72 results on '"Dussupt, V"'
Search Results
2. Assessment of mumps virus growth on various continuous cell lines by virological, immunological, molecular and morphological investigations
- Author
-
Afzal, M.A., Dussupt, V., Minor, P.D., Pipkin, P.A., Fleck, R., Hockley, D.J., and Stacey, G.N.
- Published
- 2005
- Full Text
- View/download PDF
3. Crystal structure of a human anti-ZIKV-DENV neutralizing antibody MZ24 isolated following ZPIV vaccination
- Author
-
Sankhala, R.S., primary, Dussupt, V., additional, Donofrio, G., additional, Choe, M., additional, Modjarrad, K., additional, Michael, N.L., additional, Krebs, S.J., additional, and Joyce, M.G., additional
- Published
- 2019
- Full Text
- View/download PDF
4. Crystal structure of a human anti-ZIKV-DENV neutralizing antibody MZ1 in complex with ZIKV E glycoprotein
- Author
-
Sankhala, R.S., primary, Dussupt, V., additional, Donofrio, G., additional, Choe, M., additional, Modjarrad, K., additional, Michael, N.L., additional, Krebs, S.J., additional, and Joyce, M.G., additional
- Published
- 2019
- Full Text
- View/download PDF
5. Evaluating Baculovirus as a Vector for Human Prostate Cancer Gene Therapy
- Author
-
Swift, S.L. (Stephanie), Rivera, G.C. (Guillermo), Dussupt, V. (Vincent), Leadley, R.M. (Regina), Hudson, L.C. (Lucy), Ridder, C.M.A. (Corrina) de, Kraaij, R. (Robert), Burns, J.E. (Julie), Maitland, N.J. (Norman), Georgopoulos, L.J. (Lindsay), Swift, S.L. (Stephanie), Rivera, G.C. (Guillermo), Dussupt, V. (Vincent), Leadley, R.M. (Regina), Hudson, L.C. (Lucy), Ridder, C.M.A. (Corrina) de, Kraaij, R. (Robert), Burns, J.E. (Julie), Maitland, N.J. (Norman), and Georgopoulos, L.J. (Lindsay)
- Abstract
Gene therapy represents an attractive strategy for the non-invasive treatment of prostate cancer, where current clinical interventions show limited efficacy. Here, we evaluate the use of the insect virus, baculovirus (BV), as a novel vector for human prostate cancer gene therapy. Since prostate tumours represent a heterogeneous environment, a therapeutic approach that achieves long-term regression must be capable of targeting multiple transformed cell populations. Furthermore, discrimination in the targeting of malignant compared to non-malignant cells would have value in minimising side effects. We employed a number of prostate cancer models to analyse the potential for BV to achieve these goals. In vitro, both traditional prostate cell lines as well as primary epithelial or stromal cells derived from patient prostate biopsies, in two- or three-dimensional cultures, were used. We also evaluated BV in vivo in murine prostate cancer xenograft models. BV was capable of preferentially transducing invasive malignant prostate cancer cell lines compared to early stage cancers and non-malignant samples, a restriction that was not a function of nuclear import. Of more clinical relevance, primary patient-derived prostate cancer cells were also efficiently transduced by BV, with robust rates observed in epithelial cells of basal phenotype, which expressed BV-encoded transgenes faster than epithelial cells of a more differentiated, luminal phenotype. Maximum transduction capacity was observed in stromal cells. BV was able to penetrate through three-dimensional structures, including in vitro spheroids and in vivo orthotopic xenografts. BV vectors containing a nitroreductase transgene in a gene-directed enzyme pro-drug therapy approach were capable of efficiently killing malignant prostate targets following administration of the pro-drug, CB1954. Thus, BV is capable of transducing a large proportion of prostate cell types within a heterogeneous 3-D prostate tumour, can facilitat
- Published
- 2013
- Full Text
- View/download PDF
6. Evaluating Baculovirus as a Vector for Human Prostate Cancer Gene Therapy
- Author
-
Swift, SL, Rivera, GC, Dussupt, V, Leadley, RM, Hudson, LC, de Ridder, Corrina, Kraaij, Robert, Burns, JE, Maitland, NJ, Georgopoulos, LJ, Swift, SL, Rivera, GC, Dussupt, V, Leadley, RM, Hudson, LC, de Ridder, Corrina, Kraaij, Robert, Burns, JE, Maitland, NJ, and Georgopoulos, LJ
- Abstract
Gene therapy represents an attractive strategy for the non-invasive treatment of prostate cancer, where current clinical interventions show limited efficacy. Here, we evaluate the use of the insect virus, baculovirus (BV), as a novel vector for human prostate cancer gene therapy. Since prostate tumours represent a heterogeneous environment, a therapeutic approach that achieves long-term regression must be capable of targeting multiple transformed cell populations. Furthermore, discrimination in the targeting of malignant compared to non-malignant cells would have value in minimising side effects. We employed a number of prostate cancer models to analyse the potential for BV to achieve these goals. In vitro, both traditional prostate cell lines as well as primary epithelial or stromal cells derived from patient prostate biopsies, in two-or three-dimensional cultures, were used. We also evaluated BV in vivo in murine prostate cancer xenograft models. BV was capable of preferentially transducing invasive malignant prostate cancer cell lines compared to early stage cancers and non-malignant samples, a restriction that was not a function of nuclear import. Of more clinical relevance, primary patient-derived prostate cancer cells were also efficiently transduced by BV, with robust rates observed in epithelial cells of basal phenotype, which expressed BV-encoded transgenes faster than epithelial cells of a more differentiated, luminal phenotype. Maximum transduction capacity was observed in stromal cells. BV was able to penetrate through three-dimensional structures, including in vitro spheroids and in vivo orthotopic xenografts. BV vectors containing a nitroreductase transgene in a gene-directed enzyme pro-drug therapy approach were capable of efficiently killing malignant prostate targets following administration of the pro-drug, CB1954. Thus, BV is capable of transducing a large proportion of prostate cell types within a heterogeneous 3-D prostate tumour, can facilitate
- Published
- 2013
7. Targeting Gene Therapy for Prostate Cancer
- Author
-
Maitland, N., primary, Stanbridge, L., additional, and Dussupt, V., additional
- Published
- 2004
- Full Text
- View/download PDF
8. Baculoviruses as vectors for gene therapy against human prostate cancer.
- Author
-
Stanbridge LJ, Dussupt V, and Maitland NJ
- Published
- 2003
9. SARS-CoV-2 ferritin nanoparticle vaccines produce hyperimmune equine sera with broad sarbecovirus activity.
- Author
-
Martinez EJ, Chang WC, Chen WH, Hajduczki A, Thomas PV, Jensen JL, Choe M, Sankhala RS, Peterson CE, Rees PA, Kimner J, Soman S, Kuklis C, Mendez-Rivera L, Dussupt V, King J, Corbett C, Mayer SV, Fernandes A, Murzello K, Cookenham T, Hvizdos J, Kummer L, Hart T, Lanzer K, Gambacurta J, Reagan M, Duso D, Vasan S, Collins ND, Michael NL, Krebs SJ, Gromowski GD, Modjarrad K, Kaundinya J, and Joyce MG
- Abstract
The rapid emergence of SARS-CoV-2 variants of concern (VoC) and the threat of future zoonotic sarbecovirus spillover emphasizes the need for broadly protective next-generation vaccines and therapeutics. We utilized SARS-CoV-2 spike ferritin nanoparticle (SpFN), and SARS-CoV-2 receptor binding domain ferritin nanoparticle (RFN) immunogens, in an equine model to elicit hyperimmune sera and evaluated its sarbecovirus neutralization and protection capacity. Immunized animals rapidly elicited sera with the potent neutralization of SARS-CoV-2 VoC, and SARS-CoV-1 pseudoviruses, and potent binding against receptor binding domains from sarbecovirus clades 1b, 1a, 2, 3, and 4. Purified equine polyclonal IgG provided protection against Omicron XBB.1.5 virus in the K18-hACE2 transgenic mouse model. These results suggest that SARS-CoV-2-based nanoparticle vaccines can rapidly produce a broad and protective sarbecovirus response in the equine model and that equine serum has therapeutic potential against emerging SARS-CoV-2 VoC and diverse sarbecoviruses, presenting a possible alternative or supplement to monoclonal antibody immunotherapies., Competing Interests: W.H.C, A.H., P.V.T., J.L.J., K.M. and M.G.J. are named inventors on provisional patents describing SpFN molecules. S.J.K., V.D., N.L.M., and K.M. are named inventors on provisional patents describing monoclonal antibodies against coronaviruses. M.G.J. is named as an inventor on international patent application WO/2018/081318 and U.S. patents 10,960,070, and 11,964,010 entitled “Prefusion coronavirus spike proteins and their use.” K.M. is a current employee of Pfizer and may, therefore, be a shareholder. A.F., K.Mur., and J.Kau. are former or current employees of B.S.V. The other authors declare no competing interests., (© 2024 The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
10. Mosaic vaccine-induced antibody-dependent cellular phagocytosis associated with delayed HIV-1 viral load rebound post treatment interruption.
