WO2021158627A1 - Compositions comprising nanoparticles and methods thereof - Google Patents
Compositions comprising nanoparticles and methods thereof Download PDFInfo
- Publication number
- WO2021158627A1 WO2021158627A1 PCT/US2021/016368 US2021016368W WO2021158627A1 WO 2021158627 A1 WO2021158627 A1 WO 2021158627A1 US 2021016368 W US2021016368 W US 2021016368W WO 2021158627 A1 WO2021158627 A1 WO 2021158627A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- fold
- population
- composition
- nanoparticle
- kda
- Prior art date
Links
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K9/00—Medicinal preparations characterised by special physical form
- A61K9/48—Preparations in capsules, e.g. of gelatin, of chocolate
- A61K9/50—Microcapsules having a gas, liquid or semi-solid filling; Solid microparticles or pellets surrounded by a distinct coating layer, e.g. coated microspheres, coated drug crystals
- A61K9/51—Nanocapsules; Nanoparticles
- A61K9/5107—Excipients; Inactive ingredients
- A61K9/513—Organic macromolecular compounds; Dendrimers
- A61K9/5146—Organic macromolecular compounds; Dendrimers obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyethylene glycol, polyamines, polyanhydrides
- A61K9/5153—Polyesters, e.g. poly(lactide-co-glycolide)
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/70—Carbohydrates; Sugars; Derivatives thereof
- A61K31/7084—Compounds having two nucleosides or nucleotides, e.g. nicotinamide-adenine dinucleotide, flavine-adenine dinucleotide
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/70—Carbohydrates; Sugars; Derivatives thereof
- A61K31/7088—Compounds having three or more nucleosides or nucleotides
- A61K31/7105—Natural ribonucleic acids, i.e. containing only riboses attached to adenine, guanine, cytosine or uracil and having 3'-5' phosphodiester links
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K9/00—Medicinal preparations characterised by special physical form
- A61K9/0012—Galenical forms characterised by the site of application
- A61K9/0019—Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P13/00—Drugs for disorders of the urinary system
- A61P13/12—Drugs for disorders of the urinary system of the kidneys
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K9/00—Medicinal preparations characterised by special physical form
- A61K9/48—Preparations in capsules, e.g. of gelatin, of chocolate
- A61K9/50—Microcapsules having a gas, liquid or semi-solid filling; Solid microparticles or pellets surrounded by a distinct coating layer, e.g. coated microspheres, coated drug crystals
- A61K9/5005—Wall or coating material
- A61K9/5021—Organic macromolecular compounds
- A61K9/5031—Organic macromolecular compounds obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyethylene glycol, poly(lactide-co-glycolide)
-
- B—PERFORMING OPERATIONS; TRANSPORTING
- B82—NANOTECHNOLOGY
- B82Y—SPECIFIC USES OR APPLICATIONS OF NANOSTRUCTURES; MEASUREMENT OR ANALYSIS OF NANOSTRUCTURES; MANUFACTURE OR TREATMENT OF NANOSTRUCTURES
- B82Y5/00—Nanobiotechnology or nanomedicine, e.g. protein engineering or drug delivery
Definitions
- the present disclosure provides a composition comprising a plurality of polymeric nanoparticles, wherein: (i) a polymeric nanoparticle of the plurality of polymeric nanoparticles contains a nucleic acid molecule that binds to a polypeptide; and (ii) the plurality of polymeric nanoparticles is characterize by an average diameter of no greater than about 1 micrometer, as ascertained by dynamic light scattering.
- the present disclosure provides a composition comprising a plurality of polymeric nanoparticles, wherein: (i) a nanoparticle of the plurality of nanoparticles contains a nucleic acid molecule that binds to a polypeptide; and (ii) if, in a controlled study, the nanoparticle is administered to a subject, then the nanoparticle exhibits selective targeting to a targeted tissue of the subject preferentially over a non-targeted tissue of the subject.
- medical agent coated on a surface or within a surface layer of the of particles can be present in an average quantity of at least or up to about 0.1 %, at least or up to about 0.2 %, at least or up to about 0.3 %, at least or up to about 0.4 %, at least or up to about 0.5 %, at least or up to about 0.6 %, at least or up to about 0.7 %, at least or up to about 0.8 %, at least or up to about 0.9 %, at least or up to about 1 %, at least or up to about 2 %, at least or up to about 3 %, at least or up to about 4 %, at least or up to about 5 %, at least or up to about 6 %, at least or up to about 7 %, at least or up to about 8 %, at least or up to about 9 %, at least or up to about 10 %, at least or up to about 11 %, at least or up to about 12 %, at least or up to about
- non-encapsulated ODN e.g., CpG-ODN TLR9 antagonist or control ODN
- ODN non-encapsulated ODN
- all mice received buprenorphine SR (e.g., subcutaneously (s.c.) at about 0.5-1 mg buprenorphine SR per kg weight of the animal) prior to the surgery.
- mice treated with the MNPs encapsulating the control ODN and subjected to renal IR showed severe tubular necrosis, proteinaceous casts, and increased tubular dilatation and congestion.
- mice treated with the MNPs encapsulating the CpG-ODN TLR9 antagonist at about 6 hr before renal ischemia, at the time of reperfusion, or at about 1.5 hr after perfusion exhibited decreased renal tubular necrosis, decreased congestion, and decreased cast formation, as compared to controls.
- FIG. 4 mice treated with the MNPs encapsulating the control ODN and subjected to renal IR showed severe tubular necrosis, proteinaceous casts, and increased tubular dilatation and congestion.
- mice treated with the MNPs encapsulating the CpG-ODN TLR9 antagonist at about 6 hr before renal ischemia, at the time of reperfusion, or at about 1.5 hr after perfusion exhibited decreased renal tubular necrosis, decreased congestion, and decreased cast formation, as compared to controls
- the average diameter is between about 100 nm to about 1000 nm;
- the polymeric nanoparticle comprises a population of copolymers.
- Embodiment 9 A method comprising forming a plurality of polymeric nanoparticles, wherein: (i) a nanoparticle of the plurality of nanoparticles contains a nucleic acid molecule that binds to a polypeptide; and (ii) each nanoparticle of the plurality of nanoparticles independently comprises a polymeric structure, wherein the polymeric structure comprises a population of polymers, wherein the population of polymers has an average molar mass that is at least about 10 kilodaltons (kDa), optionally wherein: (1) the average molar mass is at least about 20 kDa;
- the target moiety is a polypeptide, optionally wherein the polypeptide is a receptor polypeptide; or
- the surfactant is a polyoxyethylene-polyoxypropylene block polymer
- the nanoparticle comprises a population of copolymers.
- the nucleic acid molecule is an oligodeoxynucleotide (ODN);
- the population of copolymers is a population of poly(lactic-co-glycolic acid)- poly(ethylene glycol);
- the target moiety is a messenger ribonucleic acid.
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Pharmacology & Pharmacy (AREA)
- Medicinal Chemistry (AREA)
- Animal Behavior & Ethology (AREA)
- General Health & Medical Sciences (AREA)
- Public Health (AREA)
- Veterinary Medicine (AREA)
- Epidemiology (AREA)
- Engineering & Computer Science (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Molecular Biology (AREA)
- Dermatology (AREA)
- Physics & Mathematics (AREA)
- Biochemistry (AREA)
- Biomedical Technology (AREA)
- Nanotechnology (AREA)
- Optics & Photonics (AREA)
- Urology & Nephrology (AREA)
- Chemical Kinetics & Catalysis (AREA)
- General Chemical & Material Sciences (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Organic Chemistry (AREA)
- Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
Abstract
The present disclosure describes compositions and methods of manufacturing and using the same for a population of particles that can be used as a delivery vehicle for medical agents. The population of particles can be polymeric particles. The population of particles can selectively target or localize to a specific tissue, to deliver or localize the medical agents to the same specific tissue selectively. The population of particles can encapsulate therapeutic agents, such as a nucleic acid molecule (e.g., a cytosine-phosphate-guanosine (CpG) oligodeoxynucleotide (ODN) as a target receptor antagonist).
Description
COMPOSITIONS COMPRISING NANOPARTICLES AND METHODS THEREOF
CROSS-REFERENCE
[0001] This application claims the benefit of U.S. Patent Application No. 62/969,528, filed February 3, 2020, which is incorporated herein by reference in its entirety.
BACKGROUND
[0002] Medical agents can be administered to a subject for, e.g., diagnostics or therapeutic purposes. Therapeutic agents (e.g., small molecules, polynucleotides, polypeptides, etc.) can be administered to the subject in need thereof to treat a psychological disease or a physiological disease, such as neurological disorders or cancer. Imaging agents (e.g., dyes) can be administered to enhance a region of interest for noninvasive imaging and disease diagnostics. Some medical agents can be metabolized in the subject’s body or eliminated before adequate plasma concentrations are reached, thus resulting in low bioavailability and low efficacy. Increasing a therapeutic dose of such medical agents to compensate for the low bioavailability or efficacy can lead to other complications, e.g., kidney damage or other side-effects.
INCORPORATION BY REFERENCE
[0003] Each patent, publication, and non-patent literature cited in the application is hereby incorporated by reference in its entirety as if each was incorporated by reference individually.
SUMMARY OF THE INVENTION
[0004] In some embodiments, the present disclosure provides a composition comprising a plurality of polymeric nanoparticles, wherein: (i) a polymeric nanoparticle of the plurality of polymeric nanoparticles contains a nucleic acid molecule that binds to a polypeptide; and (ii) the plurality of polymeric nanoparticles is characterize by an average diameter of no greater than about 1 micrometer, as ascertained by dynamic light scattering.
[0005] In some embodiments, the present disclosure provides a composition comprising a plurality of polymeric nanoparticles, wherein: (i) a nanoparticle of the plurality of nanoparticles contains a nucleic acid molecule that binds to a polypeptide; and (ii) each nanoparticle of the plurality of nanoparticles independently comprises a polymeric structure, wherein the polymeric structure comprises a population of polymers, wherein the population of polymers has an average molar mass that is at least about 10 kilodaltons (kDa).
[0006] In some embodiments, the present disclosure provides a composition comprising a
plurality of polymeric nanop articles, wherein: (i) a polymeric nanoparticle of the plurality of polymeric nanoparticles contains a medical agent; and (ii) the polymeric nanoparticle is coated with a surfactant.
[0007] In some embodiments, the present disclosure provides a composition comprising a plurality of polymeric nanoparticles, wherein: (i) a nanoparticle of the plurality of nanoparticles contains a nucleic acid molecule that binds to a polypeptide; and (ii) if, in a controlled study, the nanoparticle is administered to a subject, then the nanoparticle exhibits selective targeting to a targeted tissue of the subject preferentially over a non-targeted tissue of the subject.
[0008] In some embodiments, the present disclosure provides a method comprising forming a plurality of nanoparticles, wherein: (i) a polymeric nanoparticle of the plurality of polymeric nanoparticles contains a nucleic acid molecule that binds to a polypeptide; and (ii) the plurality of polymeric nanoparticles is characterize by an average diameter of no greater than about 1 micrometer, as ascertained by dynamic light scattering.
[0009] In some embodiments, the present disclosure provides a method comprising forming a plurality of nanoparticles, wherein: (i) a nanoparticle of the plurality of nanoparticles contains a nucleic acid molecule that binds to a polypeptide; and (ii) each nanoparticle of the plurality of nanoparticles independently comprises a polymeric structure, wherein the polymeric structure comprises a population of polymers, wherein the population of polymers has an average molar mass that is at least about 10 kilodaltons (kDa).
[0010] In some embodiments, the present disclosure provides a method comprising forming a plurality of nanoparticles, wherein: (i) a polymeric nanoparticle of the plurality of polymeric nanoparticles contains a medical agent; and (ii) the polymeric nanoparticle coated with a surfactant.
[0011] In some embodiments, the present disclosure provides a method comprising forming a plurality of nanoparticles, wherein: (i) a polymeric nanoparticle of the plurality of polymeric nanoparticles contains a medical agent; and (ii) upon the contacting, the polymeric nanoparticle is coated with the surfactant.
[0012] In some embodiments, the present disclosure provides a method comprising forming a plurality of nanoparticles, wherein: (i) a nanoparticle of the plurality of nanoparticles contains a nucleic acid molecule that binds to a polypeptide; and (ii) if, in a controlled study, the nanoparticle is administered to a subject, then the nanoparticle exhibits selective targeting to a targeted tissue of the subject preferentially over a non-targeted tissue of the subject.
[0013] In some embodiments, the present disclosure provides a composition comprising a plurality of polymeric nanoparticles, wherein (i) a polymeric nanoparticle of the plurality of
polymeric nanoparticles contains a deoxyribonucleic acid molecule that binds to a target moiety; and (ii) the plurality of polymeric nanoparticles is characterized by an average diameter of no greater than about 1 micrometer, as ascertained by dynamic light scattering.
[0014] In some embodiments, the present disclosure provides a composition comprising a plurality of nanoparticles, wherein: (i) a nanoparticle of the plurality of nanoparticles contains a deoxyribonucleic acid molecule that binds to a target moiety; and (ii) each nanoparticle of the plurality of nanoparticles independently comprises a polymeric structure, wherein the polymeric structure comprises a population of polymers, wherein the population of polymers has an average molar mass that is at least about 10 kilodaltons (kDa).
[0015] In some embodiments, the present disclosure provides a composition comprising a plurality of nanoparticles, wherein: (i) a nanoparticle of the plurality of nanoparticles contains a deoxyribonucleic acid molecule that binds to a target moiety; and (ii) if, in a controlled study, the nanoparticle is administered to a subject, then the nanoparticle exhibits selective targeting to a targeted tissue of the subject preferentially over a non-targeted tissue of the subject.
[0016] In some embodiments, the present disclosure provides a method comprising forming a plurality of polymeric nanoparticles, wherein: (i) a polymeric nanoparticle of the plurality of polymeric nanoparticles contains a deoxyribonucleic acid molecule that binds to a target moiety; and (ii) the plurality of polymeric nanoparticles is characterized by an average diameter of no greater than about 1 micrometer, as ascertained by dynamic light scattering.
[0017] In some embodiments, the present disclosure provides a method comprising forming a plurality of nanoparticles, wherein: (i) a nanoparticle of the plurality of nanoparticles contains a deoxyribonucleic acid molecule that binds to a target moiety; and (ii) each nanoparticle of the plurality of nanoparticles independently comprises a polymeric structure, wherein the polymeric structure comprises a population of polymers, wherein the population of polymers has an average molar mass that is at least about 10 kilodaltons (kDa).
[0018] In some embodiments, the present disclosure provides a method comprising forming a plurality of polymeric nanoparticles, wherein: (i) a nanoparticle of the plurality of nanoparticles contains a deoxyribonucleic acid molecule that binds to a target moiety; and (ii) if, in a controlled study, the nanoparticle is administered to a subject, then the nanoparticle exhibits selective targeting to a targeted tissue of the subject preferentially over a non-targeted tissue of the subject.
BRIEF DESCRIPTION OF THE FIGURES
[0019] FIG. 1 shows histological staining of kidney sections to demonstrate co-localization of oligodeoxynucleotide (ODN)-encapsulating mesoscale nanoparticles (MNPs) and renal proximal tubular cells.
[0020] FIG. 2 shows kidney-selective targeting of MNPs as compared to other organs by brightfield and near-infrared imaging of various organs (top) and quantification of the near- infrared imaging (bottom).
[0021] FIG. 3 shows expression levels of plasma creatine (PCr), blood urea nitrogen (BUN), and neutrophil gelatinase-associated lipocalin (NGAL) messenger ribonucleic acid (mRNA) levels in kidney cells in mice treated with various MNPs or controls.
[0022] FIG. 4 shows representative hematoxylin and eosin (H&E) stained kidney sections from mice treated with various MNPs or controls.
[0023] FIG. 5 shows Jablonski score analysis of H&E stained kidney sections from mice treated with various MNPs or controls, to assess the extent of kidney tubular injury.
[0024] FIG. 6 shows representative terminal deoxynucleotidyl transferase deoxyuridine triphosphate nick end labeling (TUNEL) stained kidney sections from mice treated with various MNPs or controls.
[0025] FIG. 7 shows quantification of the TUNEL stained cells in the kidney sections from mice treated with various MNPs or controls, to assess the extent of kidney cell apoptosis.
[0026] FIG. 8 shows quantification of the caspase-3 and caspase-8 cleavage in kidney lysates from mice treated with various MNPs or controls, to assess the extent of kidney cell apoptosis. [0027] FIG. 9 shows quantification of the caspase-3 and caspase-8 cleavage in freshly isolated mouse proximal tubule cells that are treated with TLR9 activators, with or without pretreatment with various MNPs.
[0028] FIG. 10 shows representative Lymphocyte antigen 6 complex locus G6D (Ly6G) stained kidney sections from mice treated with various MNPs or controls, to assess the presence of immune cells (e.g., neutrophils) in the kidney.
[0029] FIG. 11 shows quantification of the Ly6G-positive immune cells in the kidney sections from mice treated with various MNPs or controls.
[0030] FIG. 12 shows representative F4/80 stained kidney sections from mice treated with various MNPs or controls, to assess the presence of immune cells (e.g., macrophages) in the kidney.
[0031] FIG. 13 shows quantification of the F4/80-positive immune cells in the kidney sections from mice treated with various MNPs or controls.
[0032] FIG. 14 shows quantification of various pro-inflammatory cytokine and chemokine mRNAs in kidney lysates from mice treated with various MNPs or controls.
DETAILED DESCRIPTION
[0033] The present invention provides compositions and methods of manufacturing and using the same for particles, for example, nanoparticles, such as mesoscale nanoparticles, for delivering medical agents, for example, therapeutic agents or imaging agents. The compositions of interest can be administered to a subject to deliver the medical agents for disease therapy or diagnosis. [0034] The following terms are used throughout as defined below.
[0035] As used herein and in the appended claims, singular articles such as “a” and “an” and “the” and similar referents in the context of describing the elements (especially in the context of the following claims) are to be construed to cover both the singular and the plural, unless otherwise indicated herein or clearly contradicted by context. Recitation of ranges of values herein are merely intended to serve as a shorthand method of referring individually to each separate value falling within the range, unless otherwise indicated herein, and each separate value is incorporated into the specification as if it were individually recited herein. All methods described herein can be performed in any suitable order unless otherwise indicated herein or otherwise clearly contradicted by context. The use of any and all examples, or exemplary language ( e.g ., “such as”) provided herein, is intended merely to better illuminate the embodiments and does not pose a limitation on the scope of the claims unless otherwise stated. No language in the specification should be construed as indicating any non-claimed element as essential.
[0036] As used herein, “about” will be understood by persons of ordinary skill in the art and will vary to some extent depending upon the context in which it is used. If there are uses of the term which are not clear to persons of ordinary skill in the art, given the context in which it is used, “about” will mean up to plus or minus 10% of the particular term - for example, “about 10 wt.%” would be understood to mean “9 wt.% to 11 wt.%.” It is to be understood that when “about” precedes a term, the term is to be construed as disclosing “about” the term as well as the term without modification by “about” - for example, “about 10 wt.%” discloses “9 wt.% to 11 wt.%” as well as disclosing “10 wt.%.”
[0037] The phrase “and/or” as used in the present disclosure will be understood to mean any one of the recited members individually or a combination of any two or more thereof - for example, “A, B, and/or C” would mean “A, B, C, A and B, A and C, or B and C.”
[0038] As will be understood by one skilled in the art, for any and all purposes, particularly in
terms of providing a written description, all ranges disclosed herein also encompass any and all possible subranges and combinations of subranges thereof. Any listed range can be easily recognized as sufficiently describing and enabling the same range being broken down into at least equal halves, thirds, quarters, fifths, tenths, etc. As a non-limiting example, each range discussed herein can be readily broken down into a lower third, middle third and upper third, etc. As will also be understood by one skilled in the art all language such as “up to,” “at least,” “greater than,” “less than,” and the like include the number recited and refer to ranges which can be subsequently broken down into subranges as discussed above. Finally, as will be understood by one skilled in the art, a range includes each individual member. Thus, for example, a group having 1-3 atoms refers to groups having 1, 2, or 3 atoms. Similarly, a group having 1-5 atoms refers to groups having 1, 2, 3, 4, or 5 atoms, and so forth.
Overview
[0039] In some embodiments, the invention provides a composition comprising a population of particles. In some embodiments, a particle of the population of particles comprises a medical agent. In some embodiments, the population of particles comprises a population of nanoparticles. In some embodiments, the population of nanoparticles comprises a population of mesoscale nanoparticles. In some embodiments, the medical agent is encapsulated within the nanoparticle (e.g., within a surface of the nanoparticle) or is coupled (e.g., covalently or non- covalently) to a surface of the nanoparticle. In some embodiments, the population of nanoparticles comprising (e.g., encapsulating) the medical agent is administered to a subject in need thereof. In some embodiments, the medical agent delivered to the subject via the population of nanoparticles is characterized by a mean plasma half-life that is greater than that of a control medical agent (e.g., a non-encapsulated medical agent, encapsulated by a different population of nanoparticles, etc.), as ascertained by tracing a concentration profile of the medical agent in the blood over time. In some embodiments, upon administration into a subject, the population of nanoparticles exhibits specificity towards one or more organs (e.g., kidney) over the other organs (e.g., heart, brain, lung, spleen, liver, etc.). In some embodiments, the medical agent comprises a nucleic acid molecule, e.g., a single-strand nucleic acid molecule. In some embodiments, the nucleic acid molecule comprises an oligodeoxynucleotide (ODN). In some embodiments, the nucleic acid molecule comprises an unmethylated cytosine-phosphate- guanosine (CpG) ODN.
Compositions and methods of the invention
[0040] The composition of the disclosure can comprise a population of particles. A particle of the population of particles can be a solid particle, a hollow particle, or a porous particle. A particle of the population of particles can comprise one or more layers (e.g., at least or up to about 1, at least or up to about 2, at least or up to about 3, at least or up to about 4, or at least or up to about 5 layers of the same or different materials). Non-limiting examples of a particle of the population of particles include a liposome, a micelle, an emulsion, a vesicle, a nanoparticle, a nanocapsule, a virosome, a lipoprotein, a polymersome, a liquid crystal, and mixtures thereof. A particle of the population of particles can comprise organic materials (e.g., synthetic or natural polymers, nucleotides, polynucleotides, amino acids, polypeptides, etc.) or inorganic materials, such as metals (e.g., aluminum, titanium, steel) or ceramics (e.g., metal oxides). A particle of the population of particles can have various shapes and sizes. For example, a particle can be in the shape of a sphere, cuboid, or disc, or any partial or irregular shape or combination of shapes. The particle can have a cross-section that is circular, triangular, square, rectangular, pentagonal, hexagonal, or any partial or irregular shape or combination of shapes.
[0041] The population of particles of the disclosure can comprise a metallic material. Nonlimiting examples of the metallic material include aluminum, calcium, magnesium, barium, scandium, titanium, vanadium, chromium, manganese, iron, cobalt, nickel, copper, zinc, yttrium, niobium, molybdenum, ruthenium, rhodium, silver, cadmium, actinium, and gold. In some embodiments, the particles comprise a rare earth element.
[0042] The population of particles of the present invention can comprise a ceramic material. Non-limiting examples of the ceramic material include an aluminide, boride, beryllia, carbide, chromium oxide, hydroxide, iron oxide, sulfide, nitride, mullite, kyanite, ferrite, titania zirconia, yttria, and magnesia.
[0043] The population of particles of the disclosure can comprise a polymer, e.g., a biodegradable polymer. Non-limiting examples of the polymer include polylactic acid (PLA), polyglycolic acid (PGA), polylactic-polyglycolic copolymer (PLGA), poly-D,L-lactide-co- glycolide (PLGA), PLGA-ethylene oxide fumarate, PLGA-alpha-tocopheryl succinate esterified to polyethylene glycol 1000 (PLGA-TGPS), polyanhydride poly[l,6-bis(p- carboxyphenoxy)hexane] (pCPH), poly(hydroxbutyric acid-cohydroxyvaleric acid) (PHB-PVA), polyethylene glycol (PEG)-poly (lactic acid) (PLA) copolymer (PEG-PLA), poly-e-caprolactone (PCL), poly-alkyl-cyano-acrylate (PAC), poly(ethyl)cyanoacrylate (PEC), polyisobutyl cyanoacrylate, poly-N-(2-hydroxypropyl)methacrylamide (poly(HPMA)), poly-P-R-hydroxy butyrate (PHB), poly-P-R-hydroxy alkanoate (PHA), poly-P-R-malic acid, phospholipid- cholesterol polymers, 2-dioleoyl-sn-glycero-3 -phosphatidylcholine/poly ethyleneglycol-
distearoylphosphatidylehtanolamine (DOPC/PEG-DSPE)/Cholesterol, polysaccharides, cellulose, ethyl cellulose, methyl cellulose, alginates, dextran and dextran hydrogel polymers, amylose, inulin, pectin and guar gum, chitosan, chitin, heparin, hyaluronic acid, cyclodextrin (CD)-based polyrotaxanes and polypseudorotaxanes, polyaspartates, polyglutamates, polylucine, leucine- glutamate co-polymers, polybutylene succinate (PBS), gelatin, collagens, fibrins, fibroin, polyorthoesters, polyorthoester-polyamidine copolymer, polyorthoester-diamine copolymers, polyorthoesters incorporating latent acids, poly(ethylene glycol)/poly(butylene terephthalate) copolymer, derivatives thereof, and non-covalent combinations or covalent combinations (e.g., copolymers) thereof. In an example, each particle of the population of particle comprises PLGA. [0044] The polymers of the disclosure can comprise copolymers, for example, bipolymers, terpolymers, and quaterpolymers. In some embodiments, the polymer is PLGA-PEG copolymer. The copolymers can comprise alternating copolymers, random copolymers, statistical copolymers, segmented polymers, block copolymers, multiblock copolymers, gradient copolymers, graft copolymers, star copolymers, branched copolymers, hyperbranched copolymers, and combinations thereof. The copolymers can comprise two or more species of repeating unit that are different from one another. The copolymers can comprise at least or up to 2 different species of repeating unit, at least or up to 3 different species of repeating unit, at least or up to 4 different species of repeating unit, at least or up to 5 different species of repeating unit, at least or up to 6 different species of repeating unit, at least or up to 7 different species of repeating unit, at least or up to 8 different species of repeating unit, at least or up to 9 different species of repeating unit, or at least or up to 10 different species of repeating unit.
[0045] An average molar mass (e.g., a number average molar mass as determined by gel permeation chromatography (GPC)) of a polymer (e.g., synthetic or natural polymers, polynucleotides, polypeptides, etc.) of the disclosure can be at least or up to about 1 kilodaltons (kDa), at least or up to about 2 kDa, at least or up to about 3 kDa, at least or up to about 4 kDa, at least or up to about 5 kDa, at least or up to about 6 kDa, at least or up to about 7 kDa, at least or up to about 8 kDa, at least or up to about 9 kDa, at least or up to about 10 kDa, at least or up to about 20 kDa, at least or up to about 30 kDa, at least or up to about 40 kDa, at least or up to about 50 kDa, at least or up to about 60 kDa, at least or up to about 70 kDa, at least or up to about 80 kDa, at least or up to about 90 kDa, at least or up to about 100 kDa, at least or up to about 200 kDa, at least or up to about 300 kDa, at least or up to about 400 kDa, at least or up to about 500 kDa, at least or up to about 600 kDa, at least or up to about 700 kDa, at least or up to about 800 kDa, at least or up to about 900 kDa, or at least or up to about 1,000 kDa.
[0046] In some embodiments, a number average molar mass of the polymer (e.g., PLGA-PEG)
of the disclosure can be about 10 kDa to about 100 kDa. In some embodiments, a number average molar mass of the polymer can be at least about 10 kDa. In some embodiments, a number average molar mass of the polymer can be at most about 100 kDa. In some embodiments, a number average molar mass of the polymer can be about 10 kDa to about 20 kDa, about 10 kDa to about 30 kDa, about 10 kDa to about 40 kDa, about 10 kDa to about 50 kDa, about 10 kDa to about 60 kDa, about 10 kDa to about 70 kDa, about 10 kDa to about 80 kDa, about 10 kDa to about 90 kDa, about 10 kDa to about 100 kDa, about 20 kDa to about 30 kDa, about 20 kDa to about 40 kDa, about 20 kDa to about 50 kDa, about 20 kDa to about 60 kDa, about 20 kDa to about 70 kDa, about 20 kDa to about 80 kDa, about 20 kDa to about 90 kDa, about 20 kDa to about 100 kDa, about 30 kDa to about 40 kDa, about 30 kDa to about 50 kDa, about 30 kDa to about 60 kDa, about 30 kDa to about 70 kDa, about 30 kDa to about 80 kDa, about 30 kDa to about 90 kDa, about 30 kDa to about 100 kDa, about 40 kDa to about 50 kDa, about 40 kDa to about 60 kDa, about 40 kDa to about 70 kDa, about 40 kDa to about 80 kDa, about 40 kDa to about 90 kDa, about 40 kDa to about 100 kDa, about 50 kDa to about 60 kDa, about 50 kDa to about 70 kDa, about 50 kDa to about 80 kDa, about 50 kDa to about 90 kDa, about 50 kDa to about 100 kDa, about 60 kDa to about 70 kDa, about 60 kDa to about 80 kDa, about 60 kDa to about 90 kDa, about 60 kDa to about 100 kDa, about 70 kDa to about 80 kDa, about 70 kDa to about 90 kDa, about 70 kDa to about 100 kDa, about 80 kDa to about 90 kDa, about 80 kDa to about 100 kDa, or about 90 kDa to about 100 kDa. In some embodiments, a number average molar mass of the polymer can be about 10 kDa, about 20 kDa, about 30 kDa, about 40 kDa, about 50 kDa, about 60 kDa, about 70 kDa, about 80 kDa, about 90 kDa, or about 100 kDa.
[0047] A particle of the population of particles can comprise a single material (e.g., a single polymer, such as PLGA or PLGA-PEG). In some embodiments, a particle of the population of particles comprises at least or up to about 2 different materials, at least or up to about 3 different materials, at least or up to about 4 different materials, at least or up to about 5 different materials, at least or up to about 6 different materials, at least or up to about 7 different materials, at least or up to about 8 different materials, at least or up to about 9 different materials, or at least or up to about 10 different materials. In some embodiments, the plurality of different materials can be homogeneously mixed within the particle. In some embodiments, the plurality of different materials can be heterogeneously mixed within the particle.
[0048] The particle of the population of particles can be a core-shell particle. For example, a copolymer (e.g., PLGA-PEG) comprising a first polymer (e.g., PLGA) and a second polymer (e.g., PEG) can self-assemble or precipitate to form a population of core-shell nanoparticles. In
some cases, the core can be substantially made of the first polymer (e.g., PLGA), and the shell can be substantially made of the second polymer (e.g., PEG).
[0049] In some embodiments, the population of particles of the present invention are polymeric particles (e.g., particles comprising a polymeric core). In some embodiments, the particles of the population of particles are not liposomes or micelles. In some embodiments, the particles of the population of particles are not micelles.
[0050] The population of particles in a composition (for example, single dose for administration to a subject, such as a patient in need thereof) can comprise at least or up to about 1 particle, at least or up to about 5 particles, at least or up to about 10 particles, at least or up to about 20 particles, at least or up to about 50 particles, at least or up to about 100 particles, at least or up to about 200 particles, at least or up to about 500 particles, at least or up to about 1,000 particles, at least or up to about 2,000 particles, at least or up to about 5,000 particles, at least or up to about 10,000 particles, at least or up to about 20,000 particles, at least or up to about 50,000 particles, at least or up to about 100,000 particles, at least or up to about 200,000 particles, or at least or up to about 500,000 particles.
[0051] A cross-sectional dimension (e.g., an average diameter as ascertained by dynamic light scattering (DLS) in aqueous solution, e.g., saline solution) of a particle of the population of particles can range between about 100 nanometers (nm) to about 1000 nm. The cross-sectional dimension of the particle can be at least or up to about 100 nm, at least or up to about 200 nm, at least or up to about 300 nm, at least or up to about 400 nm, at least or up to about 500 nm, at least or up to about 600 nm, at least or up to about 700 nm, at least or up to about 800 nm, at least or up to about 900 nm, or at least or up to about 1,000 nm. In some embodiments, the cross- sectional dimension of the particle (e.g., a mesoscale nanoparticle (MNP)) can be at least or up to about 200 nm, at least or up to about 250 nm, at least or up to about 300 nm, at least or up to about 350 nm, at least or up to about 400 nm, at least or up to about 450 nm, or at least or up to about 500 nm.
[0052] In some embodiments, a cross-sectional dimension (e.g., an average diameter as ascertained by DLS in aqueous solution, e.g., saline solution) of a particle of the population of particles can range between about 100 nm to about 600 nm. The average diameter of the particle of the population of particles can range between about 100 nm to about 600 nm. The average diameter of the particle of the population of particles can range between about at least about 100 nm. The average diameter of the particle of the population of particles can range between about at most about 600 nm. The average diameter of the particle of the population of particles can range between about 100 nm to about 200 nm, about 100 nm to about 300 nm, about 100 nm to
about 400 nm, about 100 nm to about 500 nm, about 100 nm to about 600 nm, about 200 nm to about 300 nm, about 200 nm to about 400 nm, about 200 nm to about 500 nm, about 200 nm to about 600 nm, about 300 nm to about 400 nm, about 300 nm to about 500 nm, about 300 nm to about 600 nm, about 400 nm to about 500 nm, about 400 nm to about 600 nm, or about 500 nm to about 600 nm. The average diameter of the particle of the population of particles can range between about 100 nm, about 200 nm, about 300 nm, about 400 nm, about 500 nm, or about 600 nm.
[0053] A mean cross-sectional dimension (e.g., an average diameter as ascertained by DLS in aqueous solution, e.g., saline solution) of the population of particles can range between about 100 nanometers (nm) to about 1000 nm. The mean cross-sectional dimension of the population of particles can be at least or up to about 100 nm, at least or up to about 200 nm, at least or up to about 300 nm, at least or up to about 400 nm, at least or up to about 500 nm, at least or up to about 600 nm, at least or up to about 700 nm, at least or up to about 800 nm, at least or up to about 900 nm, or at least or up to about 1,000 nm. In some embodiments, the mean cross- sectional dimension of the population of particles (e.g., a population of MNPs) can be at least or up to about 200 nm, at least or up to about 250 nm, at least or up to about 300 nm, at least or up to about 350 nm, at least or up to about 400 nm, at least or up to about 450 nm, or at least or up to about 500 nm. The mean cross-sectional dimension can be ascertained from a population of at least or up to about 100 particles, at least or up to about 500 particles, at least or up to about 1,000 particles, at least or up to about 2,000 particles, at least or up to about 5,000 particles, at least or up to about 10,000 particles, at least or up to about 20,000 particles, at least or up to about 50,000 particles, at least or up to about 100,000 particles, at least or up to about 200,000 particles, or at least or up to about 500,000 particles.
[0054] A surface charge (e.g., zeta potential measurement as ascertained by DLS in aqueous solution, e.g., saline solution) of a particle of the population of particles can range between about -100 millivolt (mV) to about +100 mV. The surface charge of the particle of the population of particles can be at least or up to about -100 mV, at least or up to about -90 mV, at least or up to about -80 mV, at least or up to about -70 mV, at least or up to about -60 mV, at least or up to about -50 mV, at least or up to about -45 mV, at least or up to about -40 mV, at least or up to about -35 mV, at least or up to about -30 mV, at least or up to about -25 mV, at least or up to about -20 mV, at least or up to about -15 mV, at least or up to about -10 mV, at least or up to about -5 mV, at least or up to about 0 mV, at least or up to about +5 mV, at least or up to about +10 mV, at least or up to about +15 mV, at least or up to about +20 mV, at least or up to about +25 mV, at least or up to about +30 mV, at least or up to about +35 mV, at least or up to about
+40 mV, at least or up to about +45 mV, at least or up to about +50 mV, at least or up to about +60 mV, at least or up to about +70 mV, at least or up to about +80 mV, at least or up to about +90 mV, or at least or up to about +100 mV.
[0055] The population of particles of the present invention can comprise a population of multiple-emulsion particles. The multiple-emulsion particles can be particles (e.g., polymeric particles, such as PLGA-PEG based core-shell particles as disclosed herein) comprising one or more outer layers (e.g., at least or up to about 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 outer layers). In some embodiments, the multiple-emulsion particles can be generated by (i) a first emulsion for forming polymeric particles (e.g., polymeric mesoscale nanoparticles) comprising one or more therapeutic agents in a mixture and (ii) a subsequent second emulsion to form one or more outer layers around the polymeric particles (i.e., double-emulsion particles). For example, a population of polymers (e.g., PLGA-PEG) and a population of therapeutic agents (e.g., polynucleotides) are mixed in a first mixture to form a population of polymeric nanoparticles. Subsequently, the population of nanoparticles and a surfactant (which surfactant is different than a polymer of the population of polymers) are mixed in a second mixture to form one or more outer layers around the polymeric nanoparticles. The second solution for the subsequent second emulsion can comprise a surfactant. The surfactant can comprise a non-ionic surfactant. The non-ionic surfactant may not be a small molecule. The non-ionic surfactant can be a polymer, e.g., polyoxyethylene-polyoxypropylene block copolymer (i.e., Pluronic F-68).
[0056] The population of particles of the present invention can comprise a medical agent, such as a therapeutic agent or an imaging agent. The population of particles can comprise at least or up to about 1 species of a medical agent, at least or up to about 2 species of medical agents, at least or up to about 3 species of medical agents, at least or up to about 4 species of medical agents, or at least or up to about 5 species of medical agents.
[0057] The medical agents can be present in an average quantity per particle of at least or up to about 0.1 nanogram (ng), at least or up to about 0.5 ng, at least or up to about 1 ng, at least or up to about 5 ng, at least or up to about 10 ng, at least or up to about 50 ng, at least or up to about 100 ng, at least or up to about 500 ng, at least or up to about 1 microgram (μg), at least or up to about 5 μg, at least or up to about 10 μg, at least or up to about 50 μg, at least or up to about 100 μg, at least or up to about 500 μg, or at least or up to about 1,000 μg. The medical agents can be present in an average quantity per particle of at least or up to about 0.1 %, at least or up to about 0.2 %, at least or up to about 0.3 %, at least or up to about 0.4 %, at least or up to about 0.5 %, at least or up to about 0.6 %, at least or up to about 0.7 %, at least or up to about 0.8 %, at least or up to about 0.9 %, at least or up to about 1 %, at least or up to about 2 %, at least or up to about 3
%, at least or up to about 4 %, at least or up to about 5 %, at least or up to about 6 %, at least or up to about 7 %, at least or up to about 8 %, at least or up to about 9 %, at least or up to about 10 %, at least or up to about 11 %, at least or up to about 12 %, at least or up to about 13 %, at least or up to about 14 %, at least or up to about 15 %, at least or up to about 16 %, at least or up to about 17 %, at least or up to about 18 %, at least or up to about 19 %, at least or up to about 20 %, at least or up to about 25 %, at least or up to about 30 %, at least or up to about 35 %, at least or up to about 40 %, at least or up to about 45 %, or at least or up to about 50 % by a mean weight of the particle.
[0058] The medical agents can be present in an average quantity per particle of at least or up to about 0.1 ng, at least or up to about 0.5 ng, at least or up to about 1 ng, at least or up to about 5 ng, at least or up to about 10 ng, at least or up to about 50 ng, at least or up to about 100 ng, at least or up to about 500 ng, at least or up to about 1 μg, at least or up to about 5 μg, at least or up to about 10 μg, at least or up to about 50 μg, at least or up to about 100 μg, at least or up to about 500 μg, or at least or up to about 1,000 μg in a population of particles. The medical agents can be present in an average quantity of at least or up to about 0.1 %, at least or up to about 0.2 %, at least or up to about 0.3 %, at least or up to about 0.4 %, at least or up to about 0.5 %, at least or up to about 0.6 %, at least or up to about 0.7 %, at least or up to about 0.8 %, at least or up to about 0.9 %, at least or up to about 1 %, at least or up to about 2 %, at least or up to about 3 %, at least or up to about 4 %, at least or up to about 5 %, at least or up to about 6 %, at least or up to about 7 %, at least or up to about 8 %, at least or up to about 9 %, at least or up to about 10 %, at least or up to about 11 %, at least or up to about 12 %, at least or up to about 13 %, at least or up to about 14 %, at least or up to about 15 %, at least or up to about 16 %, at least or up to about 17 %, at least or up to about 18 %, at least or up to about 19 %, at least or up to about 20 %, at least or up to about 25 %, at least or up to about 30 %, at least or up to about 35 %, at least or up to about 40 %, at least or up to about 45 %, or at least or up to about 50 % by weight of the particle in a population of particles.
[0059] In some embodiments, encapsulated medical agent (e.g., encapsulated in a particle of the population of particles) can be present in an average quantity of at least or up to about 0.1 ng, at least or up to about 0.5 ng, at least or up to about 1 ng, at least or up to about 5 ng, at least or up to about 10 ng, at least or up to about 50 ng, at least or up to about 100 ng, at least or up to about 500 ng, at least or up to about 1 μg, at least or up to about 5 μg, at least or up to about 10 μg, at least or up to about 50 μg, at least or up to about 100 μg, at least or up to about 500 μg, or at least or up to about 1,000 μg per particle or per particle in a population of particles. In some embodiments, encapsulated medical agents (e.g., encapsulated in a particle of the population of
particles) can be present in an average quantity of at least or up to about 0.1 %, at least or up to about 0.2 %, at least or up to about 0.3 %, at least or up to about 0.4 %, at least or up to about 0.5 %, at least or up to about 0.6 %, at least or up to about 0.7 %, at least or up to about 0.8 %, at least or up to about 0.9 %, at least or up to about 1 %, at least or up to about 2 %, at least or up to about 3 %, at least or up to about 4 %, at least or up to about 5 %, at least or up to about 6 %, at least or up to about 7 %, at least or up to about 8 %, at least or up to about 9 %, at least or up to about 10 %, at least or up to about 11 %, at least or up to about 12 %, at least or up to about 13 %, at least or up to about 14 %, at least or up to about 15 %, at least or up to about 16 %, at least or up to about 17 %, at least or up to about 18 %, at least or up to about 19 %, at least or up to about 20 %, at least or up to about 25 %, at least or up to about 30 %, at least or up to about 35 %, at least or up to about 40 %, at least or up to about 45 %, or at least or up to about 50 % by weight of the particle in a population of particles.
[0060] In some embodiments, medical agent coated on a surface or within a surface layer of the of particles can be present in an average quantity of at least or up to about 0.1 ng, at least or up to about 0.5 ng, at least or up to about 1 ng, at least or up to about 5 ng, at least or up to about 10 ng, at least or up to about 50 ng, at least or up to about 100 ng, at least or up to about 500 ng, at least or up to about 1 μg, at least or up to about 5 μg, at least or up to about 10 μg, at least or up to about 50 μg, at least or up to about 100 μg, at least or up to about 500 μg, or at least or up to about 1,000 μg per particle or per particle in a population of particles. In some embodiments, medical agent coated on a surface or within a surface layer of the of particles can be present in an average quantity of at least or up to about 0.1 %, at least or up to about 0.2 %, at least or up to about 0.3 %, at least or up to about 0.4 %, at least or up to about 0.5 %, at least or up to about 0.6 %, at least or up to about 0.7 %, at least or up to about 0.8 %, at least or up to about 0.9 %, at least or up to about 1 %, at least or up to about 2 %, at least or up to about 3 %, at least or up to about 4 %, at least or up to about 5 %, at least or up to about 6 %, at least or up to about 7 %, at least or up to about 8 %, at least or up to about 9 %, at least or up to about 10 %, at least or up to about 11 %, at least or up to about 12 %, at least or up to about 13 %, at least or up to about 14 %, at least or up to about 15 %, at least or up to about 16 %, at least or up to about 17 %, at least or up to about 18 %, at least or up to about 19 %, at least or up to about 20 %, at least or up to about 25 %, at least or up to about 30 %, at least or up to about 35 %, at least or up to about 40 %, at least or up to about 45 %, or at least or up to about 50 % by weight of the particle in a population of particles.
[0061] The medical agents can be present in an average quantity of at least or up to about 0.1 ng, at least or up to about 0.5 ng, at least or up to about 1 ng, at least or up to about 5 ng, at least or
up to about 10 ng, at least or up to about 50 ng, at least or up to about 100 ng, at least or up to about 500 ng, at least or up to about 1 μg, at least or up to about 5 μg, at least or up to about 10 μg, at least or up to about 50 μg, at least or up to about 100 μg, at least or up to about 500 μg, at least or up to about 1 milligram (mg), at least or up to about 5 mg, at least or up to about 10 mg, at least or up to about 50 mg, or at least or up to about 100 mg per composition. The medical agents can be present in an average quantity of at least or up to about 0.1 %, at least or up to about 0.2 %, at least or up to about 0.3 %, at least or up to about 0.4 %, at least or up to about 0.5 %, at least or up to about 0.6 %, at least or up to about 0.7 %, at least or up to about 0.8 %, at least or up to about 0.9 %, at least or up to about 1 %, at least or up to about 2 %, at least or up to about 3 %, at least or up to about 4 %, at least or up to about 5 %, at least or up to about 6 %, at least or up to about 7 %, at least or up to about 8 %, at least or up to about 9 %, at least or up to about 10 %, at least or up to about 11 %, at least or up to about 12 %, at least or up to about 13 %, at least or up to about 14 %, at least or up to about 15 %, at least or up to about 16 %, at least or up to about 17 %, at least or up to about 18 %, at least or up to about 19 %, at least or up to about 20 %, at least or up to about 25 %, at least or up to about 30 %, at least or up to about 35 %, at least or up to about 40 %, at least or up to about 45 %, or at least or up to about 50 % by weight of the composition.
[0062] In some embodiments, encapsulated medical agents (e.g., encapsulated in a particle of the population of particles of the composition) can be present in an average quantity of at least or up to about 0.1 ng, at least or up to about 0.5 ng, at least or up to about 1 ng, at least or up to about 5 ng, at least or up to about 10 ng, at least or up to about 50 ng, at least or up to about 100 ng, at least or up to about 500 ng, at least or up to about 1 μg, at least or up to about 5 μg, at least or up to about 10 μg, at least or up to about 50 μg, at least or up to about 100 μg, at least or up to about 500 μg, at least or up to about 1 milligram (mg), at least or up to about 5 mg, at least or up to about 10 mg, at least or up to about 50 mg, or at least or up to about 100 mg per composition. In some embodiments, encapsulated medical agents (e.g., encapsulated in a particle of the population of particles of the composition) can be present in an average quantity of at least or up to about 0.1 %, at least or up to about 0.2 %, at least or up to about 0.3 %, at least or up to about 0.4 %, at least or up to about 0.5 %, at least or up to about 0.6 %, at least or up to about 0.7 %, at least or up to about 0.8 %, at least or up to about 0.9 %, at least or up to about 1 %, at least or up to about 2 %, at least or up to about 3 %, at least or up to about 4 %, at least or up to about 5 %, at least or up to about 6 %, at least or up to about 7 %, at least or up to about 8 %, at least or up to about 9 %, at least or up to about 10 %, at least or up to about 11 %, at least or up to about 12 %, at least or up to about 13 %, at least or up to about 14 %, at least or up to about 15 %, at least or
up to about 16 %, at least or up to about 17 %, at least or up to about 18 %, at least or up to about 19 %, at least or up to about 20 %, at least or up to about 25 %, at least or up to about 30 %, at least or up to about 35 %, at least or up to about 40 %, at least or up to about 45 %, or at least or up to about 50 % by weight of the composition.
[0063] In some embodiments, medical agent coated on a surface or within a surface layer of the of particles can be present in an average quantity of at least or up to about 0.1 ng, at least or up to about 0.5 ng, at least or up to about 1 ng, at least or up to about 5 ng, at least or up to about 10 ng, at least or up to about 50 ng, at least or up to about 100 ng, at least or up to about 500 ng, at least or up to about 1 μg, at least or up to about 5 μg, at least or up to about 10 μg, at least or up to about 50 μg, at least or up to about 100 μg, at least or up to about 500 μg, at least or up to about 1 milligram (mg), at least or up to about 5 mg, at least or up to about 10 mg, at least or up to about 50 mg, or at least or up to about 100 mg per composition. In some embodiments, medical agent coated on a surface or within a surface layer of the of particles can be present in an average quantity of at least or up to about 0.1 %, at least or up to about 0.2 %, at least or up to about 0.3 %, at least or up to about 0.4 %, at least or up to about 0.5 %, at least or up to about 0.6 %, at least or up to about 0.7 %, at least or up to about 0.8 %, at least or up to about 0.9 %, at least or up to about 1 %, at least or up to about 2 %, at least or up to about 3 %, at least or up to about 4 %, at least or up to about 5 %, at least or up to about 6 %, at least or up to about 7 %, at least or up to about 8 %, at least or up to about 9 %, at least or up to about 10 %, at least or up to about 11 %, at least or up to about 12 %, at least or up to about 13 %, at least or up to about 14 %, at least or up to about 15 %, at least or up to about 16 %, at least or up to about 17 %, at least or up to about 18 %, at least or up to about 19 %, at least or up to about 20 %, at least or up to about 25 %, at least or up to about 30 %, at least or up to about 35 %, at least or up to about 40 %, at least or up to about 45 %, or at least or up to about 50 % by weight of the composition. [0064] A medical agent of the disclosure can be a small molecule, a lipid, a nucleic acid, a polynucleotide, an amino acid, a polypeptide (e.g., a peptide or a protein), or combinations thereof. A medical agent can comprise a therapeutic agent. A medical agent can comprise an imaging agent.
[0065] In some embodiments, a therapeutic agent can comprise one or more members selected from alkylating agents, anti-EGFR antibodies, anti-Her-2 antibodies, antimetabolites, vinca alkaloids, anthracyclines, topoisomerases, taxanes, epothilones, antibiotics, immunomodulators, immune cell antibodies, interferons, interleukins, HSP90 inhibitors, anti-androgens, antiestrogens, anti-hypercalcaemia agents, apoptosis inducers, Aurora kinase inhibitors, Bruton's tyrosine kinase inhibitors, calcineurin inhibitors, CaM kinase II inhibitors, CD45 tyrosine
phosphatase inhibitors, CDC25 phosphatase inhibitors, cyclooxygenase inhibitors, cRAF kinase inhibitors, cyclin dependent kinase inhibitors, cysteine protease inhibitors, DNA intercalators, DNA strand breakers, E3 ligase inhibitors, EGF pathway inhibitors, farnesyltransferase inhibitors, Flk-1 kinase inhibitors, glycogen synthase kinase-3 inhibitors, histone deacetylase inhibitors, I-kappa B-alpha kinase inhibitors, imidazotetrazinones, insulin tyrosine kinase inhibitors, c-Jun-N-terminal kinase inhibitors, mitogen-activated protein kinase inhibitors,
MDM2 inhibitors, MEK inhibitors, MMP inhibitors, mTor inhibitors , NGFR tyrosine kinase inhibitors, p38 MAP kinase inhibitors, p56 tyrosine kinase inhibitors, PDGF pathway inhibitors, phosphatidylinositol-3 -kinase inhibitors, phosphatase inhibitors, protein phosphatase inhibitors, PKC inhibitors, PKC delta kinase inhibitors, poly amine synthesis inhibitors, proteasome inhibitors, PTP1B inhibitors, SRC family tyrosine kinase inhibitors, Syk tyrosine kinase inhibitors, Janus (JAK-2 and/or JAK-3) tyrosine kinase inhibitors, retinoids, RNA polymerase II elongation inhibitors, Serine/Threonine kinase inhibitors, sterol biosynthesis inhibitors, VEGF pathway inhibitors, immunosuppressive agents, CYP3 A4 inhibitors, phosphorylated ribosomal protein s6 (pS6) inhibitor, anti-microbial agents, and antiemetics.
[0066] In some embodiments, a therapeutic agent can be a statin, mTOR inhibitor (e.g., RAD001 (everolimus), rapamycin, etc.), TGF-b signaling agonist, TGF-b signaling inhibitor (e.g., LY2157299 (galunisertib), SD-208, SB505124), corticosteroid, inhibitor of mitochondrial function, metabolic pathway inhibitor (e.g., STF-31, CPI-613, or Fasentin), p38 mitogen- activated protein kinases (MAPK) inhibitor, p56 tyrosine kinase inhibitor, NF-KB inhibitor (e.g., Pyrrolidine dithiocarbamate, quinazoline, BMS-345541, BAY-11-7085), adenosine receptor agonist, prostaglandin E2 agonist, phosphodiesterase 4 inhibitor, HD AC inhibitor, alpha-7 nicotine receptor antagonist, proteasome inhibitor, reactive oxygen species and DNA damage inhibitor (e.g., Amifostine (Ethyol)), p21 inhibitor (e.g., siRNA or small-molecule inhibitor), glutaminase inhibitor (e.g., BPTES, CB-839), or pS6 inhibitor.
[0067] In some embodiments, a therapeutic agent can be a nucleic acid molecule. In some embodiments, the therapeutic agent is a single-stranded nucleic acid molecule. In some embodiments, the therapeutic agent is not a double-stranded nucleic acid molecule, such as two single stranded nucleic acid molecules binding to each other to form a complex (e.g., siRNA). Alternatively, the therapeutic agent is a double-stranded nucleic acid molecule. In some embodiments, the therapeutic agent is not siRNA. In some embodiments, the therapeutic agent is a nucleic acid molecule capable of binding to polypeptide. The polypeptide can be a receptor polypeptide. The receptor polypeptide can be disposed on a membrane of a cell. For example, the receptor polypeptide is disposed on a transmembrane of a cell. In another example, the
receptor polypeptide is disposed on a membrane of an endosome. In some embodiments, the therapeutic agent is a nucleic acid molecule that is not designed to exhibit complementarity to a polynucleotide sequence (e.g., genomic sequence, mRNA sequence, etc. of a cell). In some embodiments, the therapeutic agent is a deoxyribonucleic acid (DNA) molecule (e.g., an oligodeoxynucleotide (ODN) molecule as disclosed herein). The DNA molecule can be configured to target a polynucleotide (e.g., a target gene). The targeted polynucleotide can be a DNA sequence. The targeted polynucleotide can be a RNA sequence, such as, for example, messenger RNA (mRNA). The DNA molecule as disclosed herein can be an antisense oligonucleotide.
[0068] The nucleic acid molecule (e.g., the antisense oligonucleotide) as disclosed herein can comprise sequence complementarity to a target polynucleotide sequence (e.g., mRNA sequence) by at least or up to about 50%, at least or up to about 55%, at least or up to about 60%, at least or up to about 65%, at least or up to about 70%, at least or up to about 75%, at least or up to about 80%, at least or up to about 85%, at least or up to about 90%, at least or up to about 95%, at least or up to about 99%, or about 100%.
[0069] In some embodiments, a nucleic acid molecule as disclosed herein is an oligodeoxynucleotide (ODN). An ODN can be single stranded or double stranded and the linkage can be for example phosphodiester or phosphorothioate. In some embodiments, an ODN is a strand of single-stranded synthetic nucleic acid molecule comprising at least one cytosine triphosphate deoxynucleotide followed by a guanine triphosphate deoxynucleotide connected through a phosphodiester or equivalent functional group (e.g. phosphorothioate linkage) motif (i.e., CpG), wherein the CpG is unmethylated (a CpG-ODN). The ODN strand can be from 3 to 55, for example from 12 to 24, or from 18 to 24, nucleotides long. For example, an ODN can be at least or up to about 3, at least or up to about 4, at least or up to about 5, at least or up to about 6, at least or up to about 7, at least or up to about 8, at least or up to about 9, at least or up to about 10, at least or up to about 11, at least or up to about 12, at least or up to about 13, at least or up to about 14, at least or up to about 15, at least or up to about 16, at least or up to about 17, at least or up to about 18, at least or up to about 19, at least or up to about 20, at least or up to about 21, at least or up to about 22, at least or up to about 23, or at least or up to about 24 nucleotides long. A CpG-ODN can be class A, class B, or class C.
[0070] In some embodiments, the receptor polypeptide that is targeted by the therapeutic agent as disclosed herein (e.g., CpG ODN) can be a regulator of innate immune system. The regulator of innate immune system can be a pattern recognition receptor (PRR), such as a toll-like receptor (TLR) and C-type lectin receptors (CLRs). Non-limiting examples of a TLR include TLR1,
TLR2, TLR3, TLR4, TLR5, TLR6, TLR7, TLR8, TLR9, TLR10, TLR11, TLR12, and TLR13.
In some examples, the receptor polypeptide that is targeted by the therapeutic agent as disclosed herein (e.g., CpG ODN) can be TLR9. In some embodiments, the therapeutic agent as disclosed herein can target a receptor polypeptide (e.g., TLR9) of a kidney cell (e.g., a human kidney cell). [0071] In some embodiments, the nucleic acid molecule as disclosed herein is SEQ ID NO: 1, which is the following sequence: 5 ' -TCCTGGCGGGGAAGT-3 ' (SEQ ID NO: 1). In some embodiments, the nucleic acid molecule is a TLR9 antagonist CpG-ODN.
[0072] In some embodiments, a therapeutic agent can comprise a chemotherapeutic agent. Nonlimiting examples of a chemotherapeutic agent include cisplatin, carboplatin, oxaliplatin, cyclophosphamide, altretamine, plicamydin, chlorambucil, chlormethine, ifosfamide, melphalan, carmustine, fotemustine, lomustine, streptozocin, busulfan, dacarbazine, mechlorethamine, procarbazine, temozolomide, thioTEPA, uramustine, paclitaxel, docataxel, vinblastine, vincristine, vindesine, vinorelbine, hexamethylmelamine, etoposide, teniposide, methotrexate, pemetrexed, raltitrexed, cladribine, clofarabine, fludarabine, mercaptopurine, tioguanine, capecitabine, cytarabine, fluorouracil, fluxuridine, gemcitabine, daunorubicin, doxorubicin, epirubicin, idarubicin, mitoxantrone, valrubicin, bleomycin, hydroxyurea, mitomycin, topotecan, irinotecan, aminolevulinic acid, methyl aminolevulinate, porfimer sodium, verteporfm, alitretinoin, altretamine, amsacrine, anagrelide, arsenic trioxide, asparaginase, bexarotene, bortezomib, celecoxib, denileukin, diftitox, erlotinib, estramustine, gefitinib, hydroxycarbamide, imatinib, pentostatin, masoprocol, mitotane, pegaspargase, sorafenib, sunitinib, and tretinoin, or a pharmaceutically-acceptable salt of any of the foregoing.
[0073] In some embodiments, an imaging agent can comprise radiolabels, radionuclides, radioisotopes, fluorophores, fluorochromes, dyes, metal lanthanides, paramagnetic metal ions, superparamagnetic metal oxides, ultrasound reporters, x-ray reporters, or fluorescent proteins. [0074] Non-limiting examples of radiolabels include 99mTc, 111ln, 64Cu, 67Ga, 186Re, 188Re, 153Sm, 177LU, 67CU, 123I, 124I, 125I, 11C, 13N, 150, 18F, 186Re, 188Re, 153Sm, 166Ho, 177Lu, 149Pm, 90Y, 212Bi, 103Pd, 109Pd, 159Gd, 140La, 198Au, 199Au, 169Yb, 175Yb, 165Dy, 166Dy, 67Cu, 105Rh, luAg, 89Zr, and 192Ir. Non-limiting examples of paramagnetic metal ions include Gd(III), Dy(III), Fe(III), and Mn(II). In some embodiments, Gadolinium (III) contrast agents comprise Dotarem, Gadavist, Magnevist, Omniscan, OptiMARK, and Prohance. Non-limiting examples of x-ray reporters include iodinated organic molecules and chelates of heavy metal ions of atomic numbers 57 to 83. Non-limiting examples of fluorophores (e.g., fluorochromes) include Cy5.5, Cy5 and Cy7 (GE Healthcare); AlexaFlour660, AlexaFlour680, AlexaFluor750, and AlexaFluor790
(Invitrogen); VivoTag680, VivoTag-S680, and VivoTag-S750 (VisEn Medical); Dy677, Dy682,
Dy752 and Dy780 (Dyomics); DyLight547, DyLight647 (Pierce); HiLyte Fluor 647, HiLyte Fluor 680, and HiLyte Fluor 750 (AnaSpec); IRDye 800CW, IRDye 800RS, and IRDye 700DX (Li-Cor); and ADS780WS, ADS830WS, and ADS832WS (American Dye Source) and Kodak X- SIGHT 650, Kodak X-SIGHT 691, and Kodak X-SIGHT 751 (Carestream Health).
[0075] The composition of the disclosure can comprise (i) the population of nanoparticles as disclosed herein and (ii) a dehydrating agent. A dehydrating agent can comprise a sugar (e.g., monosaccharide and/or a polysaccharide) and/or a sugar alcohol. Non-limiting examples of such a sugar include mannose, sucrose, dextrose, trehalose, lactose, and raffmose. For example, the dehydrating agent is sucrose. Non-limiting examples of such sugar alcohol include ethylene glycol, glycerol, erythritol, threitol, arabitol, xylitol, ribitol, mannitol, sorbitol, galactitol, fucitol, iditol, inositol, volemitol, isomalt, maltitol, lactitol, maltotriitol, maltotetraitol, and polyglycitol. For example, the dehydrating agent is mannitol. The population of nanoparticles (NP) and the dehydrating agent (DE) can be present in the composition in a mass-to-mass ratio of about 1 : 10,000 to about 10,000: 1 (NP:DE). The mass-mass ratio of NP:DE can be at least or up to about 1 : 10,000, at least or up to about 5,000: 1, at least or up to about 1,000: 1, at least or up to about 500: 1, at least or up to about 100: 1, at least or up to about 50:1, at least or up to about 10:1, at least or up to about 5: 1, at least or up to about 1 : 1, at least or up to about 5: 1, at least or up to about 10: 1, at least or up to about 50:1, at least or up to about 100: 1, at least or up to about 500: 1, at least or up to about 1,000: 1, at least or up to about 5,000: 1, or at least or up to about 10,000: 1. [0076] In some embodiments, the dehydrating agent (e.g., sucrose) can be present in an average quantity of at least or up to about 0.1 ng, at least or up to about 0.5 ng, at least or up to about 1 ng, at least or up to about 5 ng, at least or up to about 10 ng, at least or up to about 50 ng, at least or up to about 100 ng, at least or up to about 500 ng, at least or up to about 1 μg, at least or up to about 5 μg, at least or up to about 10 μg, at least or up to about 50 μg, at least or up to about 100 μg, at least or up to about 500 μg, at least or up to about 1 mg, at least or up to about 5 mg, at least or up to about 10 mg, at least or up to about 50 mg, at least or up to about 100 mg, at least or up to about 500 mg, or at least or up to about 1,000 mg per composition. In some embodiments, the dehydrating agent (e.g., sucrose) can be present in an average quantity of at least or up to about 0.1 %, at least or up to about 0.2 %, at least or up to about 0.3 %, at least or up to about 0.4 %, at least or up to about 0.5 %, at least or up to about 0.6 %, at least or up to about 0.7 %, at least or up to about 0.8 %, at least or up to about 0.9 %, at least or up to about 1 %, at least or up to about 2 %, at least or up to about 3 %, at least or up to about 4 %, at least or up to about 5 %, at least or up to about 6 %, at least or up to about 7 %, at least or up to about 8 %, at least or up to about 9 %, at least or up to about 10 %, at least or up to about 11 %, at least
or up to about 12 %, at least or up to about 13 %, at least or up to about 14 %, at least or up to about 15 %, at least or up to about 16 %, at least or up to about 17 %, at least or up to about 18 %, at least or up to about 19 %, at least or up to about 20 %, at least or up to about 25 %, at least or up to about 30 %, at least or up to about 35 %, at least or up to about 40 %, at least or up to about 45 %, or at least or up to about 50 % by weight of the composition.
[0077] In some embodiments, the nanoparticles of the present invention (e.g., PLGA-PEG nanoparticles) are suspended in a solution comprising the dehydrating agent (e.g., sucrose), and the resulting mixture is lyophilized and stored prior to use. In some embodiments, prior to lyophilization, the nanoparticles are suspended in a solution comprising the dehydrating agent in an average quantity of at least or up to about 0.1 %, at least or up to about 0.2 %, at least or up to about 0.3 %, at least or up to about 0.4 %, at least or up to about 0.5 %, at least or up to about 0.6 %, at least or up to about 0.7 %, at least or up to about 0.8 %, at least or up to about 0.9 %, at least or up to about 1 %, at least or up to about 2 %, at least or up to about 3 %, at least or up to about 4 %, at least or up to about 5 %, at least or up to about 6 %, at least or up to about 7 %, at least or up to about 8 %, at least or up to about 9 %, at least or up to about 10 %, at least or up to about 11 %, at least or up to about 12 %, at least or up to about 13 %, at least or up to about 14 %, at least or up to about 15 %, at least or up to about 16 %, at least or up to about 17 %, at least or up to about 18 %, at least or up to about 19 %, at least or up to about 20 %, at least or up to about 25 %, at least or up to about 30 %, at least or up to about 35 %, at least or up to about 40 %, at least or up to about 45 %, or at least or up to about 50 % by weight per volume (e.g., 1 milliliter (mL)) of the solution. In some embodiments, prior to lyophilization, the nanoparticles are suspended in a solution comprising 5 wt.% sucrose or trehalose (50 mg of the dehydrating agent per 1 mL of the solution).
[0078] In some embodiments, a presence of the hydration agent as disclosed herein (e.g., sucrose or trehalose) can reduce the degree of change (e.g., increase) of the average cross-sectional dimension of the population of nanoparticles upon lyophilization by at least or up to about 0.1 %, at least or up to about 0.2 %, at least or up to about 0.3 %, at least or up to about 0.4 %, at least or up to about 0.5 %, at least or up to about 0.6 %, at least or up to about 0.7 %, at least or up to about 0.8 %, at least or up to about 0.9 %, at least or up to about 1 %, at least or up to about 2 %, at least or up to about 3 %, at least or up to about 4 %, at least or up to about 5 %, at least or up to about 6 %, at least or up to about 7 %, at least or up to about 8 %, at least or up to about 9 %, at least or up to about 10 %, at least or up to about 11 %, at least or up to about 12 %, at least or up to about 13 %, at least or up to about 14 %, at least or up to about 15 %, at least or up to about 16 %, at least or up to about 17 %, at least or up to about 18 %, at least or up to about 19 %, at least
or up to about 20 %, at least or up to about 25 %, at least or up to about 30 %, at least or up to about 35 %, at least or up to about 40 %, at least or up to about 45 %, at least or up to about 50 %, at least or up to about 55 %, at least or up to about 60 %, at least or up to about 65 %, at least or up to about 70 %, at least or up to about 75 %, at least or up to about 80 %, at least or up to about 85 %, at least or up to about 90 %, at least or up to about 95 %, or at least or up to about 100%, as compared to lyophilization of the population of nanoparticles in absence of the hydration agent.
Uses
[0079] In a clinical or pre-clinical study, control medical agents can be administered to a subject via a control population of particles or in absence of any population of particles (e.g., in saline). As compared to the population of particles of the disclosure, the control population of particles can have one or more different features, e.g., materials, shape, cross-sectional dimension (e.g., average diameter or area), charge (e.g., surface charge, such as zeta potential), surface chemistry (e.g., hydrophobic, hydrophilic, smooth, rough, etc.), or a degradation rate (e.g., in saline, serum, whole blood, etc.). In some embodiments, the control population of particles is a population of emulsions or micelles. Similarly, control therapeutic agents, can be administered to a subject via a control population of particles or in absence of any population of particles (e.g., in saline). [0080] A composition comprising the population of particles of the disclosure can be administered to a subject in need thereof. The composition can be administered via oral (PO), intravenous (IV), intramuscular (IM), intra-arterial, intramedullary, intrathecal, subcutaneous (SQ), intraventricular, transdermal, interdermal, intradermal, rectal (PR), vaginal, intraperitoneal (IP), intragastric (IG), topical (e.g., by powders, ointments, creams, gels, lotions, and/or drops), mucosal, intranasal, buccal, enteral, vitreal, or sublingual administrations. The composition can be administered via intratracheal instillation, bronchial instillation, inhalation, oral spray, nasal spray, aerosol, or a portal vein catheter.
[0081] The composition can be administered to the subject at least or up to 1 time per day, at least or up to 2 times per day, at least or up to 3 times per day, at least or up to 4 times per day, or at least or up to 5 times per day. The population of particles of the present invention can be administered at least or up to 1 time, at least or up to 2 times, at least or up to 3 times, at least or up to 4 times, at least or up to 5 times, at least or up to 6 times, at least or up to 7 times, at least or up to 8 times, at least or up to 9 times, at least or up to 10 times, at least or up to 15 times, at least or up to 20 times over the course of several days, weeks, months, or years.
A. Biodistribution
[0082] Upon administration to the subject (e.g., via oral or intravenous administration), the population of particles can selectively target or localize in one or more types of tissue to a greater degree than in other types of tissue. In some embodiments, the population of particles can selectively localize in one or more types of tissue selected from a brain, spinal cord, heart, kidney, lung, liver, eye, pancreas, spleen, intestine, cornea, skin, bone marrow, blood, peripheral or central nerve, and connective tissue to a greater degree than in other types of tissue. In some embodiments, the population of particles can selectively localize in the kidneys to a greater degree than one or more other types of tissue, such as the, brain, spinal cord, heart, lung, liver, eye, pancreas, spleen, intestine, cornea, skin, bone marrow, blood, peripheral or central nerve, or connective tissue. In some embodiments, the population of particles can selectively localize in renal proximal tubules and/or renal distal tubules to a greater degree than one or more other types of tissue. In some embodiments, selective targeting or localization of the population of particles to one or more types of tissue is dependent on one or more features of the population of particles, e.g., shape, cross-sectional dimension (e.g., average diameter or area), charge (e.g., surface charge, such as zeta potential), surface chemistry (e.g., hydrophobic, hydrophilic, smooth, rough, etc.), or a rate of degradability of the population of particles in the subject’s body. In some embodiments, certain nanoparticles (e.g., mesoscale nanoparticles, e.g., having a cross-sectional diameter between about 300 nm to about 700 nm) can selectively target or localize in the kidneys as compared to other types of tissue (e.g., heart, lung, spleen, liver). In some embodiments, the selective targeting or localization to particular type of tissue by the population of particles of the disclosure is not dependent on surface charge or electrostatic potential of the population of particles.
[0083] In some embodiments, the population of particles of the present invention can selectively target or localize to a specific tissue without the use of any targeting agent (e.g., an antibody configured to bind to an antigen of the specific tissue). In some embodiments, the population of particles does not comprise any such targeting agent for selective targeting or localization to a specific tissue. In some embodiments, the population of particles comprises (e.g., present on the surface of the particles) one or more target agents configured to bind a biomarker (e.g., antigen) of the specific tissue selectively.
[0084] In some embodiments, selective targeting or localization of the population of particles to one or more types of tissue can be ascertained by measuring optical signal (e.g., fluorescence) of (i) the population of particles or (ii) imaging agents that are encapsulated within or coupled to the population particles.
[0085] The population of particles of the present invention can localize in one or more types of tissue (e.g., the kidneys) to a greater degree than in other types of tissue (e.g., heart, lung, spleen, liver) by at least or up to about 0.1 -fold, at least or up to about 0.2-fold, at least or up to about 0.3-fold, at least or up to about 0.4-fold, at least or up to about 0.5-fold, at least or up to about 0.6-fold, at least or up to about 0.7-fold, at least or up to about 0.8-fold, at least or up to about 0.9-fold, at least or up to about 1-fold, at least or up to about 2-fold, at least or up to about 3-fold, at least or up to about 4-fold, at least or up to about 5-fold, at least or up to about 6-fold, at least or up to about 7-fold, at least or up to about 8-fold, at least or up to about 9-fold, at least or up to about 10-fold, at least or up to about 11 -fold, at least or up to about 12-fold, at least or up to about 13-fold, at least or up to about 14-fold, at least or up to about 15-fold, at least or up to about 16-fold, at least or up to about 17-fold, at least or up to about 18-fold, at least or up to about 19-fold, at least or up to about 20-fold, at least or up to about 25-fold, at least or up to about 30-fold, at least or up to about 35-fold, at least or up to about 40-fold, at least or up to about 45-fold, at least or up to about 50-fold, at least or up to about 100-fold, at least or up to about 200-fold, at least or up to about 300-fold, at least or up to about 400-fold, at least or up to about 500-fold, at least or up to about 600-fold, at least or up to about 700-fold, at least or up to about 800-fold, at least or up to about 900-fold, or at least or up to about 1,000-fold, as ascertained by, e.g., a mean organ fluorescence normalized by a total organ weight.
[0086] In some embodiments, the medical agents of the present invention (e.g., therapeutic agents, such as ODN, e.g., CpG-ODN) that are administered to the subject via the population of particles (e.g., encapsulated therein or coupled thereto) can selectively target or localize to one or more types of tissue of the subject, as a result of selective targeting or enhanced location of the population of particles to the one or more types of tissue of the subject. In some embodiments, selective targeting or localization of the medical agents to one or more types of tissue can be ascertained by measuring the amount of the medical agents (e.g., via high-performance liquid chromatography (HPLC) detection) in various types of tissue of the subject.
[0087] Upon administration to the subject via the population of particles of the disclosure, the medical agents (e.g., therapeutic agents, such as ODN, e.g., CpG-ODN) can localize in one or more types of tissue (e.g., the kidneys) to a greater degree than in other types of tissue (e.g., heart, lung, spleen, liver) by at least or up to about 0.1 -fold, at least or up to about 0.2-fold, at least or up to about 0.3-fold, at least or up to about 0.4-fold, at least or up to about 0.5-fold, at least or up to about 0.6-fold, at least or up to about 0.7-fold, at least or up to about 0.8-fold, at least or up to about 0.9-fold, at least or up to about 1-fold, at least or up to about 2-fold, at least or up to about 3-fold, at least or up to about 4-fold, at least or up to about 5-fold, at least or up to
about 6-fold, at least or up to about 7-fold, at least or up to about 8-fold, at least or up to about 9- fold, at least or up to about 10-fold, at least or up to about 11 -fold, at least or up to about 12-fold, at least or up to about 13-fold, at least or up to about 14-fold, at least or up to about 15-fold, at least or up to about 16-fold, at least or up to about 17-fold, at least or up to about 18-fold, at least or up to about 19-fold, at least or up to about 20-fold, at least or up to about 25-fold, at least or up to about 30-fold, at least or up to about 35-fold, at least or up to about 40-fold, at least or up to about 45-fold, at least or up to about 50-fold, at least or up to about 100-fold, at least or up to about 200-fold, at least or up to about 300-fold, at least or up to about 400-fold, at least or up to about 500-fold, at least or up to about 600-fold, at least or up to about 700-fold, at least or up to about 800-fold, at least or up to about 900-fold, or at least or up to about 1,000-fold, as ascertained by measuring the amount of the medical agents (e.g., via HPLC detection) from samples derived from each of the different types of tissues.
[0088] The medical agents (e.g., therapeutic agents, such as ODN, e.g., CpG-ODN) that are administered to the subject via the population of particles of the disclosure can localize in one or more types of tissue of the subject to a greater degree as compared to the medical agents administered without the population of particles (e.g., in saline).
[0089] Upon administration to the subject, the medical agents (e.g., therapeutic agents, such as ODN, e.g., CpG-ODN) administered via the population of particles can localize in one or more types of tissue (e.g., the kidneys) to a greater degree than the medical agents without the population of particles by at least or up to about 0.1 -fold, at least or up to about 0.2-fold, at least or up to about 0.3-fold, at least or up to about 0.4-fold, at least or up to about 0.5-fold, at least or up to about 0.6-fold, at least or up to about 0.7-fold, at least or up to about 0.8-fold, at least or up to about 0.9-fold, at least or up to about 1-fold, at least or up to about 2-fold, at least or up to about 3-fold, at least or up to about 4-fold, at least or up to about 5-fold, at least or up to about 6- fold, at least or up to about 7-fold, at least or up to about 8-fold, at least or up to about 9-fold, at least or up to about 10-fold, at least or up to about 11 -fold, at least or up to about 12-fold, at least or up to about 13-fold, at least or up to about 14-fold, at least or up to about 15-fold, at least or up to about 16-fold, at least or up to about 17-fold, at least or up to about 18-fold, at least or up to about 19-fold, at least or up to about 20-fold, at least or up to about 25-fold, at least or up to about 30-fold, at least or up to about 35-fold, at least or up to about 40-fold, at least or up to about 45-fold, at least or up to about 50-fold, at least or up to about 100-fold, at least or up to about 200-fold, at least or up to about 300-fold, at least or up to about 400-fold, at least or up to about 500-fold, at least or up to about 600-fold, at least or up to about 700-fold, at least or up to about 800-fold, at least or up to about 900-fold, or at least or up to about 1,000-fold, as
ascertained by measuring the amount of the medical agents (e.g., via HPLC detection) from samples derived from the one or more types of tissue.
B. Pharmacokinetic/ Pharmacodynamic study
[0090] Bioavailability of the medical agents of the present disclosure in an animal, such as a human, mouse, rat, dog, or monkey can be defined by an area under the curve (AUC). The AUC can be an integrated measure of systemic concentrations of the medical agents over time in units of mass-time/volume (e.g., microgram-hour/milliliter, or μg-hour/ml). Alternatively or in addition, the AUC can be an integrated measure of systemic concentrations of the medical agents over a defined, measurable length of time. The AUC over the first 24 hours following administration of the medical agents (e.g., via oral or intravenous administration) can be referred to as AUCO-24. Bioavailability of the medical agents can be defined as the peak plasma concentration of the medical agents (Cmax). Bioavailability of the medical agents can be defined as the time to reach the peak plasma concentration (Tmax). Bioavailability of the medical agents can be defined by a terminal half-life (T1/2), which is the time required to divide the plasma concentration of the drug by two after reaching pseudo-equilibrium or peak plasma concentration (Tmax). Bioavailability of the medical agents can be defined by skewness, which is a characterization of the degree of asymmetry of the plasma concentration profile around the mean plasma concentration value. Bioavailability of the medical agents can be defined by a ratio of the AUC derived from oral administration (AUCPO) to the AUC derived from intravenous administration (AUCiv). Bioavailability of the medical agents can be defined by a first pass metabolism (e.g., through the liver), which can be defined by, e.g., a ratio of (i) the difference between the AUC derived from intravenous administration (AUCiv) and the AUC derived from oral administration (AUCPO) (e.g., AUCiv - AUCPO) to (ii) AUCiv. For example, the first pass metabolism can be indicative of the relative therapeutic effect of the medical agents that are orally administered relative to the same medical agents that are intravenously administered. Any one of the pharmacokinetic parameters as disclosed herein (e.g., ACU, AUCo-24, Cmax, Tmax, T1/2, skewness, bioavailability, etc.) can be any parameters suitable for describing pharmaceutical compositions of the disclosure.
[0091] The medical agents (e.g., therapeutic agents, such as ODN, e.g., CpG-ODN) administered to the subject via the population of particles of the disclosure can exhibit a mean AUC that is greater than that of the control medical agents by at least or up to about 0.1 -fold, at least or up to about 0.2-fold, at least or up to about 0.3-fold, at least or up to about 0.4-fold, at least or up to about 0.5-fold, at least or up to about 0.6-fold, at least or up to about 0.7-fold, at least or up to
about 0.8-fold, at least or up to about 0.9-fold, at least or up to about 1-fold, at least or up to about 2-fold, at least or up to about 3-fold, at least or up to about 4-fold, at least or up to about 5- fold, at least or up to about 6-fold, at least or up to about 7-fold, at least or up to about 8-fold, at least or up to about 9-fold, at least or up to about 10-fold, at least or up to about 11 -fold, at least or up to about 12-fold, at least or up to about 13 -fold, at least or up to about 14-fold, at least or up to about 15-fold, at least or up to about 16-fold, at least or up to about 17-fold, at least or up to about 18-fold, at least or up to about 19-fold, at least or up to about 20-fold, at least or up to about 25-fold, at least or up to about 30-fold, at least or up to about 35-fold, at least or up to about 40-fold, at least or up to about 45-fold, at least or up to about 50-fold, at least or up to about 100-fold, at least or up to about 200-fold, at least or up to about 300-fold, at least or up to about 400-fold, at least or up to about 500-fold, at least or up to about 600-fold, at least or up to about 700-fold, at least or up to about 800-fold, at least or up to about 900-fold, or at least or up to about 1,000-fold.
[0092] The medical agents (e.g., therapeutic agents, such as ODN, e.g., CpG-ODN) administered to the subject via the population of particles of the present invention can exhibit a mean AUCo-24 that is greater than that of the control medical agents by at least or up to about 0.1 -fold, at least or up to about 0.2-fold, at least or up to about 0.3-fold, at least or up to about 0.4-fold, at least or up to about 0.5-fold, at least or up to about 0.6-fold, at least or up to about 0.7-fold, at least or up to about 0.8-fold, at least or up to about 0.9-fold, at least or up to about 1-fold, at least or up to about 2-fold, at least or up to about 3-fold, at least or up to about 4-fold, at least or up to about 5- fold, at least or up to about 6-fold, at least or up to about 7-fold, at least or up to about 8-fold, at least or up to about 9-fold, at least or up to about 10-fold, at least or up to about 11 -fold, at least or up to about 12-fold, at least or up to about 13 -fold, at least or up to about 14-fold, at least or up to about 15-fold, at least or up to about 16-fold, at least or up to about 17-fold, at least or up to about 18-fold, at least or up to about 19-fold, at least or up to about 20-fold, at least or up to about 25-fold, at least or up to about 30-fold, at least or up to about 35-fold, at least or up to about 40-fold, at least or up to about 45-fold, at least or up to about 50-fold, at least or up to about 100-fold, at least or up to about 200-fold, at least or up to about 300-fold, at least or up to about 400-fold, at least or up to about 500-fold, at least or up to about 600-fold, at least or up to about 700-fold, at least or up to about 800-fold, at least or up to about 900-fold, or at least or up to about 1,000-fold.
[0093] The medical agents (e.g., therapeutic agents, such as ODN, e.g., CpG-ODN) administered to the subject via the population of particles of the present invention can exhibit a mean Cmax that is greater than that of the control medical agents by at least or up to about 0.1 -fold, at least or up
to about 0.2-fold, at least or up to about 0.3-fold, at least or up to about 0.4-fold, at least or up to about 0.5-fold, at least or up to about 0.6-fold, at least or up to about 0.7-fold, at least or up to about 0.8-fold, at least or up to about 0.9-fold, at least or up to about 1-fold, at least or up to about 2-fold, at least or up to about 3-fold, at least or up to about 4-fold, at least or up to about 5- fold, at least or up to about 6-fold, at least or up to about 7-fold, at least or up to about 8-fold, at least or up to about 9-fold, at least or up to about 10-fold, at least or up to about 11 -fold, at least or up to about 12-fold, at least or up to about 13 -fold, at least or up to about 14-fold, at least or up to about 15-fold, at least or up to about 16-fold, at least or up to about 17-fold, at least or up to about 18-fold, at least or up to about 19-fold, at least or up to about 20-fold, at least or up to about 25-fold, at least or up to about 30-fold, at least or up to about 35-fold, at least or up to about 40-fold, at least or up to about 45-fold, at least or up to about 50-fold, at least or up to about 100-fold, at least or up to about 200-fold, at least or up to about 300-fold, at least or up to about 400-fold, at least or up to about 500-fold, at least or up to about 600-fold, at least or up to about 700-fold, at least or up to about 800-fold, at least or up to about 900-fold, or at least or up to about 1,000-fold.
[0094] The medical agents (e.g., therapeutic agents, such as ODN, e.g., CpG-ODN) administered to the subject via the population of particles of the disclosure can exhibit a mean Cmax that is less than that of the control medical agents by at least or up to about 0.1 -fold, at least or up to about 0.2-fold, at least or up to about 0.3-fold, at least or up to about 0.4-fold, at least or up to about 0.5-fold, at least or up to about 0.6-fold, at least or up to about 0.7-fold, at least or up to about 0.8-fold, at least or up to about 0.9-fold, at least or up to about 1-fold, at least or up to about 2- fold, at least or up to about 3-fold, at least or up to about 4-fold, at least or up to about 5-fold, at least or up to about 6-fold, at least or up to about 7-fold, at least or up to about 8-fold, at least or up to about 9-fold, at least or up to about 10-fold, at least or up to about 11 -fold, at least or up to about 12-fold, at least or up to about 13 -fold, at least or up to about 14-fold, at least or up to about 15-fold, at least or up to about 16-fold, at least or up to about 17-fold, at least or up to about 18-fold, at least or up to about 19-fold, at least or up to about 20-fold, at least or up to about 25-fold, at least or up to about 30-fold, at least or up to about 35-fold, at least or up to about 40-fold, at least or up to about 45-fold, at least or up to about 50-fold, at least or up to about 100-fold, at least or up to about 200-fold, at least or up to about 300-fold, at least or up to about 400-fold, at least or up to about 500-fold, at least or up to about 600-fold, at least or up to about 700-fold, at least or up to about 800-fold, at least or up to about 900-fold, or at least or up to about 1,000-fold.
[0095] The medical agents (e.g., therapeutic agents, such as ODN, e.g., CpG-ODN) administered
to the subject via the population of particles of the disclosure can exhibit a mean Tmax that is greater than that of the control medical agents by at least or up to about 0.1 -fold, at least or up to about 0.2-fold, at least or up to about 0.3-fold, at least or up to about 0.4-fold, at least or up to about 0.5-fold, at least or up to about 0.6-fold, at least or up to about 0.7-fold, at least or up to about 0.8-fold, at least or up to about 0.9-fold, at least or up to about 1-fold, at least or up to about 2-fold, at least or up to about 3-fold, at least or up to about 4-fold, at least or up to about 5- fold, at least or up to about 6-fold, at least or up to about 7-fold, at least or up to about 8-fold, at least or up to about 9-fold, at least or up to about 10-fold, at least or up to about 11 -fold, at least or up to about 12-fold, at least or up to about 13 -fold, at least or up to about 14-fold, at least or up to about 15-fold, at least or up to about 16-fold, at least or up to about 17-fold, at least or up to about 18-fold, at least or up to about 19-fold, at least or up to about 20-fold, at least or up to about 25-fold, at least or up to about 30-fold, at least or up to about 35-fold, at least or up to about 40-fold, at least or up to about 45-fold, at least or up to about 50-fold, at least or up to about 100-fold, at least or up to about 200-fold, at least or up to about 300-fold, at least or up to about 400-fold, at least or up to about 500-fold, at least or up to about 600-fold, at least or up to about 700-fold, at least or up to about 800-fold, at least or up to about 900-fold, or at least or up to about 1,000-fold.
[0096] The medical agents (e.g., therapeutic agents, such as ODN, e.g., CpG-ODN) administered to the subject via the population of particles of the disclosure can exhibit a mean Tmax that is less than that of the control medical agents by at least or up to about 0.1 -fold, at least or up to about 0.2-fold, at least or up to about 0.3-fold, at least or up to about 0.4-fold, at least or up to about 0.5-fold, at least or up to about 0.6-fold, at least or up to about 0.7-fold, at least or up to about 0.8-fold, at least or up to about 0.9-fold, at least or up to about 1-fold, at least or up to about 2- fold, at least or up to about 3-fold, at least or up to about 4-fold, at least or up to about 5-fold, at least or up to about 6-fold, at least or up to about 7-fold, at least or up to about 8-fold, at least or up to about 9-fold, at least or up to about 10-fold, at least or up to about 11 -fold, at least or up to about 12-fold, at least or up to about 13 -fold, at least or up to about 14-fold, at least or up to about 15-fold, at least or up to about 16-fold, at least or up to about 17-fold, at least or up to about 18-fold, at least or up to about 19-fold, at least or up to about 20-fold, at least or up to about 25-fold, at least or up to about 30-fold, at least or up to about 35-fold, at least or up to about 40-fold, at least or up to about 45-fold, at least or up to about 50-fold, at least or up to about 100-fold, at least or up to about 200-fold, at least or up to about 300-fold, at least or up to about 400-fold, at least or up to about 500-fold, at least or up to about 600-fold, at least or up to about 700-fold, at least or up to about 800-fold, at least or up to about 900-fold, or at least or up
to about 1,000-fold.
[0097] The medical agents (e.g., therapeutic agents, such as ODN, e.g., CpG-ODN) administered to the subject via the population of particles of the disclosure can exhibit a mean T1/2 that is greater than that of the control medical agents by at least or up to about 0.1 -fold, at least or up to about 0.2-fold, at least or up to about 0.3-fold, at least or up to about 0.4-fold, at least or up to about 0.5-fold, at least or up to about 0.6-fold, at least or up to about 0.7-fold, at least or up to about 0.8-fold, at least or up to about 0.9-fold, at least or up to about 1-fold, at least or up to about 2-fold, at least or up to about 3-fold, at least or up to about 4-fold, at least or up to about 5- fold, at least or up to about 6-fold, at least or up to about 7-fold, at least or up to about 8-fold, at least or up to about 9-fold, at least or up to about 10-fold, at least or up to about 11 -fold, at least or up to about 12-fold, at least or up to about 13 -fold, at least or up to about 14-fold, at least or up to about 15-fold, at least or up to about 16-fold, at least or up to about 17-fold, at least or up to about 18-fold, at least or up to about 19-fold, at least or up to about 20-fold, at least or up to about 25-fold, at least or up to about 30-fold, at least or up to about 35-fold, at least or up to about 40-fold, at least or up to about 45-fold, at least or up to about 50-fold, at least or up to about 100-fold, at least or up to about 200-fold, at least or up to about 300-fold, at least or up to about 400-fold, at least or up to about 500-fold, at least or up to about 600-fold, at least or up to about 700-fold, at least or up to about 800-fold, at least or up to about 900-fold, or at least or up to about 1,000-fold.
[0098] The medical agents (e.g., therapeutic agents, such as ODN, e.g., CpG-ODN) administered to the subject via the population of particles of the disclosure can exhibit a skewness that is greater than that of the control medical agents by at least or up to about 0.1 -fold, at least or up to about 0.2-fold, at least or up to about 0.3-fold, at least or up to about 0.4-fold, at least or up to about 0.5-fold, at least or up to about 0.6-fold, at least or up to about 0.7-fold, at least or up to about 0.8-fold, at least or up to about 0.9-fold, at least or up to about 1-fold, at least or up to about 2-fold, at least or up to about 3-fold, at least or up to about 4-fold, at least or up to about 5- fold, at least or up to about 6-fold, at least or up to about 7-fold, at least or up to about 8-fold, at least or up to about 9-fold, at least or up to about 10-fold, at least or up to about 11 -fold, at least or up to about 12-fold, at least or up to about 13 -fold, at least or up to about 14-fold, at least or up to about 15-fold, at least or up to about 16-fold, at least or up to about 17-fold, at least or up to about 18-fold, at least or up to about 19-fold, at least or up to about 20-fold, at least or up to about 25-fold, at least or up to about 30-fold, at least or up to about 35-fold, at least or up to about 40-fold, at least or up to about 45-fold, or at least or up to about 50-fold.
[0099] The medical agents (e.g., therapeutic agents, such as ODN, e.g., CpG-ODN) administered
to the subject via the population of particles of the disclosure can exhibit a skewness that is less than that of the control medical agents by at least or up to about 0.1 -fold, at least or up to about 0.2-fold, at least or up to about 0.3-fold, at least or up to about 0.4-fold, at least or up to about
0.5-fold, at least or up to about 0.6-fold, at least or up to about 0.7-fold, at least or up to about
0.8-fold, at least or up to about 0.9-fold, at least or up to about 1-fold, at least or up to about 2- fold, at least or up to about 3-fold, at least or up to about 4-fold, at least or up to about 5-fold, at least or up to about 6-fold, at least or up to about 7-fold, at least or up to about 8-fold, at least or up to about 9-fold, at least or up to about 10-fold, at least or up to about 11 -fold, at least or up to about 12-fold, at least or up to about 13 -fold, at least or up to about 14-fold, at least or up to about 15-fold, at least or up to about 16-fold, at least or up to about 17-fold, at least or up to about 18-fold, at least or up to about 19-fold, at least or up to about 20-fold, at least or up to about 25-fold, at least or up to about 30-fold, at least or up to about 35-fold, at least or up to about 40-fold, at least or up to about 45-fold, or at least or up to about 50-fold.
[0100] The medical agents (e.g., therapeutic agents, such as ODN, e.g., CpG-ODN) administered to the subject via the population of particles of the disclosure can exhibit a mean bioavailability ratio (AUCPO: AUCiv) that is greater than that of the control medical agents by at least or up to about 0.1-fold, at least or up to about 0.2-fold, at least or up to about 0.3-fold, at least or up to about 0.4-fold, at least or up to about 0.5-fold, at least or up to about 0.6-fold, at least or up to about 0.7-fold, at least or up to about 0.8-fold, at least or up to about 0.9-fold, at least or up to about 1-fold, at least or up to about 2-fold, at least or up to about 3 -fold, at least or up to about 4- fold, at least or up to about 5-fold, at least or up to about 6-fold, at least or up to about 7-fold, at least or up to about 8-fold, at least or up to about 9-fold, at least or up to about 10-fold, at least or up to about 11 -fold, at least or up to about 12-fold, at least or up to about 13 -fold, at least or up to about 14-fold, at least or up to about 15-fold, at least or up to about 16-fold, at least or up to about 17-fold, at least or up to about 18-fold, at least or up to about 19-fold, at least or up to about 20-fold, at least or up to about 25-fold, at least or up to about 30-fold, at least or up to about 35-fold, at least or up to about 40-fold, at least or up to about 45-fold, at least or up to about 50-fold, at least or up to about 100-fold, at least or up to about 200-fold, at least or up to about 300-fold, at least or up to about 400-fold, at least or up to about 500-fold, at least or up to about 600-fold, at least or up to about 700-fold, at least or up to about 800-fold, at least or up to about 900-fold, or at least or up to about 1,000-fold.
[0101] The medical agents (e.g., therapeutic agents, such as ODN, e.g., CpG-ODN) administered to the subject via the population of particles of the disclosure can exhibit a mean first pass metabolism that is less than that of the control medical agents by at least or up to about 0.1 -fold,
at least or up to about 0.2-fold, at least or up to about 0.3-fold, at least or up to about 0.4-fold, at least or up to about 0.5-fold, at least or up to about 0.6-fold, at least or up to about 0.7-fold, at least or up to about 0.8-fold, at least or up to about 0.9-fold, at least or up to about 1-fold, at least or up to about 2-fold, at least or up to about 3 -fold, at least or up to about 4-fold, at least or up to about 5-fold, at least or up to about 6-fold, at least or up to about 7-fold, at least or up to about 8- fold, at least or up to about 9-fold, at least or up to about 10-fold, at least or up to about 11 -fold, at least or up to about 12-fold, at least or up to about 13 -fold, at least or up to about 14-fold, at least or up to about 15-fold, at least or up to about 16-fold, at least or up to about 17-fold, at least or up to about 18-fold, at least or up to about 19-fold, at least or up to about 20-fold, at least or up to about 25-fold, at least or up to about 30-fold, at least or up to about 35-fold, at least or up to about 40-fold, at least or up to about 45-fold, at least or up to about 50-fold, at least or up to about 100-fold, at least or up to about 200-fold, at least or up to about 300-fold, at least or up to about 400-fold, at least or up to about 500-fold, at least or up to about 600-fold, at least or up to about 700-fold, at least or up to about 800-fold, at least or up to about 900-fold, or at least or up to about 1,000-fold.
C. Additional features
[0102] A degradation rate of the medical agents (e.g., therapeutic agents, such as ODN, e.g., CpG-ODN) that are encapsulated within or coupled to the population of particles of the disclosure can exhibit a different degradation rate as compared to the control medical agents.
The degradation rate can be measured at an ambient temperature of at least or up to about 3 °C, at least or up to about 5 °C, at least or up to about 10 °C, at least or up to about 15 °C, at least or up to about 20 °C, at least or up to about 25 °C, at least or up to about 30 °C, at least or up to about 35 °C, at least or up to about 37 °C, or at least or up to about 40 °C.
[0103] The degradation rate at an ambient temperature (e.g., at least about 25 °C) of the medical agents (e.g., therapeutic agents, such as ODN, e.g., CpG ODN) that are encapsulated within or coupled to the population of particles can be lower than that of the control medical agents by at least or up to about 0.1-fold, at least or up to about 0.2-fold, at least or up to about 0.3-fold, at least or up to about 0.4-fold, at least or up to about 0.5-fold, at least or up to about 0.6-fold, at least or up to about 0.7-fold, at least or up to about 0.8-fold, at least or up to about 0.9-fold, at least or up to about 1-fold, at least or up to about 2-fold, at least or up to about 3 -fold, at least or up to about 4-fold, at least or up to about 5-fold, at least or up to about 6-fold, at least or up to about 7-fold, at least or up to about 8-fold, at least or up to about 9-fold, at least or up to about 10-fold, at least or up to about 11 -fold, at least or up to about 12-fold, at least or up to about 13-
fold, at least or up to about 14-fold, at least or up to about 15-fold, at least or up to about 16-fold, at least or up to about 17-fold, at least or up to about 18-fold, at least or up to about 19-fold, at least or up to about 20-fold, at least or up to about 25-fold, at least or up to about 30-fold, at least or up to about 35-fold, at least or up to about 40-fold, at least or up to about 45-fold, at least or up to about 50-fold, at least or up to about 100-fold, at least or up to about 200-fold, at least or up to about 300-fold, at least or up to about 400-fold, at least or up to about 500-fold, at least or up to about 600-fold, at least or up to about 700-fold, at least or up to about 800-fold, at least or up to about 900-fold, or at least or up to about 1,000-fold.
[0104] The population of particles of the disclosure can exhibit a different release rate of the medical agents (e.g., therapeutic agents, such as ODN, e.g., CpG ODN) as compared to that from a control population of particles.
[0105] The release rate of medical agents (e.g., therapeutic agents, such as ODN, e.g., CpG- ODN) from the population of particles of the present invention can be lower than that from the control population of particles by at least or up to about 0.1 -fold, at least or up to about 0.2-fold, at least or up to about 0.3-fold, at least or up to about 0.4-fold, at least or up to about 0.5-fold, at least or up to about 0.6-fold, at least or up to about 0.7-fold, at least or up to about 0.8-fold, at least or up to about 0.9-fold, at least or up to about 1-fold, at least or up to about 2-fold, at least or up to about 3-fold, at least or up to about 4-fold, at least or up to about 5-fold, at least or up to about 6-fold, at least or up to about 7-fold, at least or up to about 8-fold, at least or up to about 9- fold, at least or up to about 10-fold, at least or up to about 11 -fold, at least or up to about 12-fold, at least or up to about 13-fold, at least or up to about 14-fold, at least or up to about 15-fold, at least or up to about 16-fold, at least or up to about 17-fold, at least or up to about 18-fold, at least or up to about 19-fold, at least or up to about 20-fold, at least or up to about 25-fold, at least or up to about 30-fold, at least or up to about 35-fold, at least or up to about 40-fold, at least or up to about 45-fold, at least or up to about 50-fold, at least or up to about 100-fold, at least or up to about 200-fold, at least or up to about 300-fold, at least or up to about 400-fold, at least or up to about 500-fold, at least or up to about 600-fold, at least or up to about 700-fold, at least or up to about 800-fold, at least or up to about 900-fold, or at least or up to about 1,000-fold.
[0106] The release rate of medical agents (e.g., therapeutic agents, such as ODN, e.g., CpG- ODN) from the population of particles of the present invention can be higher than that from the control population of particles by at least or up to about 0.1 -fold, at least or up to about 0.2-fold, at least or up to about 0.3-fold, at least or up to about 0.4-fold, at least or up to about 0.5-fold, at least or up to about 0.6-fold, at least or up to about 0.7-fold, at least or up to about 0.8-fold, at least or up to about 0.9-fold, at least or up to about 1-fold, at least or up to about 2-fold, at least
or up to about 3-fold, at least or up to about 4-fold, at least or up to about 5-fold, at least or up to about 6-fold, at least or up to about 7-fold, at least or up to about 8-fold, at least or up to about 9- fold, at least or up to about 10-fold, at least or up to about 11 -fold, at least or up to about 12-fold, at least or up to about 13-fold, at least or up to about 14-fold, at least or up to about 15-fold, at least or up to about 16-fold, at least or up to about 17-fold, at least or up to about 18-fold, at least or up to about 19-fold, at least or up to about 20-fold, at least or up to about 25-fold, at least or up to about 30-fold, at least or up to about 35-fold, at least or up to about 40-fold, at least or up to about 45-fold, at least or up to about 50-fold, at least or up to about 100-fold, at least or up to about 200-fold, at least or up to about 300-fold, at least or up to about 400-fold, at least or up to about 500-fold, at least or up to about 600-fold, at least or up to about 700-fold, at least or up to about 800-fold, at least or up to about 900-fold, or at least or up to about 1,000-fold.
[0107] Upon localization into a specific type of tissue (e.g., the kidneys), the population of particles of the disclosure can release the medical agents (e.g., therapeutic agents, such as ODN, e.g., CpG-ODN) from the particles to the surrounding for at least or up to about 1 minute, at least or up to about 2 minutes, at least or up to about 3 minutes, at least or up to about 4 minutes, at least or up to about 5 minutes, at least or up to about 6 minutes, at least or up to about 7 minutes, at least or up to about 8 minutes, at least or up to about 9 minutes, at least or up to about 10 minutes, at least or up to about 15 minutes, at least or up to about 20 minutes, at least or up to about 25 minutes, at least or up to about 30 minutes, at least or up to about 40 minutes, at least or up to about 50 minutes, at least or up to about 60 minutes, at least or up to about 2 hours, at least or up to about 3 hours, at least or up to about 4 hours, at least or up to about 5 hours, at least or up to about 6 hours, at least or up to about 7 hours, at least or up to about 8 hours, at least or up to about 9 hours, at least or up to about 10 hours, at least or up to about 11 hours, at least or up to about 12 hours, at least or up to about 16 hours, at least or up to about 20 hours, at least or up to about 24 hours, at least or up to about 2 days, at least or up to about 3 days, at least or up to about 4 days, at least or up to about 5 days, at least or up to about 6 days, at least or up to about 7 days, at least or up to about 2 weeks, at least or up to about 3 weeks, at least or up to about 4 weeks, at least or up to about 2 months, at least or up to about 3 months, at least or up to about 4 months, at least or up to about 5 months, at least or up to about 6 months, at least or up to about 7 months, at least or up to about 8 months, at least or up to about 9 months, at least or up to about 10 months, at least or up to about 11 months, or at least or up to about 12 months.
D. Therapeutic applications
[0108] In some embodiments, upon administration to a subject in need thereof, the population of
particles comprising the therapeutic agents of the disclosure can protect the subject from reperfusion injury, such as ischemia reperfusion injury (e.g., renal ischemia reperfusion (AKI) injury), as compared to a control (e.g., no treatment, or non-encapsulated therapeutic agents, inactive encapsulated therapeutic agents). In some embodiments, upon administration to a subject in need thereof, the population of particles comprising the therapeutic agents can attenuate or prevent renal tubular necrosis (e.g., decreased DNA fragmentation and/or decreased caspase 3/8 activation) and/or inflammation (e.g., decreased pro-inflammatory cytokine expression), as compared to a control. In some embodiments, upon administration to a subject in need thereof, the population of particles comprising the therapeutic agents can reduce immune cell infiltration to a target tissue, such as neutrophil and/or macrophase infiltration into at least a portion of a kidney.
[0109] In some embodiments, administration of the therapeutic agents (e.g., a nucleic acid molecule, such as ODN, e.g., CpG-ODN) via the population of particles of the disclosure can reduce presence or expression level of one or more kidney injury markers (e.g., plasma creatinine (PCr), blood urea nitrogen (BUN), neutrophil gelatinase-associated lipocalin (NGAL), etc.) than a control. Treatment with the therapeutic agents via the population of particles of the disclosure can reduce the presence or expression level of the one or more kidney injury markers as compared to a control (e.g., no treatment, non-encapsulated therapeutic agents, inactive encapsulated therapeutic agents) by at least or up to about 0.1 -fold, at least or up to about 0.2- fold, at least or up to about 0.3-fold, at least or up to about 0.4-fold, at least or up to about 0.5- fold, at least or up to about 0.6-fold, at least or up to about 0.7-fold, at least or up to about 0.8- fold, at least or up to about 0.9-fold, at least or up to about 1-fold, at least or up to about 2-fold, at least or up to about 3-fold, at least or up to about 4-fold, at least or up to about 5-fold, at least or up to about 6-fold, at least or up to about 7-fold, at least or up to about 8-fold, at least or up to about 9-fold, at least or up to about 10-fold, at least or up to about 11 -fold, at least or up to about 12-fold, at least or up to about 13-fold, at least or up to about 14-fold, at least or up to about 15- fold, at least or up to about 16-fold, at least or up to about 17-fold, at least or up to about 18-fold, at least or up to about 19-fold, at least or up to about 20-fold, at least or up to about 25-fold, at least or up to about 30-fold, at least or up to about 35-fold, at least or up to about 40-fold, at least or up to about 45-fold, at least or up to about 50-fold, at least or up to about 100-fold, at least or up to about 200-fold, at least or up to about 300-fold, at least or up to about 400-fold, at least or up to about 500-fold, at least or up to about 600-fold, at least or up to about 700-fold, at least or up to about 800-fold, at least or up to about 900-fold, or at least or up to about 1,000-fold, as ascertained by Western blot analysis, gene expression assay (e.g., polymerase chain reaction
(PCR)), colorimetric/fluorometric assay, etc.
[0110] In some embodiments, upon administration of the therapeutic agents (e.g., a nucleic acid molecule, such as ODN, e.g., CpG-ODN) via the population of particles of the disclosure to a subject in need thereof, a tissue sample (e.g., a biopsy sample) of a kidney of the subject is evaluated (e.g., via histological assessment) and scored by the Jablonski scoring system, which, for example, ranges from 0 (no damage) to 4 (necrosis affecting all 3 segments of the proximal convoluted tubule). Treatment with the therapeutic agents via the population of particles of the disclosure can reduce the Jablonski score as compared to a control (e.g., no treatment, non- encapsulated therapeutic agents, inactive encapsulated therapeutic agents) by at least or up to about 0.1-fold, at least or up to about 0.2-fold, at least or up to about 0.3-fold, at least or up to about 0.4-fold, at least or up to about 0.5-fold, at least or up to about 0.6-fold, at least or up to about 0.7-fold, at least or up to about 0.8-fold, at least or up to about 0.9-fold, at least or up to about 1-fold, at least or up to about 2-fold, at least or up to about 3 -fold, at least or up to about 4- fold, at least or up to about 5-fold, at least or up to about 6-fold, at least or up to about 7-fold, at least or up to about 8-fold, at least or up to about 9-fold, at least or up to about 10-fold, at least or up to about 11 -fold, at least or up to about 12-fold, at least or up to about 13 -fold, at least or up to about 14-fold, at least or up to about 15-fold, at least or up to about 16-fold, at least or up to about 17-fold, at least or up to about 18-fold, at least or up to about 19-fold, at least or up to about 20-fold, at least or up to about 25-fold, at least or up to about 30-fold, at least or up to about 35-fold, at least or up to about 40-fold, at least or up to about 45-fold, at least or up to about 50-fold, at least or up to about 100-fold, at least or up to about 200-fold, at least or up to about 300-fold, at least or up to about 400-fold, at least or up to about 500-fold, at least or up to about 600-fold, at least or up to about 700-fold, at least or up to about 800-fold, at least or up to about 900-fold, or at least or up to about 1,000-fold, as ascertained by histological staining and assessment.
[0111] In some embodiments, upon administration of the therapeutic agents (e.g., a nucleic acid molecule, such as ODN, e.g., CpG-ODN) via the population of particles of the disclosure to a subject in need thereof, a tissue sample (e.g., a biopsy sample) of a kidney of the subject is evaluated (e.g., via histological assessment) for renal tubular apoptosis. Treatment with the therapeutic agents via the population of particles of the disclosure can reduce a number of terminal deoxynucleotidyl transferase deoxyuridine triphosphate (dUTP) nick end labeling (TUNEL) positive cells in the tissue sample as compared to a control (e.g., no treatment, non- encapsulated therapeutic agents, inactive encapsulated therapeutic agents) by at least or up to about 0.1-fold, at least or up to about 0.2-fold, at least or up to about 0.3-fold, at least or up to
about 0.4-fold, at least or up to about 0.5-fold, at least or up to about 0.6-fold, at least or up to about 0.7-fold, at least or up to about 0.8-fold, at least or up to about 0.9-fold, at least or up to about 1-fold, at least or up to about 2-fold, at least or up to about 3 -fold, at least or up to about 4- fold, at least or up to about 5-fold, at least or up to about 6-fold, at least or up to about 7-fold, at least or up to about 8-fold, at least or up to about 9-fold, at least or up to about 10-fold, at least or up to about 11 -fold, at least or up to about 12-fold, at least or up to about 13 -fold, at least or up to about 14-fold, at least or up to about 15-fold, at least or up to about 16-fold, at least or up to about 17-fold, at least or up to about 18-fold, at least or up to about 19-fold, at least or up to about 20-fold, at least or up to about 25-fold, at least or up to about 30-fold, at least or up to about 35-fold, at least or up to about 40-fold, at least or up to about 45-fold, at least or up to about 50-fold, at least or up to about 100-fold, at least or up to about 200-fold, at least or up to about 300-fold, at least or up to about 400-fold, at least or up to about 500-fold, at least or up to about 600-fold, at least or up to about 700-fold, at least or up to about 800-fold, at least or up to about 900-fold, or at least or up to about 1,000-fold.
[0112] In some embodiments, caspase (e.g., caspase 3 and/or caspase 8) activation (e.g., cleavage thereof) can be measured (e.g., by Western blot) as an indication of inflammation (e.g., TLR-mediated renal tubular inflammation and/or injury) or cell death. Upon administration of the therapeutic agents (e.g., a nucleic acid molecule, such as ODN, e.g., CpG-ODN) via the population of particles of the disclosure to a subject in need thereof, a tissue sample (e.g.,
TUNEL positive cells) of a kidney of the subject can be evaluated for caspase activity.
Treatment with the therapeutic agents via the population of particles of the disclosure can reduce cleavage of a caspase protein (e.g., caspase 3, caspase 8) in kidney cells (e.g., TUNEL positive kidney cells) as compared to a control (e.g., no treatment, non-encapsulated therapeutic agents, inactive encapsulated therapeutic agents) by at least or up to about 0.1 -fold, at least or up to about 0.2-fold, at least or up to about 0.3-fold, at least or up to about 0.4-fold, at least or up to about 0.5-fold, at least or up to about 0.6-fold, at least or up to about 0.7-fold, at least or up to about 0.8-fold, at least or up to about 0.9-fold, at least or up to about 1-fold, at least or up to about 2-fold, at least or up to about 3-fold, at least or up to about 4-fold, at least or up to about 5- fold, at least or up to about 6-fold, at least or up to about 7-fold, at least or up to about 8-fold, at least or up to about 9-fold, at least or up to about 10-fold, at least or up to about 11 -fold, at least or up to about 12-fold, at least or up to about 13 -fold, at least or up to about 14-fold, at least or up to about 15-fold, at least or up to about 16-fold, at least or up to about 17-fold, at least or up to about 18-fold, at least or up to about 19-fold, at least or up to about 20-fold, at least or up to about 25-fold, at least or up to about 30-fold, at least or up to about 35-fold, at least or up to
about 40-fold, at least or up to about 45-fold, at least or up to about 50-fold, at least or up to about 100-fold, at least or up to about 200-fold, at least or up to about 300-fold, at least or up to about 400-fold, at least or up to about 500-fold, at least or up to about 600-fold, at least or up to about 700-fold, at least or up to about 800-fold, at least or up to about 900-fold, or at least or up to about 1,000-fold.
[0113] In some embodiments, upon administration of the therapeutic agents (e.g., a nucleic acid molecule, such as ODN, e.g., CpG-ODN) via the population of particles of the disclosure to a subject in need thereof, a tissue sample (e.g., a biopsy sample) of a kidney of the subject is evaluated (e.g., via histological assessment using an antibody against, for example, Lymphocyte antigen 6 complex locus G6D (Ly6G) or F4/80) for a presence of immune cell (e.g., neutrophil, macrophage) infiltration. Treatment with the therapeutic agents via the population of particles of the disclosure can reduce the degree of immune cell infiltration into the tissue site of interest (e.g., kidney) as compared to a control (e.g., no treatment, non-encapsulated therapeutic agents, inactive encapsulated therapeutic agents) by at least or up to about 0.1 -fold, at least or up to about 0.2-fold, at least or up to about 0.3-fold, at least or up to about 0.4-fold, at least or up to about 0.5-fold, at least or up to about 0.6-fold, at least or up to about 0.7-fold, at least or up to about 0.8-fold, at least or up to about 0.9-fold, at least or up to about 1-fold, at least or up to about 2-fold, at least or up to about 3-fold, at least or up to about 4-fold, at least or up to about 5- fold, at least or up to about 6-fold, at least or up to about 7-fold, at least or up to about 8-fold, at least or up to about 9-fold, at least or up to about 10-fold, at least or up to about 11 -fold, at least or up to about 12-fold, at least or up to about 13 -fold, at least or up to about 14-fold, at least or up to about 15-fold, at least or up to about 16-fold, at least or up to about 17-fold, at least or up to about 18-fold, at least or up to about 19-fold, at least or up to about 20-fold, at least or up to about 25-fold, at least or up to about 30-fold, at least or up to about 35-fold, at least or up to about 40-fold, at least or up to about 45-fold, at least or up to about 50-fold, at least or up to about 100-fold, at least or up to about 200-fold, at least or up to about 300-fold, at least or up to about 400-fold, at least or up to about 500-fold, at least or up to about 600-fold, at least or up to about 700-fold, at least or up to about 800-fold, at least or up to about 900-fold, or at least or up to about 1,000-fold.
[0114] In some embodiments, upon administration of the therapeutic agents (e.g., a nucleic acid molecule, such as ODN, e.g., CpG-ODN) via the population of particles of the disclosure to a subject in need thereof, kidney cells of the subject is evaluated for expression of pro- inflammatory cytokine and/or chemokine (e.g., via PCR). Non-limiting examples of the pro- inflammatory cytokine and/or chemokine can include interleukin (IL) (e.g., IL-6), intercellular
adhesion molecule- 1 (ICM-1), monocyte chemoattractive protein- 1 (MCP-1), keratinocyte chemoattractant (KC), macrophage inflammatory protein-2 (MIP-2), and tumor necrosis factor- alpha (TNF-α). Treatment with the therapeutic agents via the population of particles of the disclosure can reduce expression of one or more pro-inflammatory cytokine and/or chemokines in target cells (e.g., kidney cells) as compared to a control (e.g., no treatment, non-encapsulated therapeutic agents, inactive encapsulated therapeutic agents) by at least or up to about 0.1 -fold, at least or up to about 0.2-fold, at least or up to about 0.3-fold, at least or up to about 0.4-fold, at least or up to about 0.5-fold, at least or up to about 0.6-fold, at least or up to about 0.7-fold, at least or up to about 0.8-fold, at least or up to about 0.9-fold, at least or up to about 1-fold, at least or up to about 2-fold, at least or up to about 3 -fold, at least or up to about 4-fold, at least or up to about 5-fold, at least or up to about 6-fold, at least or up to about 7-fold, at least or up to about 8- fold, at least or up to about 9-fold, at least or up to about 10-fold, at least or up to about 11 -fold, at least or up to about 12-fold, at least or up to about 13 -fold, at least or up to about 14-fold, at least or up to about 15-fold, at least or up to about 16-fold, at least or up to about 17-fold, at least or up to about 18-fold, at least or up to about 19-fold, at least or up to about 20-fold, at least or up to about 25-fold, at least or up to about 30-fold, at least or up to about 35-fold, at least or up to about 40-fold, at least or up to about 45-fold, at least or up to about 50-fold, at least or up to about 100-fold, at least or up to about 200-fold, at least or up to about 300-fold, at least or up to about 400-fold, at least or up to about 500-fold, at least or up to about 600-fold, at least or up to about 700-fold, at least or up to about 800-fold, at least or up to about 900-fold, or at least or up to about 1,000-fold.
[0115] In some embodiments, administration of the therapeutic agents (e.g., a nucleic acid molecule, such as ODN, e.g., CpG-ODN) via the population of nanoparticles of the disclosure can enhance renal functional of the subject to a greater degree than a control (e.g., no treatment, non-encapsulated therapeutic agents, inactive encapsulated therapeutic agents) by at least or up to about 0.1-fold, at least or up to about 0.2-fold, at least or up to about 0.3-fold, at least or up to about 0.4-fold, at least or up to about 0.5-fold, at least or up to about 0.6-fold, at least or up to about 0.7-fold, at least or up to about 0.8-fold, at least or up to about 0.9-fold, at least or up to about 1-fold, at least or up to about 2-fold, at least or up to about 3 -fold, at least or up to about 4- fold, at least or up to about 5-fold, at least or up to about 6-fold, at least or up to about 7-fold, at least or up to about 8-fold, at least or up to about 9-fold, at least or up to about 10-fold, at least or up to about 11 -fold, at least or up to about 12-fold, at least or up to about 13 -fold, at least or up to about 14-fold, at least or up to about 15-fold, at least or up to about 16-fold, at least or up to about 17-fold, at least or up to about 18-fold, at least or up to about 19-fold, at least or up to
about 20-fold, at least or up to about 25-fold, at least or up to about 30-fold, at least or up to about 35-fold, at least or up to about 40-fold, at least or up to about 45-fold, at least or up to about 50-fold, at least or up to about 100-fold, at least or up to about 200-fold, at least or up to about 300-fold, at least or up to about 400-fold, at least or up to about 500-fold, at least or up to about 600-fold, at least or up to about 700-fold, at least or up to about 800-fold, at least or up to about 900-fold, or at least or up to about 1,000-fold, as ascertained by evaluating a biochemical measurement of target molecules (e.g., creatinine, urea) in the serum, plasma, or urine.
[0116] In some embodiments, administration of the therapeutic agents (e.g., a nucleic acid molecule, such as ODN, e.g., CpG-ODN) via the population of nanoparticles of the disclosure can preserve or prolong renal functional of the subject for a longer duration than a control (e.g., no treatment, non-encapsulated therapeutic agents, inactive encapsulated therapeutic agents) by at least or up to about 0.1-fold, at least or up to about 0.2-fold, at least or up to about 0.3-fold, at least or up to about 0.4-fold, at least or up to about 0.5-fold, at least or up to about 0.6-fold, at least or up to about 0.7-fold, at least or up to about 0.8-fold, at least or up to about 0.9-fold, at least or up to about 1-fold, at least or up to about 2-fold, at least or up to about 3 -fold, at least or up to about 4-fold, at least or up to about 5-fold, at least or up to about 6-fold, at least or up to about 7-fold, at least or up to about 8-fold, at least or up to about 9-fold, at least or up to about 10-fold, at least or up to about 11 -fold, at least or up to about 12-fold, at least or up to about 13- fold, at least or up to about 14-fold, at least or up to about 15-fold, at least or up to about 16-fold, at least or up to about 17-fold, at least or up to about 18-fold, at least or up to about 19-fold, at least or up to about 20-fold, at least or up to about 25-fold, at least or up to about 30-fold, at least or up to about 35-fold, at least or up to about 40-fold, at least or up to about 45-fold, at least or up to about 50-fold, at least or up to about 100-fold, at least or up to about 200-fold, at least or up to about 300-fold, at least or up to about 400-fold, at least or up to about 500-fold, at least or up to about 600-fold, at least or up to about 700-fold, at least or up to about 800-fold, at least or up to about 900-fold, or at least or up to about 1,000-fold, as ascertained by evaluating a biochemical measurement of target molecules (e.g., creatinine, urea) in the serum, plasma, or urine.
[0117] In some embodiments, as compared to a control (e.g., no treatment, non-encapsulated therapeutic agents, inactive encapsulated therapeutic agents), administration of the therapeutic agents (e.g., a nucleic acid molecule, such as ODN, e.g., CpG-ODN) via the population of particles of the present invention can (i) reduce a therapeutic dose of the therapeutic agents in eliciting a desired biological effect (e.g., treating or ameliorating symptoms of a disease), (ii) reduce side-effects related to the therapeutic agents, or (iii) reduce a number of administrations of the therapeutic agents that is sufficient to elicit a therapeutic effect.
[0118] As compared to a control (e.g., no treatment, non-encapsulated therapeutic agents, inactive encapsulated therapeutic agents), administration of the therapeutic agents via the population of particles of the disclosure can reduce one or more side effects of the therapeutic agents (e.g., a nucleic acid molecule, such as ODN, e.g., CpG-ODN) by at least or up to about 0.1-fold, at least or up to about 0.2-fold, at least or up to about 0.3-fold, at least or up to about
0.4-fold, at least or up to about 0.5-fold, at least or up to about 0.6-fold, at least or up to about
0.7-fold, at least or up to about 0.8-fold, at least or up to about 0.9-fold, at least or up to about 1- fold, at least or up to about 2-fold, at least or up to about 3 -fold, at least or up to about 4-fold, at least or up to about 5-fold, at least or up to about 6-fold, at least or up to about 7-fold, at least or up to about 8-fold, at least or up to about 9-fold, at least or up to about 10-fold, at least or up to about 11 -fold, at least or up to about 12-fold, at least or up to about 13 -fold, at least or up to about 14-fold, at least or up to about 15-fold, at least or up to about 16-fold, at least or up to about 17-fold, at least or up to about 18-fold, at least or up to about 19-fold, at least or up to about 20-fold, at least or up to about 25-fold, at least or up to about 30-fold, at least or up to about 35-fold, at least or up to about 40-fold, at least or up to about 45-fold, at least or up to about 50-fold, at least or up to about 100-fold, at least or up to about 200-fold, at least or up to about 300-fold, at least or up to about 400-fold, at least or up to about 500-fold, at least or up to about 600-fold, at least or up to about 700-fold, at least or up to about 800-fold, at least or up to about 900-fold, or at least or up to about 1,000-fold.
[0119] As compared to a control (e.g., no treatment, non-encapsulated therapeutic agents, inactive encapsulated therapeutic agents), administration of the therapeutic agents via the population of particles of the disclosure can reduce the number of administrations of the therapeutic agents that is sufficient to elicit a desired therapeutic effect by at least or up to about
0.1-fold, at least or up to about 0.2-fold, at least or up to about 0.3-fold, at least or up to about
0.4-fold, at least or up to about 0.5-fold, at least or up to about 0.6-fold, at least or up to about
0.7-fold, at least or up to about 0.8-fold, at least or up to about 0.9-fold, at least or up to about 1- fold, at least or up to about 2-fold, at least or up to about 3 -fold, at least or up to about 4-fold, at least or up to about 5-fold, at least or up to about 6-fold, at least or up to about 7-fold, at least or up to about 8-fold, at least or up to about 9-fold, at least or up to about 10-fold, at least or up to about 11 -fold, at least or up to about 12-fold, at least or up to about 13 -fold, at least or up to about 14-fold, at least or up to about 15-fold, at least or up to about 16-fold, at least or up to about 17-fold, at least or up to about 18-fold, at least or up to about 19-fold, at least or up to about 20-fold, at least or up to about 25-fold, at least or up to about 30-fold, at least or up to about 35-fold, at least or up to about 40-fold, at least or up to about 45-fold, at least or up to
about 50-fold, at least or up to about 100-fold, at least or up to about 200-fold, at least or up to about 300-fold, at least or up to about 400-fold, at least or up to about 500-fold, at least or up to about 600-fold, at least or up to about 700-fold, at least or up to about 800-fold, at least or up to about 900-fold, or at least or up to about 1,000-fold.
[0120] In some embodiments, the population of particles of the disclosure can selectively target or localize to one or more kidney cells (i.e., renal cells) selected from: kidney glomerulus parietal cell, kidney glomerulus podocyte, kidney proximal tubule brush border cell, loop of Henle thin segment cell, thick ascending limb cell, kidney distal tubule cell, collecting duct principal cell, collecting duct intercalated cell, and interstitial kidney cells.
[0121] In some embodiments, a number of kidney diseases can benefit from therapeutics utilizing the population of particles of the disclosure that is capable of, e.g., site-directed accumulation, controlled temporal release, and protection of a therapeutic payload. Non-limiting examples of kidney diseases include lupus, glomerulonephritis, and renal cell carcinoma (RCC), each of which can arise in the proximal tubules of the kidneys. Alternatively, when configured to target other types of tissues (e.g., heart, lung, spleen, or liver) selectively, therapeutics utilizing the population of particles of the disclosure can be utilized to treat or ameliorate symptoms of other tissue-specific diseases.
[0122] In some embodiments, therapeutics utilizing the population of particles of the disclosure can be utilized to treat cancer. Non-limiting examples of cancer include adrenal cortical cancer, anal cancer, bile duct cancer, bladder cancer, bone cancer, brain or a nervous system cancer, breast cancer, cervical cancer, colon cancer, rectal cancer, colorectal cancer, endometrial cancer, esophageal cancer, Ewing family of tumor, eye cancer, gallbladder cancer, gastrointestinal carcinoid cancer, gastrointestinal stromal cancer, Hodgkin Disease, intestinal cancer, Kaposi Sarcoma, kidney cancer, large intestine cancer, laryngeal cancer, hypopharyngeal cancer, laryngeal and hypopharyngeal cancer, leukemia, acute lymphocytic leukemia (ALL), acute myeloid leukemia (AML), chronic lymphocytic leukemia (CLL), chronic myeloid leukemia (CML), chronic myelomonocytic leukemia (CMML), non-HCL lymphoid malignancy (hairy cell variant, splenic marginal zone lymphoma (SMZL), splenic diffuse red pulp small B-cell lymphoma (SDRPSBCL), chronic lymphocytic leukemia (CLL), prolymphocytic leukemia, low grade lymphoma, systemic mastocytosis, or splenic lymphoma/leukemia unclassifiable (SLLU)), liver cancer, lung cancer, non-small cell lung cancer, small cell lung cancer, lung carcinoid tumor, lymphoma, lymphoma of the skin, malignant mesothelioma, multiple myeloma, nasal cavity cancer, paranasal sinus cancer, nasal cavity and paranasal sinus cancer, nasopharyngeal cancer, neuroblastoma, non-Hodgkin lymphoma, oral cavity cancer, oropharyngeal cancer, oral
cavity and oropharyngeal cancer, osteosarcoma, ovarian cancer, pancreatic cancer, penile cancer, pituitary tumor, prostate cancer, retinoblastoma, rhabdomyosarcoma, salivary gland cancer, sarcoma, adult soft tissue sarcoma, skin cancer, basal cell skin cancer, squamous cell skin cancer, basal and squamous cell skin cancer, melanoma, stomach cancer, small intestine cancer, testicular cancer, thymus cancer, thyroid cancer, uterine sarcoma, uterine cancer, vaginal cancer, vulvar cancer, Waldenstrom Macroglobulinemia, and Wilms Tumor.
[0123] A cell can be in vivo. A cell can be ex vivo. A cell can be an isolated cell. A cell can be a cell inside of an organism. A cell can be an organism. A cell can be a cell in a cell culture. A cell can be one of a collection of cells. A cell can be a mammalian cell or derived from a mammalian cell. A cell can be a rodent cell or derived from a rodent cell. A cell can be a human cell or derived from a human cell. A cell can be a prokaryotic cell or derived from a prokaryotic cell. A cell can be a bacterial cell or can be derived from a bacterial cell. A cell can be an archaeal cell or derived from an archaeal cell. A cell can be a eukaryotic cell or derived from a eukaryotic cell. A cell can be a pluripotent stem cell. A cell can be a plant cell or derived from a plant cell. A cell can be an animal cell or derived from an animal cell. A cell can be an invertebrate cell or derived from an invertebrate cell. A cell can be a vertebrate cell or derived from a vertebrate cell. A cell can be a microbe cell or derived from a microbe cell. A cell can be a fungi cell or derived from a fungi cell. A cell can be from a specific organ or tissue.
[0124] A cell can be a stem cell or progenitor cell. Cells can include stem cells (e.g., adult stem cells, embryonic stem cells, iPS cells) and progenitor cells (e.g., cardiac progenitor cells, neural progenitor cells, etc.). Cells can include mammalian stem cells and progenitor cells, including rodent stem cells, rodent progenitor cells, human stem cells, human progenitor cells, etc. Clonal cells can comprise the progeny of a cell. A cell can comprise a target nucleic acid. A cell can be in a living organism. A cell can be a genetically modified cell. A cell can be a host cell.
[0125] A cell can be a diseased cell. A diseased cell can have altered metabolic, gene expression, and/or morphologic features. A diseased cell can be a cancer cell, a diabetic cell, and an apoptotic cell. A diseased cell can be a cell from a diseased subject. Illustrative diseases can include blood disorders, cancers, metabolic disorders, eye disorders, organ disorders, musculoskeletal disorders, and cardiac disease.
[0126] Non-limiting examples of cells in which a subject population of particles can be utilized include lymphoid cells, such as B cell, T cell (Cytotoxic T cell, Natural Killer T cell, Regulatory T cell, T helper cell), Natural killer cell, cytokine induced killer (CIK) cells; myeloid cells, such as granulocytes (Basophil granulocyte, Eosinophil granulocyte, Neutrophil granulocyte/Hypersegmented neutrophil), Monocyte/Macrophage, Red blood cell (Reticulocyte),
Mast cell, Thrombocyte/Megakaryocyte, Dendritic cell; cells from the endocrine system, including thyroid (Thyroid epithelial cell, Parafollicular cell), parathyroid (Parathyroid chief cell, Oxyphil cell), adrenal (Chromaffin cell), pineal (Pinealocyte) cells; cells of the nervous system, including glial cells (Astrocyte, Microglia), Magnocellular neurosecretory cell, Stellate cell, Boettcher cell, and pituitary (Gonadotrope, Corticotrope, Thyrotrope, Somatotrope, Lactotroph ); cells of the Respiratory system, including Pneumocyte (Type I pneumocyte, Type II pneumocyte), Clara cell, Goblet cell, Dust cell; cells of the circulatory system, including Myocardiocyte, Pericyte; cells of the digestive system, including stomach (Gastric chief cell, Parietal cell), Goblet cell, Paneth cell, G cells, D cells, ECL cells, I cells, K cells, S cells; enteroendocrine cells, including enterochromaffm cell, APUD cell, liver (Hepatocyte, Kupffer cell), Cartilage/bone/muscle; bone cells, including Osteoblast, Osteocyte, Osteoclast, teeth (Cementoblast, Ameloblast); cartilage cells, including Chondroblast, Chondrocyte; skin cells, including Trichocyte, Keratinocyte, Melanocyte (Nevus cell); muscle cells, including Myocyte; urinary system cells, including Podocyte, Juxtaglomerular cell, Intraglomerular mesangial cell/Extraglomerular mesangial cell, Kidney proximal tubule brush border cell, Macula densa cell; reproductive system cells, including Spermatozoon, Sertoli cell, Leydig cell, Ovum; and other cells, including Adipocyte, Fibroblast, Tendon cell, Epidermal keratinocyte (differentiating epidermal cell), Epidermal basal cell (stem cell), Keratinocyte of fingernails and toenails, Nail bed basal cell (stem cell), Medullary hair shaft cell, Cortical hair shaft cell, Cuticular hair shaft cell, Cuticular hair root sheath cell, Hair root sheath cell of Huxley's layer, Hair root sheath cell of Henle's layer, External hair root sheath cell, Hair matrix cell (stem cell), Wet stratified barrier epithelial cells, Surface epithelial cell of stratified squamous epithelium of cornea, tongue, oral cavity, esophagus, anal canal, distal urethra and vagina, basal cell (stem cell) of epithelia of cornea, tongue, oral cavity, esophagus, anal canal, distal urethra and vagina, Urinary epithelium cell (lining urinary bladder and urinary ducts), Exocrine secretory epithelial cells, Salivary gland mucous cell (polysaccharide-rich secretion), Salivary gland serous cell (glycoprotein enzyme - rich secretion), Von Ebner's gland cell in tongue (washes taste buds), Mammary gland cell (milk secretion), Lacrimal gland cell (tear secretion), Ceruminous gland cell in ear (wax secretion), Eccrine sweat gland dark cell (glycoprotein secretion), Eccrine sweat gland clear cell (small molecule secretion). Apocrine sweat gland cell (odoriferous secretion, sex -hormone sensitive), Gland of Moll cell in eyelid (specialized sweat gland), Sebaceous gland cell (lipid-rich sebum secretion), Bowman's gland cell in nose (washes olfactory epithelium), Brunner's gland cell in duodenum (enzymes and alkaline mucus), Seminal vesicle cell (secretes seminal fluid components, including fructose for swimming sperm), Prostate gland cell (secretes seminal fluid
components), Bulbourethral gland cell (mucus secretion), Bartholin's gland cell (vaginal lubricant secretion), Gland of Littre cell (mucus secretion), Uterus endometrium cell (carbohydrate secretion), Isolated goblet cell of respiratory and digestive tracts (mucus secretion), Stomach lining mucous cell (mucus secretion), Gastric gland zymogenic cell (pepsinogen secretion), Gastric gland oxyntic cell (hydrochloric acid secretion), Pancreatic acinar cell (bicarbonate and digestive enzyme secretion), Paneth cell of small intestine (lysozyme secretion), Type II pneumocyte of lung (surfactant secretion), Clara cell of lung, Hormone secreting cells, Anterior pituitary cells , Somatotropes, Lactotropes, Thyrotropes, Gonadotropes, Corticotropes, Intermediate pituitary cell, Magnocellular neurosecretory cells, Gut and respiratory tract cells , Thyroid gland cells, thyroid epithelial cell, parafollicular cell, Parathyroid gland cells, Parathyroid chief cell, Oxyphil cell, Adrenal gland cells, chromaffin cells, Ley dig cell of testes, Theca interna cell of ovarian follicle, Corpus luteum cell of ruptured ovarian follicle, Granulosa lutein cells, Theca lutein cells, Juxtaglomerular cell (renin secretion), Macula densa cell of kidney, Metabolism and storage cells, Barrier function cells (Lung, Gut, Exocrine Glands and Urogenital Tract), Kidney, Type I pneumocyte (lining air space of lung), Pancreatic duct cell (centroacinar cell), Nonstriated duct cell (of sweat gland, salivary gland, mammary gland, etc.), Duct cell (of seminal vesicle, prostate gland, etc.), Epithelial cells lining closed internal body cavities, Ciliated cells with propulsive function, Extracellular matrix secretion cells, Contractile cells; Skeletal muscle cells, stem cell, Heart muscle cells, Blood and immune system cells, Erythrocyte (red blood cell), Megakaryocyte (platelet precursor), Monocyte, Connective tissue macrophage (various types), Epidermal Langerhans cell, Osteoclast (in bone), Dendritic cell (in lymphoid tissues), Microglial cell (in central nervous system), Neutrophil granulocyte, Eosinophil granulocyte, Basophil granulocyte, Mast cell, Helper T cell, Suppressor T cell, Cytotoxic T cell, Natural Killer T cell, B cell, Natural killer cell, Reticulocyte, Stem cells and committed progenitors for the blood and immune system (various types), Pluripotent stem cells, Totipotent stem cells, Induced pluripotent stem cells, adult stem cells, Sensory transducer cells, Autonomic neuron cells, Sense organ and peripheral neuron supporting cells, Central nervous system neurons and glial cells, Lens cells, Pigment cells, Melanocyte, Retinal pigmented epithelial cell, Germ cells, Oogonium/Oocyte, Spermatid, Spermatocyte, Spermatogonium cell (stem cell for spermatocyte), Spermatozoon, Nurse cells, Ovarian follicle cell, Sertoli cell (in testis), Thymus epithelial cell, Interstitial cells, and Interstitial kidney cells.
[0127] In some embodiments of the aspects herein, methods of the disclosure are performed in a subject. A subject can be a human. A subject can be a mammal (e.g., rat, mouse, cow, dog, pig, sheep, horse). A subject can be a vertebrate or an invertebrate. A subject can be a laboratory
animal. A subject can be a patient. A subject can be suffering from a disease. A subject can display symptoms of a disease. A subject can have a disease even if not displaying symptoms. A subject can be under medical care of a caregiver (e.g., the subject is hospitalized and is treated by a physician).
EXAMPLES
Example 1: A population of nanoparticles for treating kidney injury
[0128] The present example provides a therapy for ischemic AKI with selective kidney-targeted delivery of a selective TLR9 antagonist in mice subjected to renal IR injury. Mice deficient in renal proximal tubular TLR9 were protected against renal ischemia reperfusion (IR) injury by delivering a selective TLR antagonist via a population of 300 nm to 400 nm polymer (e.g., PLGA-PEG)-based mesoscale nanoparticles (MNP) that localize to the renal tubules after intravenous injection. Mice were subjected to sham surgery or 30 min renal ischemia and reperfusion (IR) injury after receiving MNPs encapsulating selective TLR9 antagonist (e.g., unmethylated CpG-ODN TLR9 antagonist, such as ODN2088) or MNP-encapsulated with a negative control ODN. Mice treated with MNP-encapsulated TLR9 antagonist either 6 hr before renal ischemia, at the time of reperfusion, or 0.5 hr after reperfusion were protected against ischemic AKI. TLR9 antagonist-encapsulating MNPs attenuated renal tubular necrosis, attenuated, inflammation, decreased pro-inflammatory cytokine synthesis, decreased neutrophil and macrophage infiltration, and decreased apoptosis of kidney cells (as indicated by decreased DNA fragmentation and caspase 3/8 activation), as compared to, for example, negative control MNP treated mice. The present example suggests that renal proximal tubular TLR9 activation exacerbates ischemic AKI by promoting renal tubular inflammation, apoptosis, and necrosis after IR, and a therapy based on selective kidney tubular targeting of TLR9 using MNP -based drug (e.g., CpG-ODN TLR9 antagonist) delivery can treat or ameliorate the ischemic AKI.
Generation of mesoscale nanoparticles (MNPs ) that encapsulate CpG-ODN TLR9 antagonist
[0129] Mesoscale nanoparticles (MNPs) encapsulating the CpG-ODN TLR9 antagonist (e.g., ODN2088) or control ODN were formulated. Poly(lactic-co-glycolic acid) (PLGA; molecular weight between about 38 kDa and 54 kDa) was conjugated to carboxylic acid-terminated polyethylene glycol (PEG; molecular weight of about 5 kDa) to generate a population of PLGA- PEG co-polymers prior to particle formulation. The conjugated co-polymer (e.g., about 100 mg)
was dissolved in a solvent (e.g., about 2 mL acetonitrile). Following, the CpG-ODN TLR9 antagonist (e.g., about 50 μg of ODN2088) or control ODN was added (e.g., as dissolved in water) to the co-polymer solution and bath sonicated (e.g., for about 2 minutes). The resulting emulsion was added to an aqueous solution that comprises surfactants (e.g., a mixture of about 4mL of purified water and about 75 pL of Pluronic F-68) and centrifuged (e.g., at about 5400 X g for about 15 minutes). A resulting pellet of MNPs was washed (e.g., with 10 mL purified water) and centrifuged (e.g., at about 5400 X g for about 15 minutes). The pellet was resuspended (e.g., in a 2% sucrose solution). The pellet was lyophilized for storage at -20°C.
Characterization of the CpG-ODN TLR9 antagonist encapsulating MNPs.
[0130] Hydrodynamic diameter and polydispersity index (PDI) of the MNPs were characterized via dynamic light scattering (DLS) in suspension (e.g., suspended in about 10 mg/mL phosphate- buffered saline).
[0131] Loading of the CpG-ODN into MNPs was assessed by measuring an amount of free and non-encapsulated ODN. A powder of lyophilized MNPs (e.g., about 10 mg) was dissolved in a solvent (e.g., 200 pL acetonitrile) and shaken (e.g., at room temperature for 30 minutes). Subsequently, a buffer solution (e.g., 300 pL Tris-EDTA buffer) was added, and a resulting mixture was centrifuged (e.g., at about 31,000 X g for 30 minutes). A resulting supernatant was used to quantify free, non-encapsulated ODN.
[0132] Lyophilized MNPs that contain the CpG-ODN TLR9 exhibited a mean diameter of 311.7 ± 12.1 nm and a PDI of 0.316 ± 0.048. An amount of the CpG-ODN loaded to the MNPs was about 89 ng of CpG-ODN per 1 mg of MNP. Lyophilized MNPs that contain control ODN exhibited a mean diameter of 311.6 ± 5.0 nm and a PDI of 0.208 ± 0.032. An amount of control ODN loaded to the MNPs was about 56.7 ng of control ODN per 1 mg of MNP.
Renal IR injury in mice and treatment with MNPs
[0133] Based on IACUC approval, mice (e.g., about 20-25g male C57BL/6 mice) were anesthetized (e.g., via intraperitoneal injection (i.p.) of pentobarbital, such as at 50 mg pentobarbital per 1 kg body weight). Some mice received (e.g., intravenously (i.v.) administered with) MNPs encapsulating CpG-ODN TLR9 antagonist or control ODN (e.g., either about 37.5 or 75 mg of MNP per 1 kg weight of the animal, to deliver either about 3.26 or 6.5 μg of ODN per 1 kg weight of the animal) before renal ischemia (e.g., at about 5 hr before renal ischemia). Mice were subjected to right nephrectomy and left renal ischemia (e.g., about 30 min). Sham- operated animals underwent anesthesia followed by laparotomy, right nephrectomy, bowel
manipulations, and wound closure without renal ischemia. Body temperature of the animals were sustained (e.g., at about 37°C) during surgery and during recovery from anesthesia. A separate cohort of mice received (e.g., via i.v. administration) about 75 mg of MNP loaded with ODN (e.g., CpG-ODN TLR9 antagonist or control ODN) per 1 kg of the weight of the animal at the time of reperfusion or 1.5 hr after the reperfusion. Some mice received e.g., via i.v. administration) non-encapsulated ODN (e.g., CpG-ODN TLR9 antagonist or control ODN) 6 hr before renal ischemia, to determine whether systemic TLR9 blockade can protect a subject against ischemic AKI. For pain management, all mice received buprenorphine SR (e.g., subcutaneously (s.c.) at about 0.5-1 mg buprenorphine SR per kg weight of the animal) prior to the surgery.
[0134] To confirm renal tubular-preferential (or targeted) delivery of MNPs, kidney sections of mice treated with MNPs that encapsulate ODN (e.g., CpG-ODN TLR9 antagonist or control ODN) or with saline vehicle control at about 6 hr prior to the renal IR injury were stained with anti -PEG antibody (e.g., to detect the presence of PLNA-PEG MNPs), anti-PHA Lectin (PHA-L) antibody (e.g., to detect the presence of renal proximal tubular cells), and DAPI (e.g., to detect cell nucleus). As shown in FIG. 1 (e.g., 200X images shown, representative of 3 experiments), the renal proximal tubular distribution pattern for the MNPs was confirmed by co-localization of PEG staining with PHA lectin staining in mice treated with 75 mg/kg MNP. In some embodiments, almost none or minimal MNP localization to other tubular segments, endothelial cells, or mesangial cells in the glomeruli was observed. In some embodiments, mice injected with vehicle control showed no PEG staining.
[0135] Kidney-selective targeting of the MNPs as disclosed herein was also assessed. MNPs encapsulating a dye (e.g., Cy5 mimic 3,3’-diethylthiadicarbocyanine iodide (DEDC)) were prepared as disclosed herein. At 6 hr before (i) sham-surgery or (ii) 30 min renal ischemia and 24 hr reperfusion (Renal IR), the mice were administered (e.g., via i.v. administration) with the dye-loaded MNPs (e.g., at about 75 mg of MNPS per 1 kg of the animal), and various tissue samples (e.g., kidney, liver, spleen, lung and heart) were imaged for near-infrared fluorescence intensity, as an indication of the presence of the MNPs. As shown in FIG. 2 (top), representative images (e.g., overlay of brightfield and near-infrared fluorescence imaging) of kidney, liver, spleen, lung and heart (top) exhibit preferential targeting of the MNPs to the kidney as compared to other tissue samples. As shown in FIG. 2 (bottom), the average fluorescence intensity quantified from the near-infrared fluorescence intensity images exhibited at least about 30-fold kidney selectivity over other organs for mice undergone either (i) sham surgery or (ii) Renal IR (N=3; data were background subtracted and represent mean ± standard error of the mean (SEM)).
Protection against ischemic AKI in mice
[0136] Mice were injected with MNPs loaded with ODN (e.g., CpG-ODN TLR9 antagonist or control ODN), and subsequently subjected to sham-surgery (N=4) or 30 min renal ischemia and 24 hr reperfusion (RIR, N=6-7). Some mice were injected with the MNPs at about 6 hr before renal ischemia. Some mice were injected with the MNPs at the time of reperfusion or 1.5 hr after reperfusion. Some mice were intravenously injected with 5 mg/kg naked ODN (e.g., ., CpG- ODN TLR9 antagonist or control ODN) at about 6 hr before renal ischemia. Twenty-four hours after renal IR or sham surgery, plasma creatine (PCr) and blood urea nitrogen (BUN) levels were measured (e.g., via enzymatic creatinine reagent kits), and neutrophil gelatinase-associated lipocalin (NGAL) mRNA levels in kidney cells were measured (e.g., via qRT-PCR). For statistical analysis, one-way ANOVA plus Tukey’s post hoc multiple comparison test was used to detect significant changes.
[0137] As shown in FIG. 3 (*P<0.05 vs. control MNP injected mice subjected to sham surgery; #P<0.05 vs. control ODN mice subjected to renal IR; error bars represent 1 SEM; N=4-7), PCr and BUN levels were similar between sham-operated mice that were injected with MNPs encapsulated with either control ODN (i.e., ConODN MNP) or CpG-ODN TLR9 antagonist (i.e., ODN2088 MNP). Mice treated with the ConODN MNP and subjected to renal IR had significantly higher levels of PCr, BUN, and kidney NGAL mRNA, as compared to sham- operated mice. Mice treated with the CpG-ODN TLR9 antagonist loaded MNPs at about 6 hr before renal ischemia were protected against ischemic AKI compared to mice treated with the control ODN-loaded MNPs, as demonstrated by reduced levels of levels of PCr, BUN, and kidney NGAL mRNA. In addition, mice treated with the CpG-ODN TLR9 antagonist loaded MNPs (e.g., at 75mg/kg MNP per weight of the animal) at the time of reperfusion or 1.5 hr after reperfusion revealed reduced levels of PCr, BUN, and kidney NGAL mRNA as compared to controls. In contrast, naked CpG-ODN TLR9 antagonist given i.v. at about 6 hr before renal ischemia failed to protect against ischemic AKI in mice.
Reduction of renal tubular necrosis after ischemic AKI
[0138] FIG. 4 shows representative hematoxylin and eosin (H&E) staining images of kidney samples from mice treated with MNPs encapsulating ODN (e.g., e.g., CpG-ODN TLR9 antagonist or control ODN) and subjected to either (i) sham surgery or (ii) 30 min renal IR and 24 hr reperfusion (magnification 200X; N=6-7). Some mice were injected with MNPs loaded with CpG-ODN TLR9 antagonist at about 6 hr before renal ischemia. Some mice were injected
with MNPs loaded with CpG-ODN TLR9 antagonist at the time of reperfusion or 1.5 hr after reperfusion. Some mice were intravenously administered with naked CpG-ODN TLR9 antagonist (e.g., at about 5 mg/kg of ODN per weight of the animal) at about 6 hr before renal ischemia. Twenty-four hours after renal IR or sham surgery, the kidney sections stained with H&E were assessed using a grading scale of kidney necrotic IR injury to the proximal tubules (0- 4, Renal Injury Score). The renal pathologist was blinded to the experimental conditions. Deidentified slides were H&E-stained coronal cross-sections of bivalved whole kidney showing full-thickness cortex and medulla. The cortical and medullary parenchyma was evaluated in its entirety in all the microscopic fields covering the entire slide to generate the Jablonski score. [0139] As shown in FIG. 4, mice treated with the MNPs encapsulating the control ODN and subjected to renal IR showed severe tubular necrosis, proteinaceous casts, and increased tubular dilatation and congestion. In contrast, mice treated with the MNPs encapsulating the CpG-ODN TLR9 antagonist at about 6 hr before renal ischemia, at the time of reperfusion, or at about 1.5 hr after perfusion exhibited decreased renal tubular necrosis, decreased congestion, and decreased cast formation, as compared to controls. As shown in FIG. 5, kidneys from mice treated with the MNPs encapsulating the CpG-ODN TLR9 antagonist exhibited significantly reduced renal tubular injury scores, as compared to mice treated with the MNPs encapsulating the control ODN after IR. In contrast, i.v. administration of the non-encapsulated ODN (e.g., naked CpG-ODN TLR9 antagonist) at about 6 hr before renal ischemia failed to produce kidney necrosis score in mice subjected to renal IR.
[0140] Renal injury scores assessing the degree of renal tubular necrosis are also shown here (scale: 0-4) 24 hours after renal IR. *P<0.05 vs. control MNP injected mice subjected to renal IR injury. For statistical analysis, the Mann-Whitney nonparametric test was used to detect significant changes.
Attenuation of kidney cell apoptosis after ischemic AKI
[0141] Twenty-four hours after renal IR or sham surgery, kidney sections from mice treated with various MNPs or controls were stained to identify cells positive for terminal deoxynucleotidyl transferase deoxyuridine triphosphate nick end labeling (TUNEL), which staining is indicative of a presence of fragmented DNA (or renal tubular apoptosis).
[0142] FIG. 6 shows representative images of the TUNEL stained kidney sections (magnification 200X). TUNEL positive cells were detected in mice treated with (i) the MNPs encapsulating the control ODN or (ii) the non-encapsulated ODN (e.g., naked CpG-ODN TLR9 antagonist), and subjected to renal IR injury, suggestive of renal tubular apoptosis in the kidneys. In contrasted, a
decreased presence of the TUNEL positive cells were detected in mice treated with the MNPs encapsulating the CpG-ODN TLR9 antagonist, as compared to the controls.
[0143] Apoptotic TUNEL positive cells were further quantified (e.g., in 5-7 randomly chosen 200X microscope images in the corticomedullary junction) and normalized per area (e.g., per 200X field). As shown in FIG. 7 (*P<0.05 vs. control MNP injected mice subjected to sham surgerye #P<0.05 vs. control ODN mice subjected to renal IR; error bars represent 1 SEM), TUNEL positive kidney cell counts were significantly reduced in mice treated with the MNPs encapsulating the CpG-ODN TLR9 antagonist at about 6 hr prior to renal ischemia, at the time of reperfusion, or at about 1.5 hr after reperfusion, as compared to mice treated with (i) the MNPs encapsulating the control ODN or (ii) the non-encapsulated ODN (e.g., naked CpG-ODN TLR9 antagonist).
[0144] Furthermore, activation of caspase 3 and caspase 8 in kidney lysates was assessed via Western blot (e.g., by using at least primary antibodies for mouse caspase 3 and caspase 8). As shown in FIG. 8, caspase 3 and caspase 8 cleavages (as indications of caspase 3 and caspase 8 activation) were significantly reduced in mice treated with the MNPs encapsulating the CpG- ODN TLR9 antagonist at about 6 hr before renal ischemia, as compared to mice treated with the MNPs encapsulating the control ODN under similar conditions.
[0145] To show specific inhibition of the TLR9 signaling pathway in tubular cells by the MNPs as disclosed herein, freshly isolated mouse proximal tubule cells were treated with the TLR9-activating ODN (e.g., about 5 mM of naked ODN1668) for about 3 days, either with or without pretreatment of the cells with the MNPs encapsulating the CpG-ODN TLR9 antagonist (e.g., about 1 mM of encapsulated ODN2088). As shown in FIG. 9 (*P<0.05 vs. control MNP injected mice subjected to sham surgery; #P<0.05 vs. control ODN mice subjected to renal IR; error bars represent 1 SEM), the pretreatment with the MNPs encapsulating the CpG-ODN TLR9 antagonist significantly reduced the TLR-activation medicated induction of caspase 3 and caspase 8 fragmentation.
Reduction of neutrophil and macrophase infiltration in kidney after ischemic AKI [0146] Mice were treated with the MNPs as disclosed herein (or controls) and subjected to either (i) sham surgery or (ii) Renal IR. Following, kidney sections from the animals were stained for neutrophils via immunohistochemistry (e.g., by using anti -Lymphocyte antigen 6 complex locus G6D (Ly6G) antibody) and imaged for analysis. Primary IgG2a antibody (MCA1212, AbD Serotec, Raleigh, NC) was utilized as a negative isotype control. As shown in FIG. 10, kidney neutrophil infiltration (as indicated by dark brown staining) was attenuated by treating the mice
the MNPs encapsulating the CpG-ODN TLR9 antagonist at about 6 hr before renal ischemia, at the time of reperfusion, or at about 1.5 hr after reperfusion, as compared to that in mice treated with (i) the MNPs encapsulating the control ODN or (ii) the non-encapsulated ODN (e.g., naked CpG-ODN TLR9 antagonist). As shown in FIG. 11 (*P<0.05 vs. control MNP injected mice subjected to sham surgery; #P<0.05 vs. control ODN mice subjected to renal IR; error bars represent 1 SEM), quantification of the Ly6G-positive cells in the kidney sections confirmed that the kidney neutrophil infiltration was significantly decreased by treating the mice the MNPs encapsulating the CpG-ODN TLR9 antagonist at about 6 hr before renal ischemia, at the time of reperfusion, or at about 1.5 hr after reperfusion, as compared to that in mice treated with (i) the MNPs encapsulating the control ODN or (ii) the non-encapsulated ODN (e.g., naked CpG-ODN TLR9 antagonist).
[0147] The abovementioned tissue sections were also stained for macrophage via immunohistochemistry (e.g., by using anti- F4/80 antibody) and imaged for analysis. As shown in FIG. 12, kidney macrophage infiltration was higher in mice treated with the MNPs encapsulating the control ODN at about 6 hours before being subjected to renal IR, as compared to that in mice treated with the MNPs encapsulating the CpG-ODN TLR9 antagonist at about 6 hours before being subjected to renal IR. As quantified in FIG. 13 (*P<0.05 vs. control MNP injected mice subjected to sham surgery; #P<0.05 vs. control ODN mice subjected to renal IR; error bars represent 1 SEM), the kidney macrophage infiltration was significantly higher in mice treated with the MNPs encapsulating the control ODN at about 6 hours before being subjected to renal IR, as compared to that in mice treated with the MNPs encapsulating the CpG-ODN TLR9 antagonist at about 6 hours before being subjected to renal IR.
Downresulation of pro-inflammatory chemokine and cytokine induction after ischemic AKI [0148] Mice were treated with the MNPs as disclosed herein (or controls) and subjected to either (i) sham surgery or (ii) Renal IR. At about 24 hr after the sham surgery or the Renal IR, kidney lysates were collected and analyzed via quantitative RT-PCR with primers listed in Table 1 to measure expression levels of pro-inflammatory cytokine and chemokine mRNAs in the kidney (e.g., expression levels of keratinocyte-derived cytokine (KC), monocyte chemoattractive protein-1 (MCP-1), macrophage inflammatory protein-2 (MIP-2), tumor necrosis factor-a (TNF- a), interleukin-6 (IL-6), and intercellular adhesion molecule- 1 (ICAM-1)). Fold increases in the mRNAs of interest were normalized to GAPDH. As shown in FIG. 14 (*P < 0.05 vs. control MNP injected sham-operated mice; #P < 0.05 vs. control ODN injected mice subjected to renal IR injury; error bars represent 1 SEM), the expression levels of TNF-a mRNA, MCP-1 mRNA,
MIP-2 mRNA, IL-6 mRNA, and KC mRNA were significantly attenuated upon treatment with the MNPs encapsulating the CpG-ODN TLR9 antagonist, as compared to those upon treatment with (i) the MNPs encapsulating the control ODN or (ii) the non-encapsulated ODN (e.g., naked CpG-ODN TLR9 antagonist).
TABLE 1. Primers used in quantitative reverse transcription polymerase chain reactions to amplify mouse cDNAs based on published GenBank sequences. Annealing temperatures used for each primer are also provided.
EMBODIMENTS
[0149] The following non-limiting embodiments provide illustrative examples of the invention, but do not limit the scope of the invention.
[0150] Embodiment 1. A composition comprising a plurality of polymeric nanoparticles, wherein: (i) a polymeric nanoparticle of the plurality of polymeric nanoparticles contains a nucleic acid molecule that binds to a polypeptide; and (ii) the plurality of polymeric nanoparticles
is characterize by an average diameter of no greater than about 1 micrometer, as ascertained by dynamic light scattering, optionally wherein:
(1) the average diameter is from about 100 nanometers (nm) to about 1000 nm;
(2) the average diameter is from about 100 nm to about 600 nm;
(3) the average diameter is from about 200 nm to about 500 nm; and/or
(4) the polymeric nanoparticle comprises a population of copolymers.
[0151] Embodiment 2. A composition comprising a plurality of polymeric nanoparticles, wherein: (i) a nanoparticle of the plurality of nanoparticles contains a nucleic acid molecule that binds to a polypeptide; and (ii) each nanoparticle of the plurality of nanoparticles independently comprises a polymeric structure, wherein the polymeric structure comprises a population of polymers, wherein the population of polymers has an average molar mass that is at least about 10 kilodaltons (kDa), optionally wherein:
(1) the average molar mass is at least about 20 kDa;
(2) the average molar mass is from about 10 kDa to about 100 kDa;
(3) the average molar mass is from about 30 kDa to about 70 kDa; and/or
(4) the population of polymers is a population of copolymers.
[0152] Embodiment 4. A composition comprising a plurality of polymeric nanoparticles, wherein: (i) a polymeric nanoparticle of the plurality of polymeric nanoparticles contains a medical agent; and (ii) the polymeric nanoparticle is coated with a surfactant, optionally wherein:
(1) the medical agent is a therapeutic agent, optionally wherein: the therapeutic agent is a nucleic acid molecule that binds to a target moiety, optionally wherein:
(A) the target moiety is a polypeptide, optionally wherein the polypeptide is a receptor polypeptide; or
(B) the target moiety is a polynucleotide, optionally wherein the polynucleotide is a messenger RNA;
(2) the surfactant is a non-ionic surfactant;
(3) the surfactant is a polymer;
(4) the surfactant is a polyoxyethylene-polyoxypropylene block polymer; and/or
(5) the polymeric nanoparticle comprises a population of polymers, optionally wherein the population of polymers comprises a population of copolymers.
[0153] Embodiment 5. A composition comprising a plurality of polymeric nanoparticles, wherein: (i) a nanoparticle of the plurality of nanoparticles contains a nucleic acid molecule that binds to a polypeptide; and (ii) if, in a controlled study, the nanoparticle is administered to a
subject, then the nanoparticle exhibits selective targeting to a targeted tissue of the subject preferentially over a non-targeted tissue of the subject, optionally wherein:
(1) in (ii), the nanoparticle is administered intravenously;
(2) in (ii), the nanoparticle is not administered subcutaneously;
(3) the targeted tissue is a kidney; and/or
(4) the nanoparticle comprises a population of copolymers.
[0154] Embodiment 6. The composition of any one of the preceding embodiments, wherein:
(a) the population of copolymers is a population of poly(lactic-co-glycolic acid)- poly(ethylene glycol);
(b) the nucleic acid molecule is a single-stranded nucleic acid molecule;
(c) the nucleic acid molecule is an oligodeoxynucleotide (ODN);
(d) the nucleic acid molecule is an unmethylated cytosine-phosphate-guanosine (CpG)
ODN;
(e) the nucleic acid molecule is an antagonist of the polypeptide;
(f) the polypeptide is a receptor polypeptide; and/or
(g) the polypeptide is a toll-like receptor (TLR).
[0155] Embodiment 7. A method of treating a condition, the method comprising administering to a subject in need thereof a therapeutically-effective amount of the composition of any one of the preceding embodiments.
[0156] Embodiment 8. A method comprising forming a plurality of polymeric nanoparticles, wherein: (i) a polymeric nanoparticle of the plurality of polymeric nanoparticles contains a nucleic acid molecule that binds to a polypeptide; and (ii) the plurality of polymeric nanoparticles is characterize by an average diameter of no greater than about 1 micrometer, as ascertained by dynamic light scattering, optionally wherein:
(1) the average diameter is between about 100 nm to about 1000 nm;
(2) the average diameter is between about 100 nm to about 600 nm;
(3) the average diameter is between about 200 nm to about 500 nm; and/or
(4) the polymeric nanoparticle comprises a population of copolymers.
[0157] Embodiment 9. A method comprising forming a plurality of polymeric nanoparticles, wherein: (i) a nanoparticle of the plurality of nanoparticles contains a nucleic acid molecule that binds to a polypeptide; and (ii) each nanoparticle of the plurality of nanoparticles independently comprises a polymeric structure, wherein the polymeric structure comprises a population of polymers, wherein the population of polymers has an average molar mass that is at least about 10 kilodaltons (kDa), optionally wherein:
(1) the average molar mass is at least about 20 kDa;
(2) the average molar mass is from about 10 kDa to about 100 kDa;
(3) the average molar mass is from about 30 kDa to about 70 kDa; and/or
(4) the population of polymers is a population of copolymers.
[0158] Embodiment 10. A method comprising forming a plurality of polymeric nanoparticles, wherein: (i) a polymeric nanoparticle of the plurality of polymeric nanoparticles contains a medical agent; and (ii) the polymeric nanoparticle coated with a surfactant, optionally wherein:
(1) the medical agent is a therapeutic agent, optionally wherein: the therapeutic agent is a nucleic acid molecule that binds to a target moiety, optionally wherein:
(A) the target moiety is a polypeptide, optionally wherein the polypeptide is a receptor polypeptide; or
(B) the target moiety is a polynucleotide, optionally wherein the polynucleotide is a messenger RNA;
(2) the surfactant is a non-ionic surfactant;
(3) the surfactant is a polymer;
(4) the surfactant is a polyoxyethylene-polyoxypropylene block polymer; and/or
(5) the polymeric nanoparticle comprises a population of polymers, optionally wherein the population of polymers comprises a population of copolymers.
[0159] Embodiment 11. A method comprising forming a plurality of polymeric nanoparticles, wherein: (i) a polymeric nanoparticle of the plurality of polymeric nanoparticles contains a medical agent; and (ii) upon the contacting, the polymeric nanoparticle is coated with the surfactant, optionally wherein:
(1) the medical agent is a therapeutic agent, optionally wherein: the therapeutic agent is a nucleic acid molecule that binds to a target moiety, optionally wherein:
(A) the target moiety is a polypeptide, optionally wherein the polypeptide is a receptor polypeptide; or
(B) the target moiety is a polynucleotide, optionally wherein the polynucleotide is a messenger RNA;
(2) the surfactant is a non-ionic surfactant;
(3) the surfactant is a polymer;
(4) the surfactant is a polyoxyethylene-polyoxypropylene block polymer; and/or
(5) the polymeric nanoparticle comprises a population of polymers, optionally wherein
the population of polymers comprises a population of copolymers.
[0160] Embodiment 12. A method comprising forming a plurality of polymeric nanoparticles, wherein: (i) a nanoparticle of the plurality of nanoparticles contains a nucleic acid molecule that binds to a polypeptide; and (ii) if, in a controlled study, the nanoparticle is administered to a subject, then the nanoparticle exhibits selective targeting to a targeted tissue of the subject preferentially over a non-targeted tissue of the subject, optionally wherein:
(1) in (ii), the nanoparticle is administered intravenously;
(2) in (ii), the nanoparticle is not administered subcutaneously;
(3) the targeted tissue is a kidney; and/or
(4) the nanoparticle comprises a population of copolymers.
[0161] Embodiment 13. The method of any one of the preceding embodiments, wherein:
(a) the forming comprises mixing (1) the population of copolymers and (2) a population of nucleic acid molecules comprising the nucleic acid molecule, optionally wherein the population of copolymers is in a first solution and the population of nucleic acid molecules is in a second solution, wherein the first solution and the second solution are different, and optionally wherein one of the first solution and the second solution is an organic solvent, and the other of the first solution and the second solution is an aqueous solution;
(b) wherein the population of copolymers is a population of poly(lactic-co-glycolic acid)- poly(ethylene glycol);
(c) the nucleic acid molecule is a single-stranded nucleic acid molecule;
(d) the nucleic acid molecule is an oligodeoxynucleotide (ODN);
(e) the nucleic acid molecule is an unmethylated cytosine-phosphate-guanosine (CpG)
ODN;
(f) the nucleic acid molecule is an antagonist of the polypeptide;
(g) the polypeptide is a receptor polypeptide; and/or
(h) the polypeptide is a toll-like receptor (TLR).
[0162] Embodiment 14. A composition comprising a plurality of polymeric nanoparticles, wherein: (i) a polymeric nanoparticle of the plurality of polymeric nanoparticles contains a deoxyribonucleic acid molecule that binds to a target moiety; and (ii) the plurality of polymeric nanoparticles is characterized by an average diameter of no greater than about 1 micrometer, as ascertained by dynamic light scattering, optionally wherein:
(1) the average diameter is from about 100 nm to about 1000 nm.;
(2) the average diameter is from about 100 nm to about 600 nm.;
(3) the average diameter is from about 200 nm to about 500 nm; and/or
(4) the polymeric nanoparticle comprises a population of copolymers.
[0163] Embodiment 15. A composition comprising a plurality of nanoparticles, wherein: (i) a nanoparticle of the plurality of nanoparticles contains a deoxyribonucleic acid molecule that binds to a target moiety; and (ii) each nanoparticle of the plurality of nanoparticles independently comprises a polymeric structure, wherein the polymeric structure comprises a population of polymers, wherein the population of polymers has an average molar mass that is at least about 10 kilodaltons (kDa), optionally wherein:
(1) the average molar mass is at least about 20 kDa;
(2) the average molar mass is from about 10 kDa to about 100 kDa;
(3) the average molar mass is from about 30 kDa to about 70 kDa; and/or
(4) the population of polymers is a population of copolymers.
[0164] Embodiment 16. A composition comprising a plurality of nanoparticles, wherein: (i) a nanoparticle of the plurality of nanoparticles contains a deoxyribonucleic acid molecule that binds to a target moiety; and (ii) if, in a controlled study, the nanoparticle is administered to a subject, then the nanoparticle exhibits selective targeting to a targeted tissue of the subject preferentially over a non-targeted tissue of the subject, optionally wherein:
(1) in (ii), the nanoparticle is administered intravenously;
(2) in (ii), the nanoparticle is not administered subcutaneously;
(3) the targeted tissue is a kidney; and/or
(4) the nanoparticle comprises a population of copolymers.
[0165] Embodiment 17. The composition of any one of the Embodiments 14-16, wherein:
(a) the population of copolymers is a population of poly(lactic-co-glycolic acid)- poly(ethylene glycol);
(b) the deoxyribonucleic acid molecule is an antisense oligonucleotide;
(c) the deoxyribonucleic acid molecule is a cytosine-phosphate-guanosine oligodeoxynucleotide (CpG-ODN);
(d) the target moiety is a polynucleotide; and/or
(e) the target moiety is a messenger ribonucleic acid.
[0166] Embodiment 18. A method of treating a condition, the method comprising administering to a subject in need thereof a therapeutically-effective amount of the composition of any one of Embodiments 14-17.
[0167] Embodiment 19. A method comprising forming a plurality of polymeric nanoparticles, wherein: (i) a polymeric nanoparticle of the plurality of polymeric nanoparticles contains a deoxyribonucleic acid molecule that binds to a target moiety; and (ii) the plurality of polymeric
nanoparticles is characterized by an average diameter of no greater than about 1 micrometer, as ascertained by dynamic light scattering, optionally wherein:
(1) the average diameter is from about 100 nm to about 1000 nm.;
(2) the average diameter is from about 100 nm to about 600 nm.;
(3) the average diameter is from about 200 nm to about 500 nm; and/or
(4) the polymeric nanoparticle comprises a population of copolymers.
[0168] Embodiment 20. A method comprising forming a plurality of nanoparticles, wherein: (i) a nanoparticle of the plurality of nanoparticles contains a deoxyribonucleic acid molecule that binds to a target moiety; and (ii) each nanoparticle of the plurality of nanoparticles independently comprises a polymeric structure, wherein the polymeric structure comprises a population of polymers, wherein the population of polymers has an average molar mass that is at least about 10 kilodaltons (kDa), optionally wherein:
(1) the average molar mass is at least about 20 kDa;
(2) the average molar mass is from about 10 kDa to about 100 kDa;
(3) the average molar mass is from about 30 kDa to about 70 kDa; and/or
(4) the population of polymers is a population of copolymers.
[0169] Embodiment 21. A method comprising forming a plurality of polymeric nanoparticles, wherein: (i) a nanoparticle of the plurality of nanoparticles contains a deoxyribonucleic acid molecule that binds to a target moiety; and (ii) if, in a controlled study, the nanoparticle is administered to a subject, then the nanoparticle exhibits selective targeting to a targeted tissue of the subject preferentially over a non-targeted tissue of the subject, optionally wherein:
(1) in (ii), the nanoparticle is administered intravenously;
(2) in (ii), the nanoparticle is not administered subcutaneously;
(3) the targeted tissue is a kidney; and/or
(4) the nanoparticle comprises a population of copolymers.
[0170] Embodiment 22. The method of any one Embodiments 19-21, wherein:
(a) the forming comprises mixing (1) the population of copolymers and (2) a population of nucleic acid molecules comprising the nucleic acid molecule, optionally wherein the population of copolymers is in a first solution and the population of nucleic acid molecules is in a second solution, wherein the first solution and the second solution are different, and optionally wherein one of the first solution and the second solution is an organic solvent, and the other of the first solution and the second solution is an aqueous solution;
(b) wherein the population of copolymers is a population of poly(lactic-co-glycolic acid)- poly(ethylene glycol);
(c) the deoxyribonucleic acid molecule is an antisense oligonucleotide;
(d) the deoxyribonucleic acid molecule is a cytosine-phosphate-guanosine oligodeoxynucleotide (CpG-ODN);
(e) the target moiety is a polynucleotide;
(f) the target moiety is a messenger ribonucleic acid;
[0171] Each aspect and embodiment described in this disclosure can also have included or incorporated therewith such variations or aspects as disclosed in regard to any or all of the other aspects and embodiments.
[0172] The present technology is also not to be limited in terms of the particular aspects described herein, which are intended as single illustrations of individual aspects of the present technology. Many modifications and variations of this present technology can be made without departing from its spirit and scope, as will be apparent to those skilled in the art. Functionally equivalent methods within the scope of the present technology, in addition to those enumerated herein, will be apparent to those skilled in the art from the foregoing descriptions. Such modifications and variations are intended to fall within the scope of the appended claims. It is to be understood that this present technology is not limited to particular methods, reagents, compounds, compositions, labeled compounds or biological systems, which can, of course, vary. It is also to be understood that the terminology used herein is for the purpose of describing particular aspects only, and is not intended to be limiting.
[0173] The embodiments, illustratively described herein may suitably be practiced in the absence of any element or elements, limitation or limitations, not specifically disclosed herein. Thus, for example, the terms “comprising,” “including,” “containing,” etc. shall be read expansively and without limitation. Additionally, the terms and expressions employed herein have been used as terms of description and not of limitation, and there is no intention in the use of such terms and expressions of excluding any equivalents of the features shown and described or portions thereof, but it is recognized that various modifications are possible within the scope of the claimed technology. Additionally, the phrase “consisting essentially of’ will be understood to include those elements specifically recited and those additional elements that do not materially affect the basic and novel characteristics of the claimed technology. The phrase “consisting of’ excludes any element not specified.
[0174] In addition, where features or aspects of the disclosure are described in terms of Markush groups, those skilled in the art will recognize that the disclosure is also thereby described in terms of any individual member or subgroup of members of the Markush group. Each of the
narrower species and subgeneric groupings falling within the generic disclosure also form part of the invention. This includes the generic description of the invention with a proviso or negative limitation removing any subject matter from the genus, regardless of whether or not the excised material is specifically recited herein.
Claims
1. A composition comprising a plurality of polymeric nanoparticles, wherein:
(i) a polymeric nanoparticle of the plurality of polymeric nanoparticles contains a nucleic acid molecule that binds to a polypeptide; and
(ii) the plurality of polymeric nanoparticles is characterized by an average diameter of no greater than about 1 micrometer, as ascertained by dynamic light scattering.
2. The composition of claim 1, wherein the polypeptide is a receptor polypeptide.
3. The composition of claim 1, wherein the average diameter is from about 100 nm to about 1000 nm.
4. The composition of claim 1, wherein the average diameter is from about 100 nm to about 600 nm.
5. The composition of claim 1, wherein the average diameter is from about 200 nm to about 500 nm.
6. The composition of claim 1, wherein the polymeric nanoparticle comprises a population of copolymers.
7. A composition comprising a plurality of nanoparticles, wherein:
(i) a nanoparticle of the plurality of nanoparticles contains a nucleic acid molecule that binds to a polypeptide; and
(ii) each nanoparticle of the plurality of nanoparticles independently comprises a polymeric structure, wherein the polymeric structure comprises a population of polymers, wherein the population of polymers has an average molar mass that is at least about 10 kilodaltons (kDa).
8. The composition of claim 7, wherein the polypeptide is a receptor polypeptide.
9. The composition of claim 7, wherein the average molar mass is at least about 20 kDa.
10. The composition of claim 7, wherein the average molar mass is from about 10 kDa to about 100 kDa.
11. The composition of claim 7, wherein the average molar mass is from about 30 kDa to about 70 kDa.
12. The composition of claim 7, wherein the population of polymers is a population of copolymers.
13. A composition comprising a plurality of polymeric nanoparticles, wherein:
(i) a polymeric nanoparticle of the plurality of polymeric nanoparticles contains a medical agent; and
(ii) the polymeric nanoparticle is coated with a surfactant.
14. The composition of claim 13, wherein the medical agent is a therapeutic agent.
15. The composition of claim 14, wherein the therapeutic agent is a nucleic acid molecule that binds to a target moiety.
16. The composition of claim 15, wherein the target moiety is a polypeptide.
17. The composition of claim 16, wherein the polypeptide is a receptor polypeptide.
18. The composition of claim 15, wherein the target moiety is a polynucleotide.
19. The composition of claim 18, wherein the polynucleotide is a messenger RNA.
20. The composition of any one of claims 13-19, wherein the surfactant is a non-ionic surfactant.
21. The composition of any one of claims 13-19, wherein the surfactant is a polymer.
22. The composition of any one of claims 13-19, wherein the surfactant is a polyoxyethylene- polyoxypropylene block polymer.
23. The composition of any one of claims 13-19, wherein the polymeric nanoparticle comprises a population of polymers.
24. The composition of claim 23, wherein the population of polymers comprises a population of copolymers.
25. A composition comprising a plurality of nanoparticles, wherein:
(i) a nanoparticle of the plurality of nanoparticles contains a nucleic acid molecule that binds to a polypeptide; and
(ii) if, in a controlled study, the nanoparticle is administered to a subject, then the nanoparticle exhibits selective targeting to a targeted tissue of the subject preferentially over a non-targeted tissue of the subject.
26. The composition of claim 25, wherein the polypeptide is a receptor polypeptide.
27. The composition of claim 25, wherein, in (ii), the nanoparticle is administered intravenously.
28. The composition of claim 25, wherein, in (ii), the nanoparticle is not administered subcutaneously.
29. The composition of claim 25, wherein the targeted tissue is a kidney.
30. The composition of claim 25, wherein the nanoparticle comprises a population of copolymers.
31. The composition of any one of claims 6, 12, 24, and 30, wherein the population of copolymers is a population of poly(lactic-co-glycolic acid)-poly(ethylene glycol).
32. The composition of any one of claims 1-12, 15-19, and 25-30, wherein the nucleic acid molecule is a single-stranded nucleic acid molecule.
33. The composition of any one of claims 1-12, 15-19, and 25-30, wherein the nucleic acid molecule is an oligodeoxynucleotide (ODN).
34. The composition of claim 33, wherein the nucleic acid molecule is an unmethylated cytosine-phosphate-guanosine (CpG) ODN.
35. The composition of any one of claims 1-12, 16, 17, and 25-30, wherein the nucleic acid molecule is an antagonist of the polypeptide.
36. The composition of any one of claims 1-12, 16, 17, and 25-30, wherein the polypeptide is a toll-like receptor (TLR).
37. A method of treating a condition, the method comprising administering to a subject in need thereof a therapeutically-effective amount of the composition of any one of the preceding claims.
38. A method comprising forming a plurality of polymeric nanoparticles, wherein:
(i) a polymeric nanoparticle of the plurality of polymeric nanoparticles contains a nucleic acid molecule that binds to a polypeptide; and
(ii) the plurality of polymeric nanoparticles is characterized by an average diameter of no greater than about 1 micrometer, as ascertained by dynamic light scattering.
39. The method of claim 38, wherein the average diameter is from about 100 nanometers (nm) to about 1000 nm
40. The method of claim 38, wherein the average diameter is from about 100 nanometers (nm) to about 600 nm.
41. The method of claim 38, wherein the average diameter is from about 200 nm to about 500 nm.
42. The method of claim 38, wherein the polymeric nanoparticle comprises a population of copolymers.
43. A method comprising forming a plurality of nanoparticles, wherein:
(i) a nanoparticle of the plurality of nanoparticles contains a nucleic acid molecule that binds to a polypeptide; and
(ii) each nanoparticle of the plurality of nanoparticles independently comprises a polymeric structure, wherein the polymeric structure comprises a population of polymers, wherein the population of polymers has an average molar mass that is at least about 10 kilodaltons (kDa).
44. The method of claim 43, wherein the average molar mass is at least about 20 kDa.
45. The method of claim 43, wherein the average molar mass is from about 10 kDa to about 100 kDa.
46. The method of claim 43, wherein the average molar mass is from about 30 kDa to about 70 kDa.
47. The method of claim 43, wherein the population of polymers is a population of copolymers.
48. A method comprising forming a plurality of polymeric nanoparticles, wherein:
(i) a polymeric nanoparticle of the plurality of polymeric nanoparticles contains a medical agent; and
(ii) the polymeric nanoparticle coated with a surfactant.
49. A method comprising contacting a plurality of polymeric nanoparticles with a surfactant, wherein:
(i) a polymeric nanoparticle of the plurality of polymeric nanoparticles contains a medical agent; and
(ii) upon the contacting, the polymeric nanoparticle is coated with the surfactant.
50. The method of claim 48 or 49, wherein the medical agent is a therapeutic agent.
51. The method of claim 50, wherein the therapeutic agent is a nucleic acid molecule that binds to a target moiety.
52. The method of claim 51, wherein the target moiety is a polypeptide.
53. The method of claim 52, wherein the polypeptide is a receptor polypeptide.
54. The method of claim 51, wherein the target moiety is a polynucleotide.
55. The method of claim 54, wherein the polynucleotide is a messenger RNA.
56. The method of any one of claims 48-55, wherein the surfactant is a non-ionic surfactant.
57. The method of any one of claims 48-55, wherein the surfactant is a polymer.
58. The method of any one of claims 48-55, wherein the surfactant is a polyoxyethylene- polyoxypropylene block polymer.
59. The method of any one of claims 48-58, wherein the polymeric nanoparticle comprises a population of polymers.
60. The method of claim 59, wherein the population of polymers comprises a population of copolymers.
61. A method comprising forming a plurality of polymeric nanoparticles, wherein:
(i) a nanoparticle of the plurality of nanoparticles contains a nucleic acid molecule that binds to a polypeptide; and
(ii) if, in a controlled study, the nanoparticle is administered to a subject, then the nanoparticle exhibits selective targeting to a targeted tissue of the subject preferentially over a non-targeted tissue of the subject.
62. The method of claim 61, wherein, in (ii), the nanoparticle is administered intravenously.
63. The method of claim 61, wherein, in (ii), the nanoparticle is not administered subcutaneously.
64. The method of claim 61, wherein the targeted tissue is a kidney.
65. The method of claim 61, wherein the nanoparticle comprises a population of copolymers.
66. The method of any one of claims 42, 47, 60, and 65, wherein the forming comprises mixing (1) the population of copolymers and (2) a population of nucleic acid molecules comprising the nucleic acid molecule.
67. The method of claim 66, wherein the population of copolymers is in a first solution and the population of nucleic acid molecules is in a second solution, wherein the first solution and the second solution are different.
68. The method of claim 67, wherein one of the first solution and the second solution is an organic solvent, and the other of the first solution and the second solution is an aqueous solution.
69. The method of any one of claims 42, 47, 60, and 65, wherein the population of copolymers is a population of poly(lactic-co-glycolic acid)-poly(ethylene glycol).
70. The method of any one of claims 38-47, 51-55, and 61-65, wherein the nucleic acid molecule is a single-stranded nucleic acid molecule.
71. The method of any one of claims 38-47, 51-55, and 61-65, wherein the nucleic acid molecule is an oligodeoxynucleotide (ODN).
72. The method of any one of claims 38-47, 51-55, and 61-65, wherein the nucleic acid molecule is an unmethylated cytosine-phosphate-guanosine (CpG) ODN.
73. The method of any one of claims 38-47, 51-55, and 61-65, wherein the nucleic acid molecule is an antagonist of the polypeptide.
74. The method of any one of claims 38-47, 52, 53, and 61-65, wherein the polypeptide is a toll-like receptor (TLR).
75. A composition comprising a plurality of polymeric nanoparticles, wherein:
(i) a polymeric nanoparticle of the plurality of polymeric nanoparticles contains a deoxyribonucleic acid molecule that binds to a target moiety; and
(ii) the plurality of polymeric nanoparticles is characterized by an average diameter of no greater than about 1 micrometer, as ascertained by dynamic light scattering.
76. The composition of claim 75, wherein the average diameter is from about 100 nm to about 1000 nm.
77. The composition of claim 75, wherein the average diameter is from about 100 nm to about 600 nm.
78. The composition of claim 75, wherein the average diameter is from about 200 nm to about 500 nm.
79. The composition of claim 75, wherein the polymeric nanoparticle comprises a population of copolymers.
80. A composition comprising a plurality of nanoparticles, wherein:
(i) a nanoparticle of the plurality of nanoparticles contains a deoxyribonucleic acid molecule that binds to a target moiety; and
(ii) each nanoparticle of the plurality of nanoparticles independently comprises a polymeric structure, wherein the polymeric structure comprises a population of polymers, wherein the population of polymers has an average molar mass that is at least about 10 kilodaltons (kDa).
81. The composition of claim 80, wherein the average molar mass is at least about 20 kDa.
82. The composition of claim 80, wherein the average molar mass is from about 10 kDa to about 100 kDa.
83. The composition of claim 80, wherein the average molar mass is from about 30 kDa to about 70 kDa.
84. The composition of claim 80, wherein the population of polymers is a population of copolymers.
85. A composition comprising a plurality of nanoparticles, wherein:
(i) a nanoparticle of the plurality of nanoparticles contains a deoxyribonucleic acid molecule that binds to a target moