Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Norbert Pardi is active.

Publication


Featured researches published by Norbert Pardi.


Nature | 2017

Zika virus protection by a single low-dose nucleoside-modified mRNA vaccination

Norbert Pardi; Michael J. Hogan; Rebecca S. Pelc; Hiromi Muramatsu; Hanne Andersen; Christina R. DeMaso; Kimberly A. Dowd; Laura L. Sutherland; Richard M. Scearce; Robert Parks; Wendeline Wagner; Alex Granados; Jack Greenhouse; Michelle Walker; Elinor Willis; Jae-Sung Yu; Charles E. McGee; Gregory D. Sempowski; Barbara L. Mui; Ying K. Tam; Yan-Jang Huang; Dana L. Vanlandingham; Veronica M. Holmes; Harikrishnan Balachandran; Sujata Sahu; Michelle A. Lifton; Stephen Higgs; Scott E. Hensley; Thomas D. Madden; Michael J. Hope

Zika virus (ZIKV) has recently emerged as a pandemic associated with severe neuropathology in newborns and adults. There are no ZIKV-specific treatments or preventatives. Therefore, the development of a safe and effective vaccine is a high priority. Messenger RNA (mRNA) has emerged as a versatile and highly effective platform to deliver vaccine antigens and therapeutic proteins. Here we demonstrate that a single low-dose intradermal immunization with lipid-nanoparticle-encapsulated nucleoside-modified mRNA (mRNA–LNP) encoding the pre-membrane and envelope glycoproteins of a strain from the ZIKV outbreak in 2013 elicited potent and durable neutralizing antibody responses in mice and non-human primates. Immunization with 30 μg of nucleoside-modified ZIKV mRNA–LNP protected mice against ZIKV challenges at 2 weeks or 5 months after vaccination, and a single dose of 50 μg was sufficient to protect non-human primates against a challenge at 5 weeks after vaccination. These data demonstrate that nucleoside-modified mRNA–LNP elicits rapid and durable protective immunity and therefore represents a new and promising vaccine candidate for the global fight against ZIKV.


Journal of Controlled Release | 2015

Expression kinetics of nucleoside-modified mRNA delivered in lipid nanoparticles to mice by various routes.

Norbert Pardi; Steven Tuyishime; Hiromi Muramatsu; Katalin Karikó; Barbara L. Mui; Ying K. Tam; Thomas D. Madden; Michael J. Hope; Drew Weissman

In recent years, in vitro transcribed messenger RNA (mRNA) has emerged as a potential therapeutic platform. To fulfill its promise, effective delivery of mRNA to specific cell types and tissues needs to be achieved. Lipid nanoparticles (LNPs) are efficient carriers for short-interfering RNAs and have entered clinical trials. However, little is known about the potential of LNPs to deliver mRNA. Here, we generated mRNA-LNPs by incorporating HPLC purified, 1-methylpseudouridine-containing mRNA comprising codon-optimized firefly luciferase into stable LNPs. Mice were injected with 0.005-0.250mg/kg doses of mRNA-LNPs by 6 different routes and high levels of protein translation could be measured using in vivo imaging. Subcutaneous, intramuscular and intradermal injection of the LNP-encapsulated mRNA translated locally at the site of injection for up to 10days. For several days, high levels of protein production could be achieved in the lung from the intratracheal administration of mRNA. Intravenous and intraperitoneal and to a lesser extent intramuscular and intratracheal deliveries led to trafficking of mRNA-LNPs systemically resulting in active translation of the mRNA in the liver for 1-4 days. Our results demonstrate that LNPs are appropriate carriers for mRNA in vivo and have the potential to become valuable tools for delivering mRNA encoding therapeutic proteins.


Nature Communications | 2017

Administration of nucleoside-modified mRNA encoding broadly neutralizing antibody protects humanized mice from HIV-1 challenge

Norbert Pardi; Anthony Secreto; Xiaochuan Shan; Fotini Debonera; Joshua Glover; Yanjie Yi; Hiromi Muramatsu; Houping Ni; Barbara L. Mui; Ying K. Tam; Farida Shaheen; Ronald G. Collman; Katalin Karikó; Gwenn Danet-Desnoyers; Thomas D. Madden; Michael J. Hope; Drew Weissman

Monoclonal antibodies are one of the fastest growing classes of pharmaceutical products, however, their potential is limited by the high cost of development and manufacturing. Here we present a safe and cost-effective platform for in vivo expression of therapeutic antibodies using nucleoside-modified mRNA. To demonstrate feasibility and protective efficacy, nucleoside-modified mRNAs encoding the light and heavy chains of the broadly neutralizing anti-HIV-1 antibody VRC01 are generated and encapsulated into lipid nanoparticles. Systemic administration of 1.4 mg kg−1 of mRNA into mice results in ∼170 μg ml−1 VRC01 antibody concentrations in the plasma 24 h post injection. Weekly injections of 1 mg kg−1 of mRNA into immunodeficient mice maintain trough VRC01 levels above 40 μg ml−1. Most importantly, the translated antibody from a single injection of VRC01 mRNA protects humanized mice from intravenous HIV-1 challenge, demonstrating that nucleoside-modified mRNA represents a viable delivery platform for passive immunotherapy against HIV-1 with expansion to a variety of diseases.