- Author
-
Mdluli T, Slike BM, Curtis DJ, Shubin Z, Tran U, Li Y, Dussupt V, Mendez-Rivera L, Pinyakorn S, Stieh DJ, Tomaka FL, Schuitemaker H, Pau MG, Colby DJ, Kroon E, Sacdalan C, de Souza M, Phanupak N, Hsu DC, Ananworanich J, Ake JA, Trautmann L, Vasan S, Robb ML, Krebs SJ, Paquin-Proulx D, and Rolland M
- Subjects
- Humans, Male, Adult, Female, HIV Antibodies immunology, Middle Aged, Treatment Interruption, HIV-1 immunology, Phagocytosis, HIV Infections immunology, HIV Infections virology, HIV Infections drug therapy, Viral Load, AIDS Vaccines immunology, AIDS Vaccines administration & dosage
- Abstract
A heterologous Ad26/MVA vaccine was given prior to an analytic treatment interruption (ATI) in people living with HIV-1 (mainly CRF01_AE) who initiated antiretroviral treatment (ART) during acute HIV-1. We investigate the impact of Ad26/MVA vaccination on antibody (Ab)-mediated immune responses and their effect on time to viral rebound. The vaccine mainly triggers vaccine-matched binding Abs while, upon viral rebound post ATI, infection-specific CRF01_AE binding Abs increase in all participants. Binding Abs are not associated with time to viral rebound. The Ad26/MVA mosaic vaccine profile consists of correlated non-CRF01_AE binding Ab and Fc effector features, with strong Ab-dependent cellular phagocytosis (ADCP) responses. CRF01_AE-specific ADCP responses (measured either prior to or post ATI) are significantly higher in individuals with delayed viral rebound. Our results suggest that vaccines eliciting cross-reactive responses with circulating viruses in a target population could be beneficial and that ADCP responses may play a role in viral control post treatment interruption., Competing Interests: Declaration of interests The views expressed are those of the authors and should not be construed to represent the positions of the US Army, the Department of Defense, the Department of Health and Human Services, or the Henry M. Jackson Foundation for the Advancement of Military Medicine. D.J.S., F.L.T., H.S., and M.G.P. were employees of Janssen Vaccines & Prevention at the time the study was conducted and still hold stock in Johnson & Johnson. M.G.P. is an employee of Janssen Vaccines & Prevention and holds stock in Johnson & Johnson. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript., (Copyright © 2024 Henry M Jackson Foundation for the Advancement of Military Medicine, Inc. Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
11. Contemporary HIV-1 consensus Env with AI-assisted redesigned hypervariable loops promote antibody binding.
- Author
-
Bai H, Lewitus E, Li Y, Thomas PV, Zemil M, Merbah M, Peterson CE, Thuraisamy T, Rees PA, Hajduczki A, Dussupt V, Slike B, Mendez-Rivera L, Schmid A, Kavusak E, Rao M, Smith G, Frey J, Sims A, Wieczorek L, Polonis V, Krebs SJ, Ake JA, Vasan S, Bolton DL, Joyce MG, Townsley S, and Rolland M
- Subjects
- Humans, AIDS Vaccines immunology, Neutralization Tests, HEK293 Cells, Consensus Sequence, HIV Infections virology, HIV Infections immunology, Protein Binding, Epitopes immunology, HIV-1 immunology, env Gene Products, Human Immunodeficiency Virus immunology, env Gene Products, Human Immunodeficiency Virus chemistry, env Gene Products, Human Immunodeficiency Virus metabolism, HIV Antibodies immunology, Antibodies, Neutralizing immunology
- Abstract
An effective HIV-1 vaccine must elicit broadly neutralizing antibodies (bnAbs) against highly diverse Envelope glycoproteins (Env). Since Env with the longest hypervariable (HV) loops is more resistant to the cognate bnAbs than Env with shorter HV loops, we redesigned hypervariable loops for updated Env consensus sequences of subtypes B and C and CRF01_AE. Using modeling with AlphaFold2, we reduced the length of V1, V2, and V5 HV loops while maintaining the integrity of the Env structure and glycan shield, and modified the V4 HV loop. Spacers are designed to limit strain-specific targeting. All updated Env are infectious as pseudoviruses. Preliminary structural characterization suggests that the modified HV loops have a limited impact on Env's conformation. Binding assays show improved binding to modified subtype B and CRF01_AE Env but not to subtype C Env. Neutralization assays show increases in sensitivity to bnAbs, although not always consistently across clades. Strikingly, the HV loop modification renders the resistant CRF01_AE Env sensitive to 10-1074 despite the absence of a glycan at N332., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
12. Human ESCRT-I and ALIX function as scaffolding helical filaments in vivo .
- Author
-
Spada SJ, Rose KM, Sette P, O'Connor SK, Dussupt V, Siddartha Yerramilli V, Nagashima K, Sjoelund VH, Cruz P, Kabat J, Ganesan S, Smelkinson M, Nita-Lazar A, Hoyt F, Scarlata S, Hirsch V, Best SM, Grigg ME, and Bouamr F
- Abstract
The Endosomal Sorting Complex Required for Transport (ESCRT) is an evolutionarily conserved machinery that performs reverse-topology membrane scission in cells universally required from cytokinesis to budding of enveloped viruses. Upstream acting ESCRT-I and ALIX control these events and link recruitment of viral and cellular partners to late-acting ESCRT-III CHMP4 through incompletely understood mechanisms. Using structure-function analyses combined with super-resolution imaging, we show that ESCRT-I and ALIX function as distinct helical filaments in vivo . Together, they are essential for optimal structural scaffolding of HIV-1 nascent virions, the retention of viral and human genomes through defined functional interfaces, and recruitment of CHMP4 that itself assembles into corkscrew-like filaments intertwined with ESCRT-I or ALIX helices. Disruption of filament assembly or their conformationally clustered RNA binding interfaces in human cells impaired membrane abscission, resulted in major structural instability and leaked nucleic acid from nascent virions and nuclear envelopes. Thus, ESCRT-I and ALIX function as helical filaments in vivo and serve as both nucleic acid-dependent structural scaffolds as well as ESCRT-III assembly templates., Significance Statement: When cellular membranes are dissolved or breached, ESCRT is rapidly deployed to repair membranes to restore the integrity of intracellular compartments. Membrane sealing is ensured by ESCRT-III filaments assembled on the inner face of membrane; a mechanism termed inverse topology membrane scission. This mechanism, initiated by ESCRT-I and ALIX, is universally necessary for cytokinesis, wound repair, budding of enveloped viruses, and more. We show ESCRT-I and ALIX individually oligomerize into helical filaments that cluster newly discovered nucleic acid-binding interfaces and scaffold-in genomes within nascent virions and nuclear envelopes. These oligomers additionally appear to serve as ideal templates for ESCRT-III polymerization, as helical filaments of CHMP4B were found intertwined ESCRT-I or ALIX filaments in vivo . Similarly, corkscrew-like filaments of ALIX are also interwoven with ESCRT-I, supporting a model of inverse topology membrane scission that is synergistically reinforced by inward double filament scaffolding.
- Published
- 2024
- Full Text
- View/download PDF
13. Protective efficacy of a Zika purified inactivated virus vaccine candidate during pregnancy in marmosets.
- Author
-
Kim IJ, Gonzalez O, Tighe MP, Lanthier PA, Clark MJ, Travis KL, Low-Beer TC, Lanzer KG, Bernacki DT, Szaba FM, De La Barrera RA, Dussupt V, Mendez-Rivera L, Krebs SJ, Ross CN, Mdaki SD, Brasky KM, Layne-Colon D, Tardif SD, Thomas SJ, Modjarrad K, Blackman MA, and Patterson JL
- Abstract
Zika virus (ZIKV) infection during pregnancy poses significant threats to maternal and fetal health, leading to intrauterine fetal demise and severe developmental malformations that constitute congenital Zika syndrome (CZS). As such, the development of a safe and effective ZIKV vaccine is a critical public health priority. However, the safety and efficacy of such a vaccine during pregnancy remain uncertain. Historically, the conduct of clinical trials in pregnant women has been challenging. Therefore, clinically relevant animal pregnancy models are in high demand for testing vaccine efficacy. We previously reported that a marmoset pregnancy model of ZIKV infection consistently demonstrated vertical transmission from mother to fetus during pregnancy. Using this marmoset model, we also showed that vertical transmission could be prevented by pre-pregnancy vaccination with Zika purified inactivated virus (ZPIV) vaccine. Here, we further examined the efficacy of ZPIV vaccination during pregnancy. Vaccination during pregnancy elicited virus neutralizing antibody responses that were comparable to those elicited by pre-pregnancy vaccination. Vaccination also reduced placental pathology, viral burden and vertical transmission of ZIKV during pregnancy, without causing adverse effects. These results provide key insights into the safety and efficacy of ZPIV vaccination during pregnancy and demonstrate positive effects of vaccination on the reduction of ZIKV infection, an important advance in preparedness for future ZIKV outbreaks., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
14. Zika purified inactivated virus (ZPIV) vaccine reduced vertical transmission in pregnant immunocompetent mice.
- Author
-
Kim IJ, Tighe MP, Lanthier PA, Clark MJ, De La Barrera RA, Dussupt V, Mendez-Rivera L, Krebs SJ, Travis KL, Low-Beer TC, Cookenham TS, Lanzer KG, Bernacki DT, Szaba FM, Schneck AA, Ward J, Thomas SJ, Modjarrad K, and Blackman MA
- Abstract
Zika virus (ZIKV) is a significant threat to pregnant women and their fetuses as it can cause severe birth defects and congenital neurodevelopmental disorders, referred to as congenital Zika syndrome (CZS). Thus, a safe and effective ZIKV vaccine for pregnant women to prevent in utero ZIKV infection is of utmost importance. Murine models of ZIKV infection are limited by the fact that immunocompetent mice are resistant to ZIKV infection. As such, interferon-deficient mice have been used in some preclinical studies to test the efficacy of ZIKV vaccine candidates against lethal virus challenge. However, interferon-deficient mouse models have limitations in assessing the immunogenicity of vaccines, necessitating the use of immunocompetent mouse pregnancy models. Using the human stat2 knock-in (hSTAT2KI) mouse pregnancy model, we show that vaccination with a purified formalin-inactivated Zika virus (ZPIV) vaccine prior to pregnancy successfully prevented vertical transmission. In addition, maternal immunity protected offspring against postnatal challenge for up to 28 days. Furthermore, passive transfer of human IgG purified from hyper-immune sera of ZPIV vaccinees prevented maternal and fetal ZIKV infection, providing strong evidence that the neutralizing antibody response may serve as a meaningful correlate of protection., (© 2024. The Author(s).)
- 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. Safety and immunogenicity of a purified inactivated Zika virus vaccine candidate in adults primed with a Japanese encephalitis virus or yellow fever virus vaccine in the USA: a phase 1, randomised, double-blind, placebo-controlled clinical trial.
