SYSTEMS AND METHODS FOR NUCLEIC ACID EXPRESSION IN VIVO

- DNARx

The present invention provides compositions, systems, kits, and methods for expression of one or more biomolecules in a subject, human or non-human mammal, (e.g., at therapeutic levels for the extended periods of time required to produce therapeutic effects). In certain embodiments, compositions, systems, kits, and methods are provided that comprise a first composition comprising polycationic structures (e.g., empty cationic liposomes, cationic micelles, cationic emulsions, or cationic polymers) and a second composition comprising expression vectors (e.g., non-viral expression vectors not associated with liposomes or other carriers) encoding one or more biomolecules of interest.

Skip to: Description  ·  Claims  · Patent History  ·  Patent History
Description

The present application claims priority to U.S. Provisionals application 62/475,477 filed Mar. 23, 2017, which is herein incorporated by reference in its entirety.

FIELD OF THE INVENTION

The present invention provides compositions, systems, kits, and methods for expression of one or more biomolecules in a subject, human or non-human mammal, (e.g., at therapeutic levels for the extended periods of time required to produce therapeutic effects). In certain embodiments, compositions, systems, kits, and methods are provided that comprise a first composition comprising polycationic structures (e.g., empty cationic liposomes, cationic micelles, cationic emulsions, or cationic polymers) and a second composition comprising expression vectors (e.g., non-viral expression vectors not associated with liposomes or other carriers) encoding one or more biomolecules of interest.

BACKGROUND

The simplest non-viral gene delivery system uses naked expression vector DNA. Direct injection of free DNA into certain tissues, particularly muscle, has been shown to produce high levels of gene expression, and the simplicity of this approach has led to its adoption in a number of clinical protocols. In particular, this approach has been applied to the gene therapy of cancer where the DNA can be injected either directly into the tumor or can be injected into muscle cells in order to express tumor antigens that might function as a cancer vaccine.

Although direct injection of plasmid DNA has been shown to lead to gene expression, the overall level of expression is much lower than with either viral or liposomal vectors. Naked DNA is also generally thought to be unsuitable for systemic administration due to the presence of serum nucleases. As a result, direct injection of plasmid DNA appears to be limited to only a few applications involving tissues that are easily accessible to direct injection such as skin and muscle cells.

SUMMARY OF THE INVENTION

The present invention provides compositions, systems, kits, and methods for expression of one or more biomolecules in a subject, human or non-human mammal, (e.g., at therapeutic levels for the extended periods of time required to produce therapeutic effects). In certain embodiments, compositions, systems, kits, and methods are provided that comprise a first composition comprising polycationic structures (e.g., empty cationic liposomes, cationic micelles, cationic emulsions, or cationic polymers) and a second composition comprising expression vectors (e.g., non-viral expression vectors not associated with liposomes or other carriers) encoding one or more biomolecules of interest. In some embodiments, the compositions, systems, kits, and methods may employ one or more components of the compositions, systems, kits, and methods described in U.S. patent application Ser. No. 15/268,000, filed Sep. 16, 2016, the disclosure of which is herein incorporated by reference in its entirety.

In some embodiments, provided herein are compositions, kits, and/or systems comprising: a) a first composition comprising a first amount of polycationic structures liposomes, wherein the first composition is free, or essentially free, of nucleic acid molecules; and b) a second composition comprising an expression vector (e.g., one or more expression vectors that express a biomolecule of interest) (e.g., a therapeutically effective amount of one or more expression vectors). In some embodiments, the compositions, kits, and/or systems have one or more or all of the following properties: i) the ratio of the first amount of said polycationic structures to the expression vectors is 5:1 to 25:1; ii) 2.0% to 6.0%, or 2.0% to 20% (e.g., 2% . . . 5% . . . 10% . . . 15% . . . or 20%) of the first composition comprises dexamethasone palmitate and/or dexamethasone; iii) the first composition further comprises neutral lipid; and iv) the polycationic structures comprise empty liposomes, wherein the empty liposomes present in the first composition have a z-average diameter of about 20-85 nm.

In some embodiments, provided herein are aqueous compositions comprising or consisting essentially of: a) polycationic structures present in said composition at a concentration of between 500 nM and 500 mM; b) dexamethasone and/or dexamethasone palmitate present in said composition at a concentration between 1-10% of said composition; and c) a physiologically tolerable buffer, and wherein said composition is free, or essentially free, of nucleic acid molecules. In some embodiments, said polycationic structure are cationic lipids that are present as small unilamellar vesicles. In some embodiments, said physiologically tolerable buffer is selected from the group consisting of: saline buffer, 5% dextrose in water, lactated ringers buffer, and any combination thereof. In some embodiments, said polycationic structures comprise DOTAP. In some embodiments, said polycationic structures are present in said composition at a concentration of between 800 nM and 1500 nM, or between 10 mM and 100 mM.

In some embodiments, provided herein are aqueous compositions comprising or consisting essentially of: a) neutral lipids present in said composition at a concentration of between 500 nM and 500 mM; b) dexamethasone and/or dexamethasone palmitate present in said composition at a concentration between 1-10% of said composition; and c) a physiologically tolerable buffer, and wherein said composition is free, or essentially free, of nucleic acid molecules. In some embodiments, said neutral lipids are present as multi-lamellar vesicles. In some embodiments, said physiologically tolerable buffer is selected from the group consisting of: saline buffer, 5% dextrose in water, lactated ringers buffer, and any combination thereof. In some embodiments, said neutral lipids comprise DMPC. In some embodiments, said neutral lipids are present in said composition at a concentration of between 800 nM and 1500 nM, or between 10 mM and 100 mM.

In some embodiments, provided herein are aqueous compositions comprising or consisting essentially of: a) polycationic structures present in said composition at a concentration of between 500 nM and 500 mM; b) neutral lipids present in said composition at a concentration of between 500 nM and 500 mM; and c) a physiologically tolerable buffer, and wherein said composition is free, or essentially free, of nucleic acid molecules. In some embodiments, said neutral lipids are present as multi-lamellar vesicles. In some embodiments, compositions comprise neutral liposomes with (e.g., extruded to) mean diameters of 75-250 nm (e.g., 100 nm, 150 nm, 200 nm, etc.). In some embodiments, said polycationic structure are cationic lipids that are present as small unilamellar vesicles. In some embodiments, said polycationic structure are cationic lipids that are present as multilamellar vesicles. In some embodiments, compositions further comprise: d) dexamethasone, wherein said dexamethasone is present in a concentration such that it is 1-10% of said composition. In some embodiments, compositions further comprise: d) dexamethasone palmitate, wherein said dexamethasone palmitate is present in a concentration such that it is 1-10% of said composition. In some embodiments, said neutral lipids comprise DMPC. In some embodiments, said neutral lipids are present in said composition at a concentration of between 800 nM and 1300 nM, or between 10 mM and 100 mM. In some embodiments, said polycationic structures comprise DOTAP. In some embodiments, said polycationic structures are present in said composition at a concentration of between 800 nM and 1500 nM, or between 10 mM and 100 mM.

In some embodiments, provided herein are compositions, kits, and/or systems comprising: a) a first composition comprising a first amount of polycationic structures liposomes, wherein the first composition is free, or essentially free, of nucleic acid molecules; and b) a second composition comprising a therapeutically effective amount of expression vectors, wherein the expression vectors comprise circularized synthetically amplified nucleic acid, or minicircle DNA, encoding one or more biomolecules (e.g., therapeutic biomolecules). In some embodiments, the synthetically amplified nucleic acid is produced by a polymerase chain reaction. In some embodiments, no cells are used in the production of the vector (e.g., the vector is not recombinantly expressed in culture for its production). In some embodiments, the vector consists of the nucleic acid encoding one or more biomolecules and one or more promoters and enhancers operatively linked thereto.

In some embodiments, the one or more biomolecules is a therapeutic biomolecule. In some embodiments, the therapeutic biomolecule is an antibody. In some embodiments, the antibody is a broadly neutralizing antibody. In some embodiments, the broadly neutralizing antibody specifically binds to a pathogen or pathogen component. In some embodiments, the pathogen is a virus. In some embodiments, the antibody specifically binds to a tumor antigen. In some embodiments, the mAb or antigen-binding portion thereof specifically binds to a cytokine.

In some embodiments, one or more antibodies comprise a first antibody that specifically binds to a first target molecule, a second antibody that specifically binds to a second, different, target molecule, and in some embodiments, a third antibody that specifically binds to a third, different, target molecule.

In some embodiments, the one or more biomolecules comprise a CRISPR/Cas9 component (e.g., for use in gene therapy, research, or diagnostic applications).

In some embodiments, the one or more biomolecules comprise a nucleic acid (e.g., a therapeutic or diagnostic nucleic acid). In some embodiments, the nucleic acid is an antisense oligonucleotide (see e.g., 7,592,440, 7,919,472, and 9,045,754, herein incorporated by reference in their entireties). In some embodiments, the nucleic acid is a ribozyme. In some embodiments, the nucleic acid is an shRNA, miRNA, siRNA, piRNA, snoRNA, tsRNA, or srRNA.

In some embodiments, the expression vectors comprise a super enhancer regulating expression of at least one of the one or more biomolecules. In some embodiments, the expression vectors encode a first therapeutic biomolecule and a second therapeutic biomolecule, wherein said first and second therapeutic biomolecules express for different lengths of time than one another (e.g., expressed using different promoters and/or enhancers or expressed in different expression cassettes within the vectors). In certain embodiments, the expression vectors encode a first therapeutic biomolecule and a second therapeutic biomolecule, wherein said first and second therapeutic biomolecules are the same (e.g., a single vector has two expression cassettes that both express the same or different therapeutic biomolecule). In further embodiments, the expression vectors encode a first therapeutic biomolecule, a second therapeutic biomolecule, and a third therapeutic biomolecule wherein said first, second, third therapeutic biomolecules are all the same (e.g., a single vector has three expression cassettes that all express the same or different therapeutic biomolecule).

In some embodiments, the expression vectors comprise an R6K origin of replication. In some embodiments, the expression vectors are CpG-free or CpG-reduced. In some embodiments, the expression vectors are not CpG-free or CpG-reduced.

Further provided herein are methods of using the compositions, kits, and/or system described herein. For example, in some embodiments, provided herein are methods of expressing one or more therapeutic biomolecules in a subject, comprising: a) administering a first composition of the system into a subject; and b) administering a second composition of the system into said subject.

In some embodiments, provided herein are methods of expressing two or more therapeutic biomolecules in a subject for different durations, comprising: a) administering a first composition of a system into a subject; and b) administering (e.g., subsequently, within 300 minutes) a second composition of the system into the subject, wherein the vectors express first and second biomolecules each in a separate cassette or under the control of different promoters and/or enhances such that the first and second biomolecules express for different lengths of time than one another in the subject. For example, the first molecule expressed from the first cassette is present for at least seven, or 21 or 100 days following injection, whereas the biomolecule expressed from the second cassette remains present for less than seven days or fourteen 14 days.

In some embodiments, step b) of the method occurs from 1 to 400 minutes after step a) (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 12, 15, 20, 30, 40, 50, 100, 200, 300, 400 minutes or any increments therein between). In some embodiments, the biomolecule or biomolecules are expressed at a desired (e.g., therapeutic) level in the subject for at least 7 consecutive days (e.g., at least 30 days, at least 1 year or any increments therein between). In certain embodiments, the first therapeutic biomolecule expresses for at least 7 days (e.g., 7 . . . 14 . . . 21 . . . 190 . . . 365 days) at therapeutic levels, and the second therapeutic biomolecule expresses at a level that is at least 50% reduced (e.g., at least 50% . . . 65% . . . 75% . . . 90% . . . or 99% reduced) at day 7 compared to the initial expression of said second therapeutic biomolecules on day 1. In particular embodiments, the first therapeutic biomolecule expresses for at least 14 days at therapeutic levels, and the second therapeutic biomolecule expresses at a level that is at least 75% reduced at day 14 compared to the initial expression of said second therapeutic biomolecules on day 1. In additional embodiments, the first therapeutic biomolecules comprises a CRISP or a monoclonal antibody sequence (or fragment thereof, such as F(ab)2), and the second therapeutic biomolecule comprises a Cas9 protein.

In some embodiments (e.g., to increase expression levels of the desired biomolecule), the methods further comprise step c): repeating steps a) and b) one or more times.

In some embodiments, the biomolecules are therapeutic biomolecules. Any suitable or desired therapeutic biomolecule may be selected. In some embodiments, the one or more therapeutic biomolecules comprises an anti-PCSK9 monoclonal antibody that expresses at sufficient levels to reduce LDL (e.g., in a human subject). In some embodiments, the one or more therapeutic biomolecules expresses an anti-influenza A stem antigen monoclonal antibody (e.g., that broadly immunizes against a wide spectrum of influenza A strains). In some embodiments, the one or more therapeutic biomolecules expresses a combination of anti-CD20 and anti-CD47 monoclonal antibodies. In some embodiments, the one or more therapeutic biomolecules expresses an anti-PD-1 monoclonal antibody, one or more self-tumor neoantigens, and optionally one or more immunomodulatory cytokines.

In some embodiments, the present invention provides compositions, systems, kits, and methods for expression of a protein or proteins and/or biologically active nucleic acid molecule(s) in a subject (e.g., at therapeutic levels for the extended periods of time required to produce therapeutic effects in the host). In certain embodiments, systems and kits are provided that comprise a first composition comprising a first amount of polycationic structures (e.g., empty cationic liposomes, empty cationic micelles, or empty cationic emulsions), and a second composition comprising a therapeutically effective amount of expression vector(s) (e.g., non-viral expression vectors not associated with liposomes) (e.g., that are CpG-free or CpG-reduced), where the expression vectors comprise a first nucleic acid sequence encoding: i) a first therapeutic protein (or non-therapeutic protein, such as a marker protein), and/or ii) a first biologically active nucleic acid molecule. In certain embodiments, the expression vector comprises a second, third, or fourth nucleic acid sequence encoding a second, third, and/or fourth therapeutic or non-therapeutic protein, and/or a second, third, or fourth biologically active nucleic acid molecule. In some embodiments, the first nucleic acid sequences further encode a second, third, fourth, fifth, and/or sixth therapeutic protein, and/or a second, third, fourth, fifth, and/or sixth biologically active nucleic acid molecule. In other embodiments, such first and second compositions are sequentially administered (e.g., systemically) to a subject such that the therapeutic protein(s) and/or the biologically active nucleic acid molecule(s) is/are expressed in the subject (e.g., at a therapeutic level (e.g., for at least 5 or at least 50 days, or at least 100 . . . 200 . . . or at least 400 days), such that disease(s) or condition(s) is/are treated or physiological trait(s) is/are altered; at a prophylactic level (e.g., for at least 5 or at least 50 days, or at least 100 . . . 200 . . . or at least 400 days), such that disease(s), condition(s), and/or infection(s) is/are prevented).

In some embodiments, provided herein are methods of expressing a first therapeutic protein and/or a biologically active nucleic acid molecule in a subject (e.g., human or non-human mammal) comprising: a) administering (e.g., systemically) a first composition to a subject, wherein the first composition comprises first amount of polycationic structures (e.g., empty cationic liposomes, empty cationic micelles, or empty cationic emulsions) and wherein the first composition is free, or essentially free, of nucleic acid molecules (e.g., nucleic acid is un-detectable or barely detectable in the composition); and b) administering (e.g., systemically, intravascularly, etc.) a second composition to the subject (e.g., initiating within about 2 . . . 10 . . . 50 . . . 100 . . . 200 . . . 300 . . . 400 minutes of administering the first composition), wherein the second composition comprises an amount of expression vectors (e.g., non-viral expression vectors not associated with liposomes or any other carrier), wherein the expression vectors are CpG-free or CpG-reduced, wherein each of the expression vectors comprise nucleic acid sequence(s) encoding: i) first, second, third, fourth, fifth, and/or sixth therapeutic protein(s) or non-therapeutic, and/or ii) first, second, third, fourth, fifth, and/or sixth biologically active nucleic acid molecule(s). In certain embodiments, as a result of the administering the first composition and the administering the second composition, the first therapeutic or non-therapeutic protein and/or the biologically active nucleic acid molecule is/are expressed in the subject (e.g., at a therapeutic level, for at least 5 . . . 50 . . . 100 . . . 300 days . . . 400 days or longer, with respect to a disease or condition, or at an effective level sufficient to alter a physiological or disease trait). In certain embodiments, the polycationic structures (e.g., empty liposomes) present in the first composition have a z-average diameter of about 20-85 nm (e.g., 20 . . . 25 . . . 30 . . . 40 . . . 45 . . . 50 . . . 55 . . . 60 . . . 65 . . . 70 . . . 75 . . . 80 . . . 85 nm). In certain embodiments, the polycationic structures are empty liposomes with a z-average diameter of about 72-76 nm, and are small uni-lammellar vesicles.

In some embodiments, provided herein are methods of expressing a first therapeutic or non-therapeutic protein and/or a biologically active nucleic acid molecule in a subject comprising: a) administering a first composition to a subject, wherein the subject has at least one symptom of a disease or condition, or has at least physiological trait to be altered, wherein the first composition comprises a first amount of polycationic structures (e.g., empty cationic liposomes, empty cationic micelles, or empty cationic emulsions), and wherein the first composition is free, or essentially free, of nucleic acid molecules; and b) administering (or initiating administration of) a second composition to the subject within about 100 minutes or about 200 . . . or 400 minutes of administering said first composition, wherein the second composition comprises a therapeutically effective amount of expression vectors (e.g., wherein the expression vectors are CpG-free or CpG-reduced), wherein the expression vectors each comprise a first nucleic acid sequence encoding: i) a first therapeutic or non-therapeutic protein, and/or ii) a first biologically active nucleic acid molecule, c) administering dexamethasone palmitate and/or neutral lipids to the subject, either in said first and/or second composition, or present in a third composition (e.g., within 100 or 200 . . . or 400 minutes of administration of the first or second compositions). In some embodiments, as a result of the administering the first composition, the administering the second composition, and the administering of the dexamethasone palmitate and/or neutral lipids, the first therapeutic protein and/or the biologically active nucleic acid molecule is/are expressed in the subject at a therapeutic level with respect to the disease or condition, or at an effective level sufficient to alter the physiological or disease trait.

In certain embodiments, dexamethasone palmitate is in the first composition, and wherein 2.0% to 20.0% (e.g., 2.0% . . . 2.5% . . . 3.0% . . . 10% . . . 15% . . . or 20%) of the first composition comprises the dexamethasone palmitate. In certain embodiments, the dexamethasone palmitate is administered in the third composition, which is administered before the first and/or second composition is administered, or is administered after the first and/or second composition, but within 100 . . . 400 minutes thereof. In certain embodiments, the methods further comprise d) administering dexamethasone to the subject, either in the first and/or second and/or third composition, or present in a fourth composition (e.g., initiating within 100 or 300 minutes of administration of the first or second or third compositions, such as before any of the administrations or after the other administrations). In certain embodiments, the polycationic structures (e.g., empty liposomes) present in the first composition have a z-average diameter of about 20-85 nm (e.g., 20 . . . 25 . . . 30 . . . 40 . . . 45 . . . 50 . . . 55 . . . 60 . . . 65 . . . 70 . . . 75 . . . 80 . . . 85 nm). In certain embodiments, the polycationic structures are empty liposomes with a z-average diameter of about 72-76 nm, and are small uni-lammellar vesicles.

In some embodiments, A) the ratio is 10:1 to 18:1; B) 2.0% to 20.0% of the first composition comprises dexamethasone or dexamethasone palmitate; and/or C) each of the expression vectors each comprise only a single expression cassette (i.e., no other expression cassettes are present in each vector), wherein the expression cassette comprises the first nucleic acid sequence encoding the first therapeutic protein and a second nucleic acid sequence encoding a second therapeutic protein, and wherein the expression cassette encodes a self-cleaving peptide sequence (or other cleavage sequence) between the first and second nucleic acid sequences. In certain embodiments, the self-cleaving peptide comprises F2A, P2A, T2A or E2A. In some embodiments, a furin recognition site and/or (S)GSG linker are included upstream of self-cleaving peptides to enhance the cleavage efficiency. In particular embodiments, the first therapeutic protein comprises a monoclonal antibody light chain and the second therapeutic protein comprises a heavy chain of said monoclonal antibody (e.g., the light and heavy chains combine to form a monoclonal antibody fragment (e.g., Fab) or monoclonal antibody when expresses in a subject). In certain embodiments, the polycationic structures comprise empty liposomes. In particular embodiments, the empty liposomes present in said first composition have an average diameter of about 50-85 nm. In certain embodiments, the methods further comprise administering an agent or additional regulating expression vectors, either in said first and/or second composition, or present in a third composition, wherein the agent increases or decreases the expression at the therapeutic or effective level, and/or the length of time of the expression at said therapeutic or effective level, compared to when the drug agent is not administered to said subject (e.g., for therapeutics that need to be expressed for only a certain, limited amount of time). In particular embodiments, the agent is selected from colchicine, dexamethasone, dexamethasone palmitate, neutral lipids, valproic acid, theophylline, sildenafil, amlexanox, chloroquine, SAHA, and L-arginine+sildenafil.

In some embodiments, the expression vectors each further comprise a regulating nucleic acid sequence, wherein the regulating nucleic acid sequence reduces the duration of expression of the first nucleic acid sequence that would occur in the absence of said regulating nucleic acid sequence. In other embodiments, the regulating nucleic acid sequence is selected from the group consisting of: a promoter, an enhancer, a second nucleic acid sequence encoding a second protein, and/or a second biologically active nucleic acid molecule. In additional embodiments, the first amount of polycationic structures in the first composition comprises a mixture of at least a first and second different types of cationic liposomes that reduces the expression of the first therapeutic protein and/or first biologically active nucleic acid molecule compared to such expression when only said first or only said second type of cationic liposomes are employed in said method. In particular embodiments, the therapeutic protein is expressed at a level that is above 1 ug/ml (e.g., 1.1-1.5 ug/ml), and wherein said therapeutic protein is expressed at the level in said subject for at least 7 consecutive days (e.g., at least 7 . . . 21 . . . 50 . . . 100 . . . or 400 days).

In certain embodiments, provided herein are methods of expressing a first therapeutic protein and/or a biologically active nucleic acid molecule in a subject comprising: a) administering (e.g., systemically) a first composition to a subject, wherein the subject has at least one symptom of a disease or condition, is at risk of infection from one or more infectious diseases, or has at least physiological trait to be altered (e.g., level of hematopoietic stem cells), wherein the first composition comprises a first amount of polycationic structures (e.g., empty cationic liposomes, empty cationic micelles, or empty cationic emulsions), and wherein the first composition is free, or essentially free, of nucleic acid molecules; and b) administering (e.g., systemically) a second composition to the subject be initiated (or completed) within about 2 . . . 10 . . . 25 . . . 100 . . . 200 or 400 minutes of administering the first composition, wherein the second composition comprises a therapeutically effective amount of expression vectors (e.g., plasmid), wherein the expression vectors are CpG-free or CpG-reduced (e.g., the nucleic acid sequence of the expression vector has been altered to contain fewer CpG di-nucleotides than normally present in the wild-type version of the sequences in the vector) or are CpG-containing vectors (e.g, the the wild-type C-CSF is employed, which contains a plurality of CpG dinucleotides), wherein the expression vectors each comprise nucleic acid sequence(s) encoding: i) a first therapeutic protein (or first and second therapeutic proteins, or first, second, and third therapeutic proteins, etc.), and/or ii) a first biologically active nucleic acid molecule (or first and second or more biologically active nucleic acid molecules), and wherein, as a result of the administering the first composition and the administering the second composition, and wherein, as a result of administering the first and second compositions, the therapeutic protein(s) and/or the biologically active nucleic acid molecule(s) is/are expressed in the subject at a therapeutic level with respect to the disease or condition, or at an effective level sufficient to alter the physiological or disease trait.

In certain embodiments, the expression vectors are not associated with polycationic structures (e.g., empty cationic liposomes, empty cationic micelles, or empty cationic emulsions)), or other molecules, in the second composition (and there are no detectable polycationic structures present in the second composition). In other embodiments, the expression vectors are naked, non-viral, expression vectors (e.g., plasmids). In certain embodiments, the expression vectors are viral expression vectors (e.g., adeno-associated viral vector or adenovirus vector or synthetic mRNA, miRNA, ribozyme or shRNA nucleic acid vectors). In particular embodiments, the first and/or second composition is administered systemically, regionally, transcutaneously, intradermally, orally, intramuscularly, intravenously, into the gastrointestinal tract, bladder or by pulmonary inhalation, or by an intrathecal or intraventricular route.