Methods of Molecular Biology | 2013

In Vitro Transcription of Long RNA Containing Modified Nucleosides

Norbert Pardi; Hiromi Muramatsu; Drew Weissman; Katalin Karikó

The in vitro synthesis of long RNA can be accomplished using phage RNA polymerase and template DNA. However, the in vitro synthesized RNA, unlike those transcribed in vivo in cells, lacks nucleoside modifications. Introducing modified nucleosides into in vitro transcripts is important because they reduce the potential of RNA to activate RNA sensors and translation of such nucleoside-modified RNA is increased in cell lines, primary cells, and after in vivo delivery. Here, we describe the in vitro synthesis of nucleoside-modified RNA with enhanced translational capacity and reduced ability to activate immune sensors.


Methods of Molecular Biology | 2013

HPLC Purification of In Vitro Transcribed Long RNA

Drew Weissman; Norbert Pardi; Hiro Muramatsu; Katalin Karikó

In vitro transcription of DNA with phage RNA polymerases is currently the most efficient method to produce long sequence-specific RNA. While the reaction can yield large quantities of RNA, it contains impurities due to various unwanted activities of the polymerases. Here, we described an easily performed HPLC purification that removes multiple contaminants from in vitro transcribed RNA and is scalable. The purified RNA is translated at much greater levels, especially in primary cells and in vivo. HPLC purification of RNA containing modified nucleosides that suppress RNA-mediated activation of innate immune sensors leads to a non-immunogenic RNA with superior translational capacity.


Archive | 2017

Nucleoside Modified mRNA Vaccines for Infectious Diseases

Norbert Pardi; Drew Weissman

In recent years, numerous studies have demonstrated the outstanding abilities of mRNA to elicit potent immune responses against pathogens, making it a viable new platform for vaccine development (reviewed in Weissman, Expert Rev Vaccines 14:265-281, 2015; Sahin et al., Nat Rev Drug Discov 13:759-780, 2014). The incorporation of modified nucleosides in mRNA has many advantages and is currently undergoing a renaissance in the field of therapeutic protein delivery. Its use in a vaccine against infectious diseases has only begun to be described, but offers advantages for the generation of potent and long-lived antibody responses. FPLC purification and substitution of modified nucleosides in the mRNA make it non-inflammatory and highly translatable (Kariko et al., Immunity 23:165-175, 2005; Kariko et al., Mol Ther 16:1833-1840, 2008; Kariko et al., Nucleic Acids Research 39:e142, 2011) that are crucial features for therapeutic relevance. Formulation of the mRNA in lipid nanoparticles (LNPs) protects it from degradation enabling high levels of protein production for extended periods of time (Pardi et al., J Control Release, 2015). Here, we describe a simple vaccination method using LNP-encapsulated 1-methylpseudouridine-containing FPLC purified mRNA in mice. Furthermore, we describe the evaluation of antigen-specific T and B cell responses elicited by this vaccine format.


Journal of Experimental Medicine | 2018

Nucleoside-modified mRNA vaccines induce potent T follicular helper and germinal center B cell responses.

Norbert Pardi; Michael J. Hogan; Martin S. Naradikian; Kaela Parkhouse; Derek W. Cain; Letitia Jones; M. Anthony Moody; Hans Verkerke; Arpita Myles; Elinor Willis; Celia C. LaBranche; David C. Montefiori; Jenna Lobby; Kevin O. Saunders; Hua-Xin Liao; Bette Korber; Laura L. Sutherland; Richard M. Scearce; Peter Hraber; István Tombácz; Hiromi Muramatsu; Houping Ni; Daniel A Balikov; Charles Li; Barbara L. Mui; Ying K. Tam; Florian Krammer; Katalin Karikó; Patricia Polacino; Laurence C. Eisenlohr

T follicular helper (Tfh) cells are required to develop germinal center (GC) responses and drive immunoglobulin class switch, affinity maturation, and long-term B cell memory. In this study, we characterize a recently developed vaccine platform, nucleoside-modified, purified mRNA encapsulated in lipid nanoparticles (mRNA-LNPs), that induces high levels of Tfh and GC B cells. Intradermal vaccination with nucleoside-modified mRNA-LNPs encoding various viral surface antigens elicited polyfunctional, antigen-specific, CD4+ T cell responses and potent neutralizing antibody responses in mice and nonhuman primates. Importantly, the strong antigen-specific Tfh cell response and high numbers of GC B cells and plasma cells were associated with long-lived and high-affinity neutralizing antibodies and durable protection. Comparative studies demonstrated that nucleoside-modified mRNA-LNP vaccines outperformed adjuvanted protein and inactivated virus vaccines and pathogen infection. The incorporation of noninflammatory, modified nucleosides in the mRNA is required for the production of large amounts of antigen and for robust immune responses.


Vaccine | 2018

New Kids on the Block: RNA-Based Influenza Virus Vaccines

Francesco Berlanda Scorza; Norbert Pardi

RNA-based immunization strategies have emerged as promising alternatives to conventional vaccine approaches. A substantial body of published work demonstrates that RNA vaccines can elicit potent, protective immune responses against various pathogens. Consonant with its huge impact on public health, influenza virus is one of the best studied targets of RNA vaccine research. Currently licensed influenza vaccines show variable levels of protection against seasonal influenza virus strains but are inadequate against drifted and pandemic viruses. In recent years, several types of RNA vaccines demonstrated efficacy against influenza virus infections in preclinical models. Additionally, comparative studies demonstrated the superiority of some RNA vaccines over the currently used inactivated influenza virus vaccines in animal models. Based on these promising preclinical results, clinical trials have been initiated and should provide valuable information about the translatability of the impressive preclinical data to humans. This review briefly describes RNA-based vaccination strategies, summarizes published preclinical and clinical data, highlights the roadblocks that need to be overcome for clinical applications, discusses the landscape of industrial development, and shares the authors’ personal perspectives about the future of RNA-based influenza virus vaccines.


Virology | 2018

Increased surface expression of HIV-1 envelope is associated with improved antibody response in vaccinia prime/protein boost immunization

Michael J. Hogan; Angela Conde-Motter; Andrea P. O. Jordan; Lifei Yang; Brad Cleveland; Wenjin Guo; Josephine Romano; Houping Ni; Norbert Pardi; Celia C. LaBranche; David C. Montefiori; Shiu-Lok Hu; James A. Hoxie; Drew Weissman

HIV-1 envelope (Env)-based vaccines have so far largely failed to induce antibodies that prevent HIV-1 infection. One factor proposed to limit the immunogenicity of cell-associated Env is its low level of expression on the cell surface, restricting accessibility to antibodies. Using a vaccinia prime/protein boost protocol in mice, we explored the immunologic effects of mutations in the Env cytoplasmic tail (CT) that increased surface expression, including partial truncation and ablation of a tyrosine-dependent endocytosis motif. After vaccinia primes, CT-modified Envs induced up to 7-fold higher gp120-specific IgG, and after gp120 protein boosts, they elicited up to 16-fold greater Tier-1 HIV-1 neutralizing antibody titers, although results were variable between isolates. These data indicate that the immunogenicity of HIV-1 Env in a prime/boost vaccine can be enhanced in a strain-dependent manner by CT mutations that increase Env surface expression, thus highlighting the importance of the prime in this vaccine format.


Nature Communications | 2018

Nucleoside-modified mRNA immunization elicits influenza virus hemagglutinin stalk-specific antibodies

Norbert Pardi; Kaela Parkhouse; Ericka Kirkpatrick; Meagan McMahon; Seth J. Zost; Barbara L. Mui; Ying K. Tam; Katalin Karikó; Christopher J. Barbosa; Thomas D. Madden; Michael J. Hope; Florian Krammer; Scott E. Hensley; Drew Weissman

Currently available influenza virus vaccines have inadequate effectiveness and are reformulated annually due to viral antigenic drift. Thus, development of a vaccine that confers long-term protective immunity against antigenically distant influenza virus strains is urgently needed. The highly conserved influenza virus hemagglutinin (HA) stalk represents one of the potential targets of broadly protective/universal influenza virus vaccines. Here, we evaluate a potent broadly protective influenza virus vaccine candidate that uses nucleoside-modified and purified mRNA encoding full-length influenza virus HA formulated in lipid nanoparticles (LNPs). We demonstrate that immunization with HA mRNA-LNPs induces antibody responses against the HA stalk domain of influenza virus in mice, rabbits, and ferrets. The HA stalk-specific antibody response is associated with protection from homologous, heterologous, and heterosubtypic influenza virus infection in mice.The highly conserved influenza virus hemagglutinin (HA) stalk represents a potential target for a broadly protective vaccine. Here, the authors show that immunization with nucleoside-modified mRNA encoding full-length HA formulated in lipid nanoparticles elicits HA stalk-specific antibodies and protects from heterosubtypic virus infection.

Collaboration


Dive into the Norbert Pardi's collaboration.

Top Co-Authors

Avatar

Drew Weissman

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Katalin Karikó

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Ying K. Tam

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Hiromi Muramatsu

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Michael J. Hope

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Thomas D. Madden

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Houping Ni

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Michael J. Hogan

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge