Polymer nanoparticles deliver mRNA to the lung for mucosal vaccination

An inhalable platform for messenger RNA (mRNA) therapeutics would enable minimally invasive and lung-targeted delivery for a host of pulmonary diseases. Development of lung-targeted mRNA therapeutics has been limited by poor transfection efficiency and risk of vehicle-induced pathology. Here, we report an inhalable polymer-based vehicle for delivery of therapeutic mRNAs to the lung. We optimized biodegradable poly(amine-co-ester) (PACE) polyplexes for mRNA delivery using end-group modifications and polyethylene glycol. These polyplexes achieved high transfection of mRNA throughout the lung, particularly in epithelial and antigen-presenting cells. We applied this technology to develop a mucosal vaccine for severe acute respiratory syndrome coronavirus 2 and found that intranasal vaccination with spike protein–encoding mRNA polyplexes induced potent cellular and humoral adaptive immunity and protected susceptible mice from lethal viral challenge. Together, these results demonstrate the translational potential of PACE polyplexes for therapeutic delivery of mRNA to the lungs. Description Inhaled polymer nanoparticles achieve high mRNA expression in the lung and induce protective immunity against SARS-CoV-2. Editor’s summary The ability to efficiently deliver mRNA to the lung would have applications for vaccine development, gene therapy, and more. Here, Suberi et al. showed that such mRNA delivery can be accomplished by encapsulating mRNAs of interest within optimized poly(amine-co-ester) polyplexes. Polyplex-delivered mRNAs were efficiently translated into protein in the lungs of mice with limited evidence of toxicity. This platform was successfully applied as an intranasal SARS-CoV-2 vaccine, eliciting robust immune responses that conferred protection against subsequent viral challenge. These results highlight the potential of this delivery system for vaccine applications and beyond. —Courtney Malo

[1]  T. Liou,et al.  Inhaled mRNA therapy for treatment of cystic fibrosis: Interim results of a randomized, double-blind, placebo-controlled phase 1/2 clinical study. , 2023, Journal of cystic fibrosis : official journal of the European Cystic Fibrosis Society.

[2]  Daniel G. Anderson,et al.  Combinatorial design of nanoparticles for pulmonary mRNA delivery and genome editing , 2023, Nature Biotechnology.

[3]  C. Zurla,et al.  Species-agnostic polymeric formulations for inhalable messenger RNA delivery to the lung , 2022, Nature Materials.

[4]  B. Haynes,et al.  Nebulized mRNA‐Encoded Antibodies Protect Hamsters from SARS‐CoV‐2 Infection , 2022, Advanced science.

[5]  A. Iwasaki,et al.  Unadjuvanted intranasal spike vaccine elicits protective mucosal immunity against sarbecoviruses , 2022, Science.

[6]  Jeonghwan Kim,et al.  Rapid Generation of Circulating and Mucosal Decoy Human ACE2 using mRNA Nanotherapeutics for the Potential Treatment of SARS‐CoV‐2 , 2022, Advanced science.

[7]  Sixuan Li,et al.  Payload distribution and capacity of mRNA lipid nanoparticles , 2022, Nature Communications.

[8]  E. Topol,et al.  Operation Nasal Vaccine—Lightning speed to counter COVID-19 , 2022, Science Immunology.

[9]  Aaron J. Johnson,et al.  Respiratory mucosal immunity against SARS-CoV-2 following mRNA vaccination , 2022, Science Immunology.

[10]  R. Rubin COVID-19 Boosters This Fall to Include Omicron Antigen, but Questions Remain About Its Value. , 2022, JAMA.

[11]  B. Fadeel,et al.  Understanding the Role and Impact of Poly (Ethylene Glycol) (PEG) on Nanoparticle Formulation: Implications for COVID-19 Vaccines , 2022, Frontiers in Bioengineering and Biotechnology.

[12]  Qiaobing Xu,et al.  Lung-selective mRNA delivery of synthetic lipid nanoparticles for the treatment of pulmonary lymphangioleiomyomatosis , 2022, Proceedings of the National Academy of Sciences.

[13]  William F. Fadel,et al.  Waning 2-Dose and 3-Dose Effectiveness of mRNA Vaccines Against COVID-19–Associated Emergency Department and Urgent Care Encounters and Hospitalizations Among Adults During Periods of Delta and Omicron Variant Predominance — VISION Network, 10 States, August 2021–January 2022 , 2022, MMWR. Morbidity and mortality weekly report.

[14]  Matthew S. Miller,et al.  Respiratory mucosal delivery of next-generation COVID-19 vaccine provides robust protection against both ancestral and variant strains of SARS-CoV-2 , 2022, Cell.

[15]  J. Rosenecker,et al.  Nanotechnologies in Delivery of DNA and mRNA Vaccines to the Nasal and Pulmonary Mucosa , 2022, Nanomaterials.

[16]  L. Bekker,et al.  Effectiveness of BNT162b2 Vaccine against Omicron Variant in South Africa , 2021, The New England journal of medicine.

[17]  A. Iwasaki,et al.  Intranasal priming induces local lung-resident B cell populations that secrete protective mucosal antiviral IgA , 2021, Science Immunology.

[18]  A. Iwasaki,et al.  A stem-loop RNA RIG-I agonist protects against acute and chronic SARS-CoV-2 infection in mice , 2021, The Journal of experimental medicine.

[19]  B. Igyártó,et al.  The mRNA-LNP platform's lipid nanoparticle component used in preclinical vaccine studies is highly inflammatory , 2021, iScience.

[20]  Chenglin Deng,et al.  Intranasal delivery of replicating mRNA encoding neutralizing antibody against SARS-CoV-2 infection in mice , 2021, Signal Transduction and Targeted Therapy.

[21]  Rongkai Yan,et al.  The Roles of Tissue-Resident Memory T Cells in Lung Diseases , 2021, Frontiers in Immunology.

[22]  A. Iwasaki,et al.  Adaptive immune determinants of viral clearance and protection in mouse models of SARS-CoV-2 , 2021, Science Immunology.

[23]  K. Bruxvoort,et al.  Effectiveness of mRNA-1273 against delta, mu, and other emerging variants of SARS-CoV-2: test negative case-control study , 2021, BMJ.

[24]  James E. Dahlman,et al.  Optimization of lipid nanoparticles for the delivery of nebulized therapeutic mRNA to the lungs , 2021, Nature Biomedical Engineering.

[25]  S. Anzick,et al.  Intranasal ChAdOx1 nCoV-19/AZD1222 vaccination reduces viral shedding after SARS-CoV-2 D614G challenge in preclinical models , 2021, Science Translational Medicine.

[26]  R. Myers,et al.  Effectiveness of Covid-19 Vaccines against the B.1.617.2 (Delta) Variant , 2021, The New England journal of medicine.

[27]  M. C. Muenker,et al.  Impact of circulating SARS-CoV-2 variants on mRNA vaccine-induced immunity , 2021, Nature.

[28]  D. Meyerholz,et al.  Protection of K18-hACE2 mice and ferrets against SARS-CoV-2 challenge by a single-dose mucosal immunization with a parainfluenza virus 5–based COVID-19 vaccine , 2021, Science Advances.

[29]  M. Diamond,et al.  SARS-CoV-2 mRNA vaccines induce persistent human germinal centre responses , 2021, Nature.

[30]  Qun Wang,et al.  Effects of polyethylene glycol on the surface of nanoparticles for targeted drug delivery. , 2021, Nanoscale.

[31]  W. Saltzman,et al.  PEGylation of poly(amine-co-ester) polyplexes for tunable gene delivery. , 2021, Biomaterials.

[32]  W. Hinrichs,et al.  Inhaled vaccine delivery in the combat against respiratory viruses: a 2021 overview of recent developments and implications for COVID-19 , 2021, Expert review of vaccines.

[33]  D. Irvine,et al.  Exploiting albumin as a mucosal vaccine chaperone for robust generation of lung-resident memory T cells , 2021, Science Immunology.

[34]  B. Igyártó,et al.  The mRNA-LNP platform’s lipid nanoparticle component used in preclinical vaccine studies is highly inflammatory , 2021, bioRxiv.

[35]  C. Zurla,et al.  Treatment of influenza and SARS-CoV-2 infections via mRNA-encoded Cas13a in rodents , 2021, Nature Biotechnology.

[36]  Charles Y. Tan,et al.  BNT162b vaccines protect rhesus macaques from SARS-CoV-2 , 2021, Nature.

[37]  John T. Wilson,et al.  Engineering Vaccines for Tissue‐Resident Memory T Cells , 2021, Advanced therapeutics.

[38]  J. Rosenecker,et al.  In Vitro Investigations on Optimizing and Nebulization of IVT-mRNA Formulations for Potential Pulmonary-Based Alpha-1-Antitrypsin Deficiency Treatment , 2021, Pharmaceutics.

[39]  Jean Martínez,et al.  Design of PEGylated Three Ligands Silica Nanoparticles for Multi-Receptor Targeting , 2021, Nanomaterials.

[40]  D. Weissman,et al.  SARS-CoV-2 mRNA Vaccines Foster Potent Antigen-Specific Germinal Center Responses Associated with Neutralizing Antibody Generation , 2020, Immunity.

[41]  Hairui Zhang,et al.  Aerosolizable Lipid Nanoparticles for Pulmonary Delivery of mRNA through Design of Experiments , 2020, Pharmaceutics.

[42]  Michael Y. T. Chow,et al.  Inhaled RNA Therapy: From Promise to Reality , 2020, Trends in Pharmacological Sciences.

[43]  Lisa E. Gralinski,et al.  A Single-Dose Intranasal ChAd Vaccine Protects Upper and Lower Respiratory Tracts against SARS-CoV-2 , 2020, Cell.

[44]  James E. Dahlman,et al.  Treating cystic fibrosis with mRNA and CRISPR. , 2020, Human gene therapy.

[45]  Shamus P. Keeler,et al.  SARS-CoV-2 infection of hACE2 transgenic mice causes severe lung inflammation and impaired function , 2020, Nature Immunology.

[46]  A. Iwasaki,et al.  Mouse model of SARS-CoV-2 reveals inflammatory role of type I interferon signaling , 2020, The Journal of experimental medicine.

[47]  Rebecca J. Loomis,et al.  SARS-CoV-2 mRNA Vaccine Design Enabled by Prototype Pathogen Preparedness , 2020, Nature.

[48]  W. Saltzman,et al.  Polymeric vehicles for nucleic acid delivery. , 2020, Advanced drug delivery reviews.

[49]  H. Chan,et al.  Inhalation delivery technology for genome-editing of respiratory diseases , 2020, Advanced Drug Delivery Reviews.

[50]  Eric Song,et al.  Mouse model of SARS-CoV-2 reveals inflammatory role of type I interferon signaling , 2020, bioRxiv.

[51]  Qiang Cheng,et al.  Selective ORgan Targeting (SORT) nanoparticles for tissue specific mRNA delivery and CRISPR/Cas gene editing , 2020, Nature Nanotechnology.

[52]  Yucai Wang,et al.  Protein Binding Affinity of Polymeric Nanoparticles as a Direct Indicator of Their Pharmacokinetics. , 2020, ACS nano.

[53]  Fan Yang,et al.  Quantitating Endosomal Escape of a Library of Polymers for mRNA Delivery. , 2020, Nano letters.

[54]  I. Sahu,et al.  Recent Developments in mRNA-Based Protein Supplementation Therapy to Target Lung Diseases. , 2019, Molecular therapy : the journal of the American Society of Gene Therapy.

[55]  Daniel G Anderson,et al.  Delivering the Messenger: Advances in Technologies for Therapeutic mRNA Delivery. , 2019, Molecular therapy : the journal of the American Society of Gene Therapy.

[56]  Joseph Rosenecker,et al.  Self-assembled peptide–poloxamine nanoparticles enable in vitro and in vivo genome restoration for cystic fibrosis , 2019, Nature Nanotechnology.

[57]  Robert Langer,et al.  Inhaled Nanoformulated mRNA Polyplexes for Protein Production in Lung Epithelium , 2019, Advanced materials.

[58]  N. Pedemonte,et al.  Chemically modified hCFTR mRNAs recuperate lung function in a mouse model of cystic fibrosis , 2018, Scientific Reports.

[59]  T. Randall,et al.  The establishment of resident memory B cells in the lung requires local antigen encounter , 2018, Nature Immunology.

[60]  Adam Williams,et al.  Determination of T Follicular Helper Cell Fate by Dendritic Cells , 2018, Front. Immunol..

[61]  Kaitlyn Sadtler,et al.  Optimization of a Degradable Polymer-Lipid Nanoparticle for Potent Systemic Delivery of mRNA to the Lung Endothelium and Immune Cells. , 2018, Nano letters.

[62]  W. Saltzman,et al.  Tunability of Biodegradable Poly(amine- co-ester) Polymers for Customized Nucleic Acid Delivery and Other Biomedical Applications. , 2018, Biomacromolecules.

[63]  Gaurav Sahay,et al.  Lipid Nanoparticle-Delivered Chemically Modified mRNA Restores Chloride Secretion in Cystic Fibrosis. , 2018, Molecular therapy : the journal of the American Society of Gene Therapy.

