophylactic efficacy against ZIKV in rhesus monkeys. Viral escape was not detected, suggesting a relatively high bar to escape. These data demonstrate the potential for antibody-based therapy and prevention of ZIKV.
- Published
- 2018
- Full Text
- View/download PDF
34. Fetal Neuropathology in Zika Virus-Infected Pregnant Female Rhesus Monkeys.
- Author
-
Martinot AJ, Abbink P, Afacan O, Prohl AK, Bronson R, Hecht JL, Borducchi EN, Larocca RA, Peterson RL, Rinaldi W, Ferguson M, Didier PJ, Weiss D, Lewis MG, De La Barrera RA, Yang E, Warfield SK, and Barouch DH
- Subjects
- Animals, Animals, Newborn, Apoptosis, Brain diagnostic imaging, Brain pathology, Calcinosis pathology, Calcinosis veterinary, Female, Gestational Age, Macaca mulatta, Magnetic Resonance Imaging, Necrosis, Neural Stem Cells cytology, Neural Stem Cells metabolism, Neural Stem Cells virology, Neurons virology, Pregnancy, Severity of Illness Index, Vasculitis pathology, Vasculitis veterinary, Zika Virus Infection veterinary, Zika Virus Infection virology, Fetus virology, Neurons pathology, Zika Virus pathogenicity, Zika Virus Infection pathology
- Abstract
The development of interventions to prevent congenital Zika syndrome (CZS) has been limited by the lack of an established nonhuman primate model. Here we show that infection of female rhesus monkeys early in pregnancy with Zika virus (ZIKV) recapitulates many features of CZS in humans. We infected 9 pregnant monkeys with ZIKV, 6 early in pregnancy (weeks 6-7 of gestation) and 3 later in pregnancy (weeks 12-14 of gestation), and compared findings with uninfected controls. 100% (6 of 6) of monkeys infected early in pregnancy exhibited prolonged maternal viremia and fetal neuropathology, including fetal loss, smaller brain size, and histopathologic brain lesions, including microcalcifications, hemorrhage, necrosis, vasculitis, gliosis, and apoptosis of neuroprogenitor cells. High-resolution MRI demonstrated concordant lesions indicative of deep gray matter injury. We also observed spinal, ocular, and neuromuscular pathology. Our data show that vascular compromise and neuroprogenitor cell dysfunction are hallmarks of CZS pathogenesis, suggesting novel strategies to prevent and to treat this disease., (Copyright © 2018 Elsevier Inc. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
35. Rapid Cloning of Novel Rhesus Adenoviral Vaccine Vectors.
- Author
-
Abbink P, Kirilova M, Boyd M, Mercado N, Li Z, Nityanandam R, Nanayakkara O, Peterson R, Larocca RA, Aid M, Tartaglia L, Mutetwa T, Blass E, Jetton D, Maxfield LF, Borducchi EN, Badamchi-Zadeh A, Handley S, Zhao G, Virgin HW, Havenga MJ, and Barouch DH
- Subjects
- A549 Cells, Animals, Humans, Macaca mulatta, Mice, Adenoviridae genetics, Adenoviridae immunology, Adenovirus Vaccines genetics, Adenovirus Vaccines immunology, Cloning, Molecular, Genetic Vectors genetics, Genetic Vectors immunology, Immunogenicity, Vaccine genetics
- Abstract
Human and chimpanzee adenovirus vectors are being developed to circumvent preexisting antibodies against common adenovirus vectors such as Ad5. However, baseline immunity to these vectors still exists in human populations. Traditional cloning of new adenovirus vaccine vectors is a long and cumbersome process that takes 2 months or more and that requires rare unique restriction enzyme sites. Here we describe a novel, restriction enzyme-independent method for rapid cloning of new adenovirus vaccine vectors that reduces the total cloning procedure to 1 week. We developed 14 novel adenovirus vectors from rhesus monkeys that can be grown to high titers and that are immunogenic in mice. All vectors grouped with the unusual adenovirus species G and show extremely low seroprevalence in humans. Rapid cloning of novel adenovirus vectors is a promising approach for the development of new vector platforms. Rhesus adenovirus vectors may prove useful for clinical development. IMPORTANCE To overcome baseline immunity to human and chimpanzee adenovirus vectors, we developed 14 novel adenovirus vectors from rhesus monkeys. These vectors are immunogenic in mice and show extremely low seroprevalence in humans. Rhesus adenovirus vectors may prove useful for clinical development., (Copyright © 2018 Abbink et al.)
- Published
- 2018
- Full Text
- View/download PDF
36. Durability and correlates of vaccine protection against Zika virus in rhesus monkeys.
- Author
-
Abbink P, Larocca RA, Visitsunthorn K, Boyd M, De La Barrera RA, Gromowski GD, Kirilova M, Peterson R, Li Z, Nanayakkara O, Nityanandam R, Mercado NB, Borducchi EN, Chandrashekar A, Jetton D, Mojta S, Gandhi P, LeSuer J, Khatiwada S, Lewis MG, Modjarrad K, Jarman RG, Eckels KH, Thomas SJ, Michael NL, and Barouch DH
- Subjects
- Animals, Antigens, Viral immunology, DNA, Viral metabolism, Female, Macaca mulatta, Mice, Inbred BALB C, Vaccination, Viral Vaccines immunology, Zika Virus immunology, Zika Virus Infection immunology, Zika Virus Infection virology
- Abstract
An effective Zika virus (ZIKV) vaccine will require long-term durable protection. Several ZIKV vaccine candidates have demonstrated protective efficacy in nonhuman primates, but these studies have typically involved ZIKV challenge shortly after vaccination at peak immunity. We show that a single immunization with an adenovirus vector-based vaccine, as well as two immunizations with a purified inactivated virus vaccine, afforded robust protection against ZIKV challenge in rhesus monkeys at 1 year after vaccination. In contrast, two immunizations with an optimized DNA vaccine, which provided complete protection at peak immunity, resulted in reduced protective efficacy at 1 year that was associated with declining neutralizing antibody titers to subprotective levels. These data define a microneutralization log titer of 2.0 to 2.1 as the threshold required for durable protection against ZIKV challenge in this model. Moreover, our findings demonstrate that protection against ZIKV challenge in rhesus monkeys is possible for at least 1 year with a single-shot vaccine., (Copyright © 2017 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works.)
- Published
- 2017
- Full Text
- View/download PDF
37. Protection against a mixed SHIV challenge by a broadly neutralizing antibody cocktail.
- Author
-
Julg B, Liu PT, Wagh K, Fischer WM, Abbink P, Mercado NB, Whitney JB, Nkolola JP, McMahan K, Tartaglia LJ, Borducchi EN, Khatiwada S, Kamath M, LeSuer JA, Seaman MS, Schmidt SD, Mascola JR, Burton DR, Korber BT, and Barouch DH
- Subjects
- Amino Acid Sequence, Animals, Antibodies, Neutralizing chemistry, Base Sequence, Epitopes immunology, Gene Products, env chemistry, Gene Products, env genetics, HIV-1 immunology, Inhibitory Concentration 50, Macaca mulatta, Simian Acquired Immunodeficiency Syndrome virology, Antibodies, Neutralizing therapeutic use, Simian Acquired Immunodeficiency Syndrome immunology, Simian Acquired Immunodeficiency Syndrome prevention & control, Simian Immunodeficiency Virus immunology
- Abstract
HIV-1 sequence diversity presents a major challenge for the clinical development of broadly neutralizing antibodies (bNAbs) for both therapy and prevention. Sequence variation in critical bNAb epitopes has been observed in most HIV-1-infected individuals and can lead to viral escape after bNAb monotherapy in humans. We show that viral sequence diversity can limit both the therapeutic and prophylactic efficacy of bNAbs in rhesus monkeys. We first demonstrate that monotherapy with the V3 glycan-dependent antibody 10-1074, but not PGT121, results in rapid selection of preexisting viral variants containing N332/S334 escape mutations and loss of therapeutic efficacy in simian-HIV (SHIV)-SF162P3-infected rhesus monkeys. We then show that the V3 glycan-dependent antibody PGT121 alone and the V2 glycan-dependent antibody PGDM1400 alone both fail to protect against a mixed challenge with SHIV-SF162P3 and SHIV-325c. In contrast, the combination of both bNAbs provides 100% protection against this mixed SHIV challenge. These data reveal that single bNAbs efficiently select resistant viruses from a diverse challenge swarm to establish infection, demonstrating the importance of bNAb cocktails for HIV-1 prevention., (Copyright © 2017 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works.)
- Published
- 2017
- Full Text
- View/download PDF
38. Adenovirus prime, Env protein boost vaccine protects against neutralization-resistant SIVsmE660 variants in rhesus monkeys.
- Author
-
Keele BF, Li W, Borducchi EN, Nkolola JP, Abbink P, Chen B, Seaman MS, and Barouch DH
- Subjects
- Adenoviridae genetics, Animals, Antibodies, Neutralizing immunology, Female, Gene Products, env genetics, Immunity, Humoral, Immunization, Secondary, Macaca mulatta, Male, Neutralization Tests, Simian Acquired Immunodeficiency Syndrome immunology, Simian Immunodeficiency Virus immunology, Vaccines, DNA immunology, Viral Vaccines immunology, Gene Products, env immunology, Simian Acquired Immunodeficiency Syndrome prevention & control, Simian Immunodeficiency Virus pathogenicity, Vaccines, DNA pharmacology, Viral Vaccines pharmacology
- Abstract
Previous studies have shown that DNA prime, Ad5 boost vaccines protect against neutralization-sensitive but not neutralization-resistant virus variants within the SIVsmE660 swarm. Here we show that Ad prime, Env protein boost vaccines protect against neutralization-resistant SIVsmE660 variants. We perform two studies in rhesus monkeys with Ad35/Ad26 vectors expressing SIVmac239 Gag/Pol/Env with or without an AS01
B -adjuvanted SIVmac32H gp140 protein boost. In a repetitive, low-dose challenge study, we observe robust protection against acquisition of infection by both Ad Alone and Ad/Env vaccines. In a single, high-dose challenge study, only the Ad/Env vaccine affords significant protection against acquisition of infection. Analysis of transmitted/founder (T/F) viruses from this study demonstrates that the Ad/Env vaccine blocks both neutralization-sensitive and neutralization-resistant SIVsmE660 variants in rhesus monkeys with restrictive TRIM5α alleles. These data demonstrate that the adjuvanted Env protein boost is critical for protecting against high-dose SIVsmE660 challenge and for blocking neutralization-resistant viruses within the SIVsmE660 swarm.- Published
