94 results on '"Kate E Broderick"'
Search Results
2. Establishment of a Pig Influenza Challenge Model for Evaluation of Monoclonal Antibody Delivery Platforms
- Author
-
Pramila Rijal, Paul Fisher, Becky Clark, Alain Townsend, Elma Tchilian, Tiphany Chrun, Emily Bessell, Katherine Schultheis, Peter C. L. Beverley, Simon P. Graham, Trevor R.F. Smith, Jeffery K. Taubenberger, Veronica Martini, Sarah T. C. Elliott, Barbara Holzer, Heather Brown, David B. Weiner, Alejandro Núñez, John C. Kash, Ghiabe Guibinga, Ami Patel, Stephanie Ramos, Yongli Xiao, Adam McNee, Kate E. Broderick, and Matthieu Bernard
- Subjects
Swine ,Immunology ,Hemagglutinin (influenza) ,Hemagglutinin Glycoproteins, Influenza Virus ,Antibodies, Viral ,medicine.disease_cause ,Virus ,03 medical and health sciences ,Influenza A Virus, H1N1 Subtype ,0302 clinical medicine ,Orthomyxoviridae Infections ,Influenza, Human ,Pandemic ,Influenza A virus ,Animals ,Immunology and Allergy ,Medicine ,Viral shedding ,Immunotherapy and Vaccines ,biology ,business.industry ,Antibodies, Monoclonal ,respiratory system ,Antibodies, Neutralizing ,Virology ,Vaccination ,Monoclonal ,biology.protein ,business ,Viral load ,030215 immunology - Abstract
Key Points Neutralizing mAb 2–12C reduces influenza viral load and lung pathology in pigs. DNA plasmid–encoded 2–12C reduces lung pathology. The pig is a useful preclinical model for testing mAbs and mAb delivery platforms., mAbs are a possible adjunct to vaccination and drugs in treatment of influenza virus infection. However, questions remain whether small animal models accurately predict efficacy in humans. We have established the pig, a large natural host animal for influenza, with many physiological similarities to humans, as a robust model for testing mAbs. We show that a strongly neutralizing mAb (2–12C) against the hemagglutinin head administered prophylactically at 15 mg/kg reduced viral load and lung pathology after pandemic H1N1 influenza challenge. A lower dose of 1 mg/kg of 2–12C or a DNA plasmid–encoded version of 2–12C reduced pathology and viral load in the lungs but not viral shedding in nasal swabs. We propose that the pig influenza model will be useful for testing candidate mAbs and emerging delivery platforms prior to human trials.
- Published
- 2020
- Full Text
- View/download PDF
3. Active immunoprophylaxis with a synthetic DNA-encoded monoclonal anti-respiratory syncytial virus scFv-Fc fusion protein confers protection against infection and durable activity
- Author
-
David B. Weiner, Laurent Humeau, Jian Yan, Trevor R.F. Smith, Holly Pugh, Katherine Schultheis, Jing Chen, Janet Oh, Jacklyn Nguyen, Bryan S. Yung, Karuppiah Muthumani, Neil Cooch, Charles C. Reed, and Kate E. Broderick
- Subjects
electroporation ,medicine.drug_class ,viruses ,030231 tropical medicine ,Immunology ,Respiratory Syncytial Virus Infections ,Respiratory syncytial virus ,Antibodies, Viral ,Monoclonal antibody ,Virus ,03 medical and health sciences ,plasmid DNA ,0302 clinical medicine ,Immunity ,In vivo ,Respiratory Syncytial Virus Vaccines ,medicine ,Animals ,Immunology and Allergy ,Tissue Distribution ,Sigmodontinae ,030212 general & internal medicine ,Cotton rat ,Pharmacology ,DNA-encoded antibody ,biology ,Antibodies, Monoclonal ,cotton rat ,virus diseases ,respiratory system ,active immunophrophylaxis ,biology.organism_classification ,Antibodies, Neutralizing ,Virology ,Fusion protein ,single-chain antibody ,Monoclonal ,biology.protein ,Antibody ,Viral Fusion Proteins ,Research Article ,Research Paper - Abstract
Respiratory Syncytial virus (RSV) is a major threat to many vulnerable populations. There are currently no approved vaccines, and RSV remains a high unmet global medical need. Here we describe the employment of a novel synthetic DNA-encoded antibody technology platform to develop and deliver an engineered human DNA-encoded monoclonal antibody (dMAbTM) targeting the fusion protein (F) of RSV as a new approach to prevention or therapy of at risk populations. In in vivo models, a single administration of synthetic DNA-encoding the single-chain fragment variable-constant fragment (scFv-Fc) RSV-F dMAb resulted in robust and durable circulating levels of a functional antibody systemically and in mucosal tissue. In cotton rats, which are the gold-standard animals to model RSV infection, we observed sustained scFv-Fc RSV-F dMAb in the sera and lung-lavage samples, demonstrating the potential for both long-lasting immunity to RSV and effective biodistribution. The scFv-Fc RSV-F dMAb harbored in the sera exhibited RSV antigen-specific binding and potent viral neutralizing activity. Importantly, in vivo delivery of synthetic DNA-encoding, the scFv-Fc RSV-F dMAb protected animals against viral challenge. Our findings support the significance of dMAbs as a potential platform technology for durable protection against RSV disease.
- Published
- 2020
- Full Text
- View/download PDF
4. Prime-boost vaccination regimens with INO-4800 and INO-4802 augment and broaden immune responses against SARS-CoV-2 in nonhuman primates
- Author
-
Jewell Walters, Brian Nguyen, David B. Weiner, J. Joseph Kim, Trevor R.F. Smith, Abhijeet J Kulkarni, Viviane M Andrade, Alison Generotti, Emma L. Reuschel, Laurent Humeau, Dustin Elwood, Ebony N. Gary, Stephanie Ramos, Arthur Doan, Igor Maricic, Zeena Eblimit, Elizabeth Parzych, Drew Frase, Ami Patel, Katherine Schultheis, Mansi Purwar, Kate E. Broderick, Faraz I. Zaidi, and Blake Schouest
- Subjects
COVID-19 Vaccines ,Heterologous ,Biology ,DNA vaccination ,Mice ,Immune system ,Immunity ,Pandemic ,Vaccines, DNA ,Animals ,Humans ,Mice, Inbred BALB C ,General Veterinary ,General Immunology and Microbiology ,SARS-CoV-2 ,Immunogenicity ,Vaccination ,Public Health, Environmental and Occupational Health ,COVID-19 ,Viral Vaccines ,Virology ,Macaca mulatta ,Infectious Diseases ,Antibody Formation ,Molecular Medicine ,Augment - Abstract
SUMMARYThe enhanced transmissibility and immune evasion associated with emerging SARS-CoV-2 variants demands the development of next-generation vaccines capable of inducing superior protection amid a shifting pandemic landscape. Since a portion of the global population harbors some level of immunity from vaccines based on the original Wuhan-Hu-1 SARS-CoV-2 sequence or natural infection, an important question going forward is whether this immunity can be boosted by next-generation vaccines that target emerging variants while simultaneously maintaining long-term protection against existing strains. Here, we evaluated the immunogenicity of INO-4800, our synthetic DNA vaccine candidate for COVID-19 currently in clinical evaluation, and INO-4802, a next-generation DNA vaccine designed to broadly target emerging SARS-CoV-2 variants, as booster vaccines in nonhuman primates. Rhesus macaques primed over one year prior with the first-generation INO-4800 vaccine were boosted with either INO-4800 or INO-4802 in homologous or heterologous prime-boost regimens. Both boosting schedules led to an expansion of antibody responses which were characterized by improved neutralizing and ACE2 blocking activity across wild-type SARS-CoV-2 as well as multiple variants of concern. These data illustrate the durability of immunity following vaccination with INO-4800 and additionally support the use of either INO-4800 or INO-4802 in prime-boost regimens.
- Published
- 2021
5. Synthetic nucleic acid antibody prophylaxis confers rapid and durable protective immunity against Zika virus challenge
- Author
-
Gayathri Gulendran, David B. Weiner, Charles C. Reed, Sagar B. Kudchodkar, Kanika Asija, Hyeree Choi, Karuppiah Muthumani, Lucas Van Gorder, Sangya Agarwal, Piyush Borole, Don L. Siegel, Emma L. Reuschel, J. Joseph Kim, Stephanie Ramos, Kenneth E. Ugen, Gary P. Kobinger, and Kate E. Broderick
- Subjects
dMAb-DNA encoded monoclonal antibodies ,DNA vaccine ,Protective immunity ,viruses ,medicine.medical_treatment ,030231 tropical medicine ,Immunology ,Antibodies, Viral ,DNA vaccination ,Zika virus ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Nucleic Acids ,medicine ,Animals ,antibodies ,Immunology and Allergy ,030212 general & internal medicine ,Pharmacology ,Synthetic nucleic acid ,biology ,Zika Virus Infection ,business.industry ,Viral Vaccines ,Zika Virus ,Immunotherapy ,vaccination ,biology.organism_classification ,Antibodies, Neutralizing ,Virology ,Vaccination ,Flavivirus ,biology.protein ,immunotherapy ,Antibody ,business ,Research Paper - Abstract
Significant concerns have arisen over the past 3 y from the increased global spread of the mosquito-borne flavivirus, Zika. Accompanying this spread has been an increase in cases of the devastating birth defect microcephaly as well as of Guillain–Barré syndrome in adults in many affected countries. Currently there is no vaccine or therapy for this infection; however, we sought to develop a combination approach that provides more rapid and durable protection than traditional vaccination alone. A novel immune-based prophylaxis/therapy strategy entailing the facilitated delivery of a synthetic DNA consensus prME vaccine along with DNA-encoded anti-ZIKV envelope monoclonal antibodies (dMAb) were developed and evaluated for antiviral efficacy. This immediate and persistent protection strategy confers the ability to overcome shortcomings inherent with conventional active vaccination or passive immunotherapy. A collection of novel dMAbs were developed which were potent against ZIKV and could be expressed in serum within 24–48 h of in vivo administration. The DNA vaccine, from a previous development, was potent after adaptive immunity was developed, protecting against infection, brain and testes pathology in relevant mouse challenge models and in an NHP challenge. Delivery of potent dMAbs protected mice from the same murine viral challenge within days of delivery. Combined injection of dMAb and the DNA vaccine afforded rapid and long-lived protection in this challenge model, providing an important demonstration of the advantage of this synergistic approach to pandemic outbreaks.
- Published
- 2019
- Full Text
- View/download PDF
6. In Vivo Delivery of a DNA-Encoded Monoclonal Antibody Protects Non-human Primates against Zika Virus
- Author
-
Ghiabe Guibinga, Janess M. Mendoza, David B. Weiner, Laurent Humeau, Stephanie Ramos, Jian Yan, Sagar B. Kudchodkar, Trevor R.F. Smith, Kanika Asija, Jeffrey W. Allen, Piyush Borole, Jing Chen, Martina Beltramello, Ami Patel, Mamadou A. Bah, Karin Stettler, Davide Corti, Rianne Esquivel, Amy C. Durham, Hyeree Choi, Daniel H. Park, Shareef Shaheen, Kate E. Broderick, and Karuppiah Muthumani
- Subjects
Primates ,medicine.drug_class ,Antibodies, Viral ,DNA-encoded monoclonal antibody ,infectious diseases ,Monoclonal antibody ,Zika virus ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Viral Envelope Proteins ,In vivo ,antibody ,Drug Discovery ,Genetics ,medicine ,Animals ,Humans ,Neutralizing antibody ,Molecular Biology ,030304 developmental biology ,Pharmacology ,DMAb ,0303 health sciences ,biology ,Zika Virus Infection ,Antibodies, Monoclonal ,Outbreak ,DNA ,Zika Virus ,immunoprophylaxis ,protection ,biology.organism_classification ,Antibodies, Neutralizing ,Virology ,Rhesus macaque ,Infectious disease (medical specialty) ,030220 oncology & carcinogenesis ,biology.protein ,Molecular Medicine ,Original Article ,Antibody ,rhesus macaque - Abstract
Zika virus (ZIKV) infection is endemic to several world regions, and many others are at high risk for seasonal outbreaks. Synthetic DNA-encoded monoclonal antibody (DMAb) is an approach that enables in vivo delivery of highly potent mAbs to control infections. We engineered DMAb-ZK190, encoding the mAb ZK190 neutralizing antibody, which targets the ZIKV E protein DIII domain. In vivo-delivered DMAb-ZK190 achieved expression levels persisting >10 weeks in mice and >3 weeks in non-human primate (NHPs), which is protective against ZIKV infectious challenge. This study is the first demonstration of infectious disease control in NHPs following in vivo delivery of a nucleic acid-encoded antibody, supporting the importance of this new platform., Esquivel et al. deliver DNA-encoded ZK190 antibody (DMAb-ZK190) by intramuscular administration. DMAb-ZK190 targets the E protein of Zika virus, and it controls Zika virus infection in mice and non-human primates when delivered prior to infection, offering an alternative strategy for rapid protection.
- Published
- 2019
- Full Text
- View/download PDF
7. Active Immunoprophylaxis and Vaccine Augmentations Mediated by a Novel Plasmid DNA Formulation
- Author
-
Kate E. Broderick, Karuppiah Muthumani, Trevor R.F. Smith, Jacklyn Nguyen, Katherine Schultheis, Nina N. Schommer, Bryan S. Yung, David B. Weiner, and Laurent Humeau
- Subjects
Gene Expression ,Gene delivery ,Immunoglobulin G ,DNA vaccination ,Extracellular matrix ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,Immunogenicity, Vaccine ,plasmid DNA ,0302 clinical medicine ,In vivo ,Gene expression ,Vaccines, DNA ,therapeutics ,Genetics ,Animals ,Humans ,Transgenes ,Chondroitin sulfate ,gene delivery ,Muscle, Skeletal ,Molecular Biology ,030304 developmental biology ,0303 health sciences ,biology ,Electroporation ,Gene Transfer Techniques ,Original Articles ,Cell biology ,chemistry ,030220 oncology & carcinogenesis ,biology.protein ,Molecular Medicine ,Immunization ,Rabbits ,Spleen ,Plasmids - Abstract
Plasmid DNA (pDNA) gene delivery is a highly versatile technology that has the potential to address a multitude of unmet medical needs. Advances in pDNA delivery to host tissue with the employment of in vivo electroporation (EP) have led to significantly enhanced gene expression and the recent demonstration of clinical efficacy with the platform. Building upon this platform, this study reports that enzyme-mediated modification of the muscle tissue extracellular matrix structure at the site of pDNA delivery operates in a synergistic manner with EP to enhance both local and systemic gene expression further. Specifically, administration of chondroitinase ABC (Cho ABC) to the site of intramuscular delivery of pDNA led to transient disruption of chondroitin sulfate scaffolding barrier, permitting enhanced gene distribution and expression across the tissue. The employment of Cho ABC in combination with CELLECTRA® intramuscular EP resulted in increased gene expression by 5.5-fold in mice and 17.98-fold in rabbits. The study demonstrates how this protocol can be universally applied to an active prophylaxis platform to increase the in vivo production of functional immunoglobulin G, and to DNA vaccine protocols to permit drug dose sparing. The data indicate the Cho ABC formulation to be of significant value upon combination with EP to drive enhanced gene expression levels in pDNA delivery protocols.
- Published
- 2019
- Full Text
- View/download PDF
8. Novel IFN-γ ELISpot reveals robust T cell responses elicited after influenza nucleoprotein DNA vaccination in New Zealand White rabbits
- Author
-
Kate E. Broderick, Trevor R.F. Smith, Karuppiah Muthumani, Bryan S. Yung, Nikki Phanhthilath, Holly Pugh, Katherine Schultheis, and Alison Generotti
- Subjects
Enzyme-Linked Immunospot Assay ,T-Lymphocytes ,T cell ,030231 tropical medicine ,Population ,Biology ,Antiviral Agents ,DNA vaccination ,Interferon-gamma ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Antigen ,Vaccines, DNA ,medicine ,Animals ,030212 general & internal medicine ,education ,education.field_of_study ,General Veterinary ,General Immunology and Microbiology ,Viral Core Proteins ,ELISPOT ,Public Health, Environmental and Occupational Health ,RNA-Binding Proteins ,Nucleocapsid Proteins ,Vaccination ,Infectious Diseases ,medicine.anatomical_structure ,Immunization ,Influenza Vaccines ,Immunology ,Leukocytes, Mononuclear ,Molecular Medicine ,Female ,Rabbits - Abstract
The New Zealand White rabbit is a highly accessible animal model which is regularly employed in biomedical research. However, the paucity of rabbit-specific reagents available limits its use in certain fields. Specifically, the lack of a reliable T cell assay has limited its employment in immune prophylactic and therapeutic studies. To address this inadequacy, we have developed an ELISpot assay to detect cellular immune responses (IFN-γ production) after antigenic stimulation. We have applied this assay to model the T cell responses elicited by a DNA vaccine. Immunization with an influenza nucleoprotein (NP) DNA vaccine revealed strong antigen-specific T cell responses in the peripheral blood mononuclear cell population. We believe this is the first report of such an assay in rabbit species, and it will become a useful tool to monitor in vivo responses to vaccines and permit the wider adoption of this model to measure immunological responses.
