ENHANCED NANOPARTICLE DELIVERY SYSTEMS

Information

  • Patent Application
  • 20230416782
  • Publication Number
    20230416782
  • Date Filed
    December 09, 2021
    2 years ago
  • Date Published
    December 28, 2023
    5 months ago
Abstract
Disclosed are methods for the enhancement of nucleic acid delivery systems. The methods may employ treatment with a compound and/or an RNAi molecule in combination with a nucleic acid to improve nucleic acid uptake into a cell. In particular, the disclosed methods may be useful for improved gene therapy techniques.
Description
BACKGROUND

Transfer of nucleic acids, including double and single stranded DNA as well as RNA, into eukaryotic cells is the most essential step of any gene transfer, repair, or editing technology. Transfer of nucleic acids may be accomplished using many types of delivery vehicles, including cationic lipids, viral vectors and nucleic acid nanoparticles condensed with cationic polymers such as poly lysine or polyethyleneimine. However, significant costs involved in the preparation of these materials present a significant limitation in their usage as both research tools and translational applications such as gene therapy. Further, efficacy of nucleic acid transfer with or without modification of the vector remains an area in need of improvement. The instant disclosure seeks to address one or more of the aforementioned needs in the art.


BRIEF SUMMARY

Disclosed are methods for the enhancement of nucleic acid delivery systems. The methods may employ treatment with a compound and/or a nucleic acid molecule such as, for example, one or more molecules selected from RNAi, miRNA, shRNA, tRNA, siRNA, single and double stranded DNA in combination with, for example, prior to or concurrent with, administration of a nucleic acid to improve nucleic acid uptake into a cell. In particular, the disclosed methods may be useful for improved gene therapy techniques in which a disclosed RNAi and/or a disclosed compound may be administered prior to or concurrently with the gene therapy delivery vehicle containing a nucleic acid.





BRIEF DESCRIPTION OF THE DRAWINGS

This application file contains at least one drawing executed in color. Copies of this patent or patent application publication with color drawing(s) will be provided by the Office upon request and payment of the necessary fee.


Those of skill in the art will understand that the drawings, described below, are for illustrative purposes only. The drawings are not intended to limit the scope of the present teachings in any way.



FIG. 1 is a schematic of partial NNP (DNP and RNP) interactome including nucleoin, APC, and SPTAN1, which were identified by MS analysis of 2 gel bands from DNP and RNP pull downs not present in bead alone control. Lighter color circles connote interactions that enhance NNP-mediated gene transfer, while darker circles connote interactions that inhibit. (+) or (−) along arrows connote impact on interactions with DNP. (+) or (−) by pharmacological agents reflect impact on the activation of GR, CDK1, or CKII. For example, while cortisone would increase nucleolin at the membrane via GR (10), spermine would increase it through stimulation of CKII mediated phosphorylation of nucleolin. Pull downs initially conducted in primary hepatocytes and repeated three times in wd-AECs for 2 non-CF and 3 CF subjects. This DNP interactome was observed in all the hepatocyte and CF and non-CF wd-AEC studies.



FIG. 2 depicts immunoprecipitation of protein interactors of DNA nanoparticles in HeLa cells.



FIG. 3 depicts enhanced DNA nanoparticle transfection through siRNA expression.



FIG. 4 depicts transfection of human primary airway epithelia either following prior treatment with scrambled shRNA, shRNA specific for APC, or shRNA specific for SPTAN1 for 48 hours. Luciferase expression was measured two days post transfection. * connotes different from saline pretreatment (triplicates in three experiments p<0.01).



FIG. 5 depicts primary cell cultures of airway epithelia transfected with DNPs containing a plasmid coding for luciferase driven by the ubiquitin B promoter (5.4 kb). shRNA lentivirus infection was 48 hours prior to transfection while spermine (CK11 inducer) roscovitine (CDK1 inhibitor), resveratrol (CDK1 agonist), or cortisone (GR agonist) were added four hours prior to transfection. Treatments were saline (S), APC shRNA (-APC), SPTAN1 shRNA (-SPTAN1), Hydrocortisone©, spermine (Sper), roscovitine (Ros), or resveratrol (RES). Luciferase expression was measured two days post transfection. * connotes different from saline (p<0.01).



FIG. 6 is a schematic showing DNP Gene Transfer Process & Barriers to Gene Transfer. Barriers to gene transfer prevent the DNP from completing these processes. Protein-DNP interactions can affect how the DNP moves past these barriers.



FIG. 7 is a schematic showing Protein-Vector Interactions to Circumvent Intracellular Barriers.



FIG. 8 is a schematic showing Protein-Vector Interactions to Circumvent Intracellular Barriers. Concentration of cortisone vs gene expression as a percent of vehicle treatment.



FIG. 9. Interactome Analysis to Identify DNP/Protein Interactions



FIG. 10 depicts graphs showing that gene transfer of luciferase is enhanced by pharmacologic manipulation in vitro. HeLa cells were transfected with luciferase DNPs either four hours after cells were treated with drug, at the same time as cells were treated with drug, or four hours before cells were treated with drug; DNP alone, RX001 (roscovitine), RX011 (spermine), and RX008 (ruxolitinib).



FIG. 11 depicts graphs showing that gene transfer of luciferase is enhanced by pharmacologic manipulation in vitro. HeLa cells were transfected with luciferase DNPs either four hours after cells were treated with drug, at the same time as cells were treated with drug, or four hours before cells were treated with drug, DNP alone, RX012 (doxorubicin), RX013 (acetohexamide), and RX014 (sildenafil citrate).



FIG. 12. Gene transfer of luciferase is enhanced by pharmacologic manipulation in vitro. HeLa cell were treated for four hours prior to luciferase DNP administration. Drugs were obtained from a blinded plate. Labels on graphs indicate drugs position on blinded plate. *: p<0.05, **: p<0.01, ***: p<0.001, ***: p<0.0001, compared to DNP Alone group.



FIG. 13 shows that enhancement of in vivo DNP transfection by pharmaceuticals is maintained over time. Roscovitine (10 mg/kg, CDK1 inhibitor), spermine (20 mg/kg, CKII activator), and ruxolitinib (1 mg/kg, JAK inhibitor) were dosed in mice 2 hours prior to intratracheal administration of 100 μg luciferase DNP. A. BLI images taken at 2, 3, 7, and 14 days after DNP dosage of a single mouse from groups of mice given DNP only (D), roscovitine (Rs), spermine (S), and ruxolitinib (Rx). Total photon measurements were taken from the chest of each mouse, ROI outlined in red, and background luminescence was subtracted from mice saline control mice (not shown). B. Total photons collected over a 10 min BLI exposure from the ROI at each day. (n>10 for each group, mean and SD shown, p<0.05, p<0.01, p<0.001 compared to the DNP alone group) Note: At day 14, spermine was significantly different (P<0.5) from the DNP alone group as well as roscovitine (p<0.01). Mean graphed with SD, significance for each time point is in the order of ruxo, sper, rosc (top to bottom). *: p<0.05, **: p<0.01,***: p<0.001, ***: p<0.0001, compared to the DNP Alone group at the same day post treatment.



FIG. 14 shows that pharmacological manipulation of interactome proteins enhances luciferase gene transfer in vivo. Bioluminescent Image (BLI) analysis, RX001, RX011, RX008, day 2, day 3, day 7, and day 14. *: p<0.05, **: p<0.01, ***: p<0.001, ***: p<0.0001, compared to the DNP Alone group at the same day post treatment.



FIG. 15 shows that pharmacological manipulation of interactome proteins enhances luciferase gene transfer in vivo BLI image analysis and luciferase activity assay for DNP alone, RX001 (roscovitine), RX011 (spermine), and RX008 (ruxolitinib).



FIG. 16. Gene transfer of hCFTR is enhanced by pharmacologic manipulation of interactome proteins. DMP alone, RX001 (roscovitine), and RX008 (ruxolitinib), 2 days post-DNP administration, 4 days post-DNP administration and 7 days post-DNP administration.



FIG. 17 depicts pharmacological manipulations before, after, and during DNP transfection. Hela cells were given roscovitine (1 μM, CDK1 inhibitor), spermine (1 μM, CKII activator), or ruxolitinib (0.1 μM, JAK inhibitor) at various times during transfection of luciferase DNPs. A. Hela cells were dosed with drugs for 4 hours, washed, and then given luciferase DNPs for 24 hours. B. Hela cells were simultaneously given drug and luciferase DNPs for 24 hr. C. Hela cells were given luciferase DNPs for 4 hours, washed, and then given drug for 24 hr. All cells were then lysed and analyzed for luciferase activity with a light-based assay. (n=8 for each group, signifies p<0.05 and signifies p<0.001).



FIG. 18 shows pharmaceutical enhancement of DNP gene delivery efficacy in vivo. Roscovitine (10 mg/kg, CDK1 inhibitor), spermine (20 mg/kg, CKII activator), and ruxolitinib (1 mg/kg, JAK inhibitor) were dosed in mice 2 hours prior to intratracheal administration of 100 μg luciferase DNP. All data was collected 14 days post DNP administration. A. representative BLI images of mice given I) DNP only, II) Roscovitine, III) Spermine, and IV) ruxolitinib. The ROI, outlined in red, was consistently drawn on each mouse and used to quantify total photons in each mouse. B. Total photons collected over a 10 min BLI exposure from the ROI. Mice that received saline instead of DNPs (not shown) were used to subtract background from the experimental mice. C. Lungs from mice used in B were harvested immediately after BLI imaging and assayed for luciferase activity. (n>10 for each group, p<0.05, p<0.01, p<0.001)



FIG. 19 shows pharmacological manipulations before, after, and during DNP transfection. Hela cells were given roscovitine (1 μM, CDK1 inhibitor), spermine (1 μM, CKII activator), or ruxolitinib (0.1 μM, JAK inhibitor) at various times during transfection of luciferase DNPs. A. Hela cells were dosed with drugs for 4 hours, washed, and then given luciferase DNPs for 24 hours. B. Hela cells were simultaneously given drug and luciferase DNPs for 24 hr. C. Hela cells were given luciferase DNPs for 4 hours, washed, and then given drug for 24 hr. All cells were then lysed and analyzed for luciferase activity with a light-based assay. (n=8 for each group, mean and SEM shown, signifies p<0.05 and signifies p<0.001)



FIG. 20 depicts pharmaceutical enhancement of DNP gene delivery efficacy in Vivo. Roscovitine (10 mg/kg, CDK1 inhibitor), spermine (20 mg/kg, CKII activator), and ruxolitinib (1 mg/kg, JAK inhibitor) were dosed in mice 2 hours prior to intratracheal administration of 100 μg luciferase DNP. All data was collected 14 days post DNP administration. A. representative BLI images of mice given I) DNP only, II) Roscovitine, III) Spermine, and IV) ruxolitinib. The ROI, outlined in red, was consistently drawn on each mouse and used to quantify total photons in each mouse. B. Total photons collected over a 10 min BLI exposure from the ROI. Mice that received saline instead of DNPs (not shown) were used to subtract background from the experimental mice. C. Lungs from mice used in B were harvested immediately after BLI imaging and assayed for luciferase activity. (n>10 for each group, mean and SEM shown, p<0.05, p<0.01, p<0.001)



FIG. 21 shows that enhancement of in vivo DNP transfection by pharmaceuticals is maintained over time. Roscovitine (10 mg/kg, CDK1 inhibitor), spermine (20 mg/kg, CKII activator), and ruxolitinib (1 mg/kg, JAK inhibitor) were dosed in mice 2 hours prior to intratracheal administration of 100 μg luciferase DNP. A. BLI images taken at 2, 3, 7, and 14 days after DNP dosage of a single mouse from groups of mice given DNP only (D), roscovitine (Rs), spermine (S), and ruxolitinib (Rx). Total photon measurements were taken from the chest of each mouse, ROI outlined in red, and background luminescence was subtracted from mice saline control mice (not shown). B. Total photons collected over a 10 min BLI exposure from the ROI at each day. (n>10 for each group, mean and SEM shown, p<0.05, p<0.01, p<0.001 compared to the DNP alone group) Note: At day 14, spermine was significantly different (P<0.5) from the DNP alone group as well as roscovitine (p<0.01). Mean graphed with SEM, significance for each time point is in the order of ruxo, sper, rosc (top to bottom). *: p<0.05, **: p<0.01,***: p<0.001, ***: p<0.0001, compared to the DNP Alone group at the same day post treatment.



FIG. 22. CFTR Expression comparison between NHBE cells (normal human bronchial/tracheal epithelial cells), Untreated Mice and Day 2 DNP Treated Mice, and CFTR Expression comparison in Day 4 mice treated with a vehicle (DNP Alone) or drug. Day 4 data represents vector subtracted values for the PCR of CFTR.





DETAILED DESCRIPTION
Definitions

Unless otherwise noted, terms are to be understood according to conventional usage by those of ordinary skill in the relevant art. In case of conflict, the present document, including definitions, will control. Preferred methods and materials are described below, although methods and materials similar or equivalent to those described herein may be used in practice or testing of the present invention. All publications, patent applications, patents and other references mentioned herein are incorporated by reference in their entirety. The materials, methods, and examples disclosed herein are illustrative only and not intended to be limiting.


As used herein and in the appended claims, the singular forms “a,” “and,” and “the” include plural referents unless the context clearly dictates otherwise. Thus, for example, reference to “a method” includes a plurality of such methods and reference to “a dose” includes reference to one or more doses and equivalents thereof known to those skilled in the art, and so forth.


The term “about” or “approximately” means within an acceptable error range for the particular value as determined by one of ordinary skill in the art, which will depend in part on how the value is measured or determined, e.g., the limitations of the measurement system. For example, “about” may mean within 1 or more than 1 standard deviation, per the practice in the art. Alternatively, “about” may mean a range of up to 20%, or up to 10%, or up to 5%, or up to 1% of a given value. Alternatively, particularly with respect to biological systems or processes, the term may mean within an order of magnitude, preferably within 5-fold, and more preferably within 2-fold, of a value. Where particular values are described in the application and claims, unless otherwise stated the term “about” meaning within an acceptable error range for the particular value should be assumed.


As used herein, the term “effective amount” means the amount of one or more active components that is sufficient to show a desired effect. This includes both therapeutic and prophylactic effects. When applied to an individual active ingredient, administered alone, the term refers to that ingredient alone. When applied to a combination, the term refers to combined amounts of the active ingredients that result in the therapeutic effect, whether administered in combination, serially or simultaneously.


The terms “individual,” “host,” “subject,” and “patient” are used interchangeably to refer to an animal that is the object of treatment, observation and/or experiment. Generally, the term refers to a human patient, but the methods and compositions may be equally applicable to non-human subjects such as other mammals. In some embodiments, the terms refer to humans. In further embodiments, the terms may refer to children.


As used herein, a “pharmaceutically acceptable form thereof” includes any pharmaceutically acceptable salts, prodrugs, tautomers, isomers, and/or isotopically labeled derivatives of a compound provided herein, as defined below and herein.


The term “pharmaceutically acceptable salt,” as used herein, refers to a formulation of a compound that does not cause significant irritation to an organism to which it is administered and does not abrogate the biological activity and properties of the compounds described herein. As used herein, the disclosed compounds also include pharmaceutically acceptable salts thereof.


As used herein, the term “prodrug” refers to a derivative of a parent compound that requires transformation within the body in order to release the parent compound. A prodrug can be inactive when administered to a subject, but is converted in vivo to an active compound, for example, by hydrolysis (e.g., hydrolysis in blood). In certain cases, a prodrug has improved physical and/or delivery properties over the parent compound. Prodrugs are typically designed to enhance pharmaceutically and/or pharmacokinetically based properties associated with the parent compound. The advantage of a prodrug can lie in its physical properties, such as enhanced water solubility for parenteral administration at physiological pH compared to the parent compound, or it enhances absorption from the digestive tract, or it can enhance drug stability for long-term storage. (see, e.g., Bundgard, H., Design of Prodrugs (1985), pp. 7-9, 21-24 (Elsevier, Amsterdam). A discussion of prodrugs is provided in Higuchi, T., et al., “Pro-drugs as Novel Delivery Systems,” A.C.S. Symposium Series, Vol. 14, and in Bioreversible Carriers in Drug Design, ed. Edward B. Roche, American Pharmaceutical Association and Pergamon Press, 1987, both of which are incorporated in full by reference herein.


The term “prodrug” is also meant to include any covalently bonded carriers, which release the active compound in vivo when such prodrug is administered to a subject. Prodrugs of an active compound, as described herein, can be prepared by modifying functional groups present in the active compound in such a way that the modifications are cleaved, either in routine manipulation or in vivo, to the parent active compound. Prodrugs include compounds wherein a hydroxy, amino or mercapto group is bonded to any group that, when the prodrug of the active compound is administered to a subject, cleaves to form a free hydroxy, free amino or free mercapto group, respectively. Examples of prodrugs include, but are not limited to, acetate, formate and benzoate derivatives of an alcohol or acetamide, formamide and benzamide derivatives of an amine functional group in the active compound and the like. Other examples of prodrugs include compounds that comprise —NO, —NO2, —ONO, or —ONO2 moieties. Prodrugs can typically be prepared using well-known methods, such as those described in Burger's Medicinal Chemistry and Drug Discovery, 172-178, 949-982 (Manfred E. Wolff ed., 5th ed., 1995), and Design of Prodrugs (H. Bundgaard ed., Elselvier, N.Y., 1985).


For example, if a disclosed compound or a pharmaceutically acceptable form of the compound contains a carboxylic acid functional group, a prodrug can comprise a pharmaceutically acceptable ester formed by the replacement of the hydrogen atom of the acid group with a group such as (C1-C8)alkyl, (C2-C12)alkanoyloxymethyl, 1-(alkanoyloxy)ethyl having from 4 to 9 carbon atoms, 1-methyl-1-(alkanoyloxy)-ethyl having from 5 to 10 carbon atoms, alkoxycarbonyloxymethyl having from 3 to 6 carbon atoms, 1-(alkoxycarbonyloxy)ethyl having from 4 to 7 carbon atoms, 1-methyl-1-(alkoxycarbonyloxy)ethyl having from 5 to 8 carbon atoms, N-(alkoxycarbonyl)aminomethyl having from 3 to 9 carbon atoms, 1-(N-(alkoxycarbonyl)amino)ethyl having from 4 to 10 carbon atoms, 3-phthalidyl, 4-crotonolactonyl, gamma-butyrolacton-4-yl, di-N,N—(C1-C2)alkylamino(C2-C3)alkyl (such as (3-dimethylaminoethyl), carbamoyl-(C1-C2)alkyl, N,N-di(C1-C2)alkylcarbamoyl-(C1-C2)alkyl and piperidino-, pyrrolidino- or morpholino(C2-C3)alkyl.


Similarly, if a disclosed compound or a pharmaceutically acceptable form of the compound contains an alcohol functional group, a prodrug can be formed by the replacement of the hydrogen atom of the alcohol group with a group such as (C1-C6)alkanoyloxymethyl, 1-((C1-C6)alkanoyloxy)ethyl, 1-methyl-1-((C1-C6)alkanoyloxy)ethyl (C1-C6)alkoxycarbonyloxymethyl, N—(C1-C6)alkoxycarbonylaminomethyl, succinoyl, (C1-C6)alkanoyl, α-amino(C1-C4)alkanoyl, arylacyl and α-aminoacyl, or α-aminoacyl-α-aminoacyl, where each α-aminoacyl group is independently selected from the naturally occurring L-amino acids, P(O)(OH)2, —P(O)(O(C1-C6)alkyl) 2 or glycosyl (the radical resulting from the removal of a hydroxyl group of the hemiacetal form of a carbohydrate).


If a disclosed compound or a pharmaceutically acceptable form of the compound incorporates an amine functional group, a prodrug can be formed by the replacement of a hydrogen atom in the amine group with a group such as R-carbonyl, RO-carbonyl, NRR′-carbonyl where R and R′ are each independently (C1-C10)alkyl, (C3-C7)cycloalkyl, benzyl, a natural α-aminoacyl or natural α-aminoacyl-natural α-aminoacyl, —C(OH)C(O)OY1 wherein Y1 is H, (C1-C6)alkyl or benzyl, —C(OY2)Y3 wherein Y2 is (C1-C4) alkyl and Y3 is (C1-C6)alkyl, carboxy(C1-C6)alkyl, amino(C1-C4)alkyl or mono-N— or di-N,N—(C1-C6)alkylaminoalkyl, C(Y4)Y5 wherein Y4 is H or methyl and Y5 is mono-N— or di-N,N—(C1-C6)alkylamino, morpholino, piperidin-1-yl or pyrrolidin-1-yl.


The active agent may form salts, which are also within the scope of the preferred embodiments. Reference to a compound of the active agent herein is understood to include reference to salts thereof, unless otherwise indicated. The term “salt(s)”, as employed herein, denotes acidic and/or basic salts formed with inorganic and/or organic acids and bases. In addition, when an active agent contains both a basic moiety, such as, but not limited to an amine or a pyridine or imidazole ring, and an acidic moiety, such as, but not limited to a carboxylic acid, zwitterions (“inner salts”) may be formed and are included within the term “salt(s)” as used herein. Pharmaceutically acceptable (e.g., non-toxic, physiologically acceptable) salts are preferred, although other salts are also useful, e.g., in isolation or purification steps, which may be employed during preparation. Salts of the compounds of the active agent may be formed, for example, by reacting a compound of the active agent with an amount of acid or base, such as an equivalent amount, in a medium such as one in which the salt precipitates or in an aqueous medium followed by lyophilization. When the compounds are in the forms of salts, they may comprise pharmaceutically acceptable salts. Such salts may include pharmaceutically acceptable acid addition salts, pharmaceutically acceptable base addition salts, pharmaceutically acceptable metal salts, ammonium and alkylated ammonium salts. Acid addition salts include salts of inorganic acids as well as organic acids. Representative examples of suitable inorganic acids include hydrochloric, hydrobromic, hydroiodic, phosphoric, sulfuric, nitric acids and the like. Representative examples of suitable organic acids include formic, acetic, trichloroacetic, trifluoroacetic, propionic, benzoic, cinnamic, citric, fumaric, glycolic, lactic, maleic, malic, malonic, mandelic, oxalic, picric, pyruvic, salicylic, succinic, methanesulfonic, ethanesulfonic, tartaric, ascorbic, pamoic, bismethylene salicylic, ethanedisulfonic, gluconic, citraconic, aspartic, stearic, palmitic, EDTA, glycolic, p-aminobenzoic, glutamic, benzenesulfonic, p-toluenesulfonic acids, sulphates, nitrates, phosphates, perchlorates, borates, acetates, benzoates, hydroxynaphthoates, glycerophosphates, ketoglutarates and the like. Examples of metal salts include lithium, sodium, potassium, magnesium salts and the like. Examples of ammonium and alkylated ammonium salts include ammonium, methylammonium, dimethylammonium, trimethylammonium, ethylammonium, hydroxyethylammonium, diethylammonium, butylammonium, tetramethylammonium salts and the like. Examples of organic bases include lysine, arginine, guanidine, diethanolamine, choline and the like.


“Sequence identity” as used herein indicates a nucleic acid sequence that has the same nucleic acid sequence as a reference sequence, or has a specified percentage of nucleotides that are the same at the corresponding location within a reference sequence when the two sequences are optimally aligned. For example, a nucleic acid sequence may have at least 50%, 60%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% identity to the reference nucleic acid sequence. The length of comparison sequences will generally be at least 5 contiguous nucleotides, preferably at least 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, or 25 contiguous nucleotides, and most preferably the full length nucleotide sequence. Sequence identity may be measured using sequence analysis software on the default setting (e.g., Sequence Analysis Software Package of the Genetics Computer Group, University of Wisconsin Biotechnology Center, 1710 University Avenue, Madison, Wis. 53705). Such software may match similar sequences by assigning degrees of homology to various substitutions, deletions, and other modifications.


The term “NNP” refers to a Nucleic acid Nano Particle. A non-limiting example includes a complex of DNA or RNA with polymers of lysines (for example, 15-45 lysines long).


The term “DNP” refers to a DNA Nanoparticle


The term “RNP” refers to a RNA Nanoparticle


The term “Interactome” refers to the whole set of molecular interactions in a particular cell. The term specifically refers to physical interactions among molecules (such as those among proteins, also known as protein-protein interactions) but can also describe sets of indirect interactions among genes (genetic interactions).


The term “APC” refers to an adenomatous polyposis coli protein


The term “wd-AECs” refer to well-differentiated airway epithelial cells.


The term “SPTAN1” refers to Alpha II-spectrin, also known as Spectrin alpha chain, a protein that in humans is encoded by the SPTAN1 gene. Alpha II-spectrin is expressed in a variety of tissues and is highly expressed in cardiac muscle at Z-disc structures, costameres and at the sarcolemma membrane.


The term “GR” refers to a glucocorticoid receptor


The term “CDK1” refers to cyclin dependent kinase 1


The term “CKII” refers to casein kinase II


The term “Spermine” refers to a polyamine involved in cellular metabolism found in all eukaryotic cells.


The term “shRNA” refers to a small hairpin RNA or short hairpin RNA (shRNA) and is an artificial RNA molecule with a tight hairpin turn that can be used to silence target gene expression via RNA interference (RNAi)


Disclosed herein are methods for the enhancement of nucleic acid delivery systems by combination treatment with one or more compounds as disclosed herein and/or one or more RNAi molecules as disclosed herein. For example, the disclosed methods may be used with delivery of a nucleic acid such as a gene, a gene fragment, a fragment containing an active portion of a protein encoded by a gene, or the like. Further examples of nucleic acids that may be delivered include nucleic acid components of the CRISPR/CAS9, or short nucleic acids, such as microRNA or DNA or RNA oligonucleotides. The disclosed RNAi molecules and/or compounds may be administered to an individual in need of administration of a nucleic acid prior to administration of a nucleic acid delivery system, or concurrently with the administration of a nucleic acid delivery system.


In one aspect, the method may be a method for transferring a gene into a eukaryotic cell, in which the method may comprise administering a compacted nucleic acid nanoparticle and one or more agents selected from sonolisib (Steroid Lactone), LY294002 (Benzopyran), TG100115 (Pteridine), Trifluoperazine (Benzothiazine, Phenothiazine), CEP5214 (Indole Derivative, Pyrrolocarbazole), Afuresertib (Substituted Benzene, Phenethylamine, Amphetamine), Cation Vemurafenib (Aryl-Phenylketone), Suramin (Benzene Derivative, Benzanilide), Uprosertib (Benzene Derivative, Phenethylamine, Amphetamine), Flutamide, (Benzene Derivative, Trifluoromethylbenzene), Enzastaurin (Indole Derivative, N-alkylindole), Fasudil (Isoquinoline derivative), Ruboxistaurin (Macrolactam), Pentamidine (Phenol Ether), Uprosertib (Benzene Derivative, Phenethylamine, Amphetamine), Lestaurtinib (Indole derivative, Indolocarbazole), Adenine (Imidazolepyrimidine), Pimozide (Diphenylbutylpiperidine), Chlorpromazine (Phenothiazine), Afuresertib (Benzene Derivative, Phenethylamine, Amphetamine), and combinations thereof, to a eukaryotic cell.


In one aspect, the method may comprise administering an inhibitor of a protein that inhibits nanoparticle delivery uptake. In this aspect, the inhibitor may be selected from one or more of RNAi, miRNA, shRNA, tRNA, siRNA, single stranded DNA, double stranded DNA, and combinations thereof. In this aspect, the nucleic acid may inhibit synthesis of one or more proteins that inhibit nucleic acid delivery vehicle uptake. Exemplary proteins may include one or more protein selected from those of Table 1.


In one aspect, the method may comprise administering an active agent that facilitates compacted nucleic acid nanoparticle uptake into a cell. The active agent may inhibit synthesis of one or more proteins that inhibit nucleic acid delivery vehicle uptake.


In one aspect, the RNAi molecule may inhibit expression of a gene encoding a protein selected from Table 1.


In one aspect, the method may comprise administering a second active agent selected from an agent listed in Table 2 or Table 3.


In one aspect, the active agent may be selected from one or more of roscovitine, geldanamycin, acetohexamide, and ruxolitinib, or a combination thereof.


In one aspect, the nucleic acid delivery vehicle may be a nanoparticle comprising one or more of the aforementioned genes.


In one aspect, the compacted nucleic acid nanoparticle may comprise a nucleic acid plasmid and a polymer, wherein the nanoparticle may be compacted in the presence of a counter ion selected from trifluoroacetate (TFA), bromide, bicarbonate, glutamate, hydroxyl ions or combinations thereof.


In one aspect, the nucleic acid may be single or double stranded DNA, or a combination thereof.


In one aspect, the polymer may be a polycation. In one aspect, the polycation may be a lipid. In further aspects, the polycation may be a cysteine (C) containing polymer of lysine (K), such as CK30, a cysteine (C) containing polymer of arginine (R), such as CR30, or combinations thereof. In further aspects, the polycation may be selected from a cysteine (C) containing polymer of lysine (K) and arginine (R), such as C(K5R)5 or C(R5K)5 (e.g. CK15-90), polymers of arginine (e.g. CR15-90), or polymers of lysine mixed with arginine (e.g. C(KR5KR5KR5KR5KR5) or C(K5RK5RK5RK5RK5R)) conjugated to PEG and complexed with nucleic acids. In a further aspect, the polymer may be a lysine polymer, for example a polyethylene glycol (PEG)-substituted lysine polymer or polyethylenemine.


In one aspect, the compacted nucleic acid nanoparticle may have a shape selected from rod shape, ellipsoidal, spheroidal, or toroidal, and may have a diameter of from about 25 to about 400 nm in length as measured by electron microscopy.


The method, in certain aspects, may comprise the steps of

    • contacting a cell with an RNAi molecule or an active agent. The RNAi molecule or active agent may be in an amount sufficient to inhibit synthesis of one or more proteins that inhibit nucleic acid delivery vehicle uptake; and
    • contacting the eukaryotic cell with a nucleic acid delivery vehicle.


The cell may be, for example, a eukaryotic cell, derived from a human being.


In one aspect, the disclosed methods may be used to treat an individual in need of such treatment. The individual may be one in which administration a therapeutically effective amount of a protein may be advantageous to reversal, prevention, or amelioration of a disease state. The delivery of a protein may be achieved via administration of a gene, or portion of a gene that encodes an active portion of a protein, that may be subsequently expressed in the individual to provide a functional protein or functional protein fragment in a therapeutically effective amount. In this aspect, the method may comprise the steps of administering an RNAi that inhibits expression of a gene encoding a protein selected from a protein of Table 1 and/or a compound selected from Table 2 or 3, and/or an agent selected from sonolisib (Steroid Lactone), LY294002 (Benzopyran), TG100115 (Pteridine), Trifluoperazine (Benzothiazine, Phenothiazine), CEP5214 (Indole Derivative, Pyrrolocarbazole), Afuresertib (Substituted Benzene, Phenethylamine, Amphetamine), Cation Vemurafenib (Aryl-Phenylketone), Suramin (Benzene Derivative, Benzanilide), Uprosertib (Benzene Derivative, Phenethylamine, Amphetamine), Flutamide, (Benzene Derivative, Trifluoromethylbenzene), Enzastaurin (Indole Derivative, N-alkylindole), Fasudil (Isoquinoline derivative), Ruboxistaurin (Macrolactam), Pentamidine (Phenol Ether), Uprosertib (Benzene Derivative, Phenethylamine, Amphetamine), Lestaurtinib (Indole derivative, Indolocarbazole), Adenine (Imidazolepyrimidine), Pimozide (Diphenylbutylpiperidine), Chlorpromazine (Phenothiazine), Afuresertib (Benzene Derivative, Phenethylamine, Amphetamine), and combinations thereof. The RNAi or agent may be administered concurrently, before, or after administration of a drug delivery vehicle containing the nucleic acid that encodes the gene, or in some instances, the active portion of a gene, of interest.


The amount of compound and/or RNAi necessary to effect the methods of the instant disclosure may be determined by one of ordinary skill in the art. The dose administered to a subject, particularly a human, may be sufficient to effect the desired response in the subject over a reasonable period of time. The dose may be determined by the strength of the particular compound employed and the condition of the subject, as well as the body weight of the subject to be treated. The existence, nature, and extent of any adverse side effects that might accompany the administration of a particular compound also will determine the size of the dose and the particular route of administration employed with a particular patient. For example, the compounds may be therapeutically effective at low doses. Exemplary dosage ranges may be from about 0.001 mM, or less, to about 100 mM, or more, or from about 0.01, 0.05, 0.1, 0.5, 0.6, 0.7, 0.8, or 0.9 mM, to about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 20, 30, 40 50, 60, 70, 80, 90 or 100 mM. Accordingly, the compounds may be generally administered in low doses.


In one aspect, the gene is the CF gene, and the individual in need of treatment is an individual having cystic fibrosis.


In one aspect, the RNAi molecule may be one that inhibits expression of a gene encoding a protein selected from a protein of Table 1.









TABLE 1







Genes encoding proteins that modulate nucleic acid delivery vehicle uptake. The RNAi molecules of


the instant disclosure may inhibit expression of one or more of the genes listed in the table.









Uniprot ID
Gene names
Protein names





P04114
APOB
Apolipoprotein B-100 (Apo B-100) [Cleaved into:




Apolipoprotein B-48 (Apo B-48)]


P29536
LMOD1
Leiomodin-1 (64 kDa autoantigen 1D) (64 kDa




autoantigen 1D3) (64 kDa autoantigen D1) (Leiomodin,




muscle form) (Smooth muscle leiomodin) (SM-Lmod)




(Thyroid-associated ophthalmopathy autoantigen)


P68104
EEF1A1
Elongation factor 1-alpha 1 (EF-1-alpha-1) (Elongation



EEF1A
factor Tu) (EF-Tu) (Eukaryotic elongation factor 1 A-1)



EF1A
(eEF1A-1) (Leukocyte receptor cluster member 7)



LENG7


O19680

Pot. HLA-DP-alpha 1 (Aa −31 to +2) (441 is 1st base in




codon) (Fragment)


P46939
UTRN
Utrophin (Dystrophin-related protein 1) (DRP-1)



DMDL



DRP1


P08590
MYL3
Myosin light chain 3 (Cardiac myosin light chain 1)




(CMLC1) (Myosin light chain 1, slow-twitch muscle




B/ventricular isoform) (MLC1SB) (Ventricular myosin




alkali light chain) (Ventricular myosin light chain 1)




(VLCl) (Ventricular/slow twitch myosin alkali light




chain) (MLC-lV/sb)


P22695
UQCRC2
Cytochrome b-c1 complex subunit 2, mitochondrial




(Complex III subunit 2) (Core protein II) (Ubiquinol-




cytochrome-c reductase complex core protein 2)


Q16763
UBE2S
Ubiquitin-conjugating enzyme E2 S (EC 2.3.2.23) (E2



E2EPF
ubiquitin-conjugating enzyme S) (E2-EPF) (Ubiquitin



OK/SW-
carrier protein S) (Ubiquitin-conjugating enzyme E2-24



cl.73
kDa) (Ubiquitin-conjugating enzyme E2-EPF5)




(Ubiquitin-protein ligase S)


P00451
F8 F8C
Coagulation factor VIII (Antihemophilic factor) (AHF)




(Procoagulant component) [Cleaved into: Factor VIIIa




heavy chain, 200 kDa isoform; Factor VIIIa heavy chain,




92 kDa isoform; Factor VIII B chain; Factor VIIIa light




chain]


P52272
HNRNPM
Heterogeneous nuclear ribonucleoprotein M (hnRNP M)



HNRPM



NAGR1


P60660
MYL6
Myosin light polypeptide 6 (17 kDa myosin light chain)




(LC17) (Myosin light chain 3) (MLC-3) (Myosin light




chain alkali 3) (Myosin light chain A3) (Smooth muscle




and nonmuscle myosin light chain alkali 6)


P25054
APC DP2.5
Adenomatous polyposis coli protein (Protein APC)




(Deleted in polyposis 2.5)


P23458
JAK1
Tyrosine-protein kinase JAK1 (EC 2.7.10.2) (Janus



JAK1A
kinase 1) (JAK-1)



JAK1B


P13533
MYH6
Myosin-6 (Myosin heavy chain 6) (Myosin heavy chain,



MYHCA
cardiac muscle alpha isoform) (MyHC-alpha)


P61247
RPS3A
40S ribosomal protein S3a (Small ribosomal subunit



FTE1
protein eS1) (v-fos transformation effector protein) (Fte-



MFTL
1)


Q08379
GOLGA2
Golgin subfamily A member 2 (130 kDa cis-Golgi




matrix protein) (GM130) (GM130 autoantigen) (Golgin-




95)


P41219
PRPH
Peripherin (Neurofilament 4)



NEF4



PRPH1


Q99729
HNRNPAB
Heterogeneous nuclear ribonucleoprotein A/B (hnRNP



ABBP1
A/B) (APOBEC1-binding protein 1) (ABBP-1)



HNRPAB


P11277
SPTB
Spectrin beta chain, erythrocytic (Beta-I spectrin)



SPTB1


P33981
TTK MPS1
Dual specificity protein kinase TTK (EC 2.7.12.1)



MPS1L1
(Phosphotyrosine picked threonine-protein kinase) (PYT)


P11021
HSPA5
78 kDa glucose-regulated protein (GRP-78)



GRP78
(Endoplasmic reticulum lumenal Ca(2+)-binding protein




grp78) (Heat shock 70 kDa protein 5) (Immunoglobulin




heavy chain-binding protein) (BiP)


Q15552
tb protein
CACCC box-binding protein


P62913
RPL11
60S ribosomal protein L11 (CLL-associated antigen




KW-12) (Large ribosomal subunit protein uL5)


P38919
EIF4A3
Eukaryotic initiation factor 4A-III (eIF-4A-III) (eIF4A-



DDX48
III) (EC 3.6.4.13) (ATP-dependent RNA helicase



KIAA0111
DDX48) (ATP-dependent RNA helicase eIF4A-3)




(DEAD box protein 48) (Eukaryotic initiation factor 4A-




like NUK-34) (Eukaryotic translation initiation factor 4A




isoform 3) (Nuclear matrix protein 265) (NMP 265)




(hNMP 265) [Cleaved into: Eukaryotic initiation factor




4A-III, N-terminally processed]


Q12905
ILF2 NF45
Interleukin enhancer-binding factor 2 (Nuclear factor of



PRO3063
activated T-cells 45 kDa)


Q14978
NOLC1
Nucleolar and coiled-body phosphoprotein 1 (140 kDa



KIAA0035
nucleolar phosphoprotein) (Nopp140) (Hepatitis C virus



NS5ATP13
NS5A-transactivated protein 13) (HCV NS5A-




transactivated protein 13) (Nucleolar 130 kDa protein)




(Nucleolar phosphoprotein p130)


P20929
NEB
Nebulin


Q16296
4R-MAP2
Microtubule-associated protein (Fragment)


P33991
MCM4
DNA replication licensing factor MCM4 (EC 3.6.4.12)



CDC21
(CDC21 homolog) (P1-CDC21)


P49454
CENPF
Centromere protein F (CENP-F) (AH antigen)




(Kinetochore protein CENPF) (Mitosin)


Q14008
CKAP5
Cytoskeleton-associated protein 5 (Colonic and hepatic



KIAA0097
tumor overexpressed gene protein) (Ch-TOG)


Q14839
CHD4
Chromodomain-helicase-DNA-binding protein 4 (CHD-




4) (EC 3.6.4.12) (ATP-dependent helicase CHD4) (Mi-2




autoantigen 218 kDa protein) (Mi2-beta)


P55017
SLC12A3
Solute carrier family 12 member 3 (Na—Cl cotransporter)



NCC TSC
(NCC) (Na—Cl symporter) (Thiazide-sensitive sodium-




chloride cotransporter)


Q92835
INPP5D
Phosphatidylinositol 3,4,5-trisphosphate 5-phosphatase 1



SHIP SHIP1
(EC 3.1.3.86) (Inositol polyphosphate-5-phosphatase of




145 kDa) (SIP-145) (SH2 domain-containing inositol 5′-




phosphatase 1) (SH2 domain-containing inositol




phosphatase 1) (SHIP-1) (p150Ship) (hp51CN)


Q15269
PWP2
Periodic tryptophan protein 2 homolog



PWP2H


P20585
MSH3
DNA mismatch repair protein Msh3 (hMSH3)



DUC1 DUG
(Divergent upstream protein) (DUP) (Mismatch repair




protein 1) (MRP1)


Q05086
UBE3A
Ubiquitin-protein ligase E3A (EC 2.3.2.26) (E6AP



E6AP
ubiquitin-protein ligase) (HECT-type ubiquitin



EPVE6AP
transferase E3A) (Human papillomavirus E6-associated



HPVE6A
protein) (Oncogenic protein-associated protein E6-AP)




(Renal carcinoma antigen NY-REN-54)


Q92922
SMARCC1
SWI/SNF complex subunit SMARCC1 (BRG1-



BAF155
associated factor 155) (BAF155) (SWI/SNF complex




155 kDa subunit) (SWI/SNF-related matrix-associated




actin-dependent regulator of chromatin subfamily C




member 1)


P62807
HIST1H2BC
Histone H2B type 1-C/E/F/G/I (Histone H2B.1 A)



H2BFL;
(Histone H2B.a) (H2B/a) (Histone H2B.g) (H2B/g)



HIST1H2BE
(Histone H2B.h) (H2B/h) (Histone H2B.k) (H2B/k)



H2BFH;
(Histone H2B.l) (H2B/l)



HIST1H2BF



H2BFG;



HIST1H2BG



H2BFA;



HIST1H2BI



H2BFK


Q92800
EZH1
Histone-lysine N-methyltransferase EZH1 (EC 2.1.1.43)



KIAA0388
(ENX-2) (Enhancer of zeste homolog 1)


P78549
NTHL1
Endonuclease III-like protein 1 (hNTH1) (EC 3.2.2.—)



NTH1
(EC 4.2.99.18) (Bifunctional DNA N-glycosylase/DNA-



OCTS3
(apurinic or apyrimidinic site) lyase) (DNA




glycosylase/AP lyase)


Q12789
GTF3C1
General transcription factor 3C polypeptide 1 (TF3C-




alpha) (TFIIIC box B-binding subunit) (Transcription




factor IIIC 220 kDa subunit) (TFIIIC 220 kDa subunit)




(TFIIIC220) (Transcription factor IIIC subunit alpha)


O14686
KMT2D
Histone-lysine N-methyltransferase 2D (Lysine N-



ALR MLL2
methyltransferase 2D) (EC 2.1.1.43) (ALL1-related



MLL4
protein) (Myeloid/lymphoid or mixed-lineage leukemia




protein 2)


Q13304
GPR17
Uracil nucleotide/cysteinyl leukotriene receptor




(UDP/CysLT receptor) (G-protein coupled receptor 17)




(P2Y-like receptor) (R12)


Q9UQB3
CTNND2
Catenin delta-2 (Delta-catenin) (GT24) (Neural



NPRAP
plakophilin-related ARM-repeat protein) (NPRAP)




(Neurojungin)


P30519
HMOX2
Heme oxygenase 2 (HO-2) (EC 1.14.14.18)



HO2


O60437
PPL
Periplakin (190 kDa paraneoplastic pemphigus antigen)



KIAA0568
(195 kDa cornified envelope precursor protein)


Q15413
RYR3
Ryanodine receptor 3 (RYR-3) (RyR3) (Brain ryanodine



HBRR
receptor-calcium release channel) (Brain-type ryanodine




receptor) (Type 3 ryanodine receptor)


Q13618
CUL3
Cullin-3 (CUL-3)



KIAA0617


O75691
UTP20
Small subunit processome component 20 homolog



DRIM
(Down-regulated in metastasis protein) (Novel nucleolar




protein 73) (NNP73) (Protein Key-1A6)


O80743
T13D8.9
T13D8.9 protein


P38159
RBMX
RNA-binding motif protein, X chromosome



HNRPG
(Glycoprotein p43) (Heterogeneous nuclear



RBMXP1
ribonucleoprotein G) (hnRNP G) [Cleaved into: RNA-




binding motif protein, X chromosome, N-terminally




processed]


O75081
CBFA2T3
Protein CBFA2T3 (MTG8-related protein 2) (Myeloid



MTG16
translocation gene on chromosome 16 protein)



MTGR2
(hMTG16) (Zinc finger MYND domain-containing



ZMYND4
protein 4)


O95153
TSPOAP1
Peripheral-type benzodiazepine receptor-associated



BZRAP1
protein 1 (PRAX-1) (Peripheral benzodiazepine receptor-



KIAA0612
interacting protein) (PBR-IP) (RIMS-binding protein 1)



RBP1
(RIM-BP1) (TSPO-associated protein 1)



RIMBP1


P63267
ACTG2
Actin, gamma-enteric smooth muscle (Alpha-actin-3)



ACTA3
(Gamma-2-actin) (Smooth muscle gamma-actin)



ACTL3



ACTSG


P18754
RCC1
Regulator of chromosome condensation (Cell cycle



CHC1
regulatory protein) (Chromosome condensation protein




1)


Q5T081
RCC1
CHC1 protein (Regulator of chromosome condensation 1



CHC1
isoform 1) (Regulator of chromosome condensation 1,



hCG_27809
isoform CRA_b)


P13639
EEF2 EF2
Elongation factor 2 (EF-2)


Q16695
HIST3H3
Histone H3.1t (H3/t) (H3t) (H3/g)



H3FT


A8K401
PHB
Prohibitin, isoform CRA_a (cDNA FLJ78511, highly



hCG_29613
similar to Homo sapiens prohibitin (PHB), mRNA)




(cDNA, FLJ93035, Homo sapiens prohibitin (PHB),




mRNA)


P35232
PHB
Prohibitin


Q53FV0

Prohibitin variant (Fragment)


P83731
RPL24
60S ribosomal protein L24 (60S ribosomal protein L30)




(Large ribosomal subunit protein eL24)


V9HW01
HEL-S-310
Epididymis secretory protein Li 310


A0A024RCA7
RPLP2
Ribosomal protein, large, P2, isoform CRA_a



hCG_1778304


P05387
RPLP2
60S acidic ribosomal protein P2 (Large ribosomal



D11S2243E
subunit protein P2) (Renal carcinoma antigen NY-REN-



RPP2
44)


P46783
RPS10
40S ribosomal protein S10 (Small ribosomal subunit




protein eS10)


P62280
RPS11
40S ribosomal protein S11 (Small ribosomal subunit




protein uS17)


P62277
RPS13
40S ribosomal protein S13 (Small ribosomal subunit




protein uS15)


P08708
RPS17
40S ribosomal protein S17 (Small ribosomal subunit



RPS17L
protein eS17)


A8K517
RPS23
Ribosomal protein S23, isoform CRA_a (cDNA



hCG_38189
FLJ77921, highly similar to Homo sapiens ribosomal




protein S23 (RPS23), mRNA) (cDNA, FLJ92033, Homo sapiens




ribosomal protein S23 (RPS23), mRNA)


P62266
RPS23
40S ribosomal protein S23 (Small ribosomal subunit




protein uS12)


P62851
RPS25
40S ribosomal protein S25 (Small ribosomal subunit




protein eS25)


B2R491
RPS4X
40S ribosomal protein S4



hCG_18634


P62701
RPS4X
40S ribosomal protein S4, X isoform (SCR10) (Single



CCG2 RPS4
copy abundant mRNA protein) (Small ribosomal subunit



SCAR
protein eS4)


P62241
RPS8
40S ribosomal protein S8 (Small ribosomal subunit



OK/SW-
protein eS8)



cl.83


Q5JR94
RPS8
40S ribosomal protein S8



hCG_2031852


P12755
SKI
Ski oncogene (Proto-oncogene c-Ski)


A0A1L1UHR1

Sperm binding protein 1a


B3KTS5

cDNA FLJ38670 fis, clone HSYRA2000190, highly




similar to Voltage-dependent anion-selective channel




protein 1


P21796
VDAC1
Voltage-dependent anion-selective channel protein 1



VDAC
(VDAC-1) (hVDAC1) (Outer mitochondrial membrane




protein porin 1) (Plasmalemmal porin) (Porin 31HL)




(Porin 31HM)


P25490
YY1
Transcriptional represser protein YY1 (Delta



INO80S
transcription factor) (INO80 complex subunit S) (NF-E1)




(Yin and yang 1) (YY-1)


Q99996
AKAP9
A-kinase anchor protein 9 (AKAP-9) (A-kinase anchor



AKAP350
protein 350 kDa) (AKAP 350) (hgAKAP 350) (A-kinase



AKAP450
anchor protein 450 kDa) (AKAP 450) (AKAP 120-like



KIAA0803
protein) (Centrosome- and Golgi-localized PKN-




associated protein) (CG-NAP) (Protein hyperion)




(Protein kinase A-anchoring protein 9) (PRKA9) (Protein




yotiao)


P16402
HIST1H1D
Histone H1.3 (Histone H1c) (Histone H1s-2)



H1F3


Q96GY0
ZC2HC1A
Zinc finger C2HC domain-containing protein 1A



C8orf70



FAM164A



CGI-62


P02545
LMNA
Prelamin-A/C [Cleaved into: Lamin-A/C (70 kDa lamin)



LMN1
(Renal carcinoma antigen NY-REN-32)]


P20700
LMNB1
Lamin-B1



LMN2



LMNB


P14550
AKR1A1
Alcohol dehydrogenase [NADP(+)] (EC 1.1.1.2)



ALDR1
(Aldehyde reductase) (Aldo-keto reductase family 1



ALR
member A1)


V9HWI0
HEL-S-
Epididymis secretory protein Li 6 (Epididymis secretory



165mP
sperm binding protein Li 165 mP)



HEL-S-6


A0PJH2
ATP5H
ATP5H protein (Fragment)


O75947
ATP5H
ATP synthase subunit d, mitochondrial (ATPase subunit



My032
d)


P19105
MYL12A
Myosin regulatory light chain 12A (Epididymis secretory



MLCB
protein Li 24) (HEL-S-24) (MLC-2B) (Myosin RLC)



MRLC3
(Myosin regulatory light chain 2, nonsarcomeric)



RLC
(Myosin regulatory light chain MRLC3)


A0A0G2JS52

Uncharacterized protein (Fragment)


V9H0H3

Gag-Pro-Pol-Env protein


P17096
HMGA1
High mobility group protein HMG-I/HMG-Y (HMG-



HMGIY
I(Y)) (High mobility group AT-hook protein 1) (High




mobility group protein A1) (High mobility group protein




R)


O46577
COX4I1
Cytochrome c oxidase subunit 4 isoform 1,



COX4
mitochondrial (Cytochrome c oxidase polypeptide IV)




(Cytochrome c oxidase subunit IV isoform 1) (COX IV-




1) (Fragment)


Q9UIG0
BAZ1B
Tyrosine-protein kinase BAZ1B (EC 2.7.10.2)



WBSC10
(Bromodomain adjacent to zinc finger domain protein



WBSCR10
1B) (Williams syndrome transcription factor) (Williams-



WBSCR9
Beuren syndrome chromosomal region 10 protein)



WSTF
(Williams-Beuren syndrome chromosomal region 9




protein) (hWALp2)


Q9UHD8
SEPT9
Septin-9 (MLL septin-like fusion protein MSF-A) (MLL



KIAA0991
septin-like fusion protein) (Ovarian/Breast septin) (Ov/Br



MSF
septin) (Septin D1)


P62270
Rps18
40S ribosomal protein S18 (Ke-3) (Ke3)


Q561N5
Rps18
MCG23000, isoform CRA_b (Putative uncharacterized



mCG_23000
protein) (Ribosomal protein S18)


Q9NWS8
RMND1
Required for meiotic nuclear division protein 1 homolog



C6orf96


P31942
HNRNPH3
Heterogeneous nuclear ribonucleoprotein H3 (hnRNP



HNRPH3
H3) (Heterogeneous nuclear ribonucleoprotein 2H9)




(hnRNP 2H9)


Q9NZR2
LRP1B
Low-density lipoprotein receptor-related protein 1B



LRPDIT
(LRP-1B) (Low-density lipoprotein receptor-related




protein-deleted in tumor) (LRP-DIT)


Q16891
IMMT
MICOS complex subunit MIC60 (Cell proliferation-



HMP
inducing gene 4/52 protein) (Mitochondrial inner



MIC60
membrane protein) (Mitofilin) (p87/89)



MINOS2



PIG4 PIG52


A4D1N4
CHCHD3
MICOS complex subunit



hCG_2014841



tcag7.1158


Q9NX63
CHCHD3
MICOS complex subunit MIC19 (Coiled-coil-helix-



MIC19
coiled-coil-helix domain-containing protein 3)



MINOS3


Q6NTF9
RHBDD2
Rhomboid domain-containing protein 2



RHBDL7


Q6P1M9
ARMCX5
Armadillo repeat-containing X-linked protein 5


O00148
DDX39A
ATP-dependent RNA helicase DDX39A (EC 3.6.4.13)



DDX39
(DEAD box protein 39) (Nuclear RNA helicase URH49)


Q6UY01
LRRC31
Leucine-rich repeat-containing protein 31



UNQ9367/



PRO34156


Q8IYT3
CCDC170
Coiled-coil domain-containing protein 170



C6orf97


Q2L6I2
ABCF1
ABC50 protein (ATP-binding cassette, sub-family F



ABC50
(GCN20), member 1) (ATP-binding cassette, sub-family



hCG_26012
F (GCN20), member 1, isoform CRA_a)


Q8NE71
ABCF1
ATP-binding cassette sub-family F member 1 (ATP-



ABC50
binding cassette 50) (TNF-alpha-stimulated ABC




protein)


Q99459
CDC5L
Cell division cycle 5-like protein (Cdc5-like protein)



KIAA0432
(Pombe cdc5-related protein)



PCDC5RP


P35580
MYH10
Myosin-10 (Cellular myosin heavy chain, type B)




(Myosin heavy chain 10) (Myosin heavy chain, non-




muscle IIb) (Non-muscle myosin heavy chain B)




(NMMHC-B) (Non-muscle myosin heavy chain IIb)




(NMMHC II-b) (NMMHC-IIB)


P50914
RPL14
60S ribosomal protein L14 (CAG-ISL 7) (Large




ribosomal subunit protein eL14)


Q9C093
SPEF2
Sperm flagellar protein 2 (Protein KPL2)



KIAA1770



KPL2


P08729
KRT7 SCL
Keratin, type II cytoskeletal 7 (Cytokeratin-7) (CK-7)




(Keratin-7) (K7) (Sarcolectin) (Type-II keratin Kb7)


Q9BTQ7

Similar to ribosomal protein L23 (Fragment)


Q96RT7
TUBGCP6
Gamma-tubulin complex component 6 (GCP-6)



GCP6



KIAA1669


Q5M8Q0
Rpl15
Ribosomal protein L15



mCG_10029


Q9CZM2
Rpl15
60S ribosomal protein L15


Q9BS75
KLHL20
KLHL20 protein (Kelch-like 20 (Drosophila), isoform



hCG_23698
CRA_a)


P82970
HMGN5
High mobility group nucleosome-binding domain-



NSBP1
containing protein 5 (Nucleosome-binding protein 1)


A0A024QZW2
NOL7
Nucleolar protein 7, 27 kDa, isoform CRA_a



hCG_37417


Q9UMY1
NOL7
Nucleolar protein 7 (Nucleolar protein of 27 kDa)



C6orf90



NOP27


P62907
Rpl10a
60S ribosomal protein L10a


P78527
PRKDC
DNA-dependent protein kinase catalytic subunit (DNA-



HYRC
PK catalytic subunit) (DNA-PKcs) (EC 2.7.11.1)



HYRC1
(DNPK1) (p460)


B4E1W3

cDNA FLJ51732, highly similar to Peroxisomal NADH




pyrophosphatase NUDT12 (EC 3.6.1.22)


Q9BQG2
NUDT12
Peroxisomal NADH pyrophosphatase NUDT12 (EC




3.6.1.22) (Nucleoside diphosphate-linked moiety X motif




12) (Nudix motif 12)


P46779
RPL28
60S ribosomal protein L28 (Large ribosomal subunit




protein eL28)


P22626
HNRNPA2B1
Heterogeneous nuclear ribonucleoproteins A2/B1



HNRPA2B1
(hnRNP A2/B1)


Q96Q15
SMG1 ATX
Serine/threonine-protein kinase SMG1 (SMG-1) (hSMG-



KIAA0421
1) (EC 2.7.11.1) (61E3.4) (Lambda/iota protein kinase C-



LIP
interacting protein) (Lambda-interacting protein)


A0A024R4M0
RPS9
40S ribosomal protein S9 (Ribosomal protein S9,



hCG_2009111
isoform CRA_a)


P46781
RPS9
40S ribosomal protein S9 (Small ribosomal subunit




protein uS4)


Q96T23
RSF1
Remodeling and spacing factor 1 (Rsf-1) (HBV pX-



HBXAP
associated protein 8) (Hepatitis B virus X-associated



XAP8
protein) (p325 subunit of RSF chromatin-remodeling




complex)


P60709
ACTB
Actin, cytoplasmic 1 (Beta-actin) [Cleaved into: Actin,




cytoplasmic 1,N-terminally processed]


Q96RL1
UIMC1
BRCA1-A complex subunit RAP80 (Receptor-associated



RAP80
protein 80) (Retinoid X receptor-interacting protein 110)



RXRIP110
(Ubiquitin interaction motif-containing protein 1)


Q96A11
GAL3ST3
Galactose-3-O-sulfotransferase 3 (Gal3ST-3) (EC 2.8.2.—)




(Beta-galactose-3-O-sulfotransferase 3) (Gal3ST3)




(Gal-beta-1, 3-GalNAc 3′-sulfotransferase 3)


P62847
RPS24
40S ribosomal protein S24 (Small ribosomal subunit




protein eS24)


Q9NSI6
BRWD1
Bromodomain and WD repeat-containing protein 1 (WD



C21orf107
repeat-containing protein 9)



WDR9


A0A024R1X8
JUP
Junction plakoglobin, isoform CRA_a



hCG_1771506


Q96QZ7
MAGI1
Membrane-associated guanylate kinase, WW and PDZ



AIP3
domain-containing protein 1 (Atrophin-1-interacting



BAIAP1
protein 3) (AIP-3) (BAI1-associated protein 1) (BAP-1)



BAP1
(Membrane-associated guanylate kinase inverted 1)



TNRC19
(MAGI-1) (Trinucleotide repeat-containing gene 19




protein) (WW domain-containing protein 3) (WWP3)


A8K4C8
RPL13
60S ribosomal protein L13



hCG_1723872


P26373
RPL13
60S ribosomal protein L13 (Breast basic conserved



BBC1
protein 1) (Large ribosomal subunit protein eL13)



OK/SW-



cl.46


P46019
PHKA2
Phosphorylase b kinase regulatory subunit alpha, liver



PHKLA
isoform (Phosphorylase kinase alpha L subunit)



PYK


O60506
SYNCRIP
Heterogeneous nuclear ribonucleoprotein Q (hnRNP Q)



HNRPQ
(Glycine- and tyrosine-rich RNA-binding protein) (GRY-



NSAP1
RBP) (NS1-associated protein 1) (Synaptotagmin-




binding, cytoplasmic RNA-interacting protein)


Q96Q42
ALS2
Alsin (Amyotrophic lateral sclerosis 2 chromosomal



ALS2CR6
region candidate gene 6 protein) (Amyotrophic lateral



KIAA1563
sclerosis 2 protein)


Q8IYJ3
SYTL1
Synaptotagmin-like protein 1 (Exophilin-7) (Protein



SLP1
JFC1)



SB146


A0A024RDH8
RPL34
Ribosomal protein L34, isoform CRA_a



hCG_2027853


P49207
RPL34
60S ribosomal protein L34 (Large ribosomal subunit




protein eL34)


Q9P2M7
CGN
Cingulin



KIAA1319


Q96BT3
CENPT
Centromere protein T (CENP-T) (Interphase centromere



C16orf56
complex protein 22)



ICEN22


Q0VF96
CGNL1
Cingulin-like protein 1 (Junction-associated coiled-coil



JACOP
protein) (Paracingulin)



KIAA1749


Q96M95
CCDC42
Coiled-coil domain-containing protein 42



CCDC42A


P52597
HNRNPF
Heterogeneous nuclear ribonucleoprotein F (hnRNP F)



HNRPF
(Nucleolin-like protein mcs94-1) [Cleaved into:




Heterogeneous nuclear ribonucleoprotein F, N-terminally




processed]


O96008
TOMM40
Mitochondrial import receptor subunit TOM40 homolog



C19orf1
(Protein Haymaker) (Translocase of outer membrane 40



PEREC1
kDa subunit homolog) (p38.5)



TOM40


Q96BS4
FBL
FBL protein (Putative uncharacterized protein)




(Fragment)


Q9H501
ESF1
ESF1 homolog (ABT1-associated protein)



ABTAP



C20orf6



HDCMC28P


Q6PHZ2
Camk2d
Calcium/calmodulin-dependent protein kinase type II



Kiaa4163
subunit delta (CaM kinase II subunit delta) (CaMK-II




subunit delta) (EC 2.7.11.17)


Q07020
RPL18
60S ribosomal protein L18 (Large ribosomal subunit




protein eL18)


Q8TF72
SHROOM3
Protein Shroom3 (Shroom-related protein) (hShrmL)



KIAA1481



SHRML



MSTP013


Q8TE73
DNAH5
Dynein heavy chain 5, axonemal (Axonemal beta dynein



DNAHC5
heavy chain 5) (Ciliary dynein heavy chain 5)



HL1



KIAA1603


O75475
PSIP1
PC4 and SFRS1-interacting protein (CLL-associated



DFS70
antigen KW-7) (Dense fine speckles 70 kDa protein)



LEDGF
(DPS 70) (Lens epithelium-derived growth factor)



PSIP2
(Transcriptional coactivator p75/p52)


E9KL44

Epididymis tissue sperm binding protein Li 14m


P40939
HADHA
Trifunctional enzyme subunit alpha, mitochondrial (78



HADH
kDa gastrin-binding protein) (TP-alpha) [Includes: Long-




chain enoyl-CoA hydratase (EC 4.2.1.17); Long chain 3-




hydroxyacyl-CoA dehydrogenase (EC 1.1.1.211)]


Q9HB09
BCL2L12
Bcl-2-like protein 12 (Bcl2-L-12) (Bcl-2-related proline-



BPR
rich protein)


O75367
H2AFY
Core histone macro-H2A.1 (Histone macroH2A1)



MACROH2A1
(mH2A1) (Histone H2A.y) (H2A/y) (Medulloblastoma




antigen MU-MB-50.205)


Q8N6Z2
MTRF1
MTRF1 protein (Mitochondrial translational release



hCG_32761
factor 1, isoform CRA_b) (Peptide chain release factor 1,




mitochondrial)


Q8TCU4
ALMS1
Alstrom syndrome protein 1



KIAA0328


A0JNW5
UHRF1BP1L
UHRF1-binding protein 1-like



KIAA0701


O75643
SNRNP200
U5 small nuclear ribonucleoprotein 200 kDa helicase



ASCC3L1
(EC 3.6.4.13) (Activating signal cointegrator 1 complex



HELIC2
subunit 3-like 1) (BRR2 homolog) (U5 snRNP-specific



KIAA0788
200 kDa protein) (U5-200KD)


A7E2E1
SMARCA4
SWI/SNF related, matrix associated, actin dependent



hCG_29955
regulator of chromatin, subfamily a, member 4




(SWI/SNF related, matrix associated, actin dependent




regulator of chromatin, subfamily a, member 4, isoform




CRA_a) (cDNA FLJ77531, highly similar to Homo sapiens




SWI/SNF related, matrix associated, actin




dependent regulator of chromatin, subfamily a, member 4




(SMARCA4), mRNA)


P51532
SMARCA4
Transcription activator BRG1 (EC 3.6.4.—) (ATP-



BAF190A
dependent helicase SMARCA4) (BRG1-associated factor



BRG1
190A) (BAF190A) (Mitotic growth and transcription



SNF2B
activator) (Protein BRG-1) (Protein brahma homolog 1)



SNF2L4
(SNF2-beta) (SWI/SNF-related matrix-associated actin-




dependent regulator of chromatin subfamily A member




4)


O00418
EEF2K
Eukaryotic elongation factor 2 kinase (eEF-2 kinase)




(eEF-2K) (EC 2.7.11.20) (Calcium/calmodulin-




dependent eukaryotic elongation factor 2 kinase)


Q96CN4
EVI5L
EVI5-like protein (Ecotropic viral integration site 5-like




protein)


Q9H8V3
ECT2
Protein ECT2 (Epithelial cell-transforming sequence 2




oncogene)


Q5T3F8
TMEM63B
CSC1-like protein 2 (Transmembrane protein 63B)



C6orf110


Q8NAJ6

cDNA FLJ35251 fis, clone PROST2003635, weakly




similar to MULTIFUNCTIONAL AMINOACYL-TRNA




SYNTHETASE


A0A0C4DG40
SYNE1
Nesprin-1


Q8NF91
SYNE1
Nesprin-1 (Enaptin) (KASH domain-containing protein



C6orf98
1) (KASH1) (Myocyte nuclear envelope protein 1)



KIAA0796
(Myne-1) (Nuclear envelope spectrin repeat protein 1)



KIAA1262
(Synaptic nuclear envelope protein 1) (Syne-1)



KIAA1756



MYNE1


Q8TDI0
CHD5
Chromodomain-helicase-DNA-binding protein 5 (CHD-



KIAA0444
5) (EC 3.6.4.12) (ATP-dependent helicase CHD5)


Q9NU22
MDN1
Midasin (MIDAS-containing protein)



KIAA0301


Q8WXH0
SYNE2
Nesprin-2 (KASH domain-containing protein 2)



KIAA1011
(KASH2) (Nuclear envelope spectrin repeat protein 2)



NUA
(Nucleus and actin connecting element protein) (Protein




NUANCE) (Synaptic nuclear envelope protein 2) (Syne-




2)


Q9Y277
VDAC3
Voltage-dependent anion-selective channel protein 3




(VDAC-3) (hVDAC3) (Outer mitochondrial membrane




protein porin 3)


Q96QE3
ATAD5
ATPase family AAA domain-containing protein 5



C17orf41
(Chromosome fragility-associated gene 1 protein)



FRAG1


Q9BXJ9
NAA15
N-alpha-acetyltransferase 15, NatA auxiliary subunit



GA19
(Gastric cancer antigen Ga19) (N-terminal



NARG1
acetyltransferase) (NMDA receptor-regulated protein 1)



NATH
(Protein tubedown-1) (Tbdn100)



TBDN100


Q8IUE6
HIST2H2AB
Histone H2A type 2-B


Q5TZA2
CROCC
Rootletin (Ciliary rootlet coiled-coil protein)



KIAA0445


A0A024RAS2
H2AFJ
Histone H2A



hCG_1639762


Q9BTM1
H2AFJ
Histone H2A.J (H2a/j)


Q8NEN9
PDZD8
PDZ domain-containing protein 8 (Sarcoma antigen NY-



PDZK8
SAR-84/NY-SAR-104)


Q14683
SMC1A
Structural maintenance of chromosomes protein 1A



DXS423E
(SMC protein 1A) (SMC-1-alpha) (SMC-1A) (Sb1.8)



KIAA0178



SB1.8



SMC1



SMC1L1


Q68EN4
SMC1A
SMC1A protein (Fragment)


Q7Z7G8
VPS13B
Vacuolar protein sorting-associated protein 13B (Cohen



CHS1
syndrome protein 1)



COH1



KIAA0532


Q7Z7A1
CNTRL
Centriolin (Centrosomal protein 1) (Centrosomal protein



CEP1
of 110 kDa) (Cep110)



CEP110


O95613
PCNT
Pericentrin (Kendrin) (Pericentrin-B)



KIAA0402



PCNT2


A0A140VK14

Testicular secretory protein Li 14


P49448
GLUD2
Glutamate dehydrogenase 2, mitochondrial (GDH 2) (EC



GLUDP1
1.4.1.3)


Q5VTT5
MYOM3
Myomesin-3 (Myomesin family member 3)


Q7Z612

Acidic ribosomal phosphoprotein P1


O00567
NOP56
Nucleolar protein 56 (Nucleolar protein 5A)



NOL5A


Q9Y2X3
NOP58
Nucleolar protein 58 (Nucleolar protein 5)



NOL5



NOP5



HSPC120


A0A0C4DFX4

Uncharacterized protein (Fragment)


Q6ZNL4
FLJ00279
FLJ00279 protein (Fragment)


Q6ZWK7

cDNA FLJ16045 fis, clone CTONG2000042, weakly




similar to ALPHA-2-MACROGLOBULIN


Q7Z388
DPY19L4
Probable C-mannosyltransferase DPY19L4 (EC 2.4.1.—)




(Dpy-19-like protein 4) (Protein dpy-19 homolog 4)


Q5T9S5
CCDC18
Coiled-coil domain-containing protein 18 (Sarcoma




antigen NY-SAR-24)


Q6ZV73
FGD6
FYVE, RhoGEF and PH domain-containing protein 6



KIAA1362
(Zinc finger FYVE domain-containing protein 24)



ZFYVE24


P25705
ATP5A1
ATP synthase subunit alpha, mitochondrial



ATP5A



ATP5AL2



ATPM


P42285
SKIV2L2
Superkiller viralicidic activity 2-like 2 (EC 3.6.4.13)



DOB1
(ATP-dependent RNA helicase DOB1) (ATP-dependent



KIAA0052
RNA helicase SKIV2L2) (TRAMP-like complex



Mtr4
helicase)


Q00325
SLC25A3
Phosphate carrier protein, mitochondrial (Phosphate



PHC
transport protein) (PTP) (Solute carrier family 25



OK/SW-
member 3)



cl.48


P62753
RPS6
40S ribosomal protein S6 (Phosphoprotein NP33) (Small



OK/SW-cl.2
ribosomal subunit protein eS6)


Q9BW34
EEF1D
EEF1D protein (Fragment)


Q5K651
SAMD9
Sterile alpha motif domain-containing protein 9 (SAM



C7orf5
domain-containing protein 9)



DRIF1



KIAA2004



OEF1


Q6W6M6

Antigen MLAA-44


Q5T0F9
CC2D1B
Coiled-coil and C2 domain-containing protein 1B (Five



KIAA1836
prime represser element under dual repression-binding




protein 2) (FRE under dual repression-binding protein 2)




(Freud-2)


P26641
EEF1G
Elongation factor 1-gamma (EF-1-gamma) (eEF-1B



EF1G
gamma)



PRO1608


Q00839
HNRNPU
Heterogeneous nuclear ribonucleoprotein U (hnRNP U)



HNRPU
(Scaffold attachment factor A) (SAF-A) (p120) (pp120)



SAFA



U21.1


Q9Y4C4
MFHAS1
Malignant fibrous histiocytoma-amplified sequence 1



MASL1
(Malignant fibrous histiocytoma-amplified sequence with




leucine-rich tandem repeats 1)


P16050
ALOX15
Arachidonate 15-lipoxygenase (15-LOX) (15-LOX-1)



LOG15
(EC 1.13.11.33) (12/15-lipoxygenase) (Arachidonate 12-




lipoxygenase, leukocyte-type) (12-LOX) (EC 1.13.11.31)




(Arachidonate omega-6 lipoxygenase)


P16383
GCFC2
GC-rich sequence DNA-binding factor 2 (GC-rich



C2orf3 GCF
sequence DNA-binding factor) (Transcription factor 9)



TCF9
(TCF-9)


P36578
RPL4 RPL1
60S ribosomal protein L4 (60S ribosomal protein L1)




(Large ribosomal subunit protein uL4)


O76081
RGS20
Regulator of G-protein signaling 20 (RGS20) (Gz-



RGSZ1
selective GTPase-activating protein) (G(z)GAP) (Gz-



ZGAP1
GAP) (Regulator of G-protein signaling Z1) (Regulator




of Gz-selective protein signaling 1)


Q9Y6N9
USH1C
Harmonin (Antigen NY-CO-38/NY-CO-37)



AIE75
(Autoimmune enteropathy-related antigen AIE-75)




(Protein PDZ-73) (Renal carcinoma antigen NY-REN-3)




(Usher syndrome type-1C protein)


Q15149
PLEC
Plectin (PCN) (PLTN) (Hemidesmosomal protein 1)



PLEC1
(HD1) (Plectin-1)


O60333
KIF1B
Kinesin-like protein KIF1B (Klp)



KIAA0591



KIAA1448


O60462
NRP2
Neuropilin-2 (Vascular endothelial cell growth factor



VEGF165R2
165 receptor 2)


Q7Z3T9
DKFZp686J1169
Neuropilin


Q5THJ4
VPS13D
Vacuolar protein sorting-associated protein 13D



KIAA0453


Q9NRZ9
HELLS
Lymphoid-specific helicase (EC 3.6.4.—) (Proliferation-



PASG
associated SNF2-like protein) (SWI/SNF2-related



SMARCA6
matrix-associated actin-dependent regulator of chromatin



Nbla10143
subfamily A member 6)


Q96A08
HIST1H2BA
Histone H2B type 1-A (Histone H2B, testis) (TSH2B.1)



TSH2B
(hTSH2B) (Testis-specific histone H2B)


Q6UB99
ANKRD11
Ankyrin repeat domain-containing protein 11 (Ankyrin



ANCO1
repeat-containing cofactor 1)


I6L9F7
HIST1H2BM
Histone H2B (Fragment)


P02538
KRT6A
Keratin, type II cytoskeletal 6A (Cytokeratin-6A) (CK-



K6A
6A) (Cytokeratin-6D) (CK-6D) (Keratin-6A) (K6A)



KRT6D
(Type-II keratin Kb6) (allergen Horn s 5)


Q6KC79
NIPBL
Nipped-B-like protein (Delangin) (SCC2 homolog)



IDN3


Q8NBU5
ATAD1
ATPase family AAA domain-containing protein 1 (EC



FNP001
3.6.1.3) (Thorase)


E5KLM2

Mitochondrial dynamin-like 120 kDa protein


Q15772
SPEG
Striated muscle preferentially expressed protein kinase



APEG1
(EC 2.7.11.1) (Aortic preferentially expressed protein 1)



KIAA1297
(APEG-1)


O14490
DLGAP1
Disks large-associated protein 1 (DAP-1) (Guanylate



DAP1
kinase-associated protein) (hGKAP) (PSD-95/SAP90-



GKAP
binding protein 1) (SAP90/PSD-95-associated protein 1)




(SAPAP1)


Q5JSL3
DOCK11
Dedicator of cytokinesis protein 11 (Activated Cdc42-



ZIZ2
associated guanine nucleotide exchange factor) (ACG)




(Zizimin-2)


Q5VU43
PDE4DIP
Myomegalin (Cardiomyopathy-associated protein 2)



CMYA2
(Phosphodiesterase 4D-interacting protein)



KIAA0454



KIAA0477



MMGL


Q658X5
DKFZp666F1010
Putative uncharacterized protein DKFZp666F1010




(Fragment)


Q658W4
DKFZp666M0710
Putative uncharacterized protein DKFZp666M0710




(Fragment)


Q63HR1
DKFZp686P17171
Putative uncharacterized protein DKFZp686P17171


Q5VWT5
ARAP
Activation-dependent, raft-recruited ADAP-like



C1orf168
phosphoprotein


Q92614
MYO18A
Unconventional myosin-XVIIIa (Molecule associated



CD245
with JAK3 N-terminus) (MAJN) (Myosin containing a



KIAA0216
PDZ domain) (Surfactant protein receptor SP-R210) (SP-



MYSPDZ
R210)


A0A024R4A0
NCL
Nucleolin, isoform CRA_b



hCG_33980


B3KM80
NCL
Nucleolin, isoform CRA_c (cDNA FLJ10452 fis, clone



hCG_33980
NT2RP1000966, highly similar to NUCLEOLIN)


P19338
NCL
Nucleolin (Protein C23)


P35527
KRT9
Keratin, type I cytoskeletal 9 (Cytokeratin-9) (CK-9)




(Keratin-9) (K9)


Q5T655
CFAP58
Cilia- and flagella-associated protein 58 (Coiled-coil



C10orf80
domain-containing protein 147)



CCDC147


Q5TAX3
ZCCHC11
Terminal uridylyltransferase 4 (TUTase 4) (EC 2.7.7.52)



KIAA0191
(Zinc finger CCHC domain-containing protein 11)



TUT4


Q9Y6I7
WSB1
WD repeat and SOCS box-containing protein 1 (WSB-1)



SWIP1
(SOCS box-containing WD protein SWiP-1)


Q9HC77
CENPJ
Centromere protein J (CENP-J) (Centrosomal P4.1-



CPAP LAP
associated protein) (LAG-3-associated protein) (LYST-



LIP1
interacting protein 1)


Q5H8C1
FREM1
FRAS1-related extracellular matrix protein 1 (Protein



C9orf143
QBRICK)



C9orf145



C9orf154


Q8N5G2
TMEM57
Macoilin (Transmembrane protein 57)


Q58F05
NARG1
NARG1 protein (Fragment)


Q59HE3

Calpastatin isoform a variant (Fragment)


Q59GX9

Ribosomal protein L5 variant (Fragment)


Q59FF1

Insulin-like growth factor binding protein 2 variant




(Fragment)


O75116
ROCK2
Rho-associated protein kinase 2 (EC 2.7.11.1) (Rho



KIAA0619
kinase 2) (Rho-associated, coiled-coil-containing protein




kinase 2) (Rho-associated, coiled-coil-containing protein




kinase II) (ROCK-II) (p164 ROCK-2)


Q53HW2

60S acidic ribosomal protein P0 (Fragment)


Q53HR5

Elongation factor 1-alpha (Fragment)


P14136
GFAP
Glial fibrillary acidic protein (GFAP)


Q562R1
ACTBL2
Beta-actin-like protein 2 (Kappa-actin)


A0A024R2G2
FANCD2
Fanconi anemia, complementation group D2, isoform



hCG_1811443
CRA_b


Q9BXW9
FANCD2
Fanconi anemia group D2 protein (Protein FACD2)



FACD


Q86XH1
IQCA1
IQ and AAA domain-containing protein 1



IQCA


A1XBS5
FAM92A
Protein FAM92A



FAM92A1


Q9P273
TENM3
Teneurin-3 (Ten-3) (Protein Odd Oz/ten-m homolog 3)



KIAA1455
(Tenascin-M3) (Ten-m3) (Teneurin transmembrane



ODZ3
protein 3)



TNM3


Q9P2K1
CC2D2A
Coiled-coil and C2 domain-containing protein 2A



KIAA1345


Q96BT1
C3orf49
Putative uncharacterized protein C3orf49


P09651
HNRNPA1
Heterogeneous nuclear ribonucleoprotein A1 (hnRNP



HNRPA1
A1) (Helix-destabilizing protein) (Single-strand RNA-




binding protein) (hnRNP core protein A1) [Cleaved into:




Heterogeneous nuclear ribonucleoprotein A1, N-




terminally processed]


Q9P225
DNAH2
Dynein heavy chain 2, axonemal (Axonemal beta dynein



DNAHC2
heavy chain 2) (Ciliary dynein heavy chain 2) (Dynein



DNHD3
heavy chain domain-containing protein 3)



KIAA1503


Q4KM60
Rpl10a
Ribosomal protein (Fragment)



Serpina6


Q32Q62
RSL1D1
RSL1D1 protein (Fragment)


Q9H611
PIF1
ATP-dependent DNA helicase PIF1 (EC 3.6.4.12) (DNA



C15orf20
repair and recombination helicase PIF1) (PIF1/RRM3




DNA helicase-like protein)


Q86Y46
KRT73
Keratin, type II cytoskeletal 73 (Cytokeratin-73) (CK-73)



K6IRS3
(Keratin-73) (K73) (Type II inner root sheath-specific



KB36
keratin-K6irs3) (Type-II keratin Kb36)



KRT6IRS3


Q0QEN7
ATP5B
ATP synthase subunit beta (EC 3.6.3.14) (Fragment)


B3KU66

cDNA FLJ39263 fis, clone OCBBF2009571, highly




similar to ATP-dependent RNA helicase A (EC 3.6.1.—)


Q08211
DHX9
ATP-dependent RNA helicase A (RHA) (EC 3.6.4.13)



DDX9 LKP
(DEAH box protein 9) (Leukophysin) (LKP) (Nuclear



NDH2
DNA helicase II) (NDH II)


O15078
CEP290
Centrosomal protein of 290 kDa (Cep290) (Bardet-Biedl



BBS14
syndrome 14 protein) (Cancer/testis antigen 87) (CT87)



KIAA0373
(Nephrocystin-6) (Tumor antigen se2-2)



NPHP6


Q05BJ6
CEP290
CEP290 protein


Q92538
GBF1
Golgi-specific brefeldin A-resistance guanine nucleotide



KIAA0248
exchange factor 1 (BFA-resistant GEF 1)


Q4G0J3
LARP7
La-related protein 7 (La ribonucleoprotein domain family



HDCMA18P
member 7) (P-TEFb-interaction protein for 7SK stability)




(PIP7S)


Q15397
PUM3
Pumilio homolog 3 (HBV X-transactivated gene 5



cPERP-C
protein) (HBV XAg-transactivated protein 5) (Minor



KIAA0020
histocompatibility antigen HA-8) (HLA-HA8)



PUF-A



XTP5


Q7RTY7
OVCH1
Ovochymase-1 (EC 3.4.21.—)


Q5SPB7
ino80
INO80 complex subunit



si:ch211-



244p18.3


Q9Y3V2
RWDD3
RWD domain-containing protein 3 (RWD domain-



RSUME
containing sumoylation enhancer) (RSUME)


Q9HCR9
PDE11A
Dual 3′,5′-cyclic-AMP and -GMP phosphodiesterase 11A




(EC 3.1.4.35) (EC 3.1.4.53) (cAMP and cGMP




phosphodiesterase 11A)


Q9NR48
ASH1L
Histone-lysine N-methyltransferase ASH1L (EC



KIAA1420
2.1.1.43) (ASH1-like protein) (huASH1) (Absent small



KMT2H
and homeotic disks protein 1 homolog) (Lysine N-




methyltransferase 2H)


Q09428
ABCC8
ATP-binding cassette sub-family C member 8



HRINS
(Sulfonylurea receptor 1)



SUR SUR1


Q5JU67
CFAP157
Cilia- and flagella-associated protein 157



C9orf117


D3DR32
MPHOSPH1
M-phase phosphoprotein 1, isoform CRA_a



hCG_23744


G5E9G0
RPL3 ASC-1
60S ribosomal protein L3 (Ribosomal protein L3,



hCG_2015191
isoform CRA_e)


D3DS91
AKAP6
A kinase (PRKA) anchor protein 6, isoform CRA_b



hCG_1812123


A0A0A7M1X5
LMNB2
Lamin B2, isoform CRA_b (Lamin B3)



hCG_2004338


A0A024R5M9
NUMA1
Nuclear mitotic apparatus protein 1, isoform CRA_a



hCG_2017131


Q4G0X9
CCDC40
Coiled-coil domain-containing protein 40



KIAA1640


D3DTT5
TBKBP1
TBK1 binding protein 1, isoform CRA_a



hCG_1813987


G5E972
TMPO
Lamina-associated polypeptide 2, isoforms beta/gamma



hCG_2015322
(Thymopoietin, isoform CRA_d)


D6W5D1
KIAA1212
KIAA1212, isoform CRA_a



hCG_1817741


U3KQK0
HIST1H2BN
Histone H2B



hCG_1743059


D6RGI3
SEPT11
Septin 11, isoform CRA_b (Septin-11)



hCG_24410


B4DDB6
HNRPA3
Heterogeneous nuclear ribonucleoprotein A3, isoform



hCG_2005824
CRA_a (cDNA FLJ52659, highly similar to




Heterogeneous nuclear ribonucleoprotein A3) (cDNA,




FLJ79333, highly similar to Heterogeneous nuclear




ribonucleoprotein A3)


Q8TE76
MORC4
MORC family CW-type zinc finger protein 4 (Zinc



ZCW4
finger CW-type coiled-coil domain protein 2) (Zinc



ZCWCC2
finger CW-type domain protein 4)


Q8NCM8
DYNC2H1
Cytoplasmic dynein 2 heavy chain 1 (Cytoplasmic



DHC1B
dynein 2 heavy chain) (Dynein Cytoplasmic heavy chain



DHC2
2) (Dynein heavy chain 11) (hDHC11) (Dynein heavy



DNCH2
chain isotype 1B)



DYH1B



KIAA1997


Q6PIF6
MYO7B
Unconventional myosin-VIIb


Q8NB66
UNC13C
Protein unc-13 homolog C (Munc13-3)


A0A1U9X7W7

HSPA1L


P34931
HSPA1L
Heat shock 70 kDa protein 1-like (Heat shock 70 kDa




protein 1L) (Heat shock 70 kDa protein 1-Hom) (HSP70-




Hom)


A4D0S4
LAMB4
Laminin subunit beta-4 (Laminin beta-1-related protein)


Q8N309
LRRC43
Leucine-rich repeat-containing protein 43


Q8TDW7
FAT3
Protocadherin Fat 3 (hFat3) (Cadherin family member



CDHF15
15) (FAT tumor suppressor homolog 3)



KIAA1989


A5WVL9
dapk1
Death-associated protein kinase (Death-associated



si:ch211-
protein kinase 1)



66i11.1


P05141
SLC25A5
ADP/ATP translocase 2 (ADP, ATP carrier protein 2)



ANT2
(ADP, ATP carrier protein, fibroblast isoform) (Adenine




nucleotide translocate 2) (ANT 2) (Solute carrier family




25 member 5) [Cleaved into: ADP/ATP translocase 2, N-




terminally processed]


Q6NVC0
SLC25A5
SLC25A5 protein (Fragment)


P12236
SLC25A6
ADP/ATP translocase 3 (ADP, ATP carrier protein 3)



ANT3
(ADP, ATP carrier protein, isoform T2) (ANT 2)



CDABP0051
(Adenine nucleotide translocator 3) (ANT 3) (Solute




carrier family 25 member 6) [Cleaved into: ADP/ATP




translocase 3, N-terminally processed]


Q6I9V5
SLC25A6
SLC25A6 protein (Solute carrier family 25



hCG_1746794
(Mitochondrial carrier adenine nucleotide translocator),




member 6) (cDNA, FLJ92654, highly similar to Homo sapiens




solute carrier family 25 (mitochondrial carrier;




adenine nucleotide translocator), member 6 (SLC25A6),




mRNA)


Q0VGD6
HNRPR
HNRPR protein (Fragment)


A0A024R3T8
PARP1
Poly [ADP-ribose] polymerase (PARP) (EC 2.4.2.30)



hCG_14746


P09874
PARP1
Poly [ADP-ribose] polymerase 1 (PARP-1) (EC 2.4.2.30)



ADPRT
(ADP-ribosyltransferase diphtheria toxin-like 1)



PPOL
(ARTD1) (NAD(+) ADP-ribosyltransferase 1) (ADPRT




1) (Poly[ADP-ribose] synthase 1)


Q8IVF2
AHNAK2
Protein AHNAK2



C14orf78



KIAA2019


Q9BQG0
MYBBP1A
Myb-binding protein 1A



P160


A6PVS8
LRRIQ3
Leucine-rich repeat and IQ domain-containing protein 3



LRRC44
(Leucine-rich repeat-containing protein 44)


A8K6K6

cDNA FLJ76880


A8K2G7

cDNA FLJ76071, highly similar to Homo sapiens




filamin A interacting protein 1 (FILIP1), mRNA


B0AZQ4

Structural maintenance of chromosomes protein


Q9P1Z9
CCDC180
Coiled-coil domain-containing protein 180



C9orf174



KIAA1529


Q9UFH2
DNAH17
Dynein heavy chain 17, axonemal (Axonemal beta



DNAHL1
dynein heavy chain 17) (Axonemal dynein heavy chain-



DNEL2
like protein 1) (Ciliary dynein heavy chain 17) (Ciliary




dynein heavy chain-like protein 1) (Dynein light chain 2,




axonemal)


B2R5B3

Histone H2A


B2RAM8

cDNA, FLJ95007, highly similar to Homo sapiens




BRCA1 associated RING domain 1 (BARD1), mRNA


Q68CZ1
RPGRIP1L
Protein fantom (Nephrocystin-8) (RPGR-interacting



FTM
protein 1-like protein) (RPGRIP1-like protein)



KIAA1005



NPHP8


Q2QL34
MPV17L
Mpv17-like protein (M-LP homolog) (M-LPH)


Q13948
CUX1
Protein CASP



CUTL1


B3KX72

cDNA FLJ44920 fis, clone BRAMY3011501, highly




similar to Heterogeneous nuclear ribonucleoprotein U


Q9NVI7
ATAD3A
ATPase family AAA domain-containing protein 3A


B3KS36

cDNA FLJ35376 fis, clone SKMUS2004044, highly




similar to Homo sapiens ribosomal protein L3 (RPL3),




transcript variant 2, mRNA


D7EZH4

SNF2LT


Q9C0G6
DNAH6
Dynein heavy chain 6, axonemal (Axonemal beta dynein



DNAHC6
heavy chain 6) (Ciliary dynein heavy chain 6)



DNHL1



HL2



KIAA1697


O60524
NEMF
Nuclear export mediator factor NEMF (Antigen NY-CO-



SDCCAG1
1) (Serologically defined colon cancer antigen 1)


B4DWU6

cDNA FLJ51361, highly similar to Keratin, type II




cytoskeletal 6A


B4DXG0

cDNA FLJ57651, highly similar to Ketosamine-3-kinase




(EC 2.7.1.—)


B4DGN6

cDNA FLJ50007


B4DXQ8

cDNA FLJ52940, highly similar to Mortality factor 4-




like protein 2


Q7L099
RUFY3
Protein RUFY3 (RUN and FYVE domain-containing



KIAA0871
protein 3) (Rap2-interacting protein x) (RIPx) (Single




axon-regulated protein) (Singar)


Q9C099
LRRCC1
Leucine-rich repeat and coiled-coil domain-containing



CLERC
protein 1 (Centrosomal leucine-rich repeat and coiled-



KIAA1764
coil domain-containing protein)


B4DYY8

cDNA FLJ60374


Q14439
GPR176
G-protein coupled receptor 176 (HB-954)


B4DZM3

cDNA FLJ61500, highly similar to NNP-1 protein


P62318
SNRPD3
Small nuclear ribonucleoprotein Sm D3 (Sm-D3)




(snRNP core protein D3)


B4E1T1

cDNA FLJ54081, highly similar to Keratin, type II




cytoskeletal 5


B4DLB1

cDNA FLJ58017, moderately similar to Treacle protein


Q8TC59
PIWIL2
Piwi-like protein 2 (EC 3.1.26.—) (Cancer/testis antigen



HILI
80) (CT80)


Q16513
PKN2
Serine/threonine-protein kinase N2 (EC 2.7.11.13) (PKN



PRK2
gamma) (Protein kinase C-like 2) (Protein-kinase C-



PRKCL2
related kinase 2)


O75923
DYSF
Dysferlin (Dystrophy-associated fer-1-like protein) (Fer-



FER1L1
1-like protein 1)


Q5RF89
DKFZp469P0721
Putative uncharacterized protein DKFZp469P0721


Q9UBN4
TRPC4
Short transient receptor potential channel 4 (TrpC4)




(Trp-related protein 4) (hTrp-4) (hTrp4)


P62826
RAN
GTP-binding nuclear protein Ran (Androgen receptor-



ARA24
associated protein 24) (GTPase Ran) (Ras-like protein



OK/SW-
TC4) (Ras-related nuclear protein)



cl.81


Q6NTA2
HNRNPL
HNRNPL protein (Fragment)


B4DPC0

cDNA FLJ52713, moderately similar to Mus musculus




leucine rich repeat (in FLII) interacting protein 1




(Lrrfip1), mRNA


B7Z2C5

cDNA FLJ50492, highly similar to Cyclin-dependent




kinase-like 3 (EC 2.7.11.22)


Q86TI0
TBC1D1
TBC1 domain family member 1



KIAA1108


Q15233
NONO
Non-POU domain-containing octamer-binding protein



NRB54
(NonO protein) (54 kDa nuclear RNA- and DNA-binding




protein) (55 kDa nuclear protein) (DNA-binding




p52/p100 complex, 52 kDa subunit) (NMT55) (p54(nrb))




(p54nrb)


B7Z4E3
RPL31
60S ribosomal protein L31 (cDNA FLJ58908, highly




similar to 60S ribosomal protein L31)


B7Z7K9

CDNA FLJ51382


Q92833
JARID2
Protein Jumonji (Jumonji/ARID domain-containing



JMJ
protein 2)


Q8N398
VWA5B2
von Willebrand factor A domain-containing protein 5B2


Q9BVH8
VWA5B2
VWA5B2 protein (Fragment)


Q6ZU80
CEP128
Centrosomal protein of 128 kDa (Cep128)



C14orf145



C14orf61


P46013
MKI67
Proliferation marker protein Ki-67 (Antigen identified by




monoclonal antibody Ki-67) (Antigen KI-67) (Antigen




Ki67)


A2A547
Rpl19
Ribosomal protein L19


E4W6B6
RPL27
RPL27/NME2 fusion protein (Fragment)


O15050
TRANK1
TPR and ankyrin repeat-containing protein 1 (Lupus



KIAA0342
brain antigen 1 homolog)



LBA1


B3KQL5

cDNA FLJ90678 fis, clone PLACE1005736, highly




similar to Pleckstrin homology domain-containing family




A member 1


Q9HB21
PLEKHA1
Pleckstrin homology domain-containing family A



TAPP1
member 1 (PH domain-containing family A member 1)




(Tandem PH domain-containing protein 1) (TAPP-1)


O60264
SMARCA5
SWI/SNF-related matrix-associated actin-dependent



SNF2H
regulator of chromatin subfamily A member 5



WCRF135
(SWI/SNF-related matrix-associated actin-dependent




regulator of chromatin A5) (EC 3.6.4.—) (Sucrose




nonfermenting protein 2 homolog) (hSNF2H)


Q14789
GOLGB1
Golgin subfamily B member 1 (372 kDa Golgi complex-




associated protein) (GCP372) (Giantin) (Macrogolgin)


A0A087WUK2
HNRNPDL
Heterogeneous nuclear ribonucleoprotein D-like



HNRPDL
(Heterogeneous nuclear ribonucleoprotein D-like,



hCG_22986
isoform CRA_b)


O14979
HNRNPDL
Heterogeneous nuclear ribonucleoprotein D-like (hnRNP



HNRPDL
D-like) (hnRNP DL) (AU-rich element RNA-binding



JKTBP
factor) (JKT41-binding protein) (Protein laAUF1)









In one aspect, the active agent may be selected from one or more compounds as listed in Table 2.









TABLE 2







Compounds that inhibit proteins that inhibit nucleic acid delivery vehicle uptake.















CAS
Pubchem



Compound name
Structure
Source
registry
ID
PMID





Geldanamycin and derivative Alvespimycin


embedded image


Multiple, Wutech Acorn Pharma Tech Product List ZINC OWNED by, Novartis
30562-34- 6
5288382
1551101 2656616





Entasobulin


embedded image


Multiple, ZINC MedChem- express MCE ChemScene
501921- 61-5
10203597






Androstanolone/ Dihydro- testosterone


embedded image


Multiple, Sigma-Aldrich Key Organics/ BIO NET 1717 CheMall Corporation OWNED by,
12040-51- 6, 28801- 96-9, 29873-50- 5, 521-18- 6, 571-22- 2
10635, 15
1660453 8 2003561 5 2042747 6





Spermine (Spermine-RX- 11, CKII


embedded image


Multiple, Finetech Industry
71-44-3
1103

2696287
3 6534776



activation)

Limited


2042747




AK Scientific,


6




Inc. (AKSCI)


3878




Sigma-Aldrich








Cortisone


embedded image


Multiple, Ambinter LGC Standards AKos Consulting & Solutions
53-06-5
222786
24 2785665 5





Quercetin


embedded image


Multiple, BePharm Ltd. Ambinter TimTec
117-39-5, 6151-25- 3, 7255- 55-2, 73123-10- 1, 74893- 81-5
5280343
2857457 4





Acetohexamide (Acetohexamide- RX013, ABCC8 activation)


embedded image


Multiple, TargetMol Boc Sciences Angene Chemical OWNED by, Watson Lilly
8054-32- 8, 968-81- 0
1989

21249 2264568 9






Resveratrol


embedded image


Multiple, 1717 CheMall Corporation ApexBio Technology Selleckchem OWNED by, Home Aide Diagnostics, Inc.
501-36-0
445154
7497631 2849973 2 2840697 4





Doxorubicin (Doxorubicin- RX012, modulator of multiple cytosolic interaction)


embedded image


Multiple, AbovChem LLC Alsachim ABBLIS Chemicals OWNED by, Pfizer
23214-92- 8, 25316- 40-9
31703
3405 14644 2865737 2 2871837 0





Ruxolitinib (Ruxolitinib- RX008, JAK1 inhibition)


embedded image


Multiple, BePharm Ltd. AvaChem Scientific Active Biopharma OWNED by, Novartis
1092939- 17-7
25126798
1938567 2 1946827 5 2852087 1





Roscovitine/ Seliciclib (Roscovitine- RX001, CDK1 inhibition)


embedded image


Multiple, Tocris Bio- science abcr GmbH Boc Sciences OWNED by, Cyclacel Pharmaceuti- cals Inc.
186692- 44-4
5097
2696287 3 9046330 2069273 7





Sildenafil (Sildenafil Citrate-RX014, PDE11A inhibition)


embedded image


Multiple, OXCHEM CORPORATI ON MolPort Vitas-M Laboratory OWNED by, Pfizer Actavis Pharma Company
139755- 83-2, 171599- 83-0
5212
2865226 2 2853553 6 2864007 7





Teniposide/ Vumon


embedded image


Multiple, AK Scientific, Inc. AbovChem LLC Boc Sciences OWNED by, WG Critical Care, LLC Bristol Myers Squibb
23362-13- 2, 29767- 20-2, 31514-29- 1, 35317- 44-3
34698
2691615 0 2658361 1 2277170 6









Description of Agents in Table 2. Geldanamycin is a benzoquinone ansamycin that binds to the heat shock protein Hsp90 and activates a heat shock response in mammalian cells. Entasobulin is the first anticancer drug in development involving two mechanisms of action, tubulin and topoisomerase II inhibition. Entasobulin expresses different modes of action such as, pro-apoptotic and anti-angiogenic properties. Dihydrotestosterone (DHT) (INN: androstanolone) is a biologically active metabolite of the hormone testosterone, formed primarily in the prostate gland, testes, hair follicles, and adrenal glands by the enzyme 5-alpha-reductase by means of reducing the alpha 4, 5 double-bond. Dihydrotestosterone belongs to the class of compounds called androgens, also commonly called androgenic hormones or testoids. DHT is thought to be approximately 30 times more potent than testosterone because of increased affinity to the androgen receptor. Spermine is a polyamine involved in cellular metabolism found in all eukaryotic cells. The precursor for synthesis of spermine is the amino acid ornithine. It is found in a wide variety of organisms and tissues and is an essential growth factor in some bacteria. It is found as a polycation at physiological pH. Spermine is associated with nucleic acids and is thought to stabilize helical structure, particularly in viruses. Cortisone is a Corticosteroid. The mechanism of action of cortisone is as a Corticosteroid Hormone Receptor Agonist. Quercetin is a flavonoid and more specifically a flavonol and represents 60% of the total dietary flavonols intake. The term flavonoid comprises several thousand plant derived compounds sharing a common skeleton of phenyl-chromane. This basic structure allows a multitude of substitution patterns leading to several flavonoid subclasses such as flavonols, flavones, flavanones, catechins, anthocyanidins, isoflavones, dihydroflavonols and chalcones. The first generation sulfonylureas include acetohexamide, chlorpropamide, tolazamide and tolbutamide, oral hypoglycemic agents that are used in therapy of type 2 diabetes. Resveratrol (3,5,4′-trihydroxystilbene) is a polyphenolic phytoalexin. It is a stilbenoid, a derivate of stilbene, and is produced in plants with the help of the enzyme stilbene synthase. It exists as two structural isomers: cis-(Z) and trans-(E), with the trans-isomer shown in the top image. The trans- form can undergo isomerization to the cis- form when heated or exposed to ultraviolet irradiation. In a 2004 issue of Science, Dr. Sinclair of Harvard University said resveratrol is not an easy molecule to protect from oxidation. It has been claimed that it is readily degraded by exposure to light, heat, and oxygen. However, studies find that Trans-resveratrol undergoes negligible oxidation in normal atmosphere at room temperature. Doxorubicin is a drug used in cancer chemotherapy. It is an anthracycline antibiotic, closely related to the natural product daunomycin, and like all anthracyclines it intercalates DNA. It is commonly used in the treatment of a wide range of cancers, including hematological malignancies, many types of carcinoma, and soft tissue sarcomas. The drug is administered in the form of hydrochloride salt intravenously. It may be sold under the brand names Adriamycin PFS, Adriamycin RDF, or Rubex. It is photosensitive and it is often covered by an aluminum bag to prevent light from affecting it. Ruxolitinib (INCB018424) is a selective oral JAK1/JAK2 inhibitor. This agent has the potential to modulate two important kinases that may play a role in myeloproliferative neoplasms, including primary myelofibrosis. Roscovitine is a Potent and Selective Inhibitor of the Cyclin-Dependent Kinases cdc2, cdk2 and cdk5. Sildenafil is a selective PDE5 inhibitor that is used to treat erectile dysfunction and pulmonary arterial hypertension. Teniposide/Vumon is a semisynthetic derivative of podophyllotoxin with antineoplastic activity. Teniposide forms a ternary complex with the enzyme topoisomerase II and DNA, resulting in dose-dependent single- and double-stranded breaks in DNA, DNA: protein cross-links, inhibition of DNA strand religation, and cytotoxicity. This agent acts in the late S or early G phase of the cell cycle.









TABLE 3







Compounds that inhibit proteins that inhibit nucleic acid delivery vehicle uptake












#
Drug
Gene Symbol
Target
Effect
Pubmed















1
Entasobulin
TOP2B
TOP2
Inhibition




intracellular

beta


2
Memantine
GRIN3A
NR3A
Inhibition
17157509



extracellular



region


3
Teniposide
TOP2B
TOP2
Inhibition
 8967966



intracellular

beta


4
Etoposide
TOP2B
TOP2
Inhibition
1312600, 1312601,



intracellular

beta

1662724, 2158562,







2167985, 2537424,







2550587, 2849640,







7473578, 7922123,







8120864, 8295216,







8410993, 9211397,







10395485, 10809021,







11754608, 12877556,







15008514, 15084135,







15158802, 15177438,







16242334, 16903072,







17035025, 17580961,







14504921


5
INO 1001
PARP1
PARP-1
Inhibition
15523000, 18535785,



intracellular



20364863, 14523042


6
Diazoxide
ABCC8
SUR1
Activation
10419549, 11073882,



intracellular



11121575, 12023875,







12565699, 14741296,







15561900


7
Tedisamil
ABCC8
SUR1
Inhibition
10445672, 10684468,



extracellular



10829253



region


8
Glimepiride
ABCC8
SUR1
Inhibition
9779817, 10773014,



intracellular



11078468, 12819907,







20055691, 11325810


9
Epirubicin
TOP2B
TOP2
Inhibition
16322310



intracellular

beta


10
Annamycin
TOP2B
TOP2
Inhibition
15542779



intracellular

beta


11
As(,2)O(,3)
PARP1
PARP-1
Inhibition
12883267



intracellular


12
(R/S)-
ABCC8
SUR1
Inhibition
10773014, 11440368,



Repaglinide



11716850, 12196472,



extracellular



12623163, 12819907,



region



15200348, 15219283,







15380228, 15678092


13
TOP53
TOP2B
TOP2
Inhibition
11170388



intracellular

beta


14
Acetohexamide
ABCC8
SUR1
Activation
15200348, 15561903



extracellular



region


15
Elsamitrucin
TOP2B
TOP2
Inhibition
 8280493



intracellular

beta


16
Ketamine
GRIN3A
NR3A
Inhibition
17084865, 8336337,



extracellular



8941398, 9719604,



region



11937336


17
NK109
TOP2B
TOP2
Inhibition
 9303354



intracellular

beta


18
Tifenazoxide
ABCC8
SUR1
Activation
12213059, 12961066,



extracellular



14514634, 14764798,



region



15220194


19
Olaparib
PARP1
PARP-1
Inhibition
18800822, 22343925,



intracellular



23049934


20
Intoplicine
TOP2B
TOP2
Inhibition
 8043587



intracellular

beta









In one aspect, one or more compounds or a derivative thereof may be used to facilitate transfer of the nanoparticle. These include one or more of the following: (that can be administered to patients approximately 30 to 60 minutes prior to dosing with DNPs) doxorubicin, sildenafil androstanolone, acetohexamide, and teniposide, roscovitine (Imidazopyrimidine), spermine (Dialkylamine), geldanamycin (Macrolactam), ruxolitinib (Pyrrolopyrimidine), teniposide (Podophyllotoxin), sildenafil (Benzenesulfonamide), androstanolone (Anabolic Steroiod), acetohexamide (Alkyl-Phenylketone), doxorubicin (Anthracycline), sonolisib (Steroid Lactone), LY294002 (Benzopyran), TG100115 (Pteridine), Trifluoperazine (Benzothiazine, Phenothiazine), CEP5214 (Indole Derivative, Pyrrolocarbazole), Afuresertib (Substituted Benzene, Phenethylamine, Amphetamine), Cation Vemurafenib (Aryl-Phenylketone), Suramin (Benzene Derivative, Benzanilide), Uprosertib (Benzene Derivative, Phenethylamine, Amphetamine), Flutamide, (Benzene Derivative, Trifluoromethylbenzene), Enzastaurin (Indole Derivative, N-alkylindole), Fasudil (Isoquinoline derivative), Ruboxistaurin (Macrolactam), Pentamidine (Phenol Ether), Uprosertib (Benzene Derivative, Phenethylamine, Amphetamine), Lestaurtinib (Indole derivative, Indolocarbazole), Adenine (Imidazolepyrimidine), Pimozide (Diphenylbutylpiperidine), Chlorpromazine (Phenothiazine), and Afuresertib (Benzene Derivative, Phenethylamine, Amphetamine)


These individual compounds alone or in combination may be combined with the nanoparticle formulation or administered prior or post intranasal (IN) administration (e.g., 5 μg (with respect to DNA) in a 25 μl solution).


The gene transfer may occur in the context of administration to a cell in a human, i.e., administration of a vector containing a nucleic acid to a mammal, particularly a human. For example, an individual may be administered a compound and/or RNAi as disclosed herein prior to administration of a nucleic acid delivery system as known in the art (exemplary nucleic acid delivery systems are known in the art and disclosed in References 11-16). The nucleic acid may be single stranded or double stranded, or may, in certain instances, utilize multiple delivery vehicles which may employ one or the other or both.


The nanoparticle delivery vehicle may take a variety of forms. For example, in one aspect, the nucleic acid delivery vehicle may be a nanoparticle comprising said gene. In one aspect, the nucleic acid delivery vehicle may be a nanoparticle comprising a lysine polymer conjugated to PEG and complexed with a nucleic acid comprising the gene.


In one aspect, the proteins that inhibit the nucleic acid delivery vehicle uptake may be selected from keratin 13, APC protein, protocadherin 17, spectrin alph (non-erythrocytic 1), or a combination thereof.


In one aspect, a period of time exists between step a and step b. In aspects in which the nucleic acid delivery vehicle is administered following delivery of an RNAi and/or compound as disclosed herein, the nucleic acid delivery vehicle may be administered to an individual in need thereof, for example, 30 minutes, or 60 minutes, or 90 minutes, or 120 minutes following the administration of a compound and/or RNAi as disclosed herein. In the case of RNAi, in some aspects, the RNAi may be administered about 12 hours in advance of a nucleic acid delivery vehicle, about 20 hours in advance of a nucleic acid delivery vehicle, about 24 hours in advance of a nucleic acid delivery vehicle, or about 30 hours in advance of administration of the delivery vehicle.


For example, for RNAi application, patient stem cells or patient derived iPSCs are harvested and cultured and treated with RNAi against a gene in Table 1 for 24 hr. NNPs formulated to contain an expression cassette for the therapeutic gene are then added to the cells for 72 hr. Reagents and delivery vector are replaced daily. An example of the time involved for the active agent application method is; patients are treated with one or more of the compounds claimed Tables 2 and 3 about 30 to about 60 minutes prior to gene delivery vector administration. Agent treatment may be conducted one or more times before gene therapy. NNPs containing an expression cassette for the therapeutic gene may then be administered to the airways of the patient, for example, via nebulization.


In one aspect, the method may include the step of providing a reagent that facilitates transfection. In one aspect, said agent may be a cationic lipid transfection reagent (e.g. Lipofectamine or GL67), which may be mixed with a nucleic acid under a given formulation to produce a nucleic acid/lipid complex. For lipid (or protein) nucleic acid complexes, any formulation that produces lipid/nucleic acid or protein/nucleic acid complexes (of which there are 1000s) can be combined with the methods herein. This may similarly apply to protein polymers such PEGylated poly L lysine or PEI. For viral vectors, the vector may be produced in cell lines, purified and used for therapy in accordance with the disclosed methods.


Compositions comprising RNAi and/or the compounds of Tables 2 and/or 3 may be administered intranasally. In such aspect, the compositions may further comprise other agents suited for improved delivery across nasal mucosa. For example, in certain aspects, agents such as a permeation enhancer, a polymer capable of increasing mucosal adhesion of the composition, or a combination thereof may be included in the composition. In one aspect, the disclosed compositions may comprise, consist of or consist essentially of any of the aforementioned features, in any combination.


It will be appreciated by those skilled in the art that the particular method of administration will depend on a variety of factors, all of which are considered routinely when administering therapeutics, particularly in the context of gene transfer. It will also be understood, however, that the specific dose level for any given patient will depend upon a variety of factors, including, the activity of the specific compound employed, the age of the patient, the body weight of the patient, the general health of the patient, the gender of the patient, the diet of the patient, time of administration, route of administration, rate of excretion, drug combinations, and the severity of the condition undergoing therapy. It will be further appreciated by one skilled in the art that the optimal course of treatment, i.e., the mode of treatment and the daily number of doses o given for a defined number of days, can be ascertained by those skilled in the art using conventional treatment tests.


Nanoparticle Counterions


Disclosed are nanoparticles containing nucleic acids such as DNA or RNA, which may be double or single stranded, and which may be protein coding or anti-sense coding or non-coding. The nucleic acids may include analogs of RNA and/or DNA (including, for example, miRNA, shRNA, tRNA, siRNA, single and double stranded DNA) that are modified to enhance degradation in vivo.


Methods of making nanoparticles in accordance with the instant invention are known in the art. See, for example U.S. Pat. No. 8,017,577, entitled “Lyophilizable and enhanced compacted nucleic acids,” and/or “Chapter 33: Real-Time Imaging of Gene Delivery and Expression with DNA Nanoparticle Technologies” by Sun and Ziady, filed herewith, both of which are incorporated herein in their entirety by reference. Disclosed herein are alternate counterions to those disclosed in the art which are used to manufacture nucleic acid nanoparticles. Counterions of polycations used to compact nucleic acids are known to affect the shape of particles formed. Shape is associated with nuclease resistance and colloidal stability. Moreover, shape affects the suitability and efficacy of compacted nucleic acid complexes for transfecting cells by various routes into a mammalian body.


The counterion that may be used in making compacted nucleic acid complexes may also have an effect on the stability of the complexes to lyophilization. Disclosed herein are nanoparticles which are compacted using one or more counterions selected from from trifluoroacetate (TFA), bromide, bicarbonate, glutamate, aspartate, hydroxyl ions, or combinations thereof, which may be used before compaction of the nucleic acid.


Polycations may comprise polyamino acids such as polylysine and derivatives of polylysine. The polycation may contain from 15-60 lysine residues, preferably in the ranges of 15-30, 30-45, or 45-60 residues. Exemplary derivatives of polylysine are CK15, CK30, CK45, which have an additional cysteine residue attached to polylysine polymers of length 15, 30, and 45 residues, respectively. Other amino acids can be readily attached to polylysine. Other polycationic amino acid polymers can be used such as polyarginine, or copolymers of arginine and lysine. Polymers of non-protein amino acids, such as omithine or citrulline, could also be used. Any pharmaceutically approved or appropriate polycation can be used including but not limited to protamine, histones, polycationic lipids, putrescine, spermidine, spermine, peptides, and polypeptides. The polycation may also contain a targeting moiety, which is typically a ligand which binds to a receptor on a particular type of cell. The targeting ligand may be a polyamino acid or other chemical moiety. Specificity of interaction of the ligand and the receptor is important for purposes of targeting. In one aspect, the polycation may be reacted with a bifunctional PEG (e.g. PEG-maleimide (PEG-Mal) or ortho-pyridyl disulfide (OPSS) (PEG-OPSS) to allow for the addition of a targeting moiety.


In one aspect, a composition is disclosed, the composition comprising a) a compacted nucleic acid nanoparticle as described above; and b) one or more agents selected from sonolisib (Steroid Lactone), LY294002 (Benzopyran), TG100115 (Pteridine), Trifluoperazine (Benzothiazine, Phenothiazine), CEP5214 (Indole Derivative, Pyrrolocarbazole), Afuresertib (Substituted Benzene, Phenethylamine, Amphetamine), Cation Vemurafenib (Aryl-Phenylketone), Suramin (Benzene Derivative, Benzanilide), Uprosertib (Benzene Derivative, Phenethylamine, Amphetamine), Flutamide, (Benzene Derivative, Trifluoromethylbenzene), Enzastaurin (Indole Derivative, N-alkylindole), Fasudil (Isoquinoline derivative), Ruboxistaurin (Macrolactam), Pentamidine (Phenol Ether), Uprosertib (Benzene Derivative, Phenethylamine, Amphetamine), Lestaurtinib (Indole derivative, Indolocarbazole), Adenine (Imidazolepyrimidine), Pimozide (Diphenylbutylpiperidine), Chlorpromazine (Phenothiazine), Afuresertib (Benzene Derivative, Phenethylamine, Amphetamine), and combinations thereof; and optionally, one or more agents selected from Table 1 or Table 2.


Kits are also provided. In one aspect, a kit may comprise or consist essentially of agents or compositions described herein. The kit may be a package that houses a container which may contain one or more compounds or solutions containing an RNAi as disclosed herein, and also houses instructions for administering the agent or composition to a subject. In one aspect, a pharmaceutical pack or kit is provided comprising one or more containers filled with one or more composition as disclosed herein. Associated with such container(s) can be various written materials such as instructions for use, or a notice in the form prescribed by a governmental agency regulating the manufacture, use or sale of pharmaceuticals or biological products, which notice reflects approval by the agency of manufacture, use, or sale for human administration.


As there may be advantages to mixing a component of a composition described herein and a pharmaceutically acceptable carrier, excipient or vehicle near the time of use, kits in which components of the compositions are packaged separately are disclosed. For example, the kit can contain an active ingredient in a powdered or other dry form in, for example, a sterile vial or ampule and, in a separate container within the kit, a carrier, excipient, or vehicle, or a component of a carrier, excipient, or vehicle (in liquid or dry form). In one aspect, the kit can contain a component in a dry form, typically as a powder, often in a lyophilized form in, for example, a sterile vial or ampule and, in a separate container within the kit, a carrier, excipient, or vehicle, or a component of a carrier, excipient, or vehicle. Alternatively, the kit may contain a component in the form of a concentrated solution that is diluted prior to administration. Any of the components described herein, any of the carriers, excipients or vehicles described herein, and any combination of components and carriers, excipients or vehicles can be included in a kit.


Optionally, a kit may also contain instructions for preparation or use (e.g., written instructions printed on the outer container or on a leaflet placed therein) and one or more devices to aid the preparation of the solution and/or its administration to a patient (e.g., one or a plurality of syringes, needles, filters, tape, tubing (e.g., tubing to facilitate intravenous administration) alcohol swabs and/or the Band-Aid® applicator). Compositions which are more concentrated than those administered to a subject can be prepared. Accordingly, such compositions can be included in the kits with, optionally, suitable materials (e.g., water, saline, or other physiologically acceptable solutions) for dilution. Instructions included with the kit can include, where appropriate, instructions for dilution.


In other embodiments, the kits can include pre-mixed compositions and instructions for solubilizing any precipitate that may have formed during shipping or storage. Kits containing solutions of one or more of the aforementioned active agents, or pharmaceutically acceptable salts thereof, and one or more carriers, excipients or vehicles may also contain any of the materials mentioned above (e.g., any device to aid in preparing the composition for administration or in the administration per se). The instructions in these kits may describe suitable indications (e.g., a description of patients amenable to treatment) and instructions for administering the solution to a patient.


EXAMPLES

Method for Enhancing Nucleic Acid Transfer


Applicant has discovered methods for enhancing the efficiency of gene transfer through the use of interference RNA (RNAi) technology or pharmacological agents that modulate the interactome (FIG. 1) of nucleic acid nanoparticles consisting of polymers of lysine conjugated to PEG and complexed with nucleic acids. Both of these approaches have been reduced to practice and achieve significantly higher levels of gene transfer in the context of condensed DNA nanoparticle vectors, resulting in as much as 50-fold greater gene transfer efficiency. These technologies represent a significant enhancement to gene transfer technologies.


By using a novel immunocapture procedure (FIG. 2), Applicant identified protein interactors of polyethylene glycol conjugated DNA nanoparticles. This investigation revealed 474 unique proteins that interact with the nanoparticles as listed in Table 3. Many of these proteins represent a nanoparticle specific transfection interactome, but a number of proteins such as Prohibitin 1 and 2 are also involved in viral as well as liposomal gene delivery. Some of these protein interactors may be inhibiting the cellular uptake of DNA nanoparticles as well as other vectors for the delivery of nucleic acids. The interactome segregated into sites in the cell where nucleic acid particles are delivered (Table 4). In this method, Applicant used RNAi and/or pharmacological agents to modulate the particle interactome and enhance nucleic acid delivery to the nucleus (DNA) or the ribosome (RNA).









TABLE 4







Characteristics of the nucleic acid nanoparticle cellular protein interactome


















False
Percent of



Cellular

Cellular

Discovery
Dataset


Rank
Processes
Class
Localization
P Value
Rate
(%)
















1
Intermediate
Cytoskeleton
Cytosol
1.99E−14
8.68E−13
21.75



filaments







2
Translation
Translation
Ribosome
1.05E−16
1.37E−14
15.45



initiation







3
Elongation-
Translation
Ribosome
1.99E−15
1.30E−13
12.47



Termination







4
Actin filaments
Cytoskeleton
Cytosol
4.84E−08
1.58E−06
10.01


5
Chromatin
Transcription
Nucleus
7.80E−06
2.04E−04
9.71



modification







6
Spindle
Cytoskeleton
Cytosol
7.16E−04
8.53E−03
8.08



microtubules







7
mRNA
Transcription
Nucleus
1.06E−04
1.83E−03
7.71



processing







8
Cell junctions
Cell adhesion
Cell
1.21E−04
1.83E−03
7.61





membrane





9
Regulation of
Cytoskeleton
Cytosol
1.25E−04
1.83E−03
7.22



cytoskeleton








rearrangement














For RNAi application, RNAi molecules may be delivered to the cells, or in the case of delivery to an individual, to the individual, prior to the desired nucleic acid delivery vehicle. The RNAi molecules are administered in an amount sufficient to target and knock down specific cellular proteins that negatively impact the uptake of the nucleic acid delivery vehicle. RNAi decreases the cellular levels of these proteins, reducing their deleterious impact on the downstream transfer of nucleic acids. RNAi mediated knockdown of four of these proteins has been tested by Applicant, which resulted in significant enhancement of gene transfer in ¾ constructs tested. RNAi targeted to interfere with the synthesis of the 4 proteins; keratin 13 (GI: 81891678), APC protein (GI: 97535708), protocadherin 17 (GI:94538350), and spectrin alpha (non-erythrocytic 1, GI:119608216) that are deleterious to gene transfer with the DNA nanoparticles improved gene delivery (FIG. 3).