- 2017
- Full Text
- View/download PDF
39. Zika Virus Persistence in the Central Nervous System and Lymph Nodes of Rhesus Monkeys.
- Author
-
Aid M, Abbink P, Larocca RA, Boyd M, Nityanandam R, Nanayakkara O, Martinot AJ, Moseley ET, Blass E, Borducchi EN, Chandrashekar A, Brinkman AL, Molloy K, Jetton D, Tartaglia LJ, Liu J, Best K, Perelson AS, De La Barrera RA, Lewis MG, and Barouch DH
- Subjects
- Animals, Cerebrospinal Fluid virology, Inflammation immunology, Lower Gastrointestinal Tract virology, Lymph Nodes virology, Macaca mulatta, Signal Transduction, TOR Serine-Threonine Kinases metabolism, Zika Virus Infection immunology, Zika Virus Infection virology
- Abstract
Zika virus (ZIKV) is associated with severe neuropathology in neonates as well as Guillain-Barré syndrome and other neurologic disorders in adults. Prolonged viral shedding has been reported in semen, suggesting the presence of anatomic viral reservoirs. Here we show that ZIKV can persist in cerebrospinal fluid (CSF) and lymph nodes (LN) of infected rhesus monkeys for weeks after virus has been cleared from peripheral blood, urine, and mucosal secretions. ZIKV-specific neutralizing antibodies correlated with rapid clearance of virus in peripheral blood but remained undetectable in CSF for the duration of the study. Viral persistence in both CSF and LN correlated with upregulation of mechanistic target of rapamycin (mTOR), proinflammatory, and anti-apoptotic signaling pathways, as well as downregulation of extracellular matrix signaling pathways. These data raise the possibility that persistent or occult neurologic and lymphoid disease may occur following clearance of peripheral virus in ZIKV-infected individuals., (Copyright © 2017 Elsevier Inc. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
40. Ad26/MVA therapeutic vaccination with TLR7 stimulation in SIV-infected rhesus monkeys.
- Author
-
Borducchi EN, Cabral C, Stephenson KE, Liu J, Abbink P, Ng'ang'a D, Nkolola JP, Brinkman AL, Peter L, Lee BC, Jimenez J, Jetton D, Mondesir J, Mojta S, Chandrashekar A, Molloy K, Alter G, Gerold JM, Hill AL, Lewis MG, Pau MG, Schuitemaker H, Hesselgesser J, Geleziunas R, Kim JH, Robb ML, Michael NL, and Barouch DH
- Subjects
- AIDS Vaccines genetics, AIDS Vaccines immunology, Animals, Anti-Retroviral Agents administration & dosage, DNA, Viral analysis, DNA, Viral blood, Female, Genetic Vectors genetics, HIV Infections immunology, HIV Infections therapy, Immunity, Cellular, Immunity, Innate, Macaca mulatta, Male, RNA, Viral analysis, RNA, Viral blood, SAIDS Vaccines genetics, Simian Acquired Immunodeficiency Syndrome virology, Simian Immunodeficiency Virus genetics, Simian Immunodeficiency Virus growth & development, Simian Immunodeficiency Virus isolation & purification, Time Factors, Toll-Like Receptor 7 genetics, Viral Load immunology, Adenoviridae genetics, SAIDS Vaccines immunology, Simian Acquired Immunodeficiency Syndrome immunology, Simian Acquired Immunodeficiency Syndrome therapy, Simian Immunodeficiency Virus immunology, Toll-Like Receptor 7 immunology, Vaccinia virus genetics
- Abstract
The development of immunologic interventions that can target the viral reservoir in HIV-1-infected individuals is a major goal of HIV-1 research. However, little evidence exists that the viral reservoir can be sufficiently targeted to improve virologic control following discontinuation of antiretroviral therapy. Here we show that therapeutic vaccination with Ad26/MVA (recombinant adenovirus serotype 26 (Ad26) prime, modified vaccinia Ankara (MVA) boost) and stimulation of TLR7 (Toll-like receptor 7) improves virologic control and delays viral rebound following discontinuation of antiretroviral therapy in SIV-infected rhesus monkeys that began antiretroviral therapy during acute infection. Therapeutic vaccination with Ad26/MVA resulted in a marked increase in the magnitude and breadth of SIV-specific cellular immune responses in virologically suppressed, SIV-infected monkeys. TLR7 agonist administration led to innate immune stimulation and cellular immune activation. The combination of Ad26/MVA vaccination and TLR7 stimulation resulted in decreased levels of viral DNA in lymph nodes and peripheral blood, and improved virologic control and delayed viral rebound following discontinuation of antiretroviral therapy. The breadth of cellular immune responses correlated inversely with set point viral loads and correlated directly with time to viral rebound. These data demonstrate the potential of therapeutic vaccination combined with innate immune stimulation as a strategy aimed at a functional cure for HIV-1 infection.
- Published
- 2016
- Full Text
- View/download PDF
41. Adenovirus serotype 5 vaccine vectors trigger IL-27-dependent inhibitory CD4 + T cell responses that impair CD8 + T cell function.
- Author
-
Larocca RA, Provine NM, Aid M, Iampietro MJ, Borducchi EN, Badamchi-Zadeh A, Abbink P, Ng'ang'a D, Bricault CA, Blass E, Penaloza-MacMaster P, Stephenson KE, and Barouch DH
- Abstract
Adenovirus serotype 5 (Ad5) vaccine vectors elicit robust CD8
+ T cell responses, but these responses typically exhibit a partially exhausted phenotype. However, the immunologic mechanism by which Ad5 vectors induce dysfunctional CD8+ T cells has not previously been elucidated. Here we demonstrate that, following immunization of B6 mice, Ad5 vectors elicit antigen-specific IL-10+ CD4+ T cells with a distinct transcriptional profile in a dose-dependent fashion. In rhesus monkeys, we similarly observed upregulated expression of IL-10 and PD-1 by CD4+ T cells following Ad5 vaccination. These cells markedly suppressed vaccine-elicited CD8+ T cell responses in vivo and IL-10 blockade increased the frequency and functionality of antigen-specific CD8+ T cells as well as improved protective efficacy against challenge with recombinant Listeria monocytogenes . Moreover, induction of these inhibitory IL-10+ CD4+ T cells correlated with IL-27 expression and IL-27 blockade substantially improved CD4+ T cell functionality. These data highlight a role for IL-27 in the induction of inhibitory IL-10+ CD4+ T cells, which suppress CD8+ T cell magnitude and function following Ad5 vector immunization. A deeper understanding of the cytokine networks and transcriptional profiles induced by vaccine vectors should lead to strategies to improve the immunogenicity and protective efficacy of viral vector-based vaccines., Competing Interests: Competing interests: The authors declare that they have no competing interests.- Published
- 2016
- Full Text
- View/download PDF
42. Protective efficacy of multiple vaccine platforms against Zika virus challenge in rhesus monkeys.
- Author
-
Abbink P, Larocca RA, De La Barrera RA, Bricault CA, Moseley ET, Boyd M, Kirilova M, Li Z, Ng'ang'a D, Nanayakkara O, Nityanandam R, Mercado NB, Borducchi EN, Agarwal A, Brinkman AL, Cabral C, Chandrashekar A, Giglio PB, Jetton D, Jimenez J, Lee BC, Mojta S, Molloy K, Shetty M, Neubauer GH, Stephenson KE, Peron JP, Zanotto PM, Misamore J, Finneyfrock B, Lewis MG, Alter G, Modjarrad K, Jarman RG, Eckels KH, Michael NL, Thomas SJ, and Barouch DH
- Subjects
- Adenoviridae, Adoptive Transfer, Animals, Antibodies, Viral biosynthesis, Antibodies, Viral immunology, Brazil, Female, Genetic Vectors, Humans, Immunoglobulins immunology, Immunoglobulins isolation & purification, Macaca mulatta, Male, Mice, Mice, Inbred BALB C, Puerto Rico, Vaccines, DNA administration & dosage, Vaccines, Inactivated administration & dosage, Vaccines, Inactivated immunology, Viral Envelope Proteins genetics, Viral Envelope Proteins immunology, Viral Vaccines administration & dosage, Immunogenicity, Vaccine, Vaccines, DNA immunology, Viral Vaccines immunology, Zika Virus immunology, Zika Virus Infection prevention & control
- Abstract
Zika virus (ZIKV) is responsible for a major ongoing epidemic in the Americas and has been causally associated with fetal microcephaly. The development of a safe and effective ZIKV vaccine is therefore an urgent global health priority. Here we demonstrate that three different vaccine platforms protect against ZIKV challenge in rhesus monkeys. A purified inactivated virus vaccine induced ZIKV-specific neutralizing antibodies and completely protected monkeys against ZIKV strains from both Brazil and Puerto Rico. Purified immunoglobulin from vaccinated monkeys also conferred passive protection in adoptive transfer studies. A plasmid DNA vaccine and a single-shot recombinant rhesus adenovirus serotype 52 vector vaccine, both expressing ZIKV premembrane and envelope, also elicited neutralizing antibodies and completely protected monkeys against ZIKV challenge. These data support the rapid clinical development of ZIKV vaccines for humans., (Copyright © 2016, American Association for the Advancement of Science.)
- Published
- 2016
- Full Text
- View/download PDF
43. Immediate Dysfunction of Vaccine-Elicited CD8+ T Cells Primed in the Absence of CD4+ T Cells.
- Author
-
Provine NM, Larocca RA, Aid M, Penaloza-MacMaster P, Badamchi-Zadeh A, Borducchi EN, Yates KB, Abbink P, Kirilova M, Ng'ang'a D, Bramson J, Haining WN, and Barouch DH
- Subjects
- Adenoviridae genetics, Animals, CD8-Positive T-Lymphocytes physiology, Cytokines immunology, Cytotoxicity, Immunologic, Female, Gene Expression Regulation, Genetic Vectors, Immunization, Immunologic Memory, Lymphocyte Activation, Mice, Mice, Inbred C57BL, CD4-Positive T-Lymphocytes immunology, CD8-Positive T-Lymphocytes immunology, Vaccines immunology
- Abstract
CD4(+) T cell help is critical for optimal CD8(+) T cell memory differentiation and maintenance in many experimental systems. In addition, many reports have identified reduced primary CD8(+) T cell responses in the absence of CD4(+) T cell help, which often coincides with reduced Ag or pathogen clearance. In this study, we demonstrate that absence of CD4(+) T cells at the time of adenovirus vector immunization of mice led to immediate impairments in early CD8(+) T cell functionality and differentiation. Unhelped CD8(+) T cells exhibited a reduced effector phenotype, decreased ex vivo cytotoxicity, and decreased capacity to produce cytokines. This dysfunctional state was imprinted within 3 d of immunization. Unhelped CD8(+) T cells expressed elevated levels of inhibitory receptors and exhibited transcriptomic exhaustion and anergy profiles by gene set enrichment analysis. Dysfunctional, impaired effector differentiation also occurred following immunization of CD4(+) T cell-deficient mice with a poxvirus vector. This study demonstrates that following priming with viral vectors, CD4(+) T cell help is required to promote both the expansion and acquisition of effector functions by CD8(+) T cells, which is accomplished by preventing immediate dysfunction., (Copyright © 2016 by The American Association of Immunologists, Inc.)
- Published
- 2016
- Full Text
- View/download PDF
44. Vaccine protection against Zika virus from Brazil.
- Author
-
Larocca RA, Abbink P, Peron JP, Zanotto PM, Iampietro MJ, Badamchi-Zadeh A, Boyd M, Ng'ang'a D, Kirilova M, Nityanandam R, Mercado NB, Li Z, Moseley ET, Bricault CA, Borducchi EN, Giglio PB, Jetton D, Neubauer G, Nkolola JP, Maxfield LF, De La Barrera RA, Jarman RG, Eckels KH, Michael NL, Thomas SJ, and Barouch DH
- Subjects
- Adoptive Transfer, Animals, Antibodies, Neutralizing immunology, Antibodies, Viral immunology, Antibody Specificity, Brazil, CD4-Positive T-Lymphocytes immunology, CD8-Positive T-Lymphocytes immunology, Female, Gene Deletion, Humans, Immunoglobulin G immunology, Immunoglobulin G isolation & purification, Mice, Microcephaly complications, Microcephaly virology, Vaccines, DNA chemistry, Vaccines, DNA genetics, Vaccines, DNA immunology, Vaccines, Inactivated chemistry, Vaccines, Inactivated genetics, Vaccines, Inactivated immunology, Vaccines, Subunit chemistry, Vaccines, Subunit genetics, Vaccines, Subunit immunology, Viral Envelope Proteins chemistry, Viral Envelope Proteins genetics, Viral Envelope Proteins immunology, Viral Vaccines chemistry, Viral Vaccines genetics, Zika Virus chemistry, Zika Virus genetics, Zika Virus Infection complications, Zika Virus Infection immunology, Viral Vaccines immunology, Zika Virus immunology, Zika Virus Infection prevention & control, Zika Virus Infection virology
- Abstract
Zika virus (ZIKV) is a flavivirus that is responsible for the current epidemic in Brazil and the Americas. ZIKV has been causally associated with fetal microcephaly, intrauterine growth restriction, and other birth defects in both humans and mice. The rapid development of a safe and effective ZIKV vaccine is a global health priority, but very little is currently known about ZIKV immunology and mechanisms of immune protection. Here we show that a single immunization with a plasmid DNA vaccine or a purified inactivated virus vaccine provides complete protection in susceptible mice against challenge with a strain of ZIKV involved in the outbreak in northeast Brazil. This ZIKV strain has recently been shown to cross the placenta and to induce fetal microcephaly and other congenital malformations in mice. We produced DNA vaccines expressing ZIKV pre-membrane and envelope (prM-Env), as well as a series of deletion mutants. The prM-Env DNA vaccine, but not the deletion mutants, afforded complete protection against ZIKV, as measured by absence of detectable viraemia following challenge, and protective efficacy correlated with Env-specific antibody titers. Adoptive transfer of purified IgG from vaccinated mice conferred passive protection, and depletion of CD4 and CD8 T lymphocytes in vaccinated mice did not abrogate this protection. These data demonstrate that protection against ZIKV challenge can be achieved by single-shot subunit and inactivated virus vaccines in mice and that Env-specific antibody titers represent key immunologic correlates of protection. Our findings suggest that the development of a ZIKV vaccine for humans is likely to be achievable.
