122 results on '"Currier JR"'
Search Results
2. P10-12. Altered NK cell phenotype and function in Ugandans with chronic HIV-1 infection
- Author
-
Robb ML, Wabwire-Mangen F, de Souza MS, Michael NL, Marovich MA, Guwatudde D, Kijak GH, Koehler RN, Eller L, Eller MA, Currier JR, and Sandberg JK
- Subjects
Immunologic diseases. Allergy ,RC581-607 - Published
- 2009
- Full Text
- View/download PDF
3. The Pairing of Rapid Intensification Events and Eyewall Replacement Cycles in Tropical Cyclones in the Atlantic Basin from 2015 to 2020.
- Author
-
Currier Jr., John W. and Preston, Ari D.
- Subjects
- *
TROPICAL cyclones , *METEOROLOGICAL satellites , *STATISTICAL significance , *SOLAR stills , *STATISTICS - Abstract
Rapid intensification (RI) and eyewall replacement cycles (ERCs) frequently occur in intense tropical cyclones (TCs), often causing rapid, significant changes in intensity and structure. In some TCs, RI and ERCs can occur concurrently or within a short period of one another. This study investigates whether there is a link between RI and ERCs by conducting a statistical analysis of TCs that occurred in the North Atlantic basin from 2015 to 2020. The HURDAT2 dataset was used to detect RI events, while the Morphed Integrated Microwave Imagery archive by the Cooperative Institute for Meteorological Satellite Studies was used to detect ERC events. Three sets of data were constructed from this analysis: TCs with paired RI/ERC events that occurred within 24 h of each other, TCs with RI only, and TCs with ERCs only. Statistics selected for analysis within the constructed datasets were mean duration of phenomena, mean rate of intensification, and mean peak intensity. We performed t-tests to determine the statistical significance of results. The results of this study show that TCs with these paired RI/ERC events often intensified at a faster rate, intensified for longer, and ended up stronger than TCs that only experienced RI or ERCs in isolation. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
4. P10-12. Altered NK cell phenotype and function in Ugandans with chronic HIV-1 infection
- Author
-
Eller, MA, primary, Eller, L, additional, Koehler, RN, additional, Kijak, GH, additional, Guwatudde, D, additional, Marovich, MA, additional, Michael, NL, additional, de Souza, MS, additional, Wabwire-Mangen, F, additional, Robb, ML, additional, Currier, JR, additional, and Sandberg, JK, additional
- Published
- 2009
- Full Text
- View/download PDF
5. S011-05 OA. HLA-A*7401 is associated with protection from HIV-1 acquisition and disease progression in Mbeya, Tanzania
- Author
-
Koehler, RN, primary, Walsh, AM, additional, Saathoff, E, additional, Currier, JR, additional, Bautista, CT, additional, Moqueet, N, additional, Ratto-Kim, S, additional, Maboko, L, additional, Hoelscher, M, additional, Robb, ML, additional, Michael, NL, additional, McCutchan, FE, additional, Kim, JH, additional, and Kijak, GH, additional
- Published
- 2009
- Full Text
- View/download PDF
6. OA04-03. Characterization of cell-mediated immune responses generated by recombinant modified vaccinia Ankara (rMVA)-HIV-1 in a phase I vaccine trial
- Author
-
Currier, JR, primary, De Souza, M, additional, Ratto-Kim, S, additional, Paris, R, additional, Nguay, V, additional, Cox, J, additional, Earl, P, additional, Moss, B, additional, Sriplienchan, S, additional, Thongcharoen, P, additional, Kim, J, additional, Robb, M, additional, Michael, N, additional, and Marovich, M, additional
- Published
- 2009
- Full Text
- View/download PDF
7. Two Lines to Carol
- Author
-
Currier, Jr., John C.
- Subjects
Literature/writing - Abstract
Content not available due to copyright [...]
- Published
- 1972
8. Donor hepatitis C virus status does not adversely affect short-term outcomes in HCV+ recipients in renal transplantation
- Author
-
All, M.K., Light, J.A., Barhyte, D.Y., Sasaki, T.M., Currier, Jr.,, C.B., Grandas, O., and Fowlkes, D.
- Published
- 1998
9. Harmonization guidelines for HLA-peptide multimer assays derived from results of a large scale international proficiency panel of the Cancer Vaccine Consortium
- Author
-
Kunle Odunsi, Lloyd Old, Sylvia Janetzki, Michael Kalos, Leah Ben-Porat, Michael W. Pride, Pedro Romero, Holden T. Maecker, Axel Hoos, Timothy M. Clay, Cedrik M. Britten, HLA-peptide Multimer Proficiency Panel of the CVC-CRI Immune Assay Working Group, Ahmed, RK., Bain, C., Barnby-Porritt, H., Baumgartner, P., Bercovici, N., Brockstedt, D., Cerundolo, V., Channon, JY., Chikoti, P., Cox, J., Currier, JR., Curtsinger, J., Desmaretz, C., Gagnon, D., Gearhart, J., Gillanders, V., Hampl, J., Harrop, R., Hobeika, A., Kaempgen, E., Kaufman, J., Labroquere, K., Landry, C., Maeurer, M., Matijevic, M., Mueller, S., Mohanakumar, T., Olson, W., Pohla, H., Ramesh, R., Rooke, R., Samorski, R., Sékaly, RP., Schullery, D., Slingluff, C., Scheibenbogen, C., Schmitt-Händle, M., Smith, K., Speiser, D., Tarlton, A., Trautmann, L., Van Der Aa, A., Walter, S., Wolchok, J., and Yuan, J.
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,International Cooperation ,Immunology ,Harmonization ,Guidelines as Topic ,Tumor immunity ,Human leukocyte antigen ,Cancer Vaccines ,Internal medicine ,HLA-A2 Antigen ,Immunology and Allergy ,Medicine ,Humans ,Flow cytometry ,HLA-peptide multimer ,business.industry ,Clinical Laboratory Techniques ,Cancer ,Biological Assay ,Cancer Vaccines/immunology ,Clinical Laboratory Techniques/standards ,HLA-A2 Antigen/immunology ,HLA-A2 Antigen/metabolism ,Peptide Fragments/immunology ,Peptide Fragments/metabolism ,Protein Multimerization ,medicine.disease ,Protein multimerization ,Peptide Fragments ,CTL ,Original Article ,Cancer vaccine ,business - Abstract
Purpose The Cancer Vaccine Consortium of the Cancer Research Institute (CVC-CRI) conducted a multicenter HLA-peptide multimer proficiency panel (MPP) with a group of 27 laboratories to assess the performance of the assay. Experimental design Participants used commercially available HLA-peptide multimers and a well characterized common source of peripheral blood mononuclear cells (PBMC). The frequency of CD8+ T cells specific for two HLA-A2-restricted model antigens was measured by flow cytometry. The panel design allowed for participants to use their preferred staining reagents and locally established protocols for both cell labeling, data acquisition and analysis. Results We observed significant differences in both the performance characteristics of the assay and the reported frequencies of specific T cells across laboratories. These results emphasize the need to identify the critical variables important for the observed variability to allow for harmonization of the technique across institutions. Conclusions Three key recommendations emerged that would likely reduce assay variability and thus move toward harmonizing of this assay. (1) Use of more than two colors for the staining (2) collect at least 100,000 CD8 T cells, and (3) use of a background control sample to appropriately set the analytical gates. We also provide more insight into the limitations of the assay and identified additional protocol steps that potentially impact the quality of data generated and therefore should serve as primary targets for systematic analysis in future panels. Finally, we propose initial guidelines for harmonizing assay performance which include the introduction of standard operating protocols to allow for adequate training of technical staff and auditing of test analysis procedures. Electronic supplementary material The online version of this article (doi:10.1007/s00262-009-0681-z) contains supplementary material, which is available to authorized users.
- Published
- 2009
10. B cell receptor dependent enhancement of dengue virus infection.
- Author
-
Gebo C, Hardy CSC, McElvany BD, Graham NR, Lu JQ, Moradpour S, Currier JR, Friberg H, Gromowski GD, Thomas SJ, Chan GC, Diehl SA, and Waickman AT
- Abstract
Dengue virus (DENV) is the causative agent of dengue, a mosquito-borne disease that represents a significant and growing public health burden around the world. A unique pathophysiological feature of dengue is immune-mediated enhancement, wherein preexisting immunity elicited by a primary infection can enhance the severity of a subsequent infection by a heterologous DENV serotype. A leading mechanistic explanation for this phenomenon is antibody dependent enhancement (ADE), where sub-neutralizing concentrations of DENV-specific IgG antibodies facilitate entry of DENV into FcγR expressing cells such as monocytes, macrophages, and dendritic cells. Accordingly, this model posits that phagocytic mononuclear cells are the primary reservoir of DENV. However, analysis of samples from individuals experiencing acute DENV infection reveals that B cells are the largest reservoir of infected circulating cells, representing a disconnect in our understanding of immune-mediated DENV tropism. In this study, we demonstrate that the expression of a DENV-specific B cell receptor (BCR) renders cells highly susceptible to DENV infection, with the infection-enhancing activity of the membrane-restricted BCR correlating with the ADE potential of the IgG version of the antibody. In addition, we observed that the frequency of DENV-infectible B cells increases in previously flavivirus-naïve volunteers after a primary DENV infection. These findings suggest that BCR-dependent infection of B cells is a novel mechanism immune-mediated enhancement of DENV-infection., Competing Interests: The authors have declared that no competing interests exist., (Copyright: This is an open access article, free of all copyright, and may be freely reproduced, distributed, transmitted, modified, built upon, or otherwise used by anyone for any lawful purpose. The work is made available under the Creative Commons CC0 public domain dedication.)
- Published
- 2024
- Full Text
- View/download PDF
11. Long term T cell response and safety of a tetravalent dengue vaccine in healthy children.
- Author
-
Mandaric S, Friberg H, Saez-Llorens X, Borja-Tabora C, Biswal S, Escudero I, Faccin A, Gottardo R, Brose M, Roubinis N, Fladager D, DeAntonio R, Dimero JAL, Montenegro N, Folschweiller N, Currier JR, Sharma M, and Tricou V
- Abstract
As robust cellular responses are important for protection against dengue, this phase 2 study evaluated the kinetics and phenotype of T cell responses induced by TAK-003, a live-attenuated tetravalent dengue vaccine, in 4-16-year-old living in dengue-endemic countries (NCT02948829). Two hundred participants received TAK-003 on Days 1 and 90. Interferon-gamma (IFN-γ) enzyme-linked immunospot assay [ELISPOT] and intracellular cytokine staining were used to analyze T cell response and functionality, using peptide pools representing non-structural (NS) proteins NS3 and NS5 matching DENV-1, -2, -3, and -4 and DENV-2 NS1. One month after the second TAK-003 dose (Day 120), IFN-γ ELISPOT T cell response rates against any peptide pool were 97.1% (95% CI: 93.4% to 99.1%), and similar for baseline dengue seropositive (96.0%) and seronegative (98.6%) participants. IFN-γ ELISPOT T cell response rates at Day 120 were 79.8%, 90.2%, 77.3%, and 74.0%, against DENV-1, -2, -3, and -4, respectively, and remained elevated through 3 years post-vaccination. Multifunctional CD4 and CD8 T cell responses against DENV-2 NS peptides were observed, independent of baseline serostatus: CD8 T cells typically secreted IFN-γ and TNF-α whereas CD4 T cells secreted ≥ 2 of IFN-γ, IL-2 and TNF-α cytokines. NAb titers and seropositivity rates remained substantially elevated through 3 years post-vaccination. Overall, TAK-003 was well tolerated and elicited durable T cell responses against all four DENV serotypes irrespective of baseline serostatus in children and adolescents aged 4-16 years living in dengue-endemic countries. TAK-003-elicited CD4 and CD8 T cells were multifunctional and persisted up to 3 years post-vaccination., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
12. Efficacy and immunogenicity following dengue virus-1 human challenge after a tetravalent prime-boost dengue vaccine regimen: an open-label, phase 1 trial.
- Author
-
Lyke KE, Chua JV, Koren M, Friberg H, Gromowski GD, Rapaka RR, Waickman AT, Joshi S, Strauss K, McCracken MK, Gutierrez-Barbosa H, Shrestha B, Culbertson C, Bernal P, De La Barrera RA, Currier JR, Jarman RG, and Edelman R
- Subjects
- Humans, Adult, Male, Female, Young Adult, Adolescent, Middle Aged, Antibodies, Neutralizing blood, Vaccines, Attenuated immunology, Vaccines, Attenuated administration & dosage, Vaccines, Attenuated adverse effects, Vaccine Efficacy, Immunization, Secondary, Immunogenicity, Vaccine, Dengue Vaccines immunology, Dengue Vaccines adverse effects, Dengue Vaccines administration & dosage, Dengue prevention & control, Dengue immunology, Dengue Virus immunology, Antibodies, Viral blood
- Abstract
Background: Dengue human infection models (DHIMs) are important tools to down-select dengue vaccine candidates and establish tetravalent efficacy before advanced clinical field trials. We aimed to provide data for the safety and immunogenicity of DHIM and evaluate dengue vaccine efficacy., Methods: We performed an open-label, phase 1 trial at the University of Maryland (Baltimore, MD, USA). Eligible participants were healthy individuals aged 18-50 years who either previously received a tetravalent dengue purified inactivated vaccine prime followed by a live-attenuated vaccine boost (ie, the vaccinee group), or were unvaccinated flavivirus-naive participants (ie, the control group). Participants in the vaccinee group with detectable pre-challenge dengue virus-1 neutralising antibody titres and flavivirus-naive participants in the control group were inoculated with dengue virus-1 strain 45AZ5 in the deltoid region, 27-65 months following booster dosing. These participants were followed-up from days 4-16 following dengue virus-1 live virus human challenge, with daily real-time quantitative PCR specific to dengue virus-1 RNA detection, and dengue virus-1 solicited local and systemic adverse events were recorded. The primary outcomes were safety (ie, solicited local and systemic adverse events) and vaccine efficacy (ie, dengue virus-1 RNAaemia) following dengue challenge. This study is registered with ClinicalTrials.gov, number NCT04786457., Findings: In January 2021, ten eligible participants were enrolled; of whom, six (60%) were in the vaccinee group and four (40%) were in the control group. Daily quantitative PCR detected dengue virus-1 RNA in nine (90%) of ten participants (five [83%] of six in the vaccinee group and all four [100%] in the control group). The mean onset of RNAaemia occurred on day 5 (SD 1·0) in the vaccinee group versus day 8 (1·5) in the control group (95% CI 1·1-4·9; p=0·007), with a trend towards reduced RNAaemia duration in the vaccinee group compared with the control group (8·2 days vs 10·5 days; 95% CI -0·08 to 4·68; p=0·056). Mild-to-moderate symptoms (nine [90%] of ten), leukopenia (eight [89%] of nine), and elevated aminotransferases (seven [78%] of nine) were commonly observed. Severe adverse events were detected only in the vaccinee group (fever ≥38·9°C in three [50%] of six, headache in one [17%], and transient grade 4 aspartate aminotransferase elevation in one [17%]). No deaths were reported., Interpretation: Participants who had tetravalent dengue purified inactivated vaccine prime and live-attenuated vaccine boost were unprotected against dengue virus-1 infection and further showed increased clinical, immunological, and transcriptomic evidence for inflammation potentially mediated by pre-existing infection-enhancing antibodies. This study highlights the impact of small cohort, human challenge models studying dengue pathogenesis and downstream vaccine development., Funding: Military Infectious Disease Research Program and Medical Technology Enterprise Consortium and Advanced Technology International., Competing Interests: Declaration of interests KEL receives funding from the Gates Medical Research Institute and The Medical Technology Enterprise. JVC receives research funding from Gilead Sciences and BD Biosciences. RRR is funded by a Mentored Clinical Scientist Career Development Award from National Institute of Allergy and Infectious Diseases (K08AI143923) during the completion of this work, and is currently employed at Moderna and might own Moderna shares. ATW is founder and co-owner of Azimuth Biologics, and a compensated Scientific Advisory Board member for Takeda Pharmaceuticals. RE is a consultant to Takeda Vaccines. All other authors declare no competing interests., (Copyright © 2024 Elsevier Ltd. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
13. Low-dose dengue virus 3 human challenge model: a phase 1 open-label study.
- Author
-
Waickman AT, Newell K, Lu JQ, Fang H, Waldran M, Gebo C, Currier JR, Friberg H, Jarman RG, Klick MD, Ware LA, Endy TP, and Thomas SJ
- Subjects
- Humans, Adult, Male, Female, Young Adult, Cytokines blood, Cytokines metabolism, RNA, Viral blood, Seroconversion, Memory T Cells immunology, Middle Aged, Dengue Virus immunology, Dengue immunology, Dengue virology, Dengue Vaccines immunology, Dengue Vaccines administration & dosage, Dengue Vaccines adverse effects, Antibodies, Viral blood, Antibodies, Viral immunology, Viremia
- Abstract
Dengue human infection models present an opportunity to explore the potential of a vaccine, anti-viral or immuno-compound for clinical benefit in a controlled setting. Here we report the outcome of a phase 1 open-label assessment of a low-dose dengue virus 3 (DENV-3) challenge model (NCT04298138), in which nine participants received a subcutaneous inoculation with 0.5 ml of a 1.4 × 10
3 plaque-forming unit per ml suspension of the attenuated DENV-3 strain CH53489. The primary and secondary endpoints of the study were to assess the safety of this DENV-3 strain in healthy flavivirus-seronegative individuals. All participants developed RNAaemia within 7 days after inoculation with peak titre ranging from 3.13 × 104 to 7.02 × 108 genome equivalents per ml. Solicited symptoms such as fever and rash, clinical laboratory abnormalities such as lymphopenia and thrombocytopenia, and self-reported symptoms such as myalgia were consistent with mild-to-moderate dengue in all volunteers. DENV-3-specific seroconversion and memory T cell responses were observed within 14 days after inoculation as assessed by enzyme-linked immunosorbent assay and interferon-gamma-based enzyme-linked immunospot. RNA sequencing and serum cytokine analysis revealed anti-viral responses that overlapped with the period of viraemia. The magnitude and frequency of clinical and immunologic endpoints correlated with an individual's peak viral titre., (© 2024. The Author(s), under exclusive licence to Springer Nature Limited.)- Published
- 2024
- Full Text
- View/download PDF
14. Protective Role of NS1-Specific Antibodies in the Immune Response to Dengue Virus through Antibody-Dependent Cellular Cytotoxicity.
- Author
-
Sanchez-Vargas LA, Mathew A, Salje H, Sousa D, Casale NA, Farmer A, Buddhari D, Anderson K, Iamsirithaworn S, Kaewhiran S, Friberg H, Currier JR, and Rothman AL
- Abstract
Background: Dengue virus (DENV) non-structural protein 1 (NS1) has multiple functions within infected cells, on the cell surface, and in secreted form, and is highly immunogenic. Immunity from previous DENV infections is known to exert both positive and negative effects on subsequent DENV infections, but the contribution of NS1-specific antibodies to these effects is incompletely understood., Methods: We investigated the functions of NS1-specific antibodies and their significance in DENV infection. We analyzed plasma samples collected in a prospective cohort study prior to symptomatic or subclinical secondary DENV infection. We measured binding to purified recombinant NS1 protein and to NS1-expressing CEM cells, antibody-mediated NK cell activation by plate-bound NS1 protein, and antibody-dependent cellular cytotoxicity (ADCC) of NS1-expressing target cells., Results: We found that antibody responses to NS1 were highly serotype-cross-reactive and that subjects who experienced subclinical DENV infection had significantly higher antibody responses to NS1 in pre-infection plasma than subjects who experienced symptomatic infection. We observed strong positive correlations between antibody binding and NK activation., Conclusions: These findings demonstrate the involvement of NS1-specific antibodies in ADCC and provide evidence for a protective effect of NS1-specific antibodies in secondary DENV infection., (© The Author(s) 2024. Published by Oxford University Press on behalf of Infectious Diseases Society of America. All rights reserved. For commercial re-use, please contact reprints@oup.com for reprints and translation rights for reprints. All other permissions can be obtained through our RightsLink service via the Permissions link on the article page on our site—for further information please contact journals.permissions@oup.com.)
- Published
- 2024
- Full Text
- View/download PDF
15. Antibody targeting of conserved sites of vulnerability on the SARS-CoV-2 spike receptor-binding domain.
- Author
-
Sankhala RS, Dussupt V, Chen WH, Bai H, Martinez EJ, Jensen JL, Rees PA, Hajduczki A, Chang WC, Choe M, Yan L, Sterling SL, Swafford I, Kuklis C, Soman S, King J, Corbitt C, Zemil M, Peterson CE, Mendez-Rivera L, Townsley SM, Donofrio GC, Lal KG, Tran U, Green EC, Smith C, de Val N, Laing ED, Broder CC, Currier JR, Gromowski GD, Wieczorek L, Rolland M, Paquin-Proulx D, van Dyk D, Britton Z, Rajan S, Loo YM, McTamney PM, Esser MT, Polonis VR, Michael NL, Krebs SJ, Modjarrad K, and Joyce MG
- Subjects
- Humans, Angiotensin-Converting Enzyme 2 metabolism, Antibodies, Neutralizing chemistry, Antibodies, Viral chemistry, Binding Sites, Epitopes, SARS-CoV-2 metabolism, COVID-19
- Abstract
Given the continuous emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants of concern (VoCs), immunotherapeutics that target conserved epitopes on the spike (S) glycoprotein have therapeutic advantages. Here, we report the crystal structure of the SARS-CoV-2 S receptor-binding domain (RBD) at 1.95 Å and describe flexibility and distinct conformations of the angiotensin-converting enzyme 2 (ACE2)-binding site. We identify a set of SARS-CoV-2-reactive monoclonal antibodies (mAbs) with broad RBD cross-reactivity including SARS-CoV-2 Omicron subvariants, SARS-CoV-1, and other sarbecoviruses and determine the crystal structures of mAb-RBD complexes with Ab246 and CR3022 mAbs targeting the class IV site, WRAIR-2134, which binds the recently designated class V epitope, and WRAIR-2123, the class I ACE2-binding site. The broad reactivity of class IV and V mAbs to conserved regions of SARS-CoV-2 VoCs and other sarbecovirus provides a framework for long-term immunotherapeutic development strategies., Competing Interests: Declaration of interests Patent application number PCT/US 63/140,763 was filed containing mAbs described in this publication for authors S.J.K., K.M., V.D., S.M.T., G.D., and N.L.M. The status of the patent is pending, not yet published. M.G.J. and K.M. are named as inventors on International Patent Application No. WO 2021/178971 A1 entitled “Vaccines against SARS-CoV-2 and other coronaviruses.” M.G.J. is named as an inventor on International Patent Application No. WO/2018/081318 and U.S. patent 10,960,070 entitled “Prefusion Coronavirus S Proteins and Their Use.”, (Copyright © 2023 The Author(s). Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
16. Diverse array of neutralizing antibodies elicited upon Spike Ferritin Nanoparticle vaccination in rhesus macaques.
- Author
-
Sankhala RS, Lal KG, Jensen JL, Dussupt V, Mendez-Rivera L, Bai H, Wieczorek L, Mayer SV, Zemil M, Wagner DA, Townsley SM, Hajduczki A, Chang WC, Chen WH, Donofrio GC, Jian N, King HAD, Lorang CG, Martinez EJ, Rees PA, Peterson CE, Schmidt F, Hart TJ, Duso DK, Kummer LW, Casey SP, Williams JK, Kannan S, Slike BM, Smith L, Swafford I, Thomas PV, Tran U, Currier JR, Bolton DL, Davidson E, Doranz BJ, Hatziioannou T, Bieniasz PD, Paquin-Proulx D, Reiley WW, Rolland M, Sullivan NJ, Vasan S, Collins ND, Modjarrad K, Gromowski GD, Polonis VR, Michael NL, Krebs SJ, and Joyce MG
- Subjects
- Animals, Mice, Antibodies, Neutralizing, Macaca mulatta, Vaccination, Antibodies, Viral, Antibodies, Monoclonal, COVID-19 Vaccines, Ferritins, Spike Glycoprotein, Coronavirus genetics, Nanoparticles, Severe acute respiratory syndrome-related coronavirus
- Abstract
The repeat emergence of SARS-CoV-2 variants of concern (VoC) with decreased susceptibility to vaccine-elicited antibodies highlights the need to develop next-generation vaccine candidates that confer broad protection. Here we describe the antibody response induced by the SARS-CoV-2 Spike Ferritin Nanoparticle (SpFN) vaccine candidate adjuvanted with the Army Liposomal Formulation including QS21 (ALFQ) in non-human primates. By isolating and characterizing several monoclonal antibodies directed against the Spike Receptor Binding Domain (RBD), N-Terminal Domain (NTD), or the S2 Domain, we define the molecular recognition of vaccine-elicited cross-reactive monoclonal antibodies (mAbs) elicited by SpFN. We identify six neutralizing antibodies with broad sarbecovirus cross-reactivity that recapitulate serum polyclonal antibody responses. In particular, RBD mAb WRAIR-5001 binds to the conserved cryptic region with high affinity to sarbecovirus clades 1 and 2, including Omicron variants, while mAb WRAIR-5021 offers complete protection from B.1.617.2 (Delta) in a murine challenge study. Our data further highlight the ability of SpFN vaccination to stimulate cross-reactive B cells targeting conserved regions of the Spike with activity against SARS CoV-1 and SARS-CoV-2 variants., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
17. Priming with Japanese encephalitis virus or yellow fever virus vaccination led to the recognition of multiple flaviviruses without boosting antibody responses induced by an inactivated Zika virus vaccine.
