Lipid nanoparticle-based mRNA candidates elicit potent T cell responses.

The induction of a potent T cell response is essential for successful tumor immunotherapy and protection against many infectious diseases. In the past few years, mRNA vaccines have emerged as potent immune activators and inducers of a robust T cell immune response. The recent approval of the Moderna and the Pfizer/BioNTech vaccines based on lipid nanoparticles (LNP) encapsulating antigen-encoding mRNA has revolutionized the field of vaccines. The advantages of LNPs are their ease of design and formulation resulting in potent, effective, and safe vaccines. However, there is still plenty of room for improvement with respect to LNP efficacy, for instance, by optimizing the lipid composition and tuning LNP for specific purposes. mRNA delivery is known to be strongly dependent on the lipid composition of LNPs and the efficiency is mainly determined by the ionizable lipids. Besides that, cholesterol and helper lipids also play important roles in mRNA transfection potency. Here, a panel of LNP formulations was studied by keeping the ionizable lipids constant, replacing cholesterol with β-sitosterol, and changing the fusogenic helper lipid DOPE content. We studied the ability of this LNP library to induce antigen presentation and T cell proliferation to identify superior LNP candidates eliciting potent T cell immune responses. We hypothesize that using β-sitosterol and increasing DOPE content would boost the mRNA transfection on immune cells and result in enhanced immune responses. Transfection of immortal immune cell lines and bone marrow dendritic cells (BMDCs) with LNPs was studied. Delivery of mRNA coding for the model antigen ovalbumin (OVA-mRNA) to BMDCs with a number of LNP formulations, resulted in a high level of activation, as evidenced by the upregulation of the co-stimulatory receptors (CD40 and CD86) and IL-12 in BMDCs. The enhancement of BMDC activation and T cell proliferation induced by the introduction of β-sitosterol and fusogenic DOPE lipids were cell dependent. Four LNP formulations (C12-200-cho-10%DOPE, C12-200-sito-10%DOPE, cKK-E12-cho-10%DOPE and cKK-E12-sito-30%DOPE) were identified that induced robust T cell proliferation and enhanced IFN-γ, TNF-α, IL-2 expression. These results demonstrate that T cell proliferation is strongly dependent on LNP composition and promising LNP-mRNA vaccine formulations were identified.

[1]  T. Sharp,et al.  Anionic Lipid Nanoparticles Preferentially Deliver mRNA to the Hepatic Reticuloendothelial System , 2022, Advanced materials.

[2]  Yizhou Dong,et al.  Lipid Nanoparticle-mRNA Formulations for Therapeutic Applications. , 2021, Accounts of chemical research.

[3]  S. D. De Smedt,et al.  The dawn of mRNA vaccines: The COVID-19 case , 2021, Journal of Controlled Release.

[4]  Siddharth Patel,et al.  Deconvoluting Lipid Nanoparticle Structure for Messenger RNA Delivery , 2020, Nano letters.

[5]  D. Weissman,et al.  A Multi-Targeting, Nucleoside-Modified mRNA Influenza Virus Vaccine Provides Broad Protection in Mice. , 2020, Molecular therapy : the journal of the American Society of Gene Therapy.

[6]  Martha E. Zeeman,et al.  Human chimeric antigen receptor macrophages for cancer immunotherapy , 2020, Nature Biotechnology.

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

[8]  Siddharth Patel,et al.  Naturally-occurring cholesterol analogues in lipid nanoparticles induce polymorphic shape and enhance intracellular delivery of mRNA , 2020, Nature Communications.

[9]  Michael J. Mitchell,et al.  Ionizable Lipid Nanoparticle Mediated mRNA Delivery for Human CAR T Cell Engineering. , 2020, Nano letters.

[10]  M. Manoharan,et al.  The Onpattro story and the clinical translation of nanomedicines containing nucleic acid-based drugs , 2019, Nature Nanotechnology.

[11]  Robert Langer,et al.  Delivery of mRNA vaccines with heterocyclic lipids increases anti-tumor efficacy by STING-mediated immune cell activation , 2019, Nature Biotechnology.

[12]  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.

[13]  D. Weissman,et al.  Characterization of HIV-1 Nucleoside-Modified mRNA Vaccines in Rabbits and Rhesus Macaques , 2019, Molecular therapy. Nucleic acids.

[14]  G. Ebel,et al.  An mRNA Vaccine Protects Mice against Multiple Tick-Transmitted Flavivirus Infections , 2018, Cell Reports.

[15]  Florian Krammer,et al.  Nucleoside-modified mRNA immunization elicits influenza virus hemagglutinin stalk-specific antibodies , 2018, Nature Communications.

[16]  J. Bluestone,et al.  Revisiting IL-2: Biology and therapeutic prospects , 2018, Science Immunology.

[17]  Örn Almarsson,et al.  A Novel Amino Lipid Series for mRNA Delivery: Improved Endosomal Escape and Sustained Pharmacology and Safety in Non-human Primates. , 2018, Molecular therapy : the journal of the American Society of Gene Therapy.

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

[19]  Kimberly J. Hassett,et al.  Preclinical and Clinical Demonstration of Immunogenicity by mRNA Vaccines against H10N8 and H7N9 Influenza Viruses , 2017, Molecular therapy : the journal of the American Society of Gene Therapy.

[20]  Justin M. Richner,et al.  Modified mRNA Vaccines Protect against Zika Virus Infection , 2017, Cell.

[21]  Robert Langer,et al.  Lipid Nanoparticle Assisted mRNA Delivery for Potent Cancer Immunotherapy. , 2017, Nano letters.

[22]  D. Weissman,et al.  Administration of nucleoside-modified mRNA encoding broadly neutralizing antibody protects humanized mice from HIV-1 challenge , 2017, Nature Communications.

[23]  D. Weissman,et al.  Zika virus protection by a single low dose nucleoside modified mRNA vaccination , 2017, Nature.

[24]  J. Rosenecker,et al.  Nanotechnologies in delivery of mRNA therapeutics using nonviral vector-based delivery systems , 2017, Gene Therapy.

[25]  S. Kaech,et al.  The multifaceted role of CD4+ T cells in CD8+ T cell memory , 2016, Nature Reviews Immunology.

[26]  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.

[27]  T. Schlake,et al.  Sequence-engineered mRNA Without Chemical Nucleoside Modifications Enables an Effective Protein Therapy in Large Animals , 2015, Molecular therapy : the journal of the American Society of Gene Therapy.

[28]  D. O’Sullivan,et al.  Expanding the role of metabolism in T cells , 2015, Science.

[29]  Özlem Türeci,et al.  mRNA-based therapeutics — developing a new class of drugs , 2014, Nature Reviews Drug Discovery.

[30]  Daniel G. Anderson,et al.  Non-viral vectors for gene-based therapy , 2014, Nature Reviews Genetics.

[31]  Robert Langer,et al.  Lipopeptide nanoparticles for potent and selective siRNA delivery in rodents and nonhuman primates , 2014, Proceedings of the National Academy of Sciences.

[32]  Michael J. Bevan,et al.  CD8+ T Cells: Foot Soldiers of the Immune System , 2011, Immunity.

[33]  K. G. Rajeev,et al.  Rational design of cationic lipids for siRNA delivery , 2010, Nature Biotechnology.

[34]  M. Bevan,et al.  Interleukin-2 and inflammation induce distinct transcriptional programs that promote the differentiation of effector cytolytic T cells. , 2010, Immunity.

[35]  Kathryn A. Whitehead,et al.  Lipid-like materials for low-dose, in vivo gene silencing , 2010, Proceedings of the National Academy of Sciences.

[36]  Hiroki Kato,et al.  Incorporation of pseudouridine into mRNA yields superior nonimmunogenic vector with increased translational capacity and biological stability. , 2008, Molecular therapy : the journal of the American Society of Gene Therapy.

[37]  V. Appay,et al.  CD8+ T cell efficacy in vaccination and disease , 2008, Nature Medicine.

[38]  J. Sprent,et al.  Selective Stimulation of T Cell Subsets with Antibody-Cytokine Immune Complexes , 2006, Science.

[39]  G. Trinchieri,et al.  Interleukin-12 and the regulation of innate resistance and adaptive immunity , 2003, Nature Reviews Immunology.

[40]  T Salditt,et al.  An inverted hexagonal phase of cationic liposome-DNA complexes related to DNA release and delivery. , 1998, Science.