- Published
- 2016
- Full Text
- View/download PDF
45. Neutralizing antibody affords comparable protection against vaginal and rectal simian/human immunodeficiency virus challenge in macaques.
- Author
-
Moldt B, Le KM, Carnathan DG, Whitney JB, Schultz N, Lewis MG, Borducchi EN, Smith KM, Mackel JJ, Sweat SL, Hodges AP, Godzik A, Parren PW, Silvestri G, Barouch DH, and Burton DR
- Subjects
- Animals, Disease Models, Animal, Female, Macaca, Treatment Outcome, Viral Load, Antibodies, Neutralizing administration & dosage, HIV immunology, HIV Antibodies administration & dosage, HIV Infections prevention & control, Rectum immunology, Vagina immunology
- Abstract
Objective: Passive administration of broadly neutralizing antibodies has been shown to protect against both vaginal and rectal challenge in the simian/human immunodeficiency virus (SHIV)/macaque model of HIV transmission. However, the relative efficacy of antibody against the two modes of exposure is unknown and, given differences in the composition and immunology of the two tissue compartments, this is an important gap in knowledge. To investigate the significance of the challenge route for antibody-mediated protection, we performed a comparative protection study in macaques using the highly potent human monoclonal antibody, PGT126., Design: Animals were administered PGT126 at three different doses before challenged either vaginally or rectally with a single dose of SHIVSF163P3., Methods: Viral loads, PGT126 serum concentrations, and serum neutralizing titers were monitored., Results: In vaginally challenged animals, sterilizing immunity was achieved in all animals administered 10 mg/kg, in two of five animals administered 2 mg/kg and in one of five animals administered 0.4 mg/kg PGT126. Comparable protection was observed for the corresponding groups challenged rectally as sterilizing immunity was achieved in three of four animals administered 10 mg/kg, in two of four animals administered 2 mg/kg and in none of four animals administered 0.4 mg/kg PGT126. Serological analysis showed similar serum concentrations of PGT126 and serum neutralization titers in animals administered the same antibody dose., Conclusion: Our data suggest that broadly neutralizing antibody-mediated protection is not strongly dependent on the mucosal route of challenge, which indicates that a vaccine aimed to induce a neutralizing antibody response would have broadly similar efficacy against both primary transmission routes for HIV.
- Published
- 2016
- Full Text
- View/download PDF
46. Transient CD4+ T Cell Depletion Results in Delayed Development of Functional Vaccine-Elicited Antibody Responses.
- Author
-
Provine NM, Badamchi-Zadeh A, Bricault CA, Penaloza-MacMaster P, Larocca RA, Borducchi EN, Seaman MS, and Barouch DH
- Subjects
- Adenoviridae genetics, Adenoviridae immunology, Animals, Antibodies, Neutralizing blood, Antibodies, Neutralizing immunology, Antibodies, Viral blood, Antibodies, Viral immunology, B-Lymphocytes immunology, B-Lymphocytes metabolism, CD4-Positive T-Lymphocytes metabolism, Disease Models, Animal, Epitopes, T-Lymphocyte immunology, Genetic Vectors genetics, Genetic Vectors immunology, Germinal Center immunology, Immunization, Mice, Mice, Knockout, Simian Acquired Immunodeficiency Syndrome immunology, Simian Immunodeficiency Virus genetics, Simian Immunodeficiency Virus immunology, Antibodies immunology, Antibody Formation immunology, CD4-Positive T-Lymphocytes immunology, Lymphocyte Depletion methods, Vaccines immunology
- Abstract
Unlabelled: We have recently demonstrated that CD4(+)T cell help is required at the time of adenovirus (Ad) vector immunization for the development of functional CD8(+)T cell responses, but the temporal requirement for CD4(+)T cell help for the induction of antibody responses remains unclear. Here we demonstrate that induction of antibody responses in C57BL/6 mice can occur at a time displaced from the time of Ad vector immunization by depletion of CD4(+)T cells. Transient depletion of CD4(+)T cells at the time of immunization delays the development of antigen-specific antibody responses but does not permanently impair their development or induce tolerance against the transgene. Upon CD4(+)T cell recovery, transgene-specific serum IgG antibody titers develop and reach a concentration equivalent to that in undepleted control animals. These delayed antibody responses exhibit no functional defects with regard to isotype, functional avidity, expansion after boosting immunization, or the capacity to neutralize a simian immunodeficiency virus (SIV) Env-expressing pseudovirus. The development of this delayed transgene-specific antibody response is temporally linked to the expansion of de novo antigen-specific CD4(+)T cell responses, which develop after transient depletion of CD4(+)T cells. These data demonstrate that functional vaccine-elicited antibody responses can be induced even if CD4(+)T cell help is provided at a time markedly separated from the time of vaccination., Importance: CD4(+)T cells have a critical role in providing positive help signals to B cells, which promote robust antibody responses. The paradigm is that helper signals must be provided immediately upon antigen exposure, and their absence results in tolerance against the antigen. Here we demonstrate that, in contrast to the current model that the absence of CD4(+)T cell help at priming results in long-term antibody nonresponsiveness, antibody responses can be induced by adenovirus vector immunization or alum-adjuvanted protein immunization even if CD4(+)T cell help is not provided until >1 month after immunization. These data demonstrate that the time when CD4(+)T cell help signals must be provided is more dynamic and flexible than previously appreciated. These data suggest that augmentation of CD4(+)T cell helper function even after the time of vaccination can enhance vaccine-elicited antibody responses and thereby potentially enhance the immunogenicity of vaccines in immunocompromised individuals., (Copyright © 2016 Provine et al.)
- Published
- 2016
- Full Text
- View/download PDF
47. Attenuation of Replication-Competent Adenovirus Serotype 26 Vaccines by Vectorization.
- Author
-
Maxfield LF, Abbink P, Stephenson KE, Borducchi EN, Ng'ang'a D, Kirilova MM, Paulino N, Boyd M, Shabram P, Ruan Q, Patel M, and Barouch DH
- Subjects
- Adenovirus Vaccines immunology, Clinical Trials, Phase I as Topic, Gene Expression, Humans, Mutagenesis, Insertional, Serogroup, env Gene Products, Human Immunodeficiency Virus genetics, Adenovirus Vaccines genetics, Adenoviruses, Human genetics, Adenoviruses, Human physiology, Genetic Vectors, Virus Replication
- Abstract
Replication-competent adenovirus (rcAd)-based vaccine vectors may theoretically provide immunological advantages over replication-incompetent Ad vectors, but they also raise additional potential clinical and regulatory issues. We produced replication-competent Ad serotype 26 (rcAd26) vectors by adding the E1 region back into a replication-incompetent Ad26 vector backbone with the E3 or E3/E4 regions deleted. We assessed the effect of vectorization on the replicative capacity of the rcAd26 vaccines. Attenuation occurred in a stepwise fashion, with E3 deletion, E4 deletion, and human immunodeficiency virus type 1 (HIV-1) envelope (Env) gene insertion all contributing to reduced replicative capacity compared to that with the wild-type Ad26 vector. The rcAd26 vector with E3 and E4 deleted and containing the Env transgene exhibited 2.7- to 4.4-log-lower replicative capacity than that of the wild-type Ad26 in vitro. This rcAd26 vector is currently being evaluated in a phase 1 clinical trial. Attenuation as a result of vectorization and transgene insertion has implications for the clinical development of replication-competent vaccine vectors., (Copyright © 2015, Maxfield et al.)
- Published
- 2015
- Full Text
- View/download PDF
48. Protective efficacy of adenovirus/protein vaccines against SIV challenges in rhesus monkeys.
- Author
-
Barouch DH, Alter G, Broge T, Linde C, Ackerman ME, Brown EP, Borducchi EN, Smith KM, Nkolola JP, Liu J, Shields J, Parenteau L, Whitney JB, Abbink P, Ng'ang'a DM, Seaman MS, Lavine CL, Perry JR, Li W, Colantonio AD, Lewis MG, Chen B, Wenschuh H, Reimer U, Piatak M, Lifson JD, Handley SA, Virgin HW, Koutsoukos M, Lorin C, Voss G, Weijtens M, Pau MG, and Schuitemaker H
- Subjects
- Adoptive Transfer, Animals, Antibodies, Neutralizing immunology, Female, Gene Products, gag immunology, Gene Products, pol immunology, Genetic Vectors immunology, Histocompatibility Antigens Class I genetics, Histocompatibility Antigens Class I immunology, Immunization, Secondary, Macaca mulatta, Male, Simian Immunodeficiency Virus immunology, AIDS Vaccines immunology, Adenovirus Vaccines immunology, Gene Products, env immunology, HIV-1 immunology, SAIDS Vaccines immunology, Simian Acquired Immunodeficiency Syndrome prevention & control
- Abstract
Preclinical studies of viral vector-based HIV-1 vaccine candidates have previously shown partial protection against neutralization-resistant virus challenges in rhesus monkeys. In this study, we evaluated the protective efficacy of adenovirus serotype 26 (Ad26) vector priming followed by purified envelope (Env) glycoprotein boosting. Rhesus monkeys primed with Ad26 vectors expressing SIVsmE543 Env, Gag, and Pol and boosted with AS01B-adjuvanted SIVmac32H Env gp140 demonstrated complete protection in 50% of vaccinated animals against a series of repeated, heterologous, intrarectal SIVmac251 challenges that infected all controls. Protective efficacy correlated with the functionality of Env-specific antibody responses. Comparable protection was also observed with a similar Ad/Env vaccine against repeated, heterologous, intrarectal SHIV-SF162P3 challenges. These data demonstrate robust protection by Ad/Env vaccines against acquisition of neutralization-resistant virus challenges in rhesus monkeys., (Copyright © 2015, American Association for the Advancement of Science.)
- Published
- 2015
- Full Text
- View/download PDF
49. Construction and evaluation of novel rhesus monkey adenovirus vaccine vectors.
- Author
-
Abbink P, Maxfield LF, Ng'ang'a D, Borducchi EN, Iampietro MJ, Bricault CA, Teigler JE, Blackmore S, Parenteau L, Wagh K, Handley SA, Zhao G, Virgin HW, Korber B, and Barouch DH
- Subjects
- Adenoviridae genetics, Adenoviridae immunology, Adenoviridae Infections immunology, Adenoviridae Infections veterinary, Adenoviridae Infections virology, Africa South of the Sahara, Animals, Antibodies, Viral blood, Cluster Analysis, DNA, Viral chemistry, DNA, Viral genetics, Humans, Macaca mulatta, Mice, Inbred BALB C, Molecular Sequence Data, Phylogeny, Sequence Analysis, DNA, Seroepidemiologic Studies, Vaccines, Synthetic genetics, Adenoviridae classification, Adenoviridae isolation & purification, Drug Carriers isolation & purification, Genetic Vectors isolation & purification
- Abstract
Unlabelled: Adenovirus vectors are widely used as vaccine candidates for a variety of pathogens, including HIV-1. To date, human and chimpanzee adenoviruses have been explored in detail as vaccine vectors. The phylogeny of human and chimpanzee adenoviruses is overlapping, and preexisting humoral and cellular immunity to both are exhibited in human populations worldwide. More distantly related adenoviruses may therefore offer advantages as vaccine vectors. Here we describe the primary isolation and vectorization of three novel adenoviruses from rhesus monkeys. The seroprevalence of these novel rhesus monkey adenovirus vectors was extremely low in sub-Saharan Africa human populations, and these vectors proved to have immunogenicity comparable to that of human and chimpanzee adenovirus vaccine vectors in mice. These rhesus monkey adenoviruses phylogenetically clustered with the poorly described adenovirus species G and robustly stimulated innate immune responses. These novel adenoviruses represent a new class of candidate vaccine vectors., Importance: Although there have been substantial efforts in the development of vaccine vectors from human and chimpanzee adenoviruses, far less is known about rhesus monkey adenoviruses. In this report, we describe the isolation and vectorization of three novel rhesus monkey adenoviruses. These vectors exhibit virologic and immunologic characteristics that make them attractive as potential candidate vaccine vectors for both HIV-1 and other pathogens., (Copyright © 2015, American Society for Microbiology. All Rights Reserved.)
- Published
- 2015
- Full Text
- View/download PDF
50. Vaccine-elicited CD4 T cells induce immunopathology after chronic LCMV infection.
- Author
-
Penaloza-MacMaster P, Barber DL, Wherry EJ, Provine NM, Teigler JE, Parenteau L, Blackmore S, Borducchi EN, Larocca RA, Yates KB, Shen H, Haining WN, Sommerstein R, Pinschewer DD, Ahmed R, and Barouch DH
- Subjects
- Adaptive Immunity, Animals, Antibodies, Viral immunology, Antigens, Viral immunology, Arenaviridae Infections virology, CD8-Positive T-Lymphocytes immunology, Cytokines blood, Epitopes, T-Lymphocyte immunology, Immune System Diseases immunology, Immune System Diseases pathology, Immunologic Memory, Inflammation immunology, Inflammation pathology, Lymphocytic choriomeningitis virus physiology, Mice, Inbred C57BL, Multiple Organ Failure etiology, Vaccination, Viral Load, Virus Replication, Arenaviridae Infections immunology, CD4-Positive T-Lymphocytes immunology, Immune System Diseases etiology, Inflammation etiology, Lymphocytic choriomeningitis virus immunology, Viral Vaccines adverse effects, Viral Vaccines immunology
- Abstract
CD4 T cells promote innate and adaptive immune responses, but how vaccine-elicited CD4 T cells contribute to immune protection remains unclear. We evaluated whether induction of virus-specific CD4 T cells by vaccination would protect mice against infection with chronic lymphocytic choriomeningitis virus (LCMV). Immunization with vaccines that selectively induced CD4 T cell responses resulted in catastrophic inflammation and mortality after challenge with a persistent strain of LCMV. Immunopathology required antigen-specific CD4 T cells and was associated with a cytokine storm, generalized inflammation, and multi-organ system failure. Virus-specific CD8 T cells or antibodies abrogated the pathology. These data demonstrate that vaccine-elicited CD4 T cells in the absence of effective antiviral immune responses can trigger lethal immunopathology., (Copyright © 2015, American Association for the Advancement of Science.)
- Published
- 2015
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.