- Author
-
Li Y, Merbah M, Wollen-Roberts S, Beckman B, Mdluli T, Curtis DJ, Currier JR, Mendez-Rivera L, Dussupt V, Krebs SJ, De La Barrera R, Michael NL, Paquin-Proulx D, Eller MA, Koren MA, Modjarrad K, and Rolland M
- Subjects
- Humans, Yellow fever virus, Vaccines, Inactivated, Antibody Formation, Receptors, IgG, Antibodies, Neutralizing, Antibodies, Viral, Vaccination, Antigens, Viral, Cross Reactions, Zika Virus, Flavivirus, Encephalitis Virus, Japanese, Zika Virus Infection prevention & control
- Abstract
Background: Complex patterns of cross-reactivity exist between flaviviruses, yet there is no precise understanding of how sequential exposures due to flavivirus infections or vaccinations impact subsequent antibody responses., Methods: We investigated whether B cell priming from Japanese encephalitis virus (JEV) or yellow fever virus (YFV) vaccination impacted binding and functional antibody responses to flaviviruses following vaccination with a Zika virus (ZIKV) purified inactivated virus (ZPIV) vaccine. Binding antibody responses and Fc gamma receptor engagement against 23 flavivirus antigens were characterized along with neutralization titres and Fc effector responses in 75 participants at six time points., Findings: We found no evidence that priming with JEV or YFV vaccines improved the magnitude of ZPIV induced antibody responses to ZIKV. Binding antibodies and Fc gamma receptor engagement to ZIKV antigens did not differ significantly across groups, while antibody-dependent cellular phagocytosis (ADCP) and neutralizing responses were higher in the naïve group than in the JEV and YFV primed groups following the second ZPIV immunization (p ≤ 0.02). After a third dose of ZPIV, ADCP responses remained higher in the naïve group than in the primed groups. However, priming affected the quality of the response following ZPIV vaccination, as primed individuals recognized a broader array of flavivirus antigens than individuals in the naïve group., Interpretation: While a priming vaccination to either JEV or YFV did not boost ZIKV-specific responses upon ZIKV vaccination, the qualitatively different responses elicited in the primed groups highlight the complexity in the cross-reactive antibody responses to flaviviruses., Funding: This work was supported by a cooperative agreement between The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., and the U.S. Department of the Army [W81XWH-18-2-0040]. The work was also funded in part by the National Institute of Allergy and Infectious Diseases (NIAID) R01AI155983 to SJK and KM., Competing Interests: Declaration of interests Kayvon Modjarrad is an employee of Pfizer. Rafael De La Barrera is one of the inventors for the WO2017210215A1 Patent: Zika virus vaccine and methods of production. All other authors declare that they have no competing interests. The views expressed are those of the authors and should not be construed to represent the positions of the U.S. Army, the Department of Defense, or the Department of Health and Human Services., (Copyright © 2023 Henry M Jackson Foundation for the Advancement of Military Medicine, Inc. Published by Elsevier B.V. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
18. Humoral immune responses associated with control of SARS-CoV-2 breakthrough infections in a vaccinated US military population.
- Author
-
Gromowski GD, Cincotta CM, Mayer S, King J, Swafford I, McCracken MK, Coleman D, Enoch J, Storme C, Darden J, Peel S, Epperson D, McKee K, Currier JR, Okulicz J, Paquin-Proulx D, Cowden J, and Peachman K
- Subjects
- Humans, COVID-19 Vaccines, SARS-CoV-2, Immunity, Humoral, Breakthrough Infections, Antibodies, Neutralizing, Antibodies, Viral, Vaccination, COVID-19, Military Personnel
- Abstract
Background: COVID-19 vaccines have been critical for protection against severe disease following infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) but gaps remain in our understanding of the immune responses that contribute to controlling subclinical and mild infections., Methods: Vaccinated, active-duty US military service members were enrolled in a non-interventional, minimal-risk, observational study starting in May, 2021. Clinical data, serum, and saliva samples were collected from study participants and were used to characterise the humoral immune responses to vaccination and to assess its impact on clinical and subclinical infections, as well as virologic outcomes of breakthrough infections (BTI) including viral load and infection duration., Findings: The majority of VIRAMP participants had received the Pfizer COVID-19 vaccine and by January, 2022, N = 149 had a BTI. The median BTI duration (PCR+ days) was 4 days and the interquartile range was 1-8 days. Participants that were nucleocapsid seropositive prior to their BTI had significantly higher levels of binding and functional antibodies to the spike protein, shorter median duration of infections, and lower median peak viral loads compared to seronegative participants. Furthermore, levels of neutralising antibody, ACE2 blocking activity, and spike-specific IgA measured prior to BTI also correlated with the duration of infection., Interpretation: We extended previous findings and demonstrate that a subset of vaccine-induced humoral immune responses, along with nucleocapsid serostatus are associated with control of SARS-CoV-2 breakthrough infections in the upper airways., Funding: This work was funded by the DoD Joint Program Executive Office for Chemical, Biological, Radiological and Nuclear Defense (JPEO-CBRND) in collaboration with the Defense Health Agency (DHA) COVID-19 funding initiative for the VIRAMP study., Competing Interests: Declaration of interests KM (Allucent) was contracted by the US Department of Defense to conduct the VIRAMP clinical study, to perform testing on clinical study samples, and to collect epidemiologic data. JC served as the Acting Chief Medical Officer for the Vaccine Acceleration Program at the Joint Program Executive Office for Chemical, Biological, Radiological and Nuclear Defense that funded the VIRAMP clinical study, and was involved with clinical study design and establishing collaborations for analysis of samples. The other authors declare no conflicts of interest., (Published by Elsevier B.V.)
- Published
- 2023
- Full Text
- View/download PDF
19. Soluble NS1 Antagonizes IgG- and IgA- Mediated Monocytic Phagocytosis of DENV Infected Cells.
- Author
-
Waldran MJ, Wegman AD, Bahr LE, Roy NH, Currier JR, and Waickman AT
- Subjects
- Humans, Phagocytosis, Immunoglobulin G, Immunoglobulin A metabolism, Viral Nonstructural Proteins metabolism, Antibodies, Viral, Dengue, Dengue Virus
- Abstract
Dengue virus (DENV) is endemic in >100 countries, infecting an estimated 400 million individuals every year. Infection with DENV raises an antibody response primarily targeting viral structural proteins. However, DENV encodes several immunogenic nonstructural (NS) proteins, one of which, NS1, is expressed on the membrane of DENV-infected cells. IgG and IgA isotype antibodies that bind NS1 are abundant in serum following DENV infection. Our study aimed to determine if NS1-binding IgG and IgA isotype antibodies contribute to the clearance of DENV-infected cells by antibody-mediated cellular phagocytosis. We observed that both IgG and IgA isotype antibodies can facilitate monocytic uptake of DENV NS1-expressing cells in an FcγRI- and FcαRI-dependent fashion. Interestingly, this process was antagonized by the presence of soluble NS1, suggesting that the production of soluble NS1 by infected cells may serve as immunological chaff, antagonizing opsonization and clearance of DENV-infected cells., Competing Interests: Potential conflicts of interest. A. T. W. is a co-founder of Azimuth Biologics, Inc. All other authors report no potential conflicts. All authors have submitted the ICMJE Form for Disclosure of Potential Conflicts of Interest. Conflicts that the editors consider relevant to the content of the manuscript have been disclosed., (© The Author(s) 2023. Published by Oxford University Press on behalf of Infectious Diseases Society of America. All rights reserved. For permissions, please e-mail: journals.permissions@oup.com.)
- Published
- 2023
- Full Text
- View/download PDF
20. SARS-CoV-2 spike-ferritin-nanoparticle adjuvanted with ALFQ induces long-lived plasma cells and cross-neutralizing antibodies.
- Author
-
Shrivastava S, Carmen JM, Lu Z, Basu S, Sankhala RS, Chen WH, Nguyen P, Chang WC, King J, Corbitt C, Mayer S, Bolton JS, Anderson A, Swafford I, Terriquez GD, Trinh HV, Kim J, Jobe O, Paquin-Proulx D, Matyas GR, Gromowski GD, Currier JR, Bergmann-Leitner E, Modjarrad K, Michael NL, Joyce MG, Malloy AMW, and Rao M
- Abstract
This study demonstrates the impact of adjuvant on the development of T follicular helper (Tfh) and B cells, and their influence on antibody responses in mice vaccinated with SARS-CoV-2-spike-ferritin-nanoparticle (SpFN) adjuvanted with either Army Liposome Formulation containing QS-21 (SpFN + ALFQ) or Alhydrogel
® (SpFN + AH). SpFN + ALFQ increased the size and frequency of germinal center (GC) B cells in the vaccine-draining lymph nodes and increased the frequency of antigen-specific naive B cells. A single vaccination with SpFN + ALFQ resulted in a higher frequency of IL-21-producing-spike-specific Tfh and GC B cells in the draining lymph nodes and spleen, S-2P protein-specific IgM and IgG antibodies, and elicitation of robust cross-neutralizing antibodies against SARS-CoV-2 variants as early as day 7, which was enhanced by a second vaccination. This was associated with the generation of high titer, high avidity binding antibodies. The third vaccination with SpFN + ALFQ elicited high levels of neutralizing antibodies against the Omicron variant. No cross-neutralizing antibodies against Omicron were induced with SpFN + AH. These findings highlight the importance of ALFQ in orchestrating early induction of antigen-specific Tfh and GC B cell responses and long-lived plasma cells in the bone marrow. The early engagement of S-2P specific naive B cells and high titer IgM antibodies shape the development of long-term neutralization breadth., (© 2023. This is a U.S. Government work and not under copyright protection in the US; foreign copyright protection may apply.)- Published
- 2023
- Full Text
- View/download PDF
21. Protection against SARS-CoV-2 Omicron BA.1 variant challenge in macaques by prime-boost vaccination with Ad26.COV2.S and SpFN.
- Author
-
Yu J, Thomas PV, McMahan K, Jacob-Dolan C, Liu J, He X, Hope D, Martinez EJ, Chen WH, Sciacca M, Hachmann NP, Lifton M, Miller J, Powers OC, Hall K, Wu C, Barrett J, Swafford I, Currier JR, King J, Corbitt C, Chang WC, Golub E, Rees PA, Peterson CE, Hajduczki A, Hussin E, Lange C, Gong H, Matyas GR, Rao M, Paquin-Proulx D, Gromowski GD, Lewis MG, Andersen H, Davis-Gardner M, Suthar MS, Michael NL, Bolton DL, Joyce MG, Modjarrad K, and Barouch DH
- Abstract
Emerging severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants and waning immunity call for next-generation vaccine strategies. Here, we assessed the immunogenicity and protective efficacy of two SARS-CoV-2 vaccines targeting the WA1/2020 spike protein, Ad26.COV2.S (Ad26) and Spike ferritin Nanoparticle (SpFN), in nonhuman primates, delivered as either a homologous (SpFN/SpFN and Ad26/Ad26) or heterologous (Ad26/SpFN) prime-boost regimen. The Ad26/SpFN regimen elicited the highest CD4 T cell and memory B cell responses, the SpFN/SpFN regimen generated the highest binding and neutralizing antibody responses, and the Ad26/Ad26 regimen generated the most robust CD8 T cell responses. Despite these differences, protective efficacy against SARS-CoV-2 Omicron BA.1 challenge was similar for all three regimens. After challenge, all vaccinated monkeys showed significantly reduced peak and day 4 viral loads in both bronchoalveolar lavage and nasal swabs as compared with sham animals. The efficacy conferred by these three immunologically distinct vaccine regimens suggests that both humoral and cellular immunity contribute to protection against SARS-CoV-2 Omicron challenge.
- Published
- 2022
- Full Text
- View/download PDF
22. Coronavirus Antibody Responses before COVID-19 Pandemic, Africa and Thailand.
- Author
-
Li Y, Merbah M, Wollen-Roberts S, Beckman B, Mdluli T, Swafford I, Mayer SV, King J, Corbitt C, Currier JR, Liu H, Esber A, Pinyakorn S, Parikh A, Francisco LV, Phanuphak N, Maswai J, Owuoth J, Kibuuka H, Iroezindu M, Bahemana E, Vasan S, Ake JA, Modjarrad K, Gromowski G, Paquin-Proulx D, and Rolland M
- Subjects
- Humans, Antibodies, Monoclonal, Antibodies, Viral, Antibody Formation, Immunoglobulin G, Nigeria, Nucleocapsid Proteins, Pandemics, SARS-CoV-2, Spike Glycoprotein, Coronavirus, Thailand epidemiology, Africa, COVID-19 epidemiology
- Abstract
Prior immune responses to coronaviruses might affect human SARS-CoV-2 response. We screened 2,565 serum and plasma samples collected from 2013 through early 2020, before the COVID-19 pandemic began, from 2,250 persons in 4 countries in Africa (Kenya, Nigeria, Tanzania, and Uganda) and in Thailand, including persons living with HIV-1. We detected IgG responses to SARS-CoV-2 spike (S) subunit 2 protein in 1.8% of participants. Profiling against 23 coronavirus antigens revealed that responses to S, subunit 2, or subunit 1 proteins were significantly more frequent than responses to the receptor-binding domain, S-Trimer, or nucleocapsid proteins (p<0.0001). We observed similar responses in persons with or without HIV-1. Among all coronavirus antigens tested, SARS-CoV-2, SARS-CoV-1, and Middle East respiratory syndrome coronavirus antibody responses were much higher in participants from Africa than in participants from Thailand (p<0.01). We noted less pronounced differences for endemic coronaviruses. Serosurveys could affect vaccine and monoclonal antibody distribution across global populations.