- Published
- 2019
- Full Text
- View/download PDF
9. Live Virus Neutralisation of the 501Y.V1 and 501Y.V2 SARS-CoV-2 Variants following INO-4800 Vaccination of Ferrets
- Author
-
Mary Tachedjian, Kim R. Blasdell, Sarah Goldie, Alexander J. McAuley, Julian Druce, Seshadri S. Vasan, Michael J. Kuiper, Peter A. Durr, Shane Riddell, Kate E. Broderick, and Trevor R.F. Smith
- Subjects
Models, Molecular ,DNA vaccine ,0301 basic medicine ,COVID-19 Vaccines ,Immunology ,Immunization, Secondary ,biomolecular modelling ,Biology ,Antibodies, Viral ,Neutralization ,Virus ,DNA vaccination ,neutralisation ,03 medical and health sciences ,Immunogenicity, Vaccine ,0302 clinical medicine ,Immune system ,Immunity ,Pandemic ,Antigenic variation ,Animals ,Humans ,Immunology and Allergy ,Original Research ,variants ,SARS-CoV-2 antibodies ,SARS-CoV-2 ,Vaccination ,Ferrets ,COVID-19 ,RC581-607 ,Antibodies, Neutralizing ,Antigenic Variation ,Virology ,Disease Models, Animal ,030104 developmental biology ,Mutation ,Spike Glycoprotein, Coronavirus ,biology.protein ,Antibody ,Immunologic diseases. Allergy ,030217 neurology & neurosurgery ,030215 immunology - Abstract
The ongoing COVID-19 pandemic has resulted in significant global morbidity and mortality on a scale similar to the influenza pandemic of 1918. Over the course of the last few months, a number of SARS-CoV-2 variants have been identified against which vaccine-induced immune responses may be less effective. These “variants-of-concern” have garnered significant attention in the media, with discussion around their impact on the future of the pandemic and the ability of leading COVID-19 vaccines to protect against them effectively. To address concerns about emerging SARS-CoV-2 variants affecting vaccine-induced immunity, we investigated the neutralisation of representative ‘G614’, ‘501Y.V1’ and ‘501Y.V2’ virus isolates using sera from ferrets that had received prime-boost doses of the DNA vaccine, INO-4800. Neutralisation titres against G614 and 501Y.V1 were comparable, but titres against the 501Y.V2 variant were approximately 4-fold lower, similar to results reported with other nucleic acid vaccines and supported by in silico biomolecular modelling. The results confirm that the vaccine-induced neutralising antibodies generated by INO-4800 remain effective against current variants-of-concern, albeit with lower neutralisation titres against 501Y.V2 similar to other leading nucleic acid-based vaccines.
- Published
- 2021
- Full Text
- View/download PDF
10. Intradermal delivery of a synthetic DNA vaccine protects macaques from Middle East respiratory syndrome coronavirus
- Author
-
Emma L. Reuschel, Laurent Humeau, Stephanie Ramos, Jamie Lovaglio, Kevin Kim, Janess M. Mendoza, David B. Weiner, Kimberly Meade-White, Rebecca Rosenke, Karuppiah Muthumani, Faraz I. Zaidi, Kate E. Broderick, Greg Saturday, Ami Patel, Regina Stoltz, Atsushi Okumura, Dana P. Scott, Tina Thomas, Ziyang Xu, Heinz Feldmann, Patrick W. Hanley, Elaine Haddock, and Friederike Feldmann
- Subjects
0301 basic medicine ,Injections, Intradermal ,Middle East respiratory syndrome coronavirus ,viruses ,Adaptive immunity ,medicine.disease_cause ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Immunogenicity, Vaccine ,Vaccines, DNA ,Medicine ,Animals ,Infectious disease ,Vaccines ,Lung ,biology ,Transmission (medicine) ,business.industry ,Viral Vaccine ,virus diseases ,Viral Vaccines ,General Medicine ,biochemical phenomena, metabolism, and nutrition ,Acquired immune system ,Virology ,Macaca mulatta ,respiratory tract diseases ,030104 developmental biology ,medicine.anatomical_structure ,Infectious disease (medical specialty) ,030220 oncology & carcinogenesis ,Spike Glycoprotein, Coronavirus ,biology.protein ,Middle East Respiratory Syndrome Coronavirus ,Antibody ,business ,Coronavirus Infections ,Research Article - Abstract
Emerging coronaviruses from zoonotic reservoirs, including severe acute respiratory syndrome coronavirus (SARS-CoV), Middle East respiratory syndrome coronavirus (MERS-CoV), and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), have been associated with human-to-human transmission and significant morbidity and mortality. Here, we study both intradermal and intramuscular 2-dose delivery regimens of an advanced synthetic DNA vaccine candidate encoding a full-length MERS-CoV spike (S) protein, which induced potent binding and neutralizing antibodies as well as cellular immune responses in rhesus macaques. In a MERS-CoV challenge, all immunized rhesus macaques exhibited reduced clinical symptoms, lowered viral lung load, and decreased severity of pathological signs of disease compared with controls. Intradermal vaccination was dose sparing and more effective in this model at protecting animals from disease. The data support the further study of this vaccine for preventing MERS-CoV infection and transmission, including investigation of such vaccines and simplified delivery routes against emerging coronaviruses.
- Published
- 2021
11. Nucleic acid delivery of immune-focused SARS-CoV-2 nanoparticles drive rapid and potent immunogenicity capable of single-dose protection
- Author
-
Nicholas J. Tursi, Katherine Schultheis, Emma L. Reuschel, Daniel W. Kulp, Igor Maricic, Laurent Humeau, Susanne Walker, David B. Weiner, Neethu Chokkalingam, Kate E. Broderick, Kylie Konrath, Viviane Machado, Ziyang Xu, Christel Iffland, Jianqiu Du, Trevor Smith, Xizhou Zhu, Jesper Pallesen, Drew Frase, Matthew P. Sullivan, Yuanhan Wu, Alan Moore, Mansi Purwar, and Kevin Liaw
- Subjects
Immune system ,Immunization ,Antigen ,Immunity ,Immunogenicity ,medicine ,biology.protein ,Biology ,Antibody ,medicine.disease_cause ,Virology ,Epitope ,Coronavirus - Abstract
Antibodies from SARS-CoV-2 vaccines may target epitopes which reduce durability or increase the potential for escape from vaccine-induced immunity. Using a novel synthetic vaccinology pipeline, we developed rationally immune focused SARS-CoV-2 Spike-based vaccines. N-linked glycans can be employed to alter antibody responses to infection and vaccines. Utilizing computational modeling and comprehensive in vitro screening, we incorporated glycans into the Spike Receptor-Binding Domain (RBD) and assessed antigenic profiles. We developed glycan coated RBD immunogens and engineered seven multivalent configurations. Advanced DNA delivery of engineered nanoparticle vaccines rapidly elicited potent neutralizing antibodies in guinea pigs, hamsters and multiple mouse models, including human ACE2 and human B cell repertoire transgenics. RBD nanoparticles encoding wild-type and the P.1 SARS-CoV-2 variant induced high levels of cross-neutralizing antibodies. Single, low dose immunization protected against a lethal SARS-CoV-2 challenge. Single-dose coronavirus vaccines via DNA-launched nanoparticles provide a platform for rapid clinical translation of novel, potent coronavirus vaccines.
- Published
- 2021
- Full Text
- View/download PDF
12. One or two dose regimen of the SARS-CoV-2 synthetic DNA vaccine INO-4800 protects against respiratory tract disease burden in nonhuman primate challenge model
- Author
-
Mike Dennis, Karen E. Gooch, Charlotte Sarfas, Alastair Handley, Francisco J. Salguero, Kathryn A. Ryan, Stephanie Longet, Miles W. Carroll, Robert J. Watson, Holly E. Humphries, Tom Tipton, Susan A. Fotheringham, Andrew White, Laurent Humeau, Emma Rayner, Stephanie Ramos, Fergus V. Gleeson, Trevor R.F. Smith, Kate E. Broderick, Sue Charlton, Yper Hall, Gillian S. Slack, Katherine Schultheis, Sally Sharpe, and Laura Sibley
- Subjects
DNA vaccine ,COVID-19 Vaccines ,030231 tropical medicine ,Antibodies, Viral ,Article ,DNA vaccination ,Nonhuman Primate Virus Challenge ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Vaccines, DNA ,Medicine ,Animals ,Humans ,030212 general & internal medicine ,Pandemics ,General Veterinary ,General Immunology and Microbiology ,biology ,business.industry ,SARS-CoV-2 ,Viral Vaccine ,Immunogenicity ,Public Health, Environmental and Occupational Health ,COVID-19 ,Viral Vaccines ,Antibodies, Neutralizing ,Macaca mulatta ,Vaccination ,Regimen ,Infectious Diseases ,Immunology ,Spike Glycoprotein, Coronavirus ,biology.protein ,Molecular Medicine ,Nasal administration ,Antibody ,business - Abstract
Safe and effective vaccines will provide essential medical countermeasures to tackle the COVID-19 pandemic. Here, we evaluate the safety, immunogenicity and efficacy of the intradermal delivery of INO-4800, a synthetic DNA vaccine candidate encoding the SARS-CoV-2 spike protein in the rhesus macaque model. Single and 2 dose vaccination regimens were evaluated. Vaccination induces both binding and neutralizing antibodies, along with IFN-γ-producing T cells against SARS-CoV-2. Upon administration of a high viral dose (5 x 106 pfu) via the intranasal and intratracheal routes we observe significantly reduced virus load in the lung and throat, in the vaccinated animals compared to controls. 2 doses of INO-4800 is associated with more robust vaccine-induced immune responses and improved viral protection. Importantly, histopathological examination of lung tissue provides no indication of vaccine-enhanced disease following SARS-CoV-2 challenge in INO-4800 immunized animals. This vaccine candidate is currently under clinical evaluation as a 2 dose regimen.
- Published
- 2021
13. INO-4800 DNA Vaccine Induces Neutralizing Antibodies and T cell Activity Against Global SARS-CoV-2 Variants
- Author
-
Joseph Agnes, Aaron Christensen-Quick, Emma L. Reuschel, Kim Kraynyak, Jared Tur, Mammen P Mammen, Viviane M Andrade, Trevor R.F. Smith, Katherine Schultheis, Dustin Elwood, Patrick Pezzoli, Jean D. Boyer, Idania Marrero, Laurent Humeau, Stephanie Ramos, David B. Weiner, J. Joseph Kim, Trevor McMullan, Albert Sylvester, Pablo Tebas, Charles C. Reed, Kate E. Broderick, Mansi Purwar, and Richa Kalia
- Subjects
Pharmacology ,biology ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,T cell ,Immunology ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC581-607 ,biochemical phenomena, metabolism, and nutrition ,Brief Communication ,Pathogenicity ,Virology ,DNA vaccines ,DNA vaccination ,Vaccination ,Infectious Diseases ,Immune system ,medicine.anatomical_structure ,Viral infection ,Immunity ,biology.protein ,medicine ,Pharmacology (medical) ,Immunologic diseases. Allergy ,Antibody ,RC254-282 - Abstract
Global surveillance has identified emerging SARS-CoV-2 variants of concern (VOC) associated with broadened host specificity, pathogenicity, and immune evasion to vaccine-induced immunity. Here we compared humoral and cellular responses against SARS-CoV-2 VOC in subjects immunized with the DNA vaccine, INO-4800. INO-4800 vaccination induced neutralizing antibodies against all variants tested, with reduced levels detected against B.1.351. IFNγ T cell responses were fully maintained against all variants tested.
- Published
- 2021
- Full Text
- View/download PDF
14. Multivalent DNA Vaccines as A Strategy to Combat Multiple Concurrent Epidemics: Mosquito-Borne and Hemorrhagic Fever Viruses
- Author
-
Jared Tur, Sagar B. Kudchodkar, Jingjing Jiang, Ami Patel, Dustin Elwood, Kate E. Broderick, Connie S. Schmaljohn, Karuppiah Muthumani, Kathleen A. Cashman, Jian Yan, Holly Pugh, Katherine Schultheis, Preeti Bangalore, David B. Weiner, Laurent Humeau, Stephanie Ramos, and Jewell Walters
- Subjects
0301 basic medicine ,viruses ,lcsh:QR1-502 ,Immunological memory ,immunogenicity ,Antibodies, Viral ,medicine.disease_cause ,lcsh:Microbiology ,Dengue fever ,Dengue ,Mice ,0302 clinical medicine ,Vaccines, DNA ,030212 general & internal medicine ,Chikungunya ,Arenaviruses, New World ,Zika Virus Infection ,Immunogenicity ,Vaccination ,Lassa ,Ebolavirus ,in vivo electroporation ,Africa, Western ,Infectious Diseases ,Ebola ,Multivalent vaccine platform ,Female ,DNA vaccine ,Guinea Pigs ,Biology ,Article ,DNA vaccination ,03 medical and health sciences ,Marburg ,Lassa Fever ,Immune system ,Zika ,Virology ,medicine ,Animals ,Vaccines, Combined ,Epidemics ,Outbreak ,Viral Vaccines ,Zika Virus ,Dengue Virus ,Hemorrhagic Fever, Ebola ,medicine.disease ,Immunity, Humoral ,Mice, Inbred C57BL ,Culicidae ,030104 developmental biology ,Marburgvirus ,Immunization - Abstract
The emergence of multiple concurrent infectious diseases localized in the world creates a complex burden on global public health systems. Outbreaks of Ebola, Lassa, and Marburg viruses in overlapping regions of central and West Africa and the co-circulation of Zika, Dengue, and Chikungunya viruses in areas with A. aegypti mosquitos highlight the need for a rapidly deployable, safe, and versatile vaccine platform readily available to respond. The DNA vaccine platform stands out as such an application. Here, we present proof-of-concept studies from mice, guinea pigs, and non-human primates for two multivalent DNA vaccines delivered using in vivo electroporation (EP) targeting mosquito-borne (MMBV) and hemorrhagic fever (MHFV) viruses. Immunization with MMBV or MHFV vaccines via intradermal EP delivery generated robust cellular and humoral immune responses against all target viral antigens in all species. MMBV vaccine generated antigen-specific binding antibodies and IFNγ-secreting lymphocytes detected in NHPs up to six months post final immunization, suggesting induction of long-term immune memory. Serum from MHFV vaccinated NHPs demonstrated neutralizing activity in Ebola, Lassa, and Marburg pseudovirus assays indicating the potential to offer protection. Together, these data strongly support and demonstrate the versatility of DNA vaccines as a multivalent vaccine development platform for emerging infectious diseases.
- Published
- 2021
15. Safety and immunogenicity of INO-4800 DNA vaccine against SARS-CoV-2: A preliminary report of an open-label, Phase 1 clinical trial
- Author
-
Karen R. Buttigieg, Emma L. Reuschel, Trevor R.F. Smith, Jean D. Boyer, Malissa Diehl, Elisabeth Gillespie, Laurent Humeau, Stephanie Ramos, Mammen P Mammen, J. Joseph Kim, John Ervin, Yaya Dia, Miles W. Carroll, Aaron Christensen-Quick, Kate E. Broderick, Katherine Schultheis, Jan Pawlicki, Jacqueline E. Shea, Igor Maricic, Michael J. Dallas, Shu Ping Yang, Elliott Blackwood, Drew Frase, Kimberly A. Kraynyak, Mansi Purwar, Ami Shah Brown, Jessica Lee, Faraz I. Zaidi, Kevin Kim, Trevor McMullan, David B. Weiner, Joseph Agnes, Viviane M Andrade, Matthew P. Morrow, Patrick Pezzoli, Ami Patel, Albert Sylvester, and Pablo Tebas
- Subjects
DNA vaccine ,medicine.medical_specialty ,Phases of clinical research ,Phase 1 ,01 natural sciences ,Gastroenterology ,03 medical and health sciences ,0302 clinical medicine ,Antigen ,Internal medicine ,medicine ,030212 general & internal medicine ,0101 mathematics ,Neutralizing antibody ,Adverse effect ,INO-4800 ,lcsh:R5-920 ,biology ,business.industry ,SARS-CoV-2 ,Immunogenicity ,ELISPOT ,010102 general mathematics ,COVID-19 ,General Medicine ,Tolerability ,biology.protein ,Antibody ,business ,lcsh:Medicine (General) ,Research Paper - Abstract
Background A vaccine against SARS-CoV-2 is of high urgency. Here the safety and immunogenicity induced by a DNA vaccine (INO-4800) targeting the full length spike antigen of SARS-CoV-2 are described. Methods INO-4800 was evaluated in two groups of 20 participants, receiving either 1.0 mg or 2.0 mg of vaccine intradermally followed by CELLECTRA® EP at 0 and 4 weeks. Thirty-nine subjects completed both doses; one subject in the 2.0 mg group discontinued trial participation prior to receiving the second dose. ClinicalTrials.gov identifier: NCT04336410. Findings The median age was 34.5, 55% (22/40) were men and 82.5% (33/40) white. Through week 8, only 6 related Grade 1 adverse events in 5 subjects were observed. None of these increased in frequency with the second administration. No serious adverse events were reported. All 38 subjects evaluable for immunogenicity had cellular and/or humoral immune responses following the second dose of INO-4800. By week 6, 95% (36/38) of the participants seroconverted based on their responses by generating binding (ELISA) and/or neutralizing antibodies (PRNT IC50), with responder geometric mean binding antibody titers of 655.5 [95% CI (255.6, 1681.0)] and 994.2 [95% CI (395.3, 2500.3)] in the 1.0 mg and 2.0 mg groups, respectively. For neutralizing antibody, 78% (14/18) and 84% (16/19) generated a response with corresponding geometric mean titers of 102.3 [95% CI (37.4, 280.3)] and 63.5 [95% CI (39.6, 101.8)], in the respective groups. By week 8, 74% (14/19) and 100% (19/19) of subjects generated T cell responses by IFN-ɣ ELISpot assay with the median SFU per 106 PBMC of 46 [95% CI (21.1, 142.2)] and 71 [95% CI (32.2, 194.4)] in the 1.0 mg and 2.0 mg groups, respectively. Flow cytometry demonstrated a T cell response, dominated by CD8+ T cells co-producing IFN-ɣ and TNF-α, without increase in IL-4. Interpretation INO-4800 demonstrated excellent safety and tolerability and was immunogenic in 100% (38/38) of the vaccinated subjects by eliciting either or both humoral or cellular immune responses. Funding Coalition for Epidemic Preparedness Innovations (CEPI).
- Published
- 2021
16. Intradermal-delivered DNA vaccine induces durable immunity mediating a reduction in viral load in a rhesus macaque SARS-CoV-2 challenge model
- Author
-
Mohamed Abdel-Mohsen, Laurent Humeau, Stephanie Ramos, J. Joseph Kim, Arthur Doan, Edgar Tello-Ruiz, Mansi Purwar, Anthony L. Cook, Faraz I. Zaidi, Brad Finneyfrock, Diana Guimet, Yaya Dia, Kate E. Broderick, Dustin Elwood, Alan Dodson, Viviane M Andrade, Laurent Pessaint, Jewell Walters, Ami Patel, Elizabeth Parzych, Neethu Chokkalingam, Jihae Choi, Ziyang Xu, Emma L. Reuschel, Igor Maricic, Trevor R.F. Smith, Zeena Eblimit, Opeyemi S. Adeniji, Kevin Kim, Ebony N. Gary, Sophia M. Reeder, Mark G. Lewis, John Harrison, Daniel W. Kulp, Susanne Walker, Alison Generotti, David B. Weiner, Karuppiah Muthumani, Nicholas J. Tursi, Hanne Leth Andersen, Katherine Schultheis, and Pratik Bhojnagarwala
- Subjects
Male ,electroporation ,Medicine (General) ,COVID-19 Vaccines ,Injections, Intradermal ,viruses ,T-Lymphocytes ,infectious disease ,coronavirus ,Antibodies, Viral ,General Biochemistry, Genetics and Molecular Biology ,Article ,DNA vaccination ,Immune system ,R5-920 ,Immunity ,Vaccines, DNA ,Medicine ,Animals ,Neutralizing antibody ,Lung ,biology ,business.industry ,SARS-CoV-2 ,Immunogenicity ,macaque ,COVID-19 ,Viral Load ,Acquired immune system ,protection ,Antibodies, Neutralizing ,Macaca mulatta ,ID DNA vaccine ,Vaccination ,Immunology ,Spike Glycoprotein, Coronavirus ,biology.protein ,challenge ,Female ,business ,Viral load - Abstract
Coronavirus disease 2019 (COVID-19), caused by the SARS-CoV-2 virus, has had a dramatic global impact on public health and social and economic infrastructures. Here, we assess the immunogenicity and anamnestic protective efficacy in rhesus macaques of an intradermal (i.d.)-delivered SARS-CoV-2 spike DNA vaccine, INO-4800, currently being evaluated in clinical trials. Vaccination with INO-4800 induced T cell responses and induced spike antigen and RBD binding antibodies with ADCP and ADCD activity. Sera from the animals neutralized both the D614 and G614 SARS-CoV-2 pseudotype viruses. Several months after vaccination, animals were challenged with SARS-CoV-2 resulting in rapid recall of anti-SARS-CoV-2 spike protein T cell and neutralizing antibody responses. These responses were associated with lower viral loads in the lung. These studies support the immune impact of INO-4800 for inducing both humoral and cellular arms of the adaptive immune system, which are likely important for providing durable protection against COVID-19 disease., Graphical abstract, Patel et al. demonstrate that immunization with a DNA vaccine encoding the SARS-CoV-2 spike antigen, INO-4800, induces durable immune responses in rhesus macaques and is associated with reduced viral loads after challenge.
- Published
- 2020
17. SARS-CoV-2 Assays To Detect Functional Antibody Responses That Block ACE2 Recognition in Vaccinated Animals and Infected Patients
- Author
-
Kate E. Broderick, David B. Weiner, Pablo Tebas, Emma L. Reuschel, Mansi Purwar, Katherine Schultheis, Daniel W. Kulp, Karuppiah Muthumani, Neethu Chokkalingam, Ami Patel, Trevor R.F. Smith, Susanne Walker, Stephanie Ramos, Kevin Kim, Ebony N. Gary, Michaela Helble, Ziyang Xu, and Jewell Walters
- Subjects
Primates ,serological tests ,Microbiology (medical) ,COVID-19 Vaccines ,Guinea Pigs ,Pneumonia, Viral ,Enzyme-Linked Immunosorbent Assay ,Peptidyl-Dipeptidase A ,medicine.disease_cause ,Antibodies, Viral ,Immunoglobulin G ,Neutralization ,Serology ,ACE2 blocking assay ,Betacoronavirus ,Mice ,COVID-19 Testing ,Neutralization Tests ,SARS-CoV-2 vaccine ,medicine ,Animals ,Humans ,Antibodies, Blocking ,Immunoassays ,Pandemics ,Coronavirus ,biology ,business.industry ,Clinical Laboratory Techniques ,SARS-CoV-2 ,Viral Vaccine ,SARS-CoV-2 immunity ,fungi ,functional antibodies ,COVID-19 ,Viral Vaccines ,Surface Plasmon Resonance ,Vaccine efficacy ,Virology ,Antibodies, Neutralizing ,Titer ,Spike Glycoprotein, Coronavirus ,biology.protein ,Angiotensin-Converting Enzyme 2 ,Rabbits ,Antibody ,business ,Coronavirus Infections - Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has caused a global pandemic of COVID-19, resulting in cases of mild to severe respiratory distress and significant mortality. The global outbreak of this novel coronavirus has now infected >20 million people worldwide, with >5 million cases in the United States (11 August 2020). The development of diagnostic and research tools to determine infection and vaccine efficacy is critically needed., Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has caused a global pandemic of COVID-19, resulting in cases of mild to severe respiratory distress and significant mortality. The global outbreak of this novel coronavirus has now infected >20 million people worldwide, with >5 million cases in the United States (11 August 2020). The development of diagnostic and research tools to determine infection and vaccine efficacy is critically needed. We have developed multiple serologic assays using newly designed SARS-CoV-2 reagents for detecting the presence of receptor-binding antibodies in sera. The first assay is surface plasmon resonance (SPR) based and can quantitate both antibody binding to the SARS-CoV-2 spike protein and blocking to the Angiotensin-converting enzyme 2 (ACE2) receptor in a single experiment. The second assay is enzyme-linked immunosorbent assay (ELISA) based and can measure competition and blocking of the ACE2 receptor to the SARS-CoV-2 spike protein with antispike antibodies. The assay is highly versatile, and we demonstrate the broad utility of the assay by measuring antibody functionality of sera from small animals and nonhuman primates immunized with an experimental SARS-CoV-2 vaccine. In addition, we employ the assay to measure receptor blocking of sera from SARS-CoV-2-infected patients. The assay is shown to correlate with pseudovirus neutralization titers. This type of rapid, surrogate neutralization diagnostic can be employed widely to help study SARS-CoV-2 infection and assess the efficacy of vaccines.
- Published
- 2020
- Full Text
- View/download PDF
18. Experimental and in silico evidence suggests vaccines are unlikely to be affected by D614G mutation in SARS-CoV-2 spike protein
- Author
-
Mary Tachedjian, Gough G. Au, Trevor W. Drew, Sarah Edwards, Trevor R.F. Smith, Kim R. Blasdell, Sue Lowther, Rachel Layton, Matthew P. Bruce, Julian Druce, Alexander J. McAuley, Seshadri S. Vasan, Michael J. Kuiper, Timothy Poole, Glenn A. Marsh, Sarah Jane Riddell, Jennifer A. Barr, Peter A. Durr, Shawn Todd, and Kate E. Broderick
- Subjects
lcsh:Immunologic diseases. Allergy ,0301 basic medicine ,Proteases ,In silico ,Immunology ,Biology ,Brief Communication ,lcsh:RC254-282 ,Virus ,Neutralization ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,RNA polymerase ,Pharmacology (medical) ,030212 general & internal medicine ,Viral evolution ,chemistry.chemical_classification ,Pharmacology ,Vaccines ,SARS-CoV-2 ,Elastase ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Virology ,030104 developmental biology ,Infectious Diseases ,chemistry ,lcsh:RC581-607 ,Glycoprotein - Abstract
The ‘D614G’ mutation (Aspartate-to-Glycine change at position 614) of the SARS-CoV-2 spike protein has been speculated to adversely affect the efficacy of most vaccines and countermeasures that target this glycoprotein, necessitating frequent vaccine matching. Virus neutralisation assays were performed using sera from ferrets which received two doses of the INO-4800 COVID-19 vaccine, and Australian virus isolates (VIC01, SA01 and VIC31) which either possess or lack this mutation but are otherwise comparable. Through this approach, supported by biomolecular modelling of this mutation and the commonly-associated P314L mutation in the RNA-dependent RNA polymerase, we have shown that there is no experimental evidence to support this speculation. We additionally demonstrate that the putative elastase cleavage site introduced by the D614G mutation is unlikely to be accessible to proteases.
- Published
- 2020
19. Intradermal-delivered DNA vaccine provides anamnestic protection in a rhesus macaque SARS-CoV-2 challenge model
- Author
-
Arthur Doan, Ebony N. Gary, Yaya Dia, Sophia M. Reeder, Emma L. Reuschel, Anthony L. Cook, Jihae Choi, Mark G. Lewis, Diana Guimet, Laurent Pessaint, Faraz I. Zaidi, Neethu Chokkalingam, Igor Maricic, Alison Generotti, David B. Weiner, Zeena Eblimit, Dustin Elwood, Ziyang Xu, John Harrison, Alan Dodson, Laurent Humeau, Stephanie Ramos, Hanne Anderson, Pratik Bhojnagarwala, Trevor R.F. Smith, Karuppiah Muthumani, Kevin Kim, Viviane M Andrade, Elizabeth Parzych, Nicholas J. Tursi, Ami Patel, Edgar Tello-Ruiz, Mansi Purwar, Brad Finneyfrock, Katherine Schultheis, Kate E. Broderick, Susanne Walker, J. Joseph Kim, Daniel W. Kulp, and Jewell Walters
- Subjects
biology ,business.industry ,viruses ,Immunogenicity ,Acquired immune system ,Virus ,DNA vaccination ,Vaccination ,Immune system ,Immunology ,biology.protein ,Medicine ,Neutralizing antibody ,business ,Viral load - Abstract
SummaryCoronavirus disease 2019 (COVID-19), caused by the SARS-CoV-2 virus, has had a dramatic global impact on public health, social, and economic infrastructures. Here, we assess immunogenicity and anamnestic protective efficacy in rhesus macaques of the intradermal (ID)-delivered SARS-CoV-2 spike DNA vaccine, INO-4800. INO-4800 is an ID-delivered DNA vaccine currently being evaluated in clinical trials. Vaccination with INO-4800 induced T cell responses and neutralizing antibody responses against both the D614 and G614 SARS-CoV-2 spike proteins. Several months after vaccination, animals were challenged with SARS-CoV-2 resulting in rapid recall of anti-SARS-CoV-2 spike protein T and B cell responses. These responses were associated with lower viral loads in the lung and with faster nasal clearance of virus. These studies support the immune impact of INO-4800 for inducing both humoral and cellular arms of the adaptive immune system which are likely important for providing durable protection against COVID-19 disease.
- Published
- 2020
- Full Text
- View/download PDF
20. SARS-CoV-2 assays to detect functional antibody responses that block ACE2 recognition in vaccinated animals and infected patients
- Author
-
Emma L. Reuschel, David B. Weiner, Daniel W. Kulp, Jewell Walters, Mansi Purwar, Katherine Schultheis, Ebony N. Gary, Neethu Chokkalingam, Pablo Tebas, Kate E. Broderick, Stephanie Ramos, Trevor R.F. Smith, Susanne Walker, Ami Patel, Ziyang Xu, and Kevin Kim
- Subjects
biology ,business.industry ,Vaccine efficacy ,medicine.disease_cause ,Virology ,Neutralization ,Virus ,Serology ,Immune system ,Antibody receptor ,medicine ,biology.protein ,Antibody ,business ,Coronavirus - Abstract
SARS-CoV-2 (Severe Acute Respiratory Syndrome Coronavirus 2) has caused a global pandemic of COVID-19 resulting in cases of mild to severe respiratory distress and significant mortality. The global outbreak of this novel coronavirus has now infected >8 million people worldwide with >2 million cases in the US (June 17th, 2020). There is an urgent need for vaccines and therapeutics to combat the spread of this coronavirus. Similarly, the development of diagnostic and research tools to determine infection and vaccine efficacy are critically needed. Molecular assays have been developed to determine viral genetic material present in patients. Serological assays have been developed to determine humoral responses to the spike protein or receptor binding domain (RBD). Detection of functional antibodies can be accomplished through neutralization of live SARS-CoV2 virus, but requires significant expertise, an infectible stable cell line, a specialized BioSafety Level 3 (BSL-3) facility. As large numbers of people return from quarantine, it is critical to have rapid diagnostics that can be widely adopted and employed to assess functional antibody levels in the returning workforce. This type of surrogate neutralization diagnostic can also be used to assess humoral immune responses induced in patients from the large number of vaccine and immunotherapy trials currently on-going. Here we describe a rapid serological diagnostic assay for determining antibody receptor blocking and demonstrate the broad utility of the assay by measuring the antibody functionality of sera from small animals and non-human primates immunized with an experimental SARS-CoV-2 vaccine and using sera from infected patients.