In certain embodiments, the therapeutic protein or proteins and/or biologically active nucleic acid molecule or molecules is/are expressed at the therapeutic or effective level in the subject on consecutive days for at least 5 . . . 20 . . . 63 . . . 100 . . . 200 . . . 300 days . . . 1 year or more. In some embodiments, the methods further comprise: c) testing the subject (e.g., body imaging or scanning), or a sample (e.g., blood, serum, plasma, tissue, urine, etc.) from the subject, after at least 5 . . . 20 . . . 63 . . . 100 . . . 200 . . . 300 days . . . or 1 year from the administering the first and second compositions, and determining that the therapeutic protein(s) and/or biologically active nucleic acid molecule(s) is/are being expressed in the subject at the therapeutic or effective level (e.g., therapeutic levels have been sustained in the subject for a time period required to produce therapeutic and/or prophylactic effects in the subject due the single treatment of the first and second compositions). In additional embodiments, the methods further comprise: d) generating a written and/or electronic report that indicates the therapeutic protein and/or biologically active nucleic acid molecule is/are being expressed in the subject at the therapeutic or effective level (e.g., for a certain amount of time). In other embodiments, the report is sent to the treating clinician or practitioner and/or patient from a lab that conducted the test.

In some embodiments, the therapeutic protein and/or biologically active nucleic acid molecule is/are expressed at a level of at least 50 pg/ml . . . 100 . . . 500 . . . 1000 . . . 1500 . . . 4000 . . . 8000 . . . 9500 . . . 1,000,000 pg/ml (1 ug/ml) . . . 1.5 ug/ml or higher, and wherein a blood, serum, or plasma sample (or other biological sample) from the subject is assayed to determine that the therapeutic or effective level is achieved for at least 5 . . . 7 . . . 10 . . . 25 . . . 45 . . . 63 . . . 150 . . . 300 days, or longer, after the administration of the first and second compositions. In other embodiments, the therapeutic protein(s) is/are expressed at a level that is at least 50 pg/ml or at least 100 pg/ml or at least 500, 1,000,000 pg/ml (1 ug/ml) . . . 1.5 ug/ml or higher, and wherein the therapeutic protein is expressed at the level in the subject for at least 5 . . . 7 . . . 10 . . . 25 . . . 45 . . . 63 . . . 150 . . . 300 . . . 350 consecutive days. In certain embodiments, the therapeutic protein and/or biologically active nucleic acid molecule is expressed (e.g., at therapeutic levels) in the subject without clinically significant elevated toxicity (e.g., as measured by ALT (alanine aminotransferase) and/or AST (aspartate aminotransferase)) after at least 48 hours following the administration of the first and second compositions.

In certain embodiments, the therapeutic protein is human G-CSF (e.g., as encoded by SEQ ID NO:1, or sequence with at least 98% identity with SEQ ID NO:1) and is expressed in the subject at a therapeutic level of at least 100 pg/ml as measured in a blood, serum, or plasma sample, wherein the therapeutic protein is expressed in the subject for at least seven days, and wherein the disease, condition, or physiological trait is selected from the group consisting of: neutropenia caused by chemotherapy, non-elevated levels of hematopoietic stem cells in blood of a stem cell donor or recipient, heart degeneration, cerebral ischemia, amyotrophic lateral sclerosis, neutrophil deficiency diseases, and radiation exposure. In particular embodiments, the G-CSF is expressed for at least 5, or 6, or 7 days, but no more than about 10 days (e.g., using drugs, promoter/enhancer combinations, additional expression cassette within the nucleic acid vector or additional expressed proteins to limit production to about 10 days to avoid any toxic neutrophilia-related side effects by expression beyond about days). In other embodiments, the therapeutic protein is Rituximab or similar anti-CD20 antibody or antibody fragment. In some embodiments, the therapeutic protein is human Factor IX or similar protein.

In particular embodiments, the therapeutic protein or proteins and/or biologically active nucleic acid molecule or molecules is/are expressed in the subject for a sufficient amount of time at the therapeutic level to reduce or eliminate the at least one symptom (or all symptoms) without the subject having to receive any other treatment that provides the therapeutic protein(s) and/or biologically active nucleic acid molecule(s) to the subject. In further embodiments, during the sufficient time, the subject does not receive any other specific treatment (e.g., no other specific therapeutic treatment that provides the therapeutic protein or biologically active nucleic acid molecule(s) to the subject). In certain embodiments, the subject has multiple symptoms of a disease or diseases, and wherein the sufficient amount of time is such that all or substantially all of the multiple symptoms of the disease(s) and/or the condition(s) are reduced or eliminated in the subject (e.g., permanently, or for at least 20 days . . . 50 days . . . 200 days . . . 1 year or longer). In other embodiments, during the sufficient time, the subject does not receive the any other disease-specific treatment.

In some embodiments, the first amount of the polycationic structures (e.g., empty cationic liposomes, empty cationic micelles, or empty cationic emulsions) is about 0.01-70, 30-50, or 20-60, μmoles per 1 kilogram of the subject (e.g., 0.01 . . . 1 . . . 10 . . . 20 . . . 40 . . . or 60 μmoles per kilogram). In other embodiments, the ratio of the first amount of the polycationic structures (e.g., empty cationic lipids) to the therapeutically effective amount of the expression vectors is 0.5:1 to 25:1, nmoles of polycationic structures (e.g., empty cationic lipids) to 1 μg of expression vectors (e.g., 0.5:1 . . . 1:1 . . . 4:1 . . . 8:1 . . . 12:1 . . . 17:1 . . . 21:1 . . . or 25:1). In certain embodiments, the ratio of the first amount of the polycationic structures (e.g., empty cationic lipids) to the therapeutically effective amount of the expression vectors is 7:1 to 13:1, nmoles of polycationic structures (e.g., empty cationic lipids) to 1 μg of expression vectors. In particular embodiments, the therapeutically effective amount of the expression vectors is 0.001-8.0 milligrams of the expression vectors per 1 kilogram of the subject (e.g., 0.001 . . . 0.1 . . . 3.0 . . . 4.5 . . . 5.7 . . . 7.1 . . . 8.0 milligrams per kilogram). In some embodiments, the therapeutically effective amount of expression vectors is 0.001 to 1 μg per 1 kilogram of the subject (e.g., 0.001 . . . 0.01 . . . 0.1 . . . 1 μg per kilogram of subject). In certain embodiments, the therapeutically effective amount of the expression vectors is about 0.01-4.0 milligrams of the expression vectors per 1 kilogram of the subject.

In some embodiments, the first nucleic acid sequence encodes the first or first and second, or first, second, and third, therapeutic protein(s). In additional embodiments, the first nucleic acid sequence encodes the biologically active nucleic acid molecule(s). In other embodiments, the subject is a human. In additional embodiments, the expression vectors are CpG-free. In other embodiments, the expression vectors are CpG-reduced. In other embodiments, the therapeutic protein(s) is/are human protein(s) or animal protein(s).

In some embodiments, the polycationic structures do not contain cholesterol (e.g., cholesterol free empty cationic micelles or liposomes). In certain embodiments, the cationic liposomes each comprise at least 60% DOTAP and/or DPTAP (e.g., 60% . . . 75% . . . 85% . . . 95% . . . 98% . . . 100% DOTAP and/or DPTAP). In other embodiments, all or substantially all of the cationic liposomes are multi-lamellar vesicles. In further embodiments, all or substantially all of the cationic liposomes are uni-lamellar vesicles. In further embodiments, the cationic liposomes each comprise at least 99% DOTAP or 99% DPTAP. In further embodiments, the empty cationic liposomes each comprise DOTAP and cholesterol. In additional embodiments, the cationic liposomes each comprise about one-third cholesterol and about two-thirds DOTAP and/or DPTAP. In further embodiments, the first nucleic acid sequence encodes human G-CSF (e.g., as shown in SEQ ID NO:1).

In certain embodiments, the biologically active nucleic acid molecule(s) comprises sequence(s) selected from: shRNA sequence(s), miRNA sequence(s), antisense sequence(s), ribozyme(s), and/or CRISPR single guide RNA sequence(s) (sgRNA). In other embodiments, the CRISPR sgRNA comprises: i) a Cas9 nuclease-recruiting sequence (tracRNA), and ii) a target-specific sequence (crRNA) that hybridizes to a sgRNA target site. In particular embodiments, the biologically active nucleic acid molecule targets human p65 (aka, NF-kappa-B p65 or RELA). Any desired combination of one or more RNA sequences may be encoded in a vector or in combination with non-RNA encoded molecules of interest. For example, in some embodiments, multiple expression cassettes (e.g., 2, 3, 4, etc.) are included in the vector, each expressing a different molecule of interest. In some embodiments, the molecules of interest comprise multiple single CRISPR/Cas9 guide cassettes contained in the vector.

In further embodiments, each of the expression vectors further comprises a second nucleic acid sequence encoding: i) a second therapeutic protein, and/or ii) a second biologically active nucleic acid molecule. In some embodiments, each of the expression vectors further comprises a third nucleic acid sequence encoding: i) a third, and/or fourth therapeutic protein, and/or ii) a third, and/or fourth biologically active nucleic acid molecule. In further embodiments, each of the expression vectors further comprise a first promoter associated with the first nucleic acid sequence, and a second promoter associated with the second nucleic acid sequence, and wherein the first and second promoters are the same or different. In other embodiments, the therapeutic or effective expression level of the first nucleic acid sequence and/or the length of time of the therapeutic or effective expression level, is reduced compared to the expression level or the length of time, when the second nucleic acid is not present and/or expressed from the expression vectors. In other embodiments, the first nucleic acid sequence is expressed at the therapeutic level for at least 5 days, but less than 21 days (e.g., 5 . . . 7 . . . 13 . . . 16 . . . 20 . . . and 21 days). In certain embodiments, the first nucleic acid sequence encodes the therapeutic protein, and wherein the therapeutic protein comprises human G-CSF.

In other embodiments, the expression vector provides the expression at the therapeutic or effective level for a first length of time and/or at a first level of expression when each of the expression vectors comprises a first promoter and first enhancer associated with the first nucleic acid sequence, and wherein the first length of time and/or expression level is altered when a second promoter, different from the first promoter, replaces the first promoter, and/or a second enhancer, different from the second enhancer, replaces the second promoter, on the expression vectors. In other embodiments, the expression at the therapeutic or effective level for a first length of time is for at least 10 . . . 15 . . . 45 . . . 100 . . . 200 . . . 300 days, and wherein replacement with the second promoter and/or second enhancer reduces expression at the therapeutic or effective level to a second length of time that is less than 10 . . . 15 . . . 45 . . . 100 . . . 200 days. In other embodiments, each of the expression vectors comprises a first promoter and a first enhancer, and wherein the first promoter and the first enhancer cause expression at the therapeutic level for at least 5 days, but less than 21 . . . 15 . . . or 10 days. In particular embodiments, the first nucleic acid sequence encodes the therapeutic protein, and wherein the therapeutic protein comprises human G-CSF.

In some embodiments, the methods further comprise administering a drug agent or agents, either in the first and/or second composition, or present in a third composition, wherein the drug agent or agents increase or decrease the expression of the first nucleic acid (e.g., at the therapeutic or effective level, and/or the length of time of the expression at the therapeutic or effective level), compared to when the drug agent or agents are not administered to the subject. In particular embodiments, the drug agent increases the expression level of the first nucleic acid in the subject, and wherein the drug is selected from colchicine, an immunosuppressant, dexamethasone, dexamethasone palmitate, sildenafil, or L-arginine+sildenafil. In certain embodiments, the drug (e.g., dexamethasone or dexamethasone palmitate) is present at between 2.0% and 20.0% of a polycationic structure (e.g., empty cationic lipid composition), such as at 2.0% . . . 2.5% . . . 3.5% . . . 4.5% . . . 6.0% . . . 15% . . . or 20%). In other embodiments, the drug (e.g., dexamethasone or dexamethasone palmitate), is administered to the subject before or after the polycationic structure and vector compositions are administered. In certain embodiments, the polycationic structures (e.g., empty liposomes) present in the first composition have a z-average diameter of about 20-85 nm (e.g., 20 . . . 25 . . . 30 . . . 40 . . . 45 . . . 50 . . . 55 . . . 60 . . . 65 . . . 70 . . . 75 . . . 80 . . . 85 nm). In certain embodiments, the polycationic structures are empty liposomes with a z-average diameter of about 72-76 nm, and are small uni-lammellar vesicles. In some embodiments, drug agents (e.g., dexamethasone) are provided to a subject prior to dosing of the first and/or second composition, in addition to incorporating such agents into the first composition.

In other embodiments, the therapeutic protein is expressed at a level of at least two times higher (or at least 3 or 4 or 5 times higher) when the drug agent is administered to the subject compared to when the drug agent is not administered to the subject. In particular embodiments, the drug agent decreases the expression level of the first nucleic acid sequence, and wherein the drug agent is L-arginine. In further embodiments, the therapeutic protein is expressed at a level of at least two times (or at least three times or four times) lower when the drug agent is administered to the subject compared to when the drug agent is not administered to the subject. In some embodiments, the drug agent comprises an anti-inflammatory agent. In additional embodiments, the drug agent is selected from the group consisting of: amlexanox, chloroquine, valproic acid, theophylline, DHA, prostaglandin, and SAHA.

In further embodiments, the expression vectors are free of operable matrix attachment region (MAR) sequences. In certain embodiments, the expression vectors are free of operable EBNA-1 and/or EBV viral sequences. In certain embodiments the subject's blood pressure, immediately prior to said administering said first and second compositions, is not altered (e.g., no physical transfection aids are applied to the subject to attempt to increase expression of the first nucleic acid sequence). In certain embodiments, the expression vectors comprise at least one of the following: an R6K origin of replication (e.g., located in the 3′ or 5′ UTR of a gene in the vector), an hr3 enhancer, a BV3 signal sequence, a Syn21 sequence, a delta-p10 sequence, or an MITD (MHC class I trafficking signal) sequence.

In particular embodiments, the therapeutic level and/or effective level is at least 150 . . . 100 . . . 500 . . . 1000 . . . 1500 . . . 5000 . . . 1,000,000 pg/ml (1 ug/ml) . . . 1.5 ug/ml or higher, and wherein a blood, serum, or plasma sample (or other biological sample) from the subject is determined to be at the therapeutic level and/or effective level at least 7 . . . 10 . . . 25 . . . 45 . . . 63 . . . 150 . . . 300 . . . 400 days or more after the administration of the first and second compositions. In particular embodiments, the sample from the subject is tested with an ELISA assay or by mass spectrometry to determine the expression level.

In some embodiments, the methods further comprise administering a therapeutically effective amount of neutral liposomes to the subject, wherein the neutral liposomes are present in the first and/or second composition, and/are administered in a third composition, and wherein the therapeutically effective amount of neutral liposomes are administered to the subject prior to the administering the second composition. In certain embodiments, the neutral liposomes comprise at least material selected from: phospholipon 90H, hydrogenated soy PC, stearic and palmitic. In other embodiments, the therapeutically effective amount of neutral liposomes are present in the first composition or present in a third composition administered to the subject. In further embodiments, the neutral liposomes are multilamellar vesicles or extruded to 0.2 or 0.1 um. In particular embodiments, administering the therapeutically effective amount of the neutral liposomes causes expression of the first therapeutic protein and/or the biologically active nucleic acid molecule in the subject that is at least 3 . . . 4 . . . 25 . . . 100 . . . 350 . . . or 600 times higher than occurs when the neutral liposomes are not administered to the subject. In certain embodiments, the ratio of empty cationic liposomes to the neutral liposomes administered to the subject is between about 2:1 and 1:5 (e.g., 2:1 . . . 1:1 . . . 2:5 . . . 1:5).

In some embodiments, provided herein are methods of expressing a first therapeutic protein and/or a biologically active nucleic acid molecule in a subject comprising: a) administering a first composition to a subject, wherein the first composition comprises an anti-inflammatory agent; and b) administering or initiating administration of, a second composition to the subject within about 2 minutes . . . 20 minutes . . . 1 hour . . . 24 hours . . . 5 days . . . 7 days . . . 9 days or more of administering the first composition, wherein the second composition comprises a therapeutically effective amount of polyplexes, wherein each polyplex comprises an expression vector and polyethylenimine, wherein the expression vector is CpG-free or CpG-reduced, wherein each expression vector comprises a first nucleic acid sequence encoding: i) a first therapeutic protein (and/or first and second proteins), and/or ii) a first (and/or first and second) biologically active nucleic acid molecule, and wherein, as a result of administering the first composition and administering the second composition, the first therapeutic protein and/or the biologically active nucleic acid molecule is/are expressed in the subject. In further embodiments, the subject has at least one symptom of a disease or condition, or has at least one physiological trait desired to be altered, and wherein the first therapeutic protein and/or the biologically active nucleic acid molecule is expressed at a therapeutic level with respect to the disease, condition, or physiological trait to be altered. In some embodiments, the anti-inflammatory agent is selected from the group consisting of amlexanox, chloroquine, and suberanilohydroxamic acid (SAHA).

In some embodiments, the expression vector comprises a plasmid or other non-viral vector. In further certain embodiments, the administration in step b) is accomplished by systemically administering the second composition.

In some embodiments, provided herein are systems or kits comprising: a) a first composition comprising a first amount of polycationic structures (e.g., empty cationic liposomes, empty cationic micelles, or empty cationic emulsions), wherein the first composition is free, or essentially free, of nucleic acid molecules; and b) a second composition comprises a therapeutically effective amount of expression vectors (e.g., non-viral and not associated with liposomes or other carrier molecules), wherein the expression vectors are CpG-free or CpG-reduced, wherein each of the expression vectors comprises a first nucleic acid sequence encoding: i) a first therapeutic protein or non-therapeutic protein, and/or ii) a first biologically active nucleic acid molecule. In other embodiments, the expression vectors are a naked, non-viral expression vectors (e.g., plasmid). In certain embodiments, at least one of the following applies: i) wherein the ratio of the first amount of the polycationic structure (e.g., empty cationic liposome) to the therapeutically effective amount of expression vectors is 2:1 to 25:1 or 5:1 to 25:1; ii) wherein 2.0% to 20.0% of the first composition comprises dexamethasone palmitate; iii) wherein the first composition further comprises neutral lipid, and iv) wherein the polycationic structures comprise empty liposomes, and wherein the empty liposomes present in the first composition have a z-average diameter of about 20-85 nm (e.g., 20 . . . 25 . . . 30 . . . 40 . . . 45 . . . 50 . . . 55 . . . 60 . . . 65 . . . 70 . . . 75 . . . 80 . . . 85 nm). In certain embodiments, the vectors are viral vectors (e.g., AAV or adeno viral vectors). In particular embodiments, the therapeutic protein is human G-CSF (e.g., as shown in SEQ ID NO:1), or the wild-type human G-CSF which contains a plurality of CpG dinucleotides.

In particular embodiments, the first amount of the polycationic structure (e.g., empty cationic liposomes) is between 0.1 to 7.0 millimoles (e.g., 0.1 . . . 5.0 . . . 7.0 millimoles) or 1.5 and 5.0 millimoles (e.g., suitable amount for administration to a human subject). In other embodiments, the ratio of the first amount of the polycationic structure (e.g., empty cationic liposome) to the therapeutically effective amount of the expression vectors is 0.5:1 to 25:1, nmoles of empty cationic lipid to 1 μg of expression vectors (e.g., 0.5:1 . . . 1:1 . . . 5:1 . . . 10:1 . . . 15:1 . . . 25:1). In some embodiments, the ratio of the first amount of the polycationic structure (e.g., empty cationic lipid) to the therapeutically effective amount of the expression vectors is 7:1 to 13:1, nmoles of polycationic structure to 1 μg of expression vectors (e.g., 7:1 . . . 10:1 . . . or 13:1). In other embodiments, the therapeutically effective amount of the expression vectors is between 0.1 and 800 milligrams (e.g., suitable amount for administration to a human subject, such as when the vector is a plasmid). In certain embodiments, the amount is 1 . . . 25 . . . 400 . . . or 800 milligrams of expression vectors for human administration.

In other embodiments, the first nucleic acid sequence encodes the first therapeutic protein. In additional embodiments, the first nucleic acid sequence encodes the biologically active nucleic acid molecule. In particular embodiments, the expression vectors are CpG-free. In other embodiments, the expression vectors are CpG-reduced. In further embodiments, the first therapeutic protein is a human protein. In other embodiments, the first nucleic acid sequence encodes the therapeutic protein, and wherein the therapeutic protein comprises human G-CSF, Rituximab, a monoclonal antibody or monoclonal antibody fragment (e.g., Fab), or human Factor IX.

In certain embodiments, the empty cationic liposomes, micelles, or emulsions, each comprise at least 60% DOTAP and/or DPTAP (e.g., 60% . . . 75% . . . 85% . . . 95% . . . 98 . . . 100% DOTAP and/or DPTAP), and may be cholesterol-free (e.g., no detectable cholesterol in the composition). In other embodiments, all or substantially all of the empty cationic liposomes, micelles, or emulsions are multilamellar vesicles. In further embodiments, all or substantially all of the empty cationic liposomes, micelles, or emulsions are either unilamellar, multilamellar, or oligolamellar vesicles. In further embodiments, the empty cationic liposomes, micelle, or emulsions each comprise at least 99% DOTAP or at least 99% DPTAP, and may be cholesterol free. In further embodiments, the empty cationic liposomes each comprise DOTAP and/or DPTAP and cholesterol. In additional embodiments, the empty cationic liposomes, micelles, or emulsions each comprise about one-third cholesterol and about two-thirds DOTAP and/or DPTAP.

In certain embodiments, the first biologically active nucleic acid molecule comprises a sequence selected from: an siRNA or shRNA sequence, a miRNA sequence, an antisense sequence, a CRISPR multimerized single guide, and a CRISPR single guide RNA sequence (sgRNA). In other embodiments, the CRISPR sgRNA comprises: i) a Cas9 nuclease-recruiting sequence (tracRNA), and ii) a target-specific sequence (crRNA) that hybridizes to a sgRNA target site.

In further embodiments, each of the expression vectors further comprises a second nucleic acid sequence encoding: i) a second therapeutic protein, and/or ii) a second biologically active nucleic acid molecule. In further embodiments, each of the expression vectors further comprise a first promoter associated with the first nucleic acid sequence, and a second promoter associated with the second nucleic acid sequence, and wherein the first and second promoters are the same or different.

In some embodiments, the kits and systems further comprise a first container and a second container, and wherein the first composition is present in the first container and the second composition is present in the second container. In other embodiments, kits and systems further comprise a packaging component (e.g., cardboard box, plastic pouch, etc.), wherein the first container and the second container are inside the packaging component.

In certain embodiments, the kits and systems further comprise a drug agent or drug agents, wherein the drug agent(s) are present in the first and/or second compositions, or is present in a third composition. In additional embodiments, the drug agent is selected from colchicine, an immunosuppressant, dexamethasone, sildenafil, L-arginine, or L-arginine+sildenafil. In further embodiments, the drug agent comprises an anti-inflammatory agent. In further embodiments, the drug agent is selected from the group consisting of: amlexanox, valproic acid, theophylline, chloroquine, and SAHA.

In particular embodiments, the expression vectors are free of operable matrix attachment region (MAR) sequences. In additional embodiments, the expression vectors are free of operable EBNA-1 and/or EBV viral sequences.

In certain embodiments, the kits and systems further comprise a therapeutically effective amount of neutral liposomes, wherein the neutral liposomes are present in the first and/or second compositions, or is present in a third composition. In additional embodiments, the therapeutically effective amount of neutral liposomes are present in the first composition. In other embodiments, the neutral liposomes are multilamellar or oligo- or uni-lamellar vesicles. In further embodiments, the ratio of empty cationic liposomes or micelles to the neutral liposomes is between about 2:1 and 1:5 (e.g., 2:1 . . . 1:1 . . . 3:5 . . . 1:5).

In some embodiments, provided herein are a first composition and a second, separate, composition for combined use in the treatment of a disease amenable to treatment with in vivo expression of a first therapeutic protein and/or biologically active nucleic acid molecule, wherein the first composition comprises a first amount of polycationic structures (e.g., empty cationic liposomes, empty cationic micelles, or empty cationic emulsions), wherein the first composition is free, or essentially free, of nucleic acid molecules; and b) a second composition that comprises a therapeutically effective amount of expression vectors, wherein the expression vectors are CpG-free or CpG-reduced, wherein each of the expression vectors comprises a first nucleic acid sequence encoding: i) a first therapeutic protein, and/or ii) a first biologically active nucleic acid molecule.

In certain embodiments, provided herein are methods of expressing a first therapeutic protein and/or a biologically active nucleic acid molecule in a subject comprising: a) administering a first composition to a subject, wherein the first composition comprises a first amount of polycationic structures (e.g., empty cationic liposomes, empty cationic micelles, or empty cationic emulsions), and wherein the first composition is free, or essentially free, of nucleic acid molecules; and b) administering a second composition to the subject within about 100 minutes or 200 minutes of administering the first composition, wherein the second composition comprises a therapeutically effective amount of non-viral expression vectors, wherein the expression vectors are CpG-free or CpG-reduced, wherein the expression vectors each comprise a first nucleic acid sequence encoding: i) a first therapeutic protein, and/or ii) a first biologically active nucleic acid molecule, and wherein, as a result of administering the first composition and administering about the second composition, the first therapeutic protein and/or the biologically active nucleic acid molecule is/are expressed in the subject at a level above (e.g., at least 150 . . . 300 . . . 575 . . . 1000 . . . 1500 . . . 2000 . . . 5000 . . . or 1,000,000 pg/ml) (e.g., as measured in a serum sample from the subject (e.g., after 7 . . . 25 . . . 50 days from the first and second administrations).

In certain embodiments, provided herein are methods comprising: administering a composition to a subject comprising a therapeutically effective amount of non-viral expression vectors that are CpG-free or CpG-reduced, or CpG containing vectors, and comprise a first nucleic acid sequence encoding: i) a first therapeutic protein, and/or ii) a first biologically active nucleic acid molecule, and wherein, as a result of administering the first and second compositions, the first therapeutic protein and/or the biologically active nucleic acid molecule is/are expressed in the subject at a level above 100 pg/ml (e.g., at least 150 . . . 400 . . . 1200 . . . 2000 . . . 5000 . . . or more than 1,000,000 pg/ml) (e.g., as measured in a serum sample from the subject (e.g., after 7 . . . 25 . . . 50 days from the first and second administrations).

In certain embodiments, the polycationic structures comprise empty cationic liposomes, micelles, or emulsions. In other embodiments, the polycationic structures comprise one or more of the following, either alone or combined with polycationic structures: linear or branched polyethyleneimine, dendrimers (e.g., 4th generation pamaam dendrimer based on ethylene diamine, polylysine, polyarginine, and protamine sulfate), poly-lysine, and protamine sulfate. In certain embodiments, the polycationic structures are provided as a cationic emulsion. In particular embodiments, the surfactants in the emulsions are selected from: cetylpyridinium chloride, cetyltrimethylammonium bromide or the like. In other embodiments, the emulsions further comprise a neutral component, such as tweens, spans and triglycerides. In particular embodiments, the emulsions comprise a cationic lipid, such as, for example, DOTAP, DPTAP, DOTMA, or DDAB. In some embodiments, the emulsions are self-emulsifying emulsions or microemulsions (SEDDS, SMEDDS).

In some embodiments, provided herein are methods of expressing a first and second proteins and/or first and second biologically active nucleic acid molecules in a subject comprising: a) administering a first composition to a subject, wherein the subject has at least one symptom of a disease or condition, or has at least physiological trait to be altered, wherein said first composition comprises a first amount of polycationic structures, and wherein said first composition is free, or essentially free, of nucleic acid molecules; and b) administering a second composition to said subject within about 100 minutes of administering the first composition, wherein the second composition comprises a therapeutically effective amount of expression vectors, wherein the expression vectors are non-viral and are CpG-free or CpG-reduced, wherein the expression vectors each comprise: i) a first expression cassette encoding: A) a first protein, and/or B) a first biologically active nucleic acid molecule, and ii) a second expression cassette encoding: A) a second protein and/or B) a second biologically active nucleic acid molecule. In certain embodiments, as a result of the administering the first composition and the administering the second composition, the first and second proteins and/or said first and second biologically active nucleic acid molecule is/are expressed in the subject at a therapeutic level with respect to the disease or condition, or at an effective level sufficient to alter said physiological or disease trait.

In particular embodiments, the first protein comprises a monoclonal antibody light chain, and the second protein comprises a heavy chain of said monoclonal antibody. In other embodiments, the first and second expression cassettes both comprise regulatory elements. In additional embodiments, the regulatory elements are the same or different in said first and second expression cassettes.

In some embodiments, provided herein are methods of expressing a monoclonal antibody (mAb), Fab, F(ab)2, and/or scFv in a subject comprising: a) administering a first composition to a subject, wherein the subject has at least one symptom of a disease or condition, or has at least physiological trait to be altered, wherein the first composition comprises a first amount of polycationic structures, and wherein the first composition is free, or essentially free, of nucleic acid molecules; and b) administering a second composition to the subject within about 300 minutes of administering the first composition, wherein the second composition comprises a therapeutically effective amount of expression vectors encoding the mAb, the Fab, the F(ab)2, and/or scFv, and wherein, as a result of the administering the first composition and the administering the second composition, the first therapeutic protein is expressed in the subject at a therapeutic level with respect to the disease or condition, or at an effective level sufficient to alter the physiological or disease trait.

In certain embodiments, the ratio of the first amount of the polycationic structures to the therapeutically effective amount of expression vectors is 5:1 to 25:1. In further embodiments, the expression vectors are CpG-free or CpG-reduced. In other embodiments, the expression vectors contain a plurality of CpG motifs, and/or are not CpG-free or CpG-reduced. In additional embodiments, the mAb, the Fab, the F(ab)2, and/or the scFv is/are expressed at a therapeutic level in the subject for at least 7 consecutive days without any further administering. In other embodiments, the at least 7 consecutive days is at least 190 consecutive days without any further administering.

In some embodiments, the F(ab)2 is selected from the group consisting of: F(ab′)2 Afelimomab, Alacizumab pegol, Dorlimomab aritox, Erlizumab, and Igovomab. In additional embodiments, the Fab is selected from the group consisting of: Abciximab, Anatumomab mafenatox, Citatuzumab bogatox, Nacolomab tafenatox, Naptumomab estafenatox, Nofetumomab merpentan, Ranibizumab, Tadocizumab, Telimomab aritox, Arcitumomab, Bectumomab, Biciromab, Certolizumab pegol, and Sulesomab. In certain embodiments, the scFv is selected from the group consisting of: Efungumab, Oportuzumab monatox, and Pexelizumab.

In particular embodiments, the mAb is selected from the group consisting of: 3F8, 8H9, Abagovomab, Abituzumab, Abrilumab, Actoxumab, Adalimumab, Adecatumumab, Aducanumab, Afasevikumab, Afutuzumab, Alemtuzumab, Alirocumab, Altumomab pentetate, Amatuximab, Anetumab ravtansine, Anifrolumab, Anrukinzumab, Apolizumab, Ascrinvacumab, Aselizumab, Atezolizumab, Atinumab, Atlizumab, Atorolimumab, Avelumab, Bapineuzumab, Basiliximab, Bavituximab, Begelomab, Belimumab, Benralizumab, Bertilimumab, Besilesomab, Bevacizumab, Bezlotoxumab, Bimagrumab, Bimekizumab, Bivatuzumab mertansine, Bleselumab, Blontuvetmab, Blosozumab, Bococizumab, Brazikumab, Brentuximab vedotin, Briakinumab, Brodalumab, Brolucizumab, Brontictuzumab, Burosumab, Cabiralizumab, Canakinumab, Cantuzumab mertansine, Cantuzumab ravtansine, Caplacizumab, Capromab pendetide, Carlumab, Carotuximab, cBR96-doxorubicin immunoconjugate, Cedelizumab, Cergutuzumab amunaleukin, Cetuximab, Ch. 14.18, Cixutumumab, Clazakizumab, Clenoliximab, Clivatuzumab tetraxetan, Codrituzumab, Coltuximab ravtansine, Conatumumab, Concizumab, CR6261, Crenezumab, Crotedumab, Dacetuzumab, Daclizumab, Dalotuzumab, Dapirolizumab pegol, Daratumumab, Dectrekumab, Demcizumab, Denintuzumab mafodotin, Denosumab, Depatuxizumab mafodotin, Derlotuximab biotin, Detumomab, Dinutuximab, Diridavumab, Domagrozumab, Drozitumab, Duligotumab, Dupilumab, Durvalumab, Dusigitumab, Ecromeximab, Eculizumab, Edobacomab, Edrecolomab, Efalizumab, Eldelumab, Elgemtumab, Elotuzumab, Elsilimomab, Emactuzumab, Emibetuzumab, Emicizumab, Enavatuzumab, Enfortumab vedotin, Enlimomab pegol, Enoblituzumab, Enokizumab, Enoticumab, Ensituximab, Epitumomab cituxetan, and Epratuzumab.

In some embodiments, the mAb is selected from the group consisting of: Erenumab, Etaracizumab, Etrolizumab, Evinacumab, Evolocumab, Exbivirumab, Fanolesomab, Faralimomab, Farletuzumab, Fasinumab, Felvizumab, Fezakinumab, Fibatuzumab, Ficlatuzumab, Figitumumab, Firivumab, Flanvotumab, Fletikumab, Fontolizumab, Foralumab, Foravirumab, Fresolimumab, Fulranumab, Futuximab, Galcanezumab, Galiximab, Ganitumab, Gantenerumab, Gavilimomab, Gemtuzumab ozogamicin, Gevokizumab, Girentuximab, Glembatumumab vedotin, Golimumab, Gomiliximab, Guselkumab, Ibalizumab, Ibritumomab tiuxetan, Icrucumab, Idarucizumab, IMAB362, Imalumab, Imciromab, Imgatuzumab, Inclacumab, Indatuximab ravtansine, Indusatumab vedotin, Inebilizumab, Infliximab, Inolimomab, Inotuzumab ozogamicin, Intetumumab, Ipilimumab, Iratumumab, Isatuximab, Itolizumab, Ixekizumab, Keliximab, Labetuzumab, Lampalizumab, Lanadelumab, Landogrozumab, Laprituximab emtansine, Lebrikizumab, Lemalesomab, Lendalizumab, Lenzilumab, Lerdelimumab, Lexatumumab, Libivirumab, Lifastuzumab vedotin, Ligelizumab, Lilotomab satetraxetan, Lintuzumab, Lirilumab, Lodelcizumab, Lokivetmab, Lorvotuzumab mertansine, Lucatumumab, Lulizumab pegol, Lumiliximab, Lumretuzumab, MABpl, Mapatumumab, Margetuximab, Matuzumab, Mavrilimumab, Mepolizumab, Metelimumab, Milatuzumab, Minretumomab, Mirvetuximab soravtansine, Mitumomab, Mogamulizumab, Monalizumab, Morolimumab, Motavizumab, Moxetumomab pasudotox, Muromonab-CD3, Namilumab, Naratuximab emtansine, Narnatumab, Natalizumab, Navicixizumab, Navivumab, Nebacumab, Necitumumab, Nemolizumab, Nerelimomab, Nesvacumab, Nimotuzumab, Nivolumab, Obiltoxaximab, Obinutuzumab, Ocaratuzumab, Ocrelizumab, Odulimomab, Ofatumumab, Olaratumab, Olokizumab, Omalizumab, Onartuzumab, Ontuxizumab, Opicinumab, Oregovomab, Orticumab, Otelixizumab, Otlertuzumab, Oxelumab, Ozanezumab, Ozoralizumab, Pagibaximab, Palivizumab, Pamrevlumab, Panitumumab, Pankomab, Panobacumab, Parsatuzumab, Pascolizumab, and Pasotuxizumab.

In certain embodiments, the mAb is selected from the group consisting of: Pateclizumab, Patritumab, Pembrolizumab, Perakizumab, Pertuzumab, Pidilizumab, Pinatuzumab vedotin, Pintumomab, Placulumab, Plozalizumab, Pogalizumab, Polatuzumab vedotin, Ponezumab, Prezalizumab, Priliximab, Pritoxaximab, Pritumumab, Quilizumab, Racotumomab, Radretumab, Rafivirumab, Ralpancizumab, Ramucirumab, Raxibacumab, Refanezumab, Regavirumab, Reslizumab, Rilotumumab, Rinucumab, Risankizumab, Rituximab, Rivabazumab pegol, Robatumumab, Roledumab, Romosozumab, Rontalizumab, Rovalpituzumab tesirine, Rovelizumab, Ruplizumab, Sacituzumab govitecan, Samalizumab, Sapelizumab, Sarilumab, Satumomab pendetide, Secukinumab, Seribantumab, Setoxaximab, SGN-CD19A, SGN-CD33A, Sibrotuzumab, Sifalimumab, Siltuximab, Simtuzumab, Siplizumab, Sirukumab, Sofituzumab vedotin, Solanezumab, Sontuzumab, Stamulumab, Suvizumab, Tabalumab, Tacatuzumab tetraxetan, Talizumab, Tamtuvetmab, Tanezumab, Taplitumomab paptox, Tarextumab, Tefibazumab, Tenatumomab, Teneliximab, Teplizumab, Teprotumumab, Tesidolumab, Tetulomab, Tezepelumab, Ticilimumab, Tigatuzumab, Tildrakizumab, Timolumab, Tisotumab vedotin, Tocilizumab, Toralizumab, Tosatoxumab, Tovetumab, Tralokinumab, Trastuzumab, Trastuzumab emtansine, Tregalizumab, Tremelimumab, Trevogrumab, Tucotuzumab celmoleukin, Ublituximab, Ulocuplumab, Urelumab, Urtoxazumab, Ustekinumab, Utomilumab, Vadastuximab talirine, Vandortuzumab vedotin, Vantictumab, Vanucizumab, Vapaliximab, Varlilumab, Vatelizumab, Vedolizumab, Veltuzumab, Vepalimomab, Vesencumab, Visilizumab, Vobarilizumab, Volociximab, Vorsetuzumab mafodotin, Votumumab, Xentuzumab, Zalutumumab, Zanolimumab, Zatuximab, Ziralimumab, and Zolimomab aritox.

In particular embodiments, the disease or condition comprises a viral or bacterial infection. In other embodimetns, the disease or condition comprises cardiovascular disease or atherosclerosis. In further embodiments, the disease or condition comprises an autoimmune disease. In certain embodiments, the disease or condition comprises cancer (e.g., lung cancer, ovarian cancer, skin cancer, lymphoma, brain cancer, prostate cancer, pancreatic cancer, breast cancer, thyroid cancer, colon cancer, etc.). In further embodiments, the method further comprises at least one of the following: i) wherein 2.0% to 20% % of the first composition comprises dexamethasone palmitate; ii) wherein the first composition further comprises neutral lipid; and iii) wherein the polycationic structures comprise empty liposomes, and wherein the empty liposomes present in the first composition have a z-average diameter of about 20-85 nm.

In some embodiments, provided herein are systems and kits comprising: a first composition comprising a first amount of polycationic structures liposomes, wherein the first composition is free, or essentially free, of nucleic acid molecules; and b) a second composition comprising a therapeutically effective amount of expression vectors, wherein the expression vectors comprise nucleic acid sequences encoding a monoclonal antibody (mAb), Fab, F(ab)2, and/or scFv. In further embodiments, the monoclonal antibody (mAb), Fab, F(ab)2, and/or scFv is selected from those recited above and in Table 3 below.

In certain embodiments, at least one of the following applies: i) wherein the ratio of the first amount of the polycationic structures to the therapeutically effective amount of expression vectors is 5:1 to 25:1; ii) wherein 2.0% to 20% of the first composition comprises dexamethasone palmitate; iii) wherein the first composition further comprises neutral lipid; and iv) wherein the polycationic structures comprise empty liposomes, and wherein the empty liposomes present in the first composition have a z-average diameter of about 20-85 nm.

In some embodiments, the neutral lipids comprises 1,2-Dimyristoyl-SN-glycero-3-phosphocholine (DMPC; Dimyristoylphosphatidylcholine). In other embodiments, the neutral lipids are selected from: distearoyl phosphatidyl choline (DSPC), dipalmitoyl phosphatidylcholine (DPPC), palmitoyl oleoyl phosphatidylcholine (POPC), palmitoyl stearoyl phosphatidylcholine (PSPC), egg phosphatidylcholine (EPC), hydrogenated or non-hydrogenated soya phosphatidylcholine (HSPC), or sunflower phosphatidylcholine.

DESCRIPTION OF THE FIGURES

FIG. 1 the CpG-free modified nucleic sequence of h-GCSF (SEQ ID NO:1) and the amino acid sequence of h-GCSF (SEQ ID NO:2). The positions where CpG di-nucleotides have been eliminated are shown in underline in SEQ ID NO:1. These sequences are examples of modified h-GCSF that could be used with the methods, compositions, systems, and kits herein.

FIG. 2A shows that a single IV, sequential injection of cationic liposomes followed by either a dual cassette or a single expression cassette plasmid DNA vector encoding Rituximab produces long term therapeutic serum levels of Rituximab protein.

FIG. 2B shows, as described in Example 1, sera from mice sequentially injected with cationic liposomes followed by a dual cassette anti-CD20 DNA expression vector 162 and subsequently 176 days earlier (see FIG. 2A) lyses CD 20 positive Raji human B lymphoma cells as effectively as high concentrations of recombinant Rituximab monoclonal antibody protein.

FIG. 3 shows serum anti-CD20 levels produced in mice by sequential, IV cationic liposome injection followed by IV injection of either a dual cassette or a single cassette 2A containing DNA vector in mice.

FIG. 4 shows that incorporation of super enhancer elements into DNA expression vectors increases serum anti-CD20 mAb levels in mice, 24 hrs after a single IV injection of a dual cassette anti-CD20 DNA vector.

FIG. 5 shows plasmid 715.1 2a (P2A) (SEQ ID NO:3) which encodes the anti-CD20 mAb heavy and light chain cDNAs separated by a 2A self-cleaving peptide.

FIG. 6 shows plasmid 718.1 (SEQ ID NO:4), which is dual expression cassette plasmid vector that encode the anti-CD20 mAb heavy and light chain cDNAs respectively.

FIG. 7 shows plasmid 902.8 (P2A) (SEQ ID NO:5), which encodes the anti-CD20 mAb heavy and light chain cDNAs separated by a 2A self-cleaving peptide.

FIG. 8 shows plasmid p113.2 (SEQ ID NO:6), which is identical to p718.1, but includes a single super enhancer upstream of the second coding cassette.

FIG. 9 shows anti-p65 CRISPR/Cas9- and anti-p65 ribozyme-mediated knockdown of mouse NFkB-p65 protein 8 days and 1 day, respectively, after IV injection in mice.

FIG. 10 shows anti-p65 CRISPR-mediated knockdown of mouse NFkB-p65 protein 13 days after IV injection.

FIG. 11 shows anti-p65 CRISPR and anti-p65 antisense-mediated knockdown of mouse NFkB-p65 protein 13 days and 1 day, respectively, after IV injection in mice.

FIG. 12 shows anti-p65 shRNA-mediated knockdown of mouse NFkB-p65 protein 1 day after IV injection in mice.

FIG. 13 shows a ribozyme anti-p65 plasmid (SEQ ID NO:7).

FIG. 14 shows a CRISPR1 anti-p65 plasmid (SEQ ID NO:8).

FIG. 15 shows a CRISPR2 anti-p65 plasmid (SEQ ID NO:9).

FIG. 16 shows a CRISPR anti-p65 plasmid (SEQ ID NO:10).

FIGS. 17A-B show results of Example 3 which describes experiments conducted that demonstrate a single IV, sequential injection of cationic liposomes followed up by a plasmid DNA vector encoding the human G-CSF gene produces supra-therapeutic human G-CSF serum protein levels (FIG. 17A) and elevated absolute neutrophil counts (ANC) above normal ANC levels (blue line) (FIG. 17B) for at least the next 582 days in mice.

FIG. 18 shows neutrophil elevation in rat serum following sequential IV injections of DOTAP cationic liposomes followed by plasmid DNA encoding HG-CSF.

FIG. 19 shows the plasmid sequence for Anti-p65 antisense plasmid (SEQ ID NO:11).

FIG. 20 shows the plasmid sequence for an anti-mouse NFkB-p65 shRNA vector p65 shB (FIG. 20, SEQ ID NO:12).

FIG. 21 shows levels of human G-CSF in mouse serum, 24 hours after sequential IV injection of 1050 nmoles of DOTAP cationic liposomes, followed by 70 ug of either HG-CSF plasmid- or different forms of PCR generated, HG-CSF expression cassette DNA.

FIG. 22 shows levels of human G-CSF in mouse serum or plasma (left axis) and thousands per microliter absolute neutrophil counts (ANC) in whole blood (right axis) in mice for at least the next 302 days after initial injection.

FIG. 23 shows human G-CSF levels in mouse serum for 106 days following one sequential injection of cationic liposomes followed by PCR generated DNA with or without an R6K origin of replication.

FIG. 24 shows human G-CSF and corresponding absolute neutrophil counts (ANC, right axis) levels in mice injected sequentially with cationic liposomes with or without neutral lipids or dexamethasone palmitate, followed by plasmid DNA.

FIG. 25 shows the results of Example 6, which shows that use of an second enhancer increases increase human G-CSF expression in mice 1 and 8 days after sequential IV injection.

FIG. 26 shows the nucleic acid sequence of plasmid sv40-mCMVEF1 (SEQ ID NO:13).

FIG. 27 shows the nucleic acid sequence of plasmid mCMV-mCMVEF1 (SEQ ID NO:14).

FIG. 28 shows the nucleic acid sequence of plasmid mCMV-hCMVEF1 (SEQ ID NO:15).

FIG. 29 shows the nucleic acid sequence of plasmid mCMVEF1 (SEQ ID NO:16).

FIG. 30 shows mouse serum levels of human G-CSF, 24 hours after sequential IV injection of liposomes followed by plasmid DNA (first three groups in figure contain super-enhancer elements).

FIG. 31 shows the nucleic acid sequence of plasmid hr3-mCMVEF1 #2 (SEQ ID NO:17).

FIG. 32 shows the nucleic acid sequence of plasmid hr3-mcmvEF1 #5 (SEQ ID NO:18).

FIG. 33 shows the nucleic acid sequence of plasmid hr3-mcmvEF1 #18 (SEQ ID NO:19).

FIG. 34 shows plasma concentration of human Factor IX at 24 hrs after sequential IV injection of liposomes and various different FIX DNA expression plasmids.

FIG. 35 shows the nucleic acid sequence of FIX plasmid (SEQ ID NO:20).

FIG. 36 shows the nucleic acid sequence of FIX R6K1 (SEQ ID NO:21).

FIG. 37 shows the nucleic acid sequence of FIX R6K2 (SEQ ID NO:22).

FIG. 38 shows the nucleic acid sequence of FIX Superenh (SEQ ID NO:23).

FIG. 39 shows the nucleic acid sequence of FIX RNA-out (SEQ ID NO:24).

FIG. 40 shows anti-p65 CRISPR/Cas9-mediated knockdown of mouse NFkB-p65 protein 40 days after sequential IV injection in mice.

FIGS. 41A-B show immunohistochemistry stained slides from experiments on mice with one sequential IV injection of a CRISPR/Cas9 anti-NFkB p65 plasmid DNA vector. FIG. 41A shows ringers treated control, and FIG. 41B shows the CRISPR/Cas9 anti-NFkB p65 treated mouse tissue.

FIGS. 42A-D show IHC results in bone marrow of control and treated mouse 582 days after a single sequential IV injection of cationic liposomes, then an HG-CSF DNA expression vector. FIGS. 42A (20×) and 42B (60×), control bone marrow, show a diverse mix of cell types surround bony trabeculae of normal femoral medullary cavity, with dark-staining erythoid cells particularly obvious. FIGS. 42C (20×) and 42D (60×), treated bone marrow, show a monotonous nearly solid sheet of pale-staining cells replace bony trabecular elements in femoral marrow pale staining myeloid lineage cells (polymorphonuclear leukocytes) with oval, indented oval, band and segmented forms replace most other cell types within femoral marrow.

FIGS. 43A-D show IHC results in spleen tissue of control and treated mouse 582 days after a single sequential IV injection of cationic liposomes, then an HG-CSF DNA expression vector. FIGS. 43A (20×) and 43B (60×), control spleens, show red/dark portions of white (lymphoid) pulp of normal spleen showing diverse cell population. FIGS. 43C (20×) and 43D (60×), treated spleen, show pale-staining myeloid lineage cells (polymorphonuclear leukocytes) with oval, indented oval, band and segmented forms replace most other cell types.

FIGS. 44A-D show IHC results in bone marrow tissue of control and treated rat 168 days after last sequential IV injection of cationic liposomes, then an HG-CSF DNA. FIGS. 44A (20×) and 42B (60×), control bone marrow, show a diversity of cell types with round, dark staining erythroid lineage particularly obvious in femoral marrow. FIGS. 44C (20×) and 44D (60×), treated bone marrow, show pale staining myeloid lineage cells (polymorphonuclear leukocytes) with oval, indented oval, band and segmented forms predominate in femoral marrow. A few clusters of dark-staining erythroid lineage cells remain.

FIGS. 45A-B. FIG. 45A, from the same experiment as FIG. 44, shows control rat, vertebral body at 40×, while FIG. 45B shows the HGCSF rat vertebral body at 40×.

FIG. 46 shows the nucleic acid sequence of plasmid DNARx-31H4-2A (SEQ ID NO:25) which encodes anti-PCSK9 mAb heavy and light chain cDNAs separated by a 2A self-cleaving peptide.

FIG. 47 shows the nucleic acid sequence of plasmid DNARx-31H4 (SEQ ID NO:26) which is a dual expression cassette plasmid vector that encodes a different versions of anti-PCSK9 mAb heavy and light chain cDNA.

FIG. 48 shows the nucleic acid sequence of plasmid DNARx-21B12 (P2A) (SEQ ID NO:27) which encodes anti-PCSK9 mAb heavy and light chain cDNAs separated by a 2A self-cleaving peptide.

FIG. 49 shows the nucleic acid sequence of plasmid DNARx-21B12 (SEQ ID NO:28) which is a dual expression cassette plasmid vector that encodes different versions of anti-PCSK9 mAb heavy and light chain cDNAs.

FIG. 50 shows the nucleic acid sequence of plasmid DNARx-CD47-2A (P2A) (SEQ ID NO:29) which encodes anti-CD47 mAb heavy and light chain cDNAs separated by a 2A self-cleaving peptide.

FIG. 51 shows the nucleic acid sequence of plasmid DNARx-CD47 (SEQ ID NO:30) which is a dual expression cassette plasmid vector that encodes the anti-CD47 mAb heavy and light chain cDNAs respectively.

FIG. 52 shows the nucleic acid sequence of plasmid DNARx-D8-2A (SEQ ID NO:31).

FIG. 53 shows the nucleic acid sequence of plasmid DNARx-F10-2A (SEQ ID NO:32).

FIG. 54 shows the nucleic acid sequence of plasmid DNARx-A66-2A (P2A) (SEQ ID NO:33).

FIG. 55 shows the nucleic acid sequence of plasmid DNARx-D8 (SEQ ID NO:34).

FIG. 56 shows the nucleic acid sequence of plasmid DNARx-F10 (SEQ ID NO:35).

FIG. 57 shows the nucleic acid sequence of plasmid DNARx-A66 (SEQ ID NO:36).

FIG. 58 shows the nucleic acid sequence of plasmid DNARx-HA-MITD (SEQ ID NO:37).

FIG. 59 shows the nucleic acid sequence of plasmid DNARx-SEC-partial HA-MITD (SEQ ID NO:38).

FIG. 60 shows the nucleic acid sequence of plasmid DNARx-D8-2A-HA-MITD (SEQ ID NO:39).

FIG. 61 shows the nucleic acid sequence of plasmid DNARx-F10-2A-HA-MITD (SEQ ID NO:40).

FIG. 62 shows the nucleic acid sequence of plasmid DNARx-A66-2A-HA-MITD (SEQ ID NO:41).

FIG. 63 shows the nucleic acid sequence of plasmid DNARx-D8-2A-SEC-partial-HA-MITD (SEQ ID NO:42).

FIG. 64 shows the nucleic acid sequence of plasmid DNARx-F10-2A-SEC-partial-HA-MITD (SEQ ID NO:43).

FIG. 65 shows the nucleic acid sequence of plasmid DNARx-A66-2A SEC-partial-MITD (SEQ ID NO:44).

FIG. 66 shows the nucleic acid sequence of plasmid 011215 #7 (SEQ ID NO:45).

FIG. 67 shows the nucleic acid sequence of plasmid 011315 #2 (SEQ ID NO:46).

FIG. 68 shows the nucleic acid sequence of plasmid 122014 #235 (SEQ ID NO:47).

FIG. 69 shows the nucleic acid sequence of plasmid DNARx-PD1-2A (P2A) (SEQ ID NO:48).

FIG. 70 shows the nucleic acid sequence of plasmid DNARx-SEC-OVA-MITD (SEQ ID NO:49).

FIG. 71 shows the nucleic acid sequence of plasmid DNARx-SEC-gp70-MITD (SEQ ID NO:50).

FIG. 72 shows the nucleic acid sequence of plasmid DNARx-PD1-2A OVA (SEQ ID NO:51).

FIG. 73 shows the nucleic acid sequence of plasmid DNARx-PD1-2A gp70 (SEQ ID NO:52).

FIG. 74 shows the nucleic acid sequence of plasmid DNARx CD20-2A Cas9 (SEQ ID NO:53).

FIG. 75 shows the nucleic acid sequence of plasmid DNARx CD20-2A HG-CSF (SEQ ID NO:54).

FIG. 76 shows the nucleic acid sequence of plasmid p65 shA2 (SEQ ID NO:55).

FIG. 77 shows the nucleic acid sequence of plasmid PECAM sh control is SEQ ID NO:56, FIG. 77.

FIG. 78 shows anti-p65 CRISPR-mediated knockdown of mouse NFkB-p65 protein days after IV injection.

FIG. 79 shows the nucleic acid sequence of plasmid EF1/U6 RelA1 (020117 #5) (SEQ ID NO:57).

FIG. 80 shows the nucleic acid sequence of plasmid EF1/U6 RelA4 (020117 #8) (SEQ ID NO:58).

FIG. 81 shows the nucleic acid sequence of plasmid hul/EF1/U6 RelA1 (021417 #3) (SEQ ID NO:59).

FIG. 82 shows results of Example 18, which describes how the inclusion of neutral lipids (DMPC) with cationic liposomes increases serum anti-CD20 monoclonal antibody levels in mice.

FIG. 83 shows results from Example 19, which describes that employing dexamethasone palmitate with neutral liposomes further increases gene expression in vivo.

FIG. 84 shows results from Example 20, which describes how including Syn 21 and/or delta-p10 sequences into the vectors increases gene expression.

FIG. 85 shows the nucleic acid sequence of a vector construct that expresses anti-CD20 antibody, and includes Syn21 and delta-p10 sequence (SEQ ID NO:82).

FIGS. 86A-B show the results of Example 21, which shows increased G CSF expression (FIG. 86A) and increased Rituximab anti-CD20 expression (FIG. 86b) when the hr3 super enhancer is included in the plasmid.

FIGS. 87A-B show the results from Example 22, which describes that locating the R6K origin of replication in the 3′ or 5′ UTR of the Factor IX gene increased expression levels at both the 75 ug level (FIG. 87A) and the 60 ug level (FIG. 87B).

FIG. 88A shows results from Example 23, which shows long-term Rituximab expression levels at different time points over 284 days, showing long-term expression.

FIG. 88B shows results from Example 23, which shows that the anti-CD20 mouse sera was able to induce human tumor cell lysis at levels comparable to Rituximab protein.

FIG. 89 shows the results from Example 24, which shows therapeutic anti-IL-5 mAb (2B6) serum levels expressed for at least 92 days in mice.

FIG. 90 shows the nucleic acid sequence of the dual cassette, single plasmid DNA vector used in Example 24, which encodes the anti-human interleukin-5 mAb (Mepoluzimab; 2B6) heavy chain and light chain cDNAs.

FIG. 91A shows the results from Example 25, which shows that fully neutralizing anti-influenze antibody (5J8) is expressed for at least 85 days in mice.

FIG. 91B shows the results from Example 25, which shows anti-influenza antibody expressed effectively neutralizes the Ca109 epidemic influenza strain for >92 days.

FIG. 92 shows the nucleic acid sequence (SEQ ID NO:84) of the dual cassette, single plasmid DNA vector used in Example 25, which encodes the anti-influenza antibody (5J8) heavy chain and light chain cDNAs.

FIG. 93 shows the results of Example 26, which shows the expression levels in mice of anti-IL-5mAb as well as hG-CSF were at therapeutic levels for at least 66 days.

FIG. 94 shows the nucleic acids sequence (SEQ ID NO:85) of the triple cassette, single plasmid DNA vector used in Example 26, which encodes the anti-human interleukin-5 mAb (Mepoluzimab; 2B6) heavy chain and light chain cDNAs and the human G-CSF cDNA.

FIG. 95 shows results from Example 27, which shows dual-cassette cDNA for hG-CSF expression provides higher serum levels in mice than single cassette hG-CSF expression.

FIG. 96 shows results from Example 28, which shows that the dual cassette vector expressing anti-human IL-5 heavy and light chains produces higher anti-human IL-5 serum mAb levels than the single cassette anti-human IL-5 encoding DNA vector.

FIG. 97 show the results from Example 29, which shows that the dual cassette vector expressing anti-5J8 mAb produces higher anti-5J8 serum mAb levels in vivo than the single cassette anti-5J8 encoding DNA vector.

FIG. 98 shows the results from Example 30, which shows how a dual cassette single plasmid expresses different mAbs in vivo, and how two single cassette plasmids that are co-injected express different mAbs in vivo.

FIGS. 99A-B. FIG. 99A shows results from Example 31, which shows serum expression levels of the anti-human IL-5 mAb over 43 days, and FIG. 99B shows serum expression levels of the anti-influenza A mAb over 43 days.

FIG. 100 shows the results from Example 32, which shows simultaneous expression of Rituximab (anti-CD20), anti-IL5 mAb, and anti-influenza mAb, both from a single vector (left side), as well as by co-injection of three separate vectors (right side).

FIG. 101 shows the results of Example 33, which shows that a single plasmid vector expressing anti-PCSK9 mAbs reduces LDL levels in mice.

FIG. 102 shows the results of Example 34, which shows long-term reduction in LDL levels in mice expressing anti-PCSK9 mAbs.

FIG. 103 shows the results of Example 35, which shows that mice expressing the anti-PCSK9 mAbs had lower LDL levels over time compared to the control mice expressing the control anti-CD20 antibodies.

FIG. 104 show the results of Example 36, which shows expression of anti-flu FI6 mAb for about 25 days, expression of anti-flu 5J8 mAb and anti-IL4 mAB for over 100 days, and expression of Rituximab for over 275 days.

FIG. 105 shows results from Example 37, which shows good expression levels from all four plasmid doses tested.

FIG. 106 shows the results of Example 38, which shows enhanced expression of the mAb by using the BV3 signal sequence.

FIGS. 107A-D show results from Example 39. FIG. 107A shows control mouse lung tissue, and FIG. 107B shows human p53 injected mouse lung tissue stained for p53, showing that the p53 gene is widely expressed in mouse lungs. FIGS. 107C and 107D, shows stained mouse tissue showing predominate vascular endothelial cell human p53 expression in p53-injected mice.

FIG. 108 shows the nucleic acid sequence (SEQ ID NO:86) plasmid DNA vector used in Example 39, which encodes human p53.

FIG. 109 shows injection of DOTAP liposomes as SUV, 0.1 μm extruded or MLV each efficiently expresses hG-CSF in mice. Three mice per group were given sequential IV injections. The first injection contained either 800 nmol or 1000 nmol of cationic liposomes. The cationic liposomes were one of three sizes: MLV, SUV, or 0.1 micron extruded. The first injection was followed two minutes later by a second injection of plasmid vector encoding hG-CSF, injected at either 100 ug or 120 ug.

FIG. 110 shows co-injection of either DMPC or egg PC neutral liposomes as either 0.2 μm extruded or MLV each efficiently expresses hG-CSF in mice. Three mice per group were given sequential IV injections. The first injection contained 800 nmol of DOTAP SUV cationic liposomes, along with 500 nmol neutral liposomes. Neutral liposomes were either egg PC or DMPC, and were either MLV or 0.2 micron extruded. The first injection was followed two minutes later by 90 ug of plasmid vector encoding hG-CSF DNA.

DEFINITIONS

As used herein, the phrase “CpG-reduced” refers to a nucleic acid sequence or expression vector that has less CpG di-nucleotides than present in the wild-type versions of the sequence or vector. “CpG-free” means the subject nucleic acid sequence or vector does not have any CpG di-nucleotides. An initial sequence, that contains CpG dinucleotides (e.g., wild-type version of human G-CSF), may be modified to remove CpG dinucleotides by altering the nucleic acid sequence. Such CpG di-nucleotides can be suitably reduced or eliminated not just in a coding sequence, but also in the non-coding sequences, including, e.g., 5′ and 3′ untranslated regions (UTRs), promoter, enhancer, polyA, ITRs, introns, and any other sequences present in the nucleic acid molecule or vector.

As used herein, “empty liposomes” refers to liposomes that do not contain nucleic acid molecules but that may contain other bioactive molecules (e.g., liposomes that are only composed of the lipid molecules themselves, or only lipid molecules and a small molecule drug).

As used herein, “empty cationic micelles” refers to cationic micelles that do not contain nucleic acid molecules but that may contain other bioactive molecules (e.g., micelles that are only composed of lipid and surfactant molecules themselves, or only lipid and surfactant molecules and a small molecule drug).

As used herein, “empty cationic emulsions” refers to cationic emulsions or microemulsions that do not contain nucleic acid molecules but that may contain other bioactive molecules.

DETAILED DESCRIPTION

The present invention provides compositions, systems, kits, and methods for expression of one or more biomolecules in a subject, human or non-human mammal, (e.g., at therapeutic levels for the extended periods of time required to produce therapeutic effects). In certain embodiments, compositions, systems, kits, and methods are provided that comprise a first composition comprising polycationic structures (e.g., empty cationic liposomes, cationic micelles, cationic emulsions, or cationic polymers) and a second composition comprising expression vectors (e.g., non-viral expression vectors not associated with liposomes or other carriers) encoding one or more biomolecules of interest.

The present disclosure provides methods, systems, and compositions, that allow a single injection (e.g., intravenous injection) of cationic liposomes, followed shortly thereafter by injection (e.g., intravenous injection) of vectors encoding a therapeutic protein produces circulating protein levels many times (e.g., 2-20 times higher) than the therapeutic serum level for the protein for a prolonged period, such at 190 days or over 500 days. Thus, the approach provided herein allows for successful therapeutic application of systemic non-viral gene delivery.

In addition, the systems, methods, and compositions provided herein provide a versatile (e.g., non-viral) gene delivery and expression platform that can much more precisely control the duration of expression of delivered genes at therapeutic levels. This ability to control the duration of expression of delivered genes addresses a need within the gene therapy field, the ability to control the duration at which proteins are expressed at therapeutic levels. Specifically, there is now a wide and expanding spectrum of FDA-approved, recombinant, secreted human protein therapies. Different approved protein therapies must be present at therapeutic levels for very different durations in order to both effectively and safely treat patients. Recommended treatment durations of different protein therapies vary from less than two weeks (HG-CSF) to the lifetime of the patient (factor IX). For example, recombinant human G-CSF protein, Neupogen, is given daily for only the first 10 days of each three-week chemotherapy cycle. Serum HG-CSF levels return to baseline approximately 14 hours after each daily Neupogen dose. This 10 day treatment schedule is used because its neutrophil increasing effect is indicated only during this approximately 10 day period of chemotherapy-induced neutropenia. G-CSF elevation from days 11 to 21 is generally not beneficial, as the patient's own neutrophil producing capacity returns. Giving Neupogen beyond day 10 can cause toxic, neutrophilia-related side effects. In contrast, anti-TNF antibodies are routinely administered for months or years, and factor IX replacement for the lifetime of the patient. Thus, different proteins must be produced at therapeutic levels for different durations, from less than two weeks to the lifetime of the patient. Therefore, a gene therapy approach that can control the duration of gene expression at therapeutic levels it produces in patients achieves therapeutic endpoints while avoiding toxic side effects for a wide spectrum of now FDA-approved, human therapeutic proteins. Provided herein are various technologies that can be employed to provide this control.

In certain embodiments, the present disclosure employs polycationic structures (e.g., empty cationic liposomes, empty cationic micelles, or empty cationic emulsions) not containing vector DNA, which are administered to a subject prior to vector administration. In certain embodiments, the polycationic structures are cationic lipids and/or are provided as an emulsion. The present disclosure is not limited to the cationic lipids employed, which can be composed, in some embodiments, of one or more of the following: DDAB, dimethyldioctadecyl ammonium bromide; DPTAP (1,2-dipalmitoyl 3-trimethylammonium propane); DHA; prostaglandin, N-[1-(2,3-Dioloyloxy)propyl]-N,N,N-trimethylammonium methylsulfate; 1,2-diacyl-3-trimethylammonium-propanes, (including but not limited to, dioleoyl (DOTAP), dimyristoyl, dipalmitoyl, disearoyl); 1,2-diacyl-3-dimethylammonium-propanes, (including but not limited to, dioleoyl, dimyristoyl, dipalmitoyl, disearoyl) DOTMA, N[1-[2,3-bis(oleoyloxy)]propyl]-N,N,N-trimethylammoniu-m chloride; DOGS, dioctadecylamidoglycylspermine; DC-cholesterol, 3.beta.-[N-(N′,N′-dimethylaminoethane)carbamoyl]cholesterol; DOSPA, 2,3-dioleoyloxy-N-(2(sperminecarboxamido)-ethyl)-N,N-dimethyl-1-propanami-nium trifluoroacetate; 1,2-diacyl-sn-glycero-3-ethylphosphocholines (including but not limited to dioleoyl (DOEPC), dilauroyl, dimyristoyl, dipalmitoyl, distearoyl, palmitoyl-oleoyl); beta-alanyl cholesterol; CTAB, cetyl trimethyl ammonium bromide; diC14-amidine, N-t-butyl-N′-tetradecyl-3-tetradecylaminopropionamidine; 14Dea2, O,O′-ditetradecanolyl-N-(trimethylammonioacetyl) diethanolamine chloride; DOSPER, 1,3-dioleoyloxy-2-(6-carboxy-spermyl)-propylamide; N,N,N,N-tetramethyl-N,N-bis(2-hydroxylethyl)-2,3-dioleoyloxy-1,4-butan-ediammonium iodide; 1-[2-acyloxy)ethyl]2-alkyl (alkenyl)-3-(2-hydroxyethyl-) imidazolinium chloride derivatives such as 1-[2-(9(Z)-octadecenoyloxy)eth-yl]-2-(8(Z)-heptadecenyl-3-(2-hydroxyethyl)imidazolinium chloride (DOTIM), 1-[2-(hexadecanoyloxy)ethyl]-2-pentadecyl-3-(2-hydroxyethyl)imidazolinium chloride (DPTIM); 1-[2-tetradecanoyloxy)ethyl]-2-tridecyl-3-(2-hydroxyeth-yl)imidazolium chloride (DMTIM) (e.g., as described in Solodin et al. (1995) Biochem. 43:13537-13544, herein incorporated by reference); 2,3-dialkyloxypropyl quaternary ammonium compound derivates, containing a hydroxyalkyl moiety on the quaternary amine, such as 1,2-dioleoyl-3-dimethyl-hydroxyethyl ammonium bromide (DORI); 1,2-dioleyloxypropyl-3-dimethyl-hydroxyethyl ammonium bromide (DORIE); 1,2-dioleyloxypropyl-3-dimethyl-hydroxypropyl ammonium bromide (DORIE-HP), 1,2-dioleyloxypropyl-3-dimethyl-hydroxybutyl ammonium bromide (DORIE-HB); 1,2-dioleyloxypropyl-3-dimethyl-hydroxypentyl ammonium bromide (DORIE-HPe); 1,2-dimyristyloxypropyl-3-dimethyl-hydroxylethyl ammonium bromide (DMRIE); 1,2-dipalmityloxypropyl-3-dimethyl-hydroxyethyl ammonium bromide (DPRIE); 1,2-disteryloxypropyl-3-dimethyl-hydroxyethyl ammonium bromide (DSRIE) (e.g., as described in Felgner et al. (1994) J. Biol. Chem. 269:2550-2561, herein incorporated by reference in its entirety). Many of the above-mentioned lipids are available commercially from, e.g., Avanti Polar Lipids, Inc.; Sigma Chemical Co.; Molecular Probes, Inc.; Northern Lipids, Inc.; Roche Molecular Biochemicals; and Promega Corp.

In certain embodiments, the neutral lipids employed with the methods, compositions, systems, and kits includes diacylglycerophosphorylcholine wherein the acyl chains are generally at least 12 carbons in length (e.g., 12 . . . 14 . . . 20 . . . 24 . . . or more carbons in length), and may contain one or more cis or trans double bonds. Examples of said compounds include, but are not limited to, distearoyl phosphatidyl choline (DSPC), dimyristoyl phosphatidylcholine (DMPC), dipalmitoyl phosphatidylcholine (DPPC), palmitoyl oleoyl phosphatidylcholine (POPC), palmitoyl stearoyl phosphatidylcholine (PSPC), egg phosphatidylcholine (EPC), hydrogenated or non-hydrogenated soya phosphatidylcholine (HSPC), or sunflower phosphatidylcholine.

In certain embodiments, the neutral lipids include, for example, up to 70 mol diacylglycerophosphorylethanolamine/100 mol phospholipid (e.g., 10/100 mol 25/100 mol . . . 50/100 . . . 70/100 mol). In some embodiments, the diacylglycerophosphorylethanolamine has acyl chains that are generally at least 12 carbons in length (e.g., 12 . . . 14 . . . 20 . . . 24 . . . or more carbons in length), and may contain one or more cis or trans double bonds. Examples of such compounds include, but are not limited to distearoylphosphatidylethanolamine (DSPE), dimyristoylphosphatidylethanolamine (DMPE), dipalmitoylphosphatidylethanolamine (DPPE), palmitoyloleoylphosphatidylethanolamine (POPE), egg phosphatidylethanolamine (EPE), and transphosphatidylated phosphatidylethanolamine (t-EPE), which can be generated from various natural or semisynthetic phosphatidylcholines using phospholipase D.

In certain embodiments, the present disclosure employs CpG-reduced or CpG-free expression vectors. An initial sequence that contains CpG dinucleotides (e.g., wild-type version of human G-CSF), may be modified to remove CpG dinucleotides by altering the nucleic acid sequence. FIG. 1 shows a CpG-free version of human G-CSF, with sequences that have been changed to removed CpGs underlined. Such CpG di-nucleotides can be suitably reduced or eliminated not just in a coding sequence, but also in the non-coding sequences, including, e.g., 5′ and 3′ untranslated regions (UTRs), promoter, enhancer, polyA, ITRs, introns, and any other sequences present in the nucleic acid molecule or vector. CpG di-nucleotides may be located within a codon triplet for a selected amino acid. There are five amino acids (serine, proline, threonine, alanine, and arginine) that have one or more codon triplets that contain a CpG di-nucleotide. All five of these amino acids have alternative codons not containing a CpG di-nucleotide that can be changed to, to avoid the CpG but still code for the same amino acid as shown in Table 1 below. Therefore, the CpG di-nucleotides allocated within a codon triplet for a selected amino acid may be changed to a codon triplet for the same amino acid lacking a CpG di-nucleotide.

TABLE 1 DNA Codons DNA Codons Amino Acid Containing CpG Lacking CpG Serine (Ser or S) TCG TCT, TCC, TCA, AGT, AGC Proline (Pro or P) CCG CCT, CCC, CCA, Threonine (Thr or T) ACG ACA, ACT, ACC Alanine (Ala or A) GCG GCT, GCC, GCA Arginine (Arg or R) CGT, CGC, AGA, AGG CGA, CGG

In addition, within the coding region, the interface between triplets should be taken into consideration. For example, if an amino acid triplet ends in a C-nucleotide which is then followed by an amino acid triplet which can start only with a G-nucleotide (e.g., Valine, Glycine, Glutamic Acid, Alanine, Aspartic Acid), then the triplet for the first amino acid triplet is changed to one which does not end in a C-nucleotide. Methods for making CpG free sequences are shown, for example, in U.S. Pat. No. 7,244,609, which is herein incorporated by reference. A commercial service provided by INVIVOGEN is also available to produce CpG free (or reduced) nucleic acid sequences/vectors (plasmids). A commercial service provided by ThermoScientific produces CpG free nucleotide.

Provided below in Table 2 are exemplary promoters and enhancers that may be used in the vectors described herein. Such promoters, and other promoters known in the art, may be used alone or with any of the enhancers, or enhancers, known in the art. Additionally, when multiple proteins or biologically active nucleic acid molecules (e.g., two, three, four, or more) are expressed from the same vector, the same or different promoters may be used in conjunction with the subject nucleic acid sequence.

TABLE 2 Promoter Enhancer CMV human CMV EF1α mouse CMV Ferritin (Heavy/Light) Chain SV40 GRP94 Ubc U1 AP1 UbC hr3 Beta Actin IE2 PGK1 IE6 GRP78 E2-RS CAG MEF2 SV40 C/EBP TRE HNF-1

The present disclosure is not limited by the type of therapeutic proteins that is expressed. In certain embodiments, the therapeutic protein comprises an antibody or antibody fragments (e.g., F(ab) or F(ab′)2). In other embodiments, the therapeutic protein is selected from the group consisting of an anti-inflammatory protein, coagulation protein, anti-cancer protein, anti-sepsis protein, etc. Example of therapeutic proteins that can be expressed with the methods, systems, and compositions described herein include the therapeutic monoclonal antibodies (mAbs), Fabs, F(ab)2s, and scFv's that are shown in Table 3 below.

TABLE 3 Antibody Name Trade name Type Source Target Use 3F8 mab mouse GD2 ganglioside neuroblastoma 8H9 mab mouse B7-H3 neuroblastoma, sarcoma, metastatic brain cancers Abagovomab mab mouse CA-125 (imitation) ovarian cancer Abciximab ReoPro Fab chimeric CD41 (integrin platelet aggregation alpha-IIb) inhibitor Abituzumab mab humanized CD51 cancer Abrilumab mab human integrin α4β7 inflammatory bowel disease, ulcerative colitis, Crohn's disease Actoxumab mab human Clostridium Clostridium difficile difficile colitis Adalimumab Humira mab human TNF-α Rheumatoid arthritis, Crohn's Disease, Plaque Psoriasis, Psoriatic Arthritis, Ankylosing Spondylitis, Juvenile Idiopathic Arthritis, Hemolytic disease of the newborn Adecatumumab mab human EpCAM prostate and breast cancer Aducanumab mab human beta-amyloid Alzheimer's disease Afasevikumab mab human IL17A and IL17F Afelimomab F(ab′)2 mouse TNF-α sepsis Afutuzumab mab humanized CD20 lymphoma Alacizumab pegol F(ab′)2 humanized VEGFR2 cancer ALD518 humanized IL-6 rheumatoid arthritis Alemtuzumab Lemtrada, mab humanized CD52 Multiple sclerosis Campath Alirocumab mab human PCSK9 hypercholesterolemia Altumomab pentetate Hybri-ceaker mab mouse CEA colorectal cancer (diagnosis) Amatuximab mab chimeric mesothelin cancer Anatumomab Fab mouse TAG-72 non-small cell lung mafenatox carcinoma Anetumab ravtansine mab human MSLN cancer Anifrolumab mab human interferon α/β systemic lupus receptor erythematosus Anrukinzumab (= mab humanized IL-13 asthma IMA-638) Apolizumab mab humanized HLA-DR — hematological cancers Arcitumomab CEA-Scan Fab′ mouse CEA gastrointestinal cancers (diagnosis) Ascrinvacumab mab human activin receptor- cancer like kinase 1 Aselizumab mab humanized L-selectin severely injured patients (CD62L) Atezolizumab mab humanized CD274 cancer Atinumab mab human RTN4 Atlizumab (= Actemra, mab humanized IL-6 receptor rheumatoid arthritis tocilizumab) RoActemra Atorolimumab mab human Rhesus factor hemolytic disease of the newborn[citation needed] Avelumab mab human CD274 Bapineuzumab mab humanized beta amyloid Alzheimer's disease Basiliximab Simulect mab chimeric CD25 (a chain of prevention of organ IL-2 receptor) transplant rejections Bavituximab mab chimeric phosphatidylserine cancer, viral infections Bectumomab Lympho Scan Fab' mouse CD22 non-Hodgkin's lymphoma (detection) Begelomab mab mouse DPP4 Belimumab Benlysta, mab human BAFF non-Hodgkin Lympho Stat- lymphoma etc. B Benralizumab mab humanized CD125 asthma Bertilimumab mab human CCL11 (eotaxin-1) severe allergic disorders Besilesomab Scintimun mab mouse CEA-related inflammatory lesions antigen and metastases (detection) Bevacizumab Avastin mab humanized VEGF-A metastatic cancer, retinopathy of prematurity Bezlotoxumab mab human Clostridium Clostridium difficile difficile colitis Biciromab FibriScint Fab′ mouse fibrin II, beta chain thromboembolism (diagnosis) Bimagrumab mab human ACVR2B myostatin inhibitor Bimekizumab mab humanized IL 17A and IL 17F Bivatuzumab mab humanized CD44 v6 squamous cell mertansine carcinoma Bleselumab mab human CD40 Blinatumomab BiTE mouse CD19 pre-B ALL (CD19+) Blontuvetmab Blontress mab veterinary CD20 Blosozumab mab humanized SOST osteoporosis Bococizumab mab humanized neural apoptosis- dyslipidemia regulated proteinase 1 Brazikumab mab human IL23 Crohn's disease Brentuximab vedotin mab chimeric CD30 (TNFRSF8) hematologic cancers Briakinumab mab human IL-12, IL-23 psoriasis, rheumatoid arthritis, inflammatory bowel diseases, multiple sclerosis Brodalumab mab human IL-17 inflammatory diseases Brolucizumab mab humanized VEGFA wet age-related macular degeneration Brontictuzumab mab humanized Notch 1 cancer Burosumab mab human FGF 23 X-linked hypophosphatemia Cabiralizumab mab humanized CSFIR Canakinumab Ilaris mab human IL-1— rheumatoid arthritis Cantuzumab mab humanized mucin CanAg colorectal cancer etc. mertansine Cantuzumab mab humanized MUC1 cancers ravtansine Caplacizumab mab humanized VWF thrombotic thrombocytopenic purpura, thrombosis Capromab pendetide Prostascint mab mouse prostatic prostate cancer carcinoma cells (detection) indications Carlumab mab human MCP-1 oncology/immune indications Carotuximab mab chimeric endoglin Catumaxomab Removab 3funct rat/mouse EpCAM, CD3 ovarian cancer, hybrid malignant ascites, gastric cancer cBR96-doxorubicin mab humanized Lewis-Y antigen cancer immunoconjugate Cedelizumab mab humanized CD4 prevention of organ transplant rejections, treatment of autoimmune diseases Cergutuzumab mab humanized IL2 amunaleukin Certolizumab pegol Cimzia Fab′ humanized TNF-α Crohn's disease Rheumatoid arthritis axial spondyloarthritis psoriasis arthritis Cetuximab Erbitux mab chimeric EGFR metastatic colorectal cancer and head and neck cancer Ch.14.18 mab chimeric GD2 ganglioside neuroblastoma Citatuzumab bogatox Fab humanized EpCAM ovarian cancer and other solid tumors Cixutumumab mab human IGF-1 receptor solid tumors (CD221) Clazakizumab mab humanized Oryctolagus rheumatoid arthritis cumculus Clenoliximab mab chimeric CD4 rheumatoid arthritis Clivatuzumab hPAM4-Cide mab humanized MUC1 pancreatic cancer tetraxetan Codrituzumab mab humanized glypican 3 cancer Coltuximab ravtansine mab chimeric CD19 cancer Conatumumab mab human TRAIL-R2 cancer Concizumab mab humanized TFPI bleeding CR6261 mab human Influenza A infectious hemagglutinin disease/influenza A Crenezumab mab humanized 1-40-β-amyloid Alzheimer's disease Crotedumab mab human GCGR diabetes Dacetuzumab mab humanized CD40 hematologic cancers Daclizumab Zenapax mab humanized CD25 (a chain of prevention of organ IL-2 receptor) transplant rejections Dalotuzumab mab humanized IGF-1 receptor cancer etc. (CD221) Dapirolizumab pegol mab humanized CD154 (CD40L) Daratumumab mab human CD38 (cyclic ADP cancer ribose hydrolase) Dectrekumab mab human IL-13 Demcizumab mab humanized DLL4 cancer Denintuzumab mab humanized CD19 cancer mafodotin Denosumab Prolia mab human RANKL osteoporosis, bone metastases etc. Depatuxizumab mab chimeric/ EGFR cancer mafodotin humanized Derlotuximab biotin mab chimeric histone complex recurrent glioblastoma multiforme Detumomab mab mouse B-lymphoma cell lymphoma Dinutuximab mab chimeric GD2 ganglioside neuroblastoma Diridavumab mab human hemagglutinin influenza A Domagrozumab mab humanized GDF-8 Duchenne muscular dystrophy Dorlimomab aritox F(ab′)2 mouse Drozitumab mab human DR5 cancer etc. Duligotumab mab human ERBB3 (HER3) testicular cancer Dupilumab mab human IL4 atopic diseases Durvalumab mab human CD274 cancer Dusigitumab mab human ILGF2 cancer Ecromeximab mab chimeric GD3 ganglioside malignant melanoma Eculizumab Soliris mab humanized C5 paroxysmal nocturnal hemoglobinuria, atypical HUS Edobacomab mab mouse endotoxin sepsis caused by Gram- negative bacteria Edrecolomab Panorex mab mouse EpCAM colorectal carcinoma Efalizumab Raptiva mab humanized LFA-1 (CD11a) psoriasis (blocks T-cell migration) Efungumab Mycograb scFv human Hsp90 invasive Candida infection Eldelumab mab human interferon gamma- Crohn's disease, induced protein ulcerative colitis Elgemtumab mab human ERBB3 (HER3) cancer Elotuzumab mab humanized SLAMF7 multiple myeloma Elsilimomab mab mouse IL-6 Emactuzumab mab humanized CSF1R cancer Emibetuzumab mab humanized HHGFR cancer Emicizumab mab humanized activated F9, F10 haemophilia A Enavatuzumab mab humanized TWEAK receptor cancer etc. Enfortumab vedotin mab human AGS-22M6 cancer expressing Nectin-4 Enlimomab pegol mab mouse ICAM-1 (CD54) Enoblituzumab mab humanized CD276 cancer Enokizumab mab humanized IL9 asthma Enoticumab mab human DLL4 Ensituximab mab chimeric 5AC cancer Epitumomab cituxetan mab mouse episialin Epratuzumab mab humanized CD22 cancer, SLE Erenumab mab human CGRP migraine Erlizumab F(ab′)2 humanized ITGB2 (CD18) heart attack, stroke, traumatic shock Ertumaxomab Rexomun 3funct rat/mouse HER2/neu, CD3 breast cancer etc. hybrid Etaracizumab Abegrin mab humanized integrin αvβ3 melanoma, prostate cancer, ovarian cancer etc. Etrolizumab mab humanized integrin α7 β7 inflammatory bowel disease Evinacumab mab human angiopoietin 3 dyslipidemia Evolocumab mab human PCSK9 hypercholesterolemia Exbivirumab mab human hepatitis B surface hepatitis B antigen Fanolesomab NeutroSpec mab mouse CD15 appendicitis (diagnosis) Faralimomab mab mouse interferon receptor Farletuzumab mab humanized folate receptor 1 ovarian cancer Fasinumab mab human HNGF acute sciatic pain FBTA05 Lymphomun 3funct rat/mouse CD20 chronic lymphocytic hybrid leukaemia Felvizumab mab humanized respiratory respiratory syncytial syncytial virus virus infection Fezakinumab mab human IL-22 rheumatoid arthritis, psoriasis Fibatuzumab mab humanized ephrin receptor A3 Ficlatuzumab mab humanized HGF cancer etc. Figitumumab mab human IGF-1 receptor adrenocortical (CD221) carcinoma, non-small cell lung carcinoma etc. Firivumab mab human influenza A virus hemagglutinin Flanvotumab mab human TYRP1(glycoprote melanoma in 75) Fletikumab mab human IL 20 rheumatoid arthritis Fontolizumab HuZAF mab humanized IFN-γ Crohn's disease etc. Foralumab mab human CD3 epsilon Foravirumab mab human rabies virus rabies (prophylaxis) glycoprotein Fresolimumab mab human TGF-β idiopathic pulmonary fibrosis, focal segmental glomerulosclerosis, cancer Fulranumab mab human NGF pain Futuximab mab chimeric EGFR cancer Galcanezumab mab humanized calcitonin migraine Galiximab mab chimeric CD80 B-cell lymphoma Ganitumab mab human IGF-1 receptor cancer (CD221) Gantenerumab mab human beta amyloid Alzheimer's disease Gavilimomab mab mouse CD147 (basigin) graft versus host disease Gemtuzumab Mylotarg mab humanized CD33 acute myelogenous ozogamicin leukemia Gevokizumab mab humanized IL-1β diabetes etc. Girentuximab Rencarex mab chimeric carbonic anhydrase clear cell renal cell 9 (CA-IX) carcinoma[84] Glembatumumab mab human GPNMB melanoma, breast vedotin cancer Golimumab Simponi mab human TNF-α rheumatoid arthritis, psoriatic arthritis, ankylosing spondylitis Gomiliximab mab chimeric CD23 (IgE allergic asthma receptor) Guselkumab mab human IL23 psoriasis Ibalizumab mab humanized CD4 HIV infection Ibritumomab tiuxetan Zevalin mab mouse CD20 non-Hodgkin's lymphoma Icrucumab mab human VEGFR-1 cancer etc. Idarucizumab mab humanized dabigatran reversal of anticoagulant effects of dabigatran Igovomab Indimacis-125 F(ab′)2 mouse CA-125 ovarian cancer (diagnosis) IMAB362 mab human CLDN18.2 gastrointestinal adenocarcinomas and pancreatic tumor Imalumab mab human MIF cancer Imciromab Myoscint mab mouse cardiac myosin cardiac imaging Imgatuzumab mab humanized EGFR cancer Inclacumab mab human selectin P cardiovascular disease Indatuximab mab chimeric SDC1 cancer ravtansine Indusatumab vedotin mab human GUCY2C cancer Inebilizumab mab humanized CD19 cancer, systemic sclerosis, multiple sclerosis Infliximab Remicade mab chimeric TNF-α rheumatoid arthritis, ankylosing spondylitis, psoriatic arthritis, psoriasis, Crohn's disease, ulcerative colitis Inolimomab mab mouse CD25 (a chain of graft versus host disease IL-2 receptor) Inotuzumab mab humanized CD22 ALL ozogamicin Intetumumab mab human CD51 solid tumors (prostate cancer, melanoma) Ipilimumab Yervoy mab human CD152 melanoma Iratumumab mab human CD30 (TNFRSF8) Hodgkin's lymphoma Isatuximab mab chimeric CD38 cancer Itolizumab mab humanized CD6 Ixekizumab mab humanized IL 17A autoimmune diseases Keliximab mab chimeric CD4 chronic asthma Labetuzumab CEA-Cide mab humanized CEA colorectal cancer Lampalizumab mab humanized CFD geographic atrophy secondary to age-related macular degeneration Lanadelumab mab human kallikrein angioedema Landogrozumab mab humanized GDF-8 muscle wasting disorders Laprituximab mab chimeric EGFR emtansine Lebrikizumab mab humanized IL-13 asthma Lemalesomab mab mouse NCA-90 diagnostic agent (granulocyte antigen) Lendalizumab mab humanized C5 Lenzilumab mab human CSF2 Lerdelimumab mab human TGF beta 2 reduction of scarring after glaucoma surgery Lexatumumab mab human TRAIL-R2 cancer Libivirumab mab human hepatitis B surface hepatitis B antigen Lifastuzumab vedotin mab humanized phosphate-sodium cancer co-transporter Ligelizumab mab humanized IGHE severe asthma and chronic spontaneous urticaria Lilotomab satetraxetan mab mouse CD37 cancer Lintuzumab mab humanized CD33 cancer Lirilumab mab human KIR2D solid and hematological cancers Lodelcizumab mab humanized PCSK9 hypercholesterolemia Lokivetmab mab veterinary Canis lupus fandiliaris IL31 Lorvotuzumab mab humanized CD56 cancer mertansine Lucatumumab mab human CD40 multiple myeloma, non- Hodgkin's lymphoma, Hodgkin's lymphoma Lulizumab pegol mab humanized CD28 autoimmune diseases Lumiliximab mab chimeric CD23 (IgE chronic lymphocytic receptor) leukemia Lumretuzumab mab humanized ERBB3 (HER3) cancer MABp1 Xilonix mab human IL1A colorectal cancer Mapatumumab mab human TRAIL-R1 cancer Margetuximab mab humanized ch4D5 cancer Maslimomab mouse T-cell receptor Matuzumab mab humanized EGFR colorectal, lung and stomach cancer Mavrilimumab mab human GMCSF receptor rheumatoid arthritis α-chain Mepolizumab Bosatria mab humanized IL-5 asthma and white blood cell diseases Metelimumab mab human TGF beta 1 systemic scleroderma Milatuzumab mab humanized CD74 multiple myeloma and other hematological malignancies Minretumomab mab mouse TAG-72 tumor detection (and therapy—) Mirvetuximab mab chimeric folate receptor cancer soravtansine alpha Mitumomab mab mouse GD3 ganglioside small cell lung carcinoma Mogamulizumab mab humanized CCR4 cancer Monalizumab mab humanized KLRC1 Morolimumab mab human Rhesus factor Motavizumab Numax mab humanized respiratory respiratory syncytial syncytial virus virus (prevention) Moxetumomab mab mouse CD22 cancer pasudotox Muromonab-CD3 Orthoclone mab mouse CD3 prevention of organ OKT3 transplant rejections Nacolomab tafenatox Fab mouse C242 antigen colorectal cancer Namilumab mab human CSF2 Naptumomab Fab mouse 5T4 non-small cell lung estafenatox carcinoma, renal cell carcinoma Naratuximab mab chimeric CD37 emtansine Narnatumab mab human RON cancer Natalizumab Tysabri mab humanized integrin α4 multiple sclerosis, Crohn's disease Navicixizumab mab chimeric/ DLL4 humanized Navivumab mab human influenza A virus hemagglutinin HA Nebacumab mab human endotoxin sepsis Necitumumab mab human EGFR non-small cell lung carcinoma Nemolizumab mab humanized IL31RA eczema[106] Nerelimomab mab mouse TNF-α Nesvacumab mab human angiopoietin 2 cancer Nimotuzumab Theracim, mab humanized EGFR squamous cell Theraloc carcinoma, head and neck cancer, nasopharyngeal cancer, glioma Nivolumab Opdivo mab human PD-1 cancer Nofetumomab Verluma Fab mouse cancer (diagnosis) merpentan Obiltoxaximab mab chimeric Bacillus anthracis Bacillus anthracis anthrax spores Obinutuzumab Gazyva mab humanized CD20 Chronic lymphatic leukemia Ocaratuzumab mab humanized CD20 cancer Ocrelizumab mab humanized CD20 rheumatoid arthritis, lupus erythematosus etc. Odulimomab mab mouse LFA-1 (CD11a) prevention of organ transplant rejections, immunological diseases Ofatumumab Arzerra mab human CD20 chronic lymphocytic leukemia etc. Olaratumab mab human PDGF-R α cancer Olokizumab mab humanized IL6 Omalizumab Xolair mab humanized IgE Fc region allergic asthma Onartuzumab mab humanized human scatter cancer factor receptor kinase Ontuxizumab mab chimeric/ TEM1 cancer humanized Opicinumab mab human LINGO-1 multiple sclerosis Oportuzumab monatox scFv humanized EpCAM cancer Oregovomab OvaRex mab mouse CA-125 ovarian cancer Orticumab mab human oxLDL Otelixizumab mab chimeric/ CD3 diabetes mellitus type 1 humanized Otlertuzumab mab humanized CD37 cancer Oxelumab mab human OX-40 asthma Ozanezumab mab humanized NOGO-A ALS and multiple sclerosis Ozoralizumab mab humanized TNF-α inflammation Pagibaximab mab chimeric lipoteichoic acid sepsis (Staphylococcus) Palivizumab Synagis, mab humanized F protein of respiratory syncytial Abbosynagis respiratory virus (prevention) syncytial virus Pamrevlumab mab human CTGF Panitumumab Vectibix mab human EGFR colorectal cancer Pankomab mab humanized tumor specific ovarian cancer glycosylation of MUC1 Panobacumab mab human Pseudomonas Pseudomonas aeruginosa aeruginosa infection Parsatuzumab mab human EGFL7 cancer Pascolizumab mab humanized IL-4 asthma Pasotuxizumab mab chimeric/ folate hydrolase cancer humanized Pateclizumab mab humanized LTA TNF Patritumab mab human ERBB3 (HER3) cancer Pembrolizumab mab humanized PDCD1 melanoma and other cancers Pemtumomab Theragyn mouse MUC1 cancer Perakizumab mab humanized IL 17A arthritis Pertuzumab Omnitarg mab humanized HER2/neu cancer Pexelizumab scFv humanized C5 reduction of side effects of cardiac surgery Pidilizumab mab humanized PD-1 cancer and infectious diseases Pinatuzumab vedotin mab humanized CD22 cancer Pintumomab mab mouse adenocarcinoma adenocarcinoma antigen (imaging) Placulumab mab human human TNF pain and inflammatory diseases Plozalizumab mab humamized CCR2 diabetic nephropathy and arteriovenous graft patency Pogalizumab mab humanized TNFR superfamily member 4 Polatuzumab vedotin mab humanized CD79B cancer Ponezumab mab humanized human beta- Alzheimer's disease amyloid Prezalizumab mab humanized ICOSL Priliximab mab chimeric CD4 Crohn's disease, multiple sclerosis Pritoxaximab mab chimeric E. coli shiga toxin type-1 Pritumumab mab human vimentin brain cancer PRO 140 humanized CCR5 HIV infection Quilizumab mab humanized IGHE asthma Racotumomab mab mouse N- cancer glycolylneuraminic acid Radretumab mab human fibronectin extra cancer domain-B Rafivirumab mab human rabies virus rabies (prophylaxis) glycoprotein Ralpancizumab mab humanized neural apoptosis- dyslipidemia regulated proteinase 1 Ramucirumab Cyramza mab human VEGFR2 solid tumors Ranibizumab Lucentis Fab humanized VEGF-A macular degeneration (wet form) Raxibacumab mab human anthrax toxin, anthrax (prophylaxis protective antigen and treatment) Refanezumab mab humanized myelin-associated recovery of motor glycoprotein function after stroke Regavirumab mab human cytomegalovirus cytomegalovirus glycoprotein B infection Reslizumab mab humanized IL-5 inflammations of the airways, skin and gastrointestinal tract Rilotumumab mab human HGF solid tumors Rinucumab mab human platelet-derived neovascular age-related growth factor macular degeneration receptor beta Risankizumab mab humanized IL23A Rituximab MabThera, mab chimeric CD20 lymphomas, leukemias, Rituxan some autoimmune disorders Rivabazumab pegol mab humanized Pseudomonas aeruginosa type III secretion system Robatumumab mab human IGF-1 receptor cancer (CD221) Roledumab mab human RHD Romosozumab mab humanized sclerostin osteoporosis Rontalizumab mab humanized IFN-α systemic lupus erythematosus Rovalpituzumab mab humanized DLL3 tesirine Rovelizumab LeukArrest mab humanized CD11, CD18 haemorrhagic shock etc. Ruplizumab Antova mab humanized CD154 (CD40L) rheumatic diseases Sacituzumab govitecan mab humanized tumor-associated cancer calcium signal transducer 2 Samalizumab mab humanized CD200 cancer Sapelizumab mab humanized IL6R Sarilumab mab human IL6 rheumatoid arthritis, ankylosing spondylitis Satumomab pendetide mab mouse TAG-72 cancer (diagnosis) Secukinumab mab human IL 17A uveitis, rheumatoid arthritis psoriasis Seribantumab mab human ERBB3 (HER3) cancer Setoxaximab mab chimeric E. coli shiga toxin type-2 Sevirumab human cytomegalovirus cytomegalovirus infection SGN-CD19A mab humanized CD19 acute lymphoblastic leukemia and B-cell non-Hodgkin lymphoma SGN-CD33A mab humanized CD33 Acute myeloid leukemia Sibrotuzumab mab humanized FAP cancer Sifalimumab mab humanized IFN-α SLE, dermatomyositis, polymyositis Siltuximab mab chimeric IL-6 cancer Simtuzumab mab humanized LOXL2 fibrosis Siplizumab mab humanized CD2 psoriasis, graft-versus- host disease (prevention) Sirukumab mab human IL-6 rheumatoid arthritis Sofituzumab vedotin mab humanized CA-125 ovarian cancer Solanezumab mab humanized beta amyloid Alzheimer's disease Solitomab BiTE mouse EpCAM Sonepcizumab humanized sphingosine-1- choroidal and retinal phosphate neovascularization Sontuzumab mab humanized episialin Stamulumab mab human myostatin muscular dystrophy Sulesomab Leuko Scan Fab′ mouse NCA-90 osteomyelitis (imaging) (granulocyte antigen) Suvizumab mab humanized HIV-1 viral infections Tabalumab mab human BAFF B-cell cancers Tacatuzumab AFP-Cide mab humanized alpha-fetoprotein cancer tetraxetan Tadocizumab Fab humanized integrin αIIbβ3 percutaneous coronary intervention Talizumab mab humanized IgE allergic reaction Tamtuvetmab Tactress mab veterinary CD52 Tanezumab mab humanized NGF pain Taplitumomab paptox mab mouse CD19 cancer[citation needed] Tarextumab mab human Notch receptor cancer Tefibazumab Aurexis mab humanized clumping factor A Staphylococcus aureus infection Telimomab aritox Fab mouse Tenatumomab mab mouse tenascin C cancer Teneliximab mab chimeric CD40 autoimmune diseases and prevention of organ transplant rejection Teplizumab mab humanized CD3 diabetes mellitus type 1 Teprotumumab mab human IGF-1 receptor hematologic tumors (CD221) Tesidolumab mab human C5 Tetulomab mab humanized CD37 cancer[141] Tezepelumab mab human TSLP asthma, atopic dermatitis TGN1412 humanized CD28 chronic lymphocytic leukemia, rheumatoid arthritis Ticilimumab (= mab human CTLA-4 cancer tremelimumab) Tigatuzumab mab humanized TRAIL-R2 cancer Tildrakizumab mab humanized IL23 immunologically mediated inflammatory disorders Timolumab mab human AOC3 Tisotumab vedotin mab human coagulation factor III TNX-650 humanized IL-13 Hodgkin's lymphoma Tocilizumab (= Actemra, mab humanized IL-6 receptor rheumatoid arthritis atlizumab) RoActemra Toralizumab mab humanized CD154 (CD40L) rheumatoid arthritis, lupus nephritis etc. Tosatoxumab mab human Staphylococcus aureus Tositumomab Bexxar mouse CD20 follicular lymphoma Tovetumab mab human CD140a cancer Tralokinumab mab human IL-13 asthma etc. Trastuzumab Herceptin mab humanized HER2/neu breast cancer Trastuzumab Kadcyla mab humanized HER2/neu breast cancer emtansine TRBS07 Ektomab 3funct GD2 ganglioside melanoma Tregalizumab mab humanized CD4 Tremelimumab mab human CTLA-4 cancer Trevogrumab mab human growth muscle atrophy due to differentiation orthopedic disuse and factor 8 sarcopenia Tucotuzumab mab humanized EpCAM cancer celmoleukin Tuvirumab human hepatitis B virus chronic hepatitis B Ublituximab mab chimeric MS4A1 cancer Ulocuplumab mab human CXCR4 (CD184) hematologic malignancies Urelumab mab human 4-1BB (CD137) cancer etc. Urtoxazumab mab humanized Escherichia coli diarrhoea caused by E. coli Ustekinumab Stelara mab human IL-12, IL-23 multiple sclerosis, psoriasis, psoriatic arthritis Utomilumab mab human 4-1BB (CD137) cancer Vadastuximab talirine mab chimeric CD33 Vandortuzumab mab humanized STEAP1 cancer vedotin Vantictumab mab human Frizzled receptor cancer Vanucizumab mab humanized angiopoietin 2 cancer Vapaliximab mab chimeric AOC3 (VAP-1) Varlilumab mab human CD27 solid tumors and hematologic malignancies Vatelizumab mab humanized ITGA2 (CD49b) Vedolizumab Entyvio mab humanized integrin α4β7 Crohn's disease, ulcerative colitis Veltuzumab mab humanized CD20 non-Hodgkin's lymphoma Vepalimomab mab mouse AOC3 (VAP-1) inflammation Vesencumab mab human NRP1 solid malignancies Visilizumab Nuvion mab humanized CD3 Crohn's disease, ulcerative colitis Vobarilizumab mab humanized IL6R inflammatory autoimmune diseases Volociximab mab chimeric integrin α5β1 solid tumors Vorsetuzumab mab humanized CD70 cancer mafodotin Votumumab HumaSPECT mab human tumor antigen colorectal tumors CTAA16.88 Xentuzumab mab IGF1, IGF2 Zalutumumab HuMax-EGFr mab human EGFR squamous cell carcinoma of the head and neck Zanolimumab HuMax-CD4 mab human CD4 rheumatoid arthritis, psoriasis, T-cell lymphoma Zatuximab mab chimeric HER1 cancer Ziralimumab mab human CD147 (basigin) Zolimomab aritox mab mouse CD5 systemic lupus erythematosus, graft- versus-host disease

Further examples of therapeutic proteins that can be expressed with the methods, systems, and compositions described herein include the therapeutic monoclonal antibodies (mAbs), Fabs, F(ab)2s, and scFv's, such as broadly neutralizing anti-HIV monoclonals, including antibody 10-1074 (Caskey et al., Nat Med. 2017 February; 23(2):185-191, Epub 2017 Jan. 16, herein incorporated by reference in its entirety); HIV-1 antibody 3BNC117 (Scheid, et al., Nature. 2016 Jul. 28; 535(7613):556-60, herein incorporated by reference in its entirety); and VRC01 (see, e.g., Bar et al., N Engl J Med. 2016 Nov. 24; 375(21):2037-2050, herein incorporated by reference in its entirety).

In some embodiments, compositions and systems herein are provided and/or administered in doses selected to elicit a therapeutic and/or prophylactic effect in an appropriate subject (e.g., mouse, human, etc.). In some embodiments, a therapeutic dose is provided. In some embodiments, a prophylactic dose is provided. Dosing and administration regimes are tailored by the clinician, or others skilled in the pharmacological arts, based upon well-known pharmacological and therapeutic/prophylactic considerations including, but not limited to, the desired level of pharmacologic effect, the practical level of pharmacologic effect obtainable, toxicity. Generally, it is advisable to follow well-known pharmacological principles for administrating pharmaceutical agents (e.g., it is generally advisable to not change dosages by more than 50% at time and no more than every 3-4 agent half-lives). For compositions that have relatively little or no dose-related toxicity considerations, and where maximum efficacy is desired, doses in excess of the average required dose are not uncommon. This approach to dosing is commonly referred to as the “maximal dose” strategy. In certain embodiments, a dose (e.g., therapeutic of prophylactic) is about 0.01 mg/kg to about 200 mg/kg (e.g., 0.01 mg/kg, 0.02 mg/kg, 0.05 mg/kg, 0.1 mg/kg, 0.2 mg/kg, 0.5 mg/kg, 1.0 mg/kg, 2.0 mg/kg, 5.0 mg/kg, 10 mg/kg, 20 mg/kg, 50 mg/kg, 100 mg/kg, 200 mg/kg, or any ranges therebetween (e.g., 5.0 mg/kg to 100 mg/kg)). In some embodiments, a subject is between 0.1 kg (e.g., mouse) and 150 kg (e.g., human), for example, 0.1 kg, 0.2 kg, kg, 1.0 kg, 2.0 kg, 5.0 kg, 10 kg, 20 kg, 50 kg, 100 kg, 200 kg, or any ranges therebetween (e.g., 40-125 kg). In some embodiments, a dose comprises between 0.001 mg and 40,000 mg (e.g., 0.001 mg, 0.002 mg, 0.005 mg, 0.01 mg, 0.02 mg, 0.05 mg, 0.1 kg, 0.2 mg, 0.5 mg, 1.0 mg, 2.0 mg, 5.0 mg, 10 mg, 20 mg, 50 mg, 100 mg, 200 mg, 500 mg, 1,000 mg, 2,000 mg, 5,000 mg, 10,000 mg, 20,000 mg, 40,000 mg, or ragnes therebetween.

EXAMPLES

In all of the Examples below, all of the expression vectors are CpG free except for Genscript px458-relAl (SEQ ID NO:8) and Genscript px458-relA4 (SEQ ID NO:9), both of which are CpG-laden sequences which are commercially available (see, www followed by “genscript.com/CRISPR-gRNA-constructs.html.”).

Example 1 Long-Term Therapeutic Rituximab Expression

This Example describes experiments conducted that demonstrate long-term expression of monoclonal antibody Rituximab at therapeutic serum levels following a single injection of either a dual cassette or single cassette plasmid vector encoding Rituximab.

First Example

In a first example, three mice were injected per group. Each mouse received a single IV injection of 1050 nmoles of DOTAP cationic liposomes containing 2.5% dexamethasone palmitate (DP) incorporated into the liposome bilayer. This was followed two minutes later by a single IV injection of 75 ug of one of two different plasmid DNAs encoding anti-CD20 (Rituximab). Both groups were treated two hours prior to IV injection with an IP injection of dexamethasone. Plasmid 715.1 2a (P2A), shown in FIG. 5 (SEQ ID NO:3) encodes the anti-CD20 mAb heavy and light chain cDNAs separated by a 2A self-cleaving peptide. Plasmid 718.1, shown in FIG. 6 (SEQ ID NO:4), is dual expression cassette plasmid vector that encode the anti-CD20 mAb heavy and light chain cDNAs respectively. Serum levels of anti-CD20 were determined by ELISA 24 hours following injection and in 7-day intervals thereafter. The ELISA kit was purchased from Eagle Biosciences.

The results are shown in FIG. 2A. Injection of each plasmid produced serum anti-CD20 mAb protein levels approaching or above 1 at ug/ml levels, 24 hrs post injection. Serum anti-CD20 mAb protein levels are sustained within this range in both groups for at least the next 178 days. Serum anti-CD20 mAb protein levels were undetectable in mice receiving the same protocol except that the DNA vector encoded human G-CSF cDNA. These data demonstrate that dual cassette, as well as single cassette plasmid DNA vectors encoding anti-CD20 mAb can produce prolonged, sustained serum anti-CD20 mAb protein levels after a single IV injection.

Second Example

In a second example, Raji cells (5×10 4 cells/well) were plated in 96 well plates using RPMI+10% FBS medium. Next day cells were incubated with Rituximab (0.5, 1, 10 ug/ml) or mouse serum samples (20 ul/well, duplicate samples) for 1 h at room temperature. Twenty microliters of pooled normal human plasma (Innovative Research) was then added to all wells (except the Rituximab control condition) and the plates incubated for another 12 h at 37 C. Cell viability was measured using the PROMEGA Cell titer Glo reagent according to the manufacturer's instructions. Results are shown in FIG. 2B, where values are shown as percentage change from the control conditions (no treatment). Rituximab concentrations for each mouse sample rested are shown in FIG. 2B. Ctrl 1-2 samples refer to mouse serum from mice injected with a control plasmid (encoding for G-CSF). Bars represent standard deviation for duplicate samples.

Serum from mice sequentially injected with cationic liposomes, then a plasmid DNA vector encoding either anti-CD20 mAb 148 days earlier or human G-CSF was analyzed first by ELISA for concentration determination of anti-CD20. The numbers in red font placed above the bars represent the concentration of Rituxumab for the corresponding serum samples (ng/ml). Using a cell lysis assay, sera isolated from anti-CD20 DNA vector-injected mice 148 days earlier lysed CD-20+human Raji cells at a level comparable to Raji cells treated with a high concentration of recombinant Rituximab protein (Invivogen). These data (in FIG. 2B) show that anti-CD20 DNA vector-injected mice produce fully bioactive Rituximab mAb protein for at least 176 days after a single DNA vector injection.

Third Example

In a third example, three mice were injected per group. Each mouse received a single IV injection of 1000 nmoles of DOTAP cationic liposomes containing 2.5% dexamethasone palmitate (DP) incorporated into the liposome bilayer. This was followed two minutes later by a single IV injection of 75 ug of one of two different plasmid DNAs encoding anti-CD20 (Rituximab). Both groups were treated two hours prior to IV injection with an IP injection of 40 mg/kg dexamethasone. Plasmid 902.8 (P2A), shown in FIG. 7 (SEQ ID NO:5) encodes the anti-CD20 mAb heavy and light chain cDNAs separated by a 2A self-cleaving peptide. Plasmid 718.1, shown in FIG. 6 (SEQ ID NO:4), is a dual expression cassette plasmid vector that encode the anti-CD20 mAb heavy and light chain cDNAs respectively. Serum levels of anti-CD20 were determined by ELISA 24 hours following injection and in 7-day intervals thereafter. The ELISA kit was purchased from Eagle Biosciences. The results in FIG. 3 shows that injection of each plasmid produced serum anti-CD20 mAb protein levels approaching or above 1 at ug/ml levels, 24 hrs post injection. Serum anti-CD20 mAb protein levels were within this range for both groups at 57 days after injection.

Fourth Example

In a fourth example, three mice were injected per group. Each mouse received a single IV injection of 1000 nmoles of DOTAP cationic liposomes and 1000 nmoles of DMPC neutral liposomes, each containing 2.5% dexamethasone palmitate (DP) incorporated into the liposome bilayer. This was followed two minutes later by a single IV injection of 75 ug of plasmid DNA encoding anti-CD20 (Rituximab). Both groups were treated two hours prior to IV injection with an IP injection of 40 mg/kg dexamethasone. Plasmid p718.1 is a dual expression cassette plasmid vector that encodes the anti-CD20 mAb heavy and light chain cDNAs respectively. Plasmid p113.2, shown in FIG. 8 (SEQ ID NO:6), is identical, but includes a single super enhancer upstream of the second coding cassette. Serum levels of anti-CD20 were determined by ELISA 24 hours following injection. The ELISA kit was provided by Eagle Biosciences. The results are shown in FIG. 4. Both groups express anti-CD20 at 24 hrs post sequential injection. The addition of a single super enhancer element increases production of serum anti-CD20 mAb protein in mice at this time point.

Example 2 Expression of Biologically Active Nucleic Acid

This Example describes experiments conducted that demonstrate IV, sequential injection of cationic liposomes then plasmid DNA vectors encoding CPISPR/Cas9, shRNA, ribozyme or anti-sense sequences specifically targeting mouse NFkB-p65 each suppresses p65 expression in mice.

First Example

In a first example, three or four mice were injected per group. Each mouse received a single IV injection of 1000 nmoles of DOTAP cationic liposomes containing 2.5% dexamethasone palmitate (DP) incorporated into the liposome bilayer. This was followed two minutes later by a single IV injection of 75 ug of plasmid DNA encoding the indicated CRISPR- or ribozyme-based plasmids to suppress expression of endogenous mouse NFkB-p65. The plasmids used are as follows: ribozyme (FIG. 13, SEQ ID NO:7), CRISPR1 (FIG. 14, SEQ ID NO:8), CRISPR2 (FIG. 15, SEQ ID NO:9), and CRISPR (FIG. 16, SEQ ID NO:10). The control group received an CRISPR/Cas9 plasmid identical to the anti-NFkB-p65 CRISPR plasmids except the 20 bp targeting sequence targeted mouse PECAM instead. All groups were treated two hours prior to IV injection with an IP injection of 40 mg/kg dexamethasone.

Tissue preparation and anti-mouse-p65 ELISA methods were as follows. Lung lysates were generated 24 hours after injection (Anti-p65 Ribozyme) and 8 days after injection (Anti-p65 CRISPR1/2) by dissection into 500 uL of prepared 1× Triton lysis buffer on ice. Samples include both lungs per animal. Each sample was homogenized (Polytron PT 2100) for 30 seconds, pulse sonicated (Misonix XL2000 Microson Ultrasonic Cell Disruptor XL 2000), and centrifuged for 10 minutes at 4 C, and the lysate was aspirated from the tissue pellet. Protein concentration from each lysate was then determined using a BCA total protein assay purchased from Thermo Fisher. Protein normalized lysate was added to a 96 well plate ELISA from Cell Signaling Technologies (PathScan Total NF-κB p65 Sandwich ELISA Kit) in duplicate as per the manufacture's instructions. The plate was then analyzed in a (Molecular Devices Spectramax M5) plate reader. After recording absorbance from the plate, a standard curve generated using murine B16 melanoma cell supernatant was fit by 4PL analysis. Error bars represent the standard error of the mean.

The results of this example are shown in FIG. 9. These data demonstrate that anti-mouse NFkB-p65 CRISPR/Cas9—as well as ribozyme, plasmid-based targeting vectors reduce the expression of endogenous mouse p65 at 8 days and 1 day respectively following their systemic injection.

Second Example

The methods for this example are the same as above. The results are shown in FIG. 10. These data demonstrate that an anti-mouse NFkB-p65 CRISPR/Cas9, plasmid-based targeting vector reduces the expression of endogenous mouse p65, 13 days following its systemic injection in mice. Additionally, NFkB-p65 immunohistochemistry methods were performed on tissue sections from these mice.

Paraffin embedded sections of mouse lung were batch (sections from controls and treated animals) stained on a Leica Bond autostainer with a primary rabbit Mab to C-terminus of p65 (Anti-NF-kB p65 antibody [E379] (ab32536)—ABCAM). Peroxidase labeled secondary. The IHC stained slides were scanned in brightfield at 20× magnification using the Hamamatsu NanoZoomer Digital Pathology System. The digital images were then imported into Visiopharm software for quantitative analysis.

Using the Visiopharm Image Analysis module, five scattered, representative regions of lung parenchyma (ROIs) of each sample were randomly selected by the HIC image analysis technician and manually delineated for further quantitative analysis. The software converted the initial digital image into grayscale values using three features, RGB-B with a mean and polynomial smoothing filter, Contrast Red-Blue, and HDAB-DAB with minimum H&E-Eosin filter. Visiopharm software was then trained to label positive brown staining, hematoxylin counterstain, and blank space using a Bayesian classification scheme. All ROIs were processed in batch mode using this configuration to generate the desired outputs.

TABLE 2 Classification Scheme: Bayesian Area NfKb Area Tissue Total Area Measurement (μm2) (μm2) (μm2) Ratio NfKb Animal 143-39 123566.69 483901.69 607468.38 0.2034125 Animal Ringers 287497.75 467149.91 754647.69 0.3809695 Cntrl

The ratio of NFkB was determined by dividing the Area NFkB by the Total Area of lung parenchyma. The difference between the control (Ringers) and the treated lung (143-39) ratios shows an approximately 53% reduction in NfKb staining. This finding is consistent with visual observations of stained sections. The results are shown in FIG. 41. FIG. 41A shows ringers treated control, and FIG. 41B shows the CRISPR/Cas9 anti-NFkB p65 treated mice tissue. These results demonstrate that one sequential IV injection of an CRISPR/Cas9 anti-NFkB p65 plasmid DNA vector reduced p65 protein levels by more than 50% throughout the lungs of mice injected 13 days before with the anti-NFkB p65 versus control DNA vector.

Third Example

The methods for this example are the same as above. The results are shown in FIG. 11. These data demonstrate that anti-mouse NFkB-p65 CRISPR/Cas9—as well as anti-sense, plasmid-based targeting vectors (FIG. 19, SEQ ID NO:11) reduce the expression of endogenous mouse p65 at 13 days and 1 day respectively following their systemic injection.

Fourth Example

The methods for this example are the same as above. The results are shown in FIG. 12. These data demonstrate that the anti-mouse NFkB-p65 shRNA vector p65 shB (FIG. 20, SEQ ID NO:12) and plasmid p65 shA2 (SEQ ID NO:55; FIG. 76), plasmid-based targeting vector reduces the expression of endogenous mouse p65 at 1 day following its systemic injection. Control plasmid PECAM sh control is SEQ ID NO:56, FIG. 77.

Example 3 Long-Term G-CSF Expression

This Example describes experiments conducted that demonstrate a single IV, sequential injection of cationic liposomes followed up by a plasmid DNA vector encoding the human G-CSF gene produces supra-therapeutic human G-CSF serum protein levels (FIG. 17A) and elevated absolute neutrophil counts (ANC) (FIG. 17B) for at least the next 582 days in mice. Thus, a single IV sequential liposome DNA injection can produce therapeutic serum levels of the DNA vector-encoded protein for more than one and a half years in fully immune-competent mice. The two HG-CSF plasmids employed were 011215 #7 (SEQ ID NO:45; FIG. 66), and 011315 #2 (SEQ ID NO:46; FIG. 67); and the negative control was plasmid 122014 #235 (SEQ ID NO:47; FIG. 68).

In another example, plasmid encoding HG-CSF was injected into rats. Rats No. 10 and No. 12 were given one sequential injection each, while rat 14 was re-injected twice, on days 7 and 21 after initial injection. Rat No. 10 was injected IV with 3000 nmol DOTAP SUV followed by 300 ug MARless plasmid DNA encoding HG-CSF. Rat No. 12 was injected with 3 mg Dexamethasone (IP) followed by IV injections of 3000 nmol DOTAP SUV and then 300 ug MARless plasmid DNA encoding HG-CSF. Rat No. 14 was injected at the start of the experiment with 3000 nmol DOTAP SUV and then 300 ug MAR-containing plasmid DNA encoding HG-CSF. Rat 14 was later injected on day 7 with 3 mg Dexamethasone (IP) followed by IV injections of 3300 nmol DOTAP SUV and then 330 ug MAR-containing plasmid DNA encoding HG-CSF. On day 21, rat no. 14 was injected with 3 mg Dexamethasone (IP) followed by IV injections of 4400 nmol DOTAP SUV and then 330 ug MAR-containing plasmid DNA encoding HG-CSF.

Results are shown in FIG. 18, which shows neutrophil elevation in rat serum following sequential IV injections of DOTAP cationic liposomes followed by plasmid DNA encoding HG-CSF. These results demonstrate that repeated sequential IV cationic liposome injection followed by an HG-CS plasmid DNA vector can produce sustained elevation of absolute neutrophil counts well at least the next 100 days. They also show that a dexamethasone pre-injection followed by a single sequential IV cationic liposome injection followed by an HG-CSF plasmid DNA vector can also produce sustained elevation of absolute neutrophil counts.

Example 4 Administration of PCR-Generated DNA Vectors Substantially Increases Both the Level and Duration Protein Product Production

This Example describes experiments conducted that demonstrate that administration of PCR-generated DNA vectors substantially increases both the level and duration of DNA vector gene-encoded protein product production in mice when compared to plasmid DNA.

Circularized, PCR Generated DNA Vector Increases the Level of Serum Human G-CSF Production in Mice

Methods: To generate DNA expression vectors by PCR, the HG-CSF expression cassette was amplified by PCR, using a primer pair containing the corresponding enzyme restriction site or a primer pair with a stem-loop configuration (for protected linear product) using Q5 High-Fidelity Polymerase (New England Biolab). The purified PCR product was digested with the corresponding enzyme (BamHI) at 10 U/ug then heat inactivated at 85 C for 20 min. Ligation of purified digested PCR was performed at 1 or 50 ng/ul with 80 T4DNA ligase Unit/ug of digested PCR at room temperature for 1 hr, then heat inactivated at 65 C for min. For the 1 ng/ul ligation condition, the volume was reduced with Millipore filtration Ultra15 before purification. The ligated PCR product was then eluted with lactated ringers from the purification column before being subjected to the final 0.2 uM filtration. All purification steps were performed using a Purelink PCR purification kit (Thermofisher).

Results are shown in FIG. 21, which shows levels of human G-CSF in mouse serum, 24 hours after sequential IV injection of 1050 nmoles of DOTAP cationic liposomes, followed by 70 ug of either HG-CSF plasmid- or different forms of PCR generated, HG-CSF expression cassette DNA. Mice receiving circularized, PCR generated DNA show higher levels of serum HG-CSF than those receiving either linear, PCR generated DNA or plasmid DNA.

Circularized, PCR Generated DNA Vector Increases the Level and Duration of Serum Human G-CSF and ANCs. Furthermore, a Single Re-Injection of PCR Generating DNA Substantially Increases Long-Term, High-Level Human G-CSF Levels in Mice

FIG. 22 shows levels of human G-CSF in mouse serum or plasma (left axis) and thousands per microliter absolute neutrophil counts (ANC) in whole blood (right axis) in mice for at least the next 302 days after initial injection. Mice were sequentially injected IV with cationic liposomes followed by PCR generated DNA. One group was given a repeat sequential injection of lipid and PCR DNA at Day 35, yielding significantly higher serum HG-CSF levels over the next 200 days. Heparinized whole blood was analyzed for ANC, and plasma was analyzed for HG-CSF by ELISA. Mice show significantly elevated levels of neutrophils 300 days after injection. Mean ANC in control (mock and un-injected) mice are 2K/uL. Thus, a single repeat injection of PCR generated vector DNA can substantially raise serum HG-CSF levels of the vector-expressed protein product as well as ANC for extended periods.

Inclusion of R6K DNA Sequence into Circularized, PCR Generated DNA Vector Increases the Level and Duration of Serum Human G-CSF Production in Mice

Methods: 27 g mice were injected IV with DOTAP SUV cationic liposomes, followed by circularized PCR DNA encoding HG-CSF, with or without an origin of replication (R6K). Mice were subsequently bled every 7 or 14 days. FIG. 23 shows human G-CSF levels in mouse serum for 106 days following one sequential injection of cationic liposomes followed by PCR generated DNA with or without an R6K origin of replication. Thus, incorporation of selected DNA sequences, including R6K, can significantly increase serum levels of the DNA vector encoded protein for extended periods.

Example 5 Neutral Lipid and Dexamethasone Palmitate Increases Serum Levels

This Example describes experiments conducted that demonstrate that the addition of neutral lipid and dexamethasone palmitate to sequential IV administration of a human G-CSF expression vector significantly increases both human G-CSF serum levels and ANC for prolonged periods in mice.

Methods: Mice were injected with one of three different liposome preparations. 1050 nmol DOTAP SUV alone, 1050 nmol DOTAP SUV mixed with 1050 nmol DMPC neutral lipid, or 1050 nmol SUV containing 2.5% Dexamethasone Palmitate mixed with 1050 nmol DMPC. The lipid injection was followed 2 minutes later by injection of a MAR containing plasmid coding for expression of human G-CSF. Mice were subsequently bled every 7 or 14 days. Heparinized whole blood was analyzed for neutrophil counts, and plasma was analyzed for HG-CSF by ELISA. Untreated control mice are consistently <3K/uL.

FIG. 24 shows human G-CSF and corresponding absolute neutrophil counts (ANC, right axis) levels in mice injected sequentially with cationic liposomes with or without neutral lipids or Dexamethasone Palmitate, followed by plasmid DNA. Mice show significantly elevated levels of neutrophils 99 days after injection. Mice receiving neutral lipids plus Dex palmitate show the highest ANC counts over time.

Example 6 Use of a Second Enhancer to Increase Serum Levels of Expressed Protein

This Example describes experiments conducted that demonstrate that the addition of a second enhancer in a human G-CSF DNA expression plasmid can increase human G-CSF serum levels after sequential IV injection in mice.

Methods: Mice were injected first with 1000 nmol each of DOTAP containing 2.5% Dexamethasone Palmitate and DMPC containing 2.5% Dexamethasone Palmitate. This was followed two minutes later by plasmids encoding HG-CSF. The four plasmids that were employed sv40-mCMVEF1 (SEQ ID NO:13; FIG. 26); mCMV-mCMVEF1 (SEQ ID NO:14; FIG. 27); mCMV-hCMVEF1 (SEQ ID NO:15; FIG. 28); and mCMVEF1 (SEQ ID NO:16; FIG. 29). The first three expression constructs contained extra enhancer sequences. Mice were bled as previously described.

FIG. 25 shows the results of this example. As shown in this Figure, enhancer combinations increase human G-CSF expression in mice 1 and 8 days after sequential IV injection. Shown are the results of a single treatment of liposomes followed by plasmids encoding human G-CSF and containing a series of different enhancer elements in combinations of two.

Example 7 Use of Super Enhancers

This Example describes experiments conducted that demonstrate that the addition of super enhancer sequences in a human G-CSF DNA expression plasmid can increase human G-CSF serum levels after sequential IV injection in mice.

Methods: Mice were first injected with 1000 nmol each of DOTAP containing 2.5% Dexamethasone Palmitate and DMPC neutral lipid containing 2.5% Dexamethasone Palmitate. This was followed 2 minutes later by HG-CSF encoding plasmids with or without super-enhancer elements (hr3). The hr3-containing plasmids are as follows: hr3-mCMVEF1 #2 (SEQ ID NO:17; FIG. 31); hr3-mcmvEF1 #5 (SEQ ID NO:18; FIG. 32); and hr3-mcmvEF1 #18 (SEQ ID NO:19; FIG. 33). One of the groups receiving the plasmid without a super-enhancer element (4th bar) was supplemented with 2.5% human serum albumin (HSA).

In FIG. 30, shown are mouse serum levels of human G-CSF, 24 hours after sequential IV injection of liposomes followed by plasmid DNA. Plasmids of the first three groups contain super-enhancer elements. Also shown is plasmid injected together with human serum albumin (HSA). Each of the three different DNA vectors containing a super enhancer element produced higher serum human G-CSF levels then the corresponding DNA vector lacking a super enhancer.

Example 8 Use of Super Enhancers

This Example describes experiments conducted that demonstrate that the addition of super enhancer, R6K or RNA-out DNA sequence in a human factor nine DNA expression plasmid can increase human factor nine serum levels after sequential IV injection in mice.

Methods: 27 g mice were injected IV with DOTAP SUV cationic liposomes, followed by DNA encoding human Factor IX. Both plasmid and circularized PCR constructs were used, with or without an origin of replication (R6K).

FIG. 34 shows plasma concentration of human Factor IX at 24 hrs after sequential IV injection of liposomes and various different FIX DNA expression plasmids. The plasmids used were: FIX plasmid (SEQ ID NO:20; FIG. 35); FIX R6K1 (SEQ ID NO:21; FIG. 36); FIX R6K2 (SEQ ID NO:22; FIG. 37); FIX Superenh (SEQ ID NO:23; FIG. 38); and FIX RNA-out (SEQ ID NO:24; FIG. 39). In addition to the control FIX plasmid (1st bar), plasmids included two different PCR generated DNA with R6K Element, FIX plasmid containing a super-enhancer and a FIX plasmid generated using RNA-out. Each of the modified DNA vectors produced higher human FIX serum protein levels one day after injection.

Example 9 CRISPR/Cas9 Mediated Knockdown 10 Days and 40 Days after Injection

This Example describes experiments conducted that demonstrate anti-p65 CRISPR/Cas9-mediated knockdown of mouse NFkB-p65 protein 10 days and 40 days after sequential IV injection in mice.

Mouse treatment methods: Three or four mice were injected per group. Each mouse received a single IV injection of 1000 nmoles of DOTAP cationic liposomes containing 2.5% dexamethasone palmitate (DP) incorporated into the liposome bilayer. This was followed two minutes later by a single IV injection of 75 ug of plasmid DNA encoding the indicated CRISPR- or ribozyme-based plasmids to suppress expression of endogenous mouse NFkB-p65. PECAM CRISPR control is shown in SEQ ID NO:10, and p65 CRISPR RelA1 is shown in SEQ ID NO:8. Plasmid EF1/U6 RelA1 (020117 #5) (SEQ ID NO:57) is shown in FIG. 79; plasmid EF1/U6 RelA4 (020117 #8) (SEQ ID NO:58) is shown in FIG. 80; and plasmid hul/EF1/U6 RelA1 (021417 #3) (SEQ ID NO:59) is shown in FIG. 81. The control group received an CRISPR/Cas9 plasmid identical to the anti-NFkB-p65 CRISPR plasmids except the 20 bp targeting sequence targeted mouse PECAM instead. All groups were treated two hours prior to IV injection with an IP injection of 40 mg/kg dexamethasone.

Tissue preparation and anti-mouse-p65 ELISA methods. Lung lysates were generated 24 hours after injection (Anti-p65 Ribozyme) and 8 days after injection (Anti-p65 CRISPR1/2) by dissection into 500 uL of prepared 1× Triton lysis buffer on ice. Samples include both lungs per animal. Each sample was homogenized (Polytron PT 2100) for 30 seconds, pulse sonicated (Misonix XL2000 Microson Ultrasonic Cell Disruptor XL 2000), and centrifuged for 10 minutes at 4 C, and the lysate was aspirated from the tissue pellet. Protein concentration from each lysate was then determined using a BCA total protein assay purchased from Thermo Fisher. Protein normalized lysate was added to a 96 well plate ELISA from Cell Signaling Technologies (PathScan Total NF-κB p65 Sandwich ELISA Kit) in duplicate as per the manufacture's instructions. The plate was then analyzed in a (Molecular Devices Spectramax M5) plate reader. After recording absorbance from the plate, a standard curve generated using murine B16 melanoma cell supernatant was fit by 4PL analysis. Error bars represent the standard error of the mean.

Description of Results: These data, shown in FIG. 78 and FIG. 40, which demonstrate that anti-mouse NFkB-p65 CRISPR/Cas9—as well as ribozyme, plasmid-based targeting vectors reduce the expression of endogenous mouse p65, 10 and 40 days following its systemic injection in mice.

Example 10 Long-Term HG-CSF Expression in Mice

This Example describes experiments conducted that demonstrate that a mouse sacrificed 582 days after a single sequential IV injection of cationic liposomes, then an HG-CSF DNA expression vector shows very large numbers of neutrophils in spleen and bone marrow not present in control mouse. Methods were as follows. Control mouse was un-injected. Treated 27 g mouse was injected IV with 800 nmol DOTAP SUV cationic liposomes, followed by 90 ug plasmid DNA encoding hG-CSF and euthanized 582 days after injection. Mice were exsanguinated and organs preserved in 10% neutral buffered formalin. FIG. 42 shows IHC results in bone marrow. In particular, FIGS. 42a (20×) and 42b (60×), control bone marrow, show a diverse mix of cell types surround bony trabeculae of normal femoral medullary cavity, with dark-staining erythoid cells particularly obvious. FIGS. 42c (20×) and 42d (60×), treated bone marrow, show a monotonous nearly solid sheet of pale-staining cells replace bony trabecular elements in femoral marrow pale staining myeloid lineage cells (polymorphonuclear leukocytes) with oval, indented oval, band and segmented forms replace most other cell types within femoral marrow. FIG. 43 shows IHC results in spleen tissue. FIGS. 43a (20×) and 43b (60×), control spleens, show red/dark portions of white (lymphoid) pulp of normal spleen showing diverse cell population. FIGS. 43c (20×) and 43d (60×), treated spleen, show pale-staining myeloid lineage cells (pmn's) with oval, indented oval, band and segmented forms replace most other cell types.

Example 11 Long-Term HG-CSF Expression in Rat

This Example describes experiments conducted that demonstrate that a rat sacrificed 168 days after last sequential IV injection of cationic liposomes, then an HG-CSF DNA expression vector shows very large numbers of neutrophils in bone marrow not present in control rat. The methods were as follows. Control rat was un-injected. Treated rat: a 150 g female rat was injected at the start of the experiment with 3000 nmol DOTAP SUV and then 300 ug of a DNA expression vector encoding HG-CSF. The treated rat was later injected on day 7 with 3 mg dexamethasone (IP) followed by IV injections of 3300 nmol DOTAP SUV and then 330 ug of the DNA expression vector encoding HG-CSF. On day 21, the treated rat was re-injected with 3 mg dexamethasone (IP) followed by IV injections of 4400 nmol DOTAP SUV and then 330 ug of the DNA expression vector encoding HG-CSF. Rats were euthanized, exsanguinated, and organs preserved in 10% neutral buffered formalin. FIG. 44 shows IHC results in bone marrow. In particular, FIGS. 44a (20×) and 42b (60×), control bone marrow, show a diversity of cell types with round, dark staining erythroid lineage particularly obvious in femoral marrow. FIGS. 44c (20×) and 44d (60×), treated bone marrow, show pale staining myeloid lineage cells (polymorphonuclear leukocytes) with oval, indented oval, band and segmented forms predominate in femoral marrow. A few clusters of dark-staining erythroid lineage cells remain. FIG. 45a shows control rat, vertebral body at 40×, while FIG. 45b shows the HGCSF rat vertebral body at 40×.

Example 12 Expression of Anti-Human PCSK9 Monoclonal Antibody

This Example describes sequential IV injection of cationic liposomes followed by a DNA expression vector encoding anti-human PCSK9 monoclonal antibody to reduce LDL in mice. Five CD-1 mice are injected per group. For the 2 months prior to injection, mice are placed on a high cholesterol and cholic acid diet to increase LDL cholesterol (Envigo Atherogenic Teklad Diet TD.02028). Each mouse then receives a single IV injection of 1050 nmoles of DOTAP cationic liposomes containing 2.5% dexamethasone palmitate (DP) incorporated into the liposome bilayer, followed two minutes later by a single IV injection of 75 ug of one of three different plasmid DNAs encoding an anti-human PCSK9 monoclonal antibody (mAb) or a plasmid DNA encoding an anti-human CD20 monoclonal antibody as a control group. All groups are treated two hours prior to IV injection with an IP injection of dexamethasone. DNARx-31H4-2A (SEQ ID NO:25; FIG. 46) and DNARx-21B12 (P2A) (SEQ ID NO:27; FIG. 48) encodes anti-PCSK9 mAb heavy and light chain cDNAs separated by a 2A self-cleaving peptide. Plasmids DNARx-31H4 (SEQ ID NO:26; FIG. 47) and DNARx-21B12 (SEQ ID NO:28; FIG. 49) are dual expression cassette plasmid vectors that encode different versions of anti-PCSK9 mAb heavy and light chain cDNAs respectively. Serum levels of mouse LDL cholesterol are measured 1 week prior to injection, then 24 hours following injection and in 7-day intervals thereafter. The serum LDL assay is photometric, involving the enzymatic breakdown of LDL substrate in the presence of another compound to form a dye. The color intensity of the dye is then measured by absorbance assay and is performed by the UC Davis Veterinary diagnostic laboratory. It is anticipated that the LDL levels in the treated mice will be reduced, but not in the control mice.

Example 13 Expression of Anti-Human CD47 Monoclonal Antibody

This Example describes sequential IV injection of cationic liposomes followed by a DNA expression vector encoding anti-human CD47 monoclonal antibody to suppress Raji, human B cell lymphoma tumor progression in tumor-bearing nude mice. Five athymic nude mice are injected per group. Mice receive 0.1×105-2×106 Raji cells subcutaneously in the shoulder or flank. Ten to fourteen days later, or when tumors reach a volume of 70-100 mm3, each mouse receives a single IV injection of 1050 nmoles of DOTAP cationic liposomes containing 2.5% dexamethasone palmitate (DP) incorporated into the liposome bilayer, followed two minutes later by a single IV injection of 75 ug of a DNA expression plasmid encoding an anti-human CD47 monoclonal antibody or a plasmid DNA encoding an anti-human PCSK9 monoclonal antibody as a control group. All groups are treated two hours prior to IV injection with an IP injection of 40 mg/kg dexamethasone. DNARx-CD47-2A (P2A) (SEQ ID NO:29; FIG. 50) encodes anti-CD47 mAb heavy and light chain cDNAs separated by a 2A self-cleaving peptide. DNARx-CD47 (SEQ ID NO:30; FIG. 51) is a dual expression cassette plasmid vector that encodes the anti-CD47 mAb heavy and light chain cDNAs respectively. Tumor volume is measured by caliper on a weekly or twice weekly basis following DNA expression vector injection. It is anticipated that tumor volume in the treated mice will be reduced, but not in the control mice.

Example 14 Expression of Anti-Human CD47 and Anti-Human CD20 Monoclonal Antibodies

This Example describes sequential IV injection of cationic liposomes followed by a DNA expression vector encoding anti-human CD47 monoclonal antibody, anti-human CD20 monoclonal antibody or both anti-human CD47 and anti-CD20 monoclonal antibodies to suppress Raji, human B cell lymphoma tumor progression in tumor-bearing nude mice. Five athymic nude mice are injected per group. Mice receive 0.1×105-2×106 Raji cells subcutaneously in the shoulder or flank. Ten to fourteen days later, or when tumors reach a volume of 70-100 mm3, each mouse receives a single IV injection of 1050 nmoles of DOTAP cationic liposomes containing 2.5% dexamethasone palmitate (DP) incorporated into the liposome bilayer, followed two minutes later by a single IV injection of 75 ug of a DNA expression plasmid encoding an anti-human CD47 monoclonal antibody, an anti-human CD20 monoclonal antibody, anti-human CD47 plus anti-CD20 monoclonal antibodies, or a plasmid DNA encoding an anti-human PCSK9 monoclonal antibody as a control group. All groups were treated two hours prior to IV injection with an IP injection of 40 mg/kg dexamethasone.

DNARx-CD47-2A (P2A) (SEQ ID NO:29; FIG. 50) encodes anti-CD47 mAb heavy and light chain cDNAs separated by a 2A self-cleaving peptide and Plasmid 715.1 2a (P2A) (SEQ ID NO:3) encodes anti-CD20 mAb heavy and light chain cDNAs separated by a 2A self-cleaving peptide. DNARx-CD47 (SEQ ID NO:30; FIG. 51) is a dual expression cassette plasmid vector that encodes the anti-CD47 mAb heavy and light chain cDNAs. Tumor volume is measured by caliper on a weekly or twice weekly basis following DNA expression vector injection. It is anticipated that tumor volume in the treated mice will be reduced, but not in the control mice.

Example 15 Expression of Anti-Influenza Stem Antigen Monoclonal Antibodies

This Example describes sequential IV injection of cationic liposomes followed by a DNA expression vector encoding anti-influenza A stem antigen to prevent and/or treat influenza A in mice. Five C57B16 mice are injected per group. Prior to injection, mice are inoculated with 2×MLD50 of PR/8/34 (H1N1), HKx31 (H3N1) or B/Lee/40 viral strains of influenza. The respective MLD50 of a challenge virus are determined by infection of unvaccinated mice with increasing amounts of virus. The mice are monitored for weight loss and mortality for 14-20 days following infection. Each mouse then receives a single IV injection of 1050 nmoles of DOTAP cationic liposomes containing 2.5% dexamethasone palmitate (DP) incorporated into the liposome bilayer, followed two minutes later by a single IV injection of 75 ug of one of three different plasmid DNAs encoding an anti-influenza A stem antigen monoclonal antibody or a plasmid DNA encoding an anti-human CD20 monoclonal antibody as a control group. All groups are treated two hours prior to IV injection with an IP injection of 40 mg/kg dexamethasone.

Plasmids DNARx-D8-2A (SEQ ID NO:31; FIG. 52), DNARx-F10-2A (SEQ ID NO:32; FIG. 53) and DNARx-A66-2A (P2A) (SEQ ID NO:33; FIG. 54) encode anti-influenza A stem antigen mAb heavy and light chain cDNAs separated by a 2A self-cleaving peptide. Plasmids DNARx-D8 (SEQ ID NO:34; FIG. 55), DNARx-F10 (SEQ ID NO:35; FIG. 56) and DNARx-A66 (SEQ ID NO:36; FIG. 57) are dual expression cassette plasmid vectors that encode different versions of anti-influenza A stem antigen heavy and light chain cDNAs respectively. Plasmid DNARx-HA-MITD (SEQ ID NO:37; FIG. 58) encodes that HA from PR/8/34 (H1N1) with MHC class I transmembrane and cytosolic domains (MITD) and plasmid DNARx-SEC-partial HA-MITD (SEQ ID NO:38; FIG. 59) encodes that partial HA from PR/8/34 (H1N1) with MHC class I signal peptide fragment (SEC) and transmembrane and cytosolic domains (MITD). Plasmids DNARx-D8-2A-HA-MITD (SEQ ID NO:39; FIG. 60), DNARx-F10-2A-HA-MITD (SEQ ID NO:40; FIG. 61), DNARx-A66-2A-HA-MITD (SEQ ID NO:41; FIG. 62), DNARx-D8-2A-SEC-partial-HA-MITD (SEQ ID NO:42; FIG. 63), DNARx-F10-2A-SEC-partial-HA-MITD (SEQ ID NO:43; FIG. 64) and DNARx-A66-2A SEC-partial-MITD (SEQ ID NO:44; FIG. 65) are dual expression cassette plasmid in which the first expression cassette encodes anti-influenza A stem antigen mAb heavy and light chain cDNAs separated by a 2A self-cleaving peptide and the second expression cassette encodes HA from PR/8/34 (H1N1) with MHC class I transmembrane and cytosolic domains (MITD) or partial HA from PR/8/34 (H1N1) with MHC class I signal peptide fragment (SEC) and transmembrane and cytosolic domains (MITD). The presence of influenza nucleoprotein in mouse serum is detected by ELISA using influenza A- or B-specific anti-nucleoprotein antibodies (Millipore, Billerica, MA). It is anticipated that treated mice will have their influenza prevented or treated, while the controls will not.

Example 16 Expression of Anti-Mouse PD-1 Monoclonal Antibodies, Ovalbumin, and gp-70

This Example describes sequential IV injection of cationic liposomes then a DNA expression vector encoding anti-mouse PD-1 monoclonal antibody, ovalbumin, gp-70, anti-mouse PD-1 monoclonal antibody plus ovalbumin or anti-mouse PD-1 monoclonal antibody plus gp-70 to suppress B16 melanoma or CT26 colon tumor progression in tumor-bearing mice. Five C57B16 mice are injected subcutaneously in the flank with 2×105 B16 cells per animal, or five BALBC mice are injected subcutaneously in the flank with 2×105 CT26 cells per animal. At day four following inoculation, each mouse receives a single IV injection of 1050 nmoles of DOTAP cationic liposomes containing 2.5% dexamethasone palmitate (DP) incorporated into the liposome bilayer, followed two minutes later by a single IV injection of 75 ug of a DNA expression plasmid encoding an anti-mouse PL-1 monoclonal antibody, ovalbumin, gp-70, or a plasmid DNA encoding an anti-human PCSK9 monoclonal antibody as a control group. All groups are treated two hours prior to IV injection with an IP injection of 40 mg/kg dexamethasone. Plasmid DNARx-PD1-2A (P2A) (SEQ ID NO:48; FIG. 69) encodes anti-PD-1 mAb heavy and light chain cDNAs separated by a 2A self-cleaving peptide, plasmid DNARx-SEC-OVA-MITD (SEQ ID NO:49; FIG. 70) encodes the ovalbumin restricted epitope H2Kb and MHC class I signal peptide fragment (SEC) and the transmembrane and cytosolic domains (MITD), plasmid DNARx-SEC-gp70-MITD (SEQ ID NO:50; FIG. 71) encodes the H-2Ld-restricted peptide antigen AH1 and MHC class I signal peptide fragment (SEC) and the transmembrane and cytosolic domains (MITD). Plasmid DNARx-PD1-2A OVA (SEQ ID NO:51; FIG. 72) or DNARx-PD1-2A gp70 (SEQ ID NO:52; FIG. 73) is a dual expression cassette plasmid vector that encodes the anti-PD-1 mAb heavy and light chain cDNAs separated by a 2A self-cleaving peptide in the first expression cassette and either the ovalbumin restricted epitope H2Kb or H-2Ld-restricted peptide antigen AH1 and MHC class I signal peptide fragment (SEC) and the transmembrane and cytosolic domains (MITD) in the second expression cassette. Tumor volume is measured by caliper every three to four days and animals with tumors exceeding 15 mm in volumetric diameter and/or show signs of impaired health are euthanized. It is anticipated that tumor volume in the treated mice will be reduced, but not in the control mice.

Example 17 Expression of Anti-Human Anti-CD20 Monoclonal Antibodies, Human G-CSF, and Streptococcal Cas9

This Example describes sequential IV injection of cationic liposomes followed by DNA expression vectors encoding anti-human anti-CD20 monoclonal antibody, human G-CSF, streptococcal Cas9, anti-CD20 monoclonal antibody plus HG-CSF or anti-CD20 monoclonal antibody plus Cas9 in mice. Five CD-1 mice are injected per group. Each mouse receives a single IV injection of 1050 nmoles of DOTAP cationic liposomes containing 2.5% dexamethasone palmitate (DP) incorporated into the liposome bilayer, followed two minutes later by a single IV injection of 75 ug plasmid DNAs encoding an anti-human CD20 monoclonal antibody (mAb), HG-CSF, Cas9, anti-human CD20 monoclonal antibody plus HG-CSF, anti-human CD20 monoclonal antibody plus HG-CSF or a plasmid DNA encoding an anti-human CD20 monoclonal antibody plus luciferase as a control group. All groups are treated two hours prior to IV injection with an IP injection of 40 mg/kg dexamethasone. Plasmid DNARx CD20-2A Cas9 (SEQ ID NO:53; FIG. 74) or DNARx CD20-2A HG-CSF (SEQ ID NO:54; FIG. 75) is a dual expression cassette plasmid vector that the first expression cassette encodes the anti-CD20 mAb heavy and light chain cDNAs separated by a 2A self-cleaving peptide and driven by mCMV-EF1 while the second expression cassette encodes Cas9 or HG-CSF and driven by hCMV-ferritin heavy chain promoter. Serum levels of anti-CD20 and HG-CSF will be measured by specific ELISAs every seven days after injection as previously described in the methods, while Cas9 protein levels will be measured by Cas9 ELISA and/or Western blot form mouse lung lysates of previously injected mice every 7 days after injection. Both assays will be use the following validated capture antibody: MAC133 Anti-Cas9 Antibody, clone 7A9 (Millipore).

Example 18 In Vivo Expression of Anti-Human Anti-CD20 Monoclonal Antibodies is Increased with Neutral Liposomes

This Example describes how co-injecting of neutral liposomes with cationic liposomes increases mouse serum anti-CD20 monoclonal antibody levels over time versus injecting the same cationic liposomes without neutral liposomes. Three mice per group were given IP injections of Dexamethasone at a level of 40 mg/kg. Two hours later they were sequentially injected, first with 1000 nmol or 1250 nmol DOTAP SUV, with or without 1000 nmol DMPC (1,2-Dimyristoyl-SN-glycero-3-phosphocholine) neutral lipid, and then of plasmid vector containing Rituximab (anti-CD20 monoclonal antibody) cDNA. Serum levels of Rituximab protein were measured by ELISA after 24 hours and every 2-3 weeks thereafter. Results are shown in FIG. 82, which shows that the inclusion of neutral lipids with cationic liposomes increases serum anti-CD20 monoclonal antibody levels.

Example 19 In Vivo Expression of Anti-Human Anti-CD20 Monoclonal Antibodies is Increased with Neutral Liposomes and Dexamethasone Palmitate

This Example describes how incorporating dexamethasone palmitate into neutral liposomes further increases gene expression. Three mice per group were given IP injections of Dexamethasone at a level of 40 mg/kg. Two hours later they were sequentially injected, first with 1000 nmol DOTAP SUV containing 2.5% Dexamethasone Palmitate, and 1000 nmol DMPC neutral lipid containing 1, 2.5, 5, or 10% Dexamethasone Palmitate, and then 75 ug of plasmid vector containing Rituximab cDNA. Serum levels of Rituximab protein were measured by ELISA after 24 hours. The results are shown in FIG. 83, which shows that employing dexamethasone palmitate with neutral liposomes further increases gene expression in vivo.

Example 20 Inclusion of Syn 21 and/or Delta-p10 in Vector Increases In Vivo Gene Expression

This Example describes how including Syn 21 and/or delta-p10 sequences 5′ or 3′ of the anti-CD20 mAb heavy and light chain cDNA's increases serum anti-CD20 mAb levels in mice. Three mice per group were given IP injections of Dexamethasone at a level of 40 mg/kg. Two hours later they were sequentially injected, first with 1000 nmol DOTAP SUV and 1000 nmol DMPC neutral lipid, both containing 2.5% Dexamethasone Palmitate, and then of plasmid vector containing Rituximab cDNA. A representative vector construct, containing both the Syn21 and delta-p10 sequences, is shown in SEQ ID NO:82 (FIG. 85). Serum levels of Rituximab protein were measured by ELISA after 24 hours. Results are shown in FIG. 84, which shows that including Syn 21 and/or delta-p10 sequences into the vectors increases gene expression.

Example 21 Inclusion of Hr3 Super Enhancer in Vector Increases In Vivo Gene Expression

This Example describes how the addition of a five prime hr3 super enhancer sequence increases the expression of human G CSF as well as anti-CD 20 monoclonal antibody in mice. Three mice per group were given IP injections of Dexamethasone at a level of 40 mg/kg. Two hours later they were sequentially injected, first with 1000 nmol DOTAP SUV and 1000 nmol DMPC neutral lipid, both containing 2.5% Dexamethasone Palmitate, and then of plasmid vector containing human G-CSF or Rituximab cDNA. Serum levels of hG-CSF or Rituximab protein were measured by ELISA after 24 hours. The results are shown in FIG. 86, which shows increased G CSF expression (FIG. 86A) and increased Rituximab anti-CD20 expression (FIG. 86b) when the hr3 super enhancer is included in the plasmid.

Example 22 Inclusion of R6K in 3′ or 5′ UTR Region Increases In Vivo Gene Expression

This Example describes how the insertion of an R6K origin of replication sequence either in the 5′ UTR or 3′UTR of the human factor nine cDNA, increases the level of human factor nine serum levels produced in mice. Three mice per group were given IP injections of Dexamethasone at a level of 40 mg/kg. Two hours later they were sequentially injected, first with 1000 nmol DOTAP SUV and 1000 nmol DMPC neutral lipid, both containing 2.5% Dexamethasone Palmitate, and then 75 ug (FIG. 87A) or 60 ug (FIG. 87B) of plasmid vector containing Factor IX cDNA. Plasma levels of Factor IX protein were measured by ELISA after 24 hours. Results are shown in FIG. 87, which shows that locating the R6K origin of replication in the 3′ or 5′ UTR of the Factor IX gene increased expression levels at both the ug level (FIG. 87A) and the 60 ug level (FIG. 87B).

Example 23 Long-Term Anti-CD20 Antibody Expression after Single Vector Injection

This Example describes how mouse serum Rituximab levels produced 148, 232 and 284 days after a single Rituximab DNA injection remain therapeutically effective (FIG. 88A), inducing levels of CD20+human tumor cell lysis comparable to recombinant Rituximab protein. Three mice per group were given IP injections of Dexamethasone at a level of 40 mg/kg. Two hours later they were sequentially injected, first with 1050 nmol DOTAP SUV containing 2.5% Dexamethasone Palmitate, and then 75 ug of plasmid vector containing Rituximab cDNA. Serum levels of Rituximab protein were measured by ELISA after 24 hours and every 1-2 weeks thereafter. The results are shown in FIG. 88. FIG. 88A shows long-term Rituximab levels at different time points over 284 days, showing long-term expression. FIG. 88B shows that the anti-CD20 mouse sera was able to induce human tumor cell lysis at levels comparable to Rituximab protein.

Example 24 Long-Term Anti-IL5 Antibody Expression after Single Vector Injection

This Example describes how one sequential IV injection of a dual cassette, single plasmid DNA vector (SEQ ID NO:83; FIG. 90) encoding the anti-human interleukin-5 mAb (Mepoluzimab; 2B6) heavy chain and light chain cDNAs produces therapeutic anti-IL-5 mAb serum levels in mice for >92 days, as assayed by ELISA. Three mice per group were given IP injections of Dexamethasone at a level of 40 mg/kg. Two hours later they were sequentially injected, first with 1120 nmol DOTAP SUV containing 2.5% Dexamethasone Palmitate along with 1000 nmol DMPC MLB containing 5% Dexamethasone Palmitate, and then 85 ug of plasmid vector containing anti-IL-5 cDNA (2B6). Serum levels of anti-IL-5 mAb were measured by ELISA after 24 hours and every 1-2 weeks thereafter. The results are shown in FIG. 89, which shows therapeutic anti-IL-5 mAb (2B6) serum levels expressed for at least 92 days.

Example 25 Long-Term Anti-Flu Antibody Expression after Single Vector Injection

This Example describes how one sequential IV injection of a dual cassette, single plasmid DNA vector (SEQ ID NO:84, FIG. 92) encoding the anti-influenza 5J8 mAb heavy chain and light chain cDNAs produces therapeutic anti-influenza A mAb serum levels by ELISA (FIG. 91A) effectively neutralizes the Ca109 epidemic influenza strain (FIG. 91B) for >85 days. Three mice per group were given IP injections of Dexamethasone at a level of 40 mg/kg. Two hours later they were sequentially injected, first with 1120 nmol DOTAP SUV containing 2.5% Dexamethasone Palmitate along with 1000 nmol DMPC MLV containing 5% Dexamethasone Palmitate, and then 85 ug of plasmid vector containing cDNA for the anti-flu antibody. Serum levels of anti-flu protein were measured by ELISA after 24 hours and every 1-2 weeks thereafter. The neutralization methods were as follows. Serum from mice injected with anti-flu cDNA at various time points was heat-killed, diluted 1:40 in DMEM/BSA, then mixed with 100×TCID50 of X-179 Ca109 H1N1 virus. One hour later 30,000 MDCK2 cells were added to the serum/virus mixture along with TPCK-treated trypsin. Following a 16-18 hour incubation, MDCK2 cells were scored for infection using an influenza A-specific immunoassay against viral nucleoprotein.

Example 26 Long Term Expression

This Example describes how sequential IV injection of a single plasmid DNA vector (SEQ ID NO:85, FIG. 94) encoding the anti-human interleukin-5 mAb (Mepoluzimab; 2B6) heavy chain and light chain cDNAs and the human G-CSF cDNA produces therapeutic anti-IL-5mAb as well as hG-CSF serum levels by ELISA for >66 days. Three mice per group were given IP injections of Dexamethasone at a level of 40 mg/kg. Two hours later they were sequentially injected, first with 1120 nmol DOTAP SUV containing 2.5% Dexamethasone Palmitate along with 1000 nmol DMPC MLV containing 5% Dexamethasone Palmitate, and then 88 ug of plasmid vector containing cDNA for anti-IL-5 mAb and hG-CSF. Serum levels of each protein were measured by ELISA after 24 hours and every 1-3 weeks thereafter. Results are shown in FIG. 93, which shows the expression levels in mice of anti-IL-5mAb as well as hG-CSF were at therapeutic levels for at least 66 days.

Example 27 Dual Cassette Provides Increased Expression

This Example describes how a dual expression cassette, single plasmid vector containing two hG-CSF cassettes produces higher absolute neutrophil counts over time than a single cassette hG-CSF vector. Three mice per group were given IP injections of Dexamethasone at a level of 40 mg/kg. Two hours later they were sequentially injected, first with 1000 nmol DOTAP SUV and 1000 nmol DMPC MLV, both containing 2.5% Dexamethasone Palmitate, and then 75 ug of plasmid vector containing a dual-cassette cDNA for hG-CSF. Plasma levels of hG-CSF protein were measured by ELISA after 24 hours and every 1-2 weeks thereafter. Absolute Neutrophil Count (ANC) was assessed from whole blood. FIG. 95 show the results of this Example, which shows that the dual cassette expression of the same encoded protein provides higher serum levels in mice than single-cassette expression of the encoded protein.

Example 28 Dual Cassette Provides Increased Expression

This Example describes how a dual expression cassette, single plasmid vector, each cassette containing an identical anti-human IL-5 heavy and light chain mAb cDNAs separated by a porcine teschovirus-1 2A (P2A) self cleaving peptide sequence produces higher anti-human IL-5 serum mAb levels in mice than a single cassette anti-human IL-5 mAb encoding DNA vector. Three mice per group were given IP injections of Dexamethasone at a level of 40 mg/kg. Two hours later they were sequentially injected, first with 1120 nmol DOTAP SUV containing 2.5% Dexamethasone Palmitate along with 1000 nmol DMPC MLV containing 5% Dexamethasone Palmitate, and then 88 ug of plasmid vector containing cDNA for and IL-5. Serum levels of IL-5 protein were measured by ELISA after 24 hours. Results are shown in FIG. 96, which shows that the dual cassette vector expressing anti-human IL-5 heavy and light chains produces higher anti-human IL-5 serum mAb levels than the single cassette anti-human IL-5 encoding DNA vector.

Example 29 Dual Cassette Provides Increased Expression

This Example describes how a dual expression cassette, single plasmid vector, each cassette containing an identical anti-influenza A heavy and light chain monoclonal antibody 5J8 cDNAs separated by a porcine teschovirus-1 2A (P2A) self cleaving peptide sequence produces higher anti-5J8 mAb serum levels in mice than a single cassette anti-human IL-5 mAb encoding DNA vector. Three mice per group were given IP injections of Dexamethasone at a level of 40 mg/kg. Two hours later they were sequentially injected, first with 1120 nmol DOTAP SUV containing 2.5% Dexamethasone Palmitate along with 1000 nmol DMPC MLV containing 5% Dexamethasone Palmitate, and then 88 ug of plasmid vector containing cDNA for IL-5. Serum levels of IL-5 protein were measured by ELISA after 24 hours. The results are shown in FIG. 97, which shows that the dual cassette vector expressing anti-5J8 mAb produces higher anti-5J8 serum mAb levels than the single cassette anti-5J8 encoding DNA vector.

Example 30 Dual Cassette Single Plasmid Expression of Different mAbs, and Co-Injection of Two Single Cassette Plasmids Expressing Different mAbs

This Example describes how one IV injection of a dual expression cassette, single plasmid vector, one cassette containing an anti-influenza A heavy and light chain monoclonal antibody 5J8 cDNAs separated by a porcine teschovirus-1 2A (P2A) self cleaving peptide sequence, and the second cassette containing an anti-human IL-5 heavy and light chain monoclonal antibody cDNAs (2B6) separated by a porcine teschovirus-1 2A (P2A) self cleaving peptide produces significant serum levels of both monoclonal antibodies in mice. Furthermore, one IV co-injection of two different single expression cassette DNA vectors encoding the intact heavy and light chain monoclonal antibodies anti-influenza 5J8 and anti-human IL-5 (2B6) respectively also produce significant serum levels of both monoclonal antibodies in mice. Three mice per group were given IP injections of Dexamethasone at a level of 40 mg/kg. Two hours later they were sequentially injected, first with 1120 nmol DOTAP SUV containing 2.5% Dexamethasone Palmitate along with 1000 nmol DMPC MLV containing 5% Dexamethasone Palmitate, and then 88 ug of plasmid vector. Serum levels of protein were measured by ELISA after 24 hours. The results are shown in FIG. 98, which shows how a dual cassette single plasmid expresses different mAbs in vivo, and how two single cassette plasmids that are co-injected express different mAbs in vivo.

Example 31 Dual Cassette Single Plasmid Expression of Different mAbs

This Example describes how one IV injection of a dual expression cassette, single plasmid vector, one cassette containing an anti-influenza A heavy and light chain monoclonal antibody 5J8 cDNAs separated by a porcine teschovirus-1 2A (P2A) self cleaving peptide sequence and the second cassette containing an anti-human IL-5 heavy (2B6) and light chain monoclonal antibody cDNAs separated by a porcine teschovirus-1 2A (P2A) self cleaving peptide produces significant serum levels of both monoclonal antibodies in mice. Three mice per group were given IP injections of Dexamethasone at a level of 40 mg/kg. Two hours later they were sequentially injected, first with 1120 nmol DOTAP SUV containing 2.5% Dexamethasone Palmitate along with 1000 nmol DMPC MLV containing 5% Dexamethasone Palmitate, and then 88 ug of plasmid vector. Serum levels of protein were measured by ELISA after 24 hours. The results are shown in FIG. 99. FIG. 99A shows serum expression levels of the anti-human IL-5 mAb over 43 days, and FIG. 99B shows serum expression levels of the anti-influenza A mAb over 43 days.

Example 32 Triple Cassette Single Plasmid Expression of Different mAbs

This Example describes how one IV injection of a triple expression cassette, single plasmid vector, one cassette containing an anti-influenza A heavy and light chain monoclonal antibody 5J8 cDNAs separated by a porcine teschovirus-1 2A (P2A) self cleaving peptide sequence, the second cassette containing an anti-human IL-5 heavy and light chain monoclonal antibody cDNA's separated by a porcine teschovirus-1 2A (P2A) self cleaving peptide, and the third cassette containing an anti-human CD20 heavy and light chain monoclonal antibody cDNAs separated by a porcine teschovirus-1 2A (P2A) self cleaving peptide produces significant serum levels of all three different monoclonal antibodies in mice. Furthermore, one IV co-injection of three different single expression cassette DNA vectors encoding the intact heavy and light chain monoclonal antibodies: anti-influenza 5J8, anti-human IL-5, and anti-human CD20 mAbs respectively also produce significant serum levels of all three different monoclonal antibodies in mice. Three mice per group were given IP injections of Dexamethasone at a level of 40 mg/kg. Two hours later they were sequentially injected, first with 1120 nmol DOTAP SUV containing 2.5% Dexamethasone Palmitate along with 1000 nmol DMPC MLV containing 5% Dexamethasone Palmitate, and then 88 ug of plasmid vector. Serum levels of protein were measured by ELISA after 24 hours. The results are shown in FIG. 100, which shows simultaneous expression of Rituximab (anti-CD20), anti-IL5 mAb, and anti-influenza mAb, both from a single vector (left side), as well as by co-injection of three separate vectors (right side).

Example 33 Expression of Anti-PCSK9 mAbs to Reduce LDL Levels

This Example describes how one IV injection of a single plasmid vector expressing anti-PCSK9 mAbs reduces LDL levels in mice. Three mice per group were given IP injections of Dexamethasone at a level of 40 mg/kg. Two hours later they were sequentially injected, first with 1000 nmol each of DOTAP SUV and DMPC MLV, both containing 2.5% Dexamethasone Palmitate, and then 75 ug of plasmid vector. Plasma levels of LDL cholesterol were measured 15 days after injection and plotted according to proportion relative to LDL cholesterol measurements on the same mice prior to injection. FIG. 101 shows the results, which shows that a single plasmid vector expressing anti-PCSK9 mAbs reduces LDL levels in mice.

Example 34 Expression of Anti-PCSK9 mAbs Provides Long-Lasting Reduction of LDL Levels

This Example describes how expression of anti-PCSK9 mAbs in vivo provides long-lasting reduction of LDL levels. Mice were assessed for serum LDL levels prior to injection. On the day of injection, three mice per group were given IP injections of Dexamethasone at a level of 40 mg/kg. Two hours later they were sequentially injected, first with 1000 nmol DOTAP SUV and 1000 nmol DMPC MLV, both containing 2.5% Dexamethasone Palmitate, and then 75 ug of plasmid vector encoding the light and have chain of an anti-PCSK9 mAb. Serum levels of LDL cholesterol were measured every 7-21 days thereafter. The results are shown in FIG. 102, which show long-term reduction in LDL levels in mice expressing anti-PCSK9 mAbs.

Example 35 Expression of Anti-PCSK9 mAbs Reduces LDL Levels in Mice on Fatty Diet

This Example describes how expression of anti-PCSK9 mAbs in vivo provides reduction of LDL levels in mice on a fatty diet compared to control (anti-CD20 mAb expression). Mice were assessed for serum LDL levels prior to injection. On the day of injection, three mice per group were given IP injections of Dexamethasone at a level of 40 mg/kg. Two hours later they were sequentially injected, first with 1000 nmol DOTAP SUV and 1000 nmol DMPC MLV, both containing 2.5% Dexamethasone Palmitate, and then 75 ug of plasmid vector. The day after injection, mice were switched to a fatty, cholesterol-elevating diet. Serum levels of LDL cholesterol were measured every 7-14 days thereafter. FIG. 103 shows the results, which shows that mice expressing the anti-PCSK9 mAbs had lower LDL levels over time compared to the control mice expressing the control anti-CD20 antibodies.

Example 36 Durability of mAb Expression

This Example describes how the long-term expression of mAbs, including Rituximab (anti-CD20 mAb), anti-flu mAb (FI6), anti-flu mAb (5J8), and anti-IL5 mAb. Three mice per group were given IP injections of Dexamethasone at a level of 40 mg/kg. Two hours later they were sequentially injected. Sequential injections for Rituximab comprised injections of Dexamethasone at a level of 40 mg/kg. Two hours later they were sequentially injected, first with 1050 nmol DOTAP SUV containing 2.5% Dexamethasone Palmitate, and then 75 ug of plasmid vector containing Rituximab cDNA. Sequential injections for anti-flu and anti-IL-5 antibodies comprised 1120 nmol DOTAP SUV containing 2.5% Dexamethasone Palmitate along with 1000 nmol DMPC MLV containing 5% Dexamethasone Palmitate, and then 88 ug of plasmid vector. Serum levels of protein were measured by ELISA after 24 hours, and then every 7-21 days thereafter. The results are shown in FIG. 104, which shows expression of anti-flu FI6 mAb for about 25 days, expression of anti-flu 5J8 mAb and anti-IL4 mAB for over 100 days, and expression of Rituximab for over 275 days.

Example 37 Various Plasmid Vector Doses

This Example describes a comparison of expression levels from four different doses of plasmid vector expressing Rituximab. Three mice per group were given IP injections of Dexamethasone at a level of 40 mg/kg. Two hours later they were sequentially injected as follows. Liposomes were injected first, composed of 1000 nmol DMPC MLV containing 5% Dexamethasone Palmitate as well as 1000, 1080, 1170, or 1250 nmol DOTAP SUV containing 2.5% Dexamethasone Palmitate; plasmid vector was injected second in doses of 75, 81, 88, or 95 ug. Serum levels of protein were measured by ELISA after 24 hours. Results are shown in FIG. 105, which shows good expression levels from all four plasmid doses.

Example 38 Enhanced mAb Expression

This Example describes how the ALB and AZU signal sequences enhance the expression of the 5J8 mAb. Three mice per group were given IP injections of Dexamethasone at a level of 40 mg/kg. Two hours later they were sequentially injected, first with 1120 nmol DOTAP SUV containing 2.5% Dexamethasone Palmitate along with 1000 nmol DMPC MLV containing 5% Dexamethasone Palmitate, and then 88 ug of plasmid vector. Serum levels of protein were measured by ELISA after 24 hours. The results are shown in FIG. 106, which shows enhanced expression of the 5J8 mAb by using the ALB and AZU signal sequence.

Example 39 P53 Expression In Vivo

This Example describes how the human p53 gene is widely expressed in mouse lungs 24 hours after IV injection, and further how the human p53 gene is expressed predominately in vascular endothelial cells. Three mice per group were given IP injections of Dexamethasone at a level of 40 mg/kg. Two hours later they were sequentially injected, first with DOTAP SUV liposomes and DMPC neutral lipids, both at 1000 nmol with 2.5% Dex Palmitate by weight, then two minutes later, 75 ug per mouse of plasmid vector encoding human p53 (FIG. 108, SEQ ID NO:86). Lungs were harvested and processed for immunohistochemistry 24 hrs post injection. Lung sections were stained for human p53 (brown color). FIG. 107A shows control mouse lung tissue, and FIG. 107B shows human p53 injected mouse lung tissue stained for p53, showing that the p53 gene is widely expressed in mouse lungs. Lung tissue from the treated mice was dual-stained for human p53 and mouse CD31 (PECAM), a vascular endothelial cell-specific marker. Co-localization of p53 and CD31, in FIGS. 107C and 107D, shows predominate vascular endothelial cell human p53 expression in p53-injected mice. FIGS. 107C and 107D shows the same tissue section, with different stains. CD31 staining in both figures is extensive since alveolar walls are lined by continuous endothelium.

All publications and patents mentioned in the present application are herein incorporated by reference. Various modification and variation of the described methods and compositions of the invention will be apparent to those skilled in the art without departing from the scope and spirit of the invention. Although the invention has been described in connection with specific preferred embodiments, it should be understood that the invention as claimed should not be unduly limited to such specific embodiments. Indeed, various modifications of the described modes for carrying out the invention that are obvious to those skilled in the relevant fields are intended to be within the scope of the following claims.

Claims

1. A system comprising:

a) a first composition comprising a first amount of polycationic structures, wherein said first composition is free, or essentially free, of nucleic acid molecules; and
b) a second composition comprising a therapeutically effective amount of expression vectors,
wherein said expression vectors comprise nucleic acid sequences encoding one or more therapeutic biomolecules; and
at least one of the following:
i) wherein the ratio of said first amount of said polycationic structures to said therapeutically effective amount of expression vectors is 5:1 to 25:1;
ii) wherein 2.0% to 15.0% of said first composition comprises dexamethasone palmitate and/or dexamethasone;
iii) wherein said first composition further comprises neutral lipid; and
iv) wherein said polycationic structures comprise empty liposomes, and wherein said empty liposomes present in said first composition have a z-average diameter of about 20-85 nm.

2. The system of claim 1, wherein said expression vectors comprise circularized synthetically amplified nucleic acid, plasmid-based vector, or minicircle DNA.

3. The system of claim 1, wherein said one or more therapeutic biomolecules comprise one or more monoclonal antibodies (mAb), or antigen-binding portion thereof.

4. The system of claim 3, wherein said antigen-binding portion of said mAb is selected from a Fab, F(ab)2, and/or scFv.

5. The system of claim 3, wherein said mAb or antigen-binding portion thereof specifically binds to a pathogen or pathogen component, a tumor antigen, or a cytokine.

6-7. (canceled)

8. The system of claim 3, wherein said one or more mAb or antigen-binding portion thereof comprise a first mAb or antigen-binding portion thereof that specifically binds to a first target molecule and a second mAb or antigen-binding portion thereof that specifically binds to a second, different, target molecule.

9. The system of claim 8, wherein said one or more mAb or antigen-binding portion thereof comprise a first mAb or antigen-binding portion thereof that specifically binds to a first target molecule, a second mAb or antigen-binding portion thereof that specifically binds to a second target molecule, and a third mAb or antigen-binding portion thereof that specifically binds to a third target molecule, wherein said first, second, and third target molecules are different molecules.

10. The system of claim 1, wherein said one or more therapeutic biomolecules comprise one or more CRISPR/Cas9 components in one or more expression cassettes in said expression vectors.

11. The system of claim 1, wherein said one or more therapeutic biomolecules comprise a nucleic acid, optionally wherein said nucleic acid is an antisense oligonucleotide, ribozyme, an shRNA, miRNA, siRNA, piRNA, snoRNA, tsRNA, or srRNA.

12. (canceled)

13. The system of claim 1, wherein said expression vectors

(a) encode a first therapeutic biomolecule and a second therapeutic biomolecule, wherein said first and second therapeutic biomolecules: i) express for different lengths of time than one another, and/or ii) are the same;
(b) comprise at least one of the following: an R6K origin of replication, an hr3 enhancer, a BV3 signal sequence, a Syn21 sequence, a delta-p10 sequence, or an MITD (MHC class I trafficking signal) sequence;
(c) are CpG-free or CpG-reduced; or
(d) contain a plurality of CpG motifs, and/or are not CpG-free or CpG-reduced.

14-16. (canceled)

17. A method of expressing one or more therapeutic biomolecules in a subject, comprising: a) administering a first composition of a system of claim 1 into a subject; and b) administering a second composition of said system into said subject.

18. A method of expressing a monoclonal antibody (mAb), Fab, F(ab)2, and/or scFv in a subject comprising:

a) administering a first composition to a subject, wherein said first composition comprises a first amount of polycationic structures, and
wherein said first composition is free, or essentially free, of nucleic acid molecules; and
b) administering a second composition to said subject within about 300 minutes of administering said first composition,
wherein said second composition comprises a therapeutically effective amount of expression vectors encoding said mAb, said Fab, said F(ab)2, and/or scFv, and
wherein, as a result of said administering said first composition and said administering said second composition, said first therapeutic protein is expressed in said subject.

19. The method of claim 18, wherein said subject has at least one symptom of a disease or condition, or has at least physiological trait to be altered, and wherein said first therapeutic protein is expressed in said subject at a therapeutic level with respect to said disease or condition, or at an effective level sufficient to alter said physiological or disease trait.

20. The method of claim 18, wherein said first therapeutic protein is expressed in said subject at a prophylactic level with respect sufficient to prevent the subject from acquiring one or more infectious diseases.

21. An aqueous composition comprising or consisting essentially of:

a) polycationic structures or neutral lipid present in said composition at a concentration of between 500 nM and 500 mM;
b) dexamethasone and/or dexamethasone palmitate present in said composition at a concentration between 1-10% of said composition; and
c) a physiologically tolerable buffer, and
wherein said composition is free, or essentially free, of nucleic acid molecules.

22. The composition of claim 21, wherein

(a) said polycationic structure are cationic lipids that are present as small unilamellar vesicles;
(b) said polycationic structures comprise DOTAP; or
(c) said polycationic structures are present in said composition at a concentration of between 800 nM and 1500 nM, or between 10 mM and 100 mM.

23. The composition of claim 21, wherein said physiologically tolerable buffer is selected from the group consisting of: saline buffer, 5% dextrose in water, lactated ringers buffer, and any combination thereof.

24-26. (canceled)

27. The composition of claim 21, wherein said neutral lipids

(a) are present as multi-lamellar vesicles;
(b) comprise DMPC, or
(c) are present in said composition at a concentration of between 800 nM and 1500 nM, or between 10 mM and 100 mM.

28-41. (canceled)

42. A system comprising:

a) a composition of claim 18, and
b) a syringe, wherein at least part of said composition is located inside said syringe.

43. The system of claim 42, wherein said composition located inside said syringe contains a therapeutic and/or prophylactic dose of said polycationic structures.

44-47. (canceled)

Patent History
Publication number: 20240033376
Type: Application
Filed: Oct 27, 2022
Publication Date: Feb 1, 2024
Applicant: DNARx (San Francisco, CA)
Inventors: Robert James DEBS (San Francisco, CA), Timothy D. HEATH (Madison, WI), Chakkrapong HANDUMRONGKUL (Richmond, VA)
Application Number: 18/050,389
Classifications
International Classification: A61K 48/00 (20060101); C12N 15/88 (20060101); C12N 15/85 (20060101); A61K 31/7105 (20060101); A61K 39/395 (20060101); A61K 9/127 (20060101); C12N 15/10 (20060101); C07K 16/10 (20060101); C07K 16/40 (20060101); A61K 9/00 (20060101); C12N 15/87 (20060101);