- Published
- 2022
- Full Text
- View/download PDF
23. Characterization of B-cell and T-cell responses to a tetravalent dengue purified inactivated vaccine in healthy adults.
- Author
-
Friberg H, Gargulak M, Kong A, Lin L, Martinez LJ, Schmidt AC, Paris RM, Jarman RG, Diaz C, Thomas SJ, Moris P, and Currier JR
- Abstract
The increasing global impact of dengue underscores the need for a dengue virus (DENV) vaccine. We assessed B-cell and T-cell responses following vaccination with four formulations of a tetravalent dengue purified inactivated vaccine (DPIV) in dengue-primed and dengue-naive adults from two studies (NCT01666652, NCT01702857). Frequencies of DPIV-induced memory B cells specific to each DENV serotype remained high up to 12 months post-vaccination, and were higher in the dengue-primed than dengue-naive adults. A subsequent DPIV booster dose induced strong anamnestic B-cell responses. Multifunctional CD4+ T cells (predominantly expressing IL-2) were induced by DPIV, with higher frequencies in dengue-primed adults. DPIV-induced CD4+ T cells also demonstrated in vitro proliferative capacity and antigen-specific production of GM-CSF, IFN-γ, and IL-13. CD8+ T-cell responses were undetectable in dengue-naive adults and low in dengue-primed individuals. B- and T-cell responses persisted up to 12 months post-vaccination in both dengue-primed and dengue-naive adults., (© 2022. This is a U.S. Government work and not under copyright protection in the US; foreign copyright protection may apply 2022.)
- Published
- 2022
- Full Text
- View/download PDF
24. Evolution of inflammation and immunity in a dengue virus 1 human infection model.
- Author
-
Waickman AT, Lu JQ, Fang H, Waldran MJ, Gebo C, Currier JR, Ware L, Van Wesenbeeck L, Verpoorten N, Lenz O, Tambuyzer L, Herrera-Taracena G, Van Loock M, Endy TP, and Thomas SJ
- Subjects
- Humans, Viremia, Immunoglobulin M, Immunoglobulin G, Immunoglobulin A, Antibodies, Viral, Dengue Virus genetics, Dengue
- Abstract
Dengue virus (DENV) infections are major causes of morbidity and mortality throughout the tropics and subtropics. More than 400 million infections are estimated to occur every year, resulting in nearly 100 million symptomatic infections and more than 20,000 deaths. Early immune response kinetics to infection remain unclear, in large part due to the variable incubation period exhibited by the DENVs after introduction into a susceptible host. To fill this knowledge gap, we performed a comprehensive virologic and immunologic analysis of individuals experimentally infected with the underattenuated DENV-1 strain 45AZ5. This analysis captured both the kinetics and composition of the innate, humoral, and cellular immune responses elicited by experimental DENV-1 infection, as well as virologic and clinical features. We observed a robust DENV-specific immunoglobulin A (IgA) antibody response that manifested between the appearance of DENV-specific IgM and IgG in all challenged individuals, as well as the presence of a non-neutralizing/NS1-specific antibody response that was delayed relative to the appearance of DENV virion-specific humoral immunity. RNA sequencing analysis revealed discrete and temporally restricted gene modules that correlated with acute viremia and the induction of adaptive immunity. Our analysis provides a detailed description, in time and space, of the evolving matrix of DENV-elicited human inflammation and immunity and reveals several previously unappreciated immunological aspects of primary DENV-1 infection that can inform countermeasure development and evaluation.
- Published
- 2022
- Full Text
- View/download PDF
25. Dengue virus downregulates TNFR1- and TLR3-stimulated NF-κB activation by targeting RIPK1.
- Author
-
Udawatte DJ, Lang DM, Currier JR, Medin CL, and Rothman AL
- Subjects
- Humans, NF-kappa B metabolism, Receptor-Interacting Protein Serine-Threonine Kinases metabolism, Signal Transduction, Toll-Like Receptor 3 metabolism, Dengue Virus, Receptors, Tumor Necrosis Factor, Type I metabolism
- Abstract
Dengue virus (DENV) infection is the most prevalent arthropod-borne virus disease and is endemic in more than 100 countries. Several DENV proteins have been shown to target crucial human host proteins to evade innate immune responses and establish a productive infection. Here we report that the DENV NS3 protein targets RIPK1 (Receptor Interacting Protein Kinase I), a central mediator of inflammation and cell death, and decreases intracellular RIPK1 levels during DENV infection. The interaction of NS3 with RIPK1 results in the inhibition of NF-κB activation in response to TNFR or TLR3 stimulation. Also, we observed that the effects of NS3 on RIPK1 were independent of NS3 protease activity. Our data demonstrate a novel mechanism by which DENV suppresses normal cellular functions to evade host innate immune responses., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2022 Udawatte, Lang, Currier, Medin and Rothman.)
- Published
- 2022
- Full Text
- View/download PDF
26. mRNA-1273 vaccination protects against SARS-CoV-2-elicited lung inflammation in nonhuman primates.
- Author
-
Waickman AT, Victor K, Newell K, Li T, Friberg H, Foulds KE, Roederer M, Bolton DL, Currier JR, and Seder R
- Subjects
- 2019-nCoV Vaccine mRNA-1273, Animals, Humans, Inflammation, Macaca mulatta, SARS-CoV-2, Spike Glycoprotein, Coronavirus genetics, Vaccination, COVID-19 prevention & control, Viral Vaccines
- Abstract
Vaccine-elicited SARS-CoV-2 antibody responses are an established correlate of protection against viral infection in humans and nonhuman primates. However, it is less clear that vaccine-induced immunity is able to limit infection-elicited inflammation in the lower respiratory tract. To assess this, we collected bronchoalveolar lavage fluid samples after SARS-CoV-2 strain USA-WA1/2020 challenge from rhesus macaques vaccinated with mRNA-1273 in a dose-reduction study. Single-cell transcriptomic profiling revealed a broad cellular landscape 48 hours after challenge, with distinct inflammatory signatures that correlated with viral RNA burden in the lower respiratory tract. These inflammatory signatures included phagocyte-restricted expression of chemokines, such as CXCL10 and CCL3, and the broad expression of IFN-induced genes, such as MX1, ISG15, and IFIT1. Induction of these inflammatory profiles was suppressed by prior mRNA-1273 vaccination in a dose-dependent manner and negatively correlated with prechallenge serum and lung antibody titers against SARS-CoV-2 spike. These observations were replicated and validated in a second independent macaque challenge study using the B.1.351/Beta variant of SARS-CoV-2. These data support a model wherein vaccine-elicited antibody responses restrict viral replication following SARS-CoV-2 exposure, including limiting viral dissemination to the lower respiratory tract and infection-mediated inflammation and pathogenesis.
- Published
- 2022
- Full Text
- View/download PDF
27. A SARS-CoV-2 Spike Ferritin Nanoparticle Vaccine Is Protective and Promotes a Strong Immunological Response in the Cynomolgus Macaque Coronavirus Disease 2019 (COVID-19) Model.
- Author
-
Johnston SC, Ricks KM, Lakhal-Naouar I, Jay A, Subra C, Raymond JL, King HAD, Rossi F, Clements TL, Fetterer D, Tostenson S, Cincotta CM, Hack HR, Kuklis C, Soman S, King J, Peachman KK, Kim D, Chen WH, Sankhala RS, Martinez EJ, Hajduczki A, Chang WC, Choe M, Thomas PV, Peterson CE, Anderson A, Swafford I, Currier JR, Paquin-Proulx D, Jagodzinski LL, Matyas GR, Rao M, Gromowski GD, Peel SA, White L, Smith JM, Hooper JW, Michael NL, Modjarrad K, Joyce MG, Nalca A, Bolton DL, and Pitt MLM
- Abstract
The COVID-19 pandemic has had a staggering impact on social, economic, and public health systems worldwide. Vaccine development and mobilization against SARS-CoV-2 (the etiologic agent of COVID-19) has been rapid. However, novel strategies are still necessary to slow the pandemic, and this includes new approaches to vaccine development and/or delivery that will improve vaccination compliance and demonstrate efficacy against emerging variants. Here, we report on the immunogenicity and efficacy of a SARS-CoV-2 vaccine comprising stabilized, pre-fusion spike protein trimers displayed on a ferritin nanoparticle (SpFN) adjuvanted with either conventional aluminum hydroxide or the Army Liposomal Formulation QS-21 (ALFQ) in a cynomolgus macaque COVID-19 model. Vaccination resulted in robust cell-mediated and humoral responses and a significant reduction in lung lesions following SARS-CoV-2 infection. The strength of the immune response suggests that dose sparing through reduced or single dosing in primates may be possible with this vaccine. Overall, the data support further evaluation of SpFN as a SARS-CoV-2 protein-based vaccine candidate with attention to fractional dosing and schedule optimization.
- Published
- 2022
- Full Text
- View/download PDF
28. A SARS-CoV-2 ferritin nanoparticle vaccine elicits protective immune responses in nonhuman primates.
- Author
-
Joyce MG, King HAD, Elakhal-Naouar I, Ahmed A, Peachman KK, Macedo Cincotta C, Subra C, Chen RE, Thomas PV, Chen WH, Sankhala RS, Hajduczki A, Martinez EJ, Peterson CE, Chang WC, Choe M, Smith C, Lee PJ, Headley JA, Taddese MG, Elyard HA, Cook A, Anderson A, McGuckin Wuertz K, Dong M, Swafford I, Case JB, Currier JR, Lal KG, Molnar S, Nair MS, Dussupt V, Daye SP, Zeng X, Barkei EK, Staples HM, Alfson K, Carrion R, Krebs SJ, Paquin-Proulx D, Karasavva N, Polonis VR, Jagodzinski LL, Amare MF, Vasan S, Scott PT, Huang Y, Ho DD, de Val N, Diamond MS, Lewis MG, Rao M, Matyas GR, Gromowski GD, Peel SA, Michael NL, Bolton DL, and Modjarrad K
- Subjects
- Animals, Antibodies, Neutralizing, Antibodies, Viral, COVID-19 Vaccines, Ferritins, Humans, Immunity, Macaca mulatta, SARS-CoV-2, Spike Glycoprotein, Coronavirus, COVID-19, Nanoparticles
- Abstract
The emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants stresses the continued need for next-generation vaccines that confer broad protection against coronavirus disease 2019 (COVID-19). We developed and evaluated an adjuvanted SARS-CoV-2 spike ferritin nanoparticle (SpFN) vaccine in nonhuman primates. High-dose (50 μg) SpFN vaccine, given twice 28 days apart, induced a Th1-biased CD4 T cell helper response and elicited neutralizing antibodies against SARS-CoV-2 wild-type and variants of concern, as well as against SARS-CoV-1. These potent humoral and cell-mediated immune responses translated into rapid elimination of replicating virus in the upper and lower airways and lung parenchyma of nonhuman primates following high-dose SARS-CoV-2 respiratory challenge. The immune response elicited by SpFN vaccination and resulting efficacy in nonhuman primates supports the utility of SpFN as a vaccine candidate for SARS-causing betacoronaviruses.