- Published
- 2020
- Full Text
- View/download PDF
21. Immunogenicity of a DNA vaccine candidate for COVID-19
- Author
-
Elizabeth Parzych, Alison Generotti, Maria Yang, Chuan Qin, Karen R. Buttigieg, Pratik Bhojnagarwala, Dustin Elwood, Igor Maricic, J. Joseph Kim, Patrick Pezzoli, Arthur Doan, Miguel Vasquez, Edgar Tello-Ruiz, Faraz I. Zaidi, Miles W. Carroll, Yaya Dia, Yuanhan Wu, Daniel Wrapp, Jason S. McLellan, Xizhou Zhu, David B. Weiner, Nicholas J. Tursi, Ami Shah Brown, Emma L. Reuschel, Jacqueline Chu, Ami Patel, Ziyang Xu, Nianshuang Wang, Wei Deng, Daniel W. Kulp, Gan Zhao, Timothy A. Herring, Katherine Schultheis, Susanne Walker, Neethu Chokkalingam, Karuppiah Muthumani, Mamadou A. Bah, Jiun Ming Wu, Laurent Humeau, Linlin Bao, Dinah Amante, Stephanie Ramos, Jihae Choi, Ebony N. Gary, Sophia M. Reeder, Jean D. Boyer, Viviane M Andrade, Bin Wang, Daniel H. Park, Mansi Purwar, Makan Khoshnejad, Jiangning Liu, Kevin Kim, Dan Li, Ali Raza Ali, Kate E. Broderick, Trevor R.F. Smith, Jian Yan, and Jewell Walters
- Subjects
0301 basic medicine ,viruses ,General Physics and Astronomy ,medicine.disease_cause ,Mice ,0302 clinical medicine ,Vaccines, DNA ,lcsh:Science ,skin and connective tissue diseases ,Antigens, Viral ,Lung ,Coronavirus ,Mice, Inbred BALB C ,Multidisciplinary ,biology ,Immunogenicity ,virus diseases ,Cellular immunity ,030220 oncology & carcinogenesis ,Models, Animal ,Spike Glycoprotein, Coronavirus ,Middle East Respiratory Syndrome Coronavirus ,Angiotensin-Converting Enzyme 2 ,Antibody ,Coronavirus Infections ,COVID-19 Vaccines ,Middle East respiratory syndrome coronavirus ,Science ,Guinea Pigs ,Peptidyl-Dipeptidase A ,Article ,Antibodies ,General Biochemistry, Genetics and Molecular Biology ,DNA vaccines ,03 medical and health sciences ,Antigen ,Immunity ,medicine ,Animals ,SARS-CoV-2 ,fungi ,Viral Vaccines ,General Chemistry ,Antibodies, Neutralizing ,Virology ,Immunity, Humoral ,body regions ,030104 developmental biology ,Epitope mapping ,Immunization ,Immunoglobulin G ,biology.protein ,lcsh:Q ,Epitope Mapping - Abstract
The coronavirus family member, SARS-CoV-2 has been identified as the causal agent for the pandemic viral pneumonia disease, COVID-19. At this time, no vaccine is available to control further dissemination of the disease. We have previously engineered a synthetic DNA vaccine targeting the MERS coronavirus Spike (S) protein, the major surface antigen of coronaviruses, which is currently in clinical study. Here we build on this prior experience to generate a synthetic DNA-based vaccine candidate targeting SARS-CoV-2 S protein. The engineered construct, INO-4800, results in robust expression of the S protein in vitro. Following immunization of mice and guinea pigs with INO-4800 we measure antigen-specific T cell responses, functional antibodies which neutralize the SARS-CoV-2 infection and block Spike protein binding to the ACE2 receptor, and biodistribution of SARS-CoV-2 targeting antibodies to the lungs. This preliminary dataset identifies INO-4800 as a potential COVID-19 vaccine candidate, supporting further translational study., There is currently no licensed SARS-CoV-2 vaccine. Here, the authors generate an optimized DNA vaccine candidate encoding the SARS-CoV-2 spike antigen, demonstrating induction of specific T cells and neutralizing antibody responses in mice and guinea pigs. These initial results support further development of this vaccine candidate.
- Published
- 2020
- Full Text
- View/download PDF
22. Establishment of a pig influenza challenge model for evaluation of monoclonal antibody delivery platforms
- Author
-
Jeffery K. Taubenberger, Sarah T. C. Elliott, Matthieu Bernard, Barbara Holzer, Adam McNee, Trevor R.F. Smith, Katherine Schultheis, Ghiabe Guibinga, Elma Tchilian, Stephanie Ramos, Ami Patel, Becky Clark, Kate E. Broderick, Alejandro Núñez, John C. Kash, Peter C. L. Beverley, Tiphany Chrun, Yongli Xiao, Pramila Rijal, Alain Townsend, Paul Fisher, Heather Brown, David B. Weiner, Emily Bessell, and Veronica Martini
- Subjects
biology ,medicine.drug_class ,business.industry ,Hemagglutinin (influenza) ,Monoclonal antibody ,Virology ,Virus ,Vaccination ,Plasmid ,Pandemic ,medicine ,biology.protein ,Viral shedding ,business ,Viral load - Abstract
Monoclonal antibodies are a possible adjunct to vaccination and drugs in treatment of influenza virus infection. However questions remain whether small animal models accurately predict efficacy in humans. We have established the pig, a large natural host animal for influenza, with many physiological similarities to humans, as a robust model for testing monoclonal antibodies. We show that a strongly neutralizing monoclonal antibody (2-12C) against the hemagglutinin head administered prophylactically at 15 mg/kg reduced viral load and lung pathology after pandemic H1N1 influenza challenge. A lower dose of 1 mg/kg of 2-12C or a DNA plasmid encoded version of 2-12C, reduced pathology and viral load in the lungs, but not viral shedding in nasal swabs. We propose that the pig influenza model will be useful for testing candidate monoclonal antibodies and emerging delivery platforms prior to human trials.
- Published
- 2020
- Full Text
- View/download PDF
23. Rapid development of a synthetic DNA vaccine for COVID-19
- Author
-
Arthur Doan, David B. Weiner, Kevin Kim, Elizabeth Parzych, Jean D. Boyer, Yuanhan Wu, Katherine Schultheis, Karuppiah Muthumani, Igor Maricic, Mamadou A. Bah, Miguel Vasquez, Ebony N. Gary, Sophia M. Reeder, Ali Raza Ali, Faraz I. Zaidi, Jewell Walters, Kate E. Broderick, Laurent Humeau, Daniel Park, Xizhou Zhu, Alison Generotti, Stephanie Ramos, Dustin Elwood, Neethu Chokkalingam, Yaya Dia, Makan Khoshnejad, Daniel Wrapp, Susanne Walker, Nicholas J. Tursi, Emma L. Reuschel, Jian Yan, Dinah Amante, Jacqueline Chu, Ami Shah Brown, Nianshuang Wang, Maria Yang, Trevor R.F. Smith, Jason S. McLellan, Ami Patel, Jihae Choi, Timothy A. Herring, J. Joseph Kim, Daniel W. Kulp, and Patrick Pezzoli
- Subjects
Coronavirus disease 2019 (COVID-19) ,Synthetic DNA ,Computational biology ,Biology - Abstract
The coronavirus family member, SARS-CoV-2 has been identified as the causal agent for the outbreak of viral pneumonia disease, COVID-19 which first emerged in mid-December 2019 in the city of Wuhan in central China. As of February 25, 2020 there are 80,994 people infected and 2,760 deaths, and documented human-to-human transmission across multiple continents. At this time, no vaccine is available to control further dissemination of the disease. We have previously developed a synthetic DNA vaccine targeting the MERS coronavirus Spike (S) protein that was deployed in response to the MERS outbreak in South Korea. This vaccine induced potent antibody and CTL responses, and provided protection in a NHP challenge model. In the clinic, the vaccine generated humoral immunity including neutralizing antibody responses, as well as T cell immunity. Here we build on this prior work and report on the rapid development of a synthetic DNA-based vaccine targeting the major surface antigen Spike protein of SARS-CoV-2. The engineered construct, INO-4800 induced robust expression of the Spike protein in vitro, and generated antibody and T cell responses following a single immunization in mice and guinea pigs. This preliminary dataset identifies INO-4800 as a potential COVID-19 vaccine candidate, supporting further study for mobilization against this emerging disease threat.
- Published
- 2020
- Full Text
- View/download PDF
24. Adjuvant Screen Identifies Synthetic DNA-Encoding Flt3L and CD80 Immunotherapeutics as Candidates for Enhancing Anti-tumor T Cell Responses
- Author
-
Jian Yan, Tricia T Nguyen, Charles C. Reed, Trevor R.F. Smith, Amy Haseley Thorne, Kirsten N Malo, Emma L Masteller, Neil Cooch, Ashley J Wong, Laurent Humeau, and Kate E. Broderick
- Subjects
0301 basic medicine ,lcsh:Immunologic diseases. Allergy ,T-Lymphocytes ,T cell ,medicine.medical_treatment ,Immunology ,Biology ,Lymphocyte Activation ,Cancer Vaccines ,Tetraspanin 28 ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Adjuvants, Immunologic ,CD80 ,Antigens, Neoplasm ,Cell Movement ,Neoplasms ,vaccine ,Vaccines, DNA ,medicine ,Animals ,Humans ,Immunology and Allergy ,cancer ,Original Research ,Mice, Inbred BALB C ,Flt3L ,Membrane Proteins ,DNA ,Dendritic Cells ,Immunotherapy ,Dendritic cell ,030104 developmental biology ,medicine.anatomical_structure ,B7-1 Antigen ,Cancer research ,Cytokines ,Female ,Cancer vaccine ,immune ,lcsh:RC581-607 ,Adjuvant ,CD8 ,Plasmids ,030215 immunology - Abstract
Overcoming tolerance to tumor-associated antigens remains a hurdle for cancer vaccine-based immunotherapy. A strategy to enhance the anti-tumor immune response is the inclusion of adjuvants to cancer vaccine protocols. In this report, we generated and systematically screened over twenty gene-based molecular adjuvants composed of cytokines, chemokines, and T cell co-stimulators for the ability to increase anti-tumor antigen T cell immunity. We identified several robust adjuvants whose addition to vaccine formulations resulted in enhanced T cell responses targeting the cancer antigens STEAP1 and TERT. We further characterized direct T cell stimulation through CD80-Fc and indirect T cell targeting via the dendritic cell activator Flt3L-Fc. Mechanistically, intramuscular delivery of Flt3L-Fc into mice was associated with a significant increase in infiltration of dendritic cells at the site of administration and trafficking of activated dendritic cells to the draining lymph node. Gene expression analysis of the muscle tissue confirmed a significant up-regulation in genes associated with dendritic cell signaling. Addition of CD80-Fc to STEAP1 vaccine formulation mimicked the engagement provided by DCs and increased T cell responses to STEAP1 by 8-fold, significantly increasing the frequency of antigen-specific cells expressing IFNγ, TNFα, and CD107a for both CD8+ and CD4+ T cells. CD80-Fc enhanced T cell responses to multiple tumor-associated antigens including Survivin and HPV, indicating its potential as a universal adjuvant for cancer vaccines. Together, the results of our study highlight the adjuvanting effect of T cell engagement either directly, CD80-Fc, or indirectly, Flt3L-Fc, for cancer vaccines.
- Published
- 2020
- Full Text
- View/download PDF
25. Synthetic DNA-Encoded Monoclonal Antibody Delivery of Anti–CTLA-4 Antibodies Induces Tumor ShrinkageIn Vivo
- Author
-
Laurent Humeau, Alfredo Perales-Puchalt, Karuppiah Muthumani, Aspen Trautz, Trevor R.F. Smith, Elizabeth K. Duperret, Regina Stoltz, Megan C. Wise, Ami Patel, Emma L Masteller, J. Joseph Kim, David B. Weiner, and Kate E. Broderick
- Subjects
0301 basic medicine ,Cancer Research ,medicine.drug_class ,T-Lymphocytes ,medicine.medical_treatment ,Antibodies, Monoclonal, Humanized ,Monoclonal antibody ,Article ,Inhibitory Concentration 50 ,Mice ,03 medical and health sciences ,In vivo ,Cell Line, Tumor ,Neoplasms ,Animals ,Humans ,Medicine ,CTLA-4 Antigen ,Mice, Inbred BALB C ,biology ,business.industry ,Antibodies, Monoclonal ,DNA ,Immunotherapy ,Ipilimumab ,Immune checkpoint ,Mice, Inbred C57BL ,HEK293 Cells ,030104 developmental biology ,Oncology ,Immunoglobulin G ,Monoclonal ,Leukocytes, Mononuclear ,Cancer research ,biology.protein ,Antibody ,business ,Tremelimumab ,Neoplasm Transplantation ,Ex vivo ,Plasmids ,medicine.drug - Abstract
Antibody-based immune therapies targeting the T-cell checkpoint molecules CTLA-4 and PD-1 have affected cancer therapy. However, this immune therapy requires complex manufacturing and frequent dosing, limiting the global use of this treatment. Here, we focused on the development of a DNA-encoded monoclonal antibody (DMAb) approach for delivery of anti–CTLA-4 monoclonal antibodies in vivo. With this technology, engineered and formulated DMAb plasmids encoding IgG inserts were directly injected into muscle and delivered intracellularly by electroporation, leading to in vivo expression and secretion of the encoded IgG. DMAb expression from a single dose can continue for several months without the need for repeated administration. Delivery of an optimized DMAb encoding anti-mouse CTLA-4 IgG resulted in high serum levels of the antibody as well as tumor regression in Sa1N and CT26 tumor models. DNA-delivery of the anti-human CTLA-4 antibodies ipilimumab and tremelimumab in mice achieved potent peak levels of approximately 85 and 58 μg/mL, respectively. These DMAb exhibited prolonged expression, with maintenance of serum levels at or above 15 μg/mL for over a year. Anti-human CTLA-4 DMAbs produced in vivo bound to human CTLA-4 protein expressed on stimulated human peripheral blood mononuclear cells and induced T-cell activation in a functional assay ex vivo. In summary, direct in vivo expression of DMAb encoding checkpoint inhibitors serves as a novel tool for immunotherapy that could significantly improve availability and provide broader access to such therapies.Significance: DNA-encoded monoclonal antibodies represent a novel technology for delivery and expression of immune checkpoint blockade antibodies, thus expanding patient access to, and possible clinical applications of, these therapies. Cancer Res; 78(22); 6363–70. ©2018 AACR.
- Published
- 2018
- Full Text
- View/download PDF
26. In Vivo Delivery of Synthetic Human DNA-Encoded Monoclonal Antibodies Protect against Ebolavirus Infection in a Mouse Model
- Author
-
Laurent Humeau, Shihua He, Jian Yan, Xiaoying Yu, Anders Leung, Marguerite E. Gorman, Kevin Tierney, Sarah T. C. Elliott, Trevor R.F. Smith, Kate E. Broderick, Trina Racine, Daniel H. Park, Karuppiah Muthumani, Megan C. Wise, Erica Ollmann Saphire, Aubrey L. Bryan, Xiangguo Qiu, James E. Crowe, David B. Weiner, Benjamin J. Doranz, Gary P. Kobinger, Carl W. Davis, Rianne Esquivel, Jing Chen, Darwyn Kobasa, Edgar Davidson, Ami Patel, Charles C. Reed, Niranjan Y. Sardesai, and Rafi Ahmed
- Subjects
0301 basic medicine ,Zaire ebolavirus ,medicine.drug_class ,Biology ,medicine.disease_cause ,Monoclonal antibody ,General Biochemistry, Genetics and Molecular Biology ,Article ,law.invention ,03 medical and health sciences ,Epitopes ,0302 clinical medicine ,law ,In vivo ,medicine ,Animals ,Humans ,lcsh:QH301-705.5 ,Glycoproteins ,Ebolavirus ,chemistry.chemical_classification ,Mice, Inbred BALB C ,Ebola virus ,Muscles ,Antibodies, Monoclonal ,DNA ,Hemorrhagic Fever, Ebola ,Virology ,Recombinant Proteins ,Disease Models, Animal ,030104 developmental biology ,HEK293 Cells ,chemistry ,lcsh:Biology (General) ,Infectious disease (medical specialty) ,Mutagenesis ,030220 oncology & carcinogenesis ,Recombinant DNA ,Female ,Glycoprotein ,Epitope Mapping - Abstract
SUMMARY Synthetically engineered DNA-encoded monoclonal antibodies (DMAbs) are an in vivo platform for evaluation and delivery of human mAb to control against infectious disease. Here, we engineer DMAbs encoding potent anti-Zaire ebolavirus (EBOV) glycoprotein (GP) mAbs isolated from Ebola virus disease survivors. We demonstrate the development of a human IgG1 DMAb platform for in vivo EBOV-GP mAb delivery and evaluation in a mouse model. Using this approach, we show that DMAb-11 and DMAb-34 exhibit functional and molecular profiles comparable to recombinant mAb, have a wide window of expression, and provide rapid protection against lethal mouse-adapted EBOV challenge. The DMAb platform represents a simple, rapid, and reproducible approach for evaluating the activity of mAb during clinical development. DMAbs have the potential to be a mAb delivery system, which may be advantageous for protection against highly pathogenic infectious diseases, like EBOV, in resource-limited and other challenging settings., Graphical Abstract, In Brief Monoclonal antibodies are an important approach for emerging infectious disease prevention. Patel et al. demonstrate engineering and in vivo delivery of DNA-encoded monoclonal antibodies (DMAbs) targeting the Zaire ebolavirus (EBOV) glycoprotein. DMAbs protect against lethal mouse-adapted EBOV and are useful for rapid evaluation of fully human mAbs in live animal models.
- Published
- 2018
27. A Synthetic Micro-Consensus DNA Vaccine Generates Comprehensive Influenza A H3N2 Immunity and Protects Mice Against Lethal Challenge by Multiple H3N2 Viruses
- Author
-
Charles C. Reed, Bradley Garman, Ami Patel, Jian Yan, Jacqueline Chu, David B. Weiner, Sarah T. C. Elliott, Amelia A. Keaton, and Kate E. Broderick
- Subjects
0301 basic medicine ,Immunogen ,viruses ,Hemagglutinin (influenza) ,Antibodies, Viral ,Epitope ,DNA vaccination ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Antigen ,Immunity ,Influenza, Human ,Vaccines, DNA ,Genetics ,Animals ,Humans ,030212 general & internal medicine ,Molecular Biology ,Research Articles ,Mice, Inbred BALB C ,biology ,Influenza A Virus, H3N2 Subtype ,Vaccination ,virus diseases ,DNA ,Virology ,030104 developmental biology ,Influenza Vaccines ,biology.protein ,Molecular Medicine - Abstract
Influenza A H3N2 viruses circulate globally, leading to substantial morbidity and mortality. Commercially available, antigen-matched influenza vaccines must be updated frequently to match dynamic sequence variability in immune epitopes, especially within viral influenza A H3N2 hemagglutinin (H3). In an effort to create comprehensive immune responses against H3N2, four micro-consensus antigens were designed to mimic the sequence and antigenic diversity of H3. Synthetic plasmid DNA constructs were developed to express each micro-consensus immunogen and combined into a multi-antigen DNA vaccine cocktail, pH3HA. Facilitated delivery of pH3HA via intramuscular electroporation in mice induced comprehensive, potent humoral responses against diverse seasonal H3N2 viruses that circulated between 1968 and the present. Vaccination with pH3HA also induced an antigen-specific cellular cytokine response. Mice immunized with pH3HA were protected against lethal challenge using two distinct H3N2 viruses, highlighting the heterologous protection afforded by synthetic micro-consensus immunogens. These findings warrant further study of the DNA vaccine micro-consensus platform for broad protection against influenza viruses.
- Published
- 2018
- Full Text
- View/download PDF
28. DNA Vaccine–Induced Long-Lasting Cytotoxic T Cells Targeting Conserved Elements of Human Immunodeficiency Virus Gag Are Boosted Upon DNA or Recombinant Modified Vaccinia Ankara Vaccination
- Author
-
Eric G. Ramírez-Salazar, Barbara K. Felber, Bernard Moss, Linda S. Wyatt, Viraj Kulkarni, James I. Mullins, Niranjan Y. Sardesai, Frances Dayton, George N. Pavlakis, Margherita Rosati, Antonio Valentin, Yanhui Cai, Patricia L. Earl, Xintao Hu, and Kate E. Broderick
- Subjects
CD4-Positive T-Lymphocytes ,Cytotoxicity, Immunologic ,0301 basic medicine ,Modified vaccinia Ankara ,Immunogen ,viruses ,Immunization, Secondary ,HIV Infections ,Vaccinia virus ,Biology ,medicine.disease_cause ,Epitope ,DNA vaccination ,law.invention ,03 medical and health sciences ,law ,Vaccines, DNA ,Vaccinia ,Genetics ,medicine ,Animals ,Humans ,Cytotoxic T cell ,Molecular Biology ,AIDS Vaccines ,DNA ,Simian immunodeficiency virus ,Macaca mulatta ,Virology ,Vaccination ,030104 developmental biology ,HIV-1 ,Recombinant DNA ,Molecular Medicine ,Simian Immunodeficiency Virus ,T-Lymphocytes, Cytotoxic - Abstract
DNA-based vaccines able to induce efficient cytotoxic T-cell responses targeting conserved elements (CE) of human immunodeficiency virus type 1 (HIV-1) Gag have been developed. These CE were selected by stringent conservation, the ability to induce T-cell responses with broad human leukocyte antigen coverage, and the association between recognition of CE epitopes and viral control in HIV-infected individuals. Based on homology to HIV, a simian immunodeficiency virus p27gag CE DNA vaccine has also been developed. This study reports on the durability of the CE-specific T-cell responses induced by HIV and simian immunodeficiency virus CE DNA-based prime/boost vaccine regimens in rhesus macaques, and shows that the initially primed CE-specific T-cell responses were efficiently boosted by a single CE DNA vaccination after the long rest period (up to 2 years). In another cohort of animals, the study shows that a single inoculation with non-replicating recombinant Modified Vaccinia Ankara (rMVA62B) also potently boosted CE-specific responses after around 1.5 years of rest. Both CE DNA and rMVA62B booster vaccinations increased the magnitude and cytotoxicity of the CE-specific responses while maintaining the breadth of CE recognition. Env produced by rMVA62B did not negatively interfere with the recall of the Gag CE responses. rMVA62B could be beneficial to further boosting the immune response to Gag in humans. Vaccine regimens that employ CE DNA as a priming immunogen hold promise for application in HIV prevention and therapy.
- Published
- 2018
- Full Text
- View/download PDF
29. Delineating the Cellular Mechanisms Associated with Skin Electroporation
- Author
-
William B. Kiosses, Janet Oh, Katherine Schultheis, Trevor R.F. Smith, Kate E. Broderick, Amelia Wong, and Kimberly A. Kraynyak
- Subjects
0301 basic medicine ,Skin electroporation ,Necrosis ,Treatment protocol ,Guinea Pigs ,Apoptosis ,Biology ,Applied Microbiology and Biotechnology ,DNA vaccination ,03 medical and health sciences ,Immune system ,medicine ,Vaccines, DNA ,Genetics ,Animals ,Genetics(clinical) ,Genetics (clinical) ,Research Articles ,Skin ,Pharmacology ,integumentary system ,Electroporation ,030104 developmental biology ,Cancer research ,Molecular Medicine ,Female ,medicine.symptom - Abstract
The immune responses elicited following delivery of DNA vaccines to the skin has previously been shown to be significantly enhanced by the addition of electroporation (EP) to the treatment protocol. Principally, EP increases the transfection of plasmid DNA (pDNA) into the resident skin cells. In addition to increasing the levels of in vivo transfection, the physical insult induced by EP is associated with activation of innate pathways which are believed to mediate an adjuvant effect, further enhancing DNA vaccine responses. We investigated the possible mechanisms associated with this adjuvant effect, primarily focusing on the cell death pathways associated with the skin EP procedure independent of pDNA delivery. Using the minimally invasive CELLECTRA(®)-3P intradermal electroporation device that penetrates the epidermal and dermal layers of the skin, we have investigated apoptotic and necrotic cell death in relation to the vicinity of the electrode needles and electric field generated. Employing the well-established terminal deoxynucleotidyl transferase nick-end labeling assay, we detected apoptosis beginning as early as one hour after EP and peaking at the 4 h time point. The majority of the apoptotic events were detected in the epidermal region directly adjacent to the electrode needle. Using a novel propidium iodide in vivo necrotic cell death assay, we detected necrotic events concentrated in the epidermal region adjacent to the electrode. Furthermore, we detected upregulation of calreticulin expression on skin cells after EP, thus labeling these cells for uptake by dendritic cells and macrophages. These results allow us to delineate the cell death mechanisms occurring in the skin following intradermal EP independently of pDNA delivery. We believe these events contribute to the adjuvant effect observed following electroporation at the skin treatment site.
- Published
- 2018
- Full Text
- View/download PDF
30. Gag and env conserved element CE DNA vaccines elicit broad cytotoxic T cell responses targeting subdominant epitopes of HIV and SIV Able to recognize virus-infected cells in macaques
- Author
-
Niranjan Y. Sardesai, Barbara K. Felber, Antonio Valentin, Preston A. Marx, James I. Mullins, Xintao Hu, Margherita Rosati, Zhongyan Lu, Kate E. Broderick, and George N. Pavlakis
- Subjects
Male ,0301 basic medicine ,Subdominant ,viruses ,Immunology ,Simian Acquired Immunodeficiency Syndrome ,Epitopes, T-Lymphocyte ,Gene Products, gag ,Biology ,immunization ,Conserved sequences ,Virus ,Epitope ,Conserved sequence ,DNA vaccination ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,vaccine ,Vaccines, DNA ,Animals ,Immunology and Allergy ,Cytotoxic T cell ,Viremia ,Conserved Sequence ,Immunization Schedule ,AIDS Vaccines ,Pharmacology ,SAIDS Vaccines ,virus diseases ,Gene Products, env ,HIV ,DNA ,Virology ,infection ,3. Good health ,030104 developmental biology ,SIV ,Immunization ,chemistry ,030220 oncology & carcinogenesis ,Macaca ,human activities ,T-Lymphocytes, Cytotoxic ,Research Paper - Abstract
HIV sequence diversity and the propensity of eliciting immunodominant responses targeting inessential variable regions are hurdles in the development of an effective AIDS vaccine. We developed a DNA vaccine comprising conserved elements (CE) of SIV p27Gag and HIV-1 Env and found that priming vaccination with CE DNA is critical to efficiently overcome the dominance imposed by Gag and Env variable regions. Here, we show that DNA vaccinated macaques receiving the CE prime/CE+full-length DNA co-delivery booster vaccine regimens developed broad, potent and durable cytotoxic T cell responses targeting conserved protein segments of SIV Gag and HIV Env. Gag CE-specific T cells showed robust anamnestic responses upon infection with SIVmac239 which led to the identification of CE-specific cytotoxic lymphocytes able to recognize epitopes covering distinct CE on the surface of SIV infected cells in vivo. Though not controlling infection overall, we found an inverse correlation between Gag CE-specific CD8+ T cell responses and peak viremia. The T cell responses induced by the HIV Env CE immunogen were recalled in some animals upon SIV infection, leading to the identification of two cross-reactive epitopes between HIV and SIV Env based in sequence homology. These data demonstrate that a vaccine combining Gag and Env CE DNA subverted the normal immunodominance patterns, eliciting immune responses that included subdominant, highly conserved epitopes. These vaccine regimens augment cytotoxic T cell responses to highly conserved epitopes in the viral proteome and maximize response breadth. The vaccine-induced CE-specific T cells were expanded upon SIV infection, indicating that the predicted CE epitopes incorporated in the DNA vaccine are processed and exposed by infected cells in their natural context within the viral proteome.
- Published
- 2018
- Full Text
- View/download PDF
31. Broad cross-protective anti-hemagglutination responses elicited by influenza microconsensus DNA vaccine
- Author
-
Niranjan Y. Sardesai, Trina Racine, Jaemi S. Chu, Kate E. Broderick, David B. Weiner, Matthew P. Morrow, Gary P. Kobinger, Charles C. Reed, Amir S. Khan, J. Joseph Kim, and Jian Yan
- Subjects
0301 basic medicine ,Serotype ,Cross Protection ,Guinea Pigs ,Hemagglutinin Glycoproteins, Influenza Virus ,Biology ,Antibodies, Viral ,medicine.disease_cause ,Virus ,DNA vaccination ,Mice ,03 medical and health sciences ,Influenza A Virus, H1N1 Subtype ,0302 clinical medicine ,Orthomyxoviridae Infections ,Antigen ,Immunity ,Consensus Sequence ,Vaccines, DNA ,Influenza A virus ,medicine ,Animals ,030212 general & internal medicine ,Mice, Inbred BALB C ,General Veterinary ,General Immunology and Microbiology ,Ferrets ,Public Health, Environmental and Occupational Health ,Hemagglutination Inhibition Tests ,Antibodies, Neutralizing ,Macaca mulatta ,Virology ,Human morbidity ,Electroporation ,030104 developmental biology ,Infectious Diseases ,Influenza Vaccines ,Immunology ,biology.protein ,Molecular Medicine ,Antibody - Abstract
Despite the routine development and distribution of seasonal influenza vaccines, influenza remains an important pathogen contributing to significant human morbidity as well as mortality each year. The seasonal variability of influenza creates a significant issue for vaccine development of seasonal strains that can afford protection from infection or disease based on serotype matching. It is appreciated that the globular head of the HA antigen contained in the vaccines generates antibodies that result in HAI activity that are a major correlates of the protection against a particular strain. Due to seasonal genetic changes in the HA protein, however, new vaccine strains are needed to be developed continually to match the new HA antigen of that seasons virus. A distinct advantage in seasonal vaccine development would be if a small group of antigens could be developed that could span many seasons without needed to be replaced due to this genetic drift. Here we report on a synthetic microconsensus approach that relies on a small collection of 4 synthetic H1HA DNA antigens which together induce broad protective HAI immunity spanning decades of H1 influenza viruses in mice, guinea pigs and non-human primates. The protective HAI titers induced by microconsensus immunogens are fully functional in vivo as immunized ferrets were completely protected from A/Mexico/InDRE4487/2009 virus infection and morbidity associated with lethal challenge. These results are encouraging that a limited easy-to-formulate collection of invariant antigens can be developed which can span seasonal vaccine changes allowing for continued immune protection.
- Published
- 2018
- Full Text
- View/download PDF
32. A DNA vaccine delivered by dermal electroporation fully protects cynomolgus macaques against Lassa fever
- Author
-
Suzanne E. Wollen, Paul R Facemire, Kathleen A. Cashman, Niranjan Y. Sardesai, Carl I Shaia, Kate E. Broderick, Connie S. Schmaljohn, Todd M. Bell, Joshua D. Shamblin, Jeremy J. Bearss, and Eric R. Wilkinson
- Subjects
0301 basic medicine ,DNA vaccine ,electroporation ,Male ,viruses ,Arenaviridae ,Immunology ,medicine.disease_cause ,Administration, Cutaneous ,DNA vaccination ,03 medical and health sciences ,Lassa Fever ,medicine ,Vaccines, DNA ,Immunology and Allergy ,Animals ,Viremia ,Lassa fever ,Immunization Schedule ,Pharmacology ,biology ,Electroporation ,virus diseases ,RNA virus ,Viral Vaccines ,medicine.disease ,biology.organism_classification ,Virology ,Research Papers ,Survival Analysis ,Disease Models, Animal ,Macaca fascicularis ,030104 developmental biology ,Lassa virus ,Treatment Outcome - Abstract
Lassa virus (LASV) is an ambisense RNA virus in the Arenaviridae family and is the etiological agent of Lassa fever, a severe hemorrhagic disease endemic to West and Central Africa.1,2 There are no US Food and Drug Administration (FDA)-licensed vaccines available to prevent Lassa fever.1,2 in our previous studies, we developed a gene-optimized DNA vaccine that encodes the glycoprotein precursor gene of LASV (Josiah strain) and demonstrated that 3 vaccinations accompanied by dermal electroporation protected guinea pigs from LASV-associated illness and death. Here, we describe an initial efficacy experiment in cynomolgus macaque nonhuman primates (NHPs) in which we followed an identical 3-dose vaccine schedule that was successful in guinea pigs, and a follow-on experiment in which we used an accelerated vaccination strategy consisting of 2 administrations, spaced 4 weeks apart. In both studies, all of the LASV DNA-vaccinated NHPs survived challenge and none of them had measureable, sustained viremia or displayed weight loss or other disease signs post-exposure. Three of 10 mock-vaccinates survived exposure to LASV, but all of them became acutely ill post-exposure and remained chronically ill to the study end point (45 d post-exposure). Two of the 3 survivors experienced sensorineural hearing loss (described elsewhere). These results clearly demonstrate that the LASV DNA vaccine combined with dermal electroporation is a highly effective candidate for eventual use in humans.
- Published
- 2017
33. HIV Env conserved element DNA vaccine alters immunodominance in macaques
- Author
-
Niranjan Y. Sardesai, Barbara K. Felber, Siriphan Manocheewa, Xintao Hu, Antonio Valentin, Candido Alicea, Sylvie Le Gall, Jenifer Bear, Kate E. Broderick, George N. Pavlakis, Margherita Rosati, Bhabadeb Chowdhury, and James I. Mullins
- Subjects
Cytotoxicity, Immunologic ,DNA vaccine ,Env ,0301 basic medicine ,viruses ,Immunology ,Human immunodeficiency virus (HIV) ,Immunodominance ,CD8-Positive T-Lymphocytes ,variable epitopes ,Biology ,immunization ,medicine.disease_cause ,prime-boost ,Conserved sequence ,DNA vaccination ,03 medical and health sciences ,0302 clinical medicine ,Vaccines, DNA ,medicine ,Animals ,Humans ,Immunology and Allergy ,Conserved Sequence ,AIDS Vaccines ,Pharmacology ,Genetics ,subdominant epitopes ,conserved epitopes ,cytotoxic T cells ,env Gene Products, Human Immunodeficiency Virus ,HIV ,virus diseases ,Prime boost ,biology.organism_classification ,Macaca mulatta ,Research Papers ,Virology ,Rhesus macaque ,030104 developmental biology ,030220 oncology & carcinogenesis ,HIV-1 ,human activities ,rhesus macaque - Abstract
Sequence diversity and immunodominance are major obstacles in the design of an effective vaccine against HIV. HIV Env is a highly-glycosylated protein composed of ‘conserved’ and ‘variable’ regions. The latter contains immunodominant epitopes that are frequently targeted by the immune system resulting in the generation of immune escape variants. This work describes 12 regions in HIV Env that are highly conserved throughout the known HIV M Group sequences (Env CE), and are poorly immunogenic in macaques vaccinated with full-length Env expressing DNA vaccines. Two versions of plasmids encoding the 12 Env CE were generated, differing by 0–5 AA per CE to maximize the inclusion of commonly detected variants. In contrast to the full-length env DNA vaccine, vaccination of macaques with a combination of these 2 Env CE DNA induced robust, durable cellular immune responses with a significant fraction of CD8+ T cells with cytotoxic phenotype (Granzyme B+ and CD107a+). Although inefficient in generating primary responses to the CE, boosting of the Env CE DNA primed macaques with the intact env DNA vaccine potently augmented pre-existing immunity, increasing magnitude, breadth and cytotoxicity of the cellular responses. Fine mapping showed that 7 of the 12 CE elicited T cell responses. Env CE DNA also induced humoral responses able to recognize the full-length Env. Env CE plasmids are therefore capable of inducing durable responses to highly conserved regions of Env that are frequently absent after Env vaccination or immunologically subdominant. These modified antigens are candidates for use as prophylactic and therapeutic HIV vaccines.
- Published
- 2017
- Full Text
- View/download PDF
34. Characterization of guinea pig T cell responses elicited after EP-assisted delivery of DNA vaccines to the skin
- Author
-
Bryan S. Yung, Laurent Humeau, Karuppiah Muthumani, Trevor R.F. Smith, Katherine Schultheis, Janet Oh, Hubert Schaefer, and Kate E. Broderick
- Subjects
0301 basic medicine ,DNA vaccine ,Enzyme-Linked Immunospot Assay ,T cell ,T-Lymphocytes ,Population ,Guinea Pigs ,T cells ,ELISpot ,Biology ,Article ,DNA vaccination ,03 medical and health sciences ,Interferon-gamma ,Immune system ,Antigen ,Immunology and Microbiology(all) ,medicine ,Vaccines, DNA ,Animals ,education ,Antigen-presenting cell ,Skin ,education.field_of_study ,General Veterinary ,General Immunology and Microbiology ,ELISPOT ,Viral Core Proteins ,Public Health, Environmental and Occupational Health ,RNA-Binding Proteins ,Nucleocapsid Proteins ,Guinea pig ,Virology ,veterinary(all) ,Vaccination ,030104 developmental biology ,medicine.anatomical_structure ,Electroporation ,Infectious Diseases ,Influenza Vaccines ,Immunology ,Molecular Medicine ,Female - Abstract
The skin is an ideal target tissue for vaccine delivery for a number of reasons. It is highly accessible, and most importantly, enriched in professional antigen presenting cells. Possessing strong similarities to human skin physiology and displaying a defined epidermis, the guinea pig is an appropriate model to study epidermal delivery of vaccine. However, whilst we have characterized the humoral responses in the guinea pig associated with skin vaccine protocols we have yet to investigate the T cell responses. In response to this inadequacy, we developed an IFN-γ ELISpot assay to characterize the cellular immune response in the peripheral blood of guinea pigs. Using a nucleoprotein (NP) influenza pDNA vaccination regimen, we characterized host T cell responses. After delivery of the DNA vaccine to the guinea pig epidermis we detected robust and rapid T cell responses. The levels of IFN-γ spot-forming units averaged approximately 5000 per million cells after two immunizations. These responses were broad in that multiple regions across the NP antigen elicited a T cell response. Interestingly, we identified a number of NP immunodominant T cell epitopes to be conserved across an outbred guinea pig population, a phenomenon which was also observed after immunization with a RSV DNA vaccine. We believe this data enhances our understanding of the cellular immune response elicited to a vaccine in guinea pigs, and globally, will advance the use of this model for vaccine development, especially those targeting skin as a delivery site.
- Published
- 2017
- Full Text
- View/download PDF
35. In vivo delivery of synthetic DNA-encoded antibodies induces broad HIV-1-neutralizing activity
- Author
-
Ami Patel, Karuppiah Muthumani, Jingjing Jiang, David C. Montefiori, Guido Ferrari, Sarah T. C. Elliott, Edgar Tello-Ruiz, Ziyang Xu, Janess M. Mendoza, David B. Weiner, Melissa Kerkau, Charles Beck, Aspen Trautz, Paul Fisher, Daniel W. Kulp, Laurent Humeau, Sophie Kim, Stephany J. Ramos, Kate E. Broderick, Neethu Chokkalingam, Trevor R.F. Smith, and Megan C. Wise
- Subjects
0301 basic medicine ,medicine.drug_class ,Genetic enhancement ,medicine.medical_treatment ,HIV Antibodies ,Monoclonal antibody ,Neutralization ,law.invention ,03 medical and health sciences ,Antibodies, Monoclonal, Murine-Derived ,Mice ,0302 clinical medicine ,In vivo ,law ,medicine ,Animals ,Humans ,Mice, Inbred BALB C ,biology ,General Medicine ,Immunotherapy ,Virology ,Antibodies, Neutralizing ,030104 developmental biology ,HEK293 Cells ,Infectious disease (medical specialty) ,030220 oncology & carcinogenesis ,biology.protein ,Recombinant DNA ,HIV-1 ,Female ,Antibody ,Research Article - Abstract
Interventions to prevent HIV-1 infection and alternative tools in HIV cure therapy remain pressing goals. Recently, numerous broadly neutralizing HIV-1 monoclonal antibodies (bNAbs) have been developed that possess the characteristics necessary for potential prophylactic or therapeutic approaches. However, formulation complexities, especially for multiantibody deliveries, long infusion times, and production issues could limit the use of these bNAbs when deployed, globally affecting their potential application. Here, we describe an approach utilizing synthetic DNA-encoded monoclonal antibodies (dmAbs) for direct in vivo production of prespecified neutralizing activity. We designed 16 different bNAbs as dmAb cassettes and studied their activity in small and large animals. Sera from animals administered dmAbs neutralized multiple HIV-1 isolates with activity similar to that of their parental recombinant mAbs. Delivery of multiple dmAbs to a single animal led to increased neutralization breadth. Two dmAbs, PGDM1400 and PGT121, were advanced into nonhuman primates for study. High peak-circulating levels (between 6 and 34 μg/ml) of these dmAbs were measured, and the sera of all animals displayed broad neutralizing activity. The dmAb approach provides an important local delivery platform for the in vivo generation of HIV-1 bNAbs and for other infectious disease antibodies.
- Published
- 2019
36. Intradermal Synthetic DNA Vaccination Generates Leishmania -Specific T Cells in the Skin and Protection against Leishmania major
- Author
-
David B. Weiner, Jude E. Uzonna, Andrea C. Wong, Megan L. Clark, Phillip Scott, Lumena Louis, Nelson D. Glennie, Megan C. Wise, and Kate E. Broderick
- Subjects
0301 basic medicine ,biology ,Electroporation ,Immunology ,Leishmania ,biology.organism_classification ,Microbiology ,Virology ,DNA vaccination ,Vaccination ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Infectious Diseases ,Immune system ,Immunization ,Parasitology ,Leishmania major ,Intradermal injection ,030215 immunology - Abstract
Vaccination remains one of the greatest medical breakthroughs in human history and has resulted in the near eradication of many formerly lethal diseases in many countries, including the complete eradication of smallpox. However, there remain a number of diseases for which there are no or only partially effective vaccines. There are numerous hurdles in vaccine development, of which knowing the appropriate immune response to target is one of them. Recently, tissue-resident T cells have been shown to mediate high levels of protection for several infections, although the best way to induce these cells is still unclear. Here we compare the ability to generate skin-resident T cells in sites distant from the immunization site following intramuscular and intradermal injection using optimized synthetic DNA vaccines. We found that mice immunized intradermally with a synthetic consensus DNA HIV envelope vaccine by electroporation (EP) are better able to maintain durable antigen-specific cellular responses in the skin than mice immunized by the intramuscular route. We extended these studies by delivering a synthetic DNA vaccine encoding Leishmania glycosomal phosphoenolpyruvate carboxykinase (PEPCK) by EP and again found that the intradermal route was superior to the intramuscular route for generating skin-resident PEPCK-specific T cells. We observed that when challenged with Leishmania major parasites, mice immunized intradermally exhibited significant protection, while mice immunized intramuscularly did not. The protection seen in intradermally vaccinated mice supports the viability of this platform not only to generate skin-resident T cells but also to promote durable protective immune responses at relevant tissue sites.
- Published
- 2019
- Full Text
- View/download PDF
37. DNA Prime-Boost Vaccine Regimen To Increase Breadth, Magnitude, and Cytotoxicity of the Cellular Immune Responses to Subdominant Gag Epitopes of Simian Immunodeficiency Virus and HIV
- Author
-
Malcolm A. Martin, Frances Dayton, George N. Pavlakis, Bhabadeb Chowdhury, Barbara K. Felber, Margherita Rosati, Antonio Valentin, James I. Mullins, Niranjan Y. Sardesai, Candido Alicea, Xintao Hu, Rajeev Gautam, Viraj Kulkarni, and Kate E. Broderick
- Subjects
Cytotoxicity, Immunologic ,0301 basic medicine ,Subdominant ,T cell ,Immunology ,Immunization, Secondary ,Simian Acquired Immunodeficiency Syndrome ,Gene Products, gag ,HIV Infections ,Immunodominance ,Biology ,medicine.disease_cause ,Epitope ,Epitopes ,03 medical and health sciences ,Vaccines, DNA ,medicine ,Animals ,Immunology and Allergy ,Cytotoxic T cell ,HIV vaccine ,Immunization Schedule ,Immunotherapy and Vaccines ,AIDS Vaccines ,SAIDS Vaccines ,HIV ,virus diseases ,Simian immunodeficiency virus ,Macaca mulatta ,Virology ,Vaccination ,030104 developmental biology ,medicine.anatomical_structure ,Cytokines ,Simian Immunodeficiency Virus - Abstract
HIV sequence diversity and the propensity of eliciting immunodominant responses targeting variable regions of the HIV proteome are hurdles in the development of an effective AIDS vaccine. An HIV-derived conserved element (CE) p24gag plasmid DNA (pDNA) vaccine is able to redirect immunodominant responses to otherwise subdominant and often more vulnerable viral targets. By homology to the HIV immunogen, seven CE were identified in SIV p27Gag. Analysis of 31 rhesus macaques vaccinated with full-length SIV gag pDNA showed inefficient induction (58% response rate) of cellular responses targeting these CE. In contrast, all 14 macaques immunized with SIV p27CE pDNA developed robust T cell responses recognizing CE. Vaccination with p27CE pDNA was also critical for the efficient induction and increased the frequency of Ag-specific T cells with cytotoxic potential (granzyme B+ CD107a+) targeting subdominant CE epitopes, compared with the responses elicited by the p57gag pDNA vaccine. Following p27CE pDNA priming, two booster regimens, gag pDNA or codelivery of p27CE+gag pDNA, significantly increased the levels of CE-specific T cells. However, the CE+gag pDNA booster vaccination elicited significantly broader CE epitope recognition, and thus, a more profound alteration of the immunodominance hierarchy. Vaccination with HIV molecules showed that CE+gag pDNA booster regimen further expanded the breadth of HIV CE responses. Hence, SIV/HIV vaccine regimens comprising CE pDNA prime and CE+gag pDNA booster vaccination significantly increased cytotoxic T cell responses to subdominant highly conserved Gag epitopes and maximized response breadth.
- Published
- 2016
- Full Text
- View/download PDF
38. Anti-OspA DNA-Encoded Monoclonal Antibody Prevents Transmission of Spirochetes in Tick Challenge Providing Sterilizing Immunity in Mice
- Author
-
Kate E. Broderick, Aurelie Kern, David B. Weiner, Katherine P. Sullivan, Yang Wang, Jacqueline Chu, Rianne Esquivel, Seleeke Flingai, Zachary A. Schiller, Ami Patel, Mark S. Klempner, Linden T. Hu, Sangya Agarwal, and Megan C. Wise
- Subjects
0301 basic medicine ,DNA, Bacterial ,medicine.drug_class ,Lipoproteins ,Mice, SCID ,Tick ,Monoclonal antibody ,Antibodies, Monoclonal, Humanized ,Transfection ,03 medical and health sciences ,chemistry.chemical_compound ,Major Articles and Brief Reports ,Mice ,0302 clinical medicine ,Lyme disease ,Plasmid ,Ticks ,Immunity ,Borrelia ,medicine ,Immunology and Allergy ,Animals ,Humans ,030212 general & internal medicine ,Lyme Disease ,Mice, Inbred C3H ,biology ,biology.organism_classification ,medicine.disease ,bacterial infections and mycoses ,Virology ,030104 developmental biology ,Infectious Diseases ,HEK293 Cells ,chemistry ,Borrelia burgdorferi ,Antigens, Surface ,Bacterial Vaccines ,biology.protein ,Female ,Antibody ,DNA ,Bacterial Outer Membrane Proteins ,Plasmids - Abstract
We recently developed anti-OspA human immunoglobulin G1 monoclonal antibodies (HuMAbs) that are effective in preventing Borrelia transmission from ticks in a murine model. Here, we investigated a novel approach of DNA-mediated gene transfer of HuMAbs that provide protection against Lyme disease. Plasmid DNA-encoded anti-OspA HuMAbs inoculated in mice achieved a serum antibody concentration of >6 μg/mL. Among mice injected with DNA-encoded monoclonal antibodies, 75%–77% were protected against an acute challenge by Borrelia-infected ticks. Our results represent the first demonstration of employing DNA transfer as a delivery system for antibodies that block transmission of Borrelia in animal models.
