Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
ACS Nano ; 18(18): 11631-11643, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38652829

ABSTRACT

Pharmacological activation of the retinoic acid-inducible gene I (RIG-I) pathway holds promise for increasing tumor immunogenicity and improving the response to immune checkpoint inhibitors (ICIs). However, the potency and clinical efficacy of 5'-triphosphate RNA (3pRNA) agonists of RIG-I are hindered by multiple pharmacological barriers, including poor pharmacokinetics, nuclease degradation, and inefficient delivery to the cytosol where RIG-I is localized. Here, we address these challenges through the design and evaluation of ionizable lipid nanoparticles (LNPs) for the delivery of 3p-modified stem-loop RNAs (SLRs). Packaging of SLRs into LNPs (SLR-LNPs) yielded surface charge-neutral nanoparticles with a size of ∼100 nm that activated RIG-I signaling in vitro and in vivo. SLR-LNPs were safely administered to mice via both intratumoral and intravenous routes, resulting in RIG-I activation in the tumor microenvironment (TME) and the inhibition of tumor growth in mouse models of poorly immunogenic melanoma and breast cancer. Significantly, we found that systemic administration of SLR-LNPs reprogrammed the breast TME to enhance the infiltration of CD8+ and CD4+ T cells with antitumor function, resulting in enhanced response to αPD-1 ICI in an orthotopic EO771 model of triple-negative breast cancer. Therapeutic efficacy was further demonstrated in a metastatic B16.F10 melanoma model, with systemically administered SLR-LNPs significantly reducing lung metastatic burden compared to combined αPD-1 + αCTLA-4 ICI. Collectively, these studies have established SLR-LNPs as a translationally promising immunotherapeutic nanomedicine for potent and selective activation of RIG-I with the potential to enhance response to ICIs and other immunotherapeutic modalities.


Subject(s)
Immunotherapy , Nanoparticles , Animals , Female , Humans , Mice , Cell Line, Tumor , Lipids/chemistry , Mice, Inbred C57BL , Nanoparticles/chemistry , Tumor Microenvironment/drug effects
2.
J Control Release ; 345: 354-370, 2022 05.
Article in English | MEDLINE | ID: mdl-35301055

ABSTRACT

Traditional approaches to cancer vaccines elicit weak CD8+ T cell responses and have largely failed to meet clinical expectations. This is in part due to inefficient antigen cross-presentation, inappropriate selection of adjuvant and its formulation, poor vaccine pharmacokinetics, and/or suboptimal coordination of antigen and adjuvant delivery. Here, we describe a nanoparticle vaccine platform for facile co-loading and dual-delivery of antigens and nucleic acid adjuvants that elicits robust antigen-specific cellular immune responses. The nanovaccine design is based on diblock copolymers comprising a poly(ethylene glycol)-rich first block that is functionalized with reactive moieties for covalent conjugation of antigen via disulfide linkages, and a pH-responsive second block for electrostatic packaging of nucleic acids that also facilitates endosomal escape of associated vaccine cargo to the cytosol. Using polyIC, a clinically-advanced nucleic acid adjuvant, we demonstrated that endosomolytic nanoparticles promoted the cytosolic co-delivery of polyIC and protein antigen, which acted synergistically to enhance antigen cross-presentation, co-stimulatory molecule expression, and cytokine production by dendritic cells. We also found that the vaccine platform increased the accumulation of antigen and polyIC in the local draining lymph nodes. Consequently, dual-delivery of antigen and polyIC with endsomolytic nanoparticles significantly enhanced the magnitude and functionality of CD8+ T cell responses relative to a mixture of antigen and polyIC, resulting in inhibition of tumor growth in a mouse tumor model. Collectively, this work provides a proof-of-principle for a new cancer vaccine platform that strongly augments anti-tumor cellular immunity via cytosolic co-delivery of antigen and nucleic acid adjuvant.


Subject(s)
Cancer Vaccines , Nanoparticles , Adjuvants, Immunologic/pharmacology , Animals , Antigens/chemistry , CD8-Positive T-Lymphocytes , Cytosol , Dendritic Cells , Immunity, Cellular , Mice , Nanoparticles/chemistry , Ovalbumin , RNA
3.
Methods Mol Biol ; 2412: 367-398, 2022.
Article in English | MEDLINE | ID: mdl-34918256

ABSTRACT

Vaccines remain the most effective way to protect populations against deathly infectious diseases. Several disadvantages associated with the traditional vaccines that use whole pathogens have led to the development of alternative strategies including the use of recombinant subunit vaccines. Subunit vaccines are, in general, safer than whole pathogens but tend to be less immunogenic due to the lack of molecular cues that are typically found on whole pathogens. To enhance immunogenicity, the subunit antigen  can be administered with adjuvants that stimulate the innate immune system as a means to steer the quality and magnitude of the adaptive immune response. Novel classes of adjuvants are formulated using particle-based platforms such as virus-like particles, liposomes, and polymeric nanoparticles. These particle-based systems present antigens in ways reminiscent of whole pathogens. Such platforms offer several advantages that include co-delivery of antigen along with innate immune stimulators in a highly immunogenic format. Here we describe our recent efforts to synthesize, characterize, and validate two promising nanoparticle-based delivery systems and demonstrate their potential to induce antigen-specific CD8+ T cell responses, essential in clearing infection with intracellular pathogens, such as viruses and bacteria, and eradicating tumors.


Subject(s)
CD8-Positive T-Lymphocytes , Adjuvants, Immunologic , Antigens , CD8-Positive T-Lymphocytes/immunology , Nanoparticles , Vaccines , Vaccines, Subunit
4.
Front Immunol ; 12: 753472, 2021.
Article in English | MEDLINE | ID: mdl-34899704

ABSTRACT

When compartmentally mislocalized within cells, nucleic acids can be exceptionally immunostimulatory and can even trigger the immune-mediated elimination of cancer. Specifically, the accumulation of double-stranded DNA in the cytosol can efficiently promote antitumor immunity by activating the cGAMP synthase (cGAS) / stimulator of interferon genes (STING) cellular signaling pathway. Targeting this cytosolic DNA sensing pathway with interferon stimulatory DNA (ISD) is therefore an attractive immunotherapeutic strategy for the treatment of cancer. However, the therapeutic activity of ISD is limited by several drug delivery barriers, including susceptibility to deoxyribonuclease degradation, poor cellular uptake, and inefficient cytosolic delivery. Here, we describe the development of a nucleic acid immunotherapeutic, NanoISD, which overcomes critical delivery barriers that limit the activity of ISD and thereby promotes antitumor immunity through the pharmacological activation of cGAS at the forefront of the STING pathway. NanoISD is a nanoparticle formulation that has been engineered to confer deoxyribonuclease resistance, enhance cellular uptake, and promote endosomal escape of ISD into the cytosol, resulting in potent activation of the STING pathway via cGAS. NanoISD mediates the local production of proinflammatory cytokines via STING signaling. Accordingly, the intratumoral administration of NanoISD induces the infiltration of natural killer cells and T lymphocytes into murine tumors. The therapeutic efficacy of NanoISD is demonstrated in preclinical tumor models by attenuated tumor growth, prolonged survival, and an improved response to immune checkpoint blockade therapy.


Subject(s)
DNA , Drug Delivery Systems , Nanoparticles , Nucleotidyltransferases , Animals , Female , Humans , Mice , Colonic Neoplasms/therapy , Cytokines/biosynthesis , Cytokines/genetics , DNA/administration & dosage , DNA/chemical synthesis , DNA/pharmacology , DNA/therapeutic use , Drug Screening Assays, Antitumor , Endosomes/physiology , Immunotherapy/methods , Killer Cells, Natural/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Mammary Neoplasms, Experimental/therapy , Melanoma, Experimental/therapy , Membrane Proteins/physiology , Mice, Inbred C57BL , Nanoparticles/administration & dosage , Nanoparticles/therapeutic use , Neoplasms/immunology , Nucleotidyltransferases/drug effects , Signal Transduction/drug effects , T-Lymphocyte Subsets/immunology , Thionucleotides/pharmacology , Tumor Microenvironment/drug effects
5.
ACS Appl Bio Mater ; 4(2): 1640-1654, 2021 02 15.
Article in English | MEDLINE | ID: mdl-35014512

ABSTRACT

In recent years, there has been an increasing interest in designing delivery systems to enhance the efficacy of RNA-based therapeutics. Here, we have synthesized copolymers comprised of dimethylaminoethyl methacrylate (DMAEMA) or diethylaminoethyl methacrylate (DEAEMA) copolymerized with alkyl methacrylate monomers ranging from 2 to 12 carbons, and developed a high throughput workflow for rapid investigation of their applicability for mRNA delivery. The structure activity relationship revealed that the mRNA encapsulation efficiency is improved by increasing the cationic density and use of shorter alkyl side chains (2-6 carbons). Minimal cytotoxicity was observed when using DEAEMA-co-BMA (EB) polyplexes up to 18 h after dosing, independent of a poly(ethylene glycol) (PEG) first block. The lowest molecular weight polymer (EB10,250) performed best, exhibiting greater transfection than polyethyenimine (PEI) based upon the number of cells transfected and mean intensity. Conventional investigations into the performance of polymeric materials for mRNA delivery is quite tedious, consequently limiting the number of materials and formulation conditions that can be studied. The high throughput approach presented here can accelerate the screening of polymeric systems and paves the way for expanding this generalizable approach to assess various materials for mRNA delivery.


Subject(s)
Gene Transfer Techniques/standards , Genetic Therapy/methods , Polymers/chemistry , RNA, Messenger/metabolism
6.
ACS Nano ; 14(8): 9904-9916, 2020 08 25.
Article in English | MEDLINE | ID: mdl-32701257

ABSTRACT

Cancer vaccines targeting patient-specific neoantigens have emerged as a promising strategy for improving responses to immune checkpoint blockade. However, neoantigenic peptides are poorly immunogenic and inept at stimulating CD8+ T cell responses, motivating a need for new vaccine technologies that enhance their immunogenicity. The stimulator of interferon genes (STING) pathway is an endogenous mechanism by which the innate immune system generates an immunological context for priming and mobilizing neoantigen-specific T cells. Owing to this critical role in tumor immune surveillance, a synthetic cancer nanovaccine platform (nanoSTING-vax) was developed that mimics immunogenic cancer cells in its capacity to efficiently promote co-delivery of peptide antigens and the STING agonist, cGAMP. The co-loading of cGAMP and peptides into pH-responsive, endosomolytic polymersomes promoted the coordinated delivery of both cGAMP and peptide antigens to the cytosol, thereby eliciting inflammatory cytokine production, co-stimulatory marker expression, and antigen cross-presentation. Consequently, nanoSTING-vax significantly enhanced CD8+ T cell responses to a range of peptide antigens. Therapeutic immunization with nanoSTING-vax, in combination with immune checkpoint blockade, inhibited tumor growth in multiple murine tumor models, even leading to complete tumor rejection and generation of durable antitumor immune memory. Collectively, this work establishes nanoSTING-vax as a versatile platform for enhancing immune responses to neoantigen-targeted cancer vaccines.


Subject(s)
Cancer Vaccines , Neoplasms , Animals , Antigens, Neoplasm , Humans , Interferons , Mice , Neoplasms/drug therapy , Peptides
SELECTION OF CITATIONS
SEARCH DETAIL
...