[64]  Robert Langer,et al.  Ionizable Amino‐Polyesters Synthesized via Ring Opening Polymerization of Tertiary Amino‐Alcohols for Tissue Selective mRNA Delivery , 2018, Advanced materials.

[65]  Gregg A. Duncan,et al.  PEGylated enhanced cell penetrating peptide nanoparticles for lung gene therapy , 2018, Journal of controlled release : official journal of the Controlled Release Society.

[66]  D. Weissman,et al.  mRNA vaccines — a new era in vaccinology , 2018, Nature Reviews Drug Discovery.

[67]  Samuel K Lai,et al.  PEGylation for enhancing nanoparticle diffusion in mucus☆ , 2017, Advanced drug delivery reviews.

[68]  C. Rudolph,et al.  A Single Methylene Group in Oligoalkylamine-Based Cationic Polymers and Lipids Promotes Enhanced mRNA Delivery. , 2016, Angewandte Chemie.

[69]  S. Beer-Hammer,et al.  mRNA-Mediated Gene Supplementation of Toll-Like Receptors as Treatment Strategy for Asthma In Vivo , 2016, PloS one.

[70]  D. Weissman,et al.  Expression kinetics of nucleoside-modified mRNA delivered in lipid nanoparticles to mice by various routes. , 2015, Journal of controlled release : official journal of the Controlled Release Society.

[71]  Daniel G. Anderson,et al.  Optimization of Lipid Nanoparticle Formulations for mRNA Delivery in Vivo with Fractional Factorial and Definitive Screening Designs. , 2015, Nano letters.

[72]  Xiaoqun Gong,et al.  Impact of Surface Polyethylene Glycol (PEG) Density on Biodegradable Nanoparticle Transport in Mucus ex Vivo and Distribution in Vivo. , 2015, ACS nano.

[73]  Guadalupe Ortiz-Muñoz,et al.  Non-invasive Intratracheal Instillation in Mice. , 2015, Bio-protocol.

[74]  Qing Jiang,et al.  Micelles of enzymatically synthesized PEG-poly(amine-co-ester) block copolymers as pH-responsive nanocarriers for docetaxel delivery. , 2014, Colloids and surfaces. B, Biointerfaces.

[75]  Christopher E. Nelson,et al.  Balancing cationic and hydrophobic content of PEGylated siRNA polyplexes enhances endosome escape, stability, blood circulation time, and bioactivity in vivo. , 2013, ACS nano.

[76]  D. Irvine,et al.  Generation of Effector Memory T Cell–Based Mucosal and Systemic Immunity with Pulmonary Nanoparticle Vaccination , 2013, Science Translational Medicine.

[77]  T. Moninger,et al.  Adenoviral gene transfer corrects the ion transport defect in the sinus epithelia of a porcine CF model. , 2013, Molecular therapy : the journal of the American Society of Gene Therapy.

[78]  J. Rosenecker,et al.  Expression of therapeutic proteins after delivery of chemically modified mRNA in mice , 2011, Nature Biotechnology.

[79]  Guillermo Repetto,et al.  Neutral red uptake assay for the estimation of cell viability/cytotoxicity , 2008, Nature Protocols.

[80]  Robert T. Chen,et al.  Use of the inactivated intranasal influenza vaccine and the risk of Bell's palsy in Switzerland. , 2004, The New England journal of medicine.

[81]  Jung-Ki Park,et al.  Effect of polyethylene glycol on gene delivery of polyethylenimine. , 2003, Biological & pharmaceutical bulletin.

[82]  Kenneth A Howard,et al.  Importance of lateral and steric stabilization of polyelectrolyte gene delivery vectors for extended systemic circulation. , 2002, Molecular therapy : the journal of the American Society of Gene Therapy.

[83]  M. Ogris,et al.  PEGylated DNA/transferrin–PEI complexes: reduced interaction with blood components, extended circulation in blood and potential for systemic gene delivery , 1999, Gene Therapy.

[84]  Zhaozhong Jiang,et al.  Biodegradable poly(amine-co-ester) terpolymers for targeted gene delivery. , 2011, Nature materials.

[85]  Yen Cu,et al.  Controlled surface modification with poly(ethylene)glycol enhances diffusion of PLGA nanoparticles in human cervical mucus. , 2009, Molecular pharmaceutics.

[86]  A. W. E. E. K. L. Y. J. O U R N A L D E V O T E D T O T H E A D V A N C E,et al.  S C I E N C E , 2022 .