- Author
-
Koren MA, Lin L, Eckels KH, De La Barrera R, Dussupt V, Donofrio G, Sondergaard EL, Mills KT, Robb ML, Lee C, Adedeji O, Keiser PB, Curley JM, Copeland NK, Crowell TA, Hutter JN, Hamer MJ, Valencia-Ruiz A, Darden J, Peel S, Amare MF, Mebrahtu T, Costanzo M, Krebs SJ, Gromowski GD, Jarman RG, Thomas SJ, Michael NL, and Modjarrad K
- Subjects
- Adult, Female, Humans, Male, Antibodies, Neutralizing, Antibodies, Viral, Double-Blind Method, Immunogenicity, Vaccine, Vaccines, Inactivated, Yellow fever virus, Yellow Fever prevention & control, Encephalitis Virus, Japanese, Japanese Encephalitis Vaccines adverse effects, Viral Vaccines, Yellow Fever Vaccine adverse effects, Zika Virus, Zika Virus Infection prevention & control
- Abstract
Background: Zika virus infection is a threat to at-risk populations, causing major birth defects and serious neurological complications. Development of a safe and efficacious Zika virus vaccine is, therefore, a global health priority. Assessment of heterologous flavivirus vaccination is important given co-circulation of Japanese encephalitis virus and yellow fever virus with Zika virus. We investigated the effect of priming flavivirus naive participants with a licensed flavivirus vaccine on the safety and immunogenicity of a purified inactivated Zika vaccine (ZPIV)., Methods: This phase 1, placebo-controlled, double-blind trial was done at the Walter Reed Army Institute of Research Clinical Trials Center in Silver Spring, MD, USA. Eligible participants were healthy adults aged 18-49 years, with no detectable evidence of previous flavivirus exposure (by infection or vaccination), as measured by a microneutralisation assay. Individuals with serological evidence of HIV, hepatitis B, or hepatitis C infection were excluded, as were pregnant or breastfeeding women. Participants were recruited sequentially into one of three groups (1:1:1) to receive no primer, two doses of intramuscular Japanese encephalitis virus vaccine (IXIARO), or a single dose of subcutaneous yellow fever virus vaccine (YF-VAX). Within each group, participants were randomly assigned (4:1) to receive intramuscular ZPIV or placebo. Priming vaccinations were given 72-96 days before ZPIV. ZPIV was administered either two or three times, at days 0, 28, and 196-234. The primary outcome was occurrence of solicited systemic and local adverse events along with serious adverse events and adverse events of special interest. These data were analysed in all participants receiving at least one dose of ZPIV or placebo. Secondary outcomes included measurement of neutralizing antibody responses following ZPIV vaccination in all volunteers with available post-vaccination data. This trial is registered at ClinicalTrials.gov, NCT02963909., Findings: Between Nov 7, 2016, and Oct 30, 2018, 134 participants were assessed for eligibility. 21 did not meet inclusion criteria, 29 met exclusion criteria, and ten declined to participate. 75 participants were recruited and randomly assigned. 35 (47%) of 75 participants were male and 40 (53%) were female. 25 (33%) of 75 participants identified as Black or African American and 42 (56%) identified as White. These proportions and other baseline characteristics were similar between groups. There were no statistically significant differences in age, gender, race, or BMI between those who did and did not opt into the third dose. All participants received the planned priming IXIARO and YF-VAX vaccinations, but one participant who received YF-VAX dropped out before receipt of the first dose of ZPIV. 50 participants received a third dose of ZPIV or placebo, including 14 flavivirus-naive people, 17 people primed with Japanese encephalitis virus vaccine, and 19 participants primed with yellow fever vaccine. Vaccinations were well tolerated across groups. Pain at the injection site was the only adverse event reported more frequently in participants who received ZPIV than in those who received placebo (39 [65%] of 60 participants, 95% CI 51·6-76·9 who received ZPIV vs three [21·4%] of 14 who received placebo; 4·7-50·8; p=0·006). No patients had an adverse event of special interest or serious adverse event related to study treatment. At day 57, the flavivirus-naive volunteers had an 88% (63·6-98·5, 15 of 17) seroconversion rate (neutralising antibody titre ≥1:10) and geometric mean neutralising antibody titre (GMT) against Zika virus of 100·8 (39·7-255·7). In the Japanese encephalitis vaccine-primed group, the day 57 seroconversion rate was 31·6% (95% CI 12·6-56·6, six of 19) and GMT was 11·8 (6·1-22·8). Participants primed with YF-VAX had a seroconversion rate of 25% (95% CI 8·7-49·1, five of 20) and GMT of 6·6 (5·2-8·4). Humoral immune responses rose substantially following a third dose of ZPIV, with seroconversion rates of 100% (69·2-100; ten of ten), 92·9% (66·1-99·8; 13 of 14), and 60% (32·2-83·7, nine of 15) and GMTs of 511·5 (177·6-1473·6), 174·2 (51·6-587·6), and 79 (19·0-326·8) in the flavivirus naive, Japanese encephalitis vaccine-primed, and yellow fever vaccine-primed groups, respectively., Interpretation: We found ZPIV to be well tolerated in flavivirus naive and primed adults but that immunogenicity varied significantly according to antecedent flavivirus vaccination status. Immune bias towards the flavivirus antigen of initial exposure and the timing of vaccination may have impacted responses. A third ZPIV dose overcame much, but not all, of the discrepancy in immunogenicity. The results of this phase 1 clinical trial have implications for further evaluation of ZPIV's immunisation schedule and use of concomitant vaccinations., Funding: Department of Defense, Defense Health Agency; National Institute of Allergy and Infectious Diseases; and Division of Microbiology and Infectious Disease., Competing Interests: Declaration of interests SJT is a data and safety monitoring board member for the Moderna Zika trial and is compensated for his time. He has also supported the WHO R&D blueprint and vaccine TPP working groups in exchange for no compensation. SJT, KHE, and RDLB are named patent holders (WO2017210215A1) for the Zika inactivated vaccine evaluated in this clinical trial. All other authors declare no competing interests., (Copyright © 2023 Elsevier Ltd. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
19. Zika-specific neutralizing antibodies targeting inter-dimer envelope epitopes.
- Author
-
Sankhala RS, Dussupt V, Donofrio G, Gromowski GD, De La Barrera RA, Larocca RA, Mendez-Rivera L, Lee A, Choe M, Zaky W, Mantus G, Jensen JL, Chen WH, Gohain N, Bai H, McCracken MK, Mason RD, Leggat D, Slike BM, Tran U, Jian N, Abbink P, Peterson R, Mendes EA, Freitas de Oliveira Franca R, Calvet GA, Bispo de Filippis AM, McDermott A, Roederer M, Hernandez M, Albertus A, Davidson E, Doranz BJ, Rolland M, Robb ML, Lynch RM, Barouch DH, Jarman RG, Thomas SJ, Modjarrad K, Michael NL, Krebs SJ, and Joyce MG
- Subjects
- Humans, Animals, Mice, Antibodies, Neutralizing, Epitopes, Macaca mulatta, Antibodies, Viral, Antibodies, Monoclonal, Viral Envelope Proteins chemistry, Zika Virus, Zika Virus Infection, Dengue Virus, Dengue, Viral Vaccines therapeutic use
- Abstract
Zika virus (ZIKV) is an emerging pathogen that causes devastating congenital defects. The overlapping epidemiology and immunologic cross-reactivity between ZIKV and dengue virus (DENV) pose complex challenges to vaccine design, given the potential for antibody-dependent enhancement of disease. Therefore, classification of ZIKV-specific antibody targets is of notable value. From a ZIKV-infected rhesus macaque, we identify ZIKV-reactive B cells and isolate potent neutralizing monoclonal antibodies (mAbs) with no cross-reactivity to DENV. We group these mAbs into four distinct antigenic groups targeting ZIKV-specific cross-protomer epitopes on the envelope glycoprotein. Co-crystal structures of representative mAbs in complex with ZIKV envelope glycoprotein reveal envelope-dimer epitope and unique dimer-dimer epitope targeting. All four specificities are serologically identified in convalescent humans following ZIKV infection, and representative mAbs from all four groups protect against ZIKV replication in mice. These results provide key insights into ZIKV-specific antigenicity and have implications for ZIKV vaccine, diagnostic, and therapeutic development., Competing Interests: Declaration of interests S.J.K., V.D., G.D., K.M., N.M., D.H.B., M.G.J., R.S.S., and R.G.J. are named inventors on a PCT patent application WO 2019/209974 describing ZIKV neutralizing antibodies and their use. D.H.B. has received grants from Novavax and personal fees from IGM Biosciences. M.H., A.A., E.D., and B.J.D. are employees of Integral Molecular. B.J.D. is also a shareholder of the company., (Copyright © 2023 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
20. Targeting the Spike Receptor Binding Domain Class V Cryptic Epitope by an Antibody with Pan-Sarbecovirus Activity.
- Author
-
Jensen JL, Sankhala RS, Dussupt V, Bai H, Hajduczki A, Lal KG, Chang WC, Martinez EJ, Peterson CE, Golub ES, Rees PA, Mendez-Rivera L, Zemil M, Kavusak E, Mayer SV, Wieczorek L, Kannan S, Doranz BJ, Davidson E, Yang ES, Zhang Y, Chen M, Choe M, Wang L, Gromowski GD, Koup RA, Michael NL, Polonis VR, Rolland M, Modjarrad K, Krebs SJ, and Joyce MG
- Subjects
- Humans, Antibodies, Monoclonal chemistry, Antibodies, Monoclonal metabolism, Antibodies, Neutralizing chemistry, Antibodies, Neutralizing metabolism, SARS-CoV-2 chemistry, SARS-CoV-2 metabolism, Protein Domains, Crystallography, X-Ray, Protein Structure, Quaternary, Models, Molecular, Cell Line, Antibodies, Viral chemistry, Antibodies, Viral metabolism, COVID-19, Epitopes chemistry, Severe acute respiratory syndrome-related coronavirus chemistry
- Abstract
Novel therapeutic monoclonal antibodies (MAbs) must accommodate comprehensive breadth of activity against diverse sarbecoviruses and high neutralization potency to overcome emerging variants. Here, we report the crystal structure of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) receptor binding domain (RBD) in complex with MAb WRAIR-2063, a moderate-potency neutralizing antibody with exceptional sarbecovirus breadth, that targets the highly conserved cryptic class V epitope. This epitope overlaps substantially with the spike protein N-terminal domain (NTD) -interacting region and is exposed only when the spike is in the open conformation, with one or more RBDs accessible. WRAIR-2063 binds the RBD of SARS-CoV-2 WA-1, all variants of concern (VoCs), and clade 1 to 4 sarbecoviruses with high affinity, demonstrating the conservation of this epitope and potential resiliency against variation. We compare structural features of additional class V antibodies with their reported neutralization capacity to further explore the utility of the class V epitope as a pan-sarbecovirus vaccine and therapeutic target. IMPORTANCE Characterization of MAbs against SARS-CoV-2, elicited through vaccination or natural infection, has provided vital immunotherapeutic options for curbing the COVID-19 pandemic and has supplied critical insights into SARS-CoV-2 escape, transmissibility, and mechanisms of viral inactivation. Neutralizing MAbs that target the RBD but do not block ACE2 binding are of particular interest because the epitopes are well conserved within sarbecoviruses and MAbs targeting this area demonstrate cross-reactivity. The class V RBD-targeted MAbs localize to an invariant site of vulnerability, provide a range of neutralization potency, and exhibit considerable breadth against divergent sarbecoviruses, with implications for vaccine and therapeutic development., Competing Interests: The authors declare a conflict of interest. Patent application number PCT/US 63/140,763, PCT WO 2022/159839 A1 was filed containing the mAbs described in this publication for authors S.J.K., K.M., V.D., and N.L.M. 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.” The other authors declare no competing interests.
- Published
- 2023
- Full Text
- View/download PDF
21. Convalescent human IgG, but not IgM, from COVID-19 survivors confers dose-dependent protection against SARS-CoV-2 replication and disease in hamsters.
- Author
-
King HAD, Dussupt V, Mendez-Rivera L, Slike BM, Tran U, Jackson ND, Barkei E, Zemil M, Tourtellott-Fogt E, Kuklis CH, Soman S, Ahmed A, Porto M, Kitajewski C, Spence B, Benetiene D, Wieczorek L, Kar S, Gromowski G, Polonis VR, Krebs SJ, Modjarrad K, and Bolton DL
- Subjects
- Animals, Cricetinae, Humans, Pandemics, Immunoglobulin G, Antibodies, Neutralizing, Mesocricetus, Survivors, SARS-CoV-2, COVID-19
- Abstract
Introduction: Antibody therapeutic strategies have served an important role during the COVID-19 pandemic, even as their effectiveness has waned with the emergence of escape variants. Here we sought to determine the concentration of convalescent immunoglobulin required to protect against disease from SARS-CoV-2 in a Syrian golden hamster model., Methods: Total IgG and IgM were isolated from plasma of SARS-CoV-2 convalescent donors. Dose titrations of IgG and IgM were infused into hamsters 1 day prior to challenge with SARS-CoV-2 Wuhan-1., Results: The IgM preparation was found to have ~25-fold greater neutralization potency than IgG. IgG infusion protected hamsters from disease in a dose-dependent manner, with detectable serum neutralizing titers correlating with protection. Despite a higher in vitro neutralizing potency, IgM failed to protect against disease when transferred into hamsters., Discussion: This study adds to the growing body of literature that demonstrates neutralizing IgG antibodies are important for protection from SARS-CoV-2 disease, and confirms that polyclonal IgG in sera can be an effective preventative strategy if the neutralizing titers are sufficiently high. In the context of new variants, against which existing vaccines or monoclonal antibodies have reduced efficacy, sera from individuals who have recovered from infection with the emerging variant may potentially remain an efficacious tool., Competing Interests: Authors MP, CK, BS, DB and SK are employed by BIOQUAL, Inc. 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 © 2023 King, Dussupt, Mendez-Rivera, Slike, Tran, Jackson, Barkei, Zemil, Tourtellott-Fogt, Kuklis, Soman, Ahmed, Porto, Kitajewski, Spence, Benetiene, Wieczorek, Kar, Gromowski, Polonis, Krebs, Modjarrad and Bolton.)
- Published
- 2023
- Full Text
- View/download PDF
22. Shark nanobodies with potent SARS-CoV-2 neutralizing activity and broad sarbecovirus reactivity.
- Author
-
Chen WH, Hajduczki A, Martinez EJ, Bai H, Matz H, Hill TM, Lewitus E, Chang WC, Dawit L, Peterson CE, Rees PA, Ajayi AB, Golub ES, Swafford I, Dussupt V, David S, Mayer SV, Soman S, Kuklis C, Corbitt C, King J, Choe M, Sankhala RS, Thomas PV, Zemil M, Wieczorek L, Hart T, Duso D, Kummer L, Yan L, Sterling SL, Laing ED, Broder CC, Williams JK, Davidson E, Doranz BJ, Krebs SJ, Polonis VR, Paquin-Proulx D, Rolland M, Reiley WW, Gromowski GD, Modjarrad K, Dooley H, and Joyce MG
- Subjects
- Animals, Mice, Antibodies, Neutralizing, Antibodies, Viral, Epitopes, Ferritins genetics, Immunoglobulin Fc Fragments, Mice, Transgenic, SARS-CoV-2 genetics, Spike Glycoprotein, Coronavirus genetics, Sharks, COVID-19 prevention & control, Severe acute respiratory syndrome-related coronavirus, Single-Domain Antibodies
- Abstract
Despite rapid and ongoing vaccine and therapeutic development, SARS-CoV-2 continues to evolve and evade, presenting a need for next-generation diverse therapeutic modalities. Here we show that nurse sharks immunized with SARS-CoV-2 recombinant receptor binding domain (RBD), RBD-ferritin (RFN), or spike protein ferritin nanoparticle (SpFN) immunogens elicit a set of new antigen receptor antibody (IgNAR) molecules that target two non-overlapping conserved epitopes on the spike RBD. Representative shark antibody variable NAR-Fc chimeras (ShAbs) targeting either of the two epitopes mediate cell-effector functions, with high affinity to all SARS-CoV-2 viral variants of concern, including the divergent Omicron strains. The ShAbs potently cross-neutralize SARS-CoV-2 WA-1, Alpha, Beta, Delta, Omicron BA.1 and BA.5, and SARS-CoV-1 pseudoviruses, and confer protection against SARS-CoV-2 challenge in the K18-hACE2 transgenic mouse model. Structural definition of the RBD-ShAb01-ShAb02 complex enabled design and production of multi-specific nanobodies with enhanced neutralization capacity, and picomolar affinity to divergent sarbecovirus clade 1a, 1b and 2 RBD molecules. These shark nanobodies represent potent immunotherapeutics both for current use, and future sarbecovirus pandemic preparation., (© 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
23. Bivalent intra-spike binding provides durability against emergent Omicron lineages: Results from a global consortium.
- Author
-
Callaway HM, Hastie KM, Schendel SL, Li H, Yu X, Shek J, Buck T, Hui S, Bedinger D, Troup C, Dennison SM, Li K, Alpert MD, Bailey CC, Benzeno S, Bonnevier JL, Chen JQ, Chen C, Cho H, Crompton PD, Dussupt V, Entzminger KC, Ezzyat Y, Fleming JK, Geukens N, Gilbert AE, Guan Y, Han X, Harvey CJ, Hatler JM, Howie B, Hu C, Huang A, Imbrechts M, Jin A, Kamachi N, Keitany G, Klinger M, Kolls JK, Krebs SJ, Li T, Luo F, Maruyama T, Meehl MA, Mendez-Rivera L, Musa A, Okumura CJ, Rubin BER, Sato AK, Shen M, Singh A, Song S, Tan J, Trimarchi JM, Upadhyay DP, Wang Y, Yu L, Yuan TZ, Yusko E, Peters B, Tomaras G, and Saphire EO
- Subjects
- Humans, SARS-CoV-2, Ethnicity, Epitopes, Antibodies, Viral, Antibodies, Neutralizing, Neutralization Tests, COVID-19
- Abstract
The SARS-CoV-2 Omicron variant of concern (VoC) and its sublineages contain 31-36 mutations in spike and escape neutralization by most therapeutic antibodies. In a pseudovirus neutralization assay, 66 of the nearly 400 candidate therapeutics in the Coronavirus Immunotherapeutic Consortium (CoVIC) panel neutralize Omicron and multiple Omicron sublineages. Among natural immunoglobulin Gs (IgGs), especially those in the receptor-binding domain (RBD)-2 epitope community, nearly all Omicron neutralizers recognize spike bivalently, with both antigen-binding fragments (Fabs) simultaneously engaging adjacent RBDs on the same spike. Most IgGs that do not neutralize Omicron bind either entirely monovalently or have some (22%-50%) monovalent occupancy. Cleavage of bivalent-binding IgGs to Fabs abolishes neutralization and binding affinity, with disproportionate loss of activity against Omicron pseudovirus and spike. These results suggest that VoC-resistant antibodies overcome mutagenic substitution via avidity. Hence, vaccine strategies targeting future SARS-CoV-2 variants should consider epitope display with spacing and organization identical to trimeric spike., Competing Interests: Declaration of interests C.C.B. and M.D.A. are inventors on the patent application PCT/US21/33454, which covers the version of ACE2-Ig used in this study. The patent application is assigned to Emmune, Inc. M.D.A., C.C.B., and J.M.T. own stock in Emmune, Inc. Y.G. is co-founder and shareholder in Antibody BioPharm, Inc., and ChangYuan FuNeng (Shanghai) Life Technology Co., Ltd. L.Y. and Y.G. have filed patent applications on related COVID-19 antibodies. P.D.C., J.T., and H.C. are inventors on several of the antibodies described in this study., (Copyright © 2023 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
24. Antibodies as drugs-a Keystone Symposia report.
- Author
-
Cable J, Saphire EO, Hayday AC, Wiltshire TD, Mousa JJ, Humphreys DP, Breij ECW, Bruhns P, Broketa M, Furuya G, Hauser BM, Mahévas M, Carfi A, Cantaert T, Kwong PD, Tripathi P, Davis JH, Brewis N, Keyt BA, Fennemann FL, Dussupt V, Sivasubramanian A, Kim PM, Rawi R, Richardson E, Leventhal D, Wolters RM, Geuijen CAW, Sleeman MA, Pengo N, and Donnellan FR
- Subjects
- Humans, Immunotherapy, Antibodies, Bispecific therapeutic use
- Abstract
Therapeutic antibodies have broad indications across diverse disease states, such as oncology, autoimmune diseases, and infectious diseases. New research continues to identify antibodies with therapeutic potential as well as methods to improve upon endogenous antibodies and to design antibodies de novo. On April 27-30, 2022, experts in antibody research across academia and industry met for the Keystone symposium "Antibodies as Drugs" to present the state-of-the-art in antibody therapeutics, repertoires and deep learning, bispecific antibodies, and engineering., (© 2022 New York Academy of Sciences.)
- Published
- 2023
- Full Text
- View/download PDF
25. Unglycosylated Soluble SARS-CoV-2 Receptor Binding Domain (RBD) Produced in E. coli Combined with the Army Liposomal Formulation Containing QS21 (ALFQ) Elicits Neutralizing Antibodies against Mismatched Variants.
- Author
-
Balasubramaniyam A, Ryan E, Brown D, Hamza T, Harrison W, Gan M, Sankhala RS, Chen WH, Martinez EJ, Jensen JL, Dussupt V, Mendez-Rivera L, Mayer S, King J, Michael NL, Regules J, Krebs S, Rao M, Matyas GR, Joyce MG, Batchelor AH, Gromowski GD, and Dutta S
- Abstract
The emergence of novel potentially pandemic pathogens necessitates the rapid manufacture and deployment of effective, stable, and locally manufacturable vaccines on a global scale. In this study, the ability of the Escherichia coli expression system to produce the receptor binding domain (RBD) of the SARS-CoV-2 spike protein was evaluated. The RBD of the original Wuhan-Hu1 variant and of the Alpha and Beta variants of concern (VoC) were expressed in E. coli , and their biochemical and immunological profiles were compared to RBD produced in mammalian cells. The E. coli -produced RBD variants recapitulated the structural character of mammalian-expressed RBD and bound to human angiotensin converting enzyme (ACE2) receptor and a panel of neutralizing SARS-CoV-2 monoclonal antibodies. A pilot vaccination in mice with bacterial RBDs formulated with a novel liposomal adjuvant, Army Liposomal Formulation containing QS21 (ALFQ), induced polyclonal antibodies that inhibited RBD association to ACE2 in vitro and potently neutralized homologous and heterologous SARS-CoV-2 pseudoviruses. Although all vaccines induced neutralization of the non-vaccine Delta variant, only the Beta RBD vaccine produced in E. coli and mammalian cells effectively neutralized the Omicron BA.1 pseudovirus. These outcomes warrant further exploration of E. coli as an expression platform for non-glycosylated, soluble immunogens for future rapid response to emerging pandemic pathogens.
- Published
- 2022
- Full Text
- View/download PDF
26. Fc receptor engagement of HIV-1 Env-specific antibodies in mothers and infants predicts reduced vertical transmission.
- Author
-
Barrows BM, Krebs SJ, Jian N, Zemil M, Slike BM, Dussupt V, Tran U, Mendez-Rivera L, Chang D, O'Sullivan AM, Mann B, Sanders-Buell E, Shubin Z, Creegan M, Paquin-Proulx D, Ehrenberg P, Laurence-Chenine A, Srithanaviboonchai K, Thomas R, Eller MA, Ferrari G, Robb M, Rao V, Tovanabutra S, Polonis VR, and Wieczorek L
- Subjects
- Infant, Newborn, Pregnancy, Female, Infant, Humans, Receptors, IgG, HIV Antibodies, Receptors, Fc, HIV-1, HIV Infections
- Abstract
Introduction: Infants acquire maternal antibodies by Fc receptor transcytosis across the placenta during pregnancy. Fc receptors are expressed on immune cells and are important for activation of effector cell functions., Methods: In this study, we evaluated Fc receptor engagement and ADCC activity of plasma binding antibodies from human immunodeficiency virus-1 (HIV) -infected mothers and to identify factors that may contribute to protection from HIV vertical transmission., Results: HIV-specific binding and Fc receptor engagement of plasma antibodies varied between mothers by transmission status and infants by infection status. Non-transmitting (NT) mothers and HIV-uninfected infants had antibodies with higher neonatal Fc receptor (FcRn) and FcγR engagement, as compared to transmitting (T) mothers and HIV+ infants, respectively. A significant inverse correlation between plasma antibody FcRn and FcγR engagement was observed for T mothers, but not NT mothers. Conversely, a significant direct correlation was observed between plasma antibody FcRn and FcγR engagement for HIV- infants, but not for HIV+ infants. Consequently, we observed significantly higher plasma antibody ADCC potency and breadth in HIV- infants, as compared to HIV+ infants. However, no differences in overall ADCC potency and breadth were observed between mothers. FcRn-engagement of HIV-specific antibodies in both mothers and infants predicted a lack of vertical transmission of HIV., Discussion: This study indicates that HIV-uninfected infants acquire HIV-specific antibodies with greater Fc receptor engagement and thus, greater ADCC capacity., 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 Barrows, Krebs, Jian, Zemil, Slike, Dussupt, Tran, Mendez-Rivera, Chang, O’Sullivan, Mann, Sanders-Buell, Shubin, Creegan, Paquin-Proulx, Ehrenberg, Laurence-Chenine, Srithanaviboonchai, Thomas, Eller, Ferrari, Robb, Rao, Tovanabutra, Polonis and Wieczorek.)
- Published
- 2022
- Full Text
- View/download PDF
27. Evaluation of Antibody-Dependent Fc-Mediated Viral Entry, as Compared With Neutralization, in SARS-CoV-2 Infection.
- Author
-
Wieczorek L, Zemil M, Merbah M, Dussupt V, Kavusak E, Molnar S, Heller J, Beckman B, Wollen-Roberts S, Peachman KK, Darden JM, Krebs S, Rolland M, Peel SA, and Polonis VR
- Subjects
- HEK293 Cells, Humans, Immunization, Passive, Immunoglobulin Fc Fragments, Spike Glycoprotein, Coronavirus, Virus Internalization, COVID-19 Serotherapy, COVID-19 therapy, SARS-CoV-2
- Abstract
Fc-mediated virus entry has been observed for many viruses, but the characterization of this activity in convalescent plasma against SARS-CoV-2 Variants of Concern (VOC) is undefined. In this study, we evaluated Fc-mediated viral entry (FVE) on FcγRIIa-expressing HEK293 cells in the presence of SARS-CoV-2 convalescent plasma and compared it with SARS-CoV-2 pseudovirus neutralization using ACE2-expressing HEK293 cells. The plasma were collected early in the pandemic from 39 individuals. We observed both neutralization and FVE against the infecting Washington SARS-CoV-2 strain for 31% of plasmas, neutralization, but not FVE for 61% of plasmas, and no neutralization or FVE for 8% of plasmas. Neutralization titer correlated significantly with the plasma dilution at which maximum FVE was observed, indicating Fc-mediated uptake peaked as neutralization potency waned. While total Spike-specific plasma IgG levels were similar between plasma that mediated FVE and those that did not, Spike-specific plasma IgM levels were significantly higher in plasma that did not mediate FVE. Plasma neutralization titers against the Alpha (B.1.1.7), Beta (B.1.351), Gamma (P.1) and Delta (B.1.617.2) VOC were significantly lower than titers against the Washington strain, while plasma FVE activity against the VOC was either higher or similar. This is the first report to demonstrate a functional shift in convalescent plasma antibodies from neutralizing and FVE-mediating against the earlier Washington strain, to an activity mediating only FVE and no neutralization activity against the emerging VOC, specifically the Beta (B.1.351) and Gamma (P.1) VOC. It will be important to determine the in vivo relevance of these findings., 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 Wieczorek, Zemil, Merbah, Dussupt, Kavusak, Molnar, Heller, Beckman, Wollen-Roberts, Peachman, Darden, Krebs, Rolland, Peel and Polonis.)
- 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. 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
31. 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
32. 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
33. Limited Evidence for a Relationship between HIV-1 Glycan Shield Features in Early Infection and the Development of Neutralization Breadth.
- Author
-
Li Y, Bai H, Sanders-Buell E, Dussupt V, Townsley S, Donofrio G, Bose M, O'Sullivan AM, Kibuuka H, Maganga L, Nitayaphan S, Kosgei J, Pitisuttithum P, Rerks-Ngarm S, Eller LA, Michael NL, Robb ML, Ake J, Vasan S, Tovanabutra S, Krebs SJ, and Rolland M
- Subjects
- Africa, Eastern epidemiology, Antibodies, Neutralizing blood, Cohort Studies, Epitopes, Glycosylation, HIV Antibodies blood, HIV Infections immunology, HIV Infections virology, Humans, Thailand epidemiology, Antibodies, Neutralizing immunology, HIV Antibodies immunology, HIV Infections epidemiology, HIV-1 immunology, Immune Evasion immunology, Polysaccharides immunology, env Gene Products, Human Immunodeficiency Virus immunology
- Abstract
Identifying whether viral features present in acute HIV-1 infection predetermine the development of neutralization breadth is critical to vaccine design. Incorporating such features in vaccine antigens could initiate cross-reactive antibody responses that could sufficiently protect vaccinees from HIV-1 infection despite the uniqueness of each founder virus. To understand the relationship between Env determinants and the development of neutralization breadth, we focused on 197 individuals enrolled in two cohorts in Thailand and East Africa (RV144 and RV217) and followed since their diagnosis in acute or early HIV-1 infection. We analyzed the distribution of variable loop lengths and glycans, as well as the predicted density of the glycan shield, and compared these envelope features to the neutralization breadth data obtained 3 years after infection ( n = 121). Our study revealed limited evidence for glycan shield features that associate with the development of neutralization breadth. While the glycan shield tended to be denser in participants who subsequently developed breadth, no significant relationship was found between the size of glycan holes and the development of neutralization breadth. The parallel analysis of 3,000 independent Env sequences showed no evidence of directional evolution of glycan shield features since the beginning of the epidemic. Together, our results highlight that glycan shield features in acute and early HIV-1 infection may not play a role determinant enough to dictate the development of neutralization breadth and instead suggest that the glycan shield's reactive properties that are associated with immune evasion may have a greater impact. IMPORTANCE A major goal of HIV-1 vaccine research is to design vaccine candidates that elicit potent broadly neutralizing antibodies (bNAbs). Different viral features have been associated with the development of bNAbs, including the glycan shield on the surface of the HIV-1 Envelope (Env). Here, we analyzed data from two cohorts of individuals who were followed from early infection to several years after infection spanning multiple HIV-1 subtypes. We compared Env glycan features in HIV-1 sequences obtained in early infection to the potency and breadth of neutralizing antibodies measured 1 to 3 years after infection. We found limited evidence of glycan shield properties that associate with the development of neutralization breadth in these cohorts. These results may have important implications for antigen design in future vaccine strategies and emphasize that HIV-1 vaccines will need to rely on a complex set of properties to elicit neutralization breadth.
- Published
- 2021
- Full Text
- View/download PDF
34. RV144 vaccine imprinting constrained HIV-1 evolution following breakthrough infection.
- Author
-
Lewitus E, Sanders-Buell E, Bose M, O'Sullivan AM, Poltavee K, Li Y, Bai H, Mdluli T, Donofrio G, Slike B, Zhao H, Wong K, Chen L, Miller S, Lee J, Ahani B, Lepore S, Muhammad S, Grande R, Tran U, Dussupt V, Mendez-Rivera L, Nitayaphan S, Kaewkungwal J, Pitisuttithum P, Rerks-Ngarm S, O'Connell RJ, Janes H, Gilbert PB, Gramzinski R, Vasan S, Robb ML, Michael NL, Krebs SJ, Herbeck JT, Edlefsen PT, Mullins JI, Kim JH, Tovanabutra S, and Rolland M
- Abstract
The scale of the HIV-1 epidemic underscores the need for a vaccine. The multitude of circulating HIV-1 strains together with HIV-1's high evolvability hints that HIV-1 could adapt to a future vaccine. Here, we wanted to investigate the effect of vaccination on the evolution of the virus post-breakthrough infection. We analyzed 2,635 HIV-1 env sequences sampled up to a year post-diagnosis from 110 vaccine and placebo participants who became infected in the RV144 vaccine efficacy trial. We showed that the Env signature sites that were previously identified to distinguish vaccine and placebo participants were maintained over time. In addition, fewer sites were under diversifying selection in the vaccine group than in the placebo group. These results indicate that HIV-1 would possibly adapt to a vaccine upon its roll-out., (© The Author(s) 2021. Published by Oxford University Press.)
- Published
- 2021
- Full Text
- View/download PDF
35. 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
36. B cell engagement with HIV-1 founder virus envelope predicts development of broadly neutralizing antibodies.
- Author
-
Townsley SM, Donofrio GC, Jian N, Leggat DJ, Dussupt V, Mendez-Rivera L, Eller LA, Cofer L, Choe M, Ehrenberg PK, Geretz A, Gift S, Grande R, Lee A, Peterson C, Piechowiak MB, Slike BM, Tran U, Joyce MG, Georgiev IS, Rolland M, Thomas R, Tovanabutra S, Doria-Rose NA, Polonis VR, Mascola JR, McDermott AB, Michael NL, Robb ML, and Krebs SJ
- Subjects
- Cell Line, Epitopes immunology, HIV Infections virology, Humans, Viremia, env Gene Products, Human Immunodeficiency Virus immunology, B-Lymphocytes immunology, Broadly Neutralizing Antibodies, HIV Antibodies immunology, HIV Infections immunology, HIV-1 immunology, Viral Envelope immunology
- Abstract
Determining which immunological mechanisms contribute to the development of broad neutralizing antibodies (bNAbs) during HIV-1 infection is a major goal to inform vaccine design. Using samples from a longitudinal HIV-1 acute infection cohort, we found key B cell determinants within the first 14-43 days of viremia that predict the development of bNAbs years later. Individuals who develop neutralization breadth had significantly higher B cell engagement with the autologous founder HIV envelope (Env) within 1 month of initial viremia. A higher frequency of founder-Env-specific naive B cells was associated with increased B cell activation and differentiation and predictive of bNAb development. These data demonstrate that the initial B cell interaction with the founder HIV Env is important for the development of broadly neutralizing antibodies and provide evidence that events within HIV acute infection lead to downstream functional outcomes., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2021 Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
37. 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
38. Correction: RV144 HIV-1 vaccination impacts post-infection antibody responses.
- Author
-
Mdluli T, Jian N, Slike B, Paquin-Proulx D, Donofrio G, Alrubayyi A, Gift S, Grande R, Bryson M, Lee A, Dussupt V, Mendez-Rivera L, Sanders-Buell E, Chenine AL, Tran U, Li Y, Brown E, Edlefsen PT, O'Connell R, Gilbert P, Nitayaphan S, Pitisuttihum P, Rerks-Ngarm S, Robb ML, Gramzinski R, Alter G, Tovanabutra S, Georgiev IS, Ackerman ME, Polonis VR, Vasan S, Michael NL, Kim JH, Eller MA, Krebs SJ, and Rolland M
- Abstract
[This corrects the article DOI: 10.1371/journal.ppat.1009101.].
- Published
- 2021
- Full Text
- View/download PDF
39. Landscape of Monoclonal Antibodies Targeting Zika and Dengue: Therapeutic Solutions and Critical Insights for Vaccine Development.
- Author
-
Dussupt V, Modjarrad K, and Krebs SJ
- Subjects
- Amino Acid Sequence, Antibodies, Monoclonal genetics, Antibodies, Neutralizing genetics, Antibodies, Viral genetics, Antibodies, Viral immunology, Antigens, Viral chemistry, Antigens, Viral immunology, Cross Reactions, Dengue Vaccines immunology, Dimerization, Drug Design, Epitopes chemistry, Epitopes immunology, Humans, Models, Molecular, Protein Conformation, Protein Domains, Species Specificity, Structure-Activity Relationship, Viral Envelope Proteins chemistry, Viral Envelope Proteins immunology, Virion immunology, Antibodies, Monoclonal immunology, Antibodies, Neutralizing immunology, Dengue prevention & control, Viral Vaccines immunology, Zika Virus Infection prevention & control
- Abstract
The unprecedented 2015-2016 Zika outbreak in the Americas sparked global concern and drove the rapid deployment of vaccine and therapeutic countermeasures against this re-emerging pathogen. Alongside vaccine development, a number of potent neutralizing antibodies against Zika and related flaviviruses have been identified in recent years. High-throughput antibody isolation approaches have contributed to a better understanding of the B cell responses elicited following infection and/or vaccination. Structure-based approaches have illuminated species-specific and cross-protective epitopes of therapeutic value. This review will highlight previously described monoclonal antibodies with the best therapeutic potential against ZIKV and related flaviviruses, and discuss their implications for the rational design of better vaccine strategies., 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 Dussupt, Modjarrad and Krebs.)
- Published
- 2021
- Full Text
- View/download PDF
40. RV144 HIV-1 vaccination impacts post-infection antibody responses.
- Author
-
Mdluli T, Jian N, Slike B, Paquin-Proulx D, Donofrio G, Alrubayyi A, Gift S, Grande R, Bryson M, Lee A, Dussupt V, Mendez-Riveria L, Sanders-Buell E, Chenine AL, Tran U, Li Y, Brown E, Edlefsen PT, O'Connell R, Gilbert P, Nitayaphan S, Pitisuttihum P, Rerks-Ngarm S, Robb ML, Gramzinski R, Alter G, Tovanabutra S, Georgiev IS, Ackerman ME, Polonis VR, Vasan S, Michael NL, Kim JH, Eller MA, Krebs SJ, and Rolland M
- Subjects
- Adult, Antibody Formation immunology, B-Lymphocytes immunology, Female, HIV Antibodies blood, HIV-1, Humans, Immunoglobulin G immunology, Male, Middle Aged, env Gene Products, Human Immunodeficiency Virus immunology, AIDS Vaccines immunology, HIV Antibodies immunology, HIV Infections immunology, HIV Infections prevention & control
- Abstract
The RV144 vaccine efficacy clinical trial showed a reduction in HIV-1 infections by 31%. Vaccine efficacy was associated with stronger binding antibody responses to the HIV Envelope (Env) V1V2 region, with decreased efficacy as responses wane. High levels of Ab-dependent cellular cytotoxicity (ADCC) together with low plasma levels of Env-specific IgA also correlated with decreased infection risk. We investigated whether B cell priming from RV144 vaccination impacted functional antibody responses to HIV-1 following infection. Antibody responses were assessed in 37 vaccine and 63 placebo recipients at 6, 12, and 36 months following HIV diagnosis. The magnitude, specificity, dynamics, subclass recognition and distribution of the binding antibody response following infection were different in RV144 vaccine recipients compared to placebo recipients. Vaccine recipients demonstrated increased IgG1 binding specifically to V1V2, as well as increased IgG2 and IgG4 but decreased IgG3 to HIV-1 Env. No difference in IgA binding to HIV-1 Env was detected between the vaccine and placebo recipients following infection. RV144 vaccination limited the development of broadly neutralizing antibodies post-infection, but enhanced Fc-mediated effector functions indicating B cell priming by RV144 vaccination impacted downstream antibody function. However, these functional responses were not associated with clinical markers of disease progression. These data reveal that RV144 vaccination primed B cells towards specific binding and functional antibody responses following HIV-1 infection., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2020
- Full Text
- View/download PDF
41. A high-throughput multiplex assay to characterize flavivirus-specific immunoglobulins.
- Author
-
Merbah M, Wollen-Roberts S, Shubin Z, Li Y, Bai H, Dussupt V, Mendez-Rivera L, Slike B, Krebs SJ, Modjarrad K, Michael NL, and Rolland M
- Subjects
- Antibodies, Viral immunology, Antibody Specificity, Biomarkers blood, Cross Reactions, Diagnosis, Differential, Flavivirus Infections blood, Flavivirus Infections immunology, Flavivirus Infections virology, Immunoglobulins immunology, Predictive Value of Tests, Reproducibility of Results, Antibodies, Viral blood, Flavivirus immunology, Flavivirus Infections diagnosis, High-Throughput Screening Assays, Immunoglobulins blood, Serologic Tests
- Abstract
Genus Flavivirus, which includes 53 virus species, is the leading cause of arthropod-borne diseases in humans. Diagnosis of these viral diseases is complicated by their overlapping epidemiology and clinical manifestations, and the fact that cross-reactive antibody responses are frequently elicited by individuals in response to infection. We developed a bead-based immunoassay to concomitantly profile the isotype and subclass of antibody responses (five isotypes and four subclasses) in parallel with specificity against multiple antigens. Our panel included 22 envelope (E) and non-structural 1 (NS1) proteins of different flaviviruses (Zika (ZIKV), Dengue (DENV), Yellow Fever (YFV), West Nile (WNV), Japanese Encephalitis (JEV) and Tick-Borne Encephalitis (TBEV)) and the envelope protein of Chikungunya virus (CHIKV). Using 54 samples from 40 individuals with ZIKV infection that had been pre-characterized, we identified 1) stronger ZIKV responses in individuals previously exposed to flavivirus compared to flavivirus-naïve individuals; 2) different antibody isotypes depending on the stage of infection: acute, convalescent and late convalescent; 3) cross-reactive responses; and 4) a potential CHIKV infection. The assay had a broad dynamic range (>5 logs) and has the potential to distinguish antigen-specific responses induced by ZIKV infection from cross-reactive responses. The multidimensional data provided by this high-throughput antibody-profiling platform can advance our understanding of the human immune response to flaviviruses as they expand their global reach., (Copyright © 2020 The Authors. Published by Elsevier B.V. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
42. A recombinant gp145 Env glycoprotein from HIV-1 expressed in two different cell lines: Effects on glycosylation and antigenicity.
- Author
-
González-Feliciano JA, Akamine P, Capó-Vélez CM, Delgado-Vélez M, Dussupt V, Krebs SJ, Wojna V, Polonis VR, Baerga-Ortiz A, and Lasalde-Dominicci JA
- Subjects
- Adult, Animals, Antibodies, Neutralizing immunology, Antibodies, Viral blood, Antibodies, Viral immunology, Antigen-Antibody Reactions, CHO Cells, Cricetinae, Cricetulus, Female, Glycosylation, HEK293 Cells, Humans, Middle Aged, Recombinant Proteins biosynthesis, Recombinant Proteins immunology, Recombinant Proteins isolation & purification, Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization, Young Adult, env Gene Products, Human Immunodeficiency Virus genetics, env Gene Products, Human Immunodeficiency Virus metabolism, Glycopeptides analysis, HIV Antibodies immunology, HIV-1 metabolism, env Gene Products, Human Immunodeficiency Virus immunology
- Abstract
The envelope glycoprotein (Env) of the human immunodeficiency virus (HIV), has been the primary target for the development of a protective vaccine against infection. The extensive N-linked glycosylation on Env is an important consideration as it may affect efficacy, stability, and expression yields. The expression host has been shown to influence the extent and type of glycosylation that decorates the protein target. Here, we report the glycosylation profile of the candidate subtype C immunogen CO6980v0c22 gp145, which is currently in Phase I clinical trials, produced in two different host cells: CHO-K1 and Expi293F. The amino acid sequence for both glycoproteins was confirmed to be identical by peptide mass fingerprinting. However, the isoelectric point of the proteins differed; 4.5-5.5 and 6.0-7.0 for gp145 produced in CHO-K1 and Expi293F, respectively. These differences in pI were eliminated by enzymatic treatment with sialidase, indicating a large difference in the incorporation of sialic acid between hosts. This dramatic difference in the number of sialylated glycans between hosts was confirmed by analysis of PNGase F-released glycans using MALDI-ToF MS. These differences in glycosylation, however, did not greatly translate into differences in antibody recognition. Biosensor assays showed that gp145 produced in CHO-K1 had similar affinity toward the broadly neutralizing antibodies, 2G12 and PG16, as the gp145 produced in Expi293F. Additionally, both immunogens showed the same reactivity against plasma of HIV-infected patients. Taken together, these results support the notion that there are sizeable differences in the glycosylation of Env depending on the expression host. How these differences translate to vaccine efficacy remains unknown., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2020
- Full Text
- View/download PDF
43. Potent Zika and dengue cross-neutralizing antibodies induced by Zika vaccination in a dengue-experienced donor.
- Author
-
Dussupt V, Sankhala RS, Gromowski GD, Donofrio G, De La Barrera RA, Larocca RA, Zaky W, Mendez-Rivera L, Choe M, Davidson E, McCracken MK, Brien JD, Abbink P, Bai H, Bryan AL, Bias CH, Berry IM, Botero N, Cook T, Doria-Rose NA, Escuer AGI, Frimpong JA, Geretz A, Hernandez M, Hollidge BS, Jian N, Kabra K, Leggat DJ, Liu J, Pinto AK, Rutvisuttinunt W, Setliff I, Tran U, Townsley S, Doranz BJ, Rolland M, McDermott AB, Georgiev IS, Thomas R, Robb ML, Eckels KH, Barranco E, Koren M, Smith DR, Jarman RG, George SL, Stephenson KE, Barouch DH, Modjarrad K, Michael NL, Joyce MG, and Krebs SJ
- Subjects
- Animals, Antibodies, Monoclonal immunology, Antibodies, Neutralizing immunology, Antibodies, Viral immunology, Chlorocebus aethiops, Cross Reactions, Dengue Virus, Epitope Mapping, Female, Flavivirus metabolism, Humans, Immunoglobulin G chemistry, Inhibitory Concentration 50, Mice, Mice, Inbred BALB C, Mice, Inbred C57BL, Protein Binding, Protein Domains, Vaccination, Vaccines, Inactivated therapeutic use, Vero Cells, Viremia, Zika Virus, Dengue immunology, Tissue Donors, Viral Vaccines therapeutic use, Zika Virus Infection immunology, Zika Virus Infection prevention & control
- Abstract
Zika virus (ZIKV) has caused significant disease, with widespread cases of neurological pathology and congenital neurologic defects. Rapid vaccine development has led to a number of candidates capable of eliciting potent ZIKV-neutralizing antibodies (reviewed in refs.
1-3 ). Despite advances in vaccine development, it remains unclear how ZIKV vaccination affects immune responses in humans with prior flavivirus immunity. Here we show that a single-dose immunization of ZIKV purified inactivated vaccine (ZPIV)4-7 in a dengue virus (DENV)-experienced human elicited potent cross-neutralizing antibodies to both ZIKV and DENV. Using a unique ZIKV virion-based sorting strategy, we isolated and characterized multiple antibodies, including one termed MZ4, which targets a novel site of vulnerability centered on the Envelope (E) domain I/III linker region and protects mice from viremia and viral dissemination following ZIKV or DENV-2 challenge. These data demonstrate that Zika vaccination in a DENV-experienced individual can boost pre-existing flavivirus immunity and elicit protective responses against both ZIKV and DENV. ZPIV vaccination in Puerto Rican individuals with prior flavivirus experience yielded similar cross-neutralizing potency after a single vaccination, highlighting the potential benefit of ZIKV vaccination in flavivirus-endemic areas.- Published
- 2020
- Full Text
- View/download PDF
44. Multimeric Epitope-Scaffold HIV Vaccines Target V1V2 and Differentially Tune Polyfunctional Antibody Responses.
- Author
-
Hessell AJ, Powell R, Jiang X, Luo C, Weiss S, Dussupt V, Itri V, Fox A, Shapiro MB, Pandey S, Cheever T, Fuller DH, Park B, Krebs SJ, Totrov M, Haigwood NL, Kong XP, and Zolla-Pazner S
- Subjects
- Animals, Antibodies, Monoclonal immunology, Antibodies, Neutralizing immunology, Antibodies, Viral immunology, Antibody Affinity immunology, Cell Line, Exudates and Transudates, Female, Humans, Immunization, Macaca mulatta, Male, Mucous Membrane immunology, Vagina immunology, Vagina virology, AIDS Vaccines immunology, Antibody Formation immunology, Epitopes immunology, HIV Infections immunology, Protein Multimerization, env Gene Products, Human Immunodeficiency Virus immunology
- Abstract
The V1V2 region of the HIV-1 envelope is the target of several broadly neutralizing antibodies (bNAbs). Antibodies to V1V2 elicited in the RV144 clinical trial correlated with a reduced risk of HIV infection, but these antibodies were without broad neutralizing activity. Antibodies targeting V1V2 also correlated with a reduced viral load in immunized macaques challenged with simian immunodeficiency virus (SIV) or simian/human immunodeficiency virus (SHIV). To focus immune responses on V1V2, we engrafted the native, glycosylated V1V2 domain onto five different multimeric scaffold proteins and conducted comparative immunogenicity studies in macaques. Vaccinated macaques developed high titers of plasma and mucosal antibodies that targeted structurally distinct V1V2 epitopes. Plasma antibodies displayed limited neutralizing activity but were functionally active for ADCC and phagocytosis, which was detectable 1-2 years after immunizations ended. This study demonstrates that multivalent, glycosylated V1V2-scaffold protein immunogens focus the antibody response on V1V2 and are differentially effective at inducing polyfunctional antibodies with characteristics associated with protection., (Copyright © 2019 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
45. Longitudinal Analysis Reveals Early Development of Three MPER-Directed Neutralizing Antibody Lineages from an HIV-1-Infected Individual.
- Author
-
Krebs SJ, Kwon YD, Schramm CA, Law WH, Donofrio G, Zhou KH, Gift S, Dussupt V, Georgiev IS, Schätzle S, McDaniel JR, Lai YT, Sastry M, Zhang B, Jarosinski MC, Ransier A, Chenine AL, Asokan M, Bailer RT, Bose M, Cagigi A, Cale EM, Chuang GY, Darko S, Driscoll JI, Druz A, Gorman J, Laboune F, Louder MK, McKee K, Mendez L, Moody MA, O'Sullivan AM, Owen C, Peng D, Rawi R, Sanders-Buell E, Shen CH, Shiakolas AR, Stephens T, Tsybovsky Y, Tucker C, Verardi R, Wang K, Zhou J, Zhou T, Georgiou G, Alam SM, Haynes BF, Rolland M, Matyas GR, Polonis VR, McDermott AB, Douek DC, Shapiro L, Tovanabutra S, Michael NL, Mascola JR, Robb ML, Kwong PD, and Doria-Rose NA
- Subjects
- AIDS Vaccines immunology, Amino Acid Sequence, B-Lymphocytes immunology, Cell Line, HEK293 Cells, HIV Infections immunology, Humans, Leukocytes, Mononuclear, Longitudinal Studies, Antibodies, Neutralizing immunology, HIV Antibodies immunology, HIV-1 immunology
- Abstract
Lineage-based vaccine design is an attractive approach for eliciting broadly neutralizing antibodies (bNAbs) against HIV-1. However, most bNAb lineages studied to date have features indicative of unusual recombination and/or development. From an individual in the prospective RV217 cohort, we identified three lineages of bNAbs targeting the membrane-proximal external region (MPER) of the HIV-1 envelope. Antibodies RV217-VRC42.01, -VRC43.01, and -VRC46.01 used distinct modes of recognition and neutralized 96%, 62%, and 30%, respectively, of a 208-strain virus panel. All three lineages had modest levels of somatic hypermutation and normal antibody-loop lengths and were initiated by the founder virus MPER. The broadest lineage, VRC42, was similar to the known bNAb 4E10. A multimeric immunogen based on the founder MPER activated B cells bearing the unmutated common ancestor of VRC42, with modest maturation of early VRC42 intermediates imparting neutralization breadth. These features suggest that VRC42 may be a promising template for lineage-based vaccine design., (Published by Elsevier Inc.)
- Published
- 2019
- Full Text
- View/download PDF
46. HIV-1 Nucleocapsid Mimics the Membrane Adaptor Syntenin PDZ to Gain Access to ESCRTs and Promote Virus Budding.
- Author
-
Sette P, O'Connor SK, Yerramilli VS, Dussupt V, Nagashima K, Chutiraka K, Lingappa J, Scarlata S, and Bouamr F
- Subjects
- Cell Line, Humans, Protein Binding, Calcium-Binding Proteins metabolism, Cell Cycle Proteins metabolism, Endosomal Sorting Complexes Required for Transport metabolism, HIV-1 physiology, T-Lymphocytes virology, Virus Release, gag Gene Products, Human Immunodeficiency Virus metabolism
- Abstract
HIV-1 recruits cellular endosomal sorting complexes required for transport (ESCRTs) to bud virions from the membrane. Disruption of the viral nucleocapsid (NC) domain integrity affects HIV-1 budding. However, the molecular mechanisms of NC's involvement in HIV budding remain unclear. We find that NC mimics the PDZ domains of syntenin, a membrane-binding adaptor involved in cell-to-cell contact/communication, to capture the Bro1 domain of ALIX, which is an ESCRTs recruiting cellular adaptor. NC binds membranes via basic residues in either the distal or proximal zinc fingers, and NC-membrane binding is essential for Bro1 capture and HIV-1 budding. Removal of RNA enhances NC membrane binding, suggesting a dynamic competition between membrane lipids and RNA for the same binding sites in NC. Remarkably, syntenin PDZ can substitute for NC function in HIV-1 budding. Thus, NC mimics syntenin PDZs to function as a membrane-binding adaptor critical for HIV-1 budding at specific microdomains of the membrane., (Copyright © 2016 Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
47. Comparable Antigenicity and Immunogenicity of Oligomeric Forms of a Novel, Acute HIV-1 Subtype C gp145 Envelope for Use in Preclinical and Clinical Vaccine Research.
- Author
-
Wieczorek L, Krebs SJ, Kalyanaraman V, Whitney S, Tovanabutra S, Moscoso CG, Sanders-Buell E, Williams C, Slike B, Molnar S, Dussupt V, Alam SM, Chenine AL, Tong T, Hill EL, Liao HX, Hoelscher M, Maboko L, Zolla-Pazner S, Haynes BF, Pensiero M, McCutchan F, Malek-Salehi S, Cheng RH, Robb ML, VanCott T, Michael NL, Marovich MA, Alving CR, Matyas GR, Rao M, and Polonis VR
- Subjects
- AIDS Vaccines administration & dosage, AIDS Vaccines genetics, Animals, Drug Evaluation, Preclinical, HIV Antibodies immunology, HIV Infections prevention & control, HIV Infections virology, HIV-1 genetics, Humans, Leukocytes, Mononuclear immunology, Molecular Sequence Data, Neutralization Tests, Rabbits, Vaccination, env Gene Products, Human Immunodeficiency Virus administration & dosage, env Gene Products, Human Immunodeficiency Virus genetics, AIDS Vaccines immunology, HIV Infections immunology, HIV-1 immunology, env Gene Products, Human Immunodeficiency Virus immunology
- Abstract
Unlabelled: Eliciting broadly reactive functional antibodies remains a challenge in human immunodeficiency virus type 1 (HIV-1) vaccine development that is complicated by variations in envelope (Env) subtype and structure. The majority of new global HIV-1 infections are subtype C, and novel antigenic properties have been described for subtype C Env proteins. Thus, an HIV-1 subtype C Env protein (CO6980v0c22) from an infected person in the acute phase (Fiebig stage I/II) was developed as a research reagent and candidate immunogen. The gp145 envelope is a novel immunogen with a fully intact membrane-proximal external region (MPER), extended by a polylysine tail. Soluble gp145 was enriched for trimers that yielded the expected "fan blade" motifs when visualized by cryoelectron microscopy. CO6980v0c22 gp145 reacts with the 4E10, PG9, PG16, and VRC01 HIV-1 neutralizing monoclonal antibodies (MAbs), as well as the V1/V2-specific PGT121, 697, 2158, and 2297 MAbs. Different gp145 oligomers were tested for immunogenicity in rabbits, and purified dimers, trimers, and larger multimers elicited similar levels of cross-subtype binding and neutralizing antibodies to tier 1 and some tier 2 viruses. Immunized rabbit sera did not neutralize the highly resistant CO6980v0c22 pseudovirus but did inhibit the homologous infectious molecular clone in a peripheral blood mononuclear cell (PBMC) assay. This Env is currently in good manufacturing practice (GMP) production to be made available for use as a clinical research tool and further evaluation as a candidate vaccine., Importance: At present, the product pipeline for HIV vaccines is insufficient and is limited by inadequate capacity to produce large quantities of vaccine to standards required for human clinical trials. Such products are required to evaluate critical questions of vaccine formulation, route, dosing, and schedule, as well as to establish vaccine efficacy. The gp145 Env protein presented in this study forms physical trimers, binds to many of the well-characterized broad neutralizing MAbs that target conserved Env epitopes, and induce cross-subtype neutralizing antibodies as measured in both cell line and primary cell assays. This subtype C Env gp145 protein is currently undergoing good manufacturing practice production for use as a reagent for preclinical studies and for human clinical research. This product will serve as a reagent for comparative studies and may represent a next-generation candidate HIV immunogen., (Copyright © 2015, American Society for Microbiology. All Rights Reserved.)
- Published
- 2015
- Full Text
- View/download PDF
48. Evaluating baculovirus as a vector for human prostate cancer gene therapy.
- Author
-
Swift SL, Rivera GC, Dussupt V, Leadley RM, Hudson LC, Ma de Ridder C, Kraaij R, Burns JE, Maitland NJ, and Georgopoulos LJ
- Subjects
- Animals, Cell Line, Cell Line, Tumor, Cell Survival genetics, Cell Survival physiology, Flow Cytometry, Humans, Male, Mice, Mice, Nude, Microscopy, Confocal, Tumor Cells, Cultured, Xenograft Model Antitumor Assays, Baculoviridae genetics, Genetic Therapy methods, Genetic Vectors genetics, Prostatic Neoplasms therapy
- Abstract
Gene therapy represents an attractive strategy for the non-invasive treatment of prostate cancer, where current clinical interventions show limited efficacy. Here, we evaluate the use of the insect virus, baculovirus (BV), as a novel vector for human prostate cancer gene therapy. Since prostate tumours represent a heterogeneous environment, a therapeutic approach that achieves long-term regression must be capable of targeting multiple transformed cell populations. Furthermore, discrimination in the targeting of malignant compared to non-malignant cells would have value in minimising side effects. We employed a number of prostate cancer models to analyse the potential for BV to achieve these goals. In vitro, both traditional prostate cell lines as well as primary epithelial or stromal cells derived from patient prostate biopsies, in two- or three-dimensional cultures, were used. We also evaluated BV in vivo in murine prostate cancer xenograft models. BV was capable of preferentially transducing invasive malignant prostate cancer cell lines compared to early stage cancers and non-malignant samples, a restriction that was not a function of nuclear import. Of more clinical relevance, primary patient-derived prostate cancer cells were also efficiently transduced by BV, with robust rates observed in epithelial cells of basal phenotype, which expressed BV-encoded transgenes faster than epithelial cells of a more differentiated, luminal phenotype. Maximum transduction capacity was observed in stromal cells. BV was able to penetrate through three-dimensional structures, including in vitro spheroids and in vivo orthotopic xenografts. BV vectors containing a nitroreductase transgene in a gene-directed enzyme pro-drug therapy approach were capable of efficiently killing malignant prostate targets following administration of the pro-drug, CB1954. Thus, BV is capable of transducing a large proportion of prostate cell types within a heterogeneous 3-D prostate tumour, can facilitate cell death using a pro-drug approach, and shows promise as a vector for the treatment of prostate cancer.
- Published
- 2013
- Full Text
- View/download PDF
49. Identification of the HIV-1 NC binding interface in Alix Bro1 reveals a role for RNA.
- Author
-
Sette P, Dussupt V, and Bouamr F
- Subjects
- Binding Sites, Calcium-Binding Proteins genetics, Cell Cycle Proteins genetics, Cell Line, Endosomal Sorting Complexes Required for Transport genetics, HIV-1 physiology, Humans, Protein Binding, Virus Release, Calcium-Binding Proteins metabolism, Cell Cycle Proteins metabolism, Endosomal Sorting Complexes Required for Transport metabolism, Host-Pathogen Interactions, Protein Interaction Domains and Motifs, RNA, Viral metabolism, gag Gene Products, Human Immunodeficiency Virus metabolism
- Abstract
HIV-1 recruits members of ESCRT, the cell membrane fission machinery that promotes virus exit. HIV-1 Gag protein gains access to ESCRT directly by binding Alix, an ESCRT-associated protein that promotes budding. The Alix Bro1 and V domains bind Gag NC and p6 regions, respectively. Whereas V-p6 binding and function are well characterized, residues in Bro1 that interact with NC and their functional contribution to Alix-mediated HIV-1 budding are unknown. We mapped Bro1 residues that constitute the NC binding interface and found that they are critical for function. Intriguingly, residues involved in interactions on both sides of the Bro1-NC interface are positively charged, suggesting the involvement of a negatively charged cellular factor serving as a bridge. Nuclease treatment eliminated Bro1-NC interactions, revealing the involvement of RNA. These findings establish a direct role for NC in mediating interactions with ESCRT necessary for virus release and report the first evidence of RNA involvement in such recruitments.
- Published
- 2012
- Full Text
- View/download PDF
50. Two distinct binding modes define the interaction of Brox with the C-terminal tails of CHMP5 and CHMP4B.
- Author
-
Mu R, Dussupt V, Jiang J, Sette P, Rudd V, Chuenchor W, Bello NF, Bouamr F, and Xiao TS
- Subjects
- Amino Acid Sequence, Binding Sites, Cell Cycle Proteins chemistry, Cell Cycle Proteins metabolism, Endosomal Sorting Complexes Required for Transport metabolism, Endosomes metabolism, HEK293 Cells, Humans, Molecular Sequence Data, Protein Structure, Secondary, Protein Structure, Tertiary, Endosomal Sorting Complexes Required for Transport chemistry
- Abstract
Interactions of the CHMP protein carboxyl terminal tails with effector proteins play important roles in retroviral budding, cytokinesis, and multivesicular body biogenesis. Here we demonstrate that hydrophobic residues at the CHMP4B C-terminal amphipathic α helix bind a concave surface of Brox, a mammalian paralog of Alix. Unexpectedly, CHMP5 was also found to bind Brox and specifically recruit endogenous Brox to detergent-resistant membrane fractions through its C-terminal 20 residues. Instead of an α helix, the CHMP5 C-terminal tail adopts a tandem β-hairpin structure that binds Brox at the same site as CHMP4B. Additional Brox:CHMP5 interface is furnished by a unique CHMP5 hydrophobic pocket engaging the Brox residue Y348 that is not conserved among the Bro1 domains. Our studies thus unveil a β-hairpin conformation of the CHMP5 protein C-terminal tail, and provide insights into the overlapping but distinct binding profiles of ESCRT-III and the Bro1 domain proteins., (Copyright © 2012 Elsevier Ltd. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.