WO2014039961A1 - Fusion proteins for promoting an immune response, nucleic acids encoding same, and methods of making and use thereof - Google Patents

Fusion proteins for promoting an immune response, nucleic acids encoding same, and methods of making and use thereof Download PDF

Info

Publication number
WO2014039961A1
WO2014039961A1 PCT/US2013/058748 US2013058748W WO2014039961A1 WO 2014039961 A1 WO2014039961 A1 WO 2014039961A1 US 2013058748 W US2013058748 W US 2013058748W WO 2014039961 A1 WO2014039961 A1 WO 2014039961A1
Authority
WO
WIPO (PCT)
Prior art keywords
subject
composition
lmpl
immune
cells
Prior art date
Application number
PCT/US2013/058748
Other languages
French (fr)
Inventor
Geoffrey W. STONE
Sachin Gupta
Original Assignee
University Of Miami
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 University Of Miami filed Critical University Of Miami
Priority to EP13835331.3A priority Critical patent/EP2892930B1/en
Priority to US14/424,562 priority patent/US10093701B2/en
Publication of WO2014039961A1 publication Critical patent/WO2014039961A1/en
Priority to US16/115,171 priority patent/US10329329B2/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/005Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/12Viral antigens
    • A61K39/245Herpetoviridae, e.g. herpes simplex virus
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70503Immunoglobulin superfamily
    • C07K14/7051T-cell receptor (TcR)-CD3 complex
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70503Immunoglobulin superfamily
    • C07K14/70535Fc-receptors, e.g. CD16, CD32, CD64 (CD2314/705F)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70546Integrin superfamily
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/7056Lectin superfamily, e.g. CD23, CD72
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70578NGF-receptor/TNF-receptor superfamily, e.g. CD27, CD30, CD40, CD95
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70596Molecules with a "CD"-designation not provided for elsewhere
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/715Receptors; Cell surface antigens; Cell surface determinants for cytokines; for lymphokines; for interferons
    • C07K14/7156Receptors; Cell surface antigens; Cell surface determinants for cytokines; for lymphokines; for interferons for interferons [IFN]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/715Receptors; Cell surface antigens; Cell surface determinants for cytokines; for lymphokines; for interferons
    • C07K14/7158Receptors; Cell surface antigens; Cell surface determinants for cytokines; for lymphokines; for interferons for chemokines
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/72Receptors; Cell surface antigens; Cell surface determinants for hormones
    • C07K14/723G protein coupled receptor, e.g. TSHR-thyrotropin-receptor, LH/hCG receptor, FSH receptor
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N7/00Viruses; Bacteriophages; Compositions thereof; Preparation or purification thereof
    • 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
    • C07K2319/00Fusion polypeptide
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/01Fusion polypeptide containing a localisation/targetting motif
    • C07K2319/02Fusion polypeptide containing a localisation/targetting motif containing a signal sequence
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/01Fusion polypeptide containing a localisation/targetting motif
    • C07K2319/03Fusion polypeptide containing a localisation/targetting motif containing a transmembrane segment
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/16011Herpesviridae
    • C12N2710/16211Lymphocryptovirus, e.g. human herpesvirus 4, Epstein-Barr Virus
    • C12N2710/16234Use of virus or viral component as vaccine, e.g. live-attenuated or inactivated virus, VLP, viral protein

Abstract

Disclosed herein are compositions, cells, kits, and methods for inducing an immune response in a subject. The compositions can be used as vaccines or vaccine adjuvants against cancer (e.g., melanoma, glioma, prostate, breast) and infectious diseases (e.g., therapeutic and preventative vaccination for viruses), and can be used in cell-based therapies for preventing and treating disorders such as cancer and infection. The compositions, cells, kits and methods involve one or more nucleic acids that encode one or more LMPI fusion proteins (chimeric proteins), and in a typical embodiment, synergistic activation of immune responses by a combination of two or more LMPI fusion proteins.

Description

FUSION PROTEINS FOR PROMOTING AN IMMUNE RESPONSE, NUCLEIC ACIDS ENCODING SAME, AND METHODS OF MAKING AND USE THEREOF
CROSS REFERENCE TO RELATED APPLICATIONS
[0001] This application claims the benefit of U.S. Provisional Application Serial No. 61/698,109 filed on September 7, 2012, which is hereby incorporated by reference in its entirety, for all purposes, herein.
SEQUENCE LISTING
[0002] The instant application contains a Sequence Listing which has been submitted in ASCII format via EFS-Web and is hereby incorporated by reference in its entirety. Said ASCII copy, created on September 9, 2013, is named 7230-171WO_SL.txt and is 72,591 bytes in size.
FIELD OF THE INVENTION
[0003] The invention relates generally to the field of immunology. More particularly, the invention relates to fusion proteins, nucleic acids encoding fusion proteins, and methods of use thereof for vaccines and cell therapy.
BACKGROUND
[0004] Some current vaccine and cell therapy technologies are associated with the problem of properly activating immune cells for vaccination or immune therapy of patients as a treatment for cancer or infectious disease. There is thus a significant need for improved immune stimulators that provide potent activation of the immune response in patients suffering from cancer or an infectious disease, or who are at risk thereof.
SUMMARY
[0005] Disclosed herein are compositions, cells, kits, and methods for inducing an immune response in a subject. The compositions can be used as immune stimulators to increase the response to a vaccine composition (in which case the immune stimulator is described as a vaccine adjuvant) or given to patients to improve ongoing immune responses such as those directed against cancer or an established infection. Representative cancers include melanoma, glioma, prostate, breast, and HPV-related cancers and representative infections include Human Immunodeficiency Virus (HIV), hepatitis C virus, human papilloma virus (HPV), malaria, biodefense related agents, and all other infectious agents that do not currently have effective safe vaccines. These compositions for immune stimulation can also be used in cell-based therapies (e.g. dendritic cell (DC) therapies) where cells are treated ex vivo and then delivered to the subject for preventing and treating disorders such as cancer and infection. The compositions, cells, kits and methods involve one or more nucleic acids that encode one or more LMP1 fusion proteins (also referred to as protein chimeras), and in a typical embodiment provide activation of immune responses by a combination of two or more LMP1 fusion proteins. By combining multiple fusion proteins composed of the LMP1 protein aggregation (transmembrane) domain and the cytoplasmic domain of toll-like receptors (TLR), proteins, Tumor Necrosis Factor SuperFamily Receptors (TNFSFR) proteins, pattern recognition receptor (PRR) proteins, or adapter proteins that are involved in innate and/or adaptive immune signaling pathways, introduction of the fusion proteins into immune cells such as dendritic cells or other antigen presenting cells (APCs) results in higher levels of activation. This technology can be used for traditional prophylactic or therapeutic vaccines against cancer and infectious diseases, as well as cell-based therapies such as dendritic cell therapy. In the experiments described herein, combinations of fusion proteins markedly enhanced immune responses and protection from infection, and combinations of one or more TLR agonists and a stimulator of the TNFSFR protein CD40 also induced a synergistic immune activation that protected experimental mice from tumor challenge. Also in the experiments described herein, a surprising result was that LMP1-IPS 1 is effective at restricting HIV-1 replication. LMP1-IPS 1 inhibits wild-type HIV-1 replication in cell culture and in primary human CD4+ T cells. Bystander cells expressing LMP1-IPS 1 can inhibit viral replication, suggesting that LMP1-IPS 1 can be given as a therapy in HIV-infected patients. LMPl-IPS l can be given as a viral vector (i.e. lentiviral vector) targeting sites of HIV-1 infection, thereby reducing HIV-1 replication in tissue reservoirs that are normally resistant to antiretroviral drugs. The compositions, cells, kits and methods described herein address the problem of properly activating and maturing APCs such as dendritic cells for vaccination or immune therapy of patients as a treatment for cancer or infectious disease, and can also be used to develop prophylactic vaccines and other immune therapies dependent on immune activation.
[0006] Unless otherwise defined, all technical terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. [0007] As used herein, a "nucleic acid" or a "nucleic acid molecule" means a chain of two or more nucleotides such as RNA (ribonucleic acid) or DNA (deoxyribonucleic acid), and chemically-modified nucleotides. The nucleic acid molecule may be purified. A "purified" nucleic acid molecule is one that is substantially separated from other nucleic acid sequences in a cell or organism in which the nucleic acid naturally occurs (e.g., 30, 40, 50, 60, 70, 80, 90, 95, 96, 97, 98, 99, 100% free of contaminants). The terms include, e.g., a recombinant nucleic acid molecule incorporated into a vector, a plasmid, a virus, or a genome of a prokaryote or eukaryote. Examples of purified nucleic acid molecules include cDNAs, fragments of genomic nucleic acid molecules, nucleic acid molecules produced by polymerase chain reaction (PCR), nucleic acid molecules formed by restriction enzyme treatment of genomic nucleic acid molecules, recombinant nucleic acid molecules, and chemically synthesized nucleic acid molecules.
[0008] By the term "LMPl gene," is meant a native Epstein Barr virus LMPl -encoding nucleic acid sequence, e.g., the native Epstein Barr virus LMPl gene; a nucleic acid having sequences from which a LMPl cDNA can be transcribed; and/or allelic variants and homologs of the foregoing. An exemplary nucleic acid sequence of LMPl is GenBank Accession No. M58153.1. The term encompasses double- stranded DNA, single-stranded DNA, and RNA.
[0009] By the term "LMPl protein," is meant an expression product of a LMPl gene or a protein that shares at least 65% (but preferably 75, 80, 85, 90, 95, 96, 97, 98, or 99%) amino acid sequence identity with the foregoing and displays a functional activity of a native LMPl protein. A "functional activity" of a protein is any activity associated with the physiological function of the protein. LMPl consists of an N-terminal transmembrane region linked to a C-terminal cell signaling region that is analogous to the CD40 receptor on immune cells (see Figure 1A). In addition to anchoring LMPl into the membrane, the N-terminus of LMPl self-aggregates and leads to clustering of LMPl or any protein linked to the LMPl N-terminal domain. The transmembrane (aggregation) domain of LMPl protein is amino acids 1-190 of the amino acid sequence set forth in GenBank Accession No. AAA66330.1.
[00010] As used herein, "protein" and "polypeptide" are used synonymously to mean any peptide-linked chain of amino acids, regardless of length or post-translational modification, e.g., glycosylation or phosphorylation. The terms "fusion protein," "chimeric protein," and "chimera" are used interchangeably herein, and mean a protein made by translation of an artificial combination of two otherwise separated segments of sequence, e.g., by chemical synthesis or by the manipulation of isolated segments of nucleic acids by genetic engineering techniques.
[00011] When referring to a peptide, oligopeptide or protein, the terms "amino acid residue", "amino acid" and "residue" are used interchangably and, as used herein, mean an amino acid or amino acid mimetic joined covalently to at least one other amino acid or amino acid mimetic through an amide bond or amide bond mimetic.
[00012] When referring to a nucleic acid molecule, polypeptide, or infectious pathogen, the term "native" refers to a naturally-occurring (e.g., a wild-type (WT)) nucleic acid, polypeptide, or infectious pathogen.
[00013] As used herein, the term "antigen" or "immunogen" means a molecule that is specifically recognized and bound by an antibody.
[00014] The term "antibody" is meant to include polyclonal antibodies, monoclonal antibodies (mAbs), chimeric antibodies, humanized antibodies, anti-idiotypic (anti-Id) antibodies to antibodies that can be labeled in soluble or bound form, as well as fragments, regions or derivatives thereof, provided by any known technique, such as, but not limited to, enzymatic cleavage, peptide synthesis or recombinant techniques.
[00015] As used herein the term "adjuvant" means any material which enhances the humoral and/or cellular immune response.
[00016] As used herein, an "immune cell" refers to dendritic cells, macrophages, lymphocytes, mast cells, endothelial cells, lymphatic vessel cells and the like which can, when properly stimulated, serve as an antigen-presenting cell (APC) to initiate an immune response or as an effector cell of an immune response.
[00017] As used herein, the terms "displayed", "presented", or "surface exposed" are considered to be synonyms, and refer to antigens or other molecules that are present (e.g., accessible to immune site recognition) at the external surface of a structure such as a cell.
[00018] As used herein, "vaccine" includes all prophylactic and therapeutic vaccines.
[00019] By the phrase "immune response" is meant induction of antibody and/or immune cell-mediated responses specific against an antigen or antigens or allergen(s) or drug or biologic. The induction of an immune response depends on many factors, including the immunogenic constitution of the challenged organism, the chemical composition and configuration of the antigen or allergen or drug or biologic, and the manner and period of administration of the antigen or allergen or drug or biologic. An immune response has many facets, some of which are exhibited by the cells of the immune system (e.g., B-lymphocytes, T-lymphocytes, macrophages, and plasma cells). Immune system cells may participate in the immune response through interaction with an antigen or allergen or other cells of the immune system, the release of cytokines and reactivity to those cytokines. Immune responses are generally divided into two main categories - humoral and cell-mediated. The humoral component of the immune response includes production of antibodies specific for an antigen or allergen or drug or biologic. The cell- mediated component includes the generation of delayed-type hypersensitivity and cytotoxic effector cells against the antigen or allergen.
[00020] As used herein, the term "treatment" is defined as the application or administration of a therapeutic agent to a patient, or application or administration of the therapeutic agent to an isolated tissue or cell line from a patient, who has a disease, a symptom of disease or a predisposition toward a disease, with the purpose to cure, heal, alleviate, relieve, alter, remedy, ameliorate, improve or affect the disease, the symptoms of disease, or the predisposition toward disease.
[00021] As used herein, the phrase "safe and effective amount" refers to the quantity of a component which is sufficient to yield a desired therapeutic response without undue adverse side effects (such as toxicity, irritation, or allergic response) commensurate with a reasonable benefit/risk ratio when used in the manner of this invention. By "therapeutically effective amount" is meant an amount of a composition of the present invention effective to yield the desired therapeutic response, for example, an amount effective to activate an immune response in an individual. The specific safe and effective amount or therapeutically effective amount will vary with such factors as the particular condition being treated, the physical condition of the patient, the type of mammal or animal being treated, the duration of the treatment, the nature of concurrent therapy (if any), and the specific formulations employed.
[00022] The terms "patient" "subject" and "individual" are used interchangeably herein, and mean a mammalian subject (e.g., human, rodent, non-human primates, canine, bovine, ovine, equine, feline, etc.) who is to be treated, who has been treated, or who is being considered for treatment, and/or to obtain a biological sample from, with human patients being preferred. In some cases, the methods, kits, and compositions described herein find use in experimental animals, in veterinary applications for livestock, domesticated animals, and companion animals, and in the development of animal models for disease, including, but not limited to, rodents including mice, rats, and hamsters, as well as non-human primates.
[00023] Accordingly, described herein is a composition for inducing an immune response in a subject. The composition includes at least a first nucleic acid encoding at least a first fusion protein, the at least first fusion protein including a transmembrane domain of LMP1 to provide for clustering of the at least first fusion protein in cells without the need for any exogenous molecule and at least one signaling domain from at least one of: an immune activating receptor and/or an adaptor protein, the at least first nucleic acid in an amount sufficient to induce an immune response in a subject. The immune activating receptor can be, for example, a Toll-like receptor (TLR) or a TNF superfamily receptor (TNFSFR). The at least first fusion protein can include a signaling domain from a TLR and a signaling domain from a TNFSFR. In another embodiment, the at least first fusion protein can include a signaling domain from an immune activating receptor and/or a signaling domain from an adaptor protein. The immune activating receptor can be, for example, TNF Receptor SuperFamily (TNFRSF) proteins, preferably CD40, 4- IBB, RANK, TACI, OX40, CD27, GITR, LT R, and BAFFR; Toll-Like Receptors (TLR) TLR1 to TLR13, preferably TLR7; integrins; FcyRIII; Dectinl; Dectin2; NODI; NOD2; CD16; IL-2R; Type I II interferon receptor; chemokine receptors such as CCR5 and CCR7; G-protein coupled receptors (GPCRs); TREM1; the B cell receptor (BCR) complex that includes CD79A, CD79B, and Ig-alpha, etc. The adaptor protein can be, for example, one of: IPS-1; MyD88; RIG-1; MDA5; CD3 zeta chain; MyD88ATIR; TRIF; TRAM; TIRAP; MAL; BTK; RTK; RAC1; SYK; NALP3 (NLRP3); NALP3ALRR; NALP1; CARD9; DAI; IPAG; STING; Zap70; and LAT. The at least first nucleic acid can further encode an antigen. The at least first nucleic acid can be included within a viral vector. The viral vector (including the nucleic acid sequence) can be within (encapsulated by) a recombinant virion. The composition can further include a second nucleic acid encoding a second fusion protein, the second fusion protein including a transmembrane domain of LMP1 and at least one signaling domain from at least one of: an immune activating receptor and an adaptor protein. The at least first and the second nucleic acid can be included within a viral vector. Typically, the at least first nucleic acid is an amount effective for activating and maturing immune cells (e.g., dendritic cells) when administered to a subject (e.g., a human). In a composition as described herein, the at least first nucleic acid can be in an amount effective for inducing expression of cytokines and protecting primary CD4+ T cells from infection by a virus (e.g., human immunodeficiency virus (HIV)) when administered to a subject (e.g., a human). The at least first fusion protein can include a transmembrane domain of LMP1 and a signaling domain from IPS-1.
[00024] Also described herein is a vaccine formulation for preventing or treating a disease or condition in a subject including a composition as described herein and a pharmaceutically acceptable excipient. The disease or condition can be, for example, cancer or infection.
[00025] Further described herein is a vaccine adjuvant for enhancing an immune response to a vaccine. Typically the vaccine adjuvant includes a composition as described herein and an antigen or a nucleic acid encoding an antigen in an amount effective for enhancing an immune response to a vaccine and a pharmaceutically acceptable excipient. In one embodiment, the at least first nucleic acid can further encode the antigen.
[00026] Still further described herein is an immune cell transduced with a composition as described herein. In a typical embodiment, the immune cell is a dendritic cell. The immune cell can be within a vaccine formulation for preventing or treating a disease or condition (e.g., cancer or infection) in a subject.
[00027] Additionally described herein is A method of inducing an immune response against cancer or infection in a subject. The method includes administering a composition as described herein to the subject in a therapeutically effective amount for inducing an immune response against the cancer or infection in the subject. Administering the composition to the subject vaccinates the subject against cancer or infection. Inducing an immune response against the cancer or the infection in the subject can include at least one of: activating and maturing dendritic cells, and inducing expression of at least one cytokine in the subject. The cancer can be, for example, melanoma, glioma, prostate cancer, breast cancer, and the infection is selected from the group consisting of: HIV infection, hepatitis C infection or human papilloma virus infection. In one embodiment, the subject has an HIV infection, and inducing an immune response against the infection includes induction of type I interferon and protection of primary CD4+ T cells in the subject. In another embodiment, the subject has a cancerous tumor, and the at least first fusion protein includes a transmembrane domain of LMP1 and a signaling domain from IPS-1 or a signaling domain from MyD88. In this embodiment, inducing an immune response results in a decrease in growth of the cancerous tumor. [00028] Yet further described herein is a kit for preventing or treating a disease or condition in a subject. A kit includes a composition, vaccine formulation, or vaccine adjuvant as described herein; instructions for use; and packaging.
[00029] Also described herein is a composition for inducing an immune response in a subject including a nucleic acid encoding a fusion protein. In the composition, the fusion protein includes a transmembrane domain of LMPl and an amino acid sequence including two or more signaling motifs from immune activating receptors and/or adaptor proteins. The nucleic acid is in an amount sufficient to induce an immune response in a subject.
[00030] Although compositions, cells, kits, and methods similar or equivalent to those described herein can be used in the practice or testing of the present invention, suitable compositions, cells, kits, and methods are described below. All publications, patent applications, and patents mentioned herein are incorporated by reference in their entirety. In the case of conflict, the present specification, including definitions, will control. The particular embodiments discussed below are illustrative only and not intended to be limiting.
BRIEF DESCRIPTION OF THE DRAWINGS
[00031] FIG. 1 is a model of LMPl fusion protein technology as described herein.
[00032] FIG. 2 is a series of graphs of results showing that LMPl fusion proteins with TLR, PRR, CD40, and/or signaling adapter proteins are potent inducers of NF-κΒ and IFN-β responses.
[00033] FIG. 3 is a series of graphs of results showing that LMPl fusion proteins with IPS-1, TLR7, and MyD88-CD40 enhance activation and IL-6 cytokine secretion from a RAW 264.7 mouse macrophage cell line.
[00034] FIG. 4 is a series of graphs of results showing that human monocyte derived DCs transfected with LMPl-TLR fusion constructs induce activation, maturation, and cytokine secretion.
[00035] FIG. 5 is a pair of graphs of results showing that combinations of LMP1-CD40 with LMPl-TLR fusions enhanced maturation, activation, and secretion of pro-inflammatory cytokines from human DC. [00036] FIG. 6 is a series of graphs of results showing that LMPl-MyD88-CD40 fusion enhances immune responses and control of viral infection in a mouse influenza DNA vaccine model.
[00037] FIG. 7 is a pair of graphs of results showing enhanced survival and reduced tumor growth using LMP1 fusion combinations as a dendritic cell cancer therapy.
[00038] FIG. 8 is a pair of graphs of results showing that LMPl-IPS-1 is a potent inducer of NF-κΒ and IFN-β responses.
[00039] FIG. 9 is a graph of results showing that LMPl-IPS-1 enhances IL-6 secretion from RAW 264.7 mouse macrophage cell line.
[00040] FIG. 10 is a series of graphs showing that LMPl-IPS-1 enhances activation of RAW 264.7 mouse macrophage cell line.
[00041] FIG. 11 is a pair of graphs of results showing that a LMPl-IPS-1 fusion protein is able to protect cells from HIV-1 infection and/or replication.
[00042] FIG. 12 is a pair of graphs of results showing that LMPl-IPS-1 fusion protein is able to protect cells from single cycle SIV infection and/or replication.
[00043] FIG. 13 is a graph of results showing that soluble factors produced by LMPl-IPS l expressing cells can effectively inhibit HIV-1 in primary CD4+ human T cells.
[00044] FIG. 14 is a series of graphs of results showing that LMPl-IPS l induces type I interferons as well as interferon stimulated genes implicated in the inhibition of HIV-1 replication.
[00045] FIG. 15 is a pair of graphs of results showing that full length LMP1 and a LMP1- IPS-1 fusion protein are able to protect cells from HIV-1 infection, with superior protection by LMPl-IPS-1.
[00046] FIG. 16 is a schematic illustration of examples of different LMP1 fusion protein construct formulations.
DETAILED DESCRIPTION
[00047] Described herein are LMP1 fusion proteins, nucleic acids encoding the fusion proteins, viral vectors containing the nucleic acids, and cells transduced with the nucleic acids for inducing an immune response in a subject. In our previous publications (S. Gupta et al., Journal of Leukocyte Biology 90:389-398, 2011; and S. Gupta et al., Retrovirology 8:39, 2011) and a prior application (R.S. Kornbluth and G. W. Stone, PCT WO 2011/119628, now US patent application pub. No. 2013/0039942 Al and EP11760085.8), the invention of LMPl fusion proteins is described. These were described as fusion proteins that combine an LMPl transmembrane domain and a single (not more than one) signaling domain taken from protein receptors in the Tumor Necrosis Factor Receptor SuperFamily (TNFRSF). In the instant invention, LMPl fusions are made with a plurality (two or more) of signaling domains. Additionally, the instant invention presents fusion proteins of LMPl combined with a signaling domain that is not derived from a TNFRSF but instead is derived from other types of immune activating receptors such as Toll-Like Receptors (TLRs) and/or adaptor proteins that are not part of receptors at the cell surface but rather intracytoplasmic adaptor molecules that initiate signaling events downstream from cell surface receptors in the cell such as IPS-1. Collectively, the LMPl fusion proteins of the instant invention contain signaling domains selected from the TNFRSFs, TLRs, and/or adaptor molecules such that two or more signaling domains are fused with portions of LMPl. As an exception, LMPl itself has a C-terminal intracytoplasmic domain that is recognized as functionally similar to a TNFRSF signaling domain, specifically from the CD40 receptor, a member of the TNFRSFs. Consequently, fusion proteins of the instant invention are of the form LMPl transmembrane domain (LMP1TM) operatively linked to signaling domain X and operatively linked to signaling domain Y to form LMP1TM-X-Y, where the linkage is normally provided by peptide bonds that make the fusion protein a single polypeptide strand. In cases where the entire LMPl protein is used, the "X" portion is the signaling domain that naturally occurs in LMPl followed by one or more "Y" signaling domains so that again the fusion proteins contains two or more signaling domains. In yet another embodiment, the instant invention describes LMPl fusion proteins wherein a peptidic antigen (Ag) is included in the LMP fusion protein polypeptide. Such fusion proteins have the structure of Ag-LMP1TM-X-Y and may have sequences that include an internal ribosome entry site (IRES) between the sequence encoding the antigen and the sequence encoding the N-terminal transmembrane domain of LMPl. The unifying feature of all of these fusion proteins is the presence of the LMPl N-terminal transmembrane domain which (1) anchors the fusion proteins into membranes and (2) aggregates them into clusters in the cell membrane. By clustering the signaling domains, LMPl fusion proteins allow those signaling domains to engage downstream adaptor molecules that lead to altered gene expression in the nucleus. Prior to this invention, it was not known in the art that the same LMPl transmembrane domain could activate more than one signaling domain when the LMPl and two or more signaling domains are joined in tandem in a single polypeptide chain. As a modification of this concept, an LMPl fusion protein was made using Interferon Promoter Stimulator-1 (IPS-1, also called MAVS, VISA, or Cardif). In this case, fusion to LMPl provides for the aggregation of IPS-1 that is needed for cellular activation by the IPS-1 protein (F. Hou et al., Cell 146:448-461, 2011). This novel molecule, LMPl -IPS-1, has only one signaling domain but that signal domain is used in a way not previously known in the art.
[00048] Latent membrane protein- 1 (LMPl) is a gene in the Epstein-Barr Virus (EBV). As shown in Fig. 1, its N-terminus is composed of 6 contiguous transmembrane domains that anchor the protein into the membrane. The intracytoplasmic domain of LMPl is analogous to the signaling domain of the CD40 receptor, a TNFRSF. The activation of CD40 requires that it become clustered in the membrane so that its cytoplasmic signaling domain forms a supramolecular signaling complex in concert with adaptor molecules in the TNF Receptor Activating Factor (TRAF) family, e.g., TRAF2, TRAF3, TRAF6), also referred to as a signalosome. The clustering of CD40 is initiated by either a multimeric form of its ligand (CD40 ligand or CD40L) or by anti-CD40 antibodies that must be arrayed on a nearby cell via binding to Fc receptors (as reviewed in R.S. Kornbluth, M. Stempniak, and G.W. Stone, International Review of Immunology 31:279-288, 2012). However, LMPl needs no ligand or antibody to initiate signaling through its cytoplasmic domain since its N-terminal transmembrane domain spontaneously forms clusters in the cell membrane and thereby clusters the intracytoplasmic domain(s) that are connected to it via peptide bonds as a single polypeptide chain. In this sense, LMPl is said to be "constitutively activated." Likewise, fusion proteins that link the N-terminal transmembrane domain to signaling domain(s) that require clustering in order to function can also be said to be "constitutively activated" and no longer need the ligand from the receptor from which they are taken. For example, TLR7 is normally activated by viral RNA as a ligand, but LMP1-TLR7 (a fusion protein formed by linking the LMPl N-terminal transmembrane domain with the intracellular TLR7 signaling domain) is constitutively active and signals without a ligand (Fig. 3). Because LMPl fusion proteins are constitutively active, they differ significantly from "inducible" protein constructs (B.A. Hanks et al., Nature Medicine 11: 130-137, 2005; D. Spencer, B. Hanks, and K. Slawin, US 7404950 B2; D. Spencer and N. Lepteva, US 2011/0033383 Al; and D. Spencer and N. Priyadharshini, US 2010/0203067 Al). These inducible protein constructs rely upon a chemically inducible dimerization (CID) domain to produce clustering of the construct when a chemically induced dimerization agent is administered to the subject (e.g., AP20187). In marked constrast, the molecules of the instant invention spontaneously cluster in a controlled and reproducible manner by virtue of being linked to the LMP1 N-terminal transmembrane and do not require any exogenous molecule or chemical to function.
[00049] In making LMP1 fusion proteins, the signaling domains comprise one or more cytoplasmic signaling domains of immune activating receptors (e.g., TLR cytoplasmic domains and/or TNFSFR cytoplasmic domains) and/or adaptor proteins. The immune activating receptors being contemplated in this invention include, for example, TNFRSFs (e.g., CD40, 4-1BB, RANK, TACI, OX40, CD27, GITR, LT R, and BAFFR), TLR1 through TLR 13 inclusive, integrins, FcyRIII, Dectinl, Dectin2, NODI, NOD2, CD 16, IL-2R, Type MI interferon receptor, chemokine receptors such as CCR5 and CCR7, GPCRs, TREM1, and the B cell receptor (BCR) complex that includes CD79A, CD79B, Ig-alpha, and Ig-beta. The adaptor proteins being contemplated in this invention include, for example, IPS-1, RIG-1, MDA5, CD3 zeta chain, MyD88, MyD88ATIR, TRIF, TRAM, TIRAP, MAL, BTK, RTK, RAC1, SYK, NALP3 (NLRP3), NALP3ALRR, NALP1, CARD9, DAI, IPAG, STING, Zap70, and LAT. These LMP1 fusion proteins can be encoded within recombinant DNA, RNA, or viral vectors for the treatment of subjects. In some formulations, an antigen will be encoded within the same vector as the LMP1 -fusion, inducing antigen- specific immune activation. Based on the protein binding motifs within each of these cytoplasmic domains, an artificial cytoplasmic domain can be constructed that incorporates signaling domains from one or more immune activating receptors and/or one or more adapter proteins in combinations that give the greatest immune response. This artificial signaling domain is anticipated to induce greater immune activation, and more effective vaccines or immune therapy, compared to the wild- type signaling domains. An artificial cytoplasmic domain composed of intracellular signaling motifs from multiple TLRs, TNFSFR, or adapter molecules can, as a single gene product, mimic the synergy induced by multiple but separate LMP1 fusion proteins. Such a superactivating LMP1 construct can be used as a vaccine adjuvant and immune stimulator. [00050] Several of the signaling domains contemplated in this invention are part of the inflammasome pathway. These include IPS-1, NALP2, NLRP3, and AIM2. A key function of the inflammasome pathway is to activate the proteases (e.g., caspase-1) that cleave the inactive precursor of interleukin-l-beta (pro-IL-lbeta) into the active form of this cytokine (IL-lbeta). The precursor protein, pro-IL-l-beta, is not normally present in immune cells but instead the transcription of its gene and the production of the inactive protein is initiated by activating TLRs and downstream adaptor molecules for the TLR pathway. Thus, the induction of the inflammasome and cleavage of pro-IL-lbeta to IL-lbeta may be accomplished by induction of IL-lbeta gene using LMP1-TLR2, LMPl-MyD88, etc, together with caspase inflammasome induction with LMP1 fused to domains of proteins such as NALP2, NLRP3, or AIM2 which are involved in inflammasome induction. The LMP1 fusions described herein would also be effective even in the context where the APC does not express the natural receptor for the TLR agonist or TNFSFR agonist, since the LMP1 fusions already encode the receptor in a constitutively active state.
[00051] The below described preferred embodiments illustrate adaptations of these compositions, cells, kits, and methods. Nonetheless, from the description of these embodiments, other aspects of the invention can be made and/or practiced based on the description provided below.
Biological Methods
[00052] Methods involving conventional molecular biology techniques are described herein. Such techniques are generally known in the art and are described in detail in methodology treatises such as Molecular Cloning: A Laboratory Manual, 3rd ed., vol. 1-3, ed. Sambrook et al., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., (2001); and Current Protocols in Molecular Biology, ed. Ausubel et al., Greene Publishing and Wiley-Interscience, New York, (1992) (with periodic updates). Immunology techniques are generally known in the art and are described in detail in methodology treatises such as Current Protocols in Immunology, ed. Coligan et al., Greene Publishing and Wiley-Interscience, New York, (1992) (with periodic updates); Advances in Immunology, volume 93, ed. Frederick W. Alt, Academic Press, Burlington, MA, (2007); Making and Using Antibodies: A Practical Handbook, eds. Gary C. Howard and Matthew R. Kaser, CRC Press, Boca Raton, Fl, (2006); Medical Immunology, 6th ed., edited by Gabriel Virella, Informa Healthcare Press, London, England, (2007); and Harlow and Lane ANTIBODIES: A Laboratory Manual, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY, (1988). Conventional methods of gene transfer and gene therapy may also be adapted for use in the present invention. See, e.g., Gene Therapy: Principles and Applications, ed. T. Blackenstein, Springer Verlag, 1999; Gene Therapy Protocols (Methods in Molecular Medicine), ed. P.D. Robbins, Humana Press, 1997; Viral Vectors for Gene Therapy: Methods and Protocols, ed. Otto-Wilhelm Merten and Mohammed Al-Rubeai, Humana Press, 2011; and Nonviral Vectors for Gene Therapy: Methods and Protocols, ed. Mark A. Findeis, Humana Press, 2010.
Nucleic Acids and Compositions for Inducing an Immune Response in a Subject
[00053] Described herein are nucleic acids encoding LMPl fusion proteins and compositions including such nucleic acids. The nucleic acids and compositions can be administered to a subject in order to induce an immune response in the subject (e.g., an immune response to a particular antigen) or to modulate an ongoing immune response in a subject (e.g., the naturally occurring but otherwise ineffective immune response to cancer). Nucleic acid molecules encoding LMP fusion proteins and antigens as described herein may be in the form of RNA (e.g., mRNA or synthetic chemically modified RNA) or in the form of DNA (e.g., cDNA, genomic DNA, and synthetic DNA). The DNA may be double- stranded or single- stranded, and if single- stranded, may be the coding (sense) strand or non-coding (anti-sense) template strand.
[00054] In one embodiment, a composition for inducing an immune response in a subject includes at least a first nucleic acid encoding at least a first fusion protein, the at least first fusion protein including a transmembrane domain of LMPl and two or more signaling domains selected from the group of immune activating receptors and/or the group of adaptor proteins, the at least first nucleic acid in an amount sufficient to induce an immune response in a subject. Any suitable immune activating receptor can be used. For example, the immune activating receptor can be a TLR or a TNFSFR. Examples of immune activating receptors include: TLRs 1-13, RIG-1, MDA5, FcyRIII, Dectinl, Dectin2, NODI, NOD2, CD16, IL-2R, TNFSFR (e.g., CD40, 4- IBB, RANK, TACI, OX40, CD27, GITR, LT R, and BAFFR), Type MI interferon receptor, TREM1, RTK, GPCR, Integrin, CCR7, CCR5, BCR (B cell receptor complex proteins). A fusion protein may include a signaling domain from a TLR and a signaling domain from a TNFSFR. Additionally or alternatively, a fusion protein may include a signaling domain from an immune activating receptor and a signaling domain from an adaptor protein. Any suitable adaptor protein may be used. Examples of adaptor proteins include CD3 zeta chain, MyD88, MyD88ATIR, IPS-1, TRIF, TRAM, TIRAP, MAL, BTK, RAC1, SYK, NALP3 (NLRP3), NALP3ALRR, NALP1, CARD9, DAI, IPAG, STING, Zap70, and LAT. The at least first nucleic acid may further encode an antigen.
[00055] Many vectors useful for introducing exogenous nucleic acids into target mammalian cells are available. The vectors may be episomal, e.g. plasmids, virus derived vectors such cytomegalovirus, adenovirus, adeno-associated virus (AAV), lentivirus etc., or may be integrated into the target cell genome, through homologous recombination or random integration, e.g. retrovirus derived vectors such MMLV, HIV-1, ALV, etc. In a typical embodiment, the at least first nucleic acid sequence is contained within a viral vector. In such an embodiment, recombinant virions (particles) containing the viral vector are administered to the subject. Viruses are naturally evolved vehicles which efficiently deliver their genes into host cells and therefore are desirable vector systems for the delivery of therapeutic nucleic acids. Preferred viral vectors exhibit low toxicity to the host cell and produce/deliver therapeutic quantities of the nucleic acid of interest (in some embodiments, in a tissue- specific manner). Retrovirus-based vectors, Lentivirus vectors, adenovirus based vectors, AAV-based vectors, rhabdovirus (e.g. VSV) vectors, poxvirus (e.g. vaccinia) vectors, alphavirus (e.g., Venezuelan equine encephalitis, Semliki Forest virus, Sindbis virus) vectors, and Herpesvirus (e.g., CMV) vectors are examples of viral vectors that may be used. Such recombinant virions may be pseudotyped.
[00056] In some embodiments, a composition can include a second nucleic acid encoding a second fusion protein as described herein. In such an embodiment, both the first and second nucleic acids can be contained within a single vector (e.g., a single viral vector) or multiple vectors (e.g., two, three, four, five, etc., viral vectors). A single composition can include two or more nucleic acids, each nucleic acid encoding a single LMP1 fusion protein, or can include two or more nucleic acids, one or more of which encodes multiple LMP1 fusion proteins.
[00057] Typically, the at least first nucleic acid (and the second nucleic acid if present) is in an amount effective for activating immune cells (e.g., dendritic cells) when administered to a subject. If a composition is being administered to a subject to prevent or treat a viral infection (e.g., HIV), the at least first nucleic acid (and the second nucleic acid if present) may be in an amount effective for inducing expression of cytokines and protecting primary CD4+ T cells from infection by a virus when administered to a subject. In one embodiment in which the composition is being used to prevent or treat HIV infection, the at least first fusion protein may include a transmembrane domain of LMP1 and a signaling domain from IPS-1. Similarly, if a composition is being administered to a subject to prevent or treat cancer, in one embodiment, the at least first fusion protein may include a transmembrane domain of LMP1 and a signaling domain from IPS-1, and is in an amount effective for preventing growth of a tumor or decreasing the size or growth of an existing tumor in a subject. In this embodiment of preventing or treating cancer, the composition may also include a nucleic acid encoding full-length LMP1 or a portion thereof (e.g., a composition including a nucleic acid encoding an LMP1 fusion protein and full- length LMP1, or a composition including a first nucleic acid encoding an LMP1 fusion protein and a second nucleic acid encoding full-length LMP1).
[00058] Also described herein is an LMP1 fusion protein that includes the LMP1 transmembrane domain fused to an amino acid sequence that acts as a cytoplasmic domain and that includes intracellular signaling motifs from two or more (e.g., 2, 3, 4, 5, 6, etc.) TLRs, TNFSFR, or adapter molecules, as well as nucleic acids encoding such fusion proteins. Such an artificial cytoplasmic domain composed of intracellular signaling motifs from multiple TLRs, TNFSFR, or adapter molecules can, as a single gene product, mimic the synergy induced by multiple LMP1 fusion proteins. Such a superactivating LMP1 construct (see FIG. 15, top right box) can be used as a vaccine adjuvant and immune stimulator. In one embodiment, a composition for inducing an immune response in a subject includes a nucleic acid encoding a fusion protein that includes a transmembrane domain of LMP1 and an amino acid sequence having two or more signaling motifs from immune activating receptors and/or adaptor proteins. In the composition, the nucleic acid is in an amount sufficient to induce an immune response in the subject (e.g., an immune response against HIV).
[00059] Vaccine formulations and vaccine adjuvants including nucleic acids encoding LMP1 fusion proteins are also described herein. A vaccine formulation for preventing or treating a disease or condition in a subject includes a pharmaceutically acceptable excipient and at least a first nucleic acid or protein encoded by such a nucleic acid for at least a first fusion protein that includes a transmembrane domain of LMP1 and at least one signaling domain from one or more of: an immune activating receptor and/or an adaptor protein. The at least first nucleic acid is in an amount sufficient to induce an immune response in a subject. In an embodiment in which the subject has a viral infection, typically, administration of the at least first nucleic acid induces anti-viral factors that directly reduce viral replication in an infected (e.g., chronically infected) subject. In one embodiment, the composition further includes a second nucleic acid encoding a second fusion protein that includes a transmembrane domain of LMPl and at least one signaling domain from one or more of: an immune activating receptor and/or an adaptor protein. Typically, the vaccine formulation is administered to vaccinate a subject against cancer or infection. Similarly, a vaccine adjuvant for enhancing an immune response to a vaccine includes a pharmaceutically acceptable excipient and at least a first nucleic acid encoding at least a first fusion protein that includes a transmembrane domain of LMPl and at least one signaling domain from one or more of: an immune activating receptor and/or an adaptor protein, plus an antigen, in an amount effective for enhancing an immune response to a vaccine (e.g., a conventional or currently available vaccine). Alternatively, the antigen may be encoded by a separate nucleic acid (a second or third nucleic acid). In such a vaccine adjuvant, a second nucleic acid encoding a second fusion protein may be included. In another embodiment, the antigen is administered to a subject in a separate composition. A vaccine formulation or adjuvant may include immune cells (e.g., dendritic cells) transduced with one or more nucleic acids encoding one or more LMPl fusion protein as described herein.
Methods of Inducing an Immune Response in a Subject
[00060] Methods of inducing an immune response against cancer or infection in a subject are described herein. In one embodiment, the method includes administering a composition as described herein to the subject in a therapeutically effective amount for inducing an immune response against the cancer or infection in the subject. In another embodiment, the method includes administering immune cells transduced with one or more nucleic acids encoding one or more LMPl fusion proteins as described herein to the subject in a therapeutically effective amount for inducing an immune response against the cancer or infection in the subject. In some embodiments, administering the composition to the subject vaccinates the subject against an infection or cancer or enhances the pre-existing immune response to cancer or an ongoing infection. Inducing an immune response against the cancer or the infection in the subject can include activating DCs, maturing DCs, and/or inducing expression of at least one cytokine in the subject. In a method of inducing an immune response against an HIV infection, administration of the composition induces type I interferon expression and/or activity, and protection of primary CD4+ T cells in the subject. In a method of inducing an immune response against cancer in a subject, administration of the composition prevents or decreases growth of cancer cells (e.g., a tumor, leukemia cells, etc.) in the subject. In methods of inducing an immune response against cancer and methods of inducing an immune response against an HIV infection, the at least first fusion protein may include a transmembrane domain of LMP1 and a signaling domain from IPS- 1 or a signaling domain from MyD88. In such a method, the nucleic acid encoding the at least first fusion protein may also encode a full-length LMP1 protein.
[00061] An immune response may include the activation of immune cells such as dendritic cells (DCs). DCs are a part of the immune system that act as APCs. DCs process antigen material and present it on their cell surface using MHC molecules. Pattern recognition receptors, such as TLRs, assist the DCs in detecting viruses and bacteria. After a DC is activated, it migrates to the lymph nodes. DCs interact with other cells within the immune system such as T cells and B cells. Stimulated DCs produce IL-12. IL-12 helps naive CD4+ T cells obtain a T helper cell type 1 (Thl) phenotype. Cytokines cause the development of T helper cell type 1 (Thl) and T helper cell type 2 (Th2) cells from naive CD4+ T cells. The Th phenotypes each produce particular cytokines and can be identified by specific cell- surface markers.
[00062] An immune response may be mounted to an antigen or antigens from any pathogen as a result of vaccination against that antigen or antigens. In one embodiment, the antigen may be derived from, but not limited to, pathogenic bacterial, fungal, or viral organisms, including Streptococcus species, Candida species, Brucella species, Salmonella species, Shigella species, Pseudomonas species, Bordetella species, Clostridium species, Norwalk virus, Bacillus anthracis, Mycobacterium tuberculosis, HIV, Chlamydia species, human Papillomaviruses, Influenza virus, Parainfluenza viruses, Paramyxovirus species, Herpes virus, Cytomegalovirus, Varicella-Zoster virus, Epstein-Barr virus, Hepatitis viruses (including HAV, HBV, and HCV), flaviviruses (including dengue and West Nile virus), filoviruses (including Ebola), coronaviruses (including SARS and MERS), Plasmodium species, Trichomonas species, Leishmania species, Neisseria meningitides, sexually transmitted disease agents, viral encephalitis agents, protozoan disease agents, fungal disease agents, and bacterial disease agents. A subject may be treated for any infectious pathogen, including those listed herein. [00063] In some embodiments, the antigen is derived from cancer cells. The vaccine may be used against any cancer or with any other therapy or intervention for cancer. Examples of cancers include HPV-induced cervical cancers (e.g., E7/E7 tumor associated antigens (TAA)), glioma, human melanoma (e.g., TRP-1, TRP-2, gp-100, MAGE-1, MAGE-3 and / or p53), breast cancer, and prostate cancer (e.g., TSA). Similarly for lung tumors, breast tumors, and leukemia, any suitable tumor associated antigen can be used, and many have been described. Many such TAA are common between various cancers (e.g., CEA, MUC-1, Her2, CD20). In addition, tumor cells may have uncategorized antigens such as those created by gene mutations (e.g., BRAF V600E in melanoma), gene transpositions (e.g., BCR-abl in chronic myelogenous leukemia), or simply tumor rejection antigens caused by overexpression of normal genes (e.g., telomerase).
[00064] The compositions, cells, kits, vaccines, vaccine adjuvants, and methods described herein solve the problem of properly activating and maturing DCs for therapeutic vaccination of DCs into patients as a treatment for cancer or chronic infections. They may also be used to develop prophylactic vaccines and other immune therapies dependent on immune activation.
[00065] In some embodiments, prior to or after administration of a composition or transduced immune cells as described herein to a subject, the subject's immune response is analyzed or measured. Any suitable biological sample can be tested for analyzing or measuring a subject's immune response. Examples of biological samples include blood, serum, plasma, urine, saliva and tissue. The sample may be tested using any suitable protocol or assay. Examples of suitable assays include enzyme-linked immunosorbent assays (ELISAs), Western blots, flow cytometry assays, immunofluorescence assays, qPCR, microarray analysis, etc.
Dendritic Cell Therapy
[00066] Immune cells such as dendritic cells transduced with nucleic acids encoding LMP1 fusion proteins can be used as an effective reagent for cell therapy for a subject in need thereof (e.g., a human subject having cancer or chronic infection). Immune cells (e.g., human dendritic cells) transduced with a nucleic acid or composition as described herein are encompassed within the invention. LMP1 fusion proteins can costimulate the activation of the transduced DC while allowing the transduced DC to induce T cell memory with secretion of IL-12 cytokine. Nucleic acids encoding LMP fusion proteins as described herein may be transfected into immune cells such as DCs. Alternatively, a viral vector may be used to introduce such a nucleic acid into immune cells (e.g., infection of the cells by a recombinant virus). The DCs may also be transduced with an antigen (e.g., an antigen encoded by the nucleic acid encoding an LMPl fusion protein, an antigen encoded by a separate nucleic acid). Administration of the DCs transduced with one or more nucleic acids encoding one or more LMPl fusion proteins to a subject can induce an immune response against cancer or infection including secretion of IL-12 by the DCs and activation of Thl cells. An example is provided in Fig. 7.
[00067] Given that DC therapy often uses RNA encoding the antigen or immune stimulatory proteins for transfection, LMPl fusion proteins can also be transfected as RNA. RNA is a safer method of transfection compared to DNA or viral vectors, given that RNA cannot integrate into the genome or be maintained. The RNA is degraded over time, and therefore LMPl encoded as RNA would not persist in the patient after treatment.
[00068] The compositions, nucleic acids, and cells described herein may be administered in combination with any other standard cell (e.g., DC) therapy; such methods are known to the skilled artisan and described in Remington's Pharmaceutical Sciences by E. W. Martin.
Kits for Inducing an Immune Response in a Subject
[00069] Described herein are kits for inducing an immune response and preventing or treating a disease or condition in a subject and for preparing a vaccine formulation. In one embodiment, a kit for inducing an immune response in a subject includes a composition including at least a first nucleic acid encoding at least a first fusion protein, the at least first fusion protein including a transmembrane domain of LMPl and at least one signaling domain from at least one of: an immune activating receptor and/or an adaptor protein. In the composition, the at least first nucleic acid is in an amount sufficient to induce an immune response in a subject. In another embodiment, the composition includes a second nucleic acid encoding a second fusion protein, the second fusion protein including a transmembrane domain of LMPl and at least one signaling domain from at least one of: an immune activating receptor and/or an adaptor protein. In some embodiments, the at least first nucleic acid also encodes an antigen. A kit as described herein can include a vaccine formulation that includes one or more nucleic acids encoding one or more LMP fusion proteins or a formulation that includes the proteins themselves. Similarly, a kit can include a vaccine adjuvant as described herein. In a kit, the instructions generally include one or more of: a description of the composition; dosage schedule and administration for prevention or treatment (e.g., vaccination) of cancer or infectious disease; precautions; warnings; indications; counter-indications; overdosage information; adverse reactions; animal pharmacology; clinical studies; and/or references. The instructions may be printed directly on the container (when present), or as a label applied to the container, or as a separate sheet, pamphlet, card, or folder supplied in or with the container. Generally, a kit as described herein also includes packaging. In some embodiments, the kit includes a sterile container which contains a therapeutic or prophylactic composition; such containers can be boxes, ampules, bottles, vials, tubes, bags, pouches, blister-packs, or other suitable container forms known in the art. Such containers can be made of plastic, glass, laminated paper, metal foil, or other materials suitable for holding cells or medicaments.
Effective Doses
[00070] The compositions, vaccines and cells described above are preferably administered to a mammal (e.g., non-human primate, bovine, canine, rodent, human) in an effective amount, that is, an amount capable of producing a desirable result in a treated subject (e.g., delaying or preventing onset of a disease or disorder in the subject). Toxicity and therapeutic efficacy of the compositions utilized in methods described herein can be determined by standard pharmaceutical procedures. As is well known in the medical and veterinary arts, dosage for any one animal depends on many factors, including the subject's size, body surface area, age, the particular composition to be administered, time and route of administration, general health, and other drugs being administered concurrently.
[00071] The amount of the therapeutic agent to be administered varies depending upon the manner of administration, the age and body weight of the patient, and with the pathology of the disease. A composition as described herein is typically administered at a dosage that activates and matures dendritic cells, as assayed using any assay that measures activation or maturation of dendritic cells, such as analyzing expression of one or more of: IL-l , IL-Ιβ, IFN-a, IFN-β, IFN- γ, IL-2, IL-4, IL-6, IL-10, IL-12, IL-15, IL-16, IL- 17, IL-18, and TNF-alpha.
[00072] Therapeutic compositions described herein can be administered to a subject by any suitable delivery vehicle (e.g., DNA plasmid, RNA, viral vector, recombinant virions, or purified protein) and route. The administration of a composition may include a therapeutically effective amount of a vaccine formulation or vaccine adjuvant. The composition may be provided in a dosage form that is suitable for local or systemic administration (e.g., parenterally, subcutaneously, intravenously, intramuscularly, intraperitoneally, intracranially). In various embodiments, the composition may be provided in a dosage form that is suitable for oral administration or intranasal administration. The compositions and vaccines may be formulated according to conventional pharmaceutical practice (see, e.g., Remington: The Science and Practice of Pharmacy (20th ed.), ed. A. R. Gennaro, Lippincott Williams & Wilkins, (2000) and Encyclopedia of Pharmaceutical Technology, eds. J. Swarbrick and J. C. Boylan, Marcel Dekker, New York (1988-1999)).
[00073] Compositions, vaccines, vaccine adjuvants, and cells as described herein may be administered parenterally by injection, infusion or implantation (subcutaneous, intravenous, intramuscular, intraperitoneal, or the like) in dosage forms, formulations, or via suitable delivery devices or implants containing conventional, non-toxic pharmaceutically acceptable carriers and adjuvants. The formulation and preparation of such compositions are well known to those skilled in the art of pharmaceutical formulation. Formulations can be found in Remington: The Science and Practice of Pharmacy, supra.
[00074] Compositions for parenteral use may be provided in unit dosage forms (e.g., in single-dose ampoules), or in vials containing several doses and in which a suitable preservative may be added. The composition may be in the form of a solution, a suspension, an emulsion, an infusion device, or a delivery device for implantation, or it may be presented as a dry powder to be reconstituted with water or another suitable vehicle before use. Apart from the agent that activates immune cells (e.g., activates and matures dendritic cells), the composition may include suitable parenterally acceptable carriers and/or excipients. The active therapeutic agent(s) may be incorporated into microspheres, microcapsules, nanoparticles, liposomes, or the like for controlled release. Furthermore, the composition may include suspending, solubilizing, stabilizing, pH-adjusting agents, tonicity adjusting agents, and/or dispersing agents.
[00075] As indicated above, the compositions described herein may be in a form suitable for sterile injection. To prepare such a composition, the suitable active therapeutic(s) are dissolved or suspended in a parenterally acceptable liquid vehicle. Among acceptable vehicles and solvents that may be employed are water, water adjusted to a suitable pH by addition of an appropriate amount of hydrochloric acid, sodium hydroxide or a suitable buffer, 1,3-butanediol, Ringer's solution, and isotonic sodium chloride solution and dextrose solution. The aqueous formulation may also contain one or more preservatives (e.g. , methyl, ethyl or n-propyl p- hydroxybenzoate). In cases where one of the compounds is only sparingly or slightly soluble in water, a dissolution enhancing or solubilizing agent can be added, or the solvent may include 10- 60% w/w of propylene glycol or the like.
[00076] Materials for use in the preparation of microspheres and/or microcapsules are, e.g., biodegradable/bioerodible polymers such as polygalactia poly-(isobutyl cyanoacrylate), poly(2- hydroxyethyl-L-glutam- nine) and, poly(lactic acid). Biocompatible carriers that may be used when formulating a controlled release parenteral formulation are carbohydrates (e.g. , dextrans), proteins (e.g. , albumin), lipoproteins, or antibodies. Materials for use in implants can be nonbiodegradable (e.g. , polydimethyl siloxane) or biodegradable (e.g., poly(caprolactone), poly(lactic acid), poly(glycolic acid) or poly(ortho esters) or combinations thereof).
[00077] Formulations for oral use include a liquid containing the active ingredient(s) (e.g., a nucleic acid encoding an LMPl fusion protein) in a mixture with non-toxic pharmaceutically acceptable excipients.
EXAMPLES
[00078] The present invention is further illustrated by the following specific examples. The examples are provided for illustration only and should not be construed as limiting the scope of the invention in any way.
[00079] Example 1 - Constitutively active gene-encoded combinations of cell receptor signaling domains and signaling adapter molecules.
[00080] The current technology uses TLR agonist chemicals and CD40 stimulation with agonistic antibodies. These methods rely on the presence of TLR and TNRSFR on the surface of immune cells. If those receptors are not present, or are downregulated, activation will be reduced. It would be preferable to have constitutive activation of the receptors independent of native expression by the cell. In the experiments described herein, combinations of LMPl-TLRs, LMPl fused to intracellular signaling adapter molecules, and LMPl-TNFSFR were shown to induce activation of the immune response. [00081] A model of LMPl fusion protein technology is illustrated in FIG. 1. In FIG. 1(A), the structure of LMPl is shown. LMPl contains a cytoplasmic signaling domain connected to a 6-segment transmembrane domain that aggregates the protein and anchors it in the cell membrane. The compositions, cells, vaccines, kits and methods described herein are based on the demonstration that LMPl is immunostimulatory and the cytoplasmic domain can be replaced by any immune signaling domain to create novel chimeras of LMPl fusion proteins. In FIG. 1(B), the LMPl fusion protein concept is shown on the right. The LMPl transmembrane/aggregation domain can be linked to multiple intracellular signaling domains from immune activating receptors (e.g., TLR3), from adapter proteins that interact with immune activating receptors (e.g., MyD88), and/or adaptor proteins further downstream in both adaptive and innate immune activation signaling (e.g., IPS-1). In FIG. 1(C), a model of a DNA or viral vector vaccine using LMPl fusion proteins is shown. Antigen and LMPl fusion proteins are encoded within the same polypeptide strand encoded by a nucleic acid (DNA, RNA, or viral vector genome) and directed to an immune cell. Following the introduction of the LMPl fusion protein, the cell produces antigen protein that is cleaved into antigenic peptides for presentation to T cells on MHC plus a constitutive immune activation signal from the cell signaling domains in the LMPl fusion protein. This immune activation signal induces cell activation and/or maturation and/or release of pro-inflammatory cytokines, and results in the induction of an effective immune response against the pathogen.
[00082] Referring to FIG. 2, LMPl fusion proteins with TLR, PRR, CD40, and/or signaling adapter proteins are potent inducers of NF-κΒ and IFN-β responses. 293 cells were co- transfected with an NF-κΒ or IFN-β promoter Luciferase reporter construct and plasmid DNA encoding one or more human or mouse LMPl fusion proteins. All the constructs indicated are fused to the transmembrane domain of LMPl with the exception of Flag-TRAF6 and deltaRIG-I which do not contain LMPl and are shown as positive controls for NF-κΒ and IFN-β induction. The construct labeled MyD88-CD40 contains a 3-protein fusion of LMPl, the MyD88 adapter protein, and the intracellular domain of CD40. In other samples two or more LMPl fusion proteins were co-transfected into the 293 cells (i.e. MyD88 + CD40 or TLR2 + TLR3 + TLR9). Certain LMPl fusion proteins and LMPl fusion protein combinations increased NF-κΒ mediated activity compared to parent LMPl. Expression was normalized to a pcDNA3.1 empty vector control. [00083] Referring to FIG. 3, LMPl fusions with IPS-1, TLR7, and MyD88-CD40 enhance activation and IL-6 cytokine secretion from a RAW 264.7 mouse macrophage cell line. Cells were transfected with either parent vector pcDNA3.1 or various LMPl fusion constructs with murine proteins. LMPl-MyD88-CD40 refers to a single fusion protein containing the LMPl transmembrane domain, MyD88, and the intracellular domain of CD40. MyD88dTIR-CD40 refers to an LMPl fusion protein containing the LMPl intracellular domain, MyD88 with a deletion of the TIR domain, and the CD40 intracellular domain. Cells were transfected using Lipofectamine LTX reagent. Levels of cell surface proteins were measured by Mean Fluorescence Index (MFI), and cytokines by cytometric bead array. The surprising result is that LMPl-IPS-1 fusion protein generated high levels of IL-6 and induced both maturation (CD40, CCR7) and activation (CD80, CD86) markers on the macrophage cell line. LMP1-TLR7 and LMPl-MyD88-CD40 were able to generate responses superior to wildtype LMPl, suggesting these fusion proteins are particularly potent immune activators. Dashed line denotes MFI of the pcDNA3.1 control. * p<0.05, ** p<0.01, *** p<0.001.
[00084] Referring to FIG. 4, human monocyte derived DCs transfected with LMP1-TLR fusion protein constructs induce activation, maturation, and cytokine secretion. All TLR and CD40 constructs refer to LMPl fusion proteins (i.e. TLR2 refers to LMP1-TLR2 fusion). Human DCs were electroporated with RNA encoding either control Gag protein or LMPl fusions with human TLR and CD40 genes. The "Mimic" cytokine mix (consisting of a mixture of TNF, IL-1- beta, IL-6, and PGE2) was used as a positive control (with or without electroporation with Gag RNA). LMPl and LMP1-TLR7 fusions induced a significant increase in CCR7 expression as measured by mean fluorescence index. All LMPl fusions had a trend toward increased expression of the maturation marker CD40 and the activation marker CD80. All LMPl fusion protein constructs increased secretion of the pro -inflammatory cytokine TNF-alpha and had a trend toward increased secretion of IL-6. Combinations of LMP1-TLR3, LMP1-TLR7, and LMP1-TLR9 were able to induce DC maturation compared to antigen alone or Mimic cytokine mix stimulation. * p<0.05, ** p<0.01, *** p<0.001.
[00085] Referring to FIG. 5, DCs transfected with combinations of LMP1-CD40 with LMP1- TLR fusion protein RNAs enhanced maturation, activation, and secretion of pro-inflammatory cytokines from human DC. Monocyte derived DC were electroporated with control (Gag) RNA or equal mixtures of LMP1-CD40 RNA with either Gag (a non-activating protein from HIV-1) or LMP1-TLR fusions. Combinations of LMP1-TLR fusions with LMP1-CD40 significantly increased secretion of the pro-inflammatory cytokines IL-6 (LMP1-TLR3, LMP1-TLR7, LMP1- TLR9 fusions) and TNF-alpha (LMP1-TLR3 and LMP1-TLR9 fusions) compared to LMP1- CD40 plus irrelevant control RNA (Gag). These data show for the first time that fusion of a TLR cell signaling domain to the LMP1 N-terminal transmembrane domain is sufficient to engage the TLR signaling pathway to activate cells.
[00086] Referring to FIG. 6, antigen-LMPl-MyD88-CD40 fusion protein enhances immune responses and control of viral infection in a mouse influenza DNA vaccine model. C57BL/6 mice were vaccinated with lOOug of plasmid DNA by intramuscular injection into the quadriceps muscles of both hindlimbs every two weeks for a total of 3 vaccinations. Seven days following the final vaccination, some of the mice were sacrificed and immune cells were harvested from their spleens or collected from their lungs by bronchoalveolar lavage (BAL). In parallel to this study, other mice were given an intranasal challenge with a standardized dose of live influenza A PR8 virus two weeks after the last vaccination and then sacrificed 4 days later to determine the amount of influenza viruses in lung tissue as determined by tissue culture 50% infectious dose (TCID50) units. The vaccinations used plasmids expressing the nucleoprotein (NP) antigen from Influenza A PR8 strain alone, or the NP antigen plus either full-length LMP1 (which includes its cell signaling domain) or NP antigen plus LMPl-MyD88-CD40 fusion protein. For the fusions with NP antigen, the nucleic acid sequences included an internal ribosome entry sequence (IRES) placed between the NP antigen coding sequence and the LMP1 or LMPl-MyD88-CD40 fusion sequence. This results in a single mRNA strand that is read by ribosomes from its 5' cap to translate the NP antigen until the ribosome reaches the in-frame stop codon after which the mRNA is released from the ribosome but is then available via the IRES to commence translation from at an internal ATG start codon for the translation of LMP1 or LMPl-MyD88-CD40 as a polypeptide that is separate from the NP antigen. As indicated schematically in Fig. 1C using a DC as an exemplary immune cell, the NP antigen polypeptide is available for proteolytic processing into peptides presented to T cells on MHC-I or MHC-II. At the same time, the separate LMP1 or LMPl-MyD88-CD40 polypeptide is available to anchor into the membrane and activate cell signaling pathways. As shown in Fig. 6, this design resulted in a superior vaccine response to this DNA vaccine. In the upper panels, splenocytes were exposed to NP antigen, incubated overnight, and then surface stained for either CD8 or CD4 and intracellularly stained for TNF using fluorochrome-labeled antibodies, and then analyzed for CD4, CD8, and TNF production by flow cytometry. When compared to DNA vaccination a plasmid for NP antigen alone, vaccination with plasmids for NP-IRES-LMP1 or especially vaccination with NP- IRES-MyD88-CD40 elicited significantly higher levels of NP-specific CD8+ T cells in the spleen and NP-specific CD4+ T cells in the lung BAL samples. For the CD8+ T cell response, as little as 0.001 ug/ml of NP peptide could stimulate these cells, a very low amount that indicates these CD8+ T cells have very high avidity for antigen, which is recognized in the art to be an in vitro correlate of strong protective function in vivo. These ex vivo measures of immune responses were confirmed in vivo by showing (bottom panel) that DNA vaccination with NP- IRES-LMP1 or NP-IRES-MyD88-CD40 protected mice from challenge by live influenza virus (where the horizontal line indicated the geometric mean of the lung viral loads in 3-4 mice).
[00087] Referring to FIG. 7, enhanced survival and reduced tumor growth using LMP1 fusion combinations as a dendritic cell cancer therapy was demonstrated. C57BL/6 mice were injected subcutaneously on the flank with 50,000 B16-F10 melanoma cells and tumors size measured every other day. Three days post-tumor injection, mice were given a total of 1 million C57BL/6 bone marrow derived dendritic cells subcutaneously into the flank opposite the tumor. 24 hours prior to injection these dendritic cells were electroporated with RNA encoding gplOO melanoma antigen plus a combination of RNAs encoding LMP1 and/or LMP1 fusion proteins, including LMP1, LMP1-TLR3, LMP1-TLR4, LMPl-MyD88, and LMP1-IPS 1. Dendritic cells were given to mice every 3 days for a total of three treatments. As a control, mice were treated with GVAX therapy (irradiated B16-F10 cells expressing GM-CSF injected intradermally on the opposite flank every 3 days for a total of 3 treatments). Dendritic cells expressing a combination of gplOO tumor antigen plus LMP1 and LMPl-MyD88 or gplOO tumor antigen plus LMP1 and LMPl-IPS-1 were able to enhance survival (p<0.05 and p<0.01 respectively) when compared to untreated tumors. Overall tumor growth was slowed in mice treated with these LMP1 fusion combinations.
[00088] Example 2 - Control of HIV replication by a novel constitutively active "Super- PRR"
[00089] Innate immune responses are key determinants of the outcome of HF/ infection, influencing critical events in the earliest stages of infection. Innate antiviral immune defenses are triggered through the recognition of conserved pathogen associated molecular pattern (PAMP) motifs within viral products by intracellular pattern recognition receptor (PRR) proteins in infected cells. Type I interferons (IFNcc and β) are induced directly in response to viral infection, resulting in an antiviral state for the cell. IFN-β promoter stimulator (IPS-1), also known as mitochondrial antiviral signaling protein (MAVS), virus-induced signaling adaptor (VISA), and CARD adaptor inducing IFN-β (Cardif), was recently identified as an adaptor linking RIG-I and Mda5 to the downstream signaling molecules, which has been found to have roles in anti-viral immune responses. LMP1 and chimeric LMP1-CD40 has been shown to increase maturation and activation of dendritic cells and macrophages and induce strong TH1 cytokine responses. In this study, the role of chimeric LMPl-IPS-1 fusion molecule in immune activation and anti-viral immune response was examined for the prevention of HIV-1 replication.
Results
[00090] Referring to FIG. 8, LMPl-IPS-1 is a potent inducer of NF-κΒ and IFN-β responses. 293 cells were co-transfected with an NF-κΒ or IFN-β promoter luciferase reporter construct and LMPl-IPS-1 along with controls LMP1, Flag-TRAF6 and deltaRIG-I. LMPl-IPS-1 increased IFN-β mediated activity compared to parent LMP1. Expression was normalized to a pcDNA3.1 empty vector control.
[00091] Referring to FIG. 9, LMPl-IPS-1 induces high IL-6 levels from RAW 264.7 mouse macrophage cell line. Cells were transfected with either parent vector pcDNA3.1 or LMP1-IPS- 1 construct. Cells were transfected using Lipofectamine LTX reagent. Levels of cytokines were measured by cytometric bead array (CBA). The LMPl-IPS-1 fusion protein generated high levels of IL-6 from the macrophage cell line suggesting that LMPl-IPS-1 as potent immune activator. ** p<0.01.
[00092] Referring to FIG. 10, LMPl-IPS-1 causes activation of a RAW 264.7 mouse macrophage cell line. Cells were transfected with either parent vector pcDNA3.1 or LMP1-IPS- 1 construct. Cells were transfected using Lipofectamine LTX reagent. Levels of cell surface proteins was measured by Mean Fluorescence Index (MFI). The LMP1 -IPS-1 fusion protein generated both maturation (CD40, CCR7) and activation (CD80, CD86) markers on the macrophage cell line suggesting that LMP1 -IPS-1 is potent immune activator. Dashed line denotes MFI of the pcDNA3.1 control. * p<0.05, ** p<0.01, *** p<0.001. [00093] Referring to FIG. 11, LMPl-IPS-1 prevents the replication of HIV-1 in TZM-bl cells in cis as well as in trans. In Fig. 11(A), TZM-bl cells, a HeLa cell line expressing CD4, CCR5, and a HIV-1 promoter β-galactosidase reporter gene, were transfected with plasmids expressing either EGFP (control), LMP1, or LMPl-IPS-1. Cells were then infected with serial dilutions of HIV-1 BaL strain. The level of HIV-1 infection was measured by β-galactosidase ELISA assay. The LMPl-IPS-1 fusion was able to protect TZM-bl cells from infection, providing evidence that LMPl-IPS-1 is able to induce cytokines and other innate antiviral mechanisms in this cell line. In Fig. 11(B), TZM-bl cells were cultured in a transwell assay with 293T cells that were transfected with LMPl-IPS l expressing or control plasmids suggesting that soluble factors produced by LMPl-IPS l expressing cells can effectively inhibit HIV-1 replication in TZM-bl cells.
[00094] Referring to FIG. 12, LMPl-IPS-1 prevents the replication of VSV-G pseudotyped scSr in TZM-bl cells in cis as well as in trans. In FIG. 12(A), TZM-bl cells, a HeLa cell line expressing CD4, CCR5, and a HIV-1 promoter β-galactosidase reporter gene, were transfected with plasmids expressing either EGFP (control), LMP1, or LMPl-IPS-1. Cells were then infected with serial dilutions of VSV-G pseudotyped scSIV. The level of SIV infection was measured by β-galactosidase ELISA assay. The LMPl-IPS-1 fusion was able to protect TZM-bl cells from infection, providing evidence that LMPl-IPS-1 is able to induce cytokines and other innate antiviral mechanisms in this cell line independent of Env mediated fusion. In FIG. 12(B), TZM-bl cells were cultured in a transwell assay with 293T cells that were transfected with LMPl-IPS l expressing or control plasmids suggesting that soluble factors produced by LMPl- IPS l expressing cells can effectively inhibit SF/ replication in TZM-bl cells.
[00095] Referring to FIG. 13, LMPl-IPS-1 prevents HIV replication in primary CD4+ T cells. Primary human CD4+ T cells from a healthy donor were infected with HIV-BaL at MOI of 0.1 and 1, cultured in a transwell assay with 293 cells that had been previously transfected with either empty plasmid (pcDNA3.1) or LMPl-IPS l. Again, LMPl-IPS l was able to inhibit HIV-1 replication in CD4+ T cells via one or more soluble factors. *** p<0.001
[00096] Referring to FIG. 14, LMPl-IPS l induces type I Interferons as well as interferon stimulated genes (ISGs). IPS-1 is involved in the induction of type I interferon, which is known to inhibit HIV-1 replication at high concentrations. Interferon alpha and beta mRNA levels were evaluated by qPCR. We also evaluated genes involved in CCR5 blockade (MIP-lb, RANTES, IP- 10) and interferon stimulated genes that are known to restrict HIV and other viruses (Viperin, ISG56). All of these genes were upregulated in 293T cells transfected with plasmid DNA encoding LMP1-IPS 1, but not plasmid DNA for LMPl or empty plasmid control (pcDNA3.1).
[00097] Referring to FIG. 15, LMPl and LMPl-IPS-1 fusion protein are able to protect cells from HIV-1 infection. In FIG. 15(A), TZM-bl cells, a HeLa cell line expressing CD4, CCR5, and a HIV-1 promoter β-galactosidase reporter gene, were transfected with plasmids expressing either EGFP (control), LMPl, or LMPl-IPS-1. Cells were then infected with serial dilutions of HIV-1 BaL strain virus. The level of HIV-1 infection was measured by p24 ELISA assay using β-galactosidase and OD405. Transfection with DNA for the LMPl-IPS-1 fusion protein was able to protect TZM-bl cells from infection, providing evidence that LMPl-IPS-1 is able to induce cytokines and/or other innate antiviral mechanisms in this cell line. In FIG. 15(B), primary human CD4+ T cells were cultured with HIV-1 recombinant virus expressing GFP or LMPl (as described by us in S. Gupta et al., Journal of Leukocyte Biology 90:389-398, 2011). Replication as measured by p24 ELISA on day 4 was significantly reduced for HIV-LMPl compared to BaL (wildtype) and GFP viral constructs. Other cells were infected with one virus for 2 days, washed, and then superinfected with a second viral construct. Initial infection with HIV-LMPl was able to significantly reduce replication following BaL or GFP viral construct superinfection. These data suggest that LMPl is able to induce an innate immune response capable of protecting primary CD4+ T cells from HF -1 infection. Similar data was observed with unfractionated PBMC.
[00098] Referring to FIG. 16, four examples of formulations of LMPl fusion proteins are described. Example 1 is the fusion of the LMPl transmembrane and aggregation domain to a single TLR signaling domain, TNF receptor, cytokine receptor, or adapter protein. Example 2 is the direct fusion of LMPl transmembrane domain with two or more signaling domains, including the use of the LMPl signaling domain itself. Example 3 describes the co-expression of two or more LMPl fusion proteins each encoding a different signaling domain. The example is given of LMP1-BAFFR fusion protein combined with an LMPl fusion to the B cell receptor signaling domain (LMP1-BCR). Example 4 describes the combination of full-length LMPl with LMPl fusion proteins, for example LMPl + LMPl-IPS-1. [00099] Conclusions
[000100] LMPl-IPS-1 is a potent inducer of NF-κΒ and IFN-β responses. LMPl-IPS-1 causes activation and maturation of the RAW 264.7 macrophage cell line by increasing expression of surface markers such as CD80, CD86, CD40, IA/IE and CCR7. LMPl-IPS-1 induces very high levels of IL-6 from transfected RAW 264.7 cells. LMPl-IPS-1 prevents the replication of both HIV and VSV-G pseudotyped scSIV in TZM-bl cells as well as primary CD4+T cells both in cis and in trans. LMPl-IPS-1 induces type 1 interferons, interferons stimulated genes (ISGs) and chemokines genes implicated in inhibition of HIV replication. In conclusion, these studies suggest that LMPl-IPS-1 is immuno stimulatory and induces a strong anti- viral immune response for the prevention of HIV-1 infection of primary CD4+ T cells.
[000101] Example 3 - DNA and amino acid sequences for LMP1 fusion constructs: LMP1 fusions to TLR, TNFSFR or other immune activating receptors.
SEQ ID NO: l is Epstein Barr Virus latent membrane protein 1 (without the cytoplasmic domain) fused to the cytoplasmic domain of murine TLR7.
LMPl-muTLR7
Italicized/bolded sequence: LMP1 sequence (Transmembrane Domain)
Non-italicized/bolded sequence: Murine TLR7 sequence (Cytoplasmic Domain)
ATGGAACACGACCTTGAGAGGGGCCCACCGGGCCCGCGACGGCCCCCTCGAGGACCC
CCCCTCTCCTCTTCCCTAGGCCTTGCTCTCCTTCTCCTCCTCTTGGCGCTACTGTTTTG
GCTGTACATCGTTATGAGTGACTGGACTGGAGGAGCCCTCCTTGTCCTCTATTCCTTT
GCTCTCATGCTTATAATTATAATTTTGATCATCTTTATCTTCAGAAGAGACCTTCTCTGT
CCACTTGGAGCCCTTTGTATACTCCTACTGATGATCACCCTCCTGCTCATCGCTCTCTG
GAATTTGCACGGACAGGCATTGTTCCTTGGAATTGTGCTGTTCATCTTCGGGTGCTTA
CTTGTCTTAGGTATCTGGATCTACTTATTGGAGATGCTCTGGCGACTTGGTGCCACCA
TCTGGCAGCTTTTGGCCTTCTTCCTAGCCTTCTTCCTAGACCTCATCCTGCTCATTATT
GCTCTCTATCTACAACAAAACTGGTGGACTCTATTGGTTGATCTCCTTTGGCTCCTCCT
GTTTC TGGCGA TTTTAA TC TGGA TGTA TTACCA TGGA CAA CGA AC A AC A AGTC ACCTC
TTTTTCTGGGATATGTGGTACATTTATTATTTTTGGAAAGCAAAGATAAAGGGGTAT
CAGCATCTGCAATCCATGGAGTCTTGTTATGATGCTTTTATTGTGTATGACACTAAAA
ACTCAGCTGTGACAGAATGGGTTTTGCAGGAGCTGGTGGCAAAATTGGAAGATCCA
AGAGAAAAACACTTCAATTTGTGTCTAGAAGAAAGAGACTGGCTACCAGGACAGCC
AGTTCTAGAAAACCTTTCCCAGAGCATACAGCTCAGCAAAAAGACAGTGTTTGTGAT GACACAGAAATATGCTAAGACTGAGAGTTTTAAGATGGCATTTTATTTGTCTCATCA
GAGGCTCCTGGATGAAAAAGTGGATGTGATTATCTTGATATTCTTGGAAAAGCCTCT
TCAGAAGTCTAAGTTTCTTCAGCTCAGGAAGAGACTCTGCAGGAGCTCTGTCCTTGA
GTGGCCTGCAAATCCACAGGCTCACCCATACTTCTGGCAGTGCCTGAAAAATGCCCT
GACCACAGACAATCATGTGGCTTATAGTCAAATGTTCAAGGAAACAGTCTAG (SEQ
ID NO: l)
SEQ ID NO:2 is Epstein Barr Virus latent membrane protein 1 (without the cytoplasmic domain) fused to the cytoplasmic domain of murine TLR7.
Protein:
MEHDLERGPPGPRRPPRGPPLS S S LGLALLLLLLALLFWLYIVMS D WTGG ALLVLYS FAL MLIIIILIIFIFRRDLLCPLGALCILLLMITLLLIALWNLHGQALFLGIVLFIFGCLLVLGIWIY LLEMLWRLGATIWQLLAFFLAFFLDLILLIIALYLQQNWWTLLVDLLWLLLFLAILIWMY YHGQRTTS HLFFWDM WYIY YFWKAKIKG YQHLQS MES C YD AFIV YDTKNS A VTEWVL QELV AKLEDPREKHFNLCLEERD WLPGQP VLENLS QS IQLS KKTVF VMTQKY AKTES FK M AF YLS HQRLLDEKVD VIILIFLEKPLQKS KFLQLRKRLCRS S VLEWP ANPQ AHP YFWQ CLKN ALTTDNH V A YS QMFKETV (SEQ ID NO:2)
SEQ ID NO:3 is Epstein Barr Virus latent membrane protein 1 (without the cytoplasmic domain) fused to the cytoplasmic domain of murine BAFF receptor.
LMPl-muBAFFR
Italicized/bolded sequence: LMP1 sequence (Transmembrane Domain)
Non-italicized/bolded sequence: Murine BAFFR (Cytoplasmic Domain)
ATGGAACACGACCTTGAGAGGGGCCCACCGGGCCCGCGACGGCCCCCTCGAGGACCC
CCCCTCTCCTCTTCCATAGGCCTTGCTCTCCTTCTCCTGCTCTTGGCGCTACTGTTTTG
GCTGTACATCATTATGAGTAACTGGACTGGAGGAGCCCTCCTTGTCCTCTATGCCTTT
GCTCTCATGCTTGTGATTATCATTTTGATCATCTTTATCTTCAGAAGAGACCTTCTCTG
TCCACTTGGAGCCCTTTGTCTACTCCTACTGATGATCACCCTCCTGCTCATCGCTCTCT
GGAATTTGCACGGACAGGCATTGTACCTTGGAATTGTGCTGTTCATCTTCGGGTGCTT
ACTTGTCTTAGGTCTCTGGATCTACTTATTGGAGATTCTCTGGCGACTTGGTGCCACCA
TCTGGCAGCTTTTGGCCTTCTTCCTAGCCTTCTTCCTAGACATCATCCTGCTCATTATT
GCTCTCTATCTACAACAAAACTGGTGGACTCTATTGGTTGATCTCCTTTGGCTCCTCCT
GTTTCTGGCGA TTTTAA TCTGGA TGTA TTACCA TGGA CAA CGA AGTCTGGTG AGCTGG
AGGTGGCGTCAACAGCTCAGGACGGCCTCCCCAGACACTTCAGAAGGAGTCCAGCA
AGAGTCCCTGGAAAATGTCTTTGTACCCTCCTCAGAAACCCCTCATGCCTCAGCTCC
TACCTGGCCTCCGCTCAAAGAAGATGCAGACAGCGCCCTGCCACGCCACAGCGTCC
CGGTGCCCGCCACAGAACTGGGCTCCACCGAGCTGGTGACCACCAAGACAGCTGGC
CCAGAGCAATAG (SEQ ID NO:3) SEQ ID NO:4 is Epstein Barr Virus latent membrane protein 1 (without the cytoplasmic domain) fused to the cytoplasmic domain of murine BAFF receptor.
Protein:
MEHDLERGPPGPRRPPRGPPLS S S IGLALLLLLLALLFWLYIIMS NWTGG ALLVLY AF AL MLVIIILIIFIFRRDLLCPLGALCLLLLMITLLLIALWNLHGQALYLGIVLFIFGCLLVLGLW IYLLEILWRLGATIWQLLAFFLAFFLDIILLIIALYLQQNWWTLLVDLLWLLLFLAILIWM Y YHGQRS LVS WRWRQQLRT AS PDTS EG VQQES LEN VF VPS S ETPH AS APTWPPLKED A DS ALPRHS VP VP ATELGS TELVTTKT AGPEQ (SEQ ID NO:4)
SEQ ID NO:5 is Epstein Barr Virus latent membrane protein 1 (without the cytoplasmic domain) fused to the CARD domain of murine RIG-I.
LMPl-muRIGl
Italicized/bolded sequence: LMP1 sequence (Transmembrane Domain)
Non-italicized/bolded sequence: Mouse RIG1 (CARD domain)
ATGGAACACGACCTTGAGAGGGGCCCACCGGGCCCGCGACGGCCCCCTCGAGGACCC
CCCCTCTCCTCTTCCATAGGCCTTGCTCTCCTTCTCCTGCTCTTGGCGCTACTGTTTTG
GCTGTACATCATTATGAGTAACTGGACTGGAGGAGCCCTCCTTGTCCTCTATGCCTTT
GCTCTCATGCTTGTGATTATCATTTTGATCATCTTTATCTTCAGAAGAGACCTTCTCTG
TCCACTTGGAGCCCTTTGTCTACTCCTACTGATGATCACCCTCCTGCTCATCGCTCTCT
GGAATTTGCACGGACAGGCATTGTACCTTGGAATTGTGCTGTTCATCTTCGGGTGCTT
ACTTGTCTTAGGTCTCTGGATCTACTTATTGGAGATTCTCTGGCGACTTGGTGCCACCA
TCTGGCAGCTTTTGGCCTTCTTCCTAGCCTTCTTCCTAGACATCATCCTGCTCATTATT
GCTCTCTATCTACAACAAAACTGGTGGACTCTATTGGTTGATCTCCTTTGGCTCCTCCT
GTTTCTGGCGA TTTTAA TCTGGA TGTA TTACCA TGGA CAA CGA ATG ACCGCGGCGC AG
CGGCAGAATCTGCAAGCATTCAGAGACTATATCAAGAAGATTCTGGACCCCACCTA
CATCCTCAGCTACATGAGTTCCTGGCTCGAGGATGAGGAGGTGCAGTACATTCAGGC
TGAGAAGAACAACAAGGGCCCAATGGAAGCTGCCTCACTCTTCCTCCAGTACCTGTT
GAAGCTGCAGTCAGAGGGCTGGTTCCAGGCCTTTTTGGATGCCCTGTACCATGCAGG
TTACTGTGGACTTTGTGAAGCCATCGAAAGTTGGGACTTTCAAAAAATTGAAAAGTT
AGAGGAACACAGATTACTTTTAAGACGTTTAGAACCAGAATTTAAGGCCACAGTTG
ATCCAAATGATATCCTTTCTGAACTATCCGAATGTTTGATTAATCAGGAATGTGAAG
AAATCAGACAGATCCGAGACACTAAAGGGAGAATGGCAGGTGCGGAGAAGATGGC
CGAATGTCTTATCAGATCCGACAAGGAAAACTGGCCAAAGGTCTTGCAATAG (SEQ
ID NO:5)
SEQ ID NO: 6 is Epstein Barr Virus latent membrane protein 1 (without the cytoplasmic domain) fused to the CARD domain of murine RIG-I
Protein: MEHDLERGPPGPRRPPRGPPLS S S IGLALLLLLLALLFWLYIIMS NWTGG ALLVLY AF AL
MLVIIILIIFIFRRDLLCPLGALCLLLLMITLLLIALWNLHGQALYLGIVLFIFGCLLVLGLW
IYLLEILWRLGATIWQLLAFFLAFFLDIILLIIALYLQQNWWTLLVDLLWLLLFLAILIWM
YYHGQRMTAAQRQNLQAFRDYIKKILDPTYILSYMSSWLEDEEVQYIQAEKNNKGPME
AASLFLQYLLKLQSEGWFQAFLDALYHAGYCGLCEAIESWDFQKIEKLEEHRLLLRRLE
PEFKATVDPNDILSELSECLINQECEEIRQIRDTKGRMAGAEKMAECLIRSDKENWPKVL
Q (SEQ ID NO:6)
[000102] Example 4 - DNA and amino acid sequences for LMP1 fusion constructs: LMP1 fusions to immune activation adapter proteins.
SEQ ID NO:7 is Epstein Barr Virus latent membrane protein 1 (without the cytoplasmic domain) fused to human MyD88.
LMPl-huMvD88
Italicized/bolded sequence: LMP1 sequence (Transmembrane Domain)
Non-italicized/bolded sequence: human MyD88
ATGGAACACGACCTTGAGAGGGGCCCACCGGGCCCGCGACGGCCCCCTCGAGGACCC
CCCCTCTCCTCTTCCATAGGCCTTGCTCTCCTTCTCCTGCTCTTGGCGCTACTGTTTTG
GCTGTACATCATTATGAGTAACTGGACTGGAGGAGCCCTCCTTGTCCTCTATGCCTTT
GCTCTCATGCTTGTGATTATCATTTTGATCATCTTTATCTTCAGAAGAGACCTTCTCTG
TCCACTTGGAGCCCTTTGTCTACTCCTACTGATGATCACCCTCCTGCTCATCGCTCTCT
GGAATTTGCACGGACAGGCATTGTACCTTGGAATTGTGCTGTTCATCTTCGGGTGCTT
ACTTGTCTTAGGTCTCTGGATCTACTTATTGGAGATTCTCTGGCGACTTGGTGCCACCA
TCTGGCAGCTTTTGGCCTTCTTCCTAGCCTTCTTCCTAGACATCATCCTGCTCATTATT
GCTCTCTATCTACAACAAAACTGGTGGACTCTATTGGTTGATCTCCTTTGGCTCCTCCT
GTTTCTGGCGA TTTTAA TCTGGA TGTA TTACCA TGGA CAA CGA ATGGCTGC AGG AGGT
CCCGGCGCGGGGTCTGCGGCCCCGGTCTCCTCCACATCCTCCCTTCCCCTGGCTGCTC
TCAACATGCGAGTGCGGCGCCGCCTGTCTCTGTTCTTGAACGTGCGGACACAGGTGG
CGGCCGACTGGACCGCGCTGGCGGAGGAGATGGACTTTGAGTACTTGGAGATCCGG
CAACTGGAGACACAAGCGGACCCCACTGGCAGGCTGCTGGACGCCTGGCAGGGACG
CCCTGGCGCCTCTGTAGGCCGACTGCTCGAGCTGCTTACCAAGCTGGGCCGCGACGA
CGTGCTGCTGGAGCTGGGACCCAGCATTGAGGAGGATTGCCAAAAGTATATCTTGA
AGCAGCAGCAGGAGGAGGCTGAGAAGCCTTTACAGGTGGCCGCTGTAGACAGCAGT
GTCCCACGGACAGCAGAGCTGGCGGGCATCACCACACTTGATGACCCCCTGGGGCA
TATGCCTGAGCGTTTCGATGCCTTCATCTGCTATTGCCCCAGCGACATCCAGTTTGTG
CAGGAGATGATCCGGCAACTGGAACAGACAAACTATCGACTGAAGTTGTGTGTGTC
TGACCGCGATGTCCTGCCTGGCACCTGTGTCTGGTCTATTGCTAGTGAGCTCATCGA
AAAGAGGTGCCGCCGGATGGTGGTGGTTGTCTCTGATGATTACCTGCAGAGCAAGG
AATGTGACTTCCAGACCAAATTTGCACTCAGCCTCTCTCCAGGTGCCCATCAGAAGC
GACTGATCCCCATCAAGTACAAGGCAATGAAGAAAGAGTTCCCCAGCATCCTGAGG TTCATCACTGTCTGCGACTACACCAACCCCTGCACCAAATCTTGGTTCTGGACTCGC CTTGCCAAGGCCTTGTCCCTGTGA (SEQ ID NO:7)
SEQ ID NO:8 is Epstein Barr Virus latent membrane protein 1 (without the cytoplasmic domain) fused to human MyD88.
Protein:
MEHDLERGPPGPRRPPRGPPLS S S IGLALLLLLLALLFWLYIIMS NWTGG ALLVLY AF AL MLVIIILIIFIFRRDLLCPLGALCLLLLMITLLLIALWNLHGQALYLGIVLFIFGCLLVLGLW IYLLEILWRLGATIWQLLAFFLAFFLDIILLIIALYLQQNWWTLLVDLLWLLLFLAILIWM Y YHGQRM A AGGPG AGS A AP VS S TS S LPLA ALNMR VRRRLS LFLN VRTQ V A AD WT ALA EEMDFEYLEIRQLETQADPTGRLLDAWQGRPGASVGRLLELLTKLGRDDVLLELGPSIEE DCQKYILKQQQEEAEKPLQVAAVDSSVPRTAELAGITTLDDPLGHMPERFDAFICYCPSD IQF VQEMIRQLEQTN YRLKLC VS DRD VLPGTC VWS IAS ELIEKRCRRM V V V VS DD YLQS KECDFQTKFALSLSPGAHQKRLIPIKYKAMKKEFPSILRFITVCDYTNPCTKSWFWTRLA KALSL (SEQ ID NO:8)
SEQ ID NO:9 is Epstein Barr Virus latent membrane protein 1 (without the cytoplasmic domain) fused to murine IPS-1.
LMPl-muIPS-1
Italicized/bolded sequence: LMP1 sequence (Transmembrane Domain)
Non-italicized/bolded sequence: Murine IPS-1
ATGGAACACGACCTTGAGAGGGGCCCACCGGGCCCGCGACGGCCCCCTCGAGGACCC
CCCCTCTCCTCTTCCATAGGCCTTGCTCTCCTTCTCCTGCTCTTGGCGCTACTGTTTTG
GCTGTACATCATTATGAGTAACTGGACTGGAGGAGCCCTCCTTGTCCTCTATGCCTTT
GCTCTCATGCTTGTGATTATCATTTTGATCATCTTTATCTTCAGAAGAGACCTTCTCTG
TCCACTTGGAGCCCTTTGTCTACTCCTACTGATGATCACCCTCCTGCTCATCGCTCTCT
GGAATTTGCACGGACAGGCATTGTACCTTGGAATTGTGCTGTTCATCTTCGGGTGCTT
ACTTGTCTTAGGTCTCTGGATCTACTTATTGGAGATTCTCTGGCGACTTGGTGCCACCA
TCTGGCAGCTTTTGGCCTTCTTCCTAGCCTTCTTCCTAGACATCATCCTGCTCATTATT
GCTCTCTATCTACAACAAAACTGGTGGACTCTATTGGTTGATCTCCTTTGGCTCCTCCT
GTTTC TGGC GA TTTTAA TC TGGA TGTA TTA C CA TGGA CAA CGA ATG AC ATTTGCTG AG
GACAAGACCTATAAGTATATCCGAGACAACCACAGCAAGTTTTGCTGTGTTGACGTT
CTGGAGATCCTGCCTTACCTGTCCTGCCTCACAGCTAGTGACCAGGATCGACTGCGG
GCTTCCTACAGGCAGATCGGGAACCGGGACACACTCTGGGGACTCTTCAATAATCTC
CAGCGCCGGCCTGGCTGGGTGGAGGTCTTCATCCGGGCACTGCAGATCTGTGAGCTG
CCTGGGCTGGCTGATCAAGTGACTCGAGTTTATCAGAGCTACCTGCCTCCGGGGACC
TCACTCCGCTCCCTAGAGCCACTGCAGTTACCAGACTTTCCTGCTGCGGTTTCTGGAC
CCTCTGCATTTGCGCCAGGTCACAACATCCCTGACCATGGCTTACGAGAGACACCAA GTTGCCCCAAGCCTGTCCAGGACACCCAGCCACCAGAGTCCCCAGTAGAGAATTCA
GAGCAACTCCTCCAGACCAACTCCGGGGCCGTCGCGAGGATGTCTGGTGGCTCTTTG
ATACCCTCTCCTAACCAGCAGGCTCTCAGCCCTCAGCCCTCCAGAGAGCATCAAGAG
CAAGAACCAGAACTGGGTGGCGCCCACGCAGCAAATGTTGCCTCTGTTCCCATAGC
AACCTATGGACCTGTGTCTCCAACCGTTTCCTTCCAGCCCCTTCCACGTACTGCCCTG
AGGACAAACCTCTTGTCTGGGGTCACAGTATCAGCCCTATCTGCTGATACCTCTTTGT
CCTCCTCGTCCACTGGATCAGCTTTTGCAAAGGGAGCTGGTGACCAGGCCAAAGCTG
CCACCTGTTTCAGTACTACACTCACCAATTCTGTGACTACCAGCTCAGTGCCTTCTCC
CAGATTGGTCCCAGTAAAAACCATGTCTTCCAAGTTGCCCCTCAGTTCAAAGTCCAC
TGCTGCGATGACGTCTACTGTGCTCACCAATACAGCGCCATCAAAATTACCCAGCAA
CTCAGTGTATGCGGGCACAGTGCCATCCAGAGTGCCTGCTAGTGTGGCCAAAGCAC
CTGCCAACACAATACCACCTGAGAGGAACAGCAAGCAAGCCAAGGAGACCCCGGA
GGGTCCAGCAACCAAAGTCACCACTGGAGGCAACCAGACTGGACCAAATAGCAGTA
TCAGGAGCTTGCACTCTGGACCAGAGATGAGCAAGCCAGGTGTGCTGGTATCCCAG
TTGGACGAGCCATTCTCAGCCTGCTCTGTGGACCTTGCCATTAGCCCTAGCAGCTCC
TTGGTCTCAGAACCCAACCATGGTCCAGAGGAGAATGAGTATTCGTCCTTTAGAATC
CAGGTAGACGAAAGCCCCAGTGCTGATCTATTAGGAAGCCCTGAGCCACTAGCCAC
CCAGCAGCCCCAAGAAGAGGAAGAACATTGTGCCAGTTCAATGCCCTGGGCTAAGT
GGCTTGGGGCCACCAGTGCACTCTTGGCTGTATTCCTGGCAGTGATGCTGTACCGTA
GTAGGCGCCTGGCCCAGTGA (SEQ ID NO:9)
SEQ ID NO: 10 is Epstein Barr Virus latent membrane protein 1 (without the cytoplasmic domain) fused to murine IPS-1.
Protein:
MEHDLERGPPGPRRPPRGPPLS S S IGLALLLLLLALLFWLYIIMS NWTGG ALLVLY AF AL MLVIIILIIFIFRRDLLCPLGALCLLLLMITLLLIALWNLHGQALYLGIVLFIFGCLLVLGLW IYLLEILWRLGATIWQLLAFFLAFFLDIILLIIALYLQQNWWTLLVDLLWLLLFLAILIWM YYHGQRMTFAEDKTYKYIRDNHSKFCCVDVLEILPYLSCLTASDQDRLRASYRQIGNRD TLWGLFNNLQRRPGWVEVFIRALQICELPGLADQVTRVYQSYLPPGTSLRSLEPLQLPDF P A A VS GPS AFAPGHNIPDHGLRETPS CPKP VQDTQPPES P VENS EQLLQTNS G A V ARMS G GS LIPS PNQQ ALS PQPS REHQEQEPELGG AH A AN V AS VPIATYGP VS PTVS FQPLPRT ALR TNLLS G VT VS ALS ADTS LS S S S TGS AFAKG AGDQ AKA ATCFS TTLTNS VTTS S VPS PRLV P VKTMS S KLPLS S KS T A AMTS TVLTNT APS KLPS NS V Y AGT VPS RVP AS V AKAP ANTIPP ERNS KQ AKETPEGP ATKVTTGGNQTGPNS S IRS LHS GPEMS KPG VLVS QLDEPFS ACS VD LAIS PS S S LVS EPNHGPEENE YS S FRIQ VDES PS ADLLGS PEPLATQQPQEEEEHC AS S MP WAKWLG ATS ALLA VFLA VMLYRS RRLAQ (SEQ ID NO: 10)
SEQ ID NO: 11 is Epstein Barr Virus latent membrane protein 1 (without the cytoplasmic domain) fused to the cytoplasmic domain of human CD3 zeta chain.
LMPl-huCD3 zeta chain Italicized/bolded sequence: LMP1 sequence (Transmembrane Domain)
Non-italicized/bolded sequence: huCD3 zeta chain (Cytoplasmic Domain)
ATGGAACACGACCTTGAGAGGGGCCCACCGGGCCCGCGACGGCCCCCTCGAGGACCC
CCCCTCTCCTCTTCCCTAGGCCTTGCTCTCCTTCTCCTCCTCTTGGCGCTACTGTTTTG
GCTGTACATCGTTATGAGTGACTGGACTGGAGGAGCCCTCCTTGTCCTCTATTCCTTT
GCTCTCATGCTTATAATTATAATTTTGATCATCTTTATCTTCAGAAGAGACCTTCTCTGT
CCACTTGGAGCCCTTTGTATACTCCTACTGATGATCACCCTCCTGCTCATCGCTCTCTG
GAATTTGCACGGACAGGCATTGTTCCTTGGAATTGTGCTGTTCATCTTCGGGTGCTTA
CTTGTCTTAGGTATCTGGATCTACTTATTGGAGATGCTCTGGCGACTTGGTGCCACCA
TCTGGCAGCTTTTGGCCTTCTTCCTAGCCTTCTTCCTAGACCTCATCCTGCTCATTATT
GCTCTCTATCTACAACAAAACTGGTGGACTCTATTGGTTGATCTCCTTTGGCTCCTCCT
GTTTC TGGCGA TTTTAA TC TGGA TGTA TTACCA TGGA CAA CGA AG AGTG A AGTTC AGC
AGGAGCGCAGACGCCCCCGCGTACCAGCAGGGCCAGAACCAGCTCTATAACGAGCT
CAATCTAGGACGAAGAGAGGAGTACGATGTTTTGGACAAGAGACGTGGCCGGGACC
CTGAGATGGGGGGAAAGCCGCAGAGAAGGAAGAACCCTCAGGAAGGCCTGTACAA
TGAACTGCAGAAAGATAAGATGGCGGAGGCCTACAGTGAGATTGGGATGAAAGGC
GAGCGCCGGAGGGGCAAGGGGCACGATGGCCTTTACCAGGGTCTCAGTACAGCCAC
CAAGGACACCTACGACGCCCTTCACATGCAGGCCCTGCCCCCTCGCTAA (SEQ ID
NO: l l)
SEQ ID NO: 12 is Epstein Barr Virus latent membrane protein 1 (without the cytoplasmic domain) fused to the cytoplasmic domain of human CD3 zeta chain.
Protein:
MEHDLERGPPGPRRPPRGPPLS S S LGLALLLLLLALLFWLYIVMS D WTGG ALLVLYS FAL
MLIIIILIIFIFRRDLLCPLGALCILLLMITLLLIALWNLHGQALFLGIVLFIFGCLLVLGIWIY
LLEMLWRLGATIWQLLAFFLAFFLDLILLIIALYLQQNWWTLLVDLLWLLLFLAILIWMY
YHGQRRVKFSRSADAPAYQQGQNQLYNELNLGRREEYDVLDKRRGRDPEMGGKPQRR
KNPQEGLYNELQKDKMAEAYSEIGMKGERRRGKGHDGLYQGLSTATKDTYDALHMQ
ALPPR (SEQ ID NO: 12)
SEQ ID NO: 13 is Influenza NP protein co-expressed by use of an IRES sequence with Epstein Barr Virus latent membrane protein 1 (without the cytoplasmic domain) fused to murine MyD88 and the cytoplasmic domain of murine CD40 as a single 3-protein chimera.
NP-IRES-LMPl-muMvD88-muCD40
Italicized sequence: Influenza NP sequence
Non-italicized sequence: IRES
Italicized/bolded sequence: LMP1 sequence (Transmembrane Domain)
Non-italicized/bolded sequence: Murine MyD88 sequence Underlined sequence: Linker
Bolded sequence: Murine CD40 sequence (cytoplasmic domain)
A TGGCGTCTCAA GGCA CCAAA CGA TCTTA CGAA CA GA TGGA GA CTGA TGGA GAA CGCCA G
AA TGCCA CTGAAA TCA GA GCA TCCGTCGGAAAAA TGA TTGGTGGAA TTGGA CGA TTCTA CA
Ί CCAAA TGTGCA CCGAA CTCAAA CTCA GTGA TTA TGA GGGA CGGTTGA TCCAAAA CAGCTT
AA CAA TA GA GA GAA TGGTGCTCTCTGCTTTTGA CGAAA GGA GAAA TAAA TA CCTTGAA GAA
CATCCCAGTGCGGGGAAAGATCCTAAGAAAACTGGAGGACCTATATACAGGAGAGTAAAC
GGAAA GTGGA TGA GA GAA CTCA TCCTTTA TGA CAAA GAA GAAA TAA GGCGAA TCTGGCGC
CAAGCTAATAATGGTGACGATGCAACGGCTGGTCTGACTCACATGATGATCTGGCATTCCA
A TTTGAA TGA TGCAA CTTA TCA GA GGA CAA GA GCTCTTGTTCGCA CCGGAA TGGA TCCCA G
GATGTGCTCTCTGATGCAAGGTTCAACTCTCCCTAGGAGGTCTGGAGCCGCAGGTGCTGC
A GTCAAA GGA GTTGGAA CAA TGGTGA TGGAA TTGGTCA GAA TGA TCAAA CGTGGGA TCAA T
GATCGGAACTTCTGGAGGGGTGAGAATGGACGAAAAACAAGAATTGCTTATGAAAGAATG
TGCAA CA TTCTCAAA GGGAAA TTTCAAA CTGCTGCA CAAAAA GCAA TGA TGGA TCAA GTGA
GA GA GA GCCGGAA CCCA GGGAA TGCTGA GTTCGAA GA TCTCA CTTTTCTA GCA CGGTCTG
CA CTCA TA TTGA GA GGGTCGGTTGCTCA CAA GTCCTGCCTGCCTGCCTGTGTGTA TGGA C
CTGCCGTAGCCAGTGGGTACGACTTTGAAAGGGAGGGATACTCTCTAGTCGGAATAGACC
CTTTCA GA CTGCTTCAAAA CA GCCAA GTGTA CA GCCTAA TCA GA CCAAA TGA GAA TCCA GC
A CA CAA GA GTCAA CTGGT GTGGA TGGCA TGCCA TTCTGCCGCA TTTGAA GA TCTAA GA GTA
TTAA GCTTCA TCAAA GGGA CGAA GGTGCTCCCAA GA GGGAA GCTTTCCA CTA GA GGAGTT
CAAA TTGCTTCCAA TGAAAA TA TGGA GA CTA TGGAA TCAA GTA CA CTTGAA CTGA GAA GCA
GGTACTGGGCCATAAGGACCAGAAGTGGAGGAAACACCAATCAACAGAGGGCATCTGCG
GGCCAAA TCA GCA TA CAA CCTA CGTTCTCA GTA CA GA GAAA TCTCCCTTTTGA CA GAA CAA
CCA TTA TGGCA GCA TTCAA TGGGAA TA CAGAGGGGA GAA CA TCTGA CA TGA GGA CCGAAA
TCA TAA GGA TGA T GGAAA GTGCAA GA CCA GAA GA TGTGTCTTTCCA GGGGCGGGGA GTCT
TCGA GCTCTCGGA CGAAAA GGCA GCGA GCCCGA TCGTGCCTTCCTTTGA CA TGA GTAA TG
AAGGA TCTTA TTTCTTCGGA GA CAA TGCA GA GGAGTA CGA CAA 7TAAGCCCCTCTCCCTCC
CCCCCCCCTAACGTTACTGGCCGAAGCCGCTTGGAATAAGGCCGGTGTGTGTTTGTC
TATATGTGATTTTCCACCATATTGCCGTCTTTTGGCAATGTGAGGGCCCGGAAACCT
GGCCCTGTCTTCTTGACGAGCATTCCTAGGGGTCTTTCCCCTCTCGCCAAAGGAATG
CAAGGTCTGTTGAATGTCGTGAAGGAAGCAGTTCCTCTGGAAGCTTCTTGAAGACAA
ACAACGTCTGTAGCGACCCTTTGCAGGCAGCGGAACCCCCCACCTGGCGACAGGTG
CCTCTGCGGCCAAAAGCCACGTGTATAAGATACACCTGCAAAGGCGGCACAACCCC
AGTGCCACGTTGTGAGTTGGATAGTTGTGGAAAGAGTCAAATGGCTCTCCTCAAGCG
TAGTCAACAAGGGGCTGAAGGATGCCCAGAAGGTACCCCATTGTATGGGAATCTGA
TCTGGGGCCTCGGTGCACATGCTTTACATGTGTTTAGTCGAGGTTAAAAAAGCTCTA
GGCCCCCCGAACCACGGGGACGTGGTTTTCCTTTGAAAAACACGATGATAAGCTTGC
CACAAATGGAACACGACCTTGAGAGGGGCCCACCGGGCCCGCGACGGCCCCCTCGAG
GACCCCCCCTCTCCTCTTCCCTAGGCCTTGCTCTCCTTCTCCTCCTCTTGGCGCTACTG
TTTTGGCTGTACATCGTTATGAGTGACTGGACTGGAGGAGCCCTCCTTGTCCTCTATT
CCTTTGCTCTCATGCTTATAATTATAATTTTGATCATCTTTATCTTCAGAAGAGACCTTC
TCTGTCCACTTGGAGCCCTTTGTATACTCCTACTGATGATCACCCTCCTGCTCATCGCT
CTCTGGAATTTGCACGGACAGGCATTGTTCCTTGGAATTGTGCTGTTCATCTTCGGGT
GCTTACTTGTCTTAGGTATCTGGATCTACTTATTGGAGATGCTCTGGCGACTTGGTGCC ACCATCTGGCAGCTTTTGGCCTTCTTCCTAGCCTTCTTCCTAGACCTCATCCTGCTCAT
TATTGCTCTCTATCTACAACAAAACTGGTGGACTCTATTGGTTGATCTCCTTTGGCTCC
TCCTGTTTCTGGCGATTTTAATCTGGATGTATTACCATGGACAACGAATGTCTGCGGG
AGACCCCCGCGTGGGATCCGGGTCCCTGGACTCCTTCATGTTCTCCATACCCTTGGT
CGCGCTTAACGTGGGAGTGAGGCGCCGCCTATCGCTGTTCTTGAACCCTCGGACGCC
CGTGGCGGCCGACTGGACCTTGCTGGCGGAGGAGATGGGCTTCGAGTACTTGGAGA
TCCGAGAGCTGGAAACGCGCCCTGACCCCACTCGCAGTTTGTTGGATGCCTGGCAGG
GGCGCTCTGGCGCGTCTGTCGGCAGGCTGCTAGAGCTGCTGGCCTTGTTAGACCGTG
AGGATATACTGAAGGAGCTGAAGTCGCGCATCGAGGAGGACTGCCAGAAATACTTA
GGTAAGCAGCAGAACCAGGAGTCCGAGAAGCCTTTACAGGTGGCCAGAGTGGAAA
GCAGTGTCCCACAAACAAAGGAACTGGGAGGCATCACCACCCTTGATGACCCCCTA
GGACAAACGCCGGAACTTTTCGATGCCTTTATCTGCTACTGCCCCAACGATATCGAG
TTTGTGCAGGAGATGATCCGGCAACTAGAACAGACAGACTATCGGCTTAAGTTGTGT
GTGTCCGACCGTGACGTCCTGCCGGGCACCTGTGTCTGGTCCATTGCCAGCGAGCTA
ATTGAGAAAAGGTGTCGCCGCATGGTGGTGGTTGTTTCTGACGATTATCTACAGAGC
AAGGAATGTGACTTCCAGACCAAGTTTGCACTCAGCCTGTCTCCAGGTGTCCAACAG
AAGCGACTGATTCCTATTAAATACAAGGCGATGAAGAAGGACTTTCCCAGTATCCTG
CGGTTCATCACTATATGCGACTATACCAACCCTTGCACCAAGTCCTGGTTCTGGACC
CGCCTTGCCAAGGCTTTGTCCCTGGTCGAGTATATCAAAAAGGTGGTCAAGAAAC
CAAAGGATAATGAGATCTTACCCCCTGCGGCTCGACGGCAAGATCCCCAGGAG
ATGGAAGATTATCCCGGTCATAACACCGCTGCTCCAGTGCAGGAGACGCTGCA
CGGGTGTCAGCCTGTCACACAGGAGGATGGTAAAGAGAGTCGCATCTCAGTGC
AGGAGCGGCAGGTGACAGACAGCATAGCCTTGAGGCCCCTGGTCTGA (SEQ ID
NO: 13)
SEQ ID NO: 14 is Nuclear Protein (NP).
Protein:
MAS QGTKRS YEQMETDGERQN ATEIRAS VGKMIGGIGRF YIQMCTELKLS D YEGRLIQN S LTIERM VLS AFDERRNKYLEEHPS AGKDPKKTGGPIYRRVNGKWMRELILYDKEEIRRI WRQ ANNGDD ATAGLTHMMr>VHS NLND AT YQRTR ALVRTGMDPRMCS LMQGS TLPRR SGAAGAAVKGVGTMVMELVRMIKRGINDRNFWRGENGRKTRIAYERMCNILKGKFQT A AQKAMMDQ VRES RNPGN AEFEDLTFLARS ALILRGS V AHKS CLP AC V YGP A V AS G YD FEREG YS LVGIDPFRLLQNS Q V YS LIRPNENP AHKS QLVWM ACHS A AFEDLR VLS FIKGT KVLPRGKLS TRG VQIAS NENMETMES S TLELRS R YWAIRTRS GGNTNQQRAS AGQIS IQP TFS VQRNLPFDRTTIM A AFNGNTEGRTS DMRTEIIRMMES ARPED VS FQGRG VFELS DEK A AS PIVPS FDMS NEGS YFFGDN AEE YDN (SEQ ID NO: 14)
SEQ ID NO: 15 is Epstein Barr Virus latent membrane protein 1 (without the cytoplasmic domain) fused to the murine protein MyD88 fused to the cytoplasmic domain of murine CD40.
Protein:
MEHDLERGPPGPRRPPRGPPLS S S LGLALLLLLLALLFWLYIVMS D WTGG ALLVLYS FAL MLIIIILIIFIFRRDLLCPLGALCILLLMITLLLIALWNLHGQALFLGIVLFIFGCLLVLGIWIY LLEMLWRLGATIWQLLAFFLAFFLDLILLIIALYLQQNWWTLLVDLLWLLLFLAILIWMY YHGQRMS AGDPRVGS GS LDS FMFS IPLV ALN VG VRRRLS LFLNPRTP V A AD WTLLAEE MGFEYLEIRELETRPDPTRS LLD A WQGRS GAS VGRLLELLALLDREDILKELKS RIEEDC QKYLGKQQNQESEKPLQVARVESSVPQTKELGGITTLDDPLGQTPELFDAFICYCPNDIE FVQEMIRQLEQTD YRLKLC VS DRD VLPGTC VWS IAS ELIEKRCRRM V V V VS DD YLQS KE CDFQTKFALSLSPGVQQKRLIPIKYKAMKKDFPSILRFITICDYTNPCTKSWFWTRLAKA LSLVEYIKKVVKKPKDNEILPPAARRQDPQEMEDYPGHNTAAPVQETLHGCQPVTQED GKES RIS VQERQ VTDS IALRPLV (SEQ ID NO: 15)
SEQ ID NO: 16 is Epstein Barr Virus latent membrane protein 1 (without the cytoplasmic domain) fused to murine MyD88 and the cytoplasmic domain of murine CD40 as a single 3- protein chimera.
LMPl-muMvD88-muCD40
Italicized/bolded sequence: LMP1 sequence (Transmembrane Domain)
Non-italicized/bolded sequence: Murine MyD88 sequence
Underlined sequence: Linker
Bolded sequence: Murine CD40 sequence (cytoplasmic domain)
ATGGAACACGACCTTGAGAGGGGCCCACCGGGCCCGCGACGGCCCCCTCGAGGACCC
CCCCTCTCCTCTTCCCTAGGCCTTGCTCTCCTTCTCCTCCTCTTGGCGCTACTGTTTTG
GCTGTACATCGTTATGAGTGACTGGACTGGAGGAGCCCTCCTTGTCCTCTATTCCTTT
GCTCTCATGCTTATAATTATAATTTTGATCATCTTTATCTTCAGAAGAGACCTTCTCTGT
CCACTTGGAGCCCTTTGTATACTCCTACTGATGATCACCCTCCTGCTCATCGCTCTCTG
GAATTTGCACGGACAGGCATTGTTCCTTGGAATTGTGCTGTTCATCTTCGGGTGCTTA
CTTGTCTTAGGTATCTGGATCTACTTATTGGAGATGCTCTGGCGACTTGGTGCCACCA
TCTGGCAGCTTTTGGCCTTCTTCCTAGCCTTCTTCCTAGACCTCATCCTGCTCATTATT
GCTCTCTATCTACAACAAAACTGGTGGACTCTATTGGTTGATCTCCTTTGGCTCCTCCT
GTTTC TGGCGA TTTTAA TC TGGA TGTA TTACCA TGGA CAA CGA ATGTCTGCGGG AG AC
CCCCGCGTGGGATCCGGGTCCCTGGACTCCTTCATGTTCTCCATACCCTTGGTCGCGC
TTAACGTGGGAGTGAGGCGCCGCCTATCGCTGTTCTTGAACCCTCGGACGCCCGTGG
CGGCCGACTGGACCTTGCTGGCGGAGGAGATGGGCTTCGAGTACTTGGAGATCCGA
GAGCTGGAAACGCGCCCTGACCCCACTCGCAGTTTGTTGGATGCCTGGCAGGGGCG
CTCTGGCGCGTCTGTCGGCAGGCTGCTAGAGCTGCTGGCCTTGTTAGACCGTGAGGA
TATACTGAAGGAGCTGAAGTCGCGCATCGAGGAGGACTGCCAGAAATACTTAGGTA
AGCAGCAGAACCAGGAGTCCGAGAAGCCTTTACAGGTGGCCAGAGTGGAAAGCAGT
GTCCCACAAACAAAGGAACTGGGAGGCATCACCACCCTTGATGACCCCCTAGGACA
AACGCCGGAACTTTTCGATGCCTTTATCTGCTACTGCCCCAACGATATCGAGTTTGTG
CAGGAGATGATCCGGCAACTAGAACAGACAGACTATCGGCTTAAGTTGTGTGTGTC
CGACCGTGACGTCCTGCCGGGCACCTGTGTCTGGTCCATTGCCAGCGAGCTAATTGA
GAAAAGGTGTCGCCGCATGGTGGTGGTTGTTTCTGACGATTATCTACAGAGCAAGGA
ATGTGACTTCCAGACCAAGTTTGCACTCAGCCTGTCTCCAGGTGTCCAACAGAAGCG
ACTGATTCCTATTAAATACAAGGCGATGAAGAAGGACTTTCCCAGTATCCTGCGGTT CATCACTATATGCGACTATACCAACCCTTGCACCAAGTCCTGGTTCTGGACCCGCCT
TGCCAAGGCTTTGTCCCTGGTCGAGTATATCAAAAAGGTGGTCAAGAAACCAAAG
GATAATGAGATCTTACCCCCTGCGGCTCGACGGCAAGATCCCCAGGAGATGGA
AGATTATCCCGGTCATAACACCGCTGCTCCAGTGCAGGAGACGCTGCACGGGT
GTCAGCCTGTCACACAGGAGGATGGTAAAGAGAGTCGCATCTCAGTGCAGGAG
CGGCAGGTGACAGACAGCATAGCCTTGAGGCCCCTGGTCTGA (SEQ ID NO: 16)
SEQ ID NO: 17 is Epstein Barr Virus latent membrane protein 1 (without the cytoplasmic domain) fused to the murine protein MyD88 fused to the cytoplasmic domain of murine CD40.
Protein:
MEHDLERGPPGPRRPPRGPPLS S S LGLALLLLLLALLFWLYIVMS D WTGG ALLVLYS FAL MLIIIILIIFIFRRDLLCPLGALCILLLMITLLLIALWNLHGQALFLGIVLFIFGCLLVLGIWIY LLEMLWRLGATIWQLLAFFLAFFLDLILLIIALYLQQNWWTLLVDLLWLLLFLAILIWMY YHGQRMS AGDPRVGS GS LDS FMFS IPLV ALN VG VRRRLS LFLNPRTP V A AD WTLLAEE MGFEYLEIRELETRPDPTRS LLD A WQGRS GAS VGRLLELLALLDREDILKELKS RIEEDC QKYLGKQQNQESEKPLQVARVESSVPQTKELGGITTLDDPLGQTPELFDAFICYCPNDIE FVQEMIRQLEQTD YRLKLC VS DRD VLPGTC VWS IAS ELIEKRCRRM V V V VS DD YLQS KE CDFQTKFALSLSPGVQQKRLIPIKYKAMKKDFPSILRFITICDYTNPCTKSWFWTRLAKA LSLVEYIKKVVKKPKDNEILPPAARRQDPQEMEDYPGHNTAAPVQETLHGCQPVTQED GKES RIS VQERQ VTDS IALRPLV (SEQ ID NO: 17)
SEQ ID NO: 18 is Epstein Barr Virus latent membrane protein 1 (without the cytoplasmic domain) fused to human IPS-1, the cytoplasmic domain of human TLR7, and the cytoplasmic domain (CD) of Epstein Barr Virus LMP1 as a single chimera.
LMP 1 -huIPS - 1 -huTLR7-LMP 1 (CD)
Italicized/bolded sequence: LMP1 sequence (Transmembrane Domain)
Non-italicized/bolded sequence: Human IPS-1 sequence
Underlined sequence: Human TLR7 sequence (cytoplasmic domain)
Bolded sequence: LMP1 sequence (cytoplasmic domain)
ATGGAACACGACCTTGAGAGGGGCCCACCGGGCCCGCGACGGCCCCCTCGAGGACCC
CCCCTCTCCTCTTCCCTAGGCCTTGCTCTCCTTCTCCTCCTCTTGGCGCTACTGTTTTG
GCTGTACATCGTTATGAGTGACTGGACTGGAGGAGCCCTCCTTGTCCTCTATTCCTTT
GCTCTCATGCTTATAATTATAATTTTGATCATCTTTATCTTCAGAAGAGACCTTCTCTGT
CCACTTGGAGCCCTTTGTATACTCCTACTGATGATCACCCTCCTGCTCATCGCTCTCTG
GAATTTGCACGGACAGGCATTGTTCCTTGGAATTGTGCTGTTCATCTTCGGGTGCTTA
CTTGTCTTAGGTATCTGGATCTACTTATTGGAGATGCTCTGGCGACTTGGTGCCACCA
TCTGGCAGCTTTTGGCCTTCTTCCTAGCCTTCTTCCTAGACCTCATCCTGCTCATTATT
GCTCTCTATCTACAACAAAACTGGTGGACTCTATTGGTTGATCTCCTTTGGCTCCTCCT
GTTTC TGGCGA TTTTAA TC TGGA TGTA TTACCA TGGA CAA CGA ATGCCGTTTGCTG A A
GACAAGACCTATAAGTATATCTGCCGCAATTTCAGCAATTTTTGCAATGTGGATGTT GTAGAGATTCTGCCTTACCTGCCCTGCCTCACAGCAAGAGACCAGGATCGACTGCGG
GCCACCTGCACACTCTCAGGGAACCGGGACACCCTCTGGCATCTCTTCAATACCCTT
CAGCGGCGGCCCGGCTGGGTGGAGTACTTCATTGCGGCACTGAGGGGCTGTGAGCT
AGTTGATCTCGCGGACGAAGTGGCCTCTGTCTACCAGAGCTACCAGCCTCGGACCTC
GGACCGTCCCCCAGACCCACTGGAGCCACCGTCACTTCCTGCTGAGAGGCCAGGGC
CCCCCACACCTGCTGCGGCCCACAGCATCCCCTACAACAGCTGCAGAGAGAAGGAG
CCAAGTTACCCCATGCCTGTCCAGGAGACCCAGGCGCCAGAGTCCCCAGGAGAGAA
TTCAGAGCAAGCCCTGCAGACGCTCAGCCCCAGAGCCATCCCAAGGAATCCAGATG
GTGGCCCCCTGGAGTCCTCCTCTGATCTGGCAGCCCTCAGCCCTCTGACCTCCAGCG
GGCATCAGGAGCAGGACACAGAACTGGGCAGTACCCACACAGCAGGTGCGACCTCC
AGCCTCACACCATCCCGTGGGCCTGTGTCTCCATCTGTCTCCTTCCAGCCCCTGGCCC
GTTCCACCCCCAGGGCAAGCCGCTTGCCTGGACCCACAGGGTCAGTTGTATCTACTG
GCACCTCCTTCTCCTCCTCATCCCCTGGCTTGGCCTCTGCAGGGGCTGCAGAGGGTA
AACAGGGTGCAGAGAGTGACCAGGCCGAGCCTATCATCTGCTCCAGTGGGGCAGAG
GCACCTGCCAACTCTCTGCCCTCCAAAGTGCCTACCACCTTGATGCCTGTGAACACA
GTGGCCCTGAAAGTGCCTGCCAACCCAGCATCTGTCAGCACAGTGCCCTCCAAGTTG
CCAACTAGCTCAAAGCCCCCTGGTGCAGTGCCTTCTAATGCGCTCACCAATCCAGCA
CCATCCAAATTGCCCATCAACTCAACCCGTGCTGGCATGGTGCCATCCAAAGTGCCT
ACTAGCATGGTGCTCACCAAGGTGTCTGCCAGCACAGTCCCCACTGACGGGAGCAG
CAGAAATGAGGAGACCCCAGCAGCTCCAACACCCGCCGGCGCCACTGGAGGCAGCT
CAGCCTGGCTAGACAGCAGCTCTGAGAATAGGGGCCTTGGGTCGGAGCTGAGTAAG
CCTGGCGTGCTGGCATCCCAGGTAGACAGCCCGTTCTCGGGCTGCTTCGAGGATCTT
GCCATCAGTGCCAGCACCTCCTTGGGCATGGGGCCCTGCCATGGCCCAGAGGAGAA
TGAGTATAAGTCCGAGGGCACCTTTGGGATCCACGTGGCTGAGAACCCCAGCATCC
AGCTCCTGGAGGGCAACCCTGGGCCACCTGCGGACCCGGATGGCGGCCCCAGGCCA
CAAGCCGACCGGAAGTTCCAGGAGAGGGAGGTGCCATGCCACAGGCCCTCACCTGG
GGCTCTGTGGCTCCAGGTGGCTGTGACAGGGGTGCTGGTAGTCACACTCCTGGTGGT
GCTGTACCGGCGGCGTCTGCACCACCTCTATTTCTGGGATGTGTGGTATATTTACCAT
TTCTGTAAGGCCAAGATAAAGGGGTATCAGCGTCTAATATCACCAGACTGTTGCTAT
GATGCTTTTATTGTGTATGACACTAAAGACCCAGCTGTGACCGAGTGGGTTTTGGCT
GAGCTGGTGGCCAAACTGGAAGACCCAAGAGAGAAACATTTTAATTTATGTCTCGA
GGAAAGGGACTGGTTACCAGGGCAGCCAGTTCTGGAAAACCTTTCCCAGAGCATAC
AGCTTAGCAAAAAGACAGTGTTTGTGATGACAGACAAGTATGCAAAGACTGAAAAT
TTTAAGATAGCATTTTACTTGTCCCATCAGAGGCTCATGGATGAAAAAGTTGATGTG
ATTATCTTGATATTTCTTGAGAAGCCCTTTCAGAAGTCCAAGTTCCTCCAGCTCCGGA
AAAGGCTCTGTGGGAGTTCTGTCCTTGAGTGGCCAACAAACCCGCAAGCTCACCCAT
ACTTCTGGCAGTGTCTAAAGAACGCCCTGGCCACAGACAATCATGTGGCCTATAGTC
AGGTGTTCAAGGAAACGGTCCACAGTGATGAACACCACCACGATGACTCCCTCC
CGCACCCTCAACAAGCTACCGATGATTCTGGCCATGAATCTGACTCTAACTCCA
ACGAGGGCAGACACCACCTGCTCGTGAGTGGAGCCGGCGACGGACCCCCACTC
TGCTCTCAAAACCTAGGCGCACCTGGAGGTGGTCCTGACAATGGCCCACAGGA
CCCTGACAACACTGATGACAATGGCCCACAGGACCCTGACAACACTGATGACA
ATGGCCCACATGACCCGCTGCCTCAGGACCCTGACAACACTGATGACAATGGC
CCACAGGACCCTGACAACACTGATGACAATGGCCCACATGACCCGCTGCCTCA
TAGCCCTAGCGACTCTGCTGGAAATGATGGAGGCCCTCCACAATTGACGGAAG
AGGTTGAAAACAAAGGAGGTGACCAGGGCCCGCCTTTGATGACAGACGGAGGC GGCGGTCATAGTCATGATTCCGGCCATGGCGGCGGTGATCCACACCTTCCTAC GCTGCTTTTGGGTTCTTCTGGTTCCGGTGGAGATGATGACGACCCCCACGGCC CAGTTCAGCTAAGCTACTATGACTAA (SEQ ID NO: 18)
SEQ ID NO: 19 is Epstein Barr Virus latent membrane protein 1 (without the cytoplasmic domain) fused to the human IPS-1 fused to the cytoplasmic domain of human TLR7 fused to the cytoplasmic domain of Epstein Barr Virus latent membrane protein 1.
Protein:
MEHDLERGPPGPRRPPRGPPLS S S LGLALLLLLLALLFWLYIVMS D WTGG ALLVLYS FAL MLIIIILIIFIFRRDLLCPLGALCILLLMITLLLIALWNLHGQALFLGIVLFIFGCLLVLGIWIY LLEMLWRLGATIWQLLAFFLAFFLDLILLIIALYLQQNWWTLLVDLLWLLLFLAILIWMY YHGQRMPFAEDKTYKYICRNFSNFCNVDVVEILPYLPCLTARDQDRLRATCTLSGNRDT LWHLFNTLQRRPGWVE YFIA ALRGCELVDLADEV AS V YQS YQPRTS DRPPDPLEPPS LP AERPGPPTP A A AHS IP YNS CREKEPS YPMP VQETQ APES PGENS EQ ALQTLS PRAIPRNPD GGPLES S S DLA ALS PLTS S GHQEQDTELGS THT AG ATS S LTPS RGP VS PS VS FQPLARS TP RAS RLPGPTGS V VS TGTS FS S S S PGLAS AG A AEGKQG AES DQ AEPIICS S G AEAP ANS LPS KVPTTLMP VNT V ALKVP ANP AS VS T VPS KLPTS S KPPG A VPS N ALTNP APS KLPINS TRA GM VPS KVPTS M VLTKVS AS T VPTDGS S RNEETP A APTP AG ATGGS S A WLDS S S ENRGLG S ELS KPG VLAS Q VDS PFS GCFEDLAIS AS TS LGMGPCHGPEENEYKS EGTFGIH V AENPS I QLLEGNPGPPADPDGGPRPQADRKFQEREVPCHRPSPGALWLQVAVTGVLVVTLLVVL YRRRLHHLYFWDVWYIYHFCKAKIKGYQRLISPDCCYDAFIVYDTKDPAVTEWVLAEL VAKLEDPREKHFNLCLEERDWLPGQPVLENLS QS IQLS KKTVFVMTDKYAKTENFKIAF YLS HQRLMDEKVD VIILIFLEKPFQKS KFLQLRKRLCGS S VLEWPTNPQ AHP YFWQCLK NALATDNHVAYSQVFKETVHSDEHHHDDSLPHPQQATDDSGHESDSNSNEGRHHLLVS GAGDGPPLCSQNLGAPGGGPDNGPQDPDNTDDNGPQDPDNTDDNGPHDPLPQDPDNT DDNGPQDPDNTDDNGPHDPLPHSPSDSAGNDGGPPQLTEEVENKGGDQGPPLMTDGGG GHS HDS GHGGGDPHLPTLLLGS S GS GGDDDDPHGP VQLS Y YD (SEQ ID NO: 19)
Other Embodiments
[000103] Any improvement may be made in part or all of the compositions, cells, kits, and method steps. All references, including publications, patent applications, and patents, cited herein are hereby incorporated by reference. The use of any and all examples, or exemplary language (e.g., "such as") provided herein, is intended to illuminate the invention and does not pose a limitation on the scope of the invention unless otherwise claimed. Any statement herein as to the nature or benefits of the invention or of the preferred embodiments is not intended to be limiting, and the appended claims should not be deemed to be limited by such statements. More generally, no language in the specification should be construed as indicating any non-claimed element as being essential to the practice of the invention. This invention includes all modifications and equivalents of the subject matter recited in the claims appended hereto as permitted by applicable law. Moreover, any combination of the above-described elements in all possible variations thereof is encompassed by the invention unless otherwise indicated herein or otherwise clearly contraindicated by context.

Claims

What is claimed is:
1. A composition for inducing an immune response in a subject comprising at least a first nucleic acid encoding at least a first fusion protein, the at least first fusion protein comprising a transmembrane domain of LMPl to provide for clustering of the at least first fusion protein in cells without the need for any exogenous molecule and at least one signaling domain from at least one of: an immune activating receptor and/or an adaptor protein, the at least first nucleic acid in an amount sufficient to induce an immune response in a subject.
2. The composition of claim 1, wherein the immune activating receptor is a Toll-like receptor (TLR) or a TNF superfamily receptor (TNFSFR).
3. The composition of claim 2, wherein the at least first fusion protein comprises a signaling domain from a TLR and a signaling domain from a TNFSFR.
4. The composition of claim 1, wherein the at least first fusion protein comprises a signaling domain from an immune activating receptor and/or a signaling domain from an adaptor protein.
5. The composition of claim 1, wherein the immune activating receptor is selected from the group consisting of: TNF Receptor SuperFamily (TNFRSF) proteins with preference for CD40, 4- IBB, RANK, TACI, OX40, CD27, GITR, LT R, and BAFFR; Toll-Like Receptors (TLR) TLR1 to TLR13 inclusive with preference for TLR7; integrins; FcyRIII; Dectinl; Dectin2;
NODI; NOD2; CD16; IL-2R; Type I II interferon receptor; chemokine receptors such as CCR5 and CCR7; G-protein coupled receptors (GPCRs); TREM1; and the B cell receptor (BCR) complex that includes CD79A, CD79B, and Ig-alpha.
6. The composition of claim 1, wherein the adaptor protein is selected from the group consisting of: IPS-1; MyD88; RIG-1; MDA5; CD3 zeta chain; MyD88ATIR; TRIF; TRAM; TIRAP; MAL; BTK; RTK; RAC1; SYK; NALP3 (NLRP3); NALP3ALRR; NALP1; CARD9; DAI; IPAG; STING; Zap70; and LAT.
7. The composition of claim 1, wherein the at least first nucleic acid further encodes an antigen.
8. The composition of claim 1, wherein the at least first nucleic acid is comprised within a viral vector.
9. The composition of claim 8, wherein the nucleic acid sequence is comprised within a recombinant virion.
10. The composition of claim 1, further comprising a second nucleic acid encoding a second fusion protein, the second fusion protein comprising a transmembrane domain of LMPl and at least one signaling domain from at least one of: an immune activating receptor and an adaptor protein.
11. The composition of claim 10, wherein the at least first and the second nucleic acid are comprised within a viral vector.
12. The composition of claim 1 or claim 10, wherein the at least first nucleic acid is an amount effective for activating and maturing immune cells when administered to a subject.
13. The composition of claim 12, wherein the immune cells are dendritic cells and the subject is a human.
14. The composition of claim 1 or claim 10, wherein the at least first nucleic acid is an amount effective for inducing expression of cytokines and protecting primary CD4+ T cells from infection by a virus when administered to a subject.
15. The composition of claim 14, wherein the virus is human immunodeficiency virus (HIV), and the subject is a human.
16. The composition of claim 14, wherein the at least first fusion protein comprises a transmembrane domain of LMP1 and a signaling domain from IPS-1.
17. A vaccine formulation for preventing or treating a disease or condition in a subject comprising the composition of claim 1 or claim 10 and a pharmaceutically acceptable excipient.
18. The vaccine formulation of claim 17, wherein the disease or condition is cancer or infection.
19. A vaccine adjuvant for enhancing an immune response to a vaccine, the vaccine adjuvant comprising the composition of claim 1 or claim 10 and an antigen or a nucleic acid encoding an antigen in an amount effective for enhancing an immune response to a vaccine and a
pharmaceutically acceptable excipient.
20. The vaccine adjuvant of claim 19, wherein the at least first nucleic acid further encodes the antigen.
21. An immune cell transduced with the composition of claim 1 or claim 7.
22. The immune cell of claim 21, wherein the immune cell is a dendritic cell.
23. The immune cell claim 21, wherein the immune cell is comprised within a vaccine formulation for preventing or treating a disease or condition in a subject.
24. The immune cell of claim 23, wherein the disease or condition is cancer or infection.
25. A method of inducing an immune response against cancer or infection in a subject, the method comprising administering the composition of claim 1 or claim 10 to the subject in a therapeutically effective amount for inducing an immune response against the cancer or infection in the subject.
26. The method of claim 25, wherein administering the composition to the subject vaccinates the subject against cancer or infection.
27. The method of claim 25, wherein inducing an immune response against the cancer or the infection in the subject comprises at least one of: activating and maturing dendritic cells, and inducing expression of at least one cytokine in the subject.
28. The method of claim 25, wherein the cancer is selected from the group consisting of: melanoma, glioma, prostate cancer, breast cancer, and the infection is selected from the group consisting of: HIV infection, hepatitis C infection and human papilloma virus infection.
29. The method of claim 28, wherein the subject has an HIV infection, and inducing an immune response against the infection comprises induction of type I interferon and protection of primary CD4+ T cells in the subject.
30. The method of claim 28, wherein the subject has a cancerous tumor, and the at least first fusion protein comprises a transmembrane domain of LMPl and a signaling domain from IPS-1 or a signaling domain from MyD88.
31. The method of claim 30, wherein inducing an immune response results in a decrease in growth of the cancerous tumor.
32. A kit for preventing or treating a disease or condition in a subject comprising the composition of claim 1 or claim 10, the vaccine formulation of claim 17, or the vaccine adjuvant of claim 19; instructions for use; and packaging.
33. A composition for inducing an immune response in a subject comprising a nucleic acid encoding a fusion protein, the fusion protein comprising a transmembrane domain of LMPl and an amino acid sequence comprising two or more signaling motifs from immune activating receptors and/or adaptor proteins, the nucleic acid in an amount sufficient to induce an immune response in a subject.
PCT/US2013/058748 2012-09-07 2013-09-09 Fusion proteins for promoting an immune response, nucleic acids encoding same, and methods of making and use thereof WO2014039961A1 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
EP13835331.3A EP2892930B1 (en) 2012-09-07 2013-09-09 Fusion proteins for promoting an immune response, nucleic acids encoding same, and methods of making and use thereof
US14/424,562 US10093701B2 (en) 2012-09-07 2013-09-09 Fusion proteins for promoting an immune response, nucleic acids encoding same, and methods of making and use thereof
US16/115,171 US10329329B2 (en) 2012-09-07 2018-08-28 Fusion proteins for promoting an immune response, nucleic acids encoding same, and methods of making and use thereof

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201261698109P 2012-09-07 2012-09-07
US61/698,109 2012-09-07

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US14/424,562 A-371-Of-International US10093701B2 (en) 2012-09-07 2013-09-09 Fusion proteins for promoting an immune response, nucleic acids encoding same, and methods of making and use thereof
US16/115,171 Division US10329329B2 (en) 2012-09-07 2018-08-28 Fusion proteins for promoting an immune response, nucleic acids encoding same, and methods of making and use thereof

Publications (1)

Publication Number Publication Date
WO2014039961A1 true WO2014039961A1 (en) 2014-03-13

Family

ID=50237678

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2013/058748 WO2014039961A1 (en) 2012-09-07 2013-09-09 Fusion proteins for promoting an immune response, nucleic acids encoding same, and methods of making and use thereof

Country Status (3)

Country Link
US (2) US10093701B2 (en)
EP (1) EP2892930B1 (en)
WO (1) WO2014039961A1 (en)

Cited By (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2016073875A1 (en) * 2014-11-06 2016-05-12 University Of Maryland, Baltimore CD8α AND T CELL RECEPTOR VARIANTS AND METHODS OF USING SAME IN MODULATING IMMUNE CELL RESPONSES
WO2018081459A1 (en) * 2016-10-26 2018-05-03 Modernatx, Inc. Messenger ribonucleic acids for enhancing immune responses and methods of use thereof
WO2018096395A1 (en) 2016-11-28 2018-05-31 Aratinga.Bio Aio Viral vector constructs for expression of genetic adjuvants activating the sting pathway
WO2018096399A1 (en) 2016-11-28 2018-05-31 Arating.Bio Aio Viral vector constructs for expression of genetic adjuvants activating the cd40 and sting pathways
WO2019040135A1 (en) * 2017-05-12 2019-02-28 Children's Medical Center Corporation Nucleic acids that manipulate immune pathways
WO2019106432A3 (en) * 2017-11-29 2019-07-18 Aratinga.Bio Aio Viral vector constructs for expression of genetic adjuvants mimicking cd40 signaling
CN110694069A (en) * 2019-11-08 2020-01-17 重庆医科大学附属第二医院 Medicine for preventing and treating fulminant hepatitis
WO2020171680A1 (en) 2019-02-22 2020-08-27 주식회사 레모넥스 Pharmaceutical composition for immune activity or for preventing or treating cancer
KR20200103568A (en) 2019-02-22 2020-09-02 주식회사 레모넥스 Composition for immune enhancing or preventing or treating of cancer
US10793875B2 (en) 2015-12-07 2020-10-06 Nant Holdings Ip, Llc Compositions and methods for viral delivery of neoepitopes and uses thereof
US10881730B2 (en) 2017-02-01 2021-01-05 Modernatx, Inc. Immunomodulatory therapeutic MRNA compositions encoding activating oncogene mutation peptides
KR20220004588A (en) 2020-07-03 2022-01-11 주식회사 레모넥스 Rna and nucleic acid delivery systems comprising the same
US11701378B2 (en) 2020-07-03 2023-07-18 Lemonex Inc. RNA and nucleic acid carrier including the same

Families Citing this family (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP6534146B2 (en) * 2015-06-10 2019-06-26 国立大学法人 東京大学 Adjuvant for vaccine, vaccine, and method for inducing immunity
EP3359675B1 (en) 2015-10-07 2024-01-03 Nant Holdings IP, LLC Activation of immune-related signalling pathways in cells via optofection
US20180110853A1 (en) * 2016-10-21 2018-04-26 Wisconsin Alumni Research Foundation Vaccine Adjuvant
EP3694988A4 (en) 2017-10-10 2021-07-14 NantBio, Inc. Modified ec7 cells having low toxicity to viral production payloads
KR20230153529A (en) * 2021-02-19 2023-11-06 프리트 엠. 쇼드하리 Single-chain and multi-chain synthetic antigen receptors for various immune cells
JP2024512004A (en) * 2021-03-17 2024-03-18 マイエロイド・セラピューティクス,インコーポレーテッド Engineered chimeric fusion protein compositions and methods of use thereof
CA3228856A1 (en) * 2021-08-16 2023-02-23 Peter C. Demuth Compositions containing polynucleotide amphiphiles and methods of use thereof
CN114028552A (en) * 2021-10-27 2022-02-11 保定诺未科技有限公司 Monocyte-loaded LMP1 protein vaccine composition and preparation method and application thereof

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7404950B2 (en) 2003-02-18 2008-07-29 Baylor College Of Medicine Induced activation in dendritic cell
US20100203067A1 (en) 2008-09-22 2010-08-12 Baylor College Of Medicine Methods and compositions for generating an immune response by inducing cd40 and pattern recognition receptor adapters
US20110033383A1 (en) 2006-10-19 2011-02-10 David Spencer Methods and compositions for generating an immune response by inducing cd40 and pattern recognition receptors and adaptors thereof
WO2011119628A2 (en) 2010-03-23 2011-09-29 The Regents Of The University Of California Compositions and methods for self-adjuvanting vaccines against microbes and tumors
WO2012040101A1 (en) * 2010-09-21 2012-03-29 University Of Miami Compositions and methods for inducing migration by dendritic cells and an immune response

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2324044A4 (en) * 2008-08-04 2012-04-25 Univ Miami Sting (stimulator of interferon genes), a regulator of innate immune responses
CN103145849B (en) * 2013-02-18 2014-06-11 冯振卿 Chimeric antigen receptor and use thereof

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7404950B2 (en) 2003-02-18 2008-07-29 Baylor College Of Medicine Induced activation in dendritic cell
US20110033383A1 (en) 2006-10-19 2011-02-10 David Spencer Methods and compositions for generating an immune response by inducing cd40 and pattern recognition receptors and adaptors thereof
US20100203067A1 (en) 2008-09-22 2010-08-12 Baylor College Of Medicine Methods and compositions for generating an immune response by inducing cd40 and pattern recognition receptor adapters
WO2011119628A2 (en) 2010-03-23 2011-09-29 The Regents Of The University Of California Compositions and methods for self-adjuvanting vaccines against microbes and tumors
US20130039942A1 (en) 2010-03-23 2013-02-14 Richard Syd Kornbluth Compositions and Methods for Self-Adjuvanting Vaccines against Microbes and Tumors
WO2012040101A1 (en) * 2010-09-21 2012-03-29 University Of Miami Compositions and methods for inducing migration by dendritic cells and an immune response

Non-Patent Citations (20)

* Cited by examiner, † Cited by third party
Title
"Advances in Immunology", vol. 93, 2007, ACADEMIC PRESS
"Current Protocols in Immunology", 1992, GREENE PUBLISHING AND WILEY-INTERSCIENCE
"Current Protocols in Molecular Biology", 1992, GREENE PUBLISHING AND WILEY-INTERSCIENCE
"Encyclopedia of Pharmaceutical Technology", 1988, MARCEL DEKKER
"Gene Therapy Protocols (Methods in Molecular Medicine", 1997, HUMANA PRESS
"Gene Therapy: Principles and Applications", 1999, SPRINGER VERLAG
"Making and Using Antibodies: A Practical Handbook", 2006, CRC PRESS
"Medical Immunology", 2007, INFORMA HEALTHCARE PRESS
"Molecular Cloning: A Laboratory Manual", vol. 1-3, 2001, COLD SPRING HARBOR LABORATORY PRESS
"Nonviral Vectors for Gene Therapy: Methods and Protocols", 2010, HUMANA PRESS
"Remington: The Science and Practice of Pharmacy", 2000, LIPPINCOTT WILLIAMS & WILKINS
"Viral Vectors for Gene Therapy: Methods and Protocols", 2011, HUMANA PRESS
B.A. HANKS ET AL., NATURE MEDICINE, vol. 11, 2005, pages 130 - 137
F. HOU ET AL., CELL, vol. 146, 2011, pages 448 - 461
GUPTA ET AL.: "Design of vaccine adjuvants incorporating TNF superfamily ligands and TNF superfamily molecular mimics", IMMUNOLOGY RESEARCH, vol. 57, 7 November 2013 (2013-11-07), pages 303 - 310, XP055132490 *
HARLOW; LANE: "ANTIBODIES: A Laboratory Manual", 1988, COLD SPRING HARBOR LABORATORY PRESS
R.S. KORNBLUTH; M. STEMPNIAK; G.W. STONE, INTERNATIONAL REVIEW OF IMMUNOLOGY, vol. 31, 2012, pages 279 - 288
S. GUPTA ET AL., JOURNAL OF LEUKOCYTE BIOLOGY, vol. 90, 2011, pages 389 - 398
S. GUPTA ET AL., RETROVIROLOGY, vol. 8, 2011, pages 39
See also references of EP2892930A4

Cited By (24)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2016073875A1 (en) * 2014-11-06 2016-05-12 University Of Maryland, Baltimore CD8α AND T CELL RECEPTOR VARIANTS AND METHODS OF USING SAME IN MODULATING IMMUNE CELL RESPONSES
US10975137B2 (en) 2014-11-06 2021-04-13 University Of Maryland, Baltimore CD8a and t cell receptor variants and methods of using same in modulating immune cell responses
US11441160B2 (en) 2015-12-07 2022-09-13 Nant Holdings Ip, Llc Compositions and methods for viral delivery of neoepitopes and uses thereof
TWI733719B (en) * 2015-12-07 2021-07-21 美商河谷控股Ip有限責任公司 Improved compositions and methods for viral delivery of neoepitopes and uses thereof
US10793875B2 (en) 2015-12-07 2020-10-06 Nant Holdings Ip, Llc Compositions and methods for viral delivery of neoepitopes and uses thereof
WO2018081459A1 (en) * 2016-10-26 2018-05-03 Modernatx, Inc. Messenger ribonucleic acids for enhancing immune responses and methods of use thereof
CN110225976A (en) * 2016-11-28 2019-09-10 奥拉廷加Aio生物 Viral vector construct for activating the gene adjuvant of STING approach to express
WO2018096395A1 (en) 2016-11-28 2018-05-31 Aratinga.Bio Aio Viral vector constructs for expression of genetic adjuvants activating the sting pathway
JP2020500553A (en) * 2016-11-28 2020-01-16 アラティンガ.バイオ エーアイオーAratinga.Bio Aio Viral vector construct for expression of a gene adjuvant that activates the STING pathway
US11674153B2 (en) * 2016-11-28 2023-06-13 Aratinga.Bio Aio Viral vector constructs for expression of genetic adjuvants activating the CD40 and STING pathways
US20200199620A1 (en) * 2016-11-28 2020-06-25 Aratinga.Bio Aio Viral Vector Constructs for Expression of Genetic Adjuvants Activating the CD40 and STING Pathways
JP2020500554A (en) * 2016-11-28 2020-01-16 アラティンガ.バイオ エーアイオーAratinga.Bio Aio Viral vector construct for expression of gene adjuvants that activate CD40 and STING pathway
US11638752B2 (en) * 2016-11-28 2023-05-02 Aratinga.Bio Aio Viral vector constructs for expression of genetic adjuvants activating the STING pathway
WO2018096399A1 (en) 2016-11-28 2018-05-31 Arating.Bio Aio Viral vector constructs for expression of genetic adjuvants activating the cd40 and sting pathways
AU2017363967B2 (en) * 2016-11-28 2022-12-22 Ixaka France SAS Viral vector constructs for expression of genetic adjuvants activating the CD40 and sting pathways
US10881730B2 (en) 2017-02-01 2021-01-05 Modernatx, Inc. Immunomodulatory therapeutic MRNA compositions encoding activating oncogene mutation peptides
WO2019040135A1 (en) * 2017-05-12 2019-02-28 Children's Medical Center Corporation Nucleic acids that manipulate immune pathways
WO2019106432A3 (en) * 2017-11-29 2019-07-18 Aratinga.Bio Aio Viral vector constructs for expression of genetic adjuvants mimicking cd40 signaling
WO2020171680A1 (en) 2019-02-22 2020-08-27 주식회사 레모넥스 Pharmaceutical composition for immune activity or for preventing or treating cancer
EP3936118A4 (en) * 2019-02-22 2022-12-14 Lemonex Inc. Pharmaceutical composition for immune activity or for preventing or treating cancer
KR20200103568A (en) 2019-02-22 2020-09-02 주식회사 레모넥스 Composition for immune enhancing or preventing or treating of cancer
CN110694069A (en) * 2019-11-08 2020-01-17 重庆医科大学附属第二医院 Medicine for preventing and treating fulminant hepatitis
KR20220004588A (en) 2020-07-03 2022-01-11 주식회사 레모넥스 Rna and nucleic acid delivery systems comprising the same
US11701378B2 (en) 2020-07-03 2023-07-18 Lemonex Inc. RNA and nucleic acid carrier including the same

Also Published As

Publication number Publication date
US20150252080A1 (en) 2015-09-10
EP2892930A1 (en) 2015-07-15
US10093701B2 (en) 2018-10-09
EP2892930A4 (en) 2016-01-27
US10329329B2 (en) 2019-06-25
EP2892930B1 (en) 2019-08-14
US20190062380A1 (en) 2019-02-28

Similar Documents

Publication Publication Date Title
US10329329B2 (en) Fusion proteins for promoting an immune response, nucleic acids encoding same, and methods of making and use thereof
US11135284B2 (en) MERS-CoV vaccine
CN101657213B (en) A method for enhancing t cell response
JP2020169216A (en) Vaccines having antigen and interleukin-21 as adjuvant
JP6445523B2 (en) Vaccines containing biomolecular adjuvants
JP2016538246A (en) Vaccine with interleukin-33 as adjuvant
WO2012040101A1 (en) Compositions and methods for inducing migration by dendritic cells and an immune response
CN108601951A (en) Immune regulation composite for treatment
US9474800B2 (en) Expression vector encoding alphavirus replicase and the use thereof as immunological adjuvant
Shi et al. The expression of membrane protein augments the specific responses induced by SARS-CoV nucleocapsid DNA immunization
WO2012040266A2 (en) Gene-based adjuvants and compositions thereof to increase antibody production in response to gene-based vaccines
KR20190123257A (en) Viral Vector Constructs for the Expression of Gene Adjuvant Activating the STING Pathway
US20220184200A1 (en) Virus-like particles and uses thereof
AU2017363967B2 (en) Viral vector constructs for expression of genetic adjuvants activating the CD40 and sting pathways
WO2023044542A1 (en) Sars cov-2 vaccine
Pollard Evaluation of CD8⁺ T cell responses upon mRNA vaccination and the implication of type I IFN signaling

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: 13835331

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 14424562

Country of ref document: US

NENP Non-entry into the national phase

Ref country code: DE