WO2023121960A2 - Compositions and methods for delivery and production of antibodies using an rna-containing nanoparticle platform - Google Patents

Compositions and methods for delivery and production of antibodies using an rna-containing nanoparticle platform Download PDF

Info

Publication number
WO2023121960A2
WO2023121960A2 PCT/US2022/053132 US2022053132W WO2023121960A2 WO 2023121960 A2 WO2023121960 A2 WO 2023121960A2 US 2022053132 W US2022053132 W US 2022053132W WO 2023121960 A2 WO2023121960 A2 WO 2023121960A2
Authority
WO
WIPO (PCT)
Prior art keywords
antibody
spp
mrna
encoding
disclosed
Prior art date
Application number
PCT/US2022/053132
Other languages
French (fr)
Other versions
WO2023121960A3 (en
Inventor
Ross Kedl
Jay Hesselberth
Ira FLEMING
Benjamin WILLETT
Original Assignee
The Regents Of The University Of Colorado, A Body Corporate
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by The Regents Of The University Of Colorado, A Body Corporate filed Critical The Regents Of The University Of Colorado, A Body Corporate
Publication of WO2023121960A2 publication Critical patent/WO2023121960A2/en
Publication of WO2023121960A3 publication Critical patent/WO2023121960A3/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/2818Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against CD28 or CD152
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2878Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the NGF-receptor/TNF-receptor superfamily, e.g. CD27, CD30, CD40, CD95
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2887Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against CD20
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/32Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against translation products of oncogenes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/53DNA (RNA) vaccination
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/75Agonist effect on antigen
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding

Definitions

  • Embodiments of the instant disclosure relate to novel compositions and methods for delivering one or more mRNA encoding a target monoclonal antibody or fragment thereof, using an mRNA encapsulation platform technology.
  • mRNA encapsulation platform compositions disclosed herein can include at least one mRNA encoding a heavy chain and at least one mRN A encoding a light chain of a target or selected monoclonal antibody encapsulated into a lipid nanoparticle (LNP) where the encoded monoclonal antibody is capable of inducing at least one pathway.
  • LNP lipid nanoparticle
  • Therapeutically targeted monoclonal antibodies are one of the fastest growing classes of therapeutic molecules. Immunotherapy can be used in the treatment of a variety of health conditions with improved specificity and outcoming including specifically targeted conditions (e.g., chronic inflammatory diseases, a solid tumor or other targetable cancer, a specified infectious diseases).
  • specifically targeted conditions e.g., chronic inflammatory diseases, a solid tumor or other targetable cancer, a specified infectious diseases.
  • current manufacturing and purification processes cause limitations in the production capacity of therapeutic antibodies and delivery, leading to an increase in cost and reduced efficiency.
  • toxicity has been associated with the high doses of intravenous antibody delivery needed to achieve a high and sustained therapeutic plasma concentration to treat a targeted condition. Therefore, new approaches to antibody production, use, targeting and delivery are needed in the field to improve specific and non- specific responses to these agents.
  • compositions disclosed herein can include one or more complex of at least one mRNA transcript encoding at least one heavy chain and at least one light chain of an antibody to a target molecule encapsulated in a nanoparticle where the antibody induces a cellular immune response, targets a pathway and/or targets a microorganism.
  • the nanoparticle can be any nanoparticle able to deliver an mRNA encoding an antibody.
  • the nanoparticle can be a lipid nanoparticle (LNP) or another suitable nanoparticle.
  • compositions disclosed herein can include complexes including at least one mRNA encoding at least one heavy chain and at least one mRNA encoding at least one light chain of an agonistic and/or antagonistic monoclonal antibody to a pathway, targeted molecule or cell in a nanoparticle (e.g. LNP) for inducing an immune response in a subject.
  • compositions disclosed herein can include complexes including mRNA encoding one heavy chain and at least one mRNA encoding a light chain of an agonistic or antagonistic monoclonal antibody in a nanoparticle (e.g. LNP) and at least one other agent.
  • compositions disclosed herein can include complexes having at least two mRNA transcripts that encode for at least one heavy chain of an agonistic antibody and encode for at least one light chain of the agonistic antibody.
  • compositions including complexes or nanoparticles disclosed herein can include at least two mRNA transcripts encoding an antibody such as a monoclonal antibody or fragment thereof encapsulated therein.
  • mRNA transcripts disclosed herein can include self-replicating RNA transcripts encoding at least one light chain or heavy chain of a selected antibody.
  • complexes or nanoparticles disclosed herein can form part of a pharmaceutical composition that further includes at least one pharmaceutically acceptable carrier or excipient.
  • pharmaceutical compositions containing complexes such as nanoparticles disclosed herein can be administered to a subject and the nanoparticle can erode at a predetermined time to expose the molecules for encoding the agonistic and/or antagonistic antibody or fragment thereof at a site of interest, at the cellular target or systemically to a subject.
  • a full-length agonistic or antagonistic antibody can be formed in vivo after administering a pharmaceutical composition containing the antibody-encoding nanoparticles disclosed herein to the subject.
  • one or more agonistic or antagonistic antibody formed from mRNA transcripts disclosed herein can induce an immune response in a subject to a health condition to treat or prevent or reduce the onset of the health condition.
  • complexes disclosed herein can further include one or more vaccines.
  • the one or more vaccines can include one or more mRNA transcripts encoding an antigen of use against a microbe or a microbial infection and/or for treating or preventing formation of a tumor.
  • improved cellular immune responses are induced in a subject receiving a vaccine disclosed herein compared to mRNA vaccines administered to a subject in the absence of administering a complex of encapsulated mRNA transcripts disclosed herein.
  • agonistic or antagonistic antibodies directed to inducing an immune response in a subject can be formed in vivo from encoding mRNAs after administering a composition disclosed herein to the subject.
  • the subject can have or is suspected of having a condition including, but not limited to, an infection, exposed to a pathogenic organism, at risk of developing an infection, having cancer, or suspected of developing cancer or other health condition in need of a therapy for inducing an immune response to treat the health condition, reduce the risk of onset or prevent the health condition.
  • a subject in need of treatment with a composition containing mRNA-containing nanoparticles disclosed herein can have or is suspected of developing cancer, a microbial infection or exposed to a pathogenic organism (e.g. virus, bacteria, fungus, prion, protozoa), an autoimmune disorder, an allograft rejection, hematopoietic disorders or other blood or blood cell disorders or any combination thereof.
  • a pathogenic organism e.g. virus, bacteria, fungus, prion, protozoa
  • an autoimmune disorder e.g. virus, bacteria, fungus, prion, protozoa
  • an allograft rejection hematopoietic disorders or other blood or blood cell disorders or any combination thereof.
  • a full-length agonistic or antagonistic antibody formed from mRNA transcripts-containing nanoparticles disclosed herein can be a monoclonal antibody. In some embodiments, a full-length agonistic or antagonistic antibody formed from mRNA transcripts-containing nanoparticles disclosed herein can be an immune checkpoint inhibitor.
  • a full-length agonistic or antagonistic antibody formed from mRNA transcripts-containing nanoparticles disclosed herein can be one or more of an anti-HER2 antibody, an anti-EGFR antibody, an anti-VEGRF antibody, an anti-VEGF antibody, an anti-CD-20 antibody, an anti-CD-22 antibody, an anti- CD30 antibody, an anti-CD33 antibody, an anti-CD38 antibody, an anti-CD52 antibody, an anti-CTLA-4 antibody, an anti-PD-1 antibody, an anti-PD-Ll antibody, an anti-RANKL antibody, an anti-GD2 antibody, an anti-PDGFR antibody, an anti-SLAMF7 antibody, or any combination thereof.
  • a full-length or fragment thereof of an antagonistic antibody translated from mRNA transcripts-containing nanoparticles disclosed herein can be one or more of an anti- IL-1, an anti- IL-17, an anti- IL-22, an anti- IL-23, an anti- IFNy, and an anti- TNF ⁇ or a combination thereof when introduced to a subject by encapsulated formulations blocks cellular immune responses in the subject.
  • a full-length or fragment thereof of an agonistic antibody translated from mRNA transcripts-containing nanoparticles disclosed herein can be one or more of an anti- CD137 (41BB), an anti- CD134 (0X40), an anti- CD30, an anti-CD40, an anti- CD27 or any other TNFR/L family member antibody or a combination thereof when introduced to a subject by encapsulated formulations as described herein induces or enhances immune responses (e.g., cellular immune responses) in the subject.
  • a full-length agonistic antibody formed from mRNA transcripts encapsulated in nanoparticle complexes disclosed herein can encode a full-length antibody capable of targeting one or more viruses or viral proteins derived therefrom.
  • compositions disclosed herein for including with an anti-viral vaccine can include any pathogenic virus capable of infecting a human, other mammal, reptile, or bird.
  • a virus targeted by mRNA encoding viral-derived antigen vaccine(s) or a full-length anti-viral antibody vaccine formed from encapsulating nanoparticle complexes disclosed herein can include any pathogenic vims.
  • a full-length agonistic antibody formed from mRNA transcripts encapsulated in nanoparticle complexes disclosed herein can encode a full-length antibody capable of targeting one or more bacteria or bacterial proteins derived therefrom.
  • compositions disclosed herein containing nanoparticle complexes disclosed herein harboring mRNA encoding anti-bacterial vaccines can include any pathogenic bacteria capable of infecting a human, other mammal, reptile, or bird.
  • a single nanoparticle can contain a single encoding mRNA or multiple mRNAs.
  • a single nanoparticle can include an mRNA encoding a heavy chain, light chain, or combination thereof of a targeted antibodv or fragment thereof.
  • the antibody is a monoclonal antibody. In other embodiments, the antibody is a monoclonal antibody directed to induce or inhibit a pathway.
  • multiple mRNA molecules can be encapsulated in single nanoparticles encoding at least one antagonist and agonist heavy chain and light chain to generate at least two monoclonal antibodies where each monoclonal antibody has antagonist or agonist activity for inducing an immune response in the subject.
  • compositions of mixed nanoparticles are contemplated where each nanoparticle in the mix of nanoparticles encapsulates at least one mRNA encoding a heavy chain or light chain of an antibody or fragment thereof.
  • nanoparticle complexes and compositions thereof can be delivered to a subject to treat or prevent a health condition where the encoded agent (e.g., antibody or antigen or fragment thereof) can be expressed in vivo in the subject directly to a site or systemically to the subject.
  • the encoded agent e.g., antibody or antigen or fragment thereof
  • compositions including at least one nanoparticle encapsulating at least one mRNA contemplated herein.
  • pharmaceutical compositions disclosed herein can include any of the nanoparticle complexes disclosed herein and at least one pharmaceutically acceptable excipient or carrier.
  • pharmaceutical compositions disclosed herein can further include one or more additional agents for facilitating delivery or stability of the nanoparticle complexes disclosed herein.
  • other agents for improving an immune response can include one or more adjuvants or other immune-inducing agents separate from the nanoparticle complexes within the composition or as part of the one or more nanoparticle complexes such as encapsulated within the nanoparticles.
  • lipid nanoparticles encapsulating one or more mRNA encoding at least a heavy and/or light chain of any contemplated cellular immune enhancing antibody disclosed herein can be delivered in the same or different composition as a vaccine in order to enhance cellular immunity to treat or prevent a health condition in a subject.
  • lipid nanoparticles disclosed herein can be delivered separate from a vaccine such as before, after or simultaneously by the same or different mode of administration as one or more vaccines.
  • lipid nanoparticles disclosed herein can contain mRNAs for encoding a heavy and/or light chain of a selected antibody or antibodies and further include a vaccine encapsulated in the same nanoparticles.
  • mixtures of nanoparticle complexes can be combined in a single composition for a mixed nanoparticle composition delivery to the subject.
  • nanoparticles encapsulating one or more mRNA encoding at least one heavy and/or light chain of at least one of an antagonist or agonist to a cellular immune induction pathway can replace the need for an adjuvant or be used in addition to an adjuvant to induce an immune response in a subj ect.
  • a health condition to be treated or prevented by the methods and compositions disclosed herein can include cancer.
  • cancer can include a solid tumor.
  • a health condition to be treated or prevented by the methods and compositions disclosed herein can be an infection due to a pathogenic organism.
  • a pathogenic microorganism can include a virus, a bacteria, fungus, protozoan or prion.
  • compositions disclosed herein to a subject contemplated herein can be any suitable mode.
  • composition can be administered as a vaccine by subcutaneous, inhalation, intraocular, intranasal administration, or other mode of administration.
  • methods of administering compositions can include localized administration directly to a tumor site in the subject.
  • kits for storing or delivering compositions or components including, but not limited to, nanoparticles encapsulating at least one mRNA encoding an antigen, whole organism, antibody or fragment thereof for use in practicing any of the methods disclosed herein.
  • kits disclosed herein can include nanoparticle complexes, compositions, pharmaceutical compositions, vaccines, or a combination thereof.
  • kits disclosed herein can further include at least one container.
  • Figs. 1A-1B illustrate graphs demonstrating T cell responses (Fig. 1A) and IgG and neutralization titers (Fig. IB) in a mammalian (e.g., human) PBMCs in response to a SARS- CoV2 mRNA vaccine in accordance with certain embodiments of the present disclosure and demonstrate reduced T cell responses while inducing a good antibody response.
  • FIG. 2 illustrates a schematic of mRNA in vitro transcription and lipid nano particle encapsulation in accordance with certain embodiments of the present disclosure.
  • FIG. 3 illustrates a graph demonstrating an antigen specific CD8+ T cell response in mice vaccinated with lipid encapsulated antibody-encoding mRNA transcripts in accordance with certain embodiments of the present disclosure.
  • FIGs. 4A-4B illustrate images demonstrating increased antigen-specific CD8+ T cell response in wild-type mice injected alone or with increasing mRNA transcript-containing- LNPs as evaluated by dual tetramer staining of cells isolated from peripheral blood 7 days post vaccination (4A) and by quantification of CD44hi (activated) dual tetramer+ T cells (4B) in accordance with certain embodiments of the present disclosure.
  • Fig. 5 illustrates a graph demonstrating antitumor activity post vaccination with an anti-tumor vaccine plus a checkpoint blockade disclosed herein (e.g., anti-PDl antibody) in mice injected with B 16-ova tumor cells followed by injection of a combined vaccine treatment (e.g. polyIC/aCD40/2DG) with and without concomitant administration of a blocking antibody in accordance with certain embodiments of the present disclosure.
  • a checkpoint blockade disclosed herein e.g., anti-PDl antibody
  • a combined vaccine treatment e.g. polyIC/aCD40/2DG
  • FIGs. 6A-6D illustrate images demonstrating increased antigen-specific CD8+ T cell response in wild-type mice injected alone or with mRNA transcript-containing-LNPs encoding a cellular immune stimulating antibody and its activity evaluated by dual tetramer staining of cells isolated from peripheral blood several days post vaccination (6A) and by quantification of CD44hi (activated) dual tetramer+ T cells (6B) illustrating an enhanced specific T-cell response.
  • 6C and 6D illustrate further specificity and quantitation of the response in the presence of the nanocapsules encapsulating the test mRNA encoded antibody in accordance with certain embodiments of the present disclosure.
  • “individual”, “subject”, “host”, and “patient” can be used interchangeably herein and refer to any mammalian or other contemplated subject for whom diagnosis, treatment, prophylaxis or therapy is desired, for example, humans, pets, livestock, horses, birds, reptiles, or other animals.
  • treat can refer to reversing, ameliorating, or inhibiting onset or inhibiting progression of a health condition or disease or a symptom of the health condition or disease.
  • polynucleotide can include deoxyribonucleic acid (DNA) or ribonucleic acid (RNA), messenger RNA (mRNA), oligonucleotides, and the like.
  • vector refers to a nucleic acid component used to introduce polynucleotides into a cell having regulatory elements to provide expression of the heterologous nucleic acids in the cell.
  • Vectors include but are not limited to plasmid, minicircles, yeast, and/or viral genomes. In some alternatives, the vectors are plasmid, minicircles, or viral genomes. In some alternatives, the vector is a viral vector. In some embodiments, the viral vector is a lentivirus. In some alternatives, the vector is a lentiviral vector. In some embodiments, the vector is a foamy viral vector, adenoviral vectors, retroviral vectors or lentiviral vectors.
  • encoding can refer to the inherent property of specific sequences of nucleotides in a polynucleotide, such as a gene, a cDNA, or an mRNA, to serve as templates for synthesis of other polymers and macromolecules in biological processes having either a defined sequence of nucleotides (e.g., RNA, rRNA, tRNA and mRNA) or a defined sequence of amino acids and the biological properties resulting therefrom.
  • a defined sequence of nucleotides e.g., RNA, rRNA, tRNA and mRNA
  • antibody can refer to an immunoglobulin molecule, which specifically binds to a target antigen or epitope of a target antigen, receptor or other molecule.
  • Antibodies can be intact immunoglobulins derived from natural sources or from recombinant sources, or encoded by mRNA molecules contemplated herein and can be immunoreactive portions of intact immunoglobulins.
  • antibodies can also be generated from mRNA transcripts of one or more heavy and/or light chain encoding one or more full-length antibody or fragment thereon contained in a nanoparticle (e g., any biocompatible lipid nanoparticle).
  • Antibodies are typically tetramers of immunoglobulin molecules.
  • the antibodies in the present disclosure can exist in a variety of forms including, but not limited to polyclonal antibodies, monoclonal antibodies, Fv, Fab and F(ab)2, single chain antibodies, humanized antibodies, nanoantibodies, camelids, and the like.
  • immune response can refer to a process involving the activation and/or induction of an effector function in, by way of non-limiting examples, a T-cell, B-cell, natural killer (NK) cell, and/or an antigen-presenting cell (APC) response for humoral and/or cellular immunity.
  • An immune response can include, but is not limited to, any detectable antigen-specific activation and/or induction of a helper T cell or cytotoxic T cell activity or response, production of antibodies, antigen presenting cell activity or infiltration, macrophage activity or infiltration, neutrophil activity or infiltration, and the like.
  • An immune response can refer to the combination of cellular immune induction and specific activation against an antigen as contemplated herein.
  • adjuvant can refer to any molecule able to enhance an antigen-specific adaptive immune response or able to enhance an immune response when administered to a subject such as a non-specific immune response or cellular or humoral response.
  • nanoparticle encapsulated antibodies disclosed herein can behave as an adjuvant to enhance cellular immunity in a subject in need thereof.
  • autologous refers to cells derived from the patient. “Allogenic” refers to cells derived from the same species but having a different genotype. “Syngeneic” refers to cells from a genetically identical source, such as a twin, hence immunologically compatible or so closely related that transplantation does not provoke an immune response.
  • compositions disclosed herein can include one or more nanoparticle complex of at least one mRNA transcript encoding at least one heavy chain and at least one light chain of an antibody to a target molecule encapsulated in a nanoparticle where the antibody induces a cellular immune response and/or targets a microorganism in a subject.
  • nanoparticles can be any nanoparticle able to encapsulate and deliver an mRNA encoding an antibody or fragment thereof.
  • nanoparticles can include a lipid nanoparticle (LNP) or other suitable nanoparticle capable of encapsulating mRNA encoding an antibody or fragment thereof, reducing degradation of the mRNA, and permitting translation of the antibody or fragment thereof at the site and/or timing of interest.
  • LNP lipid nanoparticle
  • complexes, compositions, and methods disclosed herein are designed to encapsulate, stabilize, transport, and generate antibodies or antibody fragments at a site of interest.
  • complexes disclosed herein include, but are not limited to, mRNA transcripts encapsulated in a nanoparticle including, but not limited to, a transcript encoding a heavy chain and a transcript encoding a light chain for generating an antibody of interest for example full-length antibodies, antigen binding fragments of full-length antibodies, Fab fragments, single chain antibodies (scFv), diabodies, triabodies, mini bodies, nanobodies, single-domain antibodies, camelids, or any combination thereof.
  • the antibody or antibody fragment thereof is an agonistic or antagonistic antibody directed to a cellular immune regulating pathway or component thereof.
  • complexes disclosed herein can include nanoparticles encapsulating one or more mRNA transcripts.
  • the nanoparticles can include lipid nanoparticles (LNPs; ionizable lipid nanoparticles for example).
  • LNPs contemplated herein can encapsulate at least two or more mRNA transcripts encoding at least one targeted antibody.
  • nanoparticles e.g., LNPs
  • mRNA e.g., 1, 2, 3, 4, 5, 6, 7, 8 or more
  • nanoparticle complexes disclosed herein can include nanoparticles encapsulating one, two, three, four or more mRNA transcripts encoding at least one heavy chain and at least one light chain of an antibody.
  • complexes disclosed herein can include nanoparticles encapsulating two mRNA transcripts encoding at least one heavy chain and at least one light chain of a single antibody and can further be mixed with complexes encoding at least one heavy chain and at least one light chain of another antibody directed to the same or a different target.
  • the encoded antibody is an antagonistic antibody of a targeted cellular response pathway.
  • the encoded antibody is an agonistic antibody of a targeted cellular response pathway.
  • nanoparticle complexes disclosed herein include mRNA transcripts for encoding a heavy and a light chain of an agonistic antibody and/or a heavy and a light chain of an antagonistic antibody to a targeted cellular immune pathway.
  • a target can be a cellular immune promoting target or a cellular immune inhibiting target where the antibody binds to the target and induces cellular immunity in the subject.
  • mRNAs encoding heavy and light chain sequences of an agonistic or antagonistic antibody targeting an immunologically relevant pathway can be encapsulated in ionizable lipid nanoparticle (LNP), the components of which in part, facilitate the fusion of the LNP to an endosomal membrane, allowing access of nanoparticle constituents (the mRNA) to flow out of the nanoparticle into cytoplasm of the cell in order to form, in vivo, the agonistic or antagonistic antibody of interest.
  • LNP ionizable lipid nanoparticle
  • encapsulated mRNA transcripts disclosed herein can encode for a full-length polypeptide or peptide fragment thereof.
  • mRNA transcripts disclosed herein can encode for an antibody.
  • mRNA transcripts disclosed herein can encode for an antibody where the antibody can be a chimeric immunoglobulin.
  • mRNA transcripts disclosed herein can encode for an antibody where the antibody can be partially or fully humanized antibody.
  • mRNA transcripts disclosed herein can encode for an antibody where the antibody is a human immunoglobulin or other mammal.
  • mRNA transcripts disclosed herein can encode for a full-length antibody or an antibody fragment. In certain embodiments, mRNA transcripts disclosed herein can encode for a single chain immunoglobulin. In other embodiments, mRNA transcripts disclosed herein can encode for at least one IgGl antibody, an IgG2 antibody, an IgG3 antibody, an IgG4 antibody, or any combination thereof. In other embodiments, mRNA transcripts disclosed herein can encode for a heavy chain of an antibody. In some embodiments, mRNA transcripts disclosed herein can encode for a light chain of an antibody.
  • mRNA transcripts of nanoparticle complexes disclosed herein can include a heavy chain to light chain (H:L) ratio in these complexes in a 1 : 1; a 1.5: 1 ; a 2: 1; a 2.5: 1 ; 3: 1; 3.5: 1 ; 4: 1; 4.5: 1 or other suitable ratio of H:L.
  • the ratio of mRNA transcript encoding a heavy chain antibody is about two times the number of mRNA transcripts encoding the light chain of a target antibody or target antibodies (e.g., an anti-CD40 antibody or other cellular immune pathway inducing antibody).
  • compositions disclosed herein can include nanoparticle complexes including at least one mRNA encoding at least one heavy chain and at least one mRNA encoding at least one light chain of an agonistic or antagonistic monoclonal antibody in a single nanoparticle for inducing a cellular immune response in a subject.
  • compositions disclosed herein can include nanoparticle complexes including mRNA encoding one heavy chain and at least one mRNA encoding a light chain of an agonistic or antagonistic monoclonal antibody in one or more LNPs.
  • compositions disclosed herein can include complexes having at least two mRNA transcripts encoding a heavy chain of an agonistic antibody, and at least one of the at least two mRNA transcripts can encode for a light chain of the agonistic antibody for use in inducing cellular immunity.
  • compositions including complexes disclosed herein can include at least two mRNA transcripts encoding an antibody or fragment thereof encapsulated in an LNP.
  • nanoparticles disclosed herein can further include one or more vaccine for treating or preventing onset of a health condition.
  • lipid nanoparticles encapsulating one or more mRNA encoding at least one heavy and/or light chain of any contemplated cellular immune enhancing antibody disclosed herein can be delivered in the same or different composition as a vaccine in order to enhance cellular immunity to treat or prevent a health condition in a subject.
  • lipid nanoparticles disclosed herein can be delivered separate from a vaccine such as before, after, or simultaneously by the same or different mode of administration as one or more vaccines.
  • lipid nanoparticles disclosed herein can contain mRNAs for encoding a heavy and/or light chain of one or more selected antibody or antibodies and further include a vaccine encapsulated in the same nanoparticles.
  • mixtures of nanoparticle complexes can be combined in a single composition for a mixed nanoparticle composition delivery to the subject.
  • nanoparticles encapsulating one or more mRNA encoding at least one heavy and/or light chain of at least one of an antagonist or agonist to a cellular immune induction pathway can replace the need for an adjuvant or be used in addition to an adjuvant to induce an immune response in a subj ect.
  • a standard adjuvant such as alum or other agent can be mixed in or supplied separately from nanoparticle complexes disclosed herein and optionally, can further include a vaccine.
  • complexes including at least two mRNA transcripts for encoding a full-length antibody capable of inducing a cellular immune response can further be formulated with complexes including at least one mRNA that encodes an antigen (e.g. an mRNA vaccine against a pathogen or tumor) in the same or separate complexes in the same or separate nanoparticles.
  • nanoparticles can be mixed and administered in a single dose, or multiple doses.
  • the dose of nanoparticle complexes can be about 1.0 ⁇ g/kg to about 300 ⁇ g/kg, or about 5.0 ⁇ g/kg to about 250 ⁇ g/kg or about 10.0 ⁇ g/kg to about 200 ⁇ g/kg, or about 20.0 ⁇ g/kg to about 150 ⁇ g/kg.
  • nanoparticles encoding antibodies contemplated herein and nanoparticles encoding an antigen of use as an mRNA vaccine can be administered separately, at the same time, staggered or at different times depending on the effect.
  • nanoparticles encoding antibodies contemplated herein and nanoparticles encoding an antigen of use as an mRNA vaccine can be part of a single nanoparticle complex or a mixture of nanoparticles harboring different mRNA transcripts encoding antibodies to the same or different cellular-immune enhancing targets.
  • one or more than one dose of nanop article complexes encoding antibodies disclosed herein can be administered to the subject.
  • administration can include more than once a day, daily, every other day, bi- weekly, weekly, bi-monthly, monthly, every other month, together with or at the time of administering a vaccine (or before or after) or other suitable dosing regimen.
  • mRNA encoding the at least one mRNA transcript encoding a heavy chain and a light chain of the antibody can further include one or more of incorporation of 1 methyl-pseudouridine (ImUp); at least one SerineUCG (SerUCG) codon substitution having at least one ImUp; and an mRNA sequence that remains unstructured between about the last 40 to 55 nucleotides of the 5’UTR and about the first 20 to 40 nucleotides (nts) of the coding sequence, followed by increased secondary structure thereafter for improved stability and translation of the mRNA transcript.
  • ImUp 1 methyl-pseudouridine
  • SerineUCG SerineUCG
  • the one or more mRNA transcript can include an mRNA sequence that remains unstructured between about the last 47 nucleotides of the 5’UTR and about the first 30 nucleotides of the coding sequence, followed by increased secondary' structure thereafter for improved stability and translation of the mRNA transcript.
  • modifications to the mRNA transcripts disclosed herein can reduce degradation of the transcripts and improve translation of a target antibody.
  • complexes disclosed herein can form part of a pharmaceutical composition further including at least one pharmaceutically acceptable carrier.
  • pharmaceutical compositions containing nanoparticle complexes contemplated herein can be administered to a subject and the nanoparticle can erode at a predetermined time to allow exposure of the mRNA transcripts for encoding the agonistic or antagonistic antibody or fragment thereof at a site of interest or systemically to a subject.
  • a full-length agonistic or antagonistic antibody can be formed in vivo after administering an mRNA transcript containing nanoparticle complex-containing composition disclosed herein to the subject.
  • one or more agonistic or antagonistic antibody formed from mRNA transcripts disclosed herein can induce a cellular immune response in the subject to treat a health condition in the subject or enhance treatment of the subject or prevent or reduce onset of a condition in the subject.
  • nanoparticle complexes disclosed herein can further include one or more mRNA vaccines or other vaccines.
  • the one or more mRNA vaccines can include an mRNA vaccine having one or more mRNA transcripts encoding an antigen of use against a microbe or a microbial infection and/or for treating or preventing formation of a tumor.
  • improved cellular immune responses are induced in a subject receiving nanoparticle complexes encoding an antagonistic or agonistic antibody disclosed herein compared to vaccines or mRNA vaccines administered to a subject in the absence of administering a nanoparticle complex of encapsulated mRNA transcripts encoding an antagonistic or agonistic antibody disclosed herein directed to a cellular immune-related system (e.g., anti-CD40 antibody or anti-CTLA4 equivalent thereof).
  • a cellular immune-related system e.g., anti-CD40 antibody or anti-CTLA4 equivalent thereof.
  • mRNA encoding an antigen as part of an mRNA vaccine-containing nanoparticle can further include one or more of an alpha globin 5'-UTR, an amino-terminal enhancer of split (AES) motif, and a mitochondrial 12S RNA element.
  • mRNA encoding an antigen as part of an mRNA vaccine-containing nanoparticle includes at least 2 of an alpha globin 5'- UTR, an amino-terminal enhancer of split (AES) motif, and a mitochondrial 12S RNA element for improved stabil ity and translation of the mRNA as well as improved translation.
  • these mRNA vaccine-containing nanoparticles further include at least one mRNA encoding a light chain and/or at least one mRNA encoding a heavy chain in the same or different nanoparticle complexes.
  • the nanoparticles include lipid nanoparticles such as ionizing lipid nanoparticles or other nanoparticles capable of encapsulating mRNA for delivery and cytosolic dispersion permitting formation of fully functional antibodies.
  • agonistic or antagonistic antibodies directed to inducing a cellular immune response can be formed in vivo after administering a composition herein to a subject in need thereof, where a subject in need thereof can have or is suspected of having a condition including, but not limited to, an infection, exposed to a pathogenic organism, at risk of an infection, cancer or other health condition in need of a therapy for inducing an immune response to treat the condition.
  • a subject in need of treatment with a composition containing complexes disclosed herein can have or is suspected of developing at least one of cancer, a microbial infection or exposed to a pathogenic microbe (e.g. virus, bacteria, fungus, prion, protozoa), an autoimmune disorder, an allograft rejection, hematopoietic or hematologic disorders or any combination thereof.
  • a pathogenic microbe e.g. virus, bacteria, fungus, prion, protozoa
  • a full-length agonistic or antagonistic antibody formed from mRNA transcripts encapsulated in nanoparticle complexes disclosed herein can be a monoclonal antibody.
  • a full-length agonistic or antagonistic antibody formed from mRNA transcripts encapsulated in nanoparticle complexes disclosed herein can be an immune checkpoint inhibitor antibody.
  • an immune checkpoint inhibitor antibody can include an antibody encoded by mRNA transcripts disclosed herein that reduces or blocks proteins referred to as checkpoints that are made by certain immune system cells, such as T cells, NK cells and some cancer cells.
  • checkpoints assist in a healthy individual by preventing the immune responses from being too strong and managing the immune response.
  • checkpoints can reduce or interfere with T cells from killing cancer cells or fighting infection.
  • T cells can kill cancer cells better and can fight infection more effectively.
  • antibodies encoded by mRNA transcripts disclosed herein can include antibodies that bind to and inhibit checkpoint molecules from inhibiting or interfering with cellular immune responses.
  • examples of checkpoint proteins found on T cells or cancer cells can include, but are not limited to, PD-1, PD-L1, CTLA-4, B7-1 and B7-2.
  • immune checkpoint inhibitors can be used to treat cancer
  • a full-length agonistic or antagonistic antibody formed from mRNA transcripts encapsulated in nanoparticle complexes disclosed herein can include, but are not limited to, one or more of an anti-HER2 antibody, an anti-EGFR antibody, an anti- VEGRF antibody, an anti-VEGF antibody, an anti-CD-20 antibody, an anti-CD-22 antibody, an anti-CD30 antibody, an anti-CD33 antibody, an anti-CD38 antibody, an anti-CD52 antibody, an anti-CTLA-4 antibody, an anti-PD-1 antibody, an anti-PD-Ll antibody, an anti- 137- 1, an anti-B7-2, an anti-RANKL antibody, an anti-GD2 antibody, an anti-PDGFR antibody, an anti-SLAMF7 antibody, or any combination thereof.
  • a full-length or fragment thereof of an antagonistic antibody translated from mRNA transcript complexes disclosed herein can include, but is not limited to, one or more of an anti-IL-1, an anti-IL-17, an anti-IL-22, an anti-IL-23, an anti-IFNy, and an anti-TNFa or a combination thereof that when introduced to a subject by encapsulated mRNA transcript-containing formulations disclosed herein blocks cellular immune responses in the subject.
  • a full-length or fragment thereof of an agonistic antibody translated from mRNA transcript complexes disclosed herein can be one or more of an anti-CD137 (4 IBB), an anti- CD134 (0X40), an anti-CD30, an anti-CD40 antibody, and an anti- CD27 or other TNFR/L family member or a combination thereof when introduced to a subject by encapsulated formulations induces or enhances cellular immune responses in the subject.
  • a full-length agonistic antibody formed from mRNA transcripts of lipid nanoparticle complexes disclosed herein can be a full-length antibody or fragment thereof (e.g., a heavy and a light chain encoding mRNA or at least one antibody) capable of targeting one or more viruses or viral proteins derived therefrom.
  • nanoparticle-containing compositions disclosed herein for including with an anti-viral vaccine can include a vaccine against any pathogenic virus capable of infecting a human, other mammal, reptile, or bird.
  • a virus targeted by an mRNA antigen vaccine or a full-length agonistic antibody formed from complexes disclosed herein can include, but is not limited to, Adenoviridae, Alphaviridae, Alphaviridea, Anelloviridae, Anelloviridae, Archiviridea, Arenaviridae, Arenaviridae, Arteriviridae, Astroviridae, Astroviridae, Bornaviridae, Bunyaviridae, Bunyaviridae, Caliciviridae, Caulimoviridae, Coronaviridae, Filoviridae, Flaviviridae, Hepadnaviridae, Hepeviridae, Herpesviridae, influenza virus, Lyssavirus, Orthobunyavirus, Orthomyxoviridae, Papillomaviridae, Papovaviridae, Paramyxoviridae, Paramyxoviridae, Parvoviridae,
  • a full-length agonistic antibody formed from mRNA transcripts of lipid nanoparticle complexes disclosed herein can be a full-length antibody or fragment thereof (e.g., a heavy and a light chain encoding mRNA or at least one antibody) capable of targeting one or more bacteria or bacterial proteins derived therefrom.
  • nanoparticle-containing compositions disclosed herein can be included with an anti-bacterial vaccine that can include any vaccine against a pathogenic bacteria capable of infecting a human, other mammal, reptile, or bird.
  • a bacteria targeted by an mRNA antigen vaccine or a full-length agonistic antibody formed from complexes disclosed herein can include, but is not limited to, Alcaligenes spp. : Bacillus spp. ; Bacteriodes spp. : Bartonella spp. ; Bordetella spp. ; Borrelia spp.; Brevundimonas spp.; Brucella spp.; Burkholderia spp.; Burkholderia spp.; Campylobacter spp.; Candida spp.; Chlamydia spp.; Citrobacter spp.; Clostridium spp.
  • Clostridioides spp. Corynebacterium spp. ; Coxiella spp. ; Enter obacter spp. ; Enterobius spp. ; Escherichia coli spp. (incl. EHEC, EPEC, ETEC, EIEC, EAEC, ESBL/MRGN, DAEC) Francisella tularensis spp., Haemophilus influenzae spp.; Helicobacter spp.; Klebsiella spp.; Leclercia spp. ; Legionella spp. ; Leishmania spp. ; Leptospira spp.
  • I ⁇ euconostoc spp. Listeria spp. ; Micrococcus spp. ; Microsporum spp. ; Moraxella spp. ; Morganella spp. Mycobacterium, spp.; Mycoplasma spp.; Neisseria spp.; Orientia spp.; Pantoea spp.; Paracoccus spp.; Prevotella spp.; Propionibacterium spp.; Proteus spp.; Proteus spp.; Providencia spp.; Pseudomonas spp.; Ralstonia spp.; Rickettsia spp.; Roseomonas spp.; Salmonella spp.; Shigella spp.; Sphingomonas spp.; Staphylococcus spp.; Stenotrophomonas spp.; Strepto
  • the present disclosure includes pharmaceutical compositions.
  • pharmaceutical compositions disclosed herein can include any of the nanoparticle-containing complexes disclosed herein and at least one pharmaceutically acceptable excipient or carrier.
  • pharmaceutical compositions disclosed herein can further include one or more additional agents.
  • one or more additional agents suitable for inclusion into pharmaceutical compositions disclosed herein can include one or more more adjuvants or an anti-cancer or anti-microbial agent.
  • the present disclosure provides methods of treating a subject having a health condition.
  • methods of treating a subject having a health condition disclosed herein can include administering at least one of the compositions or pharmaceutical compositions disclosed herein to the subject.
  • a health condition to be treated by the methods and compositions disclosed herein can include cancer.
  • cancer can include a solid tumor.
  • the solid tumor can include, but is not limited to, a lung, liver, kidney, lymph node, breast, prostate, esophageal, stomach, skin, bone, intestinal, uterine or other solid tumor.
  • a health condition to be treated by the methods and compositions disclosed herein can be a microbial infection.
  • a health condition to be treated by the methods and compositions disclosed herein can be due to a pathogenic microorganism and can include a virus, a bacterium, fungus, protozoan or prion.
  • methods of administering compositions disclosed herein to a subject contemplated herein can be any suitable mode.
  • methods of administering compositions disclosed herein to a subject can include subcutaneous, intranasal, by inhalation, intraocular, intravenous, or other mode of administration at the time of, before or after administering a vaccine by the same or different mode of administration or together as a single composition or as a single composition of nanoparticles containing all agents to be delivered to the subject.
  • methods of administering compositions can include localized administration directly to a tumor site in the subject.
  • modes of administration of pharmaceutical compositions disclosed herein can include, at least one of, intravenously, intramuscularly, intranasally, by aerosol, intradermally, transdermally, topically, intrauterine, or intrarectally administering the pharmaceutical composition or other suitable mode of administration to the subject.
  • nanoparticle complexes encoding one or more antibodies can be administered by the same route as nanoparticle complexes encoding an antigen, for example a vaccine formulation.
  • nanoparticle-containing complex compositions encapsulating mRNA transcripts for encoding an agonist or antagonist can be formulated and then stored at reduced temperatures for later administration.
  • storage temperatures for compositions disclosed herein can be about -10 or less and stored for about 1 week to several months.
  • storage temperatures of ionizing lipid nanoparticle-encapsulated mRNA transcripts disclosed herein can be about -20 or colder for about 2 weeks to about 2 months or more.
  • vectors encoding one or more of the mRNA transcripts disclosed herein are contemplated.
  • vectors can include one or more conventional control elements which are operably linked to the mRNA-encoding sequence in a manner which permits its transcription, translation and/or expression in a cell.
  • operably linked sequences include both expression control sequences that are contiguous with the mRNA-encoding sequence and expression control sequences that act in trans or at a distance to control the polynucleotide- encoding sequence.
  • Expression control sequences can include appropriate transcription initiation, termination, promoter and enhancer sequences; efficient RNA processing signals such as splicing and polyadenylation (polyA) signals; sequences that stabilize cytoplasmic mRNA; sequences that enhance translation efficiency (e.g, Kozak consensus sequence); sequences that enhance protein stability; and when desired, sequences that enhance secretion of the encoded product.
  • RNA processing signals such as splicing and polyadenylation (polyA) signals
  • sequences that stabilize cytoplasmic mRNA sequences that enhance translation efficiency (e.g, Kozak consensus sequence); sequences that enhance protein stability; and when desired, sequences that enhance secretion of the encoded product.
  • a number of expression control sequences, including promoters which are native, constitutive, inducible and/or tissue-specific, are known in the art. and can be utilized herein.
  • a vector encoding a heavy chain can be different than the vector encoding the light chain in order to for example,
  • LNP complexes e.g., ionizing lipid nanoparticles
  • LNP complexes disclosed herein can encapsulate mRNA transcripts for encoding any antibody disclosed herein.
  • LNP complexes disclosed herein can encapsulate expression vectors encoding one or more mRNA transcripts herein.
  • LNP complexes disclosed herein can encapsulate at least one mRNA transcript encoding for a light chain of an antibody and at least one mRNA transcript encoding a heavy chain of an antibody.
  • LNP complexes disclosed herein can encapsulate one or more expression vectors encoding at least one mRNA transcript for a heavy chain of an antibody and one or more expression vectors encoding at least one mRNA transcript for a light chain of an antibody.
  • LNP complexes disclosed herein can encapsulate expression vectors encoding a mRNA transcript for a heavy chain of an antibody and expression vectors encoding a mRNA transcript for a light chain of an antibody in a stoichiometric ratio. In some embodiments, LNP complexes disclosed herein can encapsulate one expression vector encoding a mRNA transcript for a heavy chain of an antibody and one expression vector encoding a mRNA transcript for a light chain of an antibody (e.g., 1 heavy chain transcript to 1 light chain transcript).
  • LNP complexes disclosed herein can encapsulate two expression vectors encoding a mRNA transcript for a heavy chain of an antibody and one expression vector encoding a mRNA transcript for a light chain of an antibody (e.g., 2 heavy chain transcripts to 1 light chain transcript). In some embodiments, LNP complexes disclosed herein can encapsulate four expression vectors encoding a mRNA transcript for a heavy chain of an antibody and one expression vector encoding an mRNA transcript for a light chain of an antibody (e.g., 4 heavy chain transcripts to 1 light chain transcript).
  • LNP complexes disclosed herein can encapsulate six expression vectors encoding a mRNA transcript for a heavy chain of an antibody and one expression vector encoding a mRNA transcript for a light chain of an antibody (e.g., 6 heavy chain transcripts to 1 light chain transcript). In some embodiments, LNP complexes disclosed herein can encapsulate eight expression vectors encoding an mRNA transcript for a heavy chain of an antibody and one expression vector encoding an mRNA transcript for a light chain of an antibody (e.g., 8 heavy chain transcripts to 1 light chain transcript).
  • LNP complexes disclosed herein can encapsulate ten expression vectors encoding an mRN A transcript for a heavy chain of an antibody and one expression vector encoding a mRNA transcript for a light chain of an antibody (e.g., 10 heavy chain transcripts to 1 light chain transcript).
  • LNP complexes disclosed herein can encapsulate expression vectors encoding an mRNA transcript for a heavy chain of an antibody and expression vectors encoding an mRNA transcript for a light chain of an antibody wherein a full-length antibody can be formed after administering the composition into a cell or a tissue of a subject.
  • a full-length antibody formed herein can be a monoclonal antibody against a target molecule or an antigen.
  • a full-length antibody formed disclosed herein can be an agonistic or antagonistic antibody.
  • a full-length antibody formed herein can be an immune checkpoint inhibitor where an antibody translated from encapsulated mRNA transcripts disclosed herein can bind to or associate with the immune checkpoint inhibitor and modulate its activity.
  • an antibody translated from encapsulated mRNA transcripts disclosed herein can bind to or associate with the immune checkpoint inhibitor and partially or completely block the immune checkpoint inhibitor’s activity.
  • an antibody translated from encapsulated mRNA transcripts disclosed herein can bind to or associate with the immune checkpoint inhibitor and reversibly inhibit activity or regulate its activity for a pre-determined period.
  • an antibody translated from encapsulated mRNA transcripts disclosed herein can bind to, or associate with an immunologically relevant molecule in an agonistic mode and stimulate or modulate lymphocyte activity for use to treat a health condition.
  • a full-length antibody can include, but is not limited to, an anti-HER2 antibody, an anti-EGFR antibody, an anti-VEGRF antibody, an anti-VEGF antibody, an anti-CD-20 antibody, an anti-CD-22 antibody, an anti-CD30 antibody, an anti- CD33 antibody, an anti-CD38 antibody, an anti-CD40 antibody, an anti-CD52 antibody, an anti-CTLA-4 antibody, an anti-PD-1 antibody, an anti-PD-Ll antibody, an anti-RANKL antibody, an anti-GD2 antibody, an anti-PDGFR antibody, an anti-SLAMF7 antibody, or any combination thereof.
  • expression vectors encoding mRNA transcripts herein can be formulated into lipid nanoparticles (LNPs) for delivery into cells, tissues, and/or a subject.
  • LNPs are spherical vesicles made of ionizable lipids, which can be positively charged at low pH (enabling RNA complexation) and neutral at physiological pH (reducing potential toxic effects, as compared with positively charged lipids, such as liposomes).
  • LNPs can be taken up by cells via endocytosis, and the ionizability of the lipids at low pH (likely) can enable endosomal escape, which allows release of the cargo into the cytoplasm.
  • LNPs for use in embodiments disclosed herein can contain ionizable cationic lipids, non-cationic lipids, sterols, and/or PEG lipids components along with expression vectors encoding mRNA transcripts disclosed herein.
  • LNPs disclosed herein can have about 5- 25% non-cationic lipid.
  • LNP nanoparticle complexes disclosed herein can have about 5-20%, 5-15%, 5-10%, 10-25%, 10-20%, 10-25%, 15-25%, 15-20%, or 20-25% non-cationic lipid.
  • Non-limiting examples of non-cationic lipids include distearoylphosphatidylcholine (DSPC), dioleoylphosphatidylcholine (DOPC), dipalmitoylphosphatidylcholine (DPPC), dioleoylphosphatidylglycerol (DOPG), dipalmitoylphosphatidylglycerol (DPPG), ioleoylphosphatidylethanolamine (DOPE), palmitoyloleoylphosphatidylcholine (POPC), palmitoyloleoyl-phosphatidylethanolamine (POPE), dipalmitoyl phosphatidyl ethanolamine (DPPE), dimyristoylphosphoethanolamine (DMPE), distearoyl-phosphatidyl-ethanolamine (DSPE), sphingolipids, cerebrosides, gangliosides, 16-O-monomethyl PE, 16-O-diraethyl P, 18-1
  • mRNA transcripts disclosed herein can be encapsulated in ionizable lipid nanoparticles (LNP), the components of which facilitate fusion of the LNPs to an endosomal membrane, allowing access of nanoparticle constituents (e.g., the mRNA) to the cytoplasm of a targeted cell taking up the LNP permitting in vivo formation of the antibody or antibody fragment encoded by the mRNA cargo.
  • LNP ionizable lipid nanoparticles
  • mRNA transcripts disclosed herein can be encapsulated in DSPC, also referred to as l,2-distearoyl-sn-glycero-3 -phosphocholine, cholesterol, also referred to as Distearoylphosphatidylcholine or other similar lipid known in the art and administered to a subject alone or in combinations disclosed herein for in vivo synthesis of one or more encoded antibody or fragment thereof in the treatment of a health condition disclosed herein.
  • DSPC also referred to as l,2-distearoyl-sn-glycero-3 -phosphocholine
  • cholesterol also referred to as Distearoylphosphatidylcholine or other similar lipid known in the art and administered to a subject alone or in combinations disclosed herein for in vivo synthesis of one or more encoded antibody or fragment thereof in the treatment of a health condition disclosed herein.
  • mRNA transcripts disclosed herein can be encapsulated in DMG-PEG2000, also referred to as l,2-dimyristoyl-rac-glycero-3- methoxypolyethylene glycol-2000 or other similar lipid known in the art and administered to a subject alone or in combinations disclosed herein for in vivo synthesis of one or more encoded antibody or fragment thereof in the treatment of a health condition disclosed herein.
  • LNPs can contain a helper lipid to promote cell binding, cholesterol to fill the gaps between the lipids, and a polyethylene glycol (PEG) to reduce opsonization by serum proteins and reticuloendothelial clearance.
  • LNPs for use herein can include one or more synthetic ionizable phospholipids provided herein and at least one helper lipid.
  • LNPs for use herein can include one or more synthetic ionizable phospholipids provided herein and at least one helper lipid selected from: l,2-dioleoyl-sn-glycero-3- phosphoethanol amine (DOPE), N-methyldioctadecylamine (MDOA), 1,2-dioleoyl-3- dimethylammonium-propane (DODAP), dimethyldioctadecylammonium bromide salt (DDAB), l,2-dioleoyl-3-trimethylammonium-propane (DOTAP), and any combination thereof.
  • DOPE l,2-dioleoyl-sn-glycero-3- phosphoethanol amine
  • MDOA N-methyldioctadecylamine
  • DODAP 1,2-dioleoyl-3- dimethylammonium-propane
  • DDAB dimethyldioctadecylam
  • LNPs for use herein can include one or more synthetic ionizable phospholipids provided herein and at least one zwitterionic helper lipid (e.g., DOPE), ionizable cationic helper lipid (e.g., MDOA, DODAP), permanently cationic helper lipid (e.g., DDAB, DOTAP), or any combination thereof.
  • LNPs of use herein concern LNPs that permit optimal transport and expression of mRNAs encoding antibodies disclosed herein in order to induce or induce an enhanced cellular immune response in a subject.
  • LNPs for use in complexes disclosed herein can include one or more synthetic ionizable phospholipids provided herein and at least one cholesterol and/or a cholesterol derivative.
  • cholesterol derivative refers to any compound consisting essentially of a cholesterol structure, including additions, substitutions and/or deletions thereof.
  • cholesterol derivative herein can also include steroid hormones and bile acids as are generally recognized in the art.
  • Non-limiting examples of cholesterol derivatives suitable for use herein can include dihydrocholesterol, ent-cholesterol, epi-cholesterol, desmosterol, cholestanol, cholestanone, cholestenone, cholesteryl-2 '-hydroxy ethyl ether, cholesteryl-4'-hydroxybutyl ether, 3 ⁇ -[N- (N'N'-dimethylaminoethyl)carbamoyl cholesterol (DC-Chol), 24(S)-hydroxycholesterol, 25- hydroxycholesterol, 25(R)-27-hydroxycholesterol, 22-oxacholesterol, 23-oxacholesterol, 24- oxacholesterol, cycloartenol, 22-ketosterol, 20-hydroxysterol, 7 -hydroxy cholesterol, 19- hydroxy cholesterol, 22-hydroxy cholesterol, 25-hydroxy cholesterol, 7-dehydrocholesterol, 5a-cholest-7-en-3 ⁇ -ol, 3,6,9-trio
  • LNPs for use in certain complexes disclosed herein can include one or more synthetic ionizable phospholipids provided herein and at least one PEG or PEG-modified lipids.
  • a PEG-modified lipid, or “PEG lipid” refers to a lipid modified with polyethylene glycol (PEG). Such species can be alternately referred to as PEGylated lipids.
  • Non-limiting examples of PEG-modified lipids suitable for use herein can include PEG-modified phosphatidylethanol amines, PEG-modified phosphatidic acids, PEG-modified ceramides (PEG-CER), PEG-modified dialkylamines, PEG-modified diacylglycerols (PEG-DAG), PEG-modified dialkylglycerols, and mixtures thereof.
  • a PEG- modified lipid for use herein can be PEG-c-DOMG (R-3-[(co- methoxypoly(ethyleneglycol)2000)carbamoyl]-l,2-dimyristyloxy-propyl-3-amine poly(ethylene glycol)); PEG-DMG (l,2-dimyristoyl-rac-glycero-3-methoxypolyethylene glycol poly(ethylene glycol)); PEG-DLPE (l,2-Dilauroyl-sn-glycero-3 -phosphorylglycerol sodium salt-poly(ethylene glycol)); PEG-DMPE (dimethyl-2- (dimethylphosphino)ethylphosphine-poly(ethylene glycol)); PEG-DPPC (1,2-dipalmitoyl-sn- glycero-3-phosphocholine-poly(ethylene glycol)); PEG-DSPE (1, 2-distearoyl-sn
  • a PEG- modified lipid for uses disclosed herein can include a PEG moiety having a size of from about 1000 daltons to about 20,000 daltons.
  • a PEG-modified lipid for use can include a PEG moiety having a size of about 1000 daltons, about 2000 daltons, about 5000 daltons, about 10,000 daltons, about 15,000 daltons, or about 20,000 daltons.
  • LNPs herein can include one or more synthetic ionizable phospholipids provided herein and at least one PEG or PEG-modified lipids wherein the PEG moiety can have a size of about 2000 daltons.
  • PEG-lipids for use in making the LNPs described herein include, but are not limited to, l,2-Diacyl-sn-Glycero-3- Phosphoethanolamine-N-[Methoxy(Poly ethylene glycol)-350] (mPEG 350 PE); 1,2-Diacyl- sn-Glycero-3-Phosphoethanolamine-N-[Methoxy(Polyethylene glycol)-550] (mPEG 550 PE); l,2-Diacyl-sn-Glycero-3-Phosphoethanolamine-N-[Methoxy(Poly ethylene glycol)-750] (mPEG 750 PE); l,2-Diacyl-sn-Glycero-3-Phosphoethanolamine-N-[
  • LNPs herein can include one or more synthetic ionizable phospholipids provided herein and l,2-dimyristoyl-rac-glycero-3- methoxy(poly(ethylene glycol-2000)) (DMG-PEG2000).
  • LNP complexes disclosed herein can include one or more agents to target one or more cell types.
  • LNP complexes can be selective for one or more cell types.
  • LNP complexes can unload cargo at one or more selective cell types.
  • LNP complexes can selectively target one or more cell types.
  • LNP complexes can selectively target one or more tissue types.
  • LNP complexes disclosed herein can selectively target one or more cancers.
  • LNP size can impact behavior of lipid nanoparticles in vivo.
  • LNP complexes disclosed herein can be about 20 nm to about 1000 nm in diameter or size. In some embodiments, LNP complexes disclosed herein can be about 20 nm to about 200 nm in size. In some embodiments, LNP complexes disclosed herein can about 20 nm to about 190 nm or about 25 nm to about 190 nm in size. In some embodiments, LNP complexes disclosed herein can be about 30 nm to about 180 nm in size.
  • LNP complexes disclosed herein can be about 35 nm to about 170 nm in size. In some embodiments, LNP complexes disclosed herein can be about 40 nm to about 160 nm in size. In some embodiments, LNPs herein can be about 50 nm to about 150 nm, about 60 nm to about 140 nm, about 70 nm to about 130 nm, about 80 nm to about 120 nm, or about 90 nm to about 110 nm in size.
  • LNP complexes disclosed herein can be about 20 nm, about 25 nm, about 30 nm, about 35 nm, about 40 nm, about 45 nm, about 50 nm, about 55 nm, about 60 nm, about 65 nm, about 70 nm, about 75 nm, about 80 nm, about 85 nm, about 90 nm, about 95 nm, about 100 nm, about 105 nm, about 110 nm, about 115 nm, about 120 nm, about 125 nm, about 130 nm, about 135 nm, about 140 nm, about 145 nm, about 150 nm, about 155 nm, about 160 nm, about 165 nm, about 170 nm, about 175 nm, about 180 nm, about 185 nm, about 190 nm, about 195 nm, or about 200 nm in size or diameter.
  • an average LNP size in a LNP complex-containing composition disclosed herein can be about 20 nm to about 1000 nm (e.g., about 20, 40, 60, 80, 100, 150, 200, 250, 300, 350, 400, 450, 500, 550, 600, 650, 700, 750, 800, 850, 900, 950, 1000) in diameter in average size.
  • LNP size in a LNP composition herein can be homogenous at about 20 nm to about 1000 nm (e.g., about 20, 40, 60, 80, 100, 150, 200, 250, 300, 350, 400, 450, 500, 550, 600, 650, 700, 750, 800, 850, 900, 950, 1000) in diameter in size.
  • LNP size in a LNP composition herein be heterogeneous at about 20 nm to about 1000 nm (e.g., about 20, 40, 60, 80, 100, 150, 200, 250, 300, 350, 400, 450, 500, 550, 600, 650, 700, 750, 800, 850, 900, 950, 1000) in diameter in average size.
  • LNP size in a LNP composition herein can be heterogeneous wherein about 50% to about 99% of the LNPs average at about 20 nm to about 1000 nm (e.g., about 20, 40, 60, 80, 100, 150, 200, 250, 300, 350, 400, 450, 500, 550, 600, 650, 700, 750, 800, 850, 900, 950, 1000) in diameter in average size.
  • LNPs size in a LNP composition herein can be heterogeneous wherein about 50% to about 99% of the LNPs average at about 20 nm to about 1000 nm (e.g., about 20, 40, 60, 80, 100, 150, 200, 250, 300, 350, 400, 450, 500, 550, 600, 650, 700, 750, 800, 850, 900, 950, 1000) in diameter in average size.
  • compositions are contemplated.
  • pharmaceutical compositions can include one or more of the LNPs encapsulating mRNA transcripts disclosed herein.
  • pharmaceutical compositions herein can include one or more of the LNPs encapsulating mRNA transcripts disclosed herein and at least one pharmaceutically acceptable excipient or carrier.
  • the term “pharmaceutically acceptable” refers to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues, organs, and/or bodily fluids of a subject without excessive toxicity, irritation, allergic response, or other problems or complications commensurate with a reasonable benefit/risk ratio.
  • pharmaceutically acceptable carrier can refer to solvents, dispersion media, coatings, antibacterial agents, antifungal agents, isotonic and absorption delaying agents, or the like that are physiologically compatible.
  • Pharmaceutically acceptable carriers suitable for use herein include, but are not limited to, buffers that are well known in the art, and can be phosphate, citrate, and other organic acids; antioxidants including ascorbic acid and methionine; preservatives; low molecular weight polypeptides; proteins, such as serum albumin, gelatin, or immunoglobulins; amino acids; hydrophobic polymers; monosaccharides; disaccharides; and other carbohydrates; metal complexes; and/or non-ionic surfactants.
  • buffers that are well known in the art, and can be phosphate, citrate, and other organic acids; antioxidants including ascorbic acid and methionine; preservatives; low molecular weight polypeptides; proteins, such as serum albumin, gelatin, or immunoglobulins; amino acids; hydrophobic polymers; monosaccharides; disaccharides; and other carbohydrates; metal complexes; and/or non-ionic surfactants.
  • compositions for uses disclosed herein can be formulated for parenteral administration, such as intravenous or intravascular, bolus infusion, intrarenal introduction, intracerebroventricular injection, intra-ci sterna magna injection, intra-parenchymal injection, or a combination thereof.
  • parenteral administration such as intravenous or intravascular, bolus infusion, intrarenal introduction, intracerebroventricular injection, intra-ci sterna magna injection, intra-parenchymal injection, or a combination thereof.
  • pharmaceutical compositions for use herein can be formulated for infusion of LNP complexes encapsulating mRNA transcripts into a tumor.
  • compositions for use herein be formulated for parenteral/ infusion administration can include pharmaceutically acceptable carriers including sterile liquids, such as water and oil, including those of petroleum, animal, vegetable or synthetic origin, such as peanut oil, soybean oil, mineral oil, and the like. Saline solutions and aqueous dextrose, polyethylene glycol (PEG) and glycerol solutions can also be employed as liquid carriers, particularly for injectable solutions.
  • pharmaceutical compositions for use herein can further include additional ingredients, for example preservatives, buffers, tonicity agents, antioxidants and stabilizers, nonionic wetting or clarifying agents, viscosity -increasing agents, and the like.
  • pharmaceutical compositions described herein can be packaged in single unit dosages or in multi -dosage forms.
  • formulations suitable for parenteral/infusion administration include aqueous and non-aqueous sterile injection solutions which can contain anti-oxidants, buffers, bacteriostats and solutes which render the formulation isotonic with the blood of the intended recipient; and aqueous and non- aqueous sterile suspensions which can include suspending agents and thickening agents.
  • aqueous solutions can be suitably buffered (e.g., a pH of from about 3.0 to about 9.0).
  • compositions herein can further include one or more pharmaceutically acceptable salts.
  • pharmaceutically acceptable salts include acid addition salts (formed from a free amino group of a polypeptide with an inorganic acid, or an organic acid.
  • the salt formed with the free carboxyl groups is derived from an inorganic base, or an organic base.
  • any of the pharmaceutical compositions herein can be used in therapeutic applications, for example, treating a disease and/or a disorder in human patients, which are disclosed herein.
  • any of the pharmaceutical compositions herein can be used in therapeutic applications, for example, treating a cancer, a viral infection, a bacterial infection, an autoimmune disorder, an allograft rejection, or any combination thereof in human patients, which are also disclosed herein.
  • methods of treating or ameliorating a health condition and/or a disorder in a subject include, but are not limited to, administration of an effective amount of any the LNP complexes encapsulating mRNA transcripts and/or pharmaceutical compositions containing these LNP complexes thereof as described herein.
  • “An effective amount” as used herein refers to a dose of LNP complexes encapsulating mRNA transcripts that is sufficient to confer a therapeutic effect on a subject having or suspected of having a disease and/or a disorder herein.
  • a therapeutic effect for a subject having or suspected of having a disease and/or a disorder herein can include reducing the symptoms or consequences of the disease.
  • methods of administering LNP complexes encapsulating mRNA transcripts as disclosed herein can include placement (e.g., transplantation or implantation) of any the LNP complex encapsulating mRNA transcripts and/or pharmaceutical compositions containing these LNP complexes into a subject, by a method or route that results in at least partial localization of the introduced LNP at a desired site, such as a tumor, such that a desired effect(s) is produced.
  • a subject can be transfused with LNP complexes encapsulating mRNA transcripts disclosed herein over the course of a day, for a few hours, daily, every other day, 2 times per week, weekly, every other week, monthly, or other appropriate treatment regimen.
  • the period of full-length antibodies generated after administration of LNPs encapsulating mRNA transcripts herein to a subject can be a few hours (e.g, about 2 hours, about 6 hours, about 12 hours, about 24 hours), a few days (e.g., about 1 day, about 2 days, about 3 days, about 4 days about 5 days, about 6 days, about 7 days), weeks (e.g., about 2 weeks, about 4 weeks, about 6 weeks, about 12 weeks, about 40 weeks, about 52 weeks), to as long as several years (e.g., about 2 years, about 5 years), or even the life time of the subject.
  • a few hours e.g, about 2 hours, about 6 hours, about 12 hours, about 24 hours
  • a few days e.g., about 1 day, about 2 days, about 3 days, about 4 days about 5 days, about 6 days, about 7 days
  • weeks e.g., about 2 weeks, about 4 weeks, about 6 weeks, about 12 weeks, about 40 weeks, about 52 weeks
  • an effective amount of the LNP complex encapsulating mRNA transcripts herein can be administered via a systemic route of administration, such as an intraperitoneal or intravenous route.
  • a systemic route of administration such as an intraperitoneal or intravenous route.
  • cellular immunity can be assessed in a subject before, during or after being treated by complexes disclosed herein in order to assess level of induction and in order to design a regimen tailored to treating the subject.
  • a subject to any of the methods disclosed herein can be any subject for whom treatment or therapy is desired.
  • a subject to any of the methods disclosed herein can be any subject having or is suspected of having a health condition such as cancer or an infection or exposed to a pathogen or a disorder in need of treatment with a full-length antibody generated after administration of LNP complex encapsulating mRNA transcripts herein to induce cellular immunity in the subject.
  • a subject to any of the methods herein can be any subject having or is suspected of having a cancer, a viral infection, a bacterial infection, an autoimmune disorder, an allograft rejection, or any combination thereof.
  • a subject can be a mammal.
  • a subject can be a human patient.
  • a human subject such as an adult, child, adolescent, toddler, young adult or infant or fetus who is in need of the methods herein can be identified by routine medical examination, e.g., laboratory tests, biopsy, magnetic resonance imaging (MRI) scans, ultrasound exams, and the like.
  • MRI magnetic resonance imaging
  • a mammal can include a pet, livestock, a horse, or other mammal can be analyzed and assessed for need of a treatment disclosed herein.
  • vaccines applicable to any subject population can be administered at the same time, before, or after nanoparticle complex compositions disclosed herein encoding at least one antibody for inducing a cellular immune response are administered to the subject of the subject population (e.g., herd of livestock, horses or group of people) in order to enhance the desired response of the vaccine.
  • a subject to any of the methods herein can be any subject having or is suspected of developing cancer, a solid tumor, a metastasis, or a combination thereof.
  • tumors treated by the methods herein can be a solid tumor, a liquid or non-solid tumor.
  • a solid tumor can include, but is not limited to, breast, lung, brain, liver, kidney, eye, skin, stomach, esophageal, prostate, uterine or another solid tumor.
  • a solid tumor can be a sarcoma.
  • tumors treated by the methods herein can be a bone sarcoma, a soft-tissue sarcoma, or any combination thereof.
  • adenocarcinomas are contemplated.
  • GI tract carcinomas are contemplated for treatment herein.
  • cancers and/or resulting tumors thereof that can be treated by the methods herein can be osteosarcoma, chondrosarcoma, poorly differentiated round/spindle cell tumors, Ewing sarcoma, hemangioendothelioma, angiosarcoma, fibrosarcoma/myofibrosarcoma, chordoma, adamantinoma, liposarcoma, leiomyosarcoma, malignant peripheral nerve sheath tumor, rhabdomyosarcoma, synovial sarcoma, malignant solitary fibrous tumor, or any combination thereof.
  • tumors treated by the methods herein can be liposarcoma, atypical lipomatous tumor, dermatofibrosarcoma protuberans, malignant solitary fibrous tumor, inflammatory myofibroblastic tumor, low-grade myofibroblastic sarcoma, fibrosarcoma, myxofibrosarcoma, low-grade fibromyxoid sarcoma, giant cell tumor of soft tissues, leiomyosarcoma, malignant glomus tumor, rhabdomyosarcoma, hemangioendothelioma, angiosarcoma of soft tissue, extraskeletal osteosarcoma, gastrointestinal stromal tumor, malignant, malignant peripheral nerve sheath tumor, malignant Triton tumor, malignant granular cell tumor, malignant ossifying fibromyxoid tumor, stromal sarcoma not otherwise specified, myoepithelial carcinoma, malignant phosphaturic mesenchymal tumor, syn
  • methods of administrating LNP complexes encapsulating mRNA transcripts as disclosed herein can reduce growth of solid tumors and/or solid tumor cells. In some embodiments, methods of administrating LNP complexes encapsulating mRNA transcripts as disclosed herein can reduce tumor size of solid tumors. In some embodiments, methods of administrating LNP complexes encapsulating mRNA transcripts as disclosed herein can induce apoptosis of solid tumors and solid tumor cells. In some embodiments, methods of administrating LNP complexes encapsulating mRNA transcripts as disclosed herein can attenuate tumor growth and/or tumor expansion and/or induce tumor cell killing. In some embodiments, methods of administrating LNP complexes encapsulating mRNA transcripts as disclosed herein can reduce the risk of metastasis.
  • a subject to any of the methods disclosed herein can be any subject having or is suspected of developing an infection or exposed to a microorganism or suspected of being exposed to a microorganism.
  • a subject to any of the methods disclosed herein can be any subject having or is suspected of developing an infection resulting from an infectious microorganism such as a pathogenic microorganism including, but not limited to a virus, a bacteria, fungus, prion, protozoan or a combination thereof.
  • a subject to any of the methods disclosed herein can be any subject having or is suspected of developing or having been exposed to a virus or having an infection resulting from a virus.
  • administrating LNP complexes encapsulating mRNA transcripts as disclosed can generate at least one antibody that can target one or more proteins on surface of a virus.
  • administrating LNPs encapsulating mRNA transcripts as disclosed can generate at least one antibody that can target one or more proteins on surface of a virus belonging to the Adenoviridae, Alphaviridae, Alphaviridea, Anelloviridae, Anelloviridae, Archiviridea, Arenaviridae, Arenaviridae, Arteriviridae, Astroviridae, Astroviridae, Bornaviridae, Bunyaviridae, Bunyaviridae, Caliciviridae, Caidimoviridae, Coronaviridae, Filoviridae, Flaviviridae, Hepadnaviridae, Hepeviridae, Herpesviridae, Lyssavirus, Orthobunyavirus, Orthomyxoviridae, Papillomaviridae, Papovaviridae, Paramyxoviridae, Paramyxoviridae, Parvoviridae, Picornaviridae
  • LNP complexes encapsulating mRNA transcripts and/or pharmaceutical compositions disclosed herein can be administered in dosages and by techniques well known to those skilled in the medical and veterinary arts.
  • healthcare professions will take into consideration such factors as the age, sex, weight, and condition of the particular patient, and the composition form used for administration (e.g., solid vs. liquid). Dosages for humans or other mammals can be determined without undue experimentation by the skilled artisan, from this disclosure, and the knowledge in the art.
  • kits are contemplated of use to generate, store and/or transport the LNP complexes encapsulating mRNA transcripts disclosed herein.
  • kits can include LNP complexes encapsulating mRN A transcripts where the LNP complexes can be used immediately or frozen and stored for transport and later use.
  • a kit disclosed herein can include any of vectors encoding mRNA transcripts disclosed herein, LNP complexes encapsulating mRNA transcripts and/or pharmaceutical compositions disclosed herein.
  • kits can further include a target mRNA vaccine of use for inducing an immune response against a specific pathogenic agent or tumor.
  • kits are provided for use in treating or alleviating a targeted disease or condition treatable by use of LNP complexes encapsulating mRNA transcripts disclosed herein.
  • kits can include instructions for use in accordance with any of the methods described herein.
  • the included instructions can include a description of administration of any the LNP complexes encapsulating mRNA transcripts and/or pharmaceutical compositions described herein and optionally one or more additional therapies to treat, delay the onset, or alleviate a target disease (e.g., cancer, infection) as those described herein.
  • kits herein can further include a description of selecting an individual suitable for treatment based on identifying whether that individual has the target disease, e.g., applying one or more diagnostic methods.
  • the instructions can include a description of administering LNP complexes encapsulating mRNA transcripts to a subj ect at risk of the target disease.
  • compositions and methods for targeted cellular depletion are contemplated to also be attainable by these methods.
  • use of LNP-encapsulated mRNA for the production of biol ogically active agonistic and biologically active antagonistic/blocking antibodies have been demonstrated.
  • LNPs containing the heavy and light chains of monoclonal antibodies which mediate targeted cellular depletion can be utilized.
  • antibodies against CD20 and/or CD 19 can be encoded by LNP encapsulation of raRNAs for production of anti-CD20 and/or anti- CD19 antibodies for efficacious use against B cell lymphomas based on their capacity to induce B-cell directed depletion.
  • antibodies against molecules such as VLA4 can facilitate both blockade of activated lymphocyte trafficking as well as activated lymphocyte depletion as therapeutic modalities using nanoparticle complexes disclosed herein for in vivo delivery and synthesis of anti-VLA4 antibodies or a similar target for directed cellular depletion.
  • antibody production via mRNA encapsulated LNPs disclosed herein can be used for intervention in therapeutic modalities requiring monoclonal antibody-mediated cellular depletion.
  • kits can include instructions for using the components of the kit, for example relating to the use of LNP complexes encapsulating mRNA transcripts.
  • Instructions for using the components of the kit can generally include information as to dosage, dosing schedule, and/or route of administration for the intended treatment.
  • containers can be unit doses, bulk packages ⁇ e.g., multi-dose packages) or sub-unit doses.
  • instructions supplied in the kits of the invention can be written instructions on a label or package insert (e.g, a paper sheet included in the kit), but machine-readable instructions (e.g, instructions carried on a magnetic or optical storage disk) are also acceptable.
  • a label or package insert herein can indicate that the composition is used for treating, delaying the onset and/or alleviating the disease (e.g., cancer, infection). Instructions can be provided for practicing any of the methods described herein.
  • kits disclosed herein include suitable packaging.
  • suitable packaging includes, but is not limited to, syringes, vials, bottles, jars, flexible packaging (e.g., sealed Mylar or plastic bags), and the like.
  • packages for use in combination with a specific device such as an inhaler, nasal administration device (e.g, an atomizer) or an infusion device such as a minipump.
  • a kit can have a sterile access port (for example the container can be an intravenous solution bag or a vial having a stopper pierceable by a hypodermic injection needle).
  • the container can also have a sterile access port (for example the container can be an intravenous solution bag or a vial having a stopper pierceable by a hypodermic injection needle).
  • a sterile access port for example the container can be an intravenous solution bag or a vial having a stopper pierceable by a hypodermic injection needle.
  • at least one active agent in the composition can include an antibody generated by LNPs encapsulating mRNA transcripts disclosed herein.
  • lipid particle-encapsulated mRNA vaccine platform Commercially available representatives of a lipid particle-encapsulated mRNA vaccine platform include Modema’s and Pfizer/BioNtech’s (“PBN”) SARS-CoV2 mRNA vaccines. While both Modema and PBN vaccines induce humoral immunity and some consistent and detectable cellular responses, improved cellular response for overall improved immunity are needed in order to provide more robust responses to these vaccines and other vaccines. For example, T cell responses (e.g. both CD4 and CD8 positive T cell response) are low in magnitude when compared to humoral immunity and when compared to other cellular immunity inducing compositions.
  • Modema and Pfizer/BioNtech (“PBN”) SARS-CoV2 mRNA vaccines. While both Modema and PBN vaccines induce humoral immunity and some consistent and detectable cellular responses, improved cellular response for overall improved immunity are needed in order to provide more robust responses to these vaccines and other vaccines. For example, T cell responses (e.g. both CD4
  • T cell responses, IgG, and neutralization titers in response to the Moderna SARS-CoV2 mRNA vaccine were assessed.
  • CD4 and CD8+ T cell responses were measured 60 days post-boost using an activation-induced marker (AIM) assay.
  • AIM assays sensitively and specifically detected antigen-responsive CD4+ and CD8+ T cells after vaccination.
  • human PBMCs were collected and isolated by density gradient centrifugation. Cells were then suspended in FBS containing 10% dimethyl sulfoxide and cryopreserved in liquid nitrogen. Cryopreserved PBMCs were thawed and cultured in serum- free AIM-V media overnight for 18 hours at 37°C.
  • PBMCs were then aliquoted into wells of a 96-well plate, at a total of 1 x 10 6 cells per well.
  • PBMCs were stimulated with 10 pg/ml heat- inactivated antibiotic-killed Group A Streptococcus, strain 5448 (“Ag stimulation”).
  • Ag stimulation As a positive control, cells were stimulated with 1 ug/ml SEB (“SEB stimulation”).
  • SEB stimulation 1 ug/ml SEB (“SEB stimulation”.
  • PMBC aliquots were incubated at 37°C for 18 hours, after which supernatants were harvested and cells were analyzed by flow cytometry.
  • Serum antibody response was measured and used to assess immunogenicity.
  • a functional neutralizing antibody test was used as a serological assay to detect the amount of antibody that can effectively neutralize virus, preventing infectivity in vitro.
  • anti-receptor binding domain (RBD) IgG titers were also assessed on human PMBCs.
  • FIG. 2 illustrates a schematic diagram demonstrating a method developed herein by which specific mRNA sequences could be transcribed in vitro and packaged into LNP for in vivo administration to generate a target molecule.
  • the gene encoding ovalbumin (ova) antigen was designed and synthesized with an alpha globin 5'-UTR, an amino-terminal enhancer of split (AES) motif, and mitochondrial 12S RNA elements.
  • mRNA was produced by T7 RNA polymerase transcription from linearized plasmid templates with co-transcriptional ARCA capping and post-transcriptional polyadenylation. Column-purified mRNA transcripts were incorporated into LNPs composed of Precision Nanosystem's Neuro9 lipid mix (catalog#: NWS0009) on a microfluidic LNP mixer (e.g., Precision NanoSystems Spark).
  • RNA content of LNPs was quantified using a dye-binding assay following detergent-based dissolution of the LNPs and LNPs at concentrations of 8-15 ng/ ⁇ l were ultrafiltered for in vivo administration.
  • escalating amounts of ova injected LNPs were administered to wild type B6 mice and 7 days later the spleen was examined for the frequency of ova-specific CD8+ T cells using K b -SIINFEKL -loaded MHC class I tetramers. While all doses of ova-LNPs produced a detectable population of ova-specific T cells, the use of 1-2 ⁇ g LNP resulted in the most consistent frequency of tetramer positive T cells at -0.2-1% (Fig. 3).
  • LNP-encapsulated mRNA for the delivery of heavy and light chains of an agonistic antibody targeting an immunologically relevant pathway
  • LNP-mRNA-elicited CD8+ T cell responses to delivery 7 of these constructs encoding for an agonistic anti-CD40 antibody (FGK45) was assessed.
  • TLR/IFN + CD40 vaccine adjuvant for generating robust T cells responses, it was investigated whether similar responses might be achieved through delivery of LNP containing mRNA transcripts encoding heavy and light chain antibody sequences derived from an anti-CD40 agonist, FGK45.
  • mRNA vaccines induce a strong IFN signature shortly after injection, without wishing to be bound by theory, it was reasoned that this might alleviate the need to additionally co-deliver a TLR agonist with the LNP construct administration.
  • Constructs encoding heavy and light chain mRNA sequences of FGK45 were assembled, mRNA transcription performed, and the heavy and light chain mRNAs co-encapsulated into lipid nanoparticles (FGK-LNP) as described above for ova.
  • ova LNPs were inj ected alone or with increasing FGK-LNPs into wild-type (WT) B6 mice and the ova-specific CD8+ T cell response was evaluated by dual tetramer staining cells isolated from peripheral blood 7 days post vaccination.
  • FIG. 4A A detectable response was observed following administration of the ova-LNP alone (Fig. 4A). However, after co-administration of increasing doses of the FGK-LNP (the anti- CD40 antibody construct), the intensity of ova specific T cells was >6 fold higher than in response to ova-LNP alone (Figs. 4A and 4B). These data indicated that not only were both heavy and light chain genes for FGK effectively translated following LNP administration, but functional anti-CD40 antibody (FGK) was formed and able to induce a cellular immune response by superi or induction of a CD8+ T cell response.
  • FGK functional anti-CD40 antibody
  • H/L chain ratio Formation of functional antibody requires the heterodimeric assembly of heavy and light chain protein sequences within the ER. Given the differences in length between the heavy and light chain sequences (L chain mRNA sequence is roughly half as long as H), L protein may be in excess when a uniform molarity between the two sequences is contained within the LNP. H and L proteins unable to form heterodimeric pairs are unstable and besides being degraded by proteosomes, may also trigger the Unfolded Protein Response (UPR).
  • UPR Unfolded Protein Response
  • the UPR is important for the production and secretion of antibody from plasma cells but may or may not be beneficial for antibody production and secretion from the muscle and/or parenchymal cells producing protein following LNP-mRNA injection.
  • HZL mRNA ratio within the LNP can be altered by altering the HZL mRNA ratio within the LNP.
  • HZL molar ratios 111, 2: 1, 4: 1, 6: 1,8: 1 respectively
  • Optimized HZL ratios based on the evaluation of cDCl IL-27-GFP production and the magnitude of the CD8 T cell response, were determined as disclosed herein. It is projected that the ratio H/L chain of at least 2: 1 will have an improved production of antibody than a 1 : 1 ratio.
  • LNP-mRNAs can be taken up by muscle/ stromal cells which produce and secrete the encoded protein. The protein is then taken up by professional APCs which migrate to the local lymphoid tissue and initiate the ensuing T cell response; and 2) LNP-mRNAs are taken up directly by professional APCs which incorporate the expressed antigen into the class I presentation pathway.
  • the APC-intrinsic adjuvant effect of the LNP drives optimal APC maturation, migration and initiation of the T cell response.
  • ovalbumin-expressing tumor cell lines were evaluated as an anti -turmor cell model.
  • Ovalbumin-expressing tumor cell lines are a useful tool in the validation of any anti-tumor therapeutic intervention.
  • the discovery that tumor responsiveness to immunotherapy tracks well with the mutational burden in the tumor has reinvigorated the clinical relevance of these model antigen-expressing tumors under the guise of “neo-antigen” representation.
  • antitumor activity of vaccination plus checkpoint blockade was measured using B16 melanoma tumor cells in the ova model. In brief, B16-ova was injected S.C.
  • Fig. 5 illustrates exemplary' antitumor activity measured in the model for the three treatment regimens tested herein.
  • RNA content of LNPs was quantified using a dye- binding assay following detergent-based dissolution of the LNPs and LNPs at concentrations of 50-80 ng/ul were ultrafiltered for in vivo administration.
  • Ova-LNPs containing lug of mRNA were either injected into B6 mice alone or in combination with LNPs containing 2 ug of heavy and light chains for the anti-CTLA4 antibody 9H10.
  • the peripheral blood and spleen were examined for the frequency of ova-specific CD8+ T cells using Kb- SIINFEKL-loaded MHC class I tetramers (Fig 6). Representative dot pots of this staining on the cells from the spleen are illustrated in Fig 6A.
  • Ova-LNPs generated an antigen specific CD8+ T cell response that was approximately 0.5% of total, and 1% of CD44hi, CD8+ T cells in the spleen (Fig 6A and 6B).
  • the addition of LNPs containing mRNA encoding for the formation of anti-CTLA4 leads to a significant increase in ova-specific T cells in both blood and spleen (Fig 6A and B) in support of an enhance antigen-specific cellular immune response.
  • T cells derived from anti-CTLA4- LNP immunized mice produced T cells with an increased affinity for antigen (see for example, quantification of this observation in Fig 6D).
  • MFI mean fluorescent intensity
  • LNP-encapsulated mRNA for the production of biologically active agonistic (Example 4) and antagonistic/blocking (Example 6) antibodies provide a proof of concept.
  • LNPs containing the heavy and light chains of monoclonal antibodies which mediate cellular depletion can be utilized.
  • CD20 e g., Rituxan
  • CD19 e.g., tafasitamab
  • VLA4 e.g., natalizumab
  • VLA4 facilitate both blockade of activated lymphocyte trafficking as well as activated lymphocyte depletion as therapeutic modalities.
  • antibodies against surface proteins expressed by tumor cells can be targeted by monoclonal antibodies (e.g., Trastuzumab or similar monoclonal antibody), causing cell death and increased immunity against the tumor.
  • monoclonal antibodies e.g., Trastuzumab or similar monoclonal antibody
  • agonistic and antagonistic antibody production via mRNA encapsulated LNPs shown above strongly support the likely success of using the same technology/methods for intervention in therapeutic modalities requiring monoclonal antibody-mediated cellular depletion.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Immunology (AREA)
  • Organic Chemistry (AREA)
  • Medicinal Chemistry (AREA)
  • Biophysics (AREA)
  • General Health & Medical Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Biochemistry (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Oncology (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Peptides Or Proteins (AREA)
  • Medicinal Preparation (AREA)

Abstract

Embodiments of the instant disclosure relate to novel complexes, compositions, and methods for generating and delivering antibodies to induce a pathway in a subject. Other embodiments relate to generating antibodies for treating a health condition or inducing a pathway for inducing a cellular immune response to treat a health condition. In accordance with these embodiments, nanoparticle complexes disclosed herein include antibody-encoding mRNA transcripts, encapsulated into a lipid nanoparticle. In other embodiments, complexes disclosed herein can be administered to a subject in combination with administration of a vaccine to a subject.

Description

COMPOSITIONS AND METHODS FOR DELIVERY AND PRODUCTION OF ANTIBODIES USING AN RNA-CONTAINING NANOPARTICLE PLATFORM
PRIORITY
[0001] This International Application claims priority to U.S. Provisional Application No. 63/292,246 filed December 21, 2021. This provisional application is incorporated herein by reference in its entirety for all purposes.
GOVERNMENT FUNDING
[0002] This invention was made with government support under grant number R21 AI171061 from the National Institutes of Health (NIH/NIAID). The government has certain rights in the invention.
FIELD
[0003] Embodiments of the instant disclosure relate to novel compositions and methods for delivering one or more mRNA encoding a target monoclonal antibody or fragment thereof, using an mRNA encapsulation platform technology. In some embodiments, mRNA encapsulation platform compositions disclosed herein can include at least one mRNA encoding a heavy chain and at least one mRN A encoding a light chain of a target or selected monoclonal antibody encapsulated into a lipid nanoparticle (LNP) where the encoded monoclonal antibody is capable of inducing at least one pathway.
BACKGROUND
[0004] Therapeutically targeted monoclonal antibodies are one of the fastest growing classes of therapeutic molecules. Immunotherapy can be used in the treatment of a variety of health conditions with improved specificity and outcoming including specifically targeted conditions (e.g., chronic inflammatory diseases, a solid tumor or other targetable cancer, a specified infectious diseases). However, current manufacturing and purification processes cause limitations in the production capacity of therapeutic antibodies and delivery, leading to an increase in cost and reduced efficiency. Also, toxicity has been associated with the high doses of intravenous antibody delivery needed to achieve a high and sustained therapeutic plasma concentration to treat a targeted condition. Therefore, new approaches to antibody production, use, targeting and delivery are needed in the field to improve specific and non- specific responses to these agents.
SUMMARY [0005] Embodiments of the instant disclosure relate to novel compositions, methods for making and methods for using and/or delivering monoclonal antibodies and/or antibody- encoding complexes including at least one mRNA encoding at least one heavy chain and at least one mRNA encoding one corresponding light chain of an antibody encapsulated in an mRNA delivery platform system. In certain embodiments, compositions disclosed herein can include one or more complex of at least one mRNA transcript encoding at least one heavy chain and at least one light chain of an antibody to a target molecule encapsulated in a nanoparticle where the antibody induces a cellular immune response, targets a pathway and/or targets a microorganism. In some embodiments, the nanoparticle can be any nanoparticle able to deliver an mRNA encoding an antibody. In other embodiments, the nanoparticle can be a lipid nanoparticle (LNP) or another suitable nanoparticle.
[0006] In certain embodiments, compositions disclosed herein can include complexes including at least one mRNA encoding at least one heavy chain and at least one mRNA encoding at least one light chain of an agonistic and/or antagonistic monoclonal antibody to a pathway, targeted molecule or cell in a nanoparticle (e.g. LNP) for inducing an immune response in a subject. In other embodiments, compositions disclosed herein can include complexes including mRNA encoding one heavy chain and at least one mRNA encoding a light chain of an agonistic or antagonistic monoclonal antibody in a nanoparticle (e.g. LNP) and at least one other agent. In other embodiments, compositions disclosed herein can include complexes having at least two mRNA transcripts that encode for at least one heavy chain of an agonistic antibody and encode for at least one light chain of the agonistic antibody. In some embodiments, compositions including complexes or nanoparticles disclosed herein can include at least two mRNA transcripts encoding an antibody such as a monoclonal antibody or fragment thereof encapsulated therein. In certain embodiments, mRNA transcripts disclosed herein can include self-replicating RNA transcripts encoding at least one light chain or heavy chain of a selected antibody.
[0007] In certain embodiments and further to paragraph [0005] above, complexes or nanoparticles disclosed herein can form part of a pharmaceutical composition that further includes at least one pharmaceutically acceptable carrier or excipient. In some embodiments, pharmaceutical compositions containing complexes such as nanoparticles disclosed herein can be administered to a subject and the nanoparticle can erode at a predetermined time to expose the molecules for encoding the agonistic and/or antagonistic antibody or fragment thereof at a site of interest, at the cellular target or systemically to a subject. In accordance with these embodiments, a full-length agonistic or antagonistic antibody can be formed in vivo after administering a pharmaceutical composition containing the antibody-encoding nanoparticles disclosed herein to the subject. In other embodiments, one or more agonistic or antagonistic antibody formed from mRNA transcripts disclosed herein can induce an immune response in a subject to a health condition to treat or prevent or reduce the onset of the health condition.
[0008] In other embodiments and further to paragraphs [0005]-[0006] above, complexes disclosed herein can further include one or more vaccines. In accordance with these embodiments, the one or more vaccines can include one or more mRNA transcripts encoding an antigen of use against a microbe or a microbial infection and/or for treating or preventing formation of a tumor. In accordance with these embodiments, improved cellular immune responses are induced in a subject receiving a vaccine disclosed herein compared to mRNA vaccines administered to a subject in the absence of administering a complex of encapsulated mRNA transcripts disclosed herein.
[0009] In some embodiments and further to paragraphs [0005]-[0007] above, agonistic or antagonistic antibodies directed to inducing an immune response in a subject can be formed in vivo from encoding mRNAs after administering a composition disclosed herein to the subject. In certain embodiments, the subject can have or is suspected of having a condition including, but not limited to, an infection, exposed to a pathogenic organism, at risk of developing an infection, having cancer, or suspected of developing cancer or other health condition in need of a therapy for inducing an immune response to treat the health condition, reduce the risk of onset or prevent the health condition. In accordance with these embodiments, a subject in need of treatment with a composition containing mRNA-containing nanoparticles disclosed herein can have or is suspected of developing cancer, a microbial infection or exposed to a pathogenic organism (e.g. virus, bacteria, fungus, prion, protozoa), an autoimmune disorder, an allograft rejection, hematopoietic disorders or other blood or blood cell disorders or any combination thereof.
[0010] In some embodiments and further to paragraphs [0005 ] -[0008] above, a full-length agonistic or antagonistic antibody formed from mRNA transcripts-containing nanoparticles disclosed herein can be a monoclonal antibody. In some embodiments, a full-length agonistic or antagonistic antibody formed from mRNA transcripts-containing nanoparticles disclosed herein can be an immune checkpoint inhibitor. In certain embodiments, a full-length agonistic or antagonistic antibody formed from mRNA transcripts-containing nanoparticles disclosed herein can be one or more of an anti-HER2 antibody, an anti-EGFR antibody, an anti-VEGRF antibody, an anti-VEGF antibody, an anti-CD-20 antibody, an anti-CD-22 antibody, an anti- CD30 antibody, an anti-CD33 antibody, an anti-CD38 antibody, an anti-CD52 antibody, an anti-CTLA-4 antibody, an anti-PD-1 antibody, an anti-PD-Ll antibody, an anti-RANKL antibody, an anti-GD2 antibody, an anti-PDGFR antibody, an anti-SLAMF7 antibody, or any combination thereof. In other embodiments, a full-length or fragment thereof of an antagonistic antibody translated from mRNA transcripts-containing nanoparticles disclosed herein can be one or more of an anti- IL-1, an anti- IL-17, an anti- IL-22, an anti- IL-23, an anti- IFNy, and an anti- TNFα or a combination thereof when introduced to a subject by encapsulated formulations blocks cellular immune responses in the subject. In other embodiments, a full-length or fragment thereof of an agonistic antibody translated from mRNA transcripts-containing nanoparticles disclosed herein can be one or more of an anti- CD137 (41BB), an anti- CD134 (0X40), an anti- CD30, an anti-CD40, an anti- CD27 or any other TNFR/L family member antibody or a combination thereof when introduced to a subject by encapsulated formulations as described herein induces or enhances immune responses (e.g., cellular immune responses) in the subject.
[0011] In some embodiments and further to paragraphs [0005]-[0009] above, a full-length agonistic antibody formed from mRNA transcripts encapsulated in nanoparticle complexes disclosed herein can encode a full-length antibody capable of targeting one or more viruses or viral proteins derived therefrom. In addition, compositions disclosed herein for including with an anti-viral vaccine can include any pathogenic virus capable of infecting a human, other mammal, reptile, or bird. In accordance with these embodiments, a virus targeted by mRNA encoding viral-derived antigen vaccine(s) or a full-length anti-viral antibody vaccine formed from encapsulating nanoparticle complexes disclosed herein can include any pathogenic vims. [0012] In some embodiments and further to paragraphs [0005]-[0010] above, a full-length agonistic antibody formed from mRNA transcripts encapsulated in nanoparticle complexes disclosed herein can encode a full-length antibody capable of targeting one or more bacteria or bacterial proteins derived therefrom. In addition, compositions disclosed herein containing nanoparticle complexes disclosed herein harboring mRNA encoding anti-bacterial vaccines can include any pathogenic bacteria capable of infecting a human, other mammal, reptile, or bird.
[0013] In some embodiments and further to paragraphs [0005]-[0011 ] above, a single nanoparticle can contain a single encoding mRNA or multiple mRNAs. In accordance with these embodiments, a single nanoparticle can include an mRNA encoding a heavy chain, light chain, or combination thereof of a targeted antibodv or fragment thereof. In certain embodiments, the antibody is a monoclonal antibody. In other embodiments, the antibody is a monoclonal antibody directed to induce or inhibit a pathway. In certain embodiments, multiple mRNA molecules can be encapsulated in single nanoparticles encoding at least one antagonist and agonist heavy chain and light chain to generate at least two monoclonal antibodies where each monoclonal antibody has antagonist or agonist activity for inducing an immune response in the subject. In certain embodiments, compositions of mixed nanoparticles are contemplated where each nanoparticle in the mix of nanoparticles encapsulates at least one mRNA encoding a heavy chain or light chain of an antibody or fragment thereof. In accordance with these embodiments, nanoparticle complexes and compositions thereof can be delivered to a subject to treat or prevent a health condition where the encoded agent (e.g., antibody or antigen or fragment thereof) can be expressed in vivo in the subject directly to a site or systemically to the subject.
[0014] In certain embodiments and further to paragraphs [0005]-[0012] above, the present disclosure includes pharmaceutical compositions including at least one nanoparticle encapsulating at least one mRNA contemplated herein. In some embodiments, pharmaceutical compositions disclosed herein can include any of the nanoparticle complexes disclosed herein and at least one pharmaceutically acceptable excipient or carrier. In other embodiments, pharmaceutical compositions disclosed herein can further include one or more additional agents for facilitating delivery or stability of the nanoparticle complexes disclosed herein. In some embodiments, other agents for improving an immune response can include one or more adjuvants or other immune-inducing agents separate from the nanoparticle complexes within the composition or as part of the one or more nanoparticle complexes such as encapsulated within the nanoparticles.
[0015] In some embodiments and further to paragraphs [0005]-[0013] above, lipid nanoparticles encapsulating one or more mRNA encoding at least a heavy and/or light chain of any contemplated cellular immune enhancing antibody disclosed herein (e.g., agonist and/or antagonistic monoclonal antibody directed to enhance or inhibit a cellular immune-related pathway) can be delivered in the same or different composition as a vaccine in order to enhance cellular immunity to treat or prevent a health condition in a subject. In certain embodiments, lipid nanoparticles disclosed herein can be delivered separate from a vaccine such as before, after or simultaneously by the same or different mode of administration as one or more vaccines. In other embodiments, lipid nanoparticles disclosed herein can contain mRNAs for encoding a heavy and/or light chain of a selected antibody or antibodies and further include a vaccine encapsulated in the same nanoparticles. In other embodiments, mixtures of nanoparticle complexes can be combined in a single composition for a mixed nanoparticle composition delivery to the subject. In some embodiments, nanoparticles encapsulating one or more mRNA encoding at least one heavy and/or light chain of at least one of an antagonist or agonist to a cellular immune induction pathway can replace the need for an adjuvant or be used in addition to an adjuvant to induce an immune response in a subj ect. [0016] In certain embodiments and further to paragraphs [0005]-[0014] above, the present disclosure provides methods of treating a subject having a health condition. In some embodiments, methods of treating a subject having a health condition disclosed herein can include administering at least one of the compositions or pharmaceutical compositions containing nanoparticles encapsulating mRNA encoding molecules disclosed herein to the subject. In accordance with these embodiments, a health condition to be treated or prevented by the methods and compositions disclosed herein can include cancer. In some embodiments, cancer can include a solid tumor. In other embodiments, a health condition to be treated or prevented by the methods and compositions disclosed herein can be an infection due to a pathogenic organism. In accordance with these embodiments, a pathogenic microorganism can include a virus, a bacteria, fungus, protozoan or prion.
[0017] In some embodiments and further to paragraphs [0005]-[0015] above, methods of administering compositions disclosed herein to a subject contemplated herein can be any suitable mode. In certain embodiments, composition can be administered as a vaccine by subcutaneous, inhalation, intraocular, intranasal administration, or other mode of administration. In some embodiments, methods of administering compositions can include localized administration directly to a tumor site in the subject.
[0018] In certain embodiments and further to paragraphs [0005]-[0016] above, the present disclosure provides kits for storing or delivering compositions or components including, but not limited to, nanoparticles encapsulating at least one mRNA encoding an antigen, whole organism, antibody or fragment thereof for use in practicing any of the methods disclosed herein. In some embodiments, kits disclosed herein can include nanoparticle complexes, compositions, pharmaceutical compositions, vaccines, or a combination thereof. In other embodiments, kits disclosed herein can further include at least one container.
BRIEF DESCRIPTION OF THE DRAWINGS
[0019] The following drawings form part of the present specification and are included to further demonstrate certain embodiments of the present disclosure. Certain embodiments can be better understood by reference to one or more of these drawings in combination with the detailed description of specific embodiments presented herein. [0020] Figs. 1A-1B illustrate graphs demonstrating T cell responses (Fig. 1A) and IgG and neutralization titers (Fig. IB) in a mammalian (e.g., human) PBMCs in response to a SARS- CoV2 mRNA vaccine in accordance with certain embodiments of the present disclosure and demonstrate reduced T cell responses while inducing a good antibody response.
[0021] Fig. 2 illustrates a schematic of mRNA in vitro transcription and lipid nano particle encapsulation in accordance with certain embodiments of the present disclosure.
[0022] Fig. 3 illustrates a graph demonstrating an antigen specific CD8+ T cell response in mice vaccinated with lipid encapsulated antibody-encoding mRNA transcripts in accordance with certain embodiments of the present disclosure.
[0023] Figs. 4A-4B illustrate images demonstrating increased antigen-specific CD8+ T cell response in wild-type mice injected alone or with increasing mRNA transcript-containing- LNPs as evaluated by dual tetramer staining of cells isolated from peripheral blood 7 days post vaccination (4A) and by quantification of CD44hi (activated) dual tetramer+ T cells (4B) in accordance with certain embodiments of the present disclosure.
[0024] Fig. 5 illustrates a graph demonstrating antitumor activity post vaccination with an anti-tumor vaccine plus a checkpoint blockade disclosed herein (e.g., anti-PDl antibody) in mice injected with B 16-ova tumor cells followed by injection of a combined vaccine treatment (e.g. polyIC/aCD40/2DG) with and without concomitant administration of a blocking antibody in accordance with certain embodiments of the present disclosure.
[0025] Figs. 6A-6D illustrate images demonstrating increased antigen-specific CD8+ T cell response in wild-type mice injected alone or with mRNA transcript-containing-LNPs encoding a cellular immune stimulating antibody and its activity evaluated by dual tetramer staining of cells isolated from peripheral blood several days post vaccination (6A) and by quantification of CD44hi (activated) dual tetramer+ T cells (6B) illustrating an enhanced specific T-cell response. 6C and 6D illustrate further specificity and quantitation of the response in the presence of the nanocapsules encapsulating the test mRNA encoded antibody in accordance with certain embodiments of the present disclosure.
DEFINITIONS
[0026] Terms, unless defined herein, have meanings as commonly understood by a person of ordinary skill in the art relevant to certain embodiments disclosed herein or as applicable.
[0027] Unless otheiwise indicated, all numbers expressing quantities of agents and/or compounds, properties such as molecular weights, reaction conditions, and as disclosed herein are contemplated as being modified in all instances by the term “about.” Accordingly, unless indicated to the contrary, the numerical parameters in the specification and claims are approximations that can vary from about 10% to about 15% plus and/or minus depending upon the desired properties sought as disclosed herein. Numerical values as represented herein inherently contain standard deviations that necessarily result from the errors found in the numerical value's testing measurements.
[0028] As used herein, “individual”, “subject”, “host”, and “patient” can be used interchangeably herein and refer to any mammalian or other contemplated subject for whom diagnosis, treatment, prophylaxis or therapy is desired, for example, humans, pets, livestock, horses, birds, reptiles, or other animals.
[0029] As used herein, “treat,” “treating” or “treatm ent” can refer to reversing, ameliorating, or inhibiting onset or inhibiting progression of a health condition or disease or a symptom of the health condition or disease.
[0030] As used herein, “polynucleotide,” “nucleic acid” or “nucleic acid molecule” can include deoxyribonucleic acid (DNA) or ribonucleic acid (RNA), messenger RNA (mRNA), oligonucleotides, and the like.
[0031] As used herein, “vector”, “expression vector” or “construct” refers to a nucleic acid component used to introduce polynucleotides into a cell having regulatory elements to provide expression of the heterologous nucleic acids in the cell. Vectors include but are not limited to plasmid, minicircles, yeast, and/or viral genomes. In some alternatives, the vectors are plasmid, minicircles, or viral genomes. In some alternatives, the vector is a viral vector. In some embodiments, the viral vector is a lentivirus. In some alternatives, the vector is a lentiviral vector. In some embodiments, the vector is a foamy viral vector, adenoviral vectors, retroviral vectors or lentiviral vectors.
[0032] As used herein, “encoding” can refer to the inherent property of specific sequences of nucleotides in a polynucleotide, such as a gene, a cDNA, or an mRNA, to serve as templates for synthesis of other polymers and macromolecules in biological processes having either a defined sequence of nucleotides (e.g., RNA, rRNA, tRNA and mRNA) or a defined sequence of amino acids and the biological properties resulting therefrom.
[0033] As used herein, “antibody,” can refer to an immunoglobulin molecule, which specifically binds to a target antigen or epitope of a target antigen, receptor or other molecule. Antibodies can be intact immunoglobulins derived from natural sources or from recombinant sources, or encoded by mRNA molecules contemplated herein and can be immunoreactive portions of intact immunoglobulins. As used herein, antibodies can also be generated from mRNA transcripts of one or more heavy and/or light chain encoding one or more full-length antibody or fragment thereon contained in a nanoparticle (e g., any biocompatible lipid nanoparticle). Antibodies are typically tetramers of immunoglobulin molecules. The antibodies in the present disclosure can exist in a variety of forms including, but not limited to polyclonal antibodies, monoclonal antibodies, Fv, Fab and F(ab)2, single chain antibodies, humanized antibodies, nanoantibodies, camelids, and the like.
[0034] As used herein, “immune response,” can refer to a process involving the activation and/or induction of an effector function in, by way of non-limiting examples, a T-cell, B-cell, natural killer (NK) cell, and/or an antigen-presenting cell (APC) response for humoral and/or cellular immunity. An immune response, can include, but is not limited to, any detectable antigen-specific activation and/or induction of a helper T cell or cytotoxic T cell activity or response, production of antibodies, antigen presenting cell activity or infiltration, macrophage activity or infiltration, neutrophil activity or infiltration, and the like. An immune response can refer to the combination of cellular immune induction and specific activation against an antigen as contemplated herein.
[0035] As used herein, “adjuvant” can refer to any molecule able to enhance an antigen- specific adaptive immune response or able to enhance an immune response when administered to a subject such as a non-specific immune response or cellular or humoral response. In certain embodiments, nanoparticle encapsulated antibodies disclosed herein can behave as an adjuvant to enhance cellular immunity in a subject in need thereof.
[0036] As used herein, “autologous” refers to cells derived from the patient. “Allogenic” refers to cells derived from the same species but having a different genotype. “Syngeneic” refers to cells from a genetically identical source, such as a twin, hence immunologically compatible or so closely related that transplantation does not provoke an immune response.
DETAILED DESCRIPTION OF THE INVENTION
[0037] In the following sections, certain exemplary compositions and methods are described in order to detail certain embodiments of the invention. It will be obvious to one skilled in the art that practicing the certain embodiments does not require the employment of all or even some of the specific details outlined herein, but rather that concentrations, times and other specific details can be modified through routine experimentation. In some cases, well known methods, or components have not been included in the description.
[0038] Embodiments of the instant disclosure relate to novel compositions, formulations, complexes, methods for making and methods for delivering monoclonal antibodies and/or nanoparticle complexes including at least one rnRNA encoding at least one of a heavy or light chain of an antibody as an encapsulated mRNA delivery platform. In certain embodiments, compositions disclosed herein can include one or more nanoparticle complex of at least one mRNA transcript encoding at least one heavy chain and at least one light chain of an antibody to a target molecule encapsulated in a nanoparticle where the antibody induces a cellular immune response and/or targets a microorganism in a subject. In some embodiments, nanoparticles can be any nanoparticle able to encapsulate and deliver an mRNA encoding an antibody or fragment thereof. In other embodiments, nanoparticles can include a lipid nanoparticle (LNP) or other suitable nanoparticle capable of encapsulating mRNA encoding an antibody or fragment thereof, reducing degradation of the mRNA, and permitting translation of the antibody or fragment thereof at the site and/or timing of interest.
[0039] In certain embodiments and further to paragraph [0037] above, complexes, compositions, and methods disclosed herein are designed to encapsulate, stabilize, transport, and generate antibodies or antibody fragments at a site of interest. In some embodiments, complexes disclosed herein include, but are not limited to, mRNA transcripts encapsulated in a nanoparticle including, but not limited to, a transcript encoding a heavy chain and a transcript encoding a light chain for generating an antibody of interest for example full-length antibodies, antigen binding fragments of full-length antibodies, Fab fragments, single chain antibodies (scFv), diabodies, triabodies, mini bodies, nanobodies, single-domain antibodies, camelids, or any combination thereof. In certain embodiments, the antibody or antibody fragment thereof is an agonistic or antagonistic antibody directed to a cellular immune regulating pathway or component thereof.
[0040] In some embodiments and further to paragraphs [0037]-[0038] above, complexes disclosed herein can include nanoparticles encapsulating one or more mRNA transcripts. In certain embodiment, the nanoparticles can include lipid nanoparticles (LNPs; ionizable lipid nanoparticles for example). In some embodiments, LNPs contemplated herein can encapsulate at least two or more mRNA transcripts encoding at least one targeted antibody. In other embodiments, nanoparticles (e.g., LNPs) disclosed herein can encapsulate one, two, three, four or more mRNA (e.g., 1, 2, 3, 4, 5, 6, 7, 8 or more) transcripts encoding at least one heavy or light chain to form a nanoparticle encapsulated complex. In some embodiments, nanoparticle complexes disclosed herein can include nanoparticles encapsulating one, two, three, four or more mRNA transcripts encoding at least one heavy chain and at least one light chain of an antibody. In other embodiments, complexes disclosed herein can include nanoparticles encapsulating two mRNA transcripts encoding at least one heavy chain and at least one light chain of a single antibody and can further be mixed with complexes encoding at least one heavy chain and at least one light chain of another antibody directed to the same or a different target. In some embodiments, the encoded antibody is an antagonistic antibody of a targeted cellular response pathway. In other embodiments, the encoded antibody is an agonistic antibody of a targeted cellular response pathway. In certain embodiments, nanoparticle complexes disclosed herein include mRNA transcripts for encoding a heavy and a light chain of an agonistic antibody and/or a heavy and a light chain of an antagonistic antibody to a targeted cellular immune pathway. In some embodiments, a target can be a cellular immune promoting target or a cellular immune inhibiting target where the antibody binds to the target and induces cellular immunity in the subject. In certain embodiments, mRNAs encoding heavy and light chain sequences of an agonistic or antagonistic antibody targeting an immunologically relevant pathway can be encapsulated in ionizable lipid nanoparticle (LNP), the components of which in part, facilitate the fusion of the LNP to an endosomal membrane, allowing access of nanoparticle constituents (the mRNA) to flow out of the nanoparticle into cytoplasm of the cell in order to form, in vivo, the agonistic or antagonistic antibody of interest. [0041] In certain embodiments and further to paragraphs [0037]-[0039] above, encapsulated mRNA transcripts disclosed herein can encode for a full-length polypeptide or peptide fragment thereof. In some embodiments, mRNA transcripts disclosed herein can encode for an antibody. In other embodiments, mRNA transcripts disclosed herein can encode for an antibody where the antibody can be a chimeric immunoglobulin. In certain embodiments, mRNA transcripts disclosed herein can encode for an antibody where the antibody can be partially or fully humanized antibody. In other embodiments, mRNA transcripts disclosed herein can encode for an antibody where the antibody is a human immunoglobulin or other mammal. In some embodiments, mRNA transcripts disclosed herein can encode for a full-length antibody or an antibody fragment. In certain embodiments, mRNA transcripts disclosed herein can encode for a single chain immunoglobulin. In other embodiments, mRNA transcripts disclosed herein can encode for at least one IgGl antibody, an IgG2 antibody, an IgG3 antibody, an IgG4 antibody, or any combination thereof. In other embodiments, mRNA transcripts disclosed herein can encode for a heavy chain of an antibody. In some embodiments, mRNA transcripts disclosed herein can encode for a light chain of an antibody. In certain embodiments, mRNA transcripts of nanoparticle complexes disclosed herein can include a heavy chain to light chain (H:L) ratio in these complexes in a 1 : 1; a 1.5: 1 ; a 2: 1; a 2.5: 1 ; 3: 1; 3.5: 1 ; 4: 1; 4.5: 1 or other suitable ratio of H:L. In some embodiments, the ratio of mRNA transcript encoding a heavy chain antibody is about two times the number of mRNA transcripts encoding the light chain of a target antibody or target antibodies (e.g., an anti-CD40 antibody or other cellular immune pathway inducing antibody). [0042] In certain embodiments and further to paragraphs [0037]-[0040] above, compositions disclosed herein can include nanoparticle complexes including at least one mRNA encoding at least one heavy chain and at least one mRNA encoding at least one light chain of an agonistic or antagonistic monoclonal antibody in a single nanoparticle for inducing a cellular immune response in a subject. In other embodiments, compositions disclosed herein can include nanoparticle complexes including mRNA encoding one heavy chain and at least one mRNA encoding a light chain of an agonistic or antagonistic monoclonal antibody in one or more LNPs. In other embodiments, compositions disclosed herein can include complexes having at least two mRNA transcripts encoding a heavy chain of an agonistic antibody, and at least one of the at least two mRNA transcripts can encode for a light chain of the agonistic antibody for use in inducing cellular immunity. In some embodiments, compositions including complexes disclosed herein can include at least two mRNA transcripts encoding an antibody or fragment thereof encapsulated in an LNP. In certain embodiments, nanoparticles disclosed herein can further include one or more vaccine for treating or preventing onset of a health condition.
[0043] In some embodiments and further to paragraphs [0037]-[0041] above, lipid nanoparticles encapsulating one or more mRNA encoding at least one heavy and/or light chain of any contemplated cellular immune enhancing antibody disclosed herein (e.g., agonist and/or antagonistic monoclonal antibody directed to enhance or inhibit a cellular immune-related pathway) can be delivered in the same or different composition as a vaccine in order to enhance cellular immunity to treat or prevent a health condition in a subject. In certain embodiments, lipid nanoparticles disclosed herein can be delivered separate from a vaccine such as before, after, or simultaneously by the same or different mode of administration as one or more vaccines. In other embodiments, lipid nanoparticles disclosed herein can contain mRNAs for encoding a heavy and/or light chain of one or more selected antibody or antibodies and further include a vaccine encapsulated in the same nanoparticles. In other embodiments, mixtures of nanoparticle complexes can be combined in a single composition for a mixed nanoparticle composition delivery to the subject. In some embodiments, nanoparticles encapsulating one or more mRNA encoding at least one heavy and/or light chain of at least one of an antagonist or agonist to a cellular immune induction pathway can replace the need for an adjuvant or be used in addition to an adjuvant to induce an immune response in a subj ect. In certain embodiments, a standard adjuvant such as alum or other agent can be mixed in or supplied separately from nanoparticle complexes disclosed herein and optionally, can further include a vaccine. [0044] In some embodiments and further to paragraphs [0037]-[0042], complexes including at least two mRNA transcripts for encoding a full-length antibody capable of inducing a cellular immune response can further be formulated with complexes including at least one mRNA that encodes an antigen (e.g. an mRNA vaccine against a pathogen or tumor) in the same or separate complexes in the same or separate nanoparticles. In certain embodiments, nanoparticles can be mixed and administered in a single dose, or multiple doses. In certain embodiments, the dose of nanoparticle complexes can be about 1.0 μg/kg to about 300 μg/kg, or about 5.0 μg/kg to about 250 μg/kg or about 10.0 μg/kg to about 200 μg/kg, or about 20.0 μg/kg to about 150 μg/kg. In other embodiments, nanoparticles encoding antibodies contemplated herein and nanoparticles encoding an antigen of use as an mRNA vaccine can be administered separately, at the same time, staggered or at different times depending on the effect. In certain embodiments, nanoparticles encoding antibodies contemplated herein and nanoparticles encoding an antigen of use as an mRNA vaccine can be part of a single nanoparticle complex or a mixture of nanoparticles harboring different mRNA transcripts encoding antibodies to the same or different cellular-immune enhancing targets. In some embodiments, one or more than one dose of nanop article complexes encoding antibodies disclosed herein can be administered to the subject. In accordance with these embodiments, administration can include more than once a day, daily, every other day, bi- weekly, weekly, bi-monthly, monthly, every other month, together with or at the time of administering a vaccine (or before or after) or other suitable dosing regimen.
[0045] In some embodiments and further to paragraphs [0037]-[0043] above, mRNA encoding the at least one mRNA transcript encoding a heavy chain and a light chain of the antibody. In certain embodiments, the at least one mRNA encoding the at least one antigen can further include one or more of incorporation of 1 methyl-pseudouridine (ImUp); at least one SerineUCG (SerUCG) codon substitution having at least one ImUp; and an mRNA sequence that remains unstructured between about the last 40 to 55 nucleotides of the 5’UTR and about the first 20 to 40 nucleotides (nts) of the coding sequence, followed by increased secondary structure thereafter for improved stability and translation of the mRNA transcript. In certain embodiments, the one or more mRNA transcript can include an mRNA sequence that remains unstructured between about the last 47 nucleotides of the 5’UTR and about the first 30 nucleotides of the coding sequence, followed by increased secondary' structure thereafter for improved stability and translation of the mRNA transcript. In some embodiments, modifications to the mRNA transcripts disclosed herein can reduce degradation of the transcripts and improve translation of a target antibody.
[0046] In certain embodiments and further to paragraphs [0037]-[0045] above, complexes disclosed herein can form part of a pharmaceutical composition further including at least one pharmaceutically acceptable carrier. In some embodiments, pharmaceutical compositions containing nanoparticle complexes contemplated herein can be administered to a subject and the nanoparticle can erode at a predetermined time to allow exposure of the mRNA transcripts for encoding the agonistic or antagonistic antibody or fragment thereof at a site of interest or systemically to a subject. In accordance with these embodiments, a full-length agonistic or antagonistic antibody can be formed in vivo after administering an mRNA transcript containing nanoparticle complex-containing composition disclosed herein to the subject. In other embodiments, one or more agonistic or antagonistic antibody formed from mRNA transcripts disclosed herein can induce a cellular immune response in the subject to treat a health condition in the subject or enhance treatment of the subject or prevent or reduce onset of a condition in the subject. In other embodiments, nanoparticle complexes disclosed herein can further include one or more mRNA vaccines or other vaccines. In accordance with these embodiments, the one or more mRNA vaccines can include an mRNA vaccine having one or more mRNA transcripts encoding an antigen of use against a microbe or a microbial infection and/or for treating or preventing formation of a tumor. In accordance with these embodiments, improved cellular immune responses are induced in a subject receiving nanoparticle complexes encoding an antagonistic or agonistic antibody disclosed herein compared to vaccines or mRNA vaccines administered to a subject in the absence of administering a nanoparticle complex of encapsulated mRNA transcripts encoding an antagonistic or agonistic antibody disclosed herein directed to a cellular immune-related system (e.g., anti-CD40 antibody or anti-CTLA4 equivalent thereof).
[0047] In certain embodiments and further to paragraphs [0037]-[0046] above, mRNA encoding an antigen as part of an mRNA vaccine-containing nanoparticle can further include one or more of an alpha globin 5'-UTR, an amino-terminal enhancer of split (AES) motif, and a mitochondrial 12S RNA element. In certain embodiments, mRNA encoding an antigen as part of an mRNA vaccine-containing nanoparticle includes at least 2 of an alpha globin 5'- UTR, an amino-terminal enhancer of split (AES) motif, and a mitochondrial 12S RNA element for improved stabil ity and translation of the mRNA as well as improved translation. In certain embodiments, these mRNA vaccine-containing nanoparticles further include at least one mRNA encoding a light chain and/or at least one mRNA encoding a heavy chain in the same or different nanoparticle complexes. In accordance with these embodiments, the nanoparticles include lipid nanoparticles such as ionizing lipid nanoparticles or other nanoparticles capable of encapsulating mRNA for delivery and cytosolic dispersion permitting formation of fully functional antibodies.
[0048] In some embodiments and further to paragraphs [0037]-[0047] above, agonistic or antagonistic antibodies directed to inducing a cellular immune response can be formed in vivo after administering a composition herein to a subject in need thereof, where a subject in need thereof can have or is suspected of having a condition including, but not limited to, an infection, exposed to a pathogenic organism, at risk of an infection, cancer or other health condition in need of a therapy for inducing an immune response to treat the condition. In accordance with these embodiments, a subject in need of treatment with a composition containing complexes disclosed herein can have or is suspected of developing at least one of cancer, a microbial infection or exposed to a pathogenic microbe (e.g. virus, bacteria, fungus, prion, protozoa), an autoimmune disorder, an allograft rejection, hematopoietic or hematologic disorders or any combination thereof.
[0049] In some embodiments and further to paragraphs [0037] -[0048] above, a full-length agonistic or antagonistic antibody formed from mRNA transcripts encapsulated in nanoparticle complexes disclosed herein can be a monoclonal antibody. In some embodiments, a full-length agonistic or antagonistic antibody formed from mRNA transcripts encapsulated in nanoparticle complexes disclosed herein can be an immune checkpoint inhibitor antibody. In accordance with these embodiments, an immune checkpoint inhibitor antibody can include an antibody encoded by mRNA transcripts disclosed herein that reduces or blocks proteins referred to as checkpoints that are made by certain immune system cells, such as T cells, NK cells and some cancer cells. In certain embodiments, checkpoints assist in a healthy individual by preventing the immune responses from being too strong and managing the immune response. In certain settings, checkpoints can reduce or interfere with T cells from killing cancer cells or fighting infection. In accordance with these embodiments, when these checkpoints are blocked or inhibited, T cells can kill cancer cells better and can fight infection more effectively. In certain embodiments, antibodies encoded by mRNA transcripts disclosed herein can include antibodies that bind to and inhibit checkpoint molecules from inhibiting or interfering with cellular immune responses. In some embodiments, examples of checkpoint proteins found on T cells or cancer cells can include, but are not limited to, PD-1, PD-L1, CTLA-4, B7-1 and B7-2. In certain embodiments, immune checkpoint inhibitors can be used to treat cancer In other embodiments, a full-length agonistic or antagonistic antibody formed from mRNA transcripts encapsulated in nanoparticle complexes disclosed herein can include, but are not limited to, one or more of an anti-HER2 antibody, an anti-EGFR antibody, an anti- VEGRF antibody, an anti-VEGF antibody, an anti-CD-20 antibody, an anti-CD-22 antibody, an anti-CD30 antibody, an anti-CD33 antibody, an anti-CD38 antibody, an anti-CD52 antibody, an anti-CTLA-4 antibody, an anti-PD-1 antibody, an anti-PD-Ll antibody, an anti- 137- 1, an anti-B7-2, an anti-RANKL antibody, an anti-GD2 antibody, an anti-PDGFR antibody, an anti-SLAMF7 antibody, or any combination thereof. In other embodiments, a full-length or fragment thereof of an antagonistic antibody translated from mRNA transcript complexes disclosed herein can include, but is not limited to, one or more of an anti-IL-1, an anti-IL-17, an anti-IL-22, an anti-IL-23, an anti-IFNy, and an anti-TNFa or a combination thereof that when introduced to a subject by encapsulated mRNA transcript-containing formulations disclosed herein blocks cellular immune responses in the subject. In other embodiments, a full-length or fragment thereof of an agonistic antibody translated from mRNA transcript complexes disclosed herein can be one or more of an anti-CD137 (4 IBB), an anti- CD134 (0X40), an anti-CD30, an anti-CD40 antibody, and an anti- CD27 or other TNFR/L family member or a combination thereof when introduced to a subject by encapsulated formulations induces or enhances cellular immune responses in the subject.
[0050] In some embodiments and further to paragraphs [0037] -[0049] above, a full-length agonistic antibody formed from mRNA transcripts of lipid nanoparticle complexes disclosed herein can be a full-length antibody or fragment thereof (e.g., a heavy and a light chain encoding mRNA or at least one antibody) capable of targeting one or more viruses or viral proteins derived therefrom. In addition, nanoparticle-containing compositions disclosed herein for including with an anti-viral vaccine can include a vaccine against any pathogenic virus capable of infecting a human, other mammal, reptile, or bird. In accordance with these embodiments, a virus targeted by an mRNA antigen vaccine or a full-length agonistic antibody formed from complexes disclosed herein can include, but is not limited to, Adenoviridae, Alphaviridae, Alphaviridea, Anelloviridae, Anelloviridae, Archiviridea, Arenaviridae, Arenaviridae, Arteriviridae, Astroviridae, Astroviridae, Bornaviridae, Bunyaviridae, Bunyaviridae, Caliciviridae, Caulimoviridae, Coronaviridae, Filoviridae, Flaviviridae, Hepadnaviridae, Hepeviridae, Herpesviridae, influenza virus, Lyssavirus, Orthobunyavirus, Orthomyxoviridae, Papillomaviridae, Papovaviridae, Paramyxoviridae, Paramyxoviridae, Parvoviridae, Picornaviridae, Pleolipoviridae, Pneumoviridae, Polyomaviridae, Polyomavirus, Poxviridae, Poxviridae, Reoviridae, Retroviridae, Rhabdoviridae, Seadornavirus, Togaviridae or species, or sub-species thereof or a combination thereof or other pathogenic virus. Any pathogenic virus is contemplated to be targeted by compositions and methods disclosed herein.
[0051] In some embodiments and further to paragraphs [0037]-[0049] above, a full-length agonistic antibody formed from mRNA transcripts of lipid nanoparticle complexes disclosed herein can be a full-length antibody or fragment thereof (e.g., a heavy and a light chain encoding mRNA or at least one antibody) capable of targeting one or more bacteria or bacterial proteins derived therefrom. In other embodiments, nanoparticle-containing compositions disclosed herein can be included with an anti-bacterial vaccine that can include any vaccine against a pathogenic bacteria capable of infecting a human, other mammal, reptile, or bird. In accordance with these embodiments, a bacteria targeted by an mRNA antigen vaccine or a full-length agonistic antibody formed from complexes disclosed herein can include, but is not limited to, Alcaligenes spp. : Bacillus spp. ; Bacteriodes spp. : Bartonella spp. ; Bordetella spp. ; Borrelia spp.; Brevundimonas spp.; Brucella spp.; Burkholderia spp.; Burkholderia spp.; Campylobacter spp.; Candida spp.; Chlamydia spp.; Citrobacter spp.; Clostridium spp. ; Clostridioides spp. ; Corynebacterium spp. ; Coxiella spp. ; Enter obacter spp. ; Enterobius spp. ; Escherichia coli spp. (incl. EHEC, EPEC, ETEC, EIEC, EAEC, ESBL/MRGN, DAEC) Francisella tularensis spp., Haemophilus influenzae spp.; Helicobacter spp.; Klebsiella spp.; Leclercia spp. ; Legionella spp. ; Leishmania spp. ; Leptospira spp. ; I^euconostoc spp. ; Listeria spp. ; Micrococcus spp. ; Microsporum spp. ; Moraxella spp. ; Morganella spp. Mycobacterium, spp.; Mycoplasma spp.; Neisseria spp.; Orientia spp.; Pantoea spp.; Paracoccus spp.; Prevotella spp.; Propionibacterium spp.; Proteus spp.; Proteus spp.; Providencia spp.; Pseudomonas spp.; Ralstonia spp.; Rickettsia spp.; Roseomonas spp.; Salmonella spp.; Shigella spp.; Sphingomonas spp.; Staphylococcus spp.; Stenotrophomonas spp.; Streptococcus spp.; Treponema spp.; Vibrio spp.; Yersinia spp. or species thereof or sub- species thereof or a combination thereof or other pathogenic bacteria.
[0052] In certain embodiments and further to paragraphs [0037]-[0051] above, the present disclosure includes pharmaceutical compositions. In some embodiments, pharmaceutical compositions disclosed herein can include any of the nanoparticle-containing complexes disclosed herein and at least one pharmaceutically acceptable excipient or carrier. In some embodiments, pharmaceutical compositions disclosed herein can further include one or more additional agents. In accordance with these embodiments, one or more additional agents suitable for inclusion into pharmaceutical compositions disclosed herein can include one or more more adjuvants or an anti-cancer or anti-microbial agent. [0053] In certain embodiments and further to paragraphs [0037] - [0052] above, the present disclosure provides methods of treating a subject having a health condition. In some embodiments, methods of treating a subject having a health condition disclosed herein can include administering at least one of the compositions or pharmaceutical compositions disclosed herein to the subject. In accordance with these embodiments, a health condition to be treated by the methods and compositions disclosed herein can include cancer. In some embodiments, cancer can include a solid tumor. In some embodiments, the solid tumor can include, but is not limited to, a lung, liver, kidney, lymph node, breast, prostate, esophageal, stomach, skin, bone, intestinal, uterine or other solid tumor. In some embodiments, a health condition to be treated by the methods and compositions disclosed herein can be a microbial infection. In certain embodiments, a health condition to be treated by the methods and compositions disclosed herein can be due to a pathogenic microorganism and can include a virus, a bacterium, fungus, protozoan or prion.
[0054] In some embodiments and further to paragraphs [0037]-[0053 ] above, methods of administering compositions disclosed herein to a subject contemplated herein can be any suitable mode. In certain embodiments, methods of administering compositions disclosed herein to a subject can include subcutaneous, intranasal, by inhalation, intraocular, intravenous, or other mode of administration at the time of, before or after administering a vaccine by the same or different mode of administration or together as a single composition or as a single composition of nanoparticles containing all agents to be delivered to the subject. In some embodiments, methods of administering compositions can include localized administration directly to a tumor site in the subject. In other embodiments, modes of administration of pharmaceutical compositions disclosed herein can include, at least one of, intravenously, intramuscularly, intranasally, by aerosol, intradermally, transdermally, topically, intrauterine, or intrarectally administering the pharmaceutical composition or other suitable mode of administration to the subject. In some embodiments, nanoparticle complexes encoding one or more antibodies can be administered by the same route as nanoparticle complexes encoding an antigen, for example a vaccine formulation.
[0055] In certain embodiments disclosed herein and further to paragraphs [0037]-[0054] above, nanoparticle-containing complex compositions encapsulating mRNA transcripts for encoding an agonist or antagonist can be formulated and then stored at reduced temperatures for later administration. In some embodiments, storage temperatures for compositions disclosed herein can be about -10 or less and stored for about 1 week to several months. In other embodiments, storage temperatures of ionizing lipid nanoparticle-encapsulated mRNA transcripts disclosed herein can be about -20 or colder for about 2 weeks to about 2 months or more.
[0056] In certain embodiments and further to paragraphs [0037]-[0055] above, expression vectors encoding one or more of the mRNA transcripts disclosed herein are contemplated. In some embodiments, vectors can include one or more conventional control elements which are operably linked to the mRNA-encoding sequence in a manner which permits its transcription, translation and/or expression in a cell. As used herein, operably linked sequences include both expression control sequences that are contiguous with the mRNA-encoding sequence and expression control sequences that act in trans or at a distance to control the polynucleotide- encoding sequence. Expression control sequences can include appropriate transcription initiation, termination, promoter and enhancer sequences; efficient RNA processing signals such as splicing and polyadenylation (polyA) signals; sequences that stabilize cytoplasmic mRNA; sequences that enhance translation efficiency (e.g, Kozak consensus sequence); sequences that enhance protein stability; and when desired, sequences that enhance secretion of the encoded product. A number of expression control sequences, including promoters which are native, constitutive, inducible and/or tissue-specific, are known in the art. and can be utilized herein. In some embodiments, a vector encoding a heavy chain can be different than the vector encoding the light chain in order to for example,
[0057] In certain embodiments and further to paragraphs [0037]-[0056] above, LNP complexes (e.g., ionizing lipid nanoparticles) disclosed herein can encapsulate mRNA transcripts for encoding any antibody disclosed herein. In other embodiments, LNP complexes disclosed herein can encapsulate expression vectors encoding one or more mRNA transcripts herein. In some embodiments, LNP complexes disclosed herein can encapsulate at least one mRNA transcript encoding for a light chain of an antibody and at least one mRNA transcript encoding a heavy chain of an antibody. In some embodiments, LNP complexes disclosed herein can encapsulate one or more expression vectors encoding at least one mRNA transcript for a heavy chain of an antibody and one or more expression vectors encoding at least one mRNA transcript for a light chain of an antibody.
[0058] In some embodiments and further to paragraphs [0037]-[0057] above, LNP complexes disclosed herein can encapsulate expression vectors encoding a mRNA transcript for a heavy chain of an antibody and expression vectors encoding a mRNA transcript for a light chain of an antibody in a stoichiometric ratio. In some embodiments, LNP complexes disclosed herein can encapsulate one expression vector encoding a mRNA transcript for a heavy chain of an antibody and one expression vector encoding a mRNA transcript for a light chain of an antibody (e.g., 1 heavy chain transcript to 1 light chain transcript). In some embodiments, LNP complexes disclosed herein can encapsulate two expression vectors encoding a mRNA transcript for a heavy chain of an antibody and one expression vector encoding a mRNA transcript for a light chain of an antibody (e.g., 2 heavy chain transcripts to 1 light chain transcript). In some embodiments, LNP complexes disclosed herein can encapsulate four expression vectors encoding a mRNA transcript for a heavy chain of an antibody and one expression vector encoding an mRNA transcript for a light chain of an antibody (e.g., 4 heavy chain transcripts to 1 light chain transcript). In some embodiments, LNP complexes disclosed herein can encapsulate six expression vectors encoding a mRNA transcript for a heavy chain of an antibody and one expression vector encoding a mRNA transcript for a light chain of an antibody (e.g., 6 heavy chain transcripts to 1 light chain transcript). In some embodiments, LNP complexes disclosed herein can encapsulate eight expression vectors encoding an mRNA transcript for a heavy chain of an antibody and one expression vector encoding an mRNA transcript for a light chain of an antibody (e.g., 8 heavy chain transcripts to 1 light chain transcript). In some embodiments, LNP complexes disclosed herein can encapsulate ten expression vectors encoding an mRN A transcript for a heavy chain of an antibody and one expression vector encoding a mRNA transcript for a light chain of an antibody (e.g., 10 heavy chain transcripts to 1 light chain transcript).
[0059] In some embodiments and further to paragraphs [0037]-[0058] above, LNP complexes disclosed herein can encapsulate expression vectors encoding an mRNA transcript for a heavy chain of an antibody and expression vectors encoding an mRNA transcript for a light chain of an antibody wherein a full-length antibody can be formed after administering the composition into a cell or a tissue of a subject. In some embodiments, a full-length antibody formed herein can be a monoclonal antibody against a target molecule or an antigen. [0060] In some embodiments and further to paragraphs [0037] -[0060] above, a full-length antibody formed disclosed herein can be an agonistic or antagonistic antibody. In some embodiments, a full-length antibody formed herein can be an immune checkpoint inhibitor where an antibody translated from encapsulated mRNA transcripts disclosed herein can bind to or associate with the immune checkpoint inhibitor and modulate its activity. In certain embodiments, an antibody translated from encapsulated mRNA transcripts disclosed herein can bind to or associate with the immune checkpoint inhibitor and partially or completely block the immune checkpoint inhibitor’s activity. In some embodiments, an antibody translated from encapsulated mRNA transcripts disclosed herein can bind to or associate with the immune checkpoint inhibitor and reversibly inhibit activity or regulate its activity for a pre-determined period. In certain embodiments, an antibody translated from encapsulated mRNA transcripts disclosed herein can bind to, or associate with an immunologically relevant molecule in an agonistic mode and stimulate or modulate lymphocyte activity for use to treat a health condition. In some embodiments, a full-length antibody can include, but is not limited to, an anti-HER2 antibody, an anti-EGFR antibody, an anti-VEGRF antibody, an anti-VEGF antibody, an anti-CD-20 antibody, an anti-CD-22 antibody, an anti-CD30 antibody, an anti- CD33 antibody, an anti-CD38 antibody, an anti-CD40 antibody, an anti-CD52 antibody, an anti-CTLA-4 antibody, an anti-PD-1 antibody, an anti-PD-Ll antibody, an anti-RANKL antibody, an anti-GD2 antibody, an anti-PDGFR antibody, an anti-SLAMF7 antibody, or any combination thereof.
[0061] In certain embodiments and further to paragraphs [0037]-[0060] above, expression vectors encoding mRNA transcripts herein can be formulated into lipid nanoparticles (LNPs) for delivery into cells, tissues, and/or a subject. LNPs are spherical vesicles made of ionizable lipids, which can be positively charged at low pH (enabling RNA complexation) and neutral at physiological pH (reducing potential toxic effects, as compared with positively charged lipids, such as liposomes). LNPs can be taken up by cells via endocytosis, and the ionizability of the lipids at low pH (likely) can enable endosomal escape, which allows release of the cargo into the cytoplasm.
[0062] In some embodiments and further to paragraphs [0037]-[0061 ] above, LNPs for use in embodiments disclosed herein can contain ionizable cationic lipids, non-cationic lipids, sterols, and/or PEG lipids components along with expression vectors encoding mRNA transcripts disclosed herein. In some embodiments, LNPs disclosed herein can have about 5- 25% non-cationic lipid. In some embodiments, LNP nanoparticle complexes disclosed herein can have about 5-20%, 5-15%, 5-10%, 10-25%, 10-20%, 10-25%, 15-25%, 15-20%, or 20-25% non-cationic lipid. Non-limiting examples of non-cationic lipids include distearoylphosphatidylcholine (DSPC), dioleoylphosphatidylcholine (DOPC), dipalmitoylphosphatidylcholine (DPPC), dioleoylphosphatidylglycerol (DOPG), dipalmitoylphosphatidylglycerol (DPPG), ioleoylphosphatidylethanolamine (DOPE), palmitoyloleoylphosphatidylcholine (POPC), palmitoyloleoyl-phosphatidylethanolamine (POPE), dipalmitoyl phosphatidyl ethanolamine (DPPE), dimyristoylphosphoethanolamine (DMPE), distearoyl-phosphatidyl-ethanolamine (DSPE), sphingolipids, cerebrosides, gangliosides, 16-O-monomethyl PE, 16-O-diraethyl P, 18-1 -trans PE, 1 -stearoyl-2-oleoyl- phosphatidyethanolamine (SOPE), and the like. In certain embodiments, mRNA transcripts disclosed herein can be encapsulated in ionizable lipid nanoparticles (LNP), the components of which facilitate fusion of the LNPs to an endosomal membrane, allowing access of nanoparticle constituents (e.g., the mRNA) to the cytoplasm of a targeted cell taking up the LNP permitting in vivo formation of the antibody or antibody fragment encoded by the mRNA cargo. In some embodiments, mRNA transcripts disclosed herein can be encapsulated in DSPC, also referred to as l,2-distearoyl-sn-glycero-3 -phosphocholine, cholesterol, also referred to as Distearoylphosphatidylcholine or other similar lipid known in the art and administered to a subject alone or in combinations disclosed herein for in vivo synthesis of one or more encoded antibody or fragment thereof in the treatment of a health condition disclosed herein. In yet other embodiments, mRNA transcripts disclosed herein can be encapsulated in DMG-PEG2000, also referred to as l,2-dimyristoyl-rac-glycero-3- methoxypolyethylene glycol-2000 or other similar lipid known in the art and administered to a subject alone or in combinations disclosed herein for in vivo synthesis of one or more encoded antibody or fragment thereof in the treatment of a health condition disclosed herein. [0063] In some embodiments and further to paragraphs [0037]-[0063] above, LNPs can contain a helper lipid to promote cell binding, cholesterol to fill the gaps between the lipids, and a polyethylene glycol (PEG) to reduce opsonization by serum proteins and reticuloendothelial clearance. In some embodiments, LNPs for use herein can include one or more synthetic ionizable phospholipids provided herein and at least one helper lipid. In some embodiments, LNPs for use herein can include one or more synthetic ionizable phospholipids provided herein and at least one helper lipid selected from: l,2-dioleoyl-sn-glycero-3- phosphoethanol amine (DOPE), N-methyldioctadecylamine (MDOA), 1,2-dioleoyl-3- dimethylammonium-propane (DODAP), dimethyldioctadecylammonium bromide salt (DDAB), l,2-dioleoyl-3-trimethylammonium-propane (DOTAP), and any combination thereof. In some embodiments, LNPs for use herein can include one or more synthetic ionizable phospholipids provided herein and at least one zwitterionic helper lipid (e.g., DOPE), ionizable cationic helper lipid (e.g., MDOA, DODAP), permanently cationic helper lipid (e.g., DDAB, DOTAP), or any combination thereof. In some embodiments, LNPs of use herein concern LNPs that permit optimal transport and expression of mRNAs encoding antibodies disclosed herein in order to induce or induce an enhanced cellular immune response in a subject.
[0064] In some embodiments and further to paragraphs [0037]-[0063] above, LNPs for use in complexes disclosed herein can include one or more synthetic ionizable phospholipids provided herein and at least one cholesterol and/or a cholesterol derivative. As used herein, “cholesterol derivative” refers to any compound consisting essentially of a cholesterol structure, including additions, substitutions and/or deletions thereof. The term cholesterol derivative herein can also include steroid hormones and bile acids as are generally recognized in the art. Non-limiting examples of cholesterol derivatives suitable for use herein can include dihydrocholesterol, ent-cholesterol, epi-cholesterol, desmosterol, cholestanol, cholestanone, cholestenone, cholesteryl-2 '-hydroxy ethyl ether, cholesteryl-4'-hydroxybutyl ether, 3β-[N- (N'N'-dimethylaminoethyl)carbamoyl cholesterol (DC-Chol), 24(S)-hydroxycholesterol, 25- hydroxycholesterol, 25(R)-27-hydroxycholesterol, 22-oxacholesterol, 23-oxacholesterol, 24- oxacholesterol, cycloartenol, 22-ketosterol, 20-hydroxysterol, 7 -hydroxy cholesterol, 19- hydroxy cholesterol, 22-hydroxy cholesterol, 25-hydroxy cholesterol, 7-dehydrocholesterol, 5a-cholest-7-en-3β-ol, 3,6,9-trioxaoctan-l-ol-cholesteryl-3e-ol, dehydroergosterol, dehydroepiandrosterone, lanosterol, dihydrolanosterol, lanostenol, lumisterol, sitocalciferol, calcipotriol, coprostanol, chol ecalciferol, lupeol, ergocalciferol, 22-dihydroegocalciferol, ergosterol, brassicasterol, tomatidine, tomatine, ursolic acid, cholic acid, chenodeoxycholic acid, zymosterol, diosgenin, fucosterol, fecosterol, or fecosterol, or a salt or ester thereof.
[0065] In some embodiments and further to paragraphs [0037]-[0064] above, LNPs for use in certain complexes disclosed herein can include one or more synthetic ionizable phospholipids provided herein and at least one PEG or PEG-modified lipids. As used herein, a PEG-modified lipid, or “PEG lipid” refers to a lipid modified with polyethylene glycol (PEG). Such species can be alternately referred to as PEGylated lipids. Non-limiting examples of PEG-modified lipids suitable for use herein can include PEG-modified phosphatidylethanol amines, PEG-modified phosphatidic acids, PEG-modified ceramides (PEG-CER), PEG-modified dialkylamines, PEG-modified diacylglycerols (PEG-DAG), PEG-modified dialkylglycerols, and mixtures thereof. For example, but not limited to, a PEG- modified lipid for use herein can be PEG-c-DOMG (R-3-[(co- methoxypoly(ethyleneglycol)2000)carbamoyl]-l,2-dimyristyloxy-propyl-3-amine poly(ethylene glycol)); PEG-DMG (l,2-dimyristoyl-rac-glycero-3-methoxypolyethylene glycol poly(ethylene glycol)); PEG-DLPE (l,2-Dilauroyl-sn-glycero-3 -phosphorylglycerol sodium salt-poly(ethylene glycol)); PEG-DMPE (dimethyl-2- (dimethylphosphino)ethylphosphine-poly(ethylene glycol)); PEG-DPPC (1,2-dipalmitoyl-sn- glycero-3-phosphocholine-poly(ethylene glycol)); PEG-DSPE (1, 2-distearoyl-sn-glycero-3- phosphoethanolamine-poly(ethylene glycol)); PEG-DPPE (1, 2-dipalmitoyl-sn-glycero-3- phosphoethanolamine-N-[monomethoxy poly(ethylene glycol)), and the like.
[0066] In some embodiments and further to paragraphs [0037]-[0065] above, a PEG- modified lipid for uses disclosed herein can include a PEG moiety having a size of from about 1000 daltons to about 20,000 daltons. In some embodiments, a PEG-modified lipid for use can include a PEG moiety having a size of about 1000 daltons, about 2000 daltons, about 5000 daltons, about 10,000 daltons, about 15,000 daltons, or about 20,000 daltons. In some embodiments, LNPs herein can include one or more synthetic ionizable phospholipids provided herein and at least one PEG or PEG-modified lipids wherein the PEG moiety can have a size of about 2000 daltons. Examples of useful PEG-lipids for use in making the LNPs described herein include, but are not limited to, l,2-Diacyl-sn-Glycero-3- Phosphoethanolamine-N-[Methoxy(Poly ethylene glycol)-350] (mPEG 350 PE); 1,2-Diacyl- sn-Glycero-3-Phosphoethanolamine-N-[Methoxy(Polyethylene glycol)-550] (mPEG 550 PE); l,2-Diacyl-sn-Glycero-3-Phosphoethanolamine-N-[Methoxy(Poly ethylene glycol)-750] (mPEG 750 PE); l,2-Diacyl-sn-Glycero-3-Phosphoethanolamine-N-[Methoxy(Poly ethylene glycol)-1000] (mPEG 1000 PE); l,2-Diacyl-sn-Glycero-3-Phosphoethanolamine-N- [Methoxy (Poly ethylene glycol)-2000] (mPEG 2000 PE); l,2-Diacyl-sn-Glycero-3- Phosphoethanolamine-N-[Methoxy(Polyethylene glycol)-3000] (mPEG 3000 PE); 1,2- Diacyl-sn-Glycero-3-Phosphoethanolamine-N-[Methoxy(Poly ethylene glycol)-5000] (mPEG 5000 PE); N-Acyl-Sphingosine-l-[Succinyl(Methoxy Polyethylene Glycol) 750] (mPEG 750 Ceramide); N-Acyl-Sphingosine-l-[Succinyl(Methoxy Polyethylene Glycol) 2000] (mPEG 2000 Ceramide); and N-Acyl-Sphingosine-1 -[Succinyl (Methoxy Polyethylene Glycol) 5000] (mPEG 5000 Ceramide). In some embodiments, LNPs herein can include one or more synthetic ionizable phospholipids provided herein and l,2-dimyristoyl-rac-glycero-3- methoxy(poly(ethylene glycol-2000)) (DMG-PEG2000).
[0067] In some embodiments and further to paragraphs [0037]-[0066] above, LNP complexes disclosed herein can include one or more agents to target one or more cell types. In some embodiments, LNP complexes can be selective for one or more cell types. In some embodiments, LNP complexes can unload cargo at one or more selective cell types. In some embodiments, LNP complexes can selectively target one or more cell types. In some embodiments, LNP complexes can selectively target one or more tissue types. In some embodiments, LNP complexes disclosed herein can selectively target one or more cancers.
[0068] It is understood by one of skill in the relevant art that LNP size can impact behavior of lipid nanoparticles in vivo. In some embodiments, LNP complexes disclosed herein can be about 20 nm to about 1000 nm in diameter or size. In some embodiments, LNP complexes disclosed herein can be about 20 nm to about 200 nm in size. In some embodiments, LNP complexes disclosed herein can about 20 nm to about 190 nm or about 25 nm to about 190 nm in size. In some embodiments, LNP complexes disclosed herein can be about 30 nm to about 180 nm in size. In some embodiments, LNP complexes disclosed herein can be about 35 nm to about 170 nm in size. In some embodiments, LNP complexes disclosed herein can be about 40 nm to about 160 nm in size. In some embodiments, LNPs herein can be about 50 nm to about 150 nm, about 60 nm to about 140 nm, about 70 nm to about 130 nm, about 80 nm to about 120 nm, or about 90 nm to about 110 nm in size. In some embodiments, LNP complexes disclosed herein can be about 20 nm, about 25 nm, about 30 nm, about 35 nm, about 40 nm, about 45 nm, about 50 nm, about 55 nm, about 60 nm, about 65 nm, about 70 nm, about 75 nm, about 80 nm, about 85 nm, about 90 nm, about 95 nm, about 100 nm, about 105 nm, about 110 nm, about 115 nm, about 120 nm, about 125 nm, about 130 nm, about 135 nm, about 140 nm, about 145 nm, about 150 nm, about 155 nm, about 160 nm, about 165 nm, about 170 nm, about 175 nm, about 180 nm, about 185 nm, about 190 nm, about 195 nm, or about 200 nm in size or diameter.
[0069] In some embodiments and further to paragraphs [0037]-[0068] above, an average LNP size in a LNP complex-containing composition disclosed herein can be about 20 nm to about 1000 nm (e.g., about 20, 40, 60, 80, 100, 150, 200, 250, 300, 350, 400, 450, 500, 550, 600, 650, 700, 750, 800, 850, 900, 950, 1000) in diameter in average size. In some embodiments, LNP size in a LNP composition herein can be homogenous at about 20 nm to about 1000 nm (e.g., about 20, 40, 60, 80, 100, 150, 200, 250, 300, 350, 400, 450, 500, 550, 600, 650, 700, 750, 800, 850, 900, 950, 1000) in diameter in size. In some embodiments, LNP size in a LNP composition herein be heterogeneous at about 20 nm to about 1000 nm (e.g., about 20, 40, 60, 80, 100, 150, 200, 250, 300, 350, 400, 450, 500, 550, 600, 650, 700, 750, 800, 850, 900, 950, 1000) in diameter in average size. In some embodiments, LNP size in a LNP composition herein can be heterogeneous wherein about 50% to about 99% of the LNPs average at about 20 nm to about 1000 nm (e.g., about 20, 40, 60, 80, 100, 150, 200, 250, 300, 350, 400, 450, 500, 550, 600, 650, 700, 750, 800, 850, 900, 950, 1000) in diameter in average size.
[0070] In certain embodiments and further to paragraphs [0037]-[0069] above, pharmaceutical compositions are contemplated. In accordance with these embodiments, pharmaceutical compositions can include one or more of the LNPs encapsulating mRNA transcripts disclosed herein. In some embodiments, pharmaceutical compositions herein can include one or more of the LNPs encapsulating mRNA transcripts disclosed herein and at least one pharmaceutically acceptable excipient or carrier. As used herein, the term “pharmaceutically acceptable” refers to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues, organs, and/or bodily fluids of a subject without excessive toxicity, irritation, allergic response, or other problems or complications commensurate with a reasonable benefit/risk ratio. As used herein, the term “pharmaceutically acceptable carrier” can refer to solvents, dispersion media, coatings, antibacterial agents, antifungal agents, isotonic and absorption delaying agents, or the like that are physiologically compatible. Pharmaceutically acceptable carriers suitable for use herein, include, but are not limited to, buffers that are well known in the art, and can be phosphate, citrate, and other organic acids; antioxidants including ascorbic acid and methionine; preservatives; low molecular weight polypeptides; proteins, such as serum albumin, gelatin, or immunoglobulins; amino acids; hydrophobic polymers; monosaccharides; disaccharides; and other carbohydrates; metal complexes; and/or non-ionic surfactants.
[0071] In some embodiments and further to paragraphs [0037]-[0070] above, pharmaceutical compositions for uses disclosed herein can be formulated for parenteral administration, such as intravenous or intravascular, bolus infusion, intrarenal introduction, intracerebroventricular injection, intra-ci sterna magna injection, intra-parenchymal injection, or a combination thereof. In some embodiments, pharmaceutical compositions for use herein can be formulated for infusion of LNP complexes encapsulating mRNA transcripts into a tumor. In some embodiments, pharmaceutical compositions for use herein be formulated for parenteral/ infusion administration can include pharmaceutically acceptable carriers including sterile liquids, such as water and oil, including those of petroleum, animal, vegetable or synthetic origin, such as peanut oil, soybean oil, mineral oil, and the like. Saline solutions and aqueous dextrose, polyethylene glycol (PEG) and glycerol solutions can also be employed as liquid carriers, particularly for injectable solutions. In some embodiments, pharmaceutical compositions for use herein can further include additional ingredients, for example preservatives, buffers, tonicity agents, antioxidants and stabilizers, nonionic wetting or clarifying agents, viscosity -increasing agents, and the like. In some embodiments, pharmaceutical compositions described herein can be packaged in single unit dosages or in multi -dosage forms.
[0072] In some embodiments and further to paragraphs [0037]-[0071] above, formulations suitable for parenteral/infusion administration include aqueous and non-aqueous sterile injection solutions which can contain anti-oxidants, buffers, bacteriostats and solutes which render the formulation isotonic with the blood of the intended recipient; and aqueous and non- aqueous sterile suspensions which can include suspending agents and thickening agents. In accordance with some embodiments herein, aqueous solutions can be suitably buffered (e.g., a pH of from about 3.0 to about 9.0). The preparation of suitable parenteral/infusion formulations for use herein under sterile conditions can be readily accomplished by standard pharmaceutical techniques well known to those skilled in the art.
[0073] In some embodiments and further to paragraphs [0037]-[0072] above, pharmaceutical compositions herein can further include one or more pharmaceutically acceptable salts. Non-limiting examples of pharmaceutically acceptable salts include acid addition salts (formed from a free amino group of a polypeptide with an inorganic acid, or an organic acid. In some embodiments, the salt formed with the free carboxyl groups is derived from an inorganic base, or an organic base. In some embodiments, any of the pharmaceutical compositions herein can be used in therapeutic applications, for example, treating a disease and/or a disorder in human patients, which are disclosed herein. In some embodiments, any of the pharmaceutical compositions herein can be used in therapeutic applications, for example, treating a cancer, a viral infection, a bacterial infection, an autoimmune disorder, an allograft rejection, or any combination thereof in human patients, which are also disclosed herein.
[0074] In certain embodiments and further to paragraphs [0037]-[0073] above, methods of treating or ameliorating a health condition and/or a disorder in a subject are disclosed. In some embodiments, methods of treating or ameliorating a health condition and/or a disorder in a subject include, but are not limited to, administration of an effective amount of any the LNP complexes encapsulating mRNA transcripts and/or pharmaceutical compositions containing these LNP complexes thereof as described herein. “An effective amount” as used herein refers to a dose of LNP complexes encapsulating mRNA transcripts that is sufficient to confer a therapeutic effect on a subject having or suspected of having a disease and/or a disorder herein. In certain embodiments, a therapeutic effect for a subject having or suspected of having a disease and/or a disorder herein can include reducing the symptoms or consequences of the disease.
[0075] In some embodiments and further to paragraphs [0037]-[0074] above, methods of administering LNP complexes encapsulating mRNA transcripts as disclosed herein can include placement (e.g., transplantation or implantation) of any the LNP complex encapsulating mRNA transcripts and/or pharmaceutical compositions containing these LNP complexes into a subject, by a method or route that results in at least partial localization of the introduced LNP at a desired site, such as a tumor, such that a desired effect(s) is produced. In some embodiments, a subject can be transfused with LNP complexes encapsulating mRNA transcripts disclosed herein over the course of a day, for a few hours, daily, every other day, 2 times per week, weekly, every other week, monthly, or other appropriate treatment regimen. In accordance with the embodiments herein, the period of full-length antibodies generated after administration of LNPs encapsulating mRNA transcripts herein to a subject can be a few hours (e.g, about 2 hours, about 6 hours, about 12 hours, about 24 hours), a few days (e.g., about 1 day, about 2 days, about 3 days, about 4 days about 5 days, about 6 days, about 7 days), weeks (e.g., about 2 weeks, about 4 weeks, about 6 weeks, about 12 weeks, about 40 weeks, about 52 weeks), to as long as several years (e.g., about 2 years, about 5 years), or even the life time of the subject. In some embodiments, an effective amount of the LNP complex encapsulating mRNA transcripts herein can be administered via a systemic route of administration, such as an intraperitoneal or intravenous route. In certain embodiments, cellular immunity can be assessed in a subject before, during or after being treated by complexes disclosed herein in order to assess level of induction and in order to design a regimen tailored to treating the subject.
[0076] In some embodiments and further to paragraphs [0037]-[0075] above, a subject to any of the methods disclosed herein can be any subject for whom treatment or therapy is desired. In some embodiments, a subject to any of the methods disclosed herein can be any subject having or is suspected of having a health condition such as cancer or an infection or exposed to a pathogen or a disorder in need of treatment with a full-length antibody generated after administration of LNP complex encapsulating mRNA transcripts herein to induce cellular immunity in the subject. In some embodiments, a subject to any of the methods herein can be any subject having or is suspected of having a cancer, a viral infection, a bacterial infection, an autoimmune disorder, an allograft rejection, or any combination thereof. In some embodiments, a subject can be a mammal. In some embodiments, a subject can be a human patient. In some embodiments, a human subject such as an adult, child, adolescent, toddler, young adult or infant or fetus who is in need of the methods herein can be identified by routine medical examination, e.g., laboratory tests, biopsy, magnetic resonance imaging (MRI) scans, ultrasound exams, and the like. In other embodiments, a mammal can include a pet, livestock, a horse, or other mammal can be analyzed and assessed for need of a treatment disclosed herein. In other embodiments, vaccines applicable to any subject population can be administered at the same time, before, or after nanoparticle complex compositions disclosed herein encoding at least one antibody for inducing a cellular immune response are administered to the subject of the subject population (e.g., herd of livestock, horses or group of people) in order to enhance the desired response of the vaccine. [0077] In some embodiments and further to paragraphs [0037]-[0076] above, a subject to any of the methods herein can be any subject having or is suspected of developing cancer, a solid tumor, a metastasis, or a combination thereof. In some embodiments, tumors treated by the methods herein can be a solid tumor, a liquid or non-solid tumor. In accordance with these embodiments, a solid tumor can include, but is not limited to, breast, lung, brain, liver, kidney, eye, skin, stomach, esophageal, prostate, uterine or another solid tumor. In some embodiments, a solid tumor can be a sarcoma. In some embodiments, tumors treated by the methods herein can be a bone sarcoma, a soft-tissue sarcoma, or any combination thereof. In some embodiments, adenocarcinomas are contemplated. In some embodiments, GI tract carcinomas are contemplated for treatment herein. In some embodiments, cancers and/or resulting tumors thereof that can be treated by the methods herein can be osteosarcoma, chondrosarcoma, poorly differentiated round/spindle cell tumors, Ewing sarcoma, hemangioendothelioma, angiosarcoma, fibrosarcoma/myofibrosarcoma, chordoma, adamantinoma, liposarcoma, leiomyosarcoma, malignant peripheral nerve sheath tumor, rhabdomyosarcoma, synovial sarcoma, malignant solitary fibrous tumor, or any combination thereof. In some embodiments, tumors treated by the methods herein can be liposarcoma, atypical lipomatous tumor, dermatofibrosarcoma protuberans, malignant solitary fibrous tumor, inflammatory myofibroblastic tumor, low-grade myofibroblastic sarcoma, fibrosarcoma, myxofibrosarcoma, low-grade fibromyxoid sarcoma, giant cell tumor of soft tissues, leiomyosarcoma, malignant glomus tumor, rhabdomyosarcoma, hemangioendothelioma, angiosarcoma of soft tissue, extraskeletal osteosarcoma, gastrointestinal stromal tumor, malignant, malignant peripheral nerve sheath tumor, malignant Triton tumor, malignant granular cell tumor, malignant ossifying fibromyxoid tumor, stromal sarcoma not otherwise specified, myoepithelial carcinoma, malignant phosphaturic mesenchymal tumor, synovial sarcoma, epithelioid sarcoma, alveolar soft part sarcoma, clear cell sarcoma of soft tissue, extraskeletal myxoid chondrosarcoma, extraskeletal Ewing sarcoma, desmoplastic small round cell tumor, extrarenal rhabdoid tumor, perivascular epithelioid cell tumor, intimal sarcoma, undifferentiated spindle cell sarcoma, undifferentiated pleomorphic sarcoma, undifferentiated round cell sarcoma, undifferentiated epithelioid sarcoma, undifferentiated sarcoma, not otherwise specified, or any combination thereof.
[0078] In some embodiments and further to paragraphs [0037]-[0077] above, methods of administrating LNP complexes encapsulating mRNA transcripts as disclosed herein can reduce growth of solid tumors and/or solid tumor cells. In some embodiments, methods of administrating LNP complexes encapsulating mRNA transcripts as disclosed herein can reduce tumor size of solid tumors. In some embodiments, methods of administrating LNP complexes encapsulating mRNA transcripts as disclosed herein can induce apoptosis of solid tumors and solid tumor cells. In some embodiments, methods of administrating LNP complexes encapsulating mRNA transcripts as disclosed herein can attenuate tumor growth and/or tumor expansion and/or induce tumor cell killing. In some embodiments, methods of administrating LNP complexes encapsulating mRNA transcripts as disclosed herein can reduce the risk of metastasis.
[0079] In some embodiments and further to paragraphs [0037]-[0078] above, a subject to any of the methods disclosed herein can be any subject having or is suspected of developing an infection or exposed to a microorganism or suspected of being exposed to a microorganism. In some embodiments, a subject to any of the methods disclosed herein can be any subject having or is suspected of developing an infection resulting from an infectious microorganism such as a pathogenic microorganism including, but not limited to a virus, a bacteria, fungus, prion, protozoan or a combination thereof. In some embodiments, a subject to any of the methods disclosed herein can be any subject having or is suspected of developing or having been exposed to a virus or having an infection resulting from a virus. In some embodiments, administrating LNP complexes encapsulating mRNA transcripts as disclosed can generate at least one antibody that can target one or more proteins on surface of a virus. In some embodiments, administrating LNPs encapsulating mRNA transcripts as disclosed can generate at least one antibody that can target one or more proteins on surface of a virus belonging to the Adenoviridae, Alphaviridae, Alphaviridea, Anelloviridae, Anelloviridae, Archiviridea, Arenaviridae, Arenaviridae, Arteriviridae, Astroviridae, Astroviridae, Bornaviridae, Bunyaviridae, Bunyaviridae, Caliciviridae, Caidimoviridae, Coronaviridae, Filoviridae, Flaviviridae, Hepadnaviridae, Hepeviridae, Herpesviridae, Lyssavirus, Orthobunyavirus, Orthomyxoviridae, Papillomaviridae, Papovaviridae, Paramyxoviridae, Paramyxoviridae, Parvoviridae, Picornaviridae, Pleolipoviridae, Pneumoviridae, Polyomaviridae, Polyomavirus, Poxviridae, Poxviridae, Reoviridae, Retroviridae, Rhabdoviridae, Seadornavirus, Togaviridae, family, other pathogenic virus family or combination thereof. In other embodiments, mRNA vaccines against one or more pathogenic viruses can be mixed with one or more nanoparticles encoding at least one immune checkpoint inhibitor antibody or separately administered to a subject in need thereof.
[0080] In certain embodiments and further to paragraphs [0037]-[0079] above, LNP complexes encapsulating mRNA transcripts and/or pharmaceutical compositions disclosed herein can be administered in dosages and by techniques well known to those skilled in the medical and veterinary arts. In accordance with these embodiments, healthcare professions will take into consideration such factors as the age, sex, weight, and condition of the particular patient, and the composition form used for administration (e.g., solid vs. liquid). Dosages for humans or other mammals can be determined without undue experimentation by the skilled artisan, from this disclosure, and the knowledge in the art.
[0081] In some embodiments and further to paragraphs [0037]-[0080] above, kits are contemplated of use to generate, store and/or transport the LNP complexes encapsulating mRNA transcripts disclosed herein. In some embodiments, kits can include LNP complexes encapsulating mRN A transcripts where the LNP complexes can be used immediately or frozen and stored for transport and later use. In some embodiments, a kit disclosed herein can include any of vectors encoding mRNA transcripts disclosed herein, LNP complexes encapsulating mRNA transcripts and/or pharmaceutical compositions disclosed herein. In other embodiments, kits can further include a target mRNA vaccine of use for inducing an immune response against a specific pathogenic agent or tumor.
[0082] In some embodiments and further to paragraphs [0037]-[0080] above, kits are provided for use in treating or alleviating a targeted disease or condition treatable by use of LNP complexes encapsulating mRNA transcripts disclosed herein. In some embodiments, kits can include instructions for use in accordance with any of the methods described herein. The included instructions can include a description of administration of any the LNP complexes encapsulating mRNA transcripts and/or pharmaceutical compositions described herein and optionally one or more additional therapies to treat, delay the onset, or alleviate a target disease (e.g., cancer, infection) as those described herein. In some embodiments, kits herein can further include a description of selecting an individual suitable for treatment based on identifying whether that individual has the target disease, e.g., applying one or more diagnostic methods. In some embodiments, the instructions can include a description of administering LNP complexes encapsulating mRNA transcripts to a subj ect at risk of the target disease.
[0083] In other embodiments, based on verification and proof of concept disclosed herein regarding in vivo synthesis of antibody or antibody fragment-encoding mRNA encapsulated nanoparticle complexes disclosed herein, compositions and methods for targeted cellular depletion are contemplated to also be attainable by these methods. In this disclosure, use of LNP-encapsulated mRNA for the production of biol ogically active agonistic and biologically active antagonistic/blocking antibodies have been demonstrated. In certain embodiments, LNPs containing the heavy and light chains of monoclonal antibodies which mediate targeted cellular depletion can be utilized. In one embodiment, antibodies against CD20 and/or CD 19 can be encoded by LNP encapsulation of raRNAs for production of anti-CD20 and/or anti- CD19 antibodies for efficacious use against B cell lymphomas based on their capacity to induce B-cell directed depletion. In another embodiment, antibodies against molecules such as VLA4 can facilitate both blockade of activated lymphocyte trafficking as well as activated lymphocyte depletion as therapeutic modalities using nanoparticle complexes disclosed herein for in vivo delivery and synthesis of anti-VLA4 antibodies or a similar target for directed cellular depletion. In accordance with these embodiments, antibody production via mRNA encapsulated LNPs disclosed herein can be used for intervention in therapeutic modalities requiring monoclonal antibody-mediated cellular depletion.
[0084] In some embodiments and further to paragraphs [0037]-[0083] above, kits can include instructions for using the components of the kit, for example relating to the use of LNP complexes encapsulating mRNA transcripts. Instructions for using the components of the kit can generally include information as to dosage, dosing schedule, and/or route of administration for the intended treatment. In some embodiments, containers can be unit doses, bulk packages {e.g., multi-dose packages) or sub-unit doses. In some embodiments, instructions supplied in the kits of the invention can be written instructions on a label or package insert (e.g, a paper sheet included in the kit), but machine-readable instructions (e.g, instructions carried on a magnetic or optical storage disk) are also acceptable.
[0085] In certain embodiments and further to paragraph [0084] above, a label or package insert herein can indicate that the composition is used for treating, delaying the onset and/or alleviating the disease (e.g., cancer, infection). Instructions can be provided for practicing any of the methods described herein.
[0086] In certain embodiments and further to paragraphs [0084]-[0085] above, kits disclosed herein include suitable packaging. Suitable packaging includes, but is not limited to, syringes, vials, bottles, jars, flexible packaging (e.g., sealed Mylar or plastic bags), and the like. Also contemplated herein are packages for use in combination with a specific device, such as an inhaler, nasal administration device (e.g, an atomizer) or an infusion device such as a minipump. A kit can have a sterile access port (for example the container can be an intravenous solution bag or a vial having a stopper pierceable by a hypodermic injection needle). The container can also have a sterile access port (for example the container can be an intravenous solution bag or a vial having a stopper pierceable by a hypodermic injection needle). In some embodiments, at least one active agent in the composition can include an antibody generated by LNPs encapsulating mRNA transcripts disclosed herein. EXAMPLES
[0087] The following examples are included to illustrate certain embodiments. It should be appreciated by those of skill in the art that the techniques disclosed in the examples which follow represent techniques discovered to function in the practice of the claimed methods, compositions, and apparatus. However, those of skill in the art should, in light of the present disclosure, appreciate that changes can be made to certain examples or some embodiments which are disclosed and still obtain a like or similar result without departing from the spirit and scope of the invention.
Example 1
[0088] Commercially available representatives of a lipid particle-encapsulated mRNA vaccine platform include Modema’s and Pfizer/BioNtech’s (“PBN”) SARS-CoV2 mRNA vaccines. While both Modema and PBN vaccines induce humoral immunity and some consistent and detectable cellular responses, improved cellular response for overall improved immunity are needed in order to provide more robust responses to these vaccines and other vaccines. For example, T cell responses (e.g. both CD4 and CD8 positive T cell response) are low in magnitude when compared to humoral immunity and when compared to other cellular immunity inducing compositions.
[0089] In one exemplary method, T cell responses, IgG, and neutralization titers in response to the Moderna SARS-CoV2 mRNA vaccine were assessed. CD4 and CD8+ T cell responses were measured 60 days post-boost using an activation-induced marker (AIM) assay. AIM assays sensitively and specifically detected antigen-responsive CD4+ and CD8+ T cells after vaccination. In brief, human PBMCs were collected and isolated by density gradient centrifugation. Cells were then suspended in FBS containing 10% dimethyl sulfoxide and cryopreserved in liquid nitrogen. Cryopreserved PBMCs were thawed and cultured in serum- free AIM-V media overnight for 18 hours at 37°C. PBMCs were then aliquoted into wells of a 96-well plate, at a total of 1 x 106 cells per well. PBMCs were stimulated with 10 pg/ml heat- inactivated antibiotic-killed Group A Streptococcus, strain 5448 (“Ag stimulation”). As a positive control, cells were stimulated with 1 ug/ml SEB (“SEB stimulation”). PMBC aliquots were incubated at 37°C for 18 hours, after which supernatants were harvested and cells were analyzed by flow cytometry.
[0090] Serum antibody response was measured and used to assess immunogenicity. In this example, a functional neutralizing antibody test was used as a serological assay to detect the amount of antibody that can effectively neutralize virus, preventing infectivity in vitro. Fifty- percent (50%) Focus Reduction Neutralization — FNRT50 - assays, which uses immunostaining to visualize “plaques” referred to as foci, were performed on human PMBC prepared as described herein. Similarly, anti-receptor binding domain (RBD) IgG titers were also assessed on human PMBCs.
[0091] Data demonstrated that CD4+ and CD8+ responses averaged around 1-300 cells per million PBMCs (~0.1-0.5% of total CD8s) (Fig. 1A). In contrast, total RBD-specific IgG and neutralizing antibody titers observed close to the same timepoint for the same donor were superior by orders of magnitude (Fig. IB). These data demonstrated a disproportionate favoring of the humoral response by adjuvanted subunit vaccines in comparison to cellular immune responses.
[0092] With minimal instigation, a robust antibody response was achieved with mobilization of about 10-20,000 plasmablasts, resulting in milligrams of antibody produced by the hour. In sharp contrast to this “action at a distance” mechanism of the antibody response, the biology of protection regarding cytotoxic T cells can be compared to hand-to-hand combat wherein the engagement of overwhelming numbers was a critical determinant of victory. Reasonable estimates derived from animal studies indicated the frequency of CD8+ T cells necessary to promote therapeutic efficacy against an established tumor or infection is about 106-107 T cells per cm3 of tissue. This is a targeted number that had not be demonstrated as achieved by any subunit vaccine platform (e.g., a lipid nanoparticle (LNP)-encapsulated mRNA platform as disclosed herein). An additional intervention or approach was needed in the field for improved application and outcome of mRNA vaccine technology into clinical areas to render a robust T cell immunity for improved therapeutic efficacy.
Example 2
[0093] In another exemplary method, T cell responses to a model antigen were assessed in mice. Fig. 2 illustrates a schematic diagram demonstrating a method developed herein by which specific mRNA sequences could be transcribed in vitro and packaged into LNP for in vivo administration to generate a target molecule.
Example 3
[0094] In one exemplary example of these methods, the gene encoding ovalbumin (ova) antigen was designed and synthesized with an alpha globin 5'-UTR, an amino-terminal enhancer of split (AES) motif, and mitochondrial 12S RNA elements. mRNA was produced by T7 RNA polymerase transcription from linearized plasmid templates with co-transcriptional ARCA capping and post-transcriptional polyadenylation. Column-purified mRNA transcripts were incorporated into LNPs composed of Precision Nanosystem's Neuro9 lipid mix (catalog#: NWS0009) on a microfluidic LNP mixer (e.g., Precision NanoSystems Spark). RNA content of LNPs was quantified using a dye-binding assay following detergent-based dissolution of the LNPs and LNPs at concentrations of 8-15 ng/μl were ultrafiltered for in vivo administration. After validating in vitro mRNA translation using mRNA encoding GFP, escalating amounts of ova injected LNPs were administered to wild type B6 mice and 7 days later the spleen was examined for the frequency of ova-specific CD8+ T cells using Kb-SIINFEKL -loaded MHC class I tetramers. While all doses of ova-LNPs produced a detectable population of ova-specific T cells, the use of 1-2 μg LNP resulted in the most consistent frequency of tetramer positive T cells at -0.2-1% (Fig. 3).
Example 4
[0095] In one exemplary method, use of LNP-encapsulated mRNA for the delivery of heavy and light chains of an agonistic antibody targeting an immunologically relevant pathway, LNP-mRNA-elicited CD8+ T cell responses to delivery7 of these constructs encoding for an agonistic anti-CD40 antibody (FGK45) was assessed. Given the success of the combined TLR/IFN + CD40 vaccine adjuvant for generating robust T cells responses, it was investigated whether similar responses might be achieved through delivery of LNP containing mRNA transcripts encoding heavy and light chain antibody sequences derived from an anti-CD40 agonist, FGK45. Because mRNA vaccines induce a strong IFN signature shortly after injection, without wishing to be bound by theory, it was reasoned that this might alleviate the need to additionally co-deliver a TLR agonist with the LNP construct administration. Constructs encoding heavy and light chain mRNA sequences of FGK45 were assembled, mRNA transcription performed, and the heavy and light chain mRNAs co-encapsulated into lipid nanoparticles (FGK-LNP) as described above for ova. One pg of ova LNPs were inj ected alone or with increasing FGK-LNPs into wild-type (WT) B6 mice and the ova-specific CD8+ T cell response was evaluated by dual tetramer staining cells isolated from peripheral blood 7 days post vaccination.
[0096] A detectable response was observed following administration of the ova-LNP alone (Fig. 4A). However, after co-administration of increasing doses of the FGK-LNP (the anti- CD40 antibody construct), the intensity of ova specific T cells was >6 fold higher than in response to ova-LNP alone (Figs. 4A and 4B). These data indicated that not only were both heavy and light chain genes for FGK effectively translated following LNP administration, but functional anti-CD40 antibody (FGK) was formed and able to induce a cellular immune response by superi or induction of a CD8+ T cell response.
Considerations
[0097] H/L chain ratio. Formation of functional antibody requires the heterodimeric assembly of heavy and light chain protein sequences within the ER. Given the differences in length between the heavy and light chain sequences (L chain mRNA sequence is roughly half as long as H), L protein may be in excess when a uniform molarity between the two sequences is contained within the LNP. H and L proteins unable to form heterodimeric pairs are unstable and besides being degraded by proteosomes, may also trigger the Unfolded Protein Response (UPR). The UPR is important for the production and secretion of antibody from plasma cells but may or may not be beneficial for antibody production and secretion from the muscle and/or parenchymal cells producing protein following LNP-mRNA injection. In this example, ultimate production of fully functional antibody can be enhanced by altering the HZL mRNA ratio within the LNP. Four different HZL molar ratios will be evaluated (111, 2: 1, 4: 1, 6: 1,8: 1 respectively) using the optimized mRNA template/transcription method disclosed herein. Optimized HZL ratios, based on the evaluation of cDCl IL-27-GFP production and the magnitude of the CD8 T cell response, were determined as disclosed herein. It is projected that the ratio H/L chain of at least 2: 1 will have an improved production of antibody than a 1 : 1 ratio. [0098] Separate vs co-delivery of LNP components. Some exemplary methods disclosed herein can utilize antigen and FGK separately packaged into LNPs and then mixed prior to injection in vivo. Certain theories regarding how LNP-mRNAs best generate T cell responses were considered: 1) LNP-mRNAs can be taken up by muscle/ stromal cells which produce and secrete the encoded protein. The protein is then taken up by professional APCs which migrate to the local lymphoid tissue and initiate the ensuing T cell response; and 2) LNP-mRNAs are taken up directly by professional APCs which incorporate the expressed antigen into the class I presentation pathway. The APC-intrinsic adjuvant effect of the LNP (e.g., the encoded anti- CD40 stimulus) drives optimal APC maturation, migration and initiation of the T cell response. While real biology is almost certainly mixture of both models, when 1) predominates, co- packaging of antigen and FGK into the same LNPs is not necessary. When 2) predominates, co-packaging is demonstrably better for DC activation and CD8+ T cell responses generation. Accordingly, antigen and optimized FGK mRNA are separately or jointly incorporated into LNPs. Dose escalating injections are utilized and cDCl IL-27-GFP production and the magnitude of the CD8 T cell response are used for selecting the preferred method.
Example 5
[0099] In another exemplary method, to highlight the potential of LNP-mRNA for delivery of anti-CTLA4 and anti-PD1-mediated immunotherapy, ovalbumin-expressing tumor cell lines were evaluated as an anti -turmor cell model. Ovalbumin-expressing tumor cell lines are a useful tool in the validation of any anti-tumor therapeutic intervention. The discovery that tumor responsiveness to immunotherapy tracks well with the mutational burden in the tumor has reinvigorated the clinical relevance of these model antigen-expressing tumors under the guise of “neo-antigen” representation. To demonstrate effectiveness of the model, antitumor activity of vaccination plus checkpoint blockade was measured using B16 melanoma tumor cells in the ova model. In brief, B16-ova was injected S.C. and 7 days later the mice were immunized against ovalbumin using a novel combined vaccine treatment (polyIC/aCD40/2DG), with and without concomitant administration of anti-PDl blocking antibody. Tumor growth was monitored over time bay measuring tumor area up to 25 days post-implantation. Fig. 5 illustrates exemplary' antitumor activity measured in the model for the three treatment regimens tested herein.
Example 6
[0100] In one exemplary method, use of LNP-encapsulated mRNA to produce an antibody capable of blocking CTLA4 (9H10), immunologically relevant "immune checkpoint" pathway was investigated. CleanCap AG capped mRNA, including Nl -methylpseudouridine-5'- triphosphate substitutions, was produced by T7 RNA polymerase transcription from linearized plasmid templates. Column-purified mRNA transcripts were incorporated into LNPs composed of Precision Nanosystem's Neuro9 lipid mix (catalog#: NWS0009) on a microfluidic LNP mixer (e.g., Precision NanoSystems Spark). RNA content of LNPs was quantified using a dye- binding assay following detergent-based dissolution of the LNPs and LNPs at concentrations of 50-80 ng/ul were ultrafiltered for in vivo administration. Ova-LNPs containing lug of mRNA were either injected into B6 mice alone or in combination with LNPs containing 2 ug of heavy and light chains for the anti-CTLA4 antibody 9H10. Ten days later, the peripheral blood and spleen were examined for the frequency of ova-specific CD8+ T cells using Kb- SIINFEKL-loaded MHC class I tetramers (Fig 6). Representative dot pots of this staining on the cells from the spleen are illustrated in Fig 6A. Ova-LNPs generated an antigen specific CD8+ T cell response that was approximately 0.5% of total, and 1% of CD44hi, CD8+ T cells in the spleen (Fig 6A and 6B). The addition of LNPs containing mRNA encoding for the formation of anti-CTLA4 leads to a significant increase in ova-specific T cells in both blood and spleen (Fig 6A and B) in support of an enhance antigen-specific cellular immune response. Further, by analyzing the tetramer mean fluorescent intensity (MFI) on T cells with a fixed level of T cell receptor (Fig 6C) as a proxy for TCR affinity 1, T cells derived from anti-CTLA4- LNP immunized mice produced T cells with an increased affinity for antigen (see for example, quantification of this observation in Fig 6D).
[0101] These experimental examples (increased frequency of antigen specific T cells illustrated in Figs. 6A and 6B, and increased TCR affinity of the responding cell-Fig 6C) are consistent with the conclusion that delivery of the heavy and light chain mRNAs of the anti- CTLA4 antibody via LNPs results in the formation of functionally active antibody and blockade of the CTLA4 pathway and is proof of concept that nanoparticles encapsulating antagonistic antibodies and/or agonistic antibodies as demonstrated herein targeted to a selected pathway can be delivered and expressed to induce, inhibit or block the targeted pathway for inducing an enhanced cellular immune response. As demonstrated herein, examples of LNP-encapsulated delivery of heavy and light chain antibody sequences were provided which produce antibodies capable of agonizing (FGK45) or antagonizing (9H10) immunologically relevant pathways (CD40 and CTLA4, respectively).
Example 7
[0102] These examples of formation and use of LNP-encapsulated mRNA for the production of biologically active agonistic (Example 4) and antagonistic/blocking (Example 6) antibodies provide a proof of concept. In another exemplary method, LNPs containing the heavy and light chains of monoclonal antibodies which mediate cellular depletion can be utilized. For example, antibodies against CD20 (e g., Rituxan) or CD19 (e.g., tafasitamab) have been well described in the literature for efficacious use against B cell lymphomas based on their capacity to induce B cell depletion. Similarly, antibodies against molecules such as VLA4 (e.g., natalizumab) facilitate both blockade of activated lymphocyte trafficking as well as activated lymphocyte depletion as therapeutic modalities.
[0103] In other exemplary methods, antibodies against surface proteins expressed by tumor cells (Her2/neu) can be targeted by monoclonal antibodies (e.g., Trastuzumab or similar monoclonal antibody), causing cell death and increased immunity against the tumor. These examples of agonistic and antagonistic antibody production via mRNA encapsulated LNPs shown above strongly support the likely success of using the same technology/methods for intervention in therapeutic modalities requiring monoclonal antibody-mediated cellular depletion.
The foregoing discussion of the disclosure has been presented for purposes of illustration and description. The foregoing is not intended to limit the disclosure to the form or forms disclosed herein. Although the description of the disclosure has included description of one or more embodiments and certain variations and modifications, other variations and modifications are within the scope of the disclosure, e.g., as can be within the skill and knowledge of those in the art, after understanding the present disclosure. It is intended to obtain rights which include alternative embodiments to the extent permitted, including alternate, interchangeable and/or equivalent structures, functions, ranges or steps to those claimed, whether or not such alternate, interchangeable and/or equivalent structures, functions, ranges or steps are disclosed herein, and without intending to publicly dedicate any patentable subject matter.

Claims

WHAT IS CLAIMED IS:
1. A complex comprising: at least one mRNA transcript, wherein at least one of the at least one mRNA transcript encodes for a heavy chain of an antibody or fragment thereof, or at least one of the at least one mRNA transcript encodes for a light chain of the antibody or fragment thereof, wherein the heavy chain and the light chain or fragment thereof comprise separate transcripts and encode a single antibody or fragment of a single antibody thereof and wherein the encoded antibody or fragment thereof modifies a pathway; and at least one coating encapsulating the at least one mRNA transcript wherein the at least one coating encapsulating the at least one mRNA transcript comprises a lipid to form a lipid- coated nanoparticle comprising the at least one mRNA transcript.
2. The complex according to claim 1, wherein the pathway comprises a cellular immune response pathway and wherein the at least one mRNA transcript encoding a light chain or heavy chain encodes a light chain or heavy chain of an antibody when assembled is capable of inducing a cellular immune response.
3. The complex according to claim 1 or 2, wherein the coating encapsulating the at least one mRNA transcript comprises an ionizing lipid nanoparticle (LNP).
4. The complex according to claim 1, wherein the complex further comprises at least one mRNA encoding at least one antigen.
5. The complex according to claim 4, wherein the at least one antigen comprises at least one antigen for inducing an immune response against a pathogenic organism, a tumor, or other antigen capable of inducing an immune response in a subject or a combination thereof.
6. The complex according to claim 4 or 5, wherein the mRNA encoding the at least one antigen further comprises one or more of an alpha globin 5'-UTR, an amino-terminal enhancer of split (AES) motif, and a mitochondrial 12S RNA element.
7. The complex according to any one of claims 4-6, wherein the mRNA encoding the at least one mRNA transcript encoding a heavy chain or a light chain of the antibody and optionally, the at least one mRNA encoding the at least one antigen further comprise one or more of incorporation of 1 methyl-pseudouridine (ImU15); at least one SerineUCG (SerUCG) codon substitution having at least one ImU13; and an mRNA sequence that remains unstructured between about the last 40 to 55 nucleotides (nts) of the 5’UTR and about the first 20 to 40 nts of the coding sequence, followed by increased secondary' structure thereafter compared to a native molecule.
8. The complex according to claim 4, wherein the at least one mRNA encoding the at least one antigen further comprises at least one serine codon substituted for SerUCG
9. The complex according to any one of claims 1-8, wherein the mRNA encoding the at least one mRNA transcript comprises at least two mRNA transcripts encoding at least one heavy chain and at least one light chain of the antibody within a single coated nanoparticle.
10. The complex according to claim 9, wherein the mRNA encoding the at least one mRNA transcript comprises at least two mRNA transcripts encoding at least one heavy chain and at least one light chain of the antibody comprise a ratio of mRNA encoding the at least one heavy chain of about a 1.5 : 1 ; 2: 1 , 2.5 : 1 ; 3 : 1 ; 3.5 : 1 or 4: 1 to the mRNA encoding the at least one light chain of the antibody.
11. The complex according to claim 1, further comprising mixing the complex of lipid- coated nanoparticles containing at least one mRNA transcript encoding at least one light chain or at least one heavy chain of an antibody or fragment thereof, with at least one mRNA transcript encoding at least one antigen to form a mixture of nanoparticles, wherein the mRNA transcripts are contained within the same or different nanoparticles.
12. The complex according to claim 1 or claim 2, further comprising at least 2 mRNA transcripts, each mRNA transcript of the at least 2 mRNA transcripts encode a heavy chain or a light chain of the antibody to form a full-length antibody upon release from the lipid-coated nanoparticle.
13. The complex according to any one of claims 1 to 3, wherein the antibody comprises an antagonistic antibody for inhibiting or completely turning off a cellular immune pathway.
14. The complex according to any one of claims 1 to 3, wherein the antibody comprises an agonistic antibody capable of inducing a cellular immune pathway.
15. The complex according to any one of claims 1-14, wherein the antibody comprises a monoclonal antibody.
16. The complex according to claim 1 or 2, wherein the antibody comprises an immune checkpoint inhibitor; and optionally comprises a monoclonal antibody.
17. The complex according to claim 1 or 2, wherein the antibody comprises at least one antibody of a full length or fragment thereof of an anti-HER2 antibody, an anti-EGFR antibody, an anti-VEGRF antibody, an anti-VEGF antibody, an anti-CD-20 antibody, an anti-CD-22 antibody, an anti-CD30 antibody, an anti-CD33 antibody, an anti-CD38 antibody, an anti- CD52 antibody, an anti-CTLA-4 antibody, an anti-PD-1 antibody, an anti-PD-Ll antibody, an anti-B7-l, an anti-B7-2, an anti-RANKL antibody, an anti-GD2 antibody, an anti-PDGFR antibody, an anti-SLAMF7 antibody or a combination of these antibodies are encoded by mRNA transcripts.
18. The complex according to claim 1 or 2, wherein the antibody comprises at least one antibody of a full length or fragment thereof of an anti-IL-1, an anti-IL-17, an anti-IL-22, an anti-IL-23, an anti-IFNy, an anti-TNFa, an anti-CD137 (41BB), an anti-CD40, an anti-CD134 (0X40), and an anti-CD27 or other cytokine specific antibody or a combination of these antibodies are encoded by mRNA transcripts.
19. The complex according to claim 17 or 18, wherein the antibody comprises at least two full length or fragments thereof of at least two different antibodies capable of inducing a cellular immune response.
20. The complex according to any one of claims 4-6, wherein the mRNA encoding the at least one antigen comprises a pathogen, a pathogenic agent, or a polypeptide derived from the pathogen.
21. The complex according to claim 20, wherein the pathogen, a pathogenic agent, or a polypeptide derived from the pathogen comprises one or more of a virus, bacteria, fungus, prion, protozoan or agent or polypeptide derived therefrom.
22. The complex according to claim 21, wherein the virus, viral agent or polypeptide derived from a vims comprises one or more viruses comprising Adenoviridae, Alphaviridae, Alphaviridea, Anelloviridae, Anelloviridae, Archiviridea, Arenaviridae, Arenaviridae, Arteriviridae, Astroviridae, Astroviridae, Bornaviridae, Bunyaviridae, Bunyaviridae, Caliciviridae, Caulimoviridae, Coronaviridae, Filoviridae, Flaviviridae, Hepadnaviridae, Hepeviridae, Herpesviridae, Lyssavirus, Orthobunyavirus, Orthomyxoviridae, Papillomaviridae, Papovaviridae, Paramyxoviridae, Paramyxoviridae, Parvoviridae, Picornaviridae, Pleolipoviridae, Pneumoviridae, Polyomaviridae, Polyomavirus, Poxviridae, Poxviridae, Reoviridae, Retroviridae, Rhabdoviridae, Seadornavirus, or Togaviridae family or other pathogenic vims family or combination thereof.
23. The complex according to claim 21, wherein the vims, viral agent or polypeptide derived from the vims comprises an enveloped or non-enveloped vims, agent or polypeptide derived therefrom.
24. The complex according to claim 21 , wherein the bacteria, bacterial agent or polypeptide derived from a bacteria comprises one or more of Acinetobacter spp.; Actinomycetes spp.; Aeromonas spp. ; Alcaligenes spp. ; Bacillus spp. ; Bacteriodes spp. ; Bartonella spp. ; Bordetella spp. ; Borrelia spp. ; Brevundimonas spp. ; Brucella spp. ; Burkholderia spp. ; Burkholderia spp. ; Campylobacter spp.; Candida spp.; Chlamydia spp.; Citrobacter spp.; Clostridium spp.; Clostridioides spp.; Corynebacterium spp.; Coxiella spp.; Enterobacter spp.; Enter obius spp. Escherichia coli spp. (incl. EHEC, EPEC, ETEC, EIEC, EAEC, ESBL/MRGN, DAEC) Francisella tularensis spp., Haemophilus influenzae spp.; Helicobacter spp.; Klebsiella spp.; Leclercia spp.; Legionella spp.; Leishmania spp.; Leptospira spp.; Leuconostoc spp.; Listeria spp.; Micrococcus spp.; Microsporum spp.; Moraxella spp.; Morganella spp.
Mycobacterium spp.; Mycoplasma spp.; Neisseria spp.; Orientia spp.; Pantoea spp.; Paracoccus spp. ; Prevotella spp. ; Propionibacterium spp. ; Proteus spp. ; Proteus spp. ;
Providencia spp.; Pseudomonas spp.; Ralstonia spp.; Rickettsia spp.; Roseomonas spp.; Salmonella spp.; Shigella spp.; Sphingomonas spp.; Staphylococcus spp.; Stenotrophomonas spp-;
Streptococcus spp.; Treponema spp.; Vibrio spp.; Yersinia spp. or a combination thereof.
25. A pharmaceutical composition, comprising the complex according to any one of claims 1-24, and a pharmaceutically acceptable excipient.
26. The pharmaceutical composition according to claim 25, further comprising one or more additional agents.
27. The pharmaceutical composition according to claim 26, wherein the one or more additional agents comprise one or more of an immunostimulant, immunosuppressant, or anti- microbial agent.
28. A method for treating a subject having a health condition, comprising administering to the subject having a health condition a composition according to any one of claims 25-27.
29. The method according to claim 28, wherein the health condition comprises cancer.
30. The method according to claim 29, wherein the cancer comprises a solid tumor.
31. The method according to claim 28, wherein the health condition comprises a pathogenic organism infection, or exposure or suspected exposure to a pathogenic organism.
32. The method according to any one of claims 28-31, further comprising administering a vaccine or immunogenic compositions specific to the health condition being treated to the subject at least one of: before, during or after administering the composition.
33. The method according to claim 32, wherein the vaccine comprises an anti-microbial or anti-tumor vaccine
34. The method according to any one of claims 28-33, wherein administering comprises intravenous, intranasal, intraocular, by inhalation, intramuscular, by aerosol, intradermal, transdermal, topical, intrauterine, or intrarectal administration or other suitable mode of administration to the subject.
35. The method according to claim 34, wherein administering the composition comprises administering the composition directly to a tumor or site of infection in the subject.
36. A kit comprising at least one complex according to any one of claims 1 -24, at least one pharmaceutical composition according to any one of claims 25-27, and at least one container.
37. The kit according to claim 36, further comprising a vaccine.
PCT/US2022/053132 2021-12-21 2022-12-16 Compositions and methods for delivery and production of antibodies using an rna-containing nanoparticle platform WO2023121960A2 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US202163292246P 2021-12-21 2021-12-21
US63/292,246 2021-12-21

Publications (2)

Publication Number Publication Date
WO2023121960A2 true WO2023121960A2 (en) 2023-06-29
WO2023121960A3 WO2023121960A3 (en) 2023-08-31

Family

ID=85157335

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2022/053132 WO2023121960A2 (en) 2021-12-21 2022-12-16 Compositions and methods for delivery and production of antibodies using an rna-containing nanoparticle platform

Country Status (1)

Country Link
WO (1) WO2023121960A2 (en)

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5874082A (en) * 1992-07-09 1999-02-23 Chiron Corporation Humanized anti-CD40 monoclonal antibodies and fragments capable of blocking B cell proliferation

Also Published As

Publication number Publication date
WO2023121960A3 (en) 2023-08-31

Similar Documents

Publication Publication Date Title
TWI755763B (en) Methods for treating allergy and enhancing allergen-specific immunotherapy by administering an il-4r inhibitor
KR101323540B1 (en) Therapeutic agent for cancer
US20130071403A1 (en) Synergistic anti-tumor efficacy using alloantigen combination immunotherapy
JP2018021047A (en) Cancer immunotherapy
US20150272980A1 (en) Dosing and Administration of Oligonucleotide Cancer Therapies
JP2018537536A (en) Compositions and methods for the treatment of HER2 positive metastatic breast cancer
US20230226169A1 (en) Multilamellar rna nanoparticle vaccine against sars-cov-2
US20190015504A1 (en) Combination therapy with cd4 lymphocyte depletion and mtor inhibitors
US20180128833A1 (en) Methods of treating with tumor membrane vesicle-based immunotherapy and predicting therapeutic response thereto
Kwon et al. Induction of immunological memory response by vaccination with TM4SF5 epitope-CpG-DNA-liposome complex in a mouse hepatocellular carcinoma model
WO2023121960A2 (en) Compositions and methods for delivery and production of antibodies using an rna-containing nanoparticle platform
US20140341971A1 (en) Composition for targeting dendritic cells
US20200055947A1 (en) Bi-specific activators for tumor therapy
CN118055775A (en) Vaccine composition for anticancer comprising HSP90 antigen peptide and use thereof
JP6799296B2 (en) New therapeutic agent for prionoid diseases
WO2020049534A1 (en) Sting agonist and combination therapy thereof for the treatment of cancer
US20210023236A1 (en) Multi-targeted, tunable, sustained delivery of payloads to charged avascular tissues
WO2024092352A1 (en) Compositions that recruit and activate antigen presenting cells
WO2023172989A2 (en) Epo receptor agonists and antagonists
JP2024520477A (en) Multicomponent chemical compositions of peptide-based neo-antigen vaccines
KR20210097101A (en) Targeting Moiety-Drug Implanted Immune Cell Compositions and Methods of Use

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 22851187

Country of ref document: EP

Kind code of ref document: A2