- Published
- 2018
39. Control of Heterologous Simian Immunodeficiency Virus SIVsmE660 Infection by DNA and Protein Coimmunization Regimens Combined with Different Toll-Like-Receptor-4-Based Adjuvants in Macaques
- Author
-
Shakti Singh, Steven G. Reed, Jeffrey D. Lifson, Georgia D. Tomaras, Jenifer Bear, Mangala Rao, Brandon F. Keele, Guido Ferrari, Christopher B. Fox, Margherita Rosati, Vanessa M. Hirsch, Celia C. LaBranche, Bhabadeb Chowdhury, Frances Dayton, George N. Pavlakis, Eric G. Ramírez-Salazar, Candido Alicea, Hung V. Trinh, Galit Alter, David Venzon, Kate E. Broderick, Jishnu Das, Xiaoying Shen, Christopher Hamlin, Antonio Valentin, Barbara K. Felber, David C. Montefiori, Xintao Hu, Rami Doueiri, Niranjan Y. Sardesai, and Sandra J. Sivananthan
- Subjects
0301 basic medicine ,SIVmac251 ,viruses ,medicine.medical_treatment ,Simian Acquired Immunodeficiency Syndrome ,medicine.disease_cause ,Antibodies, Viral ,TRIM-5α ,repeated low-dose rectal challenge ,0302 clinical medicine ,V2 responses ,vaccine ,SIVsmE660 T/F ,Vaccines, DNA ,cyclic V2 ,030212 general & internal medicine ,TLR4 ,Neutralizing antibody ,reduced risk of infection ,ADNP ,TLR7 ,Antibody-dependent cell-mediated cytotoxicity ,mucosal responses ,SAIDS Vaccines ,neutralizing antibody ,QS21 ,Vaccination ,scaffolded gp70-V1V2 ,SIVsmE660 ,viremia control ,Simian Immunodeficiency Virus ,ADCC ,Adjuvant ,ADCD ,binding antibody ,Immunology ,Mutation, Missense ,linear peptide ,Biology ,immunization ,Microbiology ,Virus ,03 medical and health sciences ,adjuvant ,Adjuvants, Immunologic ,Virology ,Vaccines and Antiviral Agents ,medicine ,Animals ,systems serology ,correlate of viremia control ,humoral responses ,HIV ,Gene Products, env ,DNA ,Simian immunodeficiency virus ,A/K variant ,Vaccine efficacy ,vaccination ,Antibodies, Neutralizing ,Immunity, Humoral ,Toll-Like Receptor 4 ,030104 developmental biology ,Amino Acid Substitution ,T cell responses ,Insect Science ,biology.protein ,Macaca ,protein ,Ab glycosylation structures ,acquisition delay ,rhesus macaque - Abstract
An effective AIDS vaccine continues to be of paramount importance for the control of the pandemic, and it has been proven to be an elusive target. Vaccine efficacy trials and macaque challenge studies indicate that protection may be the result of combinations of many parameters. We show that a combination of DNA and protein vaccinations applied at the same time provides rapid and robust cellular and humoral immune responses and evidence for a reduced risk of infection. Vaccine-induced neutralizing antibodies and Env V2-specific antibodies at mucosal sites contribute to the delay of SIVsmE660 acquisition, and genetic makeup (TRIM-5α) affects the effectiveness of the vaccine. These data are important for the design of better vaccines and may also affect other vaccine platforms., We developed a method of simultaneous vaccination with DNA and protein resulting in robust and durable cellular and humoral immune responses with efficient dissemination to mucosal sites and protection against simian immunodeficiency virus (SIV) infection. To further optimize the DNA-protein coimmunization regimen, we tested a SIVmac251-based vaccine formulated with either of two Toll-like receptor 4 (TLR4) ligand-based liposomal adjuvant formulations (TLR4 plus TLR7 [TLR4+7] or TLR4 plus QS21 [TLR4+QS21]) in macaques. Although both vaccines induced humoral responses of similar magnitudes, they differed in their functional quality, including broader neutralizing activity and effector functions in the TLR4+7 group. Upon repeated heterologous SIVsmE660 challenge, a trend of delayed viral acquisition was found in vaccinees compared to controls, which reached statistical significance in animals with the TRIM-5α-resistant (TRIM-5α R) allele. Vaccinees were preferentially infected by an SIVsmE660 transmitted/founder virus carrying neutralization-resistant A/K mutations at residues 45 and 47 in Env, demonstrating a strong vaccine-induced sieve effect. In addition, the delay in virus acquisition directly correlated with SIVsmE660-specific neutralizing antibodies. The presence of mucosal V1V2 IgG binding antibodies correlated with a significantly decreased risk of virus acquisition in both TRIM-5α R and TRIM-5α-moderate/sensitive (TRIM-5α M/S) animals, although this vaccine effect was more prominent in animals with the TRIM-5α R allele. These data support the combined contribution of immune responses and genetic background to vaccine efficacy. Humoral responses targeting V2 and SIV-specific T cell responses correlated with viremia control. In conclusion, the combination of DNA and gp120 Env protein vaccine regimens using two different adjuvants induced durable and potent cellular and humoral responses contributing to a lower risk of infection by heterologous SIV challenge. IMPORTANCE An effective AIDS vaccine continues to be of paramount importance for the control of the pandemic, and it has been proven to be an elusive target. Vaccine efficacy trials and macaque challenge studies indicate that protection may be the result of combinations of many parameters. We show that a combination of DNA and protein vaccinations applied at the same time provides rapid and robust cellular and humoral immune responses and evidence for a reduced risk of infection. Vaccine-induced neutralizing antibodies and Env V2-specific antibodies at mucosal sites contribute to the delay of SIVsmE660 acquisition, and genetic makeup (TRIM-5α) affects the effectiveness of the vaccine. These data are important for the design of better vaccines and may also affect other vaccine platforms.
- Published
- 2018
40. Nucleic acid-based vaccines targeting respiratory syncytial virus: Delivering the goods
- Author
-
Katherine Schultheis, Kate E. Broderick, and Trevor R.F. Smith
- Subjects
0301 basic medicine ,Rsv vaccine ,viruses ,DNA/RNA vaccine ,Immunology ,Target population ,Disease ,Respiratory Syncytial Virus Infections ,Biology ,Respiratory syncytial virus ,Antibodies, Viral ,Virus ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Immune system ,Drug Delivery Systems ,Immunogenicity, Vaccine ,Th2 Cells ,Respiratory Syncytial Virus Vaccines ,Vaccines, DNA ,Immunology and Allergy ,Animals ,Humans ,030212 general & internal medicine ,Child ,Pharmacology ,Immunogenicity ,Vaccination ,Infant ,vaccine delivery ,Virology ,030104 developmental biology ,Respiratory Syncytial Virus, Human ,DNA, Viral ,Nucleic acid ,Commentary ,RNA, Viral ,Female - Abstract
Respiratory syncytial virus (RSV) is a massive medical burden on a global scale. Infants, children and the elderly represent the vulnerable populations. Currently there is no approved vaccine to protect against the disease. Vaccine development has been hindered by several factors including vaccine enhanced disease (VED) associated with formalin-inactivated RSV vaccines, inability of target populations to raise protective immune responses after vaccination or natural viral infection, and a lack of consensus concerning the most appropriate virus-associated target antigen. However, with recent advances in the molecular understanding of the virus, and design of highly characterized vaccines with enhanced immunogenicity there is new belief a RSV vaccine is possible. One promising approach is nucleic acid-based vaccinology. Both DNA and mRNA RSV vaccines are showing promising results in clinically relevant animal models, supporting their transition into humans. Here we will discuss this strategy to target RSV, and the ongoing studies to advance the nucleic acid vaccine platform as a viable option to protect vulnerable populations from this important disease.
- Published
- 2017
41. An engineered bispecific DNA-encoded IgG antibody protects against Pseudomonas aeruginosa in a pneumonia challenge model
- Author
-
Antonio DiGiandomenico, Karuppiah Muthumani, Ashley E. Keller, Niranjan Y. Sardesai, Leslie Wachter-Rosati, Lily Cheng, Janess M. Mendoza, David B. Weiner, Laurent Humeau, Stephanie Ramos, C. Kendall Stover, Daniel H. Park, Jingjing Jiang, Seleeke Flingai, Amir S. Khan, Kate E. Broderick, Sarah T. C. Elliott, Jian Yan, Ami Patel, Katherine Schultheis, Megan C. Wise, and Trevor R.F. Smith
- Subjects
0301 basic medicine ,medicine.drug_class ,Science ,Antibiotics ,General Physics and Astronomy ,Biology ,Monoclonal antibody ,medicine.disease_cause ,Protein Engineering ,General Biochemistry, Genetics and Molecular Biology ,Immunoglobulin G ,Article ,Microbiology ,03 medical and health sciences ,Mice ,Antibiotic resistance ,Immune system ,In vivo ,Antibodies, Bispecific ,medicine ,Pneumonia, Bacterial ,Animals ,Humans ,Pseudomonas Infections ,lcsh:Science ,Mice, Inbred BALB C ,Multidisciplinary ,Pseudomonas aeruginosa ,Antibodies, Monoclonal ,General Chemistry ,Antibodies, Bacterial ,030104 developmental biology ,HEK293 Cells ,Immunology ,biology.protein ,lcsh:Q ,Antibody - Abstract
The impact of broad-spectrum antibiotics on antimicrobial resistance and disruption of the beneficial microbiome compels the urgent investigation of bacteria-specific approaches such as antibody-based strategies. Among these, DNA-delivered monoclonal antibodies (DMAbs), produced by muscle cells in vivo, potentially allow the prevention or treatment of bacterial infections circumventing some of the hurdles of protein IgG delivery. Here, we optimize DNA-delivered monoclonal antibodies consisting of two potent human IgG clones, including a non-natural bispecific IgG1 candidate, targeting Pseudomonas aeruginosa. The DNA-delivered monoclonal antibodies exhibit indistinguishable potency compared to bioprocessed IgG and protect against lethal pneumonia in mice. The DNA-delivered monoclonal antibodies decrease bacterial colonization of organs and exhibit enhanced adjunctive activity in combination with antibiotics. These studies support DNA-delivered monoclonal antibodies delivery as a potential strategy to augment the host immune response to prevent serious bacterial infections, and represent a significant advancement toward broader practical delivery of monoclonal antibody immunotherapeutics for additional infectious pathogens., DNA-delivered monoclonal antibodies (DMAbs) can be produced by muscle cells in vivo, potentially allowing prevention or treatment of infectious diseases. Here, the authors show that two DMAbs targeting Pseudomonas aeruginosa proteins confer protection against lethal pneumonia in mice.
- Published
- 2017
42. DMAb inoculation of synthetic cross reactive antibodies protects against lethal influenza A and B infections
- Author
-
Qing Zhu, Janess M. Mendoza, Sarah T. C. Elliott, David B. Weiner, Ami Patel, Trevor R.F. Smith, Leslie Wachter-Rosati, Jian Yan, Megan C. Wise, Seleeke Flingai, Nicole L. Kallewaard, Karuppiah Muthumani, Daniel H. Park, Ebony Benjamin, Kate E. Broderick, Josephine M. McAuliffe, Niranjan Y. Sardesai, Laurent Humeau, Stephanie Ramos, and Katherine Schultheis
- Subjects
0301 basic medicine ,medicine.drug_class ,Immunology ,Biology ,Monoclonal antibody ,Article ,Virus ,Microbiology ,03 medical and health sciences ,0302 clinical medicine ,Plasmid ,In vivo ,Pandemic ,medicine ,Pharmacology (medical) ,RC254-282 ,Pharmacology ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC581-607 ,Virology ,3. Good health ,Vaccination ,030104 developmental biology ,Infectious Diseases ,Immunization ,030220 oncology & carcinogenesis ,biology.protein ,Immunologic diseases. Allergy ,Antibody - Abstract
Influenza virus remains a significant public health threat despite innovative vaccines and antiviral drugs. A major limitation to current vaccinations and therapies against influenza virus is pathogenic diversity generated by shift and drift. A simple, cost-effective passive immunization strategy via in vivo production of cross-protective antibody molecules may augment existing vaccines and antiviral drugs in seasonal and pandemic outbreaks. We engineered synthetic plasmid DNA to encode two novel and broadly cross-protective monoclonal antibodies targeting influenza A and B. We utilized enhanced in vivo delivery of these plasmid DNA-encoded monoclonal antibody (DMAb) constructs and show that this strategy induces robust levels of functional antibodies directed against influenza A and B viruses in mouse sera. Mice receiving a single inoculation with anti-influenza A DMAb survive lethal Group 1 H1 and Group 2 H3 influenza A challenges, while inoculation with anti-influenza B DMAb yields protection against lethal Victoria and Yamagata lineage influenza B morbidity and mortality. Furthermore, these two DMAbs can be delivered coordinately resulting in exceptionally broad protection against both influenza A and B. We demonstrate this protection is similar to that achieved by conventional protein antibody delivery. DMAbs warrant further investigation as a novel immune therapy platform with distinct advantages for sustained immunoprophylaxis against influenza., Nucleic acid delivery: Instant, wide-ranging protection against influenza A and B A novel innoculation technique involving the injection of antibody-producing plasmid DNA has shown to be effective against influenza in mice. The flu is responsible for up to half a million deaths each year and up to five million cases of severe disease, while also posing a substantial pandemic threat, even with our current repertoire of vaccines. A team of researchers led by Sarah Elliott and David Weiner of The Wistar Institute of Anatomy and Biology, Philadelphia, developed potent plasmid-based constructs that, once injected, entered hosts’ cells and utilized cellular machinery to encode antibodies protective against a range of influenza A and B subtypes. DNA inoculation conferred acute protection from disease, with treated individuals also being immune to subsequent exposure. This approach warrants further investigation as an alternative technology for practical delivery of monoclonal antibody therapeutics.
- Published
- 2017
- Full Text
- View/download PDF
43. Development of an intradermal DNA vaccine delivery strategy to achieve single-dose immunity against respiratory syncytial virus
- Author
-
Katherine Schultheis, Kate E. Broderick, Trevor R.F. Smith, Niranjan Y. Sardesai, Kevin C. Yim, David B. Weiner, Matthew P. Morrow, Laurent Humeau, Kimberly A. Kraynyak, Jay Mccoy, and Karuppiah Muthumani
- Subjects
0301 basic medicine ,viruses ,Respiratory Syncytial Virus Infections ,Virus ,Article ,DNA vaccination ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Antigen ,Immunity ,Respiratory Syncytial Virus Vaccines ,Vaccines, DNA ,Animals ,030212 general & internal medicine ,Cotton rat ,Sigmodontinae ,Lung ,General Veterinary ,General Immunology and Microbiology ,biology ,Public Health, Environmental and Occupational Health ,respiratory system ,biology.organism_classification ,Virology ,Vaccination ,Disease Models, Animal ,030104 developmental biology ,Infectious Diseases ,Electroporation ,Treatment Outcome ,Immunology ,Molecular Medicine ,Female - Abstract
Respiratory syncytial virus (RSV) is a massive medical burden in infants, children and the elderly worldwide, and an effective, safe RSV vaccine remains an unmet need. Here we assess a novel vaccination strategy based on the intradermal delivery of a SynCon® DNA-based vaccine encoding engineered RSV-F antigen using a surface electroporation device (SEP) to target epidermal cells, in clinically relevant experimental models. We demonstrate the ability of this strategy to elicit robust immune responses. Importantly we demonstrate complete resistance to pulmonary infection at a single low dose of vaccine in the cotton rat RSV/A challenge model. In contrast to the formalin-inactivated RSV (FI-RSV) vaccine, there was no enhanced lung inflammation upon virus challenge after DNA vaccination. In summary the data presented outline the pre-clinical development of a highly efficacious, tolerable and safe non-replicating vaccine delivery strategy.
- Published
- 2017
44. Electroporation-enhanced delivery of nucleic acid vaccines
- Author
-
Laurent Humeau and Kate E. Broderick
- Subjects
Pharmacology ,Clinical Trials as Topic ,Electroporation ,Immunology ,Cell ,RNA ,Biology ,Virology ,DNA vaccination ,Cell biology ,chemistry.chemical_compound ,Nucleic Acid Vaccines ,Immune system ,medicine.anatomical_structure ,chemistry ,Drug Discovery ,Vaccines, DNA ,Nucleic acid ,medicine ,Animals ,Humans ,Molecular Medicine ,DNA - Abstract
The naked delivery of nucleic acid vaccines is notoriously inefficient, and an enabling delivery technology is required to direct efficiently these constructs intracellularly. A delivery technology capable of enhancing nucleic acid uptake in both cells in tissues and in culture is electroporation (EP). EP is a physical delivery mechanism that increases the permeability of mammalian cell membranes and allows the trafficking of large macromolecules into the cell. EP has now been used extensively in the clinic and been shown to be an effective method to increase both the uptake of the construct and the breadth and magnitude of the resulting immune responses. Excitingly, 2014 saw the announcement of the first EP-enhanced DNA vaccine Phase II trial demonstrating clinical efficacy. This review seeks to introduce the reader to EP as a technology to enhance the delivery of DNA and RNA vaccines and highlight several published clinical trials using this delivery modality.
- Published
- 2014
- Full Text
- View/download PDF
45. Electroporation mediated DNA vaccination directly to a mucosal surface results in improved immune responses
- Author
-
Trevor R.F. Smith, Niranjan Y. Sardesai, Gleb Kichaev, Jay Mccoy, Dinah Amante, Janess M. Mendoza, and Kate E. Broderick
- Subjects
Immunoglobulin A ,Guinea Pigs ,Immunology ,Antibodies, Viral ,Immunoglobulin G ,DNA vaccination ,Immune system ,Vaccines, DNA ,medicine ,Administration, Mucosal ,Animals ,Immunology and Allergy ,Oral mucosa ,Immunity, Mucosal ,Pharmacology ,Mice, Inbred BALB C ,biology ,Immunogenicity ,Electroporation ,Vaccination ,Mouth Mucosa ,medicine.anatomical_structure ,Influenza Vaccines ,Vagina ,biology.protein ,Female ,Research Paper - Abstract
In vivo electroporation (EP) has been shown to be a highly efficient non-viral method for enhancing DNA vaccine delivery and immunogenicity, when the site of immunization is the skin or muscle of animals and humans. However, the route of entry for many microbial pathogens is via the mucosal surfaces of the human body. We have previously reported on minimally invasive, surface and contactless EP devices for enhanced DNA delivery to dermal tissue. Robust antibody responses were induced following vaccine delivery in several tested animal models using these devices. Here, we investigated extending the modality of the surface device to efficiently deliver DNA vaccines to mucosal tissue. Initially, we demonstrated reporter gene expression in the epithelial layer of buccal mucosa in a guinea pig model. There was minimal tissue damage in guinea pig mucosal tissue resulting from EP. Delivery of a DNA vaccine encoding influenza virus nucleoprotein (NP) of influenza H1N1 elicited robust and sustained systemic IgG antibody responses following EP-enhanced delivery in the mucosa. Upon further analysis, IgA antibody responses were detected in vaginal washes and sustained cellular immune responses were detected in animals immunized at the oral mucosa with the surface EP device. This data confirms that DNA delivery and EP targeting mucosal tissue directly results in both robust and sustainable humoral as well as cellular immune responses without tissue damage. These responses are seen both in the mucosa and systemically in the blood. Direct DNA vaccine delivery enhanced by EP in mucosa may have important clinical applications for delivery of prophylactic and therapeutic DNA vaccines against diseases such as HIV, HPV and pneumonia that enter at mucosal sites and require both cellular and humoral immune responses for protection.
- Published
- 2013
- Full Text
- View/download PDF
46. Elucidating the Kinetics of Expression and Immune Cell Infiltration Resulting from Plasmid Gene Delivery Enhanced by Surface Dermal Electroporation
- Author
-
Dinah Amante, Christine L Knott, Trevor R.F. Smith, Janess M. Mendoza, William B. Kiosses, Gleb Kichaev, Niranjan Y. Sardesai, and Kate E. Broderick
- Subjects
DNA vaccine ,electroporation ,Pathology ,medicine.medical_specialty ,Immunology ,lcsh:Medicine ,Gene delivery ,Biology ,infiltration ,Article ,DNA vaccination ,Green fluorescent protein ,Immune system ,Antigen ,Drug Discovery ,medicine ,Pharmacology (medical) ,intradermal ,Pharmacology ,Reporter gene ,Electroporation ,lcsh:R ,Cell biology ,Infectious Diseases ,medicine.anatomical_structure ,kinetics ,Keratinocyte - Abstract
The skin is an attractive tissue for vaccination in a clinical setting due to the accessibility of the target, the ease of monitoring and most importantly the immune competent nature of the dermal tissue. While skin electroporation offers an exciting and novel future methodology for the delivery of DNA vaccines in the clinic, little is known about the actual mechanism of the approach and the elucidation of the resulting immune responses. To further understand the mechanism of this platform, the expression kinetics and localization of a reporter plasmid delivered via a surface dermal electroporation (SEP) device as well as the effect that this treatment would have on the resident immune cells in that tissue was investigated. Initially a time course (day 0 to day 21) of enhanced gene delivery with electroporation (EP) was performed to observe the localization of green fluorescent protein (GFP) expression and the kinetics of its appearance as well as clearance. Using gross imaging, GFP expression was not detected on the surface of the skin until 8 h post treatment. However, histological analysis by fluorescent microscopy revealed GFP positive cells as early as 1 h after plasmid delivery and electroporation. Peak GFP expression was observed at 24 h and the expression was maintained in skin for up to seven days. Using an antibody specific for a keratinocyte cell surface marker, reporter gene positive keratinocytes in the epidermis were identified. H&, E staining of treated skin sections demonstrated an influx of monocytes and granulocytes at the EP site starting at 4 h and persisting up to day 14 post treatment. Immunological staining revealed a significant migration of lymphocytic cells to the EP site, congregating around cells expressing the delivered antigen. In conclusion, this study provides insights into the expression kinetics following EP enhanced DNA delivery targeting the dermal space. These findings may have implications in the future to design efficient DNA vaccination strategies for the clinic.
- Published
- 2013
47. Enhanced Efficacy of a Codon-Optimized DNA Vaccine Encoding the Glycoprotein Precursor Gene of Lassa Virus in a Guinea Pig Disease Model When Delivered by Dermal Electroporation
- Author
-
Amy C. Shurtleff, Eric R. Wilkinson, Todd M. Bell, Kristin Spik, Carl I Shaia, Connie S. Schmaljohn, Catherine V. Badger, Niranjan Y. Sardesai, Kate E. Broderick, Kathleen A. Cashman, and Mary C. Guttieri
- Subjects
Immunology ,lcsh:Medicine ,Viremia ,medicine.disease_cause ,Article ,Virus ,DNA vaccination ,vaccine ,Drug Discovery ,medicine ,Pharmacology (medical) ,Lassa virus ,arenavirus ,Lassa fever ,Pharmacology ,Arenavirus ,biology ,business.industry ,Electroporation ,lcsh:R ,guinea pigs ,dermal electroporation ,vaccination ,medicine.disease ,biology.organism_classification ,Virology ,Vaccination ,Infectious Diseases ,business - Abstract
Lassa virus (LASV) causes a severe, often fatal, hemorrhagic fever endemic to West Africa. Presently, there are no FDA-licensed medical countermeasures for this disease. In a pilot study, we constructed a DNA vaccine (pLASV-GPC) that expressed the LASV glycoprotein precursor gene (GPC). This plasmid was used to vaccinate guinea pigs (GPs) using intramuscular electroporation as the delivery platform. Vaccinated GPs were protected from lethal infection (5/6) with LASV compared to the controls. However, vaccinated GPs experienced transient viremia after challenge, although lower than the mock-vaccinated controls. In a follow-on study, we developed a new device that allowed for both the vaccine and electroporation pulse to be delivered to the dermis. We also codon-optimized the GPC sequence of the vaccine to enhance expression in GPs. Together, these innovations resulted in enhanced efficacy of the vaccine. Unlike the pilot study where neutralizing titers were not detected until after virus challenge, modest neutralizing titers were detected in guinea pigs before challenge, with escalating titers detected after challenge. The vaccinated GPs were never ill and were not viremic at any timepoint. The combination of the codon-optimized vaccine and dermal electroporation delivery is a worthy candidate for further development.
- Published
- 2013
- Full Text
- View/download PDF
48. A Heterologous Prime/Boost Vaccination Strategy Enhances the Immunogenicity of Therapeutic Vaccines for Hepatitis C Virus
- Author
-
Lars Frelin, Emilie Jacquier, Anne Fournillier, Anette Brass, Niranjan Y. Sardesai, Estelle Gerossier, Fredrik Holmström, Kate E. Broderick, Jean-Yves Bonnefoy, Matti Sällberg, Geneviève Inchauspé, and Gustaf Ahlén
- Subjects
electroporation ,CD4-Positive T-Lymphocytes ,Viral Hepatitis Vaccines ,Genotype ,Immunization, Secondary ,Mice, Transgenic ,Hepacivirus ,Biology ,CD8-Positive T-Lymphocytes ,DNA vaccination ,Viral vector ,chemistry.chemical_compound ,Major Articles and Brief Reports ,Interferon-gamma ,Mice ,Antigen ,Aldesleukin ,Vaccines, DNA ,Immunology and Allergy ,Animals ,Tumor Necrosis Factor-alpha ,Ribavirin ,Immunogenicity ,prime/boost ,Vaccination ,Virology ,Hepatitis C ,Mice, Inbred C57BL ,Infectious Diseases ,chemistry ,Immunology ,Viruses ,HCV ,Antibody Formation ,Interleukin-2 ,therapeutic vaccine ,Viral load - Abstract
After acute hepatitis C virus (HCV) infection, 20% of individuals clear the virus, which is dependent on sustained Th1 CD4+ T lymphocyte–mediated responses together with polyfunctional CD8+ T cells [1–4]. HCV is highly efficient in establishing persistent infections, and the specific T-cell responses are impaired during chronic infection [5–8] due to appearance of escape mutants [4, 9], inhibitory effects exerted by viral proteins [10], or expression of coinhibitory receptors resulting in T-cell exhaustion [11, 12]. One way to prime T cells and/or reactivate impaired T cells is to express HCV antigens under a more “immunogenic” setting than natural infection where massive antigen expression appears in the tolerogenic liver environment. Therapeutic vaccination has been tested with some success in HCV-infected patients showing evidence of T-cell activation [13, 14]. Vectored vaccines tailored to generate robust T-cell immunity have recently reached the clinic. MVATG16643, a modified virus Ankara (MVA)–based vaccine [15], has shown in a phase I trial good safety and the capacity to induce interferon γ (IFN-γ)–producing T cells with significant although transient viral load decrease in chronically infected patients [16]. Used in combination with pegylated (PEG)–interferon α (IFN-α)/ribavirin in a phase II clinical trial, MVATG16643 resulted in a significant early viral response [17]. The DNA vaccine ChronVac-C [18] has also shown the capacity to induce T cells, which had transient effects on viral load in a phase I/IIa trial [19], and it has been suggested that it improves cure rates when given before PEG–IFN-α/ribavirin treatment [19]. The combined use of DNA vaccines with viral vectors in a prime/boost regimen has been proven useful for enhancing response levels in clinical studies [20–22]. Similarly, in vivo electroporation (EP)–mediated delivery of DNA vaccines either as stand-alone or in a prime/boost setting with viral vectors has also served to enhance the development of polyfunctional CD8+ T-cell responses [23, 24]. Here we have performed a proof-of-concept study to define the extent of improvement that could result from a prime/boost approach based on ChronVac-C and MVATG16643—2 individual vaccines currently in the clinic. These 2 vaccine regimens have been optimized extensively individually previously, and we wanted to investigate whether the combination of 2 regimens could confer additional benefits over the individual regimens. This study thus combines for the first time in the HCV setting approaches previously shown individually to enhance immunogenicity (ie, DNA vaccine delivery with in vivo EP and prime/boost using viral vectors). We performed an exhaustive evaluation of this concept in wild-type C57BL/6J and human leucocyte antigen (HLA)–A2 transgenic mice. This strategy was found very potent for the improvement of polyfunctional CD4+ and CD8+ HCV-specific responses and resulted in a significant increase of epitope recognition.
- Published
- 2013
49. The p40 Subunit of Interleukin (IL)-12 Promotes Stabilization and Export of the p35 Subunit
- Author
-
Gen-Mu Zhang, Rashmi Jalah, Niranjan Y. Sardesai, Viraj Kulkarni, Candido Alicea, Margherita Rosati, Cristina Bergamaschi, Brunda Ganneru, George N. Pavlakis, Guy R. Pilkington, Antonio Valentin, Barbara K. Felber, Kate E. Broderick, Bhabadeb Chowdhury, and Rachel Kelly Beach
- Subjects
Protein subunit ,Interleukin 5 receptor alpha subunit ,Wild type ,Cell Biology ,Biology ,Biochemistry ,Molecular biology ,law.invention ,Interleukin 10 receptor, alpha subunit ,Plasmid ,law ,Recombinant DNA ,Molecular Biology ,Gene ,Interleukin 12 receptor, beta 1 subunit - Abstract
IL-12 is a 70-kDa heterodimeric cytokine composed of the p35 and p40 subunits. To maximize cytokine production from plasmid DNA, molecular steps controlling IL-12p70 biosynthesis at the posttranscriptional and posttranslational levels were investigated. We show that the combination of RNA/codon-optimized gene sequences and fine-tuning of the relative expression levels of the two subunits within a cell resulted in increased production of the IL-12p70 heterodimer. We found that the p40 subunit plays a critical role in enhancing the stability, intracellular trafficking, and export of the p35 subunit. This posttranslational regulation mediated by the p40 subunit is conserved in mammals. Based on these findings, dual gene expression vectors were generated, producing an optimal ratio of the two subunits, resulting in a ∼1 log increase in human, rhesus, and murine IL-12p70 production compared with vectors expressing the wild type sequences. Such optimized DNA plasmids also produced significantly higher levels of systemic bioactive IL-12 upon in vivo DNA delivery in mice compared with plasmids expressing the wild type sequences. A single therapeutic injection of an optimized murine IL-12 DNA plasmid showed significantly more potent control of tumor development in the B16 melanoma cancer model in mice. Therefore, the improved IL-12p70 DNA vectors have promising potential for in vivo use as molecular vaccine adjuvants and in cancer immunotherapy.
- Published
- 2013
- Full Text
- View/download PDF
50. DNA and virus particle vaccination protects against acquisition and confers control of viremia upon heterologous simian immunodeficiency virus challenge
- Author
-
Agneta von Gegerfelt, David Venzon, Kate E. Broderick, Georgia D. Tomaras, Wensheng Huang, Vainav Patel, Vanessa M. Hirsch, Welkin E. Johnson, William T. Williams, Rama Rao Amara, Viraj Kulkarni, Brandon F. Keele, Yongjun Guan, Jeffrey D. Lifson, Xiaoying Shen, Michael Piatak, Antonio Valentin, Margherita Rosati, George N. Pavlakis, Rashmi Jalah, Harriet L. Robinson, Barbara K. Felber, Julian W. Bess, Niranjan Y. Sardesai, and Candido Alicea
- Subjects
Simian Acquired Immunodeficiency Syndrome ,Heterologous ,Viremia ,Biology ,Antibodies, Viral ,Virus ,DNA vaccination ,Immune system ,medicine ,Animals ,Immunity, Cellular ,Multidisciplinary ,Viral Vaccine ,Rectum ,Virion ,virus diseases ,Viral Vaccines ,Viral Load ,Biological Sciences ,medicine.disease ,Macaca mulatta ,Virology ,Vaccination ,Immunoglobulin G ,DNA, Viral ,Immunology ,Simian Immunodeficiency Virus ,Viral load - Abstract
We have previously shown that macaques vaccinated with DNA vectors expressing SIVmac239 antigens developed potent immune responses able to reduce viremia upon high-dose SIVmac251 challenge. To further improve vaccine-induced immunity and protection, we combined the SIVmac239 DNA vaccine with protein immunization using inactivated SIVmac239 viral particles as protein source. Twenty-six weeks after the last vaccination, the animals were challenged intrarectally at weekly intervals with a titrated dose of the heterologous SIVsmE660. Two of DNA-protein coimmunized macaques did not become infected after 14 challenges, but all controls were infected by 11 challenges. Vaccinated macaques showed modest protection from SIVsmE660 acquisition compared with naïve controls ( P = 0.050; stratified for TRIM5α genotype). Vaccinees had significantly lower peak (1.6 log, P = 0.0048) and chronic phase viremia ( P = 0.044), with 73% of the vaccinees suppressing viral replication to levels below assay detection during the 40-wk follow-up. Vaccine-induced immune responses associated significantly with virus control: binding antibody titers and the presence of rectal IgG to SIVsmE660 Env correlated with delayed SIVsmE660 acquisition; SIV-specific cytotoxic T cells, prechallenge CD4 + effector memory, and postchallenge CD8 + transitional memory cells correlated with control of viremia. Thus, SIVmac239 DNA and protein-based vaccine protocols were able to achieve high, persistent, broad, and effective cellular and humoral immune responses able to delay heterologous SIVsmE660 infection and to provide long-term control of viremia. These studies support a role of DNA and protein-based vaccines for development of an efficacious HIV/AIDS vaccine.
- Published
- 2013
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.