- Published
- 2022
- Full Text
- View/download PDF
29. SARS-CoV-2 ferritin nanoparticle vaccines elicit broad SARS coronavirus immunogenicity.
- Author
-
Joyce MG, Chen WH, Sankhala RS, Hajduczki A, Thomas PV, Choe M, Martinez EJ, Chang WC, Peterson CE, Morrison EB, Smith C, Chen RE, Ahmed A, Wieczorek L, Anderson A, Case JB, Li Y, Oertel T, Rosado L, Ganesh A, Whalen C, Carmen JM, Mendez-Rivera L, Karch CP, Gohain N, Villar Z, McCurdy D, Beck Z, Kim J, Shrivastava S, Jobe O, Dussupt V, Molnar S, Tran U, Kannadka CB, Soman S, Kuklis C, Zemil M, Khanh H, Wu W, Cole MA, Duso DK, Kummer LW, Lang TJ, Muncil SE, Currier JR, Krebs SJ, Polonis VR, Rajan S, McTamney PM, Esser MT, Reiley WW, Rolland M, de Val N, Diamond MS, Gromowski GD, Matyas GR, Rao M, Michael NL, and Modjarrad K
- Abstract
The need for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) next-generation vaccines has been highlighted by the rise of variants of concern (VoCs) and the long-term threat of emerging coronaviruses. Here, we design and characterize four categories of engineered nanoparticle immunogens that recapitulate the structural and antigenic properties of the prefusion SARS-CoV-2 spike (S), S1, and receptor-binding domain (RBD). These immunogens induce robust S binding, ACE2 inhibition, and authentic and pseudovirus neutralizing antibodies against SARS-CoV-2. A spike-ferritin nanoparticle (SpFN) vaccine elicits neutralizing titers (ID
50 > 10,000) following a single immunization, whereas RBD-ferritin nanoparticle (RFN) immunogens elicit similar responses after two immunizations and also show durable and potent neutralization against circulating VoCs. Passive transfer of immunoglobulin G (IgG) purified from SpFN- or RFN-immunized mice protects K18-hACE2 transgenic mice from a lethal SARS-CoV-2 challenge. Furthermore, S-domain nanoparticle immunization elicits ACE2-blocking activity and ID50 neutralizing antibody titers >2,000 against SARS-CoV-1, highlighting the broad response elicited by these immunogens., Competing Interests: Declaration of interests M.G.J. and K.M. are named as inventors on international patent application WO/2021/178971 A1 entitled “Vaccines against SARS-CoV-2 and other coronaviruses.” M.G.J. is named as an inventor on international patent application WO/2018/081318 and U.S. patent 10,960,070 entitled “Prefusion coronavirus spike proteins and their use.” Z.B. is named as an inventor on U.S. patent 10,434,167 entitled “Non-toxic adjuvant formulation comprising a monophosphoryl lipid A (MPLA)-containing liposome composition and a saponin.” Z.B. and G.R.M. are named as inventors on U.S. patent application 16/607,917 entitled “Compositions and methods for vaccine delivery.” M.S.D. is a consultant for Inbios, Vir Biotechnology, Fortress Biotech, and Carnival Corporation and on the scientific advisory boards of Moderna and Immunome. The Diamond laboratory has received funding support from sponsored research agreements from Moderna, Vir Biotechnology, Kaleido, and Emergent BioSolutions. S.R., P.M.M., and M.T.E. are employees of AstraZeneca and currently hold AstraZeneca stock or stock options. Z.B. is currently employed at Pfizer., (Copyright © 2021. Published by Elsevier Inc.)- Published
- 2021
- Full Text
- View/download PDF
30. SARS-CoV-2 ferritin nanoparticle vaccine induces robust innate immune activity driving polyfunctional spike-specific T cell responses.
- Author
-
Carmen JM, Shrivastava S, Lu Z, Anderson A, Morrison EB, Sankhala RS, Chen WH, Chang WC, Bolton JS, Matyas GR, Michael NL, Joyce MG, Modjarrad K, Currier JR, Bergmann-Leitner E, Malloy AMW, and Rao M
- Abstract
The emergence of variants of concern, some with reduced susceptibility to COVID-19 vaccines underscores consideration for the understanding of vaccine design that optimizes induction of effective cellular and humoral immune responses. We assessed a SARS-CoV-2 spike-ferritin nanoparticle (SpFN) immunogen paired with two distinct adjuvants, Alhydrogel
® or Army Liposome Formulation containing QS-21 (ALFQ) for unique vaccine evoked immune signatures. Recruitment of highly activated multifaceted antigen-presenting cells to the lymph nodes of SpFN+ALFQ vaccinated mice was associated with an increased frequency of polyfunctional spike-specific memory CD4+ T cells and Kb spike-(539-546)-specific long-lived memory CD8+ T cells with effective cytolytic function and distribution to the lungs. The presence of this epitope in SARS-CoV, suggests that generation of cross-reactive T cells may be induced against other coronavirus strains. Our study reveals that a nanoparticle vaccine, combined with a potent adjuvant that effectively engages innate immune cells, enhances SARS-CoV-2-specific durable adaptive immune T cell responses., (© 2021. This is a U.S. government work and not under copyright protection in the U.S.; foreign copyright protection may apply.)- Published
- 2021
- Full Text
- View/download PDF
31. Low-dose in vivo protection and neutralization across SARS-CoV-2 variants by monoclonal antibody combinations.
- Author
-
Dussupt V, Sankhala RS, Mendez-Rivera L, Townsley SM, Schmidt F, Wieczorek L, Lal KG, Donofrio GC, Tran U, Jackson ND, Zaky WI, Zemil M, Tritsch SR, Chen WH, Martinez EJ, Ahmed A, Choe M, Chang WC, Hajduczki A, Jian N, Peterson CE, Rees PA, Rutkowska M, Slike BM, Selverian CN, Swafford I, Teng IT, Thomas PV, Zhou T, Smith CJ, Currier JR, Kwong PD, Rolland M, Davidson E, Doranz BJ, Mores CN, Hatziioannou T, Reiley WW, Bieniasz PD, Paquin-Proulx D, Gromowski GD, Polonis VR, Michael NL, Modjarrad K, Joyce MG, and Krebs SJ
- Subjects
- Amino Acid Sequence, Animals, Antibodies, Monoclonal administration & dosage, Antibodies, Monoclonal metabolism, Antibodies, Neutralizing chemistry, Antibodies, Neutralizing metabolism, Antibodies, Viral immunology, Antibodies, Viral metabolism, Binding Sites genetics, COVID-19 metabolism, COVID-19 prevention & control, Disease Models, Animal, Dose-Response Relationship, Drug, Epitope Mapping, Epitopes chemistry, Epitopes immunology, Epitopes metabolism, Humans, Mice, Transgenic, Neutralization Tests, Protein Binding, Protein Conformation, SARS-CoV-2 genetics, SARS-CoV-2 metabolism, Sequence Homology, Amino Acid, Spike Glycoprotein, Coronavirus genetics, Spike Glycoprotein, Coronavirus metabolism, Survival Analysis, Antibodies, Monoclonal immunology, Antibodies, Neutralizing immunology, COVID-19 immunology, SARS-CoV-2 immunology, Spike Glycoprotein, Coronavirus immunology
- Abstract
Prevention of viral escape and increased coverage against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants of concern require therapeutic monoclonal antibodies (mAbs) targeting multiple sites of vulnerability on the coronavirus spike glycoprotein. Here we identify several potent neutralizing antibodies directed against either the N-terminal domain (NTD) or the receptor-binding domain (RBD) of the spike protein. Administered in combinations, these mAbs provided low-dose protection against SARS-CoV-2 infection in the K18-human angiotensin-converting enzyme 2 mouse model, using both neutralization and Fc effector antibody functions. The RBD mAb WRAIR-2125, which targets residue F486 through a unique heavy-chain and light-chain pairing, demonstrated potent neutralizing activity against all major SARS-CoV-2 variants of concern. In combination with NTD and other RBD mAbs, WRAIR-2125 also prevented viral escape. These data demonstrate that NTD/RBD mAb combinations confer potent protection, likely leveraging complementary mechanisms of viral inactivation and clearance., (© 2021. The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
32. Monomeric IgA Antagonizes IgG-Mediated Enhancement of DENV Infection.
- Author
-
Wegman AD, Fang H, Rothman AL, Thomas SJ, Endy TP, McCracken MK, Currier JR, Friberg H, Gromowski GD, and Waickman AT
- Subjects
- Animals, Antibodies, Monoclonal metabolism, Antibodies, Neutralizing metabolism, Antibodies, Viral metabolism, Antibody Specificity, Chlorocebus aethiops, Dengue metabolism, Dengue virology, Dengue Virus metabolism, Dengue Virus pathogenicity, Host-Pathogen Interactions, Humans, Immunity, Humoral, Immunoglobulin A metabolism, Immunoglobulin Class Switching, Immunoglobulin G metabolism, K562 Cells, Vero Cells, Antibodies, Monoclonal immunology, Antibodies, Neutralizing immunology, Antibodies, Viral immunology, Antibody-Dependent Enhancement, Dengue immunology, Dengue Virus immunology, Immunoglobulin A immunology, Immunoglobulin G immunology, Opsonization
- Abstract
Dengue virus (DENV) is a prevalent human pathogen, infecting approximately 400 million individuals per year and causing symptomatic disease in approximately 100 million. A distinct feature of dengue is the increased risk for severe disease in some individuals with preexisting DENV-specific immunity. One proposed mechanism for this phenomenon is antibody-dependent enhancement (ADE), in which poorly-neutralizing IgG antibodies from a prior infection opsonize DENV to increase infection of F
c gamma receptor-bearing cells. While IgM and IgG are the most commonly studied DENV-reactive antibody isotypes, our group and others have described the induction of DENV-specific serum IgA responses during dengue. We hypothesized that monomeric IgA would be able to neutralize DENV without the possibility of ADE. To test this, we synthesized IgG and IgA versions of two different DENV-reactive monoclonal antibodies. We demonstrate that isotype-switching does not affect the antigen binding and neutralization properties of the two mAbs. We show that DENV-reactive IgG, but not IgA, mediates ADE in Fc gamma receptor-positive K562 cells. Furthermore, we show that IgA potently antagonizes the ADE activity of IgG. These results suggest that levels of DENV-reactive IgA induced by DENV infection might regulate the overall IgG mediated ADE activity of DENV-immune plasma in vivo , and may serve as a predictor of disease risk., Competing Interests: Authors ADW, MM, JC, HF, GG, and ATW are co-inventors on the provisional patent “IgA monoclonal antibodies as a prophylactic and therapeutic treatment for acute flavivirus infection”. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2021 Wegman, Fang, Rothman, Thomas, Endy, McCracken, Currier, Friberg, Gromowski and Waickman.)- Published
- 2021
- Full Text
- View/download PDF
33. A SARS-CoV-2 spike ferritin nanoparticle vaccine protects hamsters against Alpha and Beta virus variant challenge.
- Author
-
Wuertz KM, Barkei EK, Chen WH, Martinez EJ, Lakhal-Naouar I, Jagodzinski LL, Paquin-Proulx D, Gromowski GD, Swafford I, Ganesh A, Dong M, Zeng X, Thomas PV, Sankhala RS, Hajduczki A, Peterson CE, Kuklis C, Soman S, Wieczorek L, Zemil M, Anderson A, Darden J, Hernandez H, Grove H, Dussupt V, Hack H, de la Barrera R, Zarling S, Wood JF, Froude JW, Gagne M, Henry AR, Mokhtari EB, Mudvari P, Krebs SJ, Pekosz AS, Currier JR, Kar S, Porto M, Winn A, Radzyminski K, Lewis MG, Vasan S, Suthar M, Polonis VR, Matyas GR, Boritz EA, Douek DC, Seder RA, Daye SP, Rao M, Peel SA, Joyce MG, Bolton DL, Michael NL, and Modjarrad K
- Abstract
The emergence of SARS-CoV-2 variants of concern (VOC) requires adequate coverage of vaccine protection. We evaluated whether a SARS-CoV-2 spike ferritin nanoparticle vaccine (SpFN), adjuvanted with the Army Liposomal Formulation QS21 (ALFQ), conferred protection against the Alpha (B.1.1.7), and Beta (B.1.351) VOCs in Syrian golden hamsters. SpFN-ALFQ was administered as either single or double-vaccination (0 and 4 week) regimens, using a high (10 μg) or low (0.2 μg) dose. Animals were intranasally challenged at week 11. Binding antibody responses were comparable between high- and low-dose groups. Neutralizing antibody titers were equivalent against WA1, B.1.1.7, and B.1.351 variants following two high dose vaccinations. Dose-dependent SpFN-ALFQ vaccination protected against SARS-CoV-2-induced disease and viral replication following intranasal B.1.1.7 or B.1.351 challenge, as evidenced by reduced weight loss, lung pathology, and lung and nasal turbinate viral burden. These data support the development of SpFN-ALFQ as a broadly protective, next-generation SARS-CoV-2 vaccine., (© 2021. This is a U.S. government work and not under copyright protection in the U.S.; foreign copyright protection may apply.)
- Published
- 2021
- Full Text
- View/download PDF
34. Non-structural protein 1-specific antibodies directed against Zika virus in humans mediate antibody-dependent cellular cytotoxicity.
- Author
-
Sanchez Vargas LA, Adam A, Masterson M, Smith M, Lyski ZL, Dowd KA, Pierson TC, Messer WB, Currier JR, and Mathew A
- Subjects
- Cells, Cultured, Cross Reactions immunology, Humans, Leukocytes, Mononuclear immunology, Leukocytes, Mononuclear virology, Viral Vaccines immunology, Zika Virus Infection virology, Antibodies, Neutralizing immunology, Antibodies, Viral immunology, Antibody-Dependent Cell Cytotoxicity immunology, Viral Nonstructural Proteins immunology, Zika Virus immunology, Zika Virus Infection immunology
- Abstract
There is growing interest in understanding antibody (Ab) function beyond neutralization. The non-structural protein 1 (NS1) of Zika virus (ZIKV) is an attractive candidate for an effective vaccine as Abs against NS1, unlike the envelope or premembrane, do not carry the risk of mediating antibody-dependent enhancement. Our aim was to evaluate whether ZIKV NS1 Abs elicited following natural infection in humans can mediate antibody-dependent cellular cytotoxicity (ADCC). We evaluated the isotype specificity of ZIKV-specific Abs in immune sera and supernatants from stimulated immune PBMC and found that Abs against ZIKV NS1 and virus-like particles were predominantly of the IgG1 isotype. Using a recently developed FluoroSpot assay, we found robust frequencies of NS1-specific Ab-secreting cells in PBMC of individuals who were naturally infected with ZIKV. We developed assays to measure both natural killer cell activation by flow cytometry and target cell lysis of ZIKV NS1-expressing cells using an image cytometry assay in the presence of ZIKV NS1 Abs. Our data indicate efficient opsonization of ZIKV NS1-expressing CEM-NK
R cell lines using ZIKV-immune but not ZIKV-naïve sera, a prerequisite of ADCC. Furthermore, sera from immune donors were able to induce both NK cell degranulation and lysis of ZIKV NS1 CEM-NKR cells in vitro. Our data suggest that ADCC is a possible mechanism for ZIKV NS1 Abs to eliminate virally infected target cells., (© 2021 The Authors. Immunology published by John Wiley & Sons Ltd.)- Published
- 2021
- Full Text
- View/download PDF
35. Efficacy and breadth of adjuvanted SARS-CoV-2 receptor-binding domain nanoparticle vaccine in macaques.
- Author
-
King HAD, Joyce MG, Lakhal-Naouar I, Ahmed A, Cincotta CM, Subra C, Peachman KK, Hack HR, Chen RE, Thomas PV, Chen WH, Sankhala RS, Hajduczki A, Martinez EJ, Peterson CE, Chang WC, Choe M, Smith C, Headley JA, Elyard HA, Cook A, Anderson A, Wuertz KM, Dong M, Swafford I, Case JB, Currier JR, Lal KG, Amare MF, Dussupt V, Molnar S, Daye SP, Zeng X, Barkei EK, Alfson K, Staples HM, Carrion R, Krebs SJ, Paquin-Proulx D, Karasavvas N, Polonis VR, Jagodzinski LL, Vasan S, Scott PT, Huang Y, Nair MS, Ho DD, de Val N, Diamond MS, Lewis MG, Rao M, Matyas GR, Gromowski GD, Peel SA, Michael NL, Modjarrad K, and Bolton DL
- Subjects
- Adjuvants, Immunologic administration & dosage, Animals, Antibodies, Neutralizing biosynthesis, Antibodies, Neutralizing immunology, Antibodies, Viral biosynthesis, Antibodies, Viral immunology, COVID-19 prevention & control, COVID-19 Vaccines immunology, Ferritins chemistry, SARS-CoV-2 metabolism, T-Lymphocytes immunology, COVID-19 virology, COVID-19 Vaccines administration & dosage, Macaca mulatta immunology, Nanoparticles chemistry, Receptors, Virus metabolism, SARS-CoV-2 immunology
- Abstract
Emergence of novel variants of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) underscores the need for next-generation vaccines able to elicit broad and durable immunity. Here we report the evaluation of a ferritin nanoparticle vaccine displaying the receptor-binding domain of the SARS-CoV-2 spike protein (RFN) adjuvanted with Army Liposomal Formulation QS-21 (ALFQ). RFN vaccination of macaques using a two-dose regimen resulted in robust, predominantly Th1 CD4+ T cell responses and reciprocal peak mean serum neutralizing antibody titers of 14,000 to 21,000. Rapid control of viral replication was achieved in the upper and lower airways of animals after high-dose SARS-CoV-2 respiratory challenge, with undetectable replication within 4 d in seven of eight animals receiving 50 µg of RFN. Cross-neutralization activity against SARS-CoV-2 variant B.1.351 decreased only approximately twofold relative to WA1/2020. In addition, neutralizing, effector antibody and cellular responses targeted the heterotypic SARS-CoV-1, highlighting the broad immunogenicity of RFN-ALFQ for SARS-CoV-like Sarbecovirus vaccine development., Competing Interests: Competing interest statement: M.G.J. and K.M. are named as inventors on International Patent Application no. WO/2021/21405 entitled “Vaccines against SARS-CoV-2 and other coronaviruses.” M.G.J. is named as an inventor on International Patent Application no. WO/2018/081318 entitled “Prefusion Coronavirus Spike Proteins and Their Use.” M.S.D. is a consultant for Inbios, Vir Biotechnology, Fortress Biotech, and Carnival Corporation and is on the Scientific Advisory Boards of Moderna and Immunome. The M.S.D. laboratory has received funding support in sponsored research agreements from Moderna, Vir Biotechnology, and Emergent BioSolutions., (Copyright © 2021 the Author(s). Published by PNAS.)
- Published
- 2021
- Full Text
- View/download PDF
36. Stochastic Model of the Adaptive Immune Response Predicts Disease Severity and Captures Enhanced Cross-Reactivity in Natural Dengue Infections.
- Author
-
Nguyen HD, Chaudhury S, Waickman AT, Friberg H, Currier JR, and Wallqvist A
- Subjects
- Child, Dengue diagnosis, Dengue virology, Dengue Virus growth & development, Dengue Virus pathogenicity, Host-Pathogen Interactions, Humans, Kinetics, Patient Acuity, Serogroup, Stochastic Processes, Viral Load, Adaptive Immunity, Antibodies, Viral immunology, Cross Reactions, Dengue immunology, Dengue Virus immunology, Models, Immunological
- Abstract
The dengue virus circulates as four distinct serotypes, where a single serotype infection is typically asymptomatic and leads to acquired immunity against that serotype. However, the developed immunity to one serotype is thought to underlie the severe manifestation of the disease observed in subsequent infections from a different serotype. We developed a stochastic model of the adaptive immune response to dengue infections. We first delineated the mechanisms initiating and sustaining adaptive immune responses during primary infections. We then contrasted these immune responses during secondary infections of either a homotypic or heterotypic serotype to understand the role of pre-existing and reactivated immune pathways on disease severity. Comparison of non-symptomatic and severe cases from heterotypic infections demonstrated that overproduction of specific antibodies during primary infection induces an enhanced population of cross-reactive antibodies during secondary infection, ultimately leading to severe disease manifestations. In addition, the level of disease severity was found to correlate with immune response kinetics, which was dependent on beginning lymphocyte levels. Our results detail the contribution of specific lymphocytes and antibodies to immunity and memory recall that lead to either protective or pathological outcomes, allowing for the understanding and determination of mechanisms of protective immunity., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2021 Nguyen, Chaudhury, Waickman, Friberg, Currier and Wallqvist.)
- Published
- 2021
- Full Text
- View/download PDF
37. Longitudinal Analysis of Dengue Virus-Specific Memory T Cell Responses and Their Association With Clinical Outcome in Subsequent DENV Infection.
- Author
-
Sanchez-Vargas LA, Anderson KB, Srikiatkhachorn A, Currier JR, Friberg H, Endy TP, Fernandez S, Mathew A, and Rothman AL
- Subjects
- Adolescent, Child, Cytokines biosynthesis, Dengue etiology, Female, Humans, Longitudinal Studies, Male, Severity of Illness Index, Dengue immunology, Dengue Virus immunology, Memory T Cells immunology
- Abstract
Memory T cells resulting from primary dengue virus (DENV) infection are hypothesized to influence the clinical outcome of subsequent DENV infection. However, the few studies involving prospectively collected blood samples have found weak and inconsistent associations with outcome and variable temporal trends in DENV-specific memory T cell responses between subjects. This study used both ex-vivo and cultured ELISPOT assays to further evaluate the associations between DENV serotype-cross-reactive memory T cells and severity of secondary infection. Using ex-vivo ELISPOT assays, frequencies of memory T cells secreting IFN-γ in response to DENV structural and non-structural peptide pools were low in PBMC from multiple time points prior to symptomatic secondary DENV infection and showed a variable response to infection. There were no differences in responses between subjects who were not hospitalized (NH, n=6) and those who were hospitalized with dengue hemorrhagic fever (hDHF, n=4). In contrast, responses in cultured ELISPOT assays were more reliably detectable prior to secondary infection and showed more consistent increases after infection. Responses in cultured ELISPOT assays were higher in individuals with hDHF (n=8) compared to NH (n=9) individuals before the secondary infection, with no difference between these groups after infection. These data demonstrate an association of pre-existing DENV-specific memory responses with the severity of illness in subsequent DENV infection, and suggest that frequencies of DENV-reactive T cells measured after short-term culture may be of particular importance for assessing the risk for more severe dengue disease., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2021 Sanchez-Vargas, Anderson, Srikiatkhachorn, Currier, Friberg, Endy, Fernandez, Mathew and Rothman.)
- Published
- 2021
- Full Text
- View/download PDF
38. A SARS-CoV-2 spike ferritin nanoparticle vaccine protects against heterologous challenge with B.1.1.7 and B.1.351 virus variants in Syrian golden hamsters.
- Author
-
Wuertz KM, Barkei EK, Chen WH, Martinez EJ, Lakhal-Naouar I, Jagodzinski LL, Paquin-Proulx D, Gromowski GD, Swafford I, Ganesh A, Dong M, Zeng X, Thomas PV, Sankhala RS, Hajduczki A, Peterson CE, Kuklis C, Soman S, Wieczorek L, Zemil M, Anderson A, Darden J, Hernandez H, Grove H, Dussupt V, Hack H, de la Barrera R, Zarling S, Wood JF, Froude JW, Gagne M, Henry AR, Mokhtari EB, Mudvari P, Krebs SJ, Pekosz AS, Currier JR, Kar S, Porto M, Winn A, Radzyminski K, Lewis MG, Vasan S, Suthar M, Polonis VR, Matyas GR, Boritz EA, Douek DC, Seder RA, Daye SP, Rao M, Peel SA, Joyce MG, Bolton DL, Michael NL, and Modjarrad K
- Abstract
The emergence of SARS-CoV-2 variants of concern (VOC) requires adequate coverage of vaccine protection. We evaluated whether a spike ferritin nanoparticle vaccine (SpFN), adjuvanted with the Army Liposomal Formulation QS21 (ALFQ), conferred protection against the B.1.1.7 and B.1.351 VOCs in Syrian golden hamsters. SpFN-ALFQ was administered as either single or double-vaccination (0 and 4 week) regimens, using a high (10 μg) or low (0.2 μg) immunogen dose. Animals were intranasally challenged at week 11. Binding antibody responses were comparable between high- and low-dose groups. Neutralizing antibody titers were equivalent against WA1, B.1.1.7, and B.1.351 variants following two high dose two vaccinations. SpFN-ALFQ vaccination protected against SARS-CoV-2-induced disease and viral replication following intranasal B.1.1.7 or B.1.351 challenge, as evidenced by reduced weight loss, lung pathology, and lung and nasal turbinate viral burden. These data support the development of SpFN-ALFQ as a broadly protective, next-generation SARS-CoV-2 vaccine.
- Published
- 2021
- Full Text
- View/download PDF
39. Immunogenicity of a Live-Attenuated Dengue Vaccine Using a Heterologous Prime-Boost Strategy in a Phase 1 Randomized Clinical Trial.
- Author
-
Lin L, Koren MA, Paolino KM, Eckels KH, De La Barrera R, Friberg H, Currier JR, Gromowski GD, Aronson NE, Keiser PB, Sklar MJ, Sondergaard EL, Jasper LE, Endy TP, Jarman RG, and Thomas SJ
- Subjects
- Antibodies, Neutralizing blood, Antibodies, Viral blood, Humans, Vaccines, Attenuated immunology, Vaccines, Combined immunology, Dengue prevention & control, Dengue Vaccines immunology, Immunogenicity, Vaccine
- Abstract
Background: Dengue is a global health problem and the development of a tetravalent dengue vaccine with durable protection is a high priority. A heterologous prime-boost strategy has the advantage of eliciting immune responses through different mechanisms and therefore may be superior to homologous prime-boost strategies for generating durable tetravalent immunity., Methods: In this phase 1 first-in-human heterologous prime-boost study, 80 volunteers were assigned to 4 groups and received a tetravalent dengue virus (DENV-1-4) purified inactivated vaccine (TDENV-PIV) with alum adjuvant and a tetravalent dengue virus (DENV-1-4) live attenuated vaccine (TDENV-LAV) in different orders and dosing schedules (28 or 180 days apart)., Results: All vaccination regimens had acceptable safety profiles and there were no vaccine-related serious adverse events. TDEN-PIV followed by TDEN-LAV induced higher neutralizing antibody titers and a higher rate of tetravalent seroconversions compared to TDEN-LAV followed by TDEN-PIV. Both TDEN-PIV followed by TDEN-LAV groups demonstrated 100% tetravalent seroconversion 28 days following the booster dose, which was maintained for most of these subjects through the day 180 measurement., Conclusions: A heterologous prime-boost vaccination strategy for dengue merits additional evaluation for safety, immunogenicity, and potential for clinical benefit., Clinical Trials Registration: NCT02239614., (Published by Oxford University Press for the Infectious Diseases Society of America 2020.)
- Published
- 2021
- Full Text
- View/download PDF
40. Enhanced dengue vaccine virus replication and neutralizing antibody responses in immune primed rhesus macaques.
- Author
-
McCracken MK, Kuklis CH, Kannadka CB, Barvir DA, Sanborn MA, Waickman AT, Siegfried HC, Victor KA, Hatch KL, De La Barrera R, Walls SD, Rutvisuttinunt W, Currier JR, Friberg H, Jarman RG, and Gromowski GD
- Abstract
Antibody-dependent enhancement (ADE) is suspected to influence dengue virus (DENV) infection, but the role ADE plays in vaccination strategies incorporating live attenuated virus components is less clear. Using a heterologous prime-boost strategy in rhesus macaques, we examine the effect of priming with DENV purified inactivated vaccines (PIVs) on a tetravalent live attenuated vaccine (LAV). Sera exhibited low-level neutralizing antibodies (NAb) post PIV priming, yet moderate to high in vitro ADE activity. Following LAV administration, the PIV primed groups exhibited DENV-2 LAV peak viremias up to 1,176-fold higher than the mock primed group, and peak viremia correlated with in vitro ADE. Furthermore, PIV primed groups had more balanced and higher DENV-1-4 NAb seroconversion and titers than the mock primed group following LAV administration. These results have implications for the development of effective DENV vaccine prime-boost strategies and for our understanding of the role played by ADE in modulating DENV replication.
- Published
- 2021
- Full Text
- View/download PDF
41. Persistent COVID-19 Symptoms Minimally Impact the Development of SARS-CoV-2-Specific T Cell Immunity.
- Author
-
Fang H, Wegman AD, Ripich K, Friberg H, Currier JR, Thomas SJ, Endy TP, and Waickman AT
- Subjects
- Adult, Aged, Aged, 80 and over, Antibodies, Viral immunology, COVID-19 immunology, COVID-19 metabolism, Coronavirus 229E, Human immunology, Cross Reactions, Epitopes, T-Lymphocyte immunology, Female, Humans, Male, Middle Aged, New York epidemiology, SARS-CoV-2 pathogenicity, T-Lymphocytes immunology, Post-Acute COVID-19 Syndrome, COVID-19 complications, Immunity, Cellular immunology, SARS-CoV-2 immunology
- Abstract
SARS-CoV-2 represents an unprecedented public health challenge. While the majority of SARS-CoV-2-infected individuals with mild-to-moderate COVID-19 resolve their infection with few complications, some individuals experience prolonged symptoms lasting for weeks after initial diagnosis. Persistent viral infections are commonly accompanied by immunologic dysregulation, but it is unclear if persistent COVID-19 impacts the development of virus-specific cellular immunity. To this end, we analyzed SARS-CoV-2-specific cellular immunity in convalescent COVID-19 patients who experienced eight days or fewer of COVID-19 symptoms or symptoms persisting for 18 days or more. We observed that persistent COVID-19 symptoms were not associated with the development of an overtly dysregulated cellular immune response. Furthermore, we observed that reactivity against the N protein from SARS-CoV-2 correlates with the amount of reactivity against the seasonal human coronaviruses 229E and NL63. These results provide insight into the processes that regulate the development of cellular immunity against SARS-CoV-2 and related human coronaviruses.
- Published
- 2021
- Full Text
- View/download PDF
42. Efficacy of a Broadly Neutralizing SARS-CoV-2 Ferritin Nanoparticle Vaccine in Nonhuman Primates.
- Author
-
Joyce MG, King HAD, Naouar IE, Ahmed A, Peachman KK, Cincotta CM, Subra C, Chen RE, Thomas PV, Chen WH, Sankhala RS, Hajduczki A, Martinez EJ, Peterson CE, Chang WC, Choe M, Smith C, Lee PJ, Headley JA, Taddese MG, Elyard HA, Cook A, Anderson A, McGuckin-Wuertz K, Dong M, Swafford I, Case JB, Currier JR, Lal KG, O'Connell RJ, Molnar S, Nair MS, Dussupt V, Daye SP, Zeng X, Barkei EK, Staples HM, Alfson K, Carrion R, Krebs SJ, Paquin-Proulx D, Karasavva N, Polonis VR, Jagodzinski LL, Amare MF, Vasan S, Scott PT, Huang Y, Ho DD, de Val N, Diamond MS, Lewis MG, Rao M, Matyas GR, Gromowski GD, Peel SA, Michael NL, Bolton DL, and Modjarrad K
- Abstract
The emergence of novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants stresses the continued need for next-generation vaccines that confer broad protection against coronavirus disease 2019 (COVID-19). We developed and evaluated an adjuvanted SARS-CoV-2 Spike Ferritin Nanoparticle (SpFN) vaccine in nonhuman primates (NHPs). High-dose (50 µ g) SpFN vaccine, given twice within a 28 day interval, induced a Th1-biased CD4 T cell helper response and a peak neutralizing antibody geometric mean titer of 52,773 against wild-type virus, with activity against SARS-CoV-1 and minimal decrement against variants of concern. Vaccinated animals mounted an anamnestic response upon high-dose SARS-CoV-2 respiratory challenge that translated into rapid elimination of replicating virus in their upper and lower airways and lung parenchyma. SpFN's potent and broad immunogenicity profile and resulting efficacy in NHPs supports its utility as a candidate platform for SARS-like betacoronaviruses., One-Sentence Summary: A SARS-CoV-2 Spike protein ferritin nanoparticle vaccine, co-formulated with a liposomal adjuvant, elicits broad neutralizing antibody responses that exceed those observed for other major vaccines and rapidly protects against respiratory infection and disease in the upper and lower airways and lung tissue of nonhuman primates.
- Published
- 2021
- Full Text
- View/download PDF
43. A Phase 1, Open-Label Assessment of a Dengue Virus-1 Live Virus Human Challenge Strain.
- Author
-
Endy TP, Wang D, Polhemus ME, Jarman RG, Jasper LE, Gromowski G, Lin L, De La Barra RA, Friberg H, Currier JR, Abbott M, Ware L, Klick M, Paolino KM, Blair DC, Eckels K, Rutvisuttinunt W, and Thomas SJ
- Subjects
- Dengue immunology, Dengue virology, Dengue Vaccines administration & dosage, Dengue Vaccines adverse effects, Healthy Volunteers, Humans, Outcome Assessment, Health Care, Vaccination, Vaccines, Virus-Like Particle administration & dosage, Vaccines, Virus-Like Particle adverse effects, Viremia immunology, Viremia prevention & control, Viremia virology, Dengue prevention & control, Dengue Vaccines immunology, Dengue Virus immunology, Vaccines, Virus-Like Particle immunology
- Abstract
Background: Dengue human infection models (DHIM) have been used as a safe means to test the viability of prophylaxis and therapeutics., Methods: A phase 1 study of 12 healthy adult volunteers using a challenge virus, DENV-1-LVHC strain 45AZ5, was performed. A dose escalating design was used to determine the safety and performance profile of the challenge virus. Subjects were evaluated extensively until 28 days and then out to 6 months., Results: Twelve subjects received the challenge virus: 6 with 0.5 mL of 6.5 × 103 plaque-forming units (PFU)/mL (low-dose group) and 6 with 0.5 mL of 6.5 × 104 PFU/mL (mid-dose group). All except 1 in the low-dose group developed detectable viremia. For all subjects the mean incubation period was 5.9 days (range 5-9 days) and mean time of viremia was 6.8 days (range 3-9 days). Mean peak for all subjects was 1.6 × 107 genome equivalents (GE)/mL (range 4.6 × 103 to 5 × 107 GE/mL). There were no serious adverse events or long-term safety signals noted., Conclusions: We conclude that DENV-1-LVHC was well-tolerated, resulted in an uncomplicated dengue illness, and may be a suitable DHIM for therapeutic and prophylactic product testing., Clinical Trials Registration: NCT02372175., (© The Author(s) 2020. Published by Oxford University Press for the Infectious Diseases Society of America. All rights reserved. For permissions, e-mail: journals.permissions@oup.com.)
- Published
- 2021
- Full Text
- View/download PDF
44. Temporally integrated single cell RNA sequencing analysis of PBMC from experimental and natural primary human DENV-1 infections.
- Author
-
Waickman AT, Friberg H, Gromowski GD, Rutvisuttinunt W, Li T, Siegfried H, Victor K, McCracken MK, Fernandez S, Srikiatkhachorn A, Ellison D, Jarman RG, Thomas SJ, Rothman AL, Endy T, and Currier JR
- Subjects
- Animals, Cell Line, Dengue virology, Humans, Immunity genetics, Inflammation genetics, Inflammation virology, Leukocytes, Mononuclear immunology, Leukocytes, Mononuclear virology, Sequence Analysis, RNA, Single-Cell Analysis, Dengue immunology, Dengue Virus immunology, Gene Expression Regulation
- Abstract
Dengue human infection studies present an opportunity to address many longstanding questions in the field of flavivirus biology. However, limited data are available on how the immunological and transcriptional response elicited by an attenuated challenge virus compares to that associated with a wild-type DENV infection. To determine the kinetic transcriptional signature associated with experimental primary DENV-1 infection and to assess how closely this profile correlates with the transcriptional signature accompanying natural primary DENV-1 infection, we utilized scRNAseq to analyze PBMC from individuals enrolled in a DENV-1 human challenge study and from individuals experiencing a natural primary DENV-1 infection. While both experimental and natural primary DENV-1 infection resulted in overlapping patterns of inflammatory gene upregulation, natural primary DENV-1 infection was accompanied with a more pronounced suppression in gene products associated with protein translation and mitochondrial function, principally in monocytes. This suggests that the immune response elicited by experimental and natural primary DENV infection are similar, but that natural primary DENV-1 infection has a more pronounced impact on basic cellular processes to induce a multi-layered anti-viral state., Competing Interests: I have read the journal's policy and the authors of this manuscript have the following competing interests:A.T.W reports grants from Military Infectious Disease Research Program, during the conduct of the study. A.L.R. reports grants from National Institute of Allergy and Infectious Diseases, during the conduct of the study. S.J.T reports other support from US DoD, other support from GSK, during the conduct of the study; personal fees and other support from GSK Vaccines, personal fees and other support from Takeda, personal fees and other support from Merck, personal fees and other support from PrimeVax, personal fees and other support from Themisbio, personal fees and other support from Chugai Pharma, personal fees and other support from Cormac Life Sciences, personal fees and other support from HHS NVPO / Tunnel Govt Services, personal fees and other support from Janssen, other support from GreenMark Partners. In addition, S.J.T has a patent US10086061B2 (combined flavivirus vaccines) issued. J.R.C reports grants from the Congressionally Directed Medical Research Program during the conduct of the study. All other authors have nothing to disclose.
- Published
- 2021
- Full Text
- View/download PDF
45. Next-generation sequencing of 11 HLA loci in a large dengue vaccine cohort from the Philippines.
- Author
-
Geretz A, Cofer L, Ehrenberg PK, Currier JR, Yoon IK, Alera MTP, Jarman R, Rothman AL, and Thomas R
- Subjects
- Alleles, Clinical Trials, Phase III as Topic, Exons genetics, Gene Frequency genetics, Genotype, Genotyping Techniques methods, High-Throughput Nucleotide Sequencing methods, Humans, Introns genetics, Philippines, Prospective Studies, Dengue genetics, Dengue immunology, Dengue Vaccines immunology, Genetic Loci genetics, HLA Antigens genetics
- Abstract
HLA genotyping by next-generation sequencing (NGS) has evolved with significant advancements in the last decade. Here we describe full-length HLA genotyping of 11 loci in 612 individuals comprising a dengue vaccine cohort from Cebu province in the Philippines. The multi-locus individual tagging NGS (MIT-NGS) method that we developed initially for genotyping 4-6 loci in one MiSeq run was expanded to 11 loci including HLA-A, B, C, DPA1, DPB1, DQA1, DQB1, DRB1, and DRB3/4/5. This change did not affect the overall coverage or depth of the sequencing reads. HLA alleles with frequencies greater than 10% were A*11:01:01, A*24:02:01, A*24:07:01, A*34:01:01, B*38:02:01, B*15:35, B*35:05:01, C*07:02:01, C*04:01:01, DPA1*02:02:02, DPB1*05:01:01, DPB1*01:01:01, DQA1*01:02:01, DQA1*06:01:01, DQB1*05:02:01, DQB1*03:01:01, DRB1*15:02:01, DRB1*12:02:01, DRB3*03:01:03, DRB4*01:03:01, and DRB5*01:01:01. Improvements in sequencing library preparation provide uniform and even coverage across all exons and introns. This has led to a marked reduction in allele imbalance and dropout. Furthermore, including more loci, such as DRB3/4/5, decreases cross-mapping and incorrect allele assignment at the DRB1 locus. The increased number of loci sequenced for each sample does not reduce the number of samples that can be multiplexed on a single MiSeq run and is therefore more cost-efficient. We believe that such improvements will help HLA genotyping by NGS to gain momentum over other conventional methods by increasing confidence in the calls., Competing Interests: Declaration of Competing Interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2020 American Society for Histocompatibility and Immunogenetics. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
46. Safety and Immunogenicity of an AS03 B -Adjuvanted Inactivated Tetravalent Dengue Virus Vaccine Administered on Varying Schedules to Healthy U.S. Adults: A Phase 1/2 Randomized Study.
- Author
-
Lin L, Lyke KE, Koren M, Jarman RG, Eckels KH, Lepine E, McArthur MA, Currier JR, Friberg H, Moris P, Keiser PB, De La Barrera R, Vaughn DW, Paris RM, Thomas SJ, and Schmidt AC
- Subjects
- Adult, Dengue immunology, Dengue virology, Dengue Vaccines adverse effects, Dengue Vaccines biosynthesis, Female, Healthy Volunteers, Humans, Immunogenicity, Vaccine, Male, Middle Aged, Patient Safety, Vaccines, Attenuated, Vaccines, Subunit, Antibodies, Neutralizing biosynthesis, Antibodies, Viral biosynthesis, Dengue prevention & control, Dengue Vaccines administration & dosage, Dengue Virus immunology, Vaccination methods
- Abstract
Dengue disease and its causative agents, the dengue viruses (DENV-1-4), cause high morbidity in tropical and subtropical regions. We evaluated three dosing regimens of the investigational tetravalent AS03
B -adjuvanted dengue-purified inactivated vaccine (DPIV+AS03B ). In this phase 1/2, observer-blind, placebo-controlled study (NCT02421367), 140 healthy adults were randomized 1:1:2 to receive DPIV+AS03B according to the following regimens: 0-1 month (M), 0-1-6 M, or 0-3 M. Participants received DPIV+AS03B or placebo at M0, M1, M3, and M6 according to their dosing schedule. Primary objectives were 1) to evaluate the safety of DPIV+AS03B for 28 days (D) after each dose; 2) to demonstrate the added value of a booster dose (0-1-6 M versus 0-1 M) based on neutralizing antibody titers to each DENV type (DENV-1-4) at 28 D after the last dose; and, if this objective was met, 3) to demonstrate the benefit of a longer interval between the first and second doses (0-1 M versus 0-3 M). Adverse events (AEs) within 7 D after vaccination tended to be more frequent after DPIV+AS03B doses than placebo; the number of grade 3 AEs was low (≤ 4.5% after DPIV+AS03B ; ≤ 2.9% after placebo), with no obvious differences across groups. Within 28 D following each dose, the frequency of unsolicited AEs after DPIV+AS03B appeared higher for three-dose (0-1-6 M) than two-dose (0-1 M and 0-3 M) regimens. No serious AEs were considered related to vaccination, and no potential immune-mediated diseases were reported during the study. All three schedules were well tolerated. Both primary immunogenicity objectives were demonstrated. The 0-3 M and 0-1-6 M regimens were more immunogenic than the 0-1 M regimen.- Published
- 2020
- Full Text
- View/download PDF
47. Analysis of cell-associated DENV RNA by oligo(dT) primed 5' capture scRNAseq.
- Author
-
Sanborn MA, Li T, Victor K, Siegfried H, Fung C, Rothman AL, Srikiatkhachorn A, Fernandez S, Ellison D, Jarman RG, Friberg H, Maljkovic Berry I, Currier JR, and Waickman AT
- Subjects
- B-Lymphocytes metabolism, DNA Primers genetics, DNA, Complementary genetics, Dengue Virus genetics, Dengue Virus pathogenicity, Dengue Virus physiology, Genome, Viral genetics, Humans, Leukocytes, Mononuclear metabolism, Oligodeoxyribonucleotides genetics, RNA, Viral genetics, Dengue genetics, Sequence Analysis, RNA methods
- Abstract
Dengue is one of the most widespread vector-borne viral diseases in the world. However, the size, heterogeneity, and temporal dynamics of the cell-associated viral reservoir during acute dengue virus (DENV) infection remains unclear. In this study, we analyzed cells infected in vitro with DENV and PBMC from an individual experiencing a natural DENV infection utilizing 5' capture single cell RNA sequencing (scRNAseq). Both positive- and negative-sense DENV RNA was detected in reactions containing either an oligo(dT) primer alone, or in reactions supplemented with a DENV-specific primer. The addition of a DENV-specific primer did not increase the total amount of DENV RNA captured or the fraction of cells identified as containing DENV RNA. However, inclusion of a DENV-specific cDNA primer did increase the viral genome coverage immediately 5' to the primer binding site. Furthermore, while the majority of intracellular DENV sequence captured in this analysis mapped to the 5' end of the viral genome, distinct patterns of enhanced coverage within the DENV polyprotein coding region were observed. The 5' capture scRNAseq analysis of PBMC not only recapitulated previously published reports by detecting virally infected memory and naïve B cells, but also identified cell-associated genomic variants not observed in contemporaneous serum samples. These results demonstrate that oligo(dT) primed 5' capture scRNAseq can detect DENV RNA and quantify virus-infected cells in physiologically relevant conditions, and provides insight into viral sequence variability within infected cells.
- Published
- 2020
- Full Text
- View/download PDF
48. Route of inoculation and mosquito vector exposure modulate dengue virus replication kinetics and immune responses in rhesus macaques.
- Author
-
McCracken MK, Gromowski GD, Garver LS, Goupil BA, Walker KD, Friberg H, Currier JR, Rutvisuttinunt W, Hinton KL, Christofferson RC, Mores CN, Vanloubbeeck Y, Lorin C, Malice MP, Thomas SJ, Jarman RG, Vaughn DW, Putnak JR, and Warter L
- Subjects
- Animals, Dengue virology, Dengue Virus physiology, Disease Models, Animal, Female, Kinetics, Macaca mulatta immunology, Mosquito Vectors virology, RNA, Viral blood, Salivary Glands virology, Vaccination, Viral Load, Viremia prevention & control, Virus Replication, Antibodies, Neutralizing blood, Antibodies, Viral blood, Dengue immunology
- Abstract
Dengue virus (DENV) is transmitted by infectious mosquitoes during blood-feeding via saliva containing biologically-active proteins. Here, we examined the effect of varying DENV infection modality in rhesus macaques in order to improve the DENV nonhuman primate (NHP) challenge model. NHPs were exposed to DENV-1 via subcutaneous or intradermal inoculation of virus only, intradermal inoculation of virus and salivary gland extract, or infectious mosquito feeding. The infectious mosquito feeding group exhibited delayed onset of viremia, greater viral loads, and altered clinical and immune responses compared to other groups. After 15 months, NHPs in the subcutaneous and infectious mosquito feeding groups were re-exposed to either DENV-1 or DENV-2. Viral replication and neutralizing antibody following homologous challenge were suggestive of sterilizing immunity, whereas heterologous challenge resulted in productive, yet reduced, DENV-2 replication and boosted neutralizing antibody. These results show that a more transmission-relevant exposure modality resulted in viral replication closer to that observed in humans., Competing Interests: I have read the journal's policy and the authors of this manuscript have the following competing interests: Yannick Vanloubbeeck, Clarisse Lorin, Marie-Pierre Malice, David Vaughn, and Lucile Warter are employees of the GSK group of companies.
- Published
- 2020
- Full Text
- View/download PDF
49. Transcriptional and clonal characterization of B cell plasmablast diversity following primary and secondary natural DENV infection.
- Author
-
Waickman AT, Gromowski GD, Rutvisuttinunt W, Li T, Siegfried H, Victor K, Kuklis C, Gomootsukavadee M, McCracken MK, Gabriel B, Mathew A, Grinyo I Escuer A, Fouch ME, Liang J, Fernandez S, Davidson E, Doranz BJ, Srikiatkhachorn A, Endy T, Thomas SJ, Ellison D, Rothman AL, Jarman RG, Currier JR, and Friberg H
- Subjects
- B-Lymphocytes cytology, Cells, Cultured, Dengue genetics, Humans, Immunity, Humoral, Immunoglobulins metabolism, RNA-Seq, Single-Cell Analysis, Transcriptome, B-Lymphocytes immunology, Dengue immunology, Immunoglobulins genetics
- Abstract
Antibody-mediated humoral immunity is thought to play a central role in mediating the immunopathogenesis of acute DENV infection, but limited data are available on the diversity, specificity, and functionality of the antibody response at the molecular level elicited by primary or secondary DENV infection. In order to close this functional gap in our understanding of DENV-specific humoral immunity, we utilized high-throughput single cell RNA sequencing to investigate B cells circulating in both primary and secondary natural DENV infections. We captured full-length paired immunoglobulin receptor sequence data from 9,027 B cells from a total of 6 subjects, including 2,717 plasmablasts. In addition to IgG and IgM class-switched cells, we unexpectedly found a high proportion of the DENV-elicited plasmablasts expressing IgA, principally in individuals with primary DENV infections. These IgA class-switched cells were extensively hypermutated even in individuals with a serologically confirmed primary DENV infection. Utilizing a combination of conventional biochemical assays and high-throughput shotgun mutagenesis, we determined that DENV-reactive IgA class-switched antibodies represent a significant fraction of DENV-reactive Igs generated in response to DENV infection, and that they exhibit a comparable epitope specificity to DENV-reactive IgG antibodies. These results provide insight into the molecular-level diversity of DENV-elicited humoral immunity and identify a heretofore unappreciated IgA plasmablast response to DENV infection., Competing Interests: Declaration of Competing Interest A.T.W reports grants from Military Infectious Disease Research Program, during the conduct of the study. B.J.D, E.D., M.E.F, A.G., J.L., and A.L.R. reports grants from National Institute of Allergy and Infectious Diseases, during the conduct of the study. B.J.D. reports other support from Integral Molecular, outside the submitted work. S.J.T reports other support from US DoD, other support from GSK, during the conduct of the study; personal fees and other support from GSK Vaccines, personal fees and other support from Takeda, personal fees and other support from Merck, personal fees and other support from PrimeVax, personal fees and other support from Themisbio, personal fees and other support from Chugai Pharma, personal fees and other support from Cormac Life Sciences, personal fees and other support from HHS NVPO / Tunnel Govt Services, personal fees and other support from Janssen, other support from GreenMark Partners, personal fees from Tremeau Pharma, outside the submitted work; In addition, Dr. Thomas has a patent US10086061B2 (combined flavivirus vaccines) issued. All other authors have nothing to disclose., (Copyright © 2020 The Authors. Published by Elsevier B.V. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
50. The Effects of Japanese Encephalitis Vaccine and Accelerated Dosing Scheduling on the Immunogenicity of the Chimeric Yellow Fever Derived Tetravalent Dengue Vaccine: A Phase II, Randomized, Open-Label, Single-Center Trial in Adults Aged 18 to 45 Years in the United States.
- Author
-
Glass A, Polhemus M, Wang D, Jarman RG, Thomas SJ, Friberg H, Currier JR, Bonaparte M, De La Barra R, Princiotta MF, Abbott M, Cuzzo B, Machabert T, Sridhar S, and Endy TP
- Subjects
- Adolescent, Adult, Dengue Vaccines adverse effects, Dengue Vaccines immunology, Female, Humans, Immunization Schedule, Immunophenotyping, Japanese Encephalitis Vaccines adverse effects, Japanese Encephalitis Vaccines immunology, Male, Middle Aged, Young Adult, Antibodies, Neutralizing blood, Antibodies, Viral blood, Dengue Vaccines administration & dosage, Japanese Encephalitis Vaccines administration & dosage
- Abstract
Background: Dengue is a global health problem requiring an effective, safe dengue vaccine., Methods: We report the results of a phase II, randomized, open-label, single-center trial in adults aged 18 to 45 years in the United States designed to explore the effects of the Chimeric Yellow Fever Derived Tetravalent Dengue Vaccine (CYD-TDV, Dengvaxia) when administered on its designated schedule (months 0, 6, and 12) or on an accelerated dosing schedule (months 0, 2, and 6) and/or given before, or concomitantly with, a vaccine against Japanese encephalitis (JE)., Results: Based on dengue virus serotype-specific neutralizing antibody (NAb), the accelerated dosing schedule was comparable to the 0, 6, and 12-month schedule. Giving JE vaccine concurrently with CYD-TDV did not result in an increase in overall NAb titers. Immunophenotyping of peripheral blood mononuclear cells revealed an increase in activated CD8+ T cells after CYD-TDV vaccination, a phenomenon that was greatest for the JE vaccine primed., Conclusions: We conclude that an accelerated dosing schedule of CYD-TDV results in essentially equivalent dengue serotype-specific NAb titers as the currently used schedule, and there may be an early benefit in antibody titers and activated CD8+ T cells by the administration of the JE vaccine before CYD-TDV vaccination., (© The Author(s) 2019. Published by Oxford University Press for the Infectious Diseases Society of America. All rights reserved. For permissions, e-mail: journals.permissions@oup.com.)
- Published
- 2020
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.