In addition to, or separately, pharmacological agents that modulate the DNP interactome can enhance nucleic acid transfer to the nucleus or the ribosome (in the case of RNA delivery). Applicant found 13 compounds and their derivatives that modulate 71 interactors (see Table 2) that can be administered to patients about 30 to about 60 minutes prior to dosing with DNPs. These are classified by cellular site of action. For example, Doxorubicin and Sildenafil will act to inhibit or promote interactions in the cytosol. Androstanolone will modulate interactions at the ribosome. Acetohexamide will promote non-nucleolin mediated interactions at the cell membrane. Ruxolitinib and Teniposide may be used to modulate nuclear interactions. Applicant's data also points to the importance to the interaction with nucleolin and how modulation of this interaction at the plasma membrane greatly impacts gene transfer with DNPs (FIGS. 3-5). Modulation of these cellular interactions is expected to have different impacts on RNPs vs. DNPs, as the cellular compartment targets for these formulations of NNPs vary (ribosome vs. nucleus, respectively). For example, drugs that promote cellular entry may benefit both DNPs and RNPs. However, drugs that diminish interactions at the ribosome would be expected to only benefit DNPs. Conversely, drugs that diminish nuclear interactions should benefit RNPs.


Table 2 and 3 outlines compounds may be used to modulate nucleolin associated nucleic acid nanoparticle (NNP) uptake. Nucleolin translocation to the cell surface may be promoted by IP injections of roscovitine (inhibits CDK1 at 10 mg/kg), spermine (induces CKII at 50 mg/kg), geldanamycin (inhibits HSP90 interaction with nucleolin at 15 mg/kg), or hydrocortisone (increases GR shuttling of nucleolin to the cell surface at 7.5 mg/kg) into animals 60 min prior to a 25 μl intranasal (IN) administration of 5 μg (with respect to DNA) NNPs containing the CFTR gene, as has been previously reported(1). Control groups injected with either DMSO instead of pharmacological agents, and NNPs containing the transgene with no drug may be used. CF mice may receive NPD measurements 1 week before treatment (a background/baseline measurement) and then 4, 7, and 14 days after transfection with CFTR-containing NNPs applied to the nose, as previously reported (1). Two weeks following transfection mice will be sacrificed, and the lungs may be harvested, paraffin imbeded, and sectioned for immunohistochemistry, and sections probed with the CF3 or 24:1 anti-CFTR Ab that does not cross react with mouse cftr, as previously reported (1). Studies can be duplicated in F508del and S489X homozygote mice.


Use in Research. The RNAi and/or pharmacological approaches to enhancing gene transfer may be developed as an additive to current gene transfer and transfection vectors. For example, it may be used as a supplemental additive to the cationic lipid transfection reagent Lipofectamine, enabling either greater gene transfer or decreased amounts of transfection reagent used, resulting in either reduced costs or enhanced efficiency. Alternatively, pharmacological and RNAi treatment may be employed prior to or concurrent with the delivery of viral vectors in in vitro or ex vivo gene transfer applications. This may allow more cellular gene modification and higher expression of therapeutic transgenes within these cells, or decreased viral titer needed to provide curative levels of cell modification. This may increase the efficacy of these genes or reduce the associated costs with producing sufficient amount of virus, which is currently a significant obstacle in gene therapy protocols.


Use in human therapy. CF is the most common inherited recessive disorders in Caucasians, and advances in small molecule therapy have not significantly benefitted a large majority of the patients. Gene therapy (repair or replacement) offers a potential of corrective therapy for the disease regardless of mutation type. The disclosed methods may be useful for enhancing corrective DNA and/or RNA delivery with a synthetic vector to airway epithelial cells, the most affected cells in CF. The present example relates to the biology of NNPs, a vector that has been shown to have partial efficacy in correcting CFTR in CF patients (2). Applicant has found 71 specific protein interactors (for example, some of the interactors and associated regulation are shown in FIG. 2, others are listed in Table 1) that help define the biology of the particles in cells and can be targeted with 13 FDA approved drugs (Table 2). Other compounds are listed in Table 3.


Applicant has demonstrated that modulating the NNP interactome can enhance gene transfer by 10-50 fold, the highest levels of enhancement ever achieved in two decades of modifying and examining DNP-based vectors (see FIGS. 3 and 4). Based on this result and given the fact that DNPs have achieved partial clinical correction in a Phase 1 trial in CF patients (2), the methods of the instant disclosure have the potential to provide pharmacological agents that can enhance gene transfer to fully therapeutic levels in humans. While airway epithelial cells are the primary site of disease and the most important gene therapy target in CF, a better understanding of the determinants of successful gene transfer into these cells will significantly benefit gene delivery for a number of other diseases, including chronic obstructive pulmonary disease (COPD; ˜12,000,000 patients in the USA), and epithelial lung cancers (200,000 patients in the USA). The instant disclosure provides a novel approach to implementation of NNP biology. Findings in airway epithelia will likely be relevant to other cell targets where NNPs have succeeded, including cells in the brain (3-6) and retina (7-10), and may be relevant to the cellular uptake of other non-viral polyplex-based vectors as well as viral and liposomal vectors.


In a therapeutic context, siRNA can be applied to human cells ex vivo or pharmacological agents to humans directly before or during gene delivery to optimize gene transfer obtained with DNA/RNA nanoparticles, and potential with liposomal and viral vectors as well.


REFERENCE LIST



  • 1. Ziady A G, Kelley T J, Milliken E, Ferkol T, Davis P B. Functional evidence of CFTR gene transfer in nasal epithelium of cystic fibrosis mice in vivo following luminal application of DNA complexes targeted to the serpin-enzyme complex receptor. Mol. Ther. 2002 April; 5(4):413-9

  • 2. Konstan M W, Davis P B, Wagener J S, Hilliard K A, Stern R C, Milgram U, Kowalczyk T H, Hyatt S L, Fink T L, Gedeon C R, et al. Compacted DNA nanoparticles administered to the nasal mucosa of cystic fibrosis subjects are safe and demonstrate partial to complete cystic fibrosis transmembrane regulator reconstitution. Hum. Gene Ther. 2004 December; 15(12):1255-69

  • 3. Yurek D M, Fletcher A M, Smith G M, Seroogy K B, Ziady A G, Molter J, Kowalczyk T H, Padegimas L, Cooper M J. Long-term transgene expression in the central nervous system using DNA nanoparticles. Mol. Ther. 2009 April; 17(4):641-50

  • 4. Yurek D M, Flectcher A M, Kowalczyk T H, Padegimas L, Cooper M J. Compacted DNA nanoparticle gene transfer of GDNF to the rat striatum enhances the survival of grafted fetal dopamine neurons. Cell Transplant. 2009; 18(10):1183-96. PMCID:PMC3031110

  • 5. Yurek D M, Fletcher A M, McShane M, Kowalczyk T H, Padegimas L, Weatherspoon M R, Kaytor M D, Cooper M J, Ziady A G. DNA Nanoparticles: Detection of Long-term Transgene Activity In Brain Using Bioluminescence Imaging Mol. Imaging 2011 Apr. 26;

  • 6. Fletcher A M, Kowalczyk T H, Padegimas L, Cooper M J, Yurek D M. Transgene expression in the striatum following intracerebral injections of DNA nanoparticles encoding for human glial cell line-derived neurotrophic factor. Neuroscience 2011 Oct. 27; 194:220-6. PMCID:PMC3408714

  • 7. Farjo R, Skaggs J, Quiambao A B, Cooper M J, Naash M I. Efficient non-viral ocular gene transfer with compacted DNA nanoparticles. PLoS. One. 2006; 1:e38. PMCID:PMC1762345

  • 8. Ding X Q, Quiambao A B, Fitzgerald J B, Cooper M J, Conley S M, Naash M I. Ocular delivery of compacted DNA-nanoparticles does not elicit toxicity in the mouse retina. PLoS. One. 2009; 4(10):e7410. PMCID:PMC2756629

  • 9. Cai X, Conley S M, Nash Z, Fliesler S J, Cooper M J, Naash M I. Gene delivery to mitotic and postmitotic photoreceptors via compacted DNA nanoparticles results in improved phenotype in a mouse model of retinitis pigmentosa. FASEB J. 2010 April; 24(4):1178-91. PMCID:PMC2845431

  • 10. Koirala A, Makkia R S, Conley S M, Cooper M J, Naash M I. S/MAR-containing DNA nanoparticles promote persistent RPE gene expression and improvement in RPE65-associated LCA. Hum. Mol. Genet. 2013 Apr. 15; 22(8):1632-42. PMCID:PMC3605833

  • 11. Ziady A G, Davis P B, Konstan M W. Non-viral gene transfer therapy for cystic fibrosis. Expert. Opin. Biol. Ther. 2003 June; 3(3):449-58

  • 12. Ziady A G, Davis P B. Current prospects for gene therapy of cystic fibrosis. Curr. Opin. Pharmacol. 2006 October; 6(5):515-21

  • 13. Ahmed H, Shubina-Oleinik O, Holt J R. Emerging Gene Therapies for Genetic Hearing Loss. J. Assoc. Res. Otolaryngol. 2017 Aug. 16;

  • 14. Naso M F, Tomkowicz B, Perry W L, III, Strohl W R. Adeno-Associated Virus (AAV) as a Vector for Gene Therapy. BioDrugs. 2017 Jul. 1. PMCID:PMC5548848

  • 15. Huang J, Wang Y, Zhao J. CRISPR Editing in Biological and Biomedical Investigation. J. Cell Physiol 2017 Aug. 8;

  • 16. Zhang X, Wang L, Liu M, Li D. CRISPR/Cas9 system: a powerful technology for in vivo and ex vivo gene therapy. Sci. China Life Sci. 2017 May; 60(5):468-75



All percentages and ratios are calculated by weight unless otherwise indicated.


All percentages and ratios are calculated based on the total composition unless otherwise indicated.


It should be understood that every maximum numerical limitation given throughout this specification includes every lower numerical limitation, as if such lower numerical limitations were expressly written herein. Every minimum numerical limitation given throughout this specification will include every higher numerical limitation, as if such higher numerical limitations were expressly written herein. Every numerical range given throughout this specification will include every narrower numerical range that falls within such broader numerical range, as if such narrower numerical ranges were all expressly written herein.


The dimensions and values disclosed herein are not to be understood as being strictly limited to the exact numerical values recited. Instead, unless otherwise specified, each such dimension is intended to mean both the recited value and a functionally equivalent range surrounding that value. For example, a dimension disclosed as “20 mm” is intended to mean “about 20 mm.”


Every document cited herein, including any cross referenced or related patent or application, is hereby incorporated herein by reference in its entirety unless expressly excluded or otherwise limited. All accessioned information (e.g., as identified by PUBMED, PUBCHEM, NCBI, UNIPROT, or EBI accession numbers) and publications in their entireties are incorporated into this disclosure by reference in order to more fully describe the state of the art as known to those skilled therein as of the date of this disclosure. The citation of any document is not an admission that it is prior art with respect to any invention disclosed or claimed herein or that it alone, or in any combination with any other reference or references, teaches, suggests or discloses any such invention. Further, to the extent that any meaning or definition of a term in this document conflicts with any meaning or definition of the same term in a document incorporated by reference, the meaning or definition assigned to that term in this document shall govern.


While particular embodiments of the present invention have been illustrated and described, it would be obvious to those skilled in the art that various other changes and modifications may be made without departing from the spirit and scope of the invention. It is therefore intended to cover in the appended claims all such changes and modifications that are within the scope of this invention.

Claims
  • 1. A method for transferring a gene into a eukaryotic cell, comprising administering a compacted nucleic acid nanoparticle; and one or more active agent selected from sonolisib (Steroid Lactone), LY294002 (Benzopyran), TG100115 (Pteridine), Trifluoperazine (Benzothiazine, Phenothiazine), CEP5214 (Indole Derivative, Pyrrolocarbazole), Afuresertib (Substituted Benzene, Phenethylamine, Amphetamine), Cation Vemurafenib (Aryl-Phenylketone), Suramin (Benzene Derivative, Benzanilide), Uprosertib (Benzene Derivative, Phenethylamine, Amphetamine), Flutamide, (Benzene Derivative, Trifluoromethylbenzene), Enzastaurin (Indole Derivative, N-alkylindole), Fasudil (Isoquinoline derivative), Ruboxistaurin (Macrolactam), Pentamidine (Phenol Ether), Uprosertib (Benzene Derivative, Phenethylamine, Amphetamine), Lestaurtinib (Indole derivative, Indolocarbazole), Adenine (Imidazolepyrimidine), Pimozide (Diphenylbutylpiperidine), Chlorpromazine (Phenothiazine), Afuresertib (Benzene Derivative, Phenethylamine, Amphetamine), and combinations thereof, to a eukaryotic cell.
  • 2. The method of claim 1, further comprising administering an inhibitor of a protein that inhibits nanoparticle delivery uptake, said inhibitor being a nucleic acid selected from one or more of RNAi, miRNA, shRNA, tRNA, siRNA, single stranded DNA, double stranded DNA, and combinations thereof, and wherein said nucleic acid inhibits synthesis of one or more proteins that inhibit nucleic acid delivery vehicle uptake, preferably wherein said one or more protein is selected from Table 1.
  • 3. The method of claim 1, further comprising administering an active agent that facilitates compacted nucleic acid nanoparticle uptake into a cell, wherein said active agent inhibits synthesis of one or more proteins that inhibit nucleic acid delivery vehicle uptake.
  • 4. The method of claim 2, wherein said inhibitor is RNAi, and wherein said RNAi molecule inhibits expression of a gene encoding a protein selected from Table 1.
  • 5. The method of claim 1, further comprising administering a second active agent selected from an agent listed in Table 2 or Table 3.
  • 6. The method of claim 1, wherein said active agent is selected from roscovitine, geldanamycin, acetohexamide, and ruxolitinib, or a combination thereof.
  • 7. The method of claim 1, wherein said nucleic acid nanoparticle comprises said gene.
  • 8. The method of claim 1, wherein said compacted nucleic acid nanoparticle comprises a nucleic acid plasmid and a polymer, wherein said nanoparticle is compacted in the presence of a counter ion selected from trifluoroacetate (TFA), bromide, bicarbonate, glutamate, hydroxyl ions or combinations thereof.
  • 9. The method of claim 8, wherein said nucleic acid is single or double stranded DNA.
  • 10. The method of claim 8, wherein said polymer is a polycation.
  • 11. The method of claim 10, wherein said polycation is a lipid.
  • 12. The method of claim 10, wherein said polycation is selected from a cysteine (C) containing polymer of lysine (K), a cysteine (C) containing polymer of arginine (R), or combinations thereof.
  • 13. The method of claim 10, wherein said polycation is selected from a cysteine (C) containing polymer of lysine (K) and arginine (R), or polymers of lysine mixed with arginine, conjugated to PEG and complexed with nucleic acids.
  • 14. The method of claim 8, wherein said polymer is a lysine polymer, preferably a polyethylene glycol (PEG)-substituted lysine polymer or polyethylenemine.
  • 15. The method of claim 1, wherein said compacted nucleic acid nanoparticle has a shape selected from rod shape, ellipsoidal, spheroidal, or toroidal.
  • 16. The method of claim 1, wherein said compacted nucleic acid nanoparticle has a diameter of about 25 to about 400 nm in length as measured by electron microscopy.
  • 17. A composition comprising a) a compacted nucleic acid nanoparticle of claim 8; andb) one or more agents selected from sonolisib (Steroid Lactone), LY294002 (Benzopyran), TG100115 (Pteridine), Trifluoperazine (Benzothiazine, Phenothiazine), CEP5214 (Indole Derivative, Pyrrolocarbazole), Afuresertib (Substituted Benzene, Phenethylamine, Amphetamine), Cation Vemurafenib (Aryl-Phenylketone), Suramin (Benzene Derivative, Benzanilide), Uprosertib (Benzene Derivative, Phenethylamine, Amphetamine), Flutamide, (Benzene Derivative, Trifluoromethylbenzene), Enzastaurin (Indole Derivative, N-alkylindole), Fasudil (Isoquinoline derivative), Ruboxistaurin (Macrolactam), Pentamidine (Phenol Ether), Uprosertib (Benzene Derivative, Phenethylamine, Amphetamine), Lestaurtinib (Indole derivative, Indolocarbazole), Adenine (Imidazolepyrimidine), Pimozide (Diphenylbutylpiperidine), Chlorpromazine (Phenothiazine), Afuresertib (Benzene Derivative, Phenethylamine, Amphetamine), and combinations thereof.
  • 18. The composition of claim 17, further comprising one or more agents selected from Table 1 or Table 2.
CROSS REFERENCE TO RELATED APPLICATION

This application claims the benefit of and priority to U.S. Provisional Application No. 63/123,519, entitled “Enhanced Nanoparticle Delivery Systems,” filed Dec. 10, 2020, the contents of which are incorporated by reference in its entirety for all purposes.

STATEMENT REGARDING FEDERALLY-SPONSORED RESEARCH

This invention was made with government support under EB023800 awarded by the National Institutes of Health. The government has certain rights in this invention.

PCT Information
Filing Document Filing Date Country Kind
PCT/US2021/062624 12/9/2021 WO
Provisional Applications (1)
Number Date Country
63123519 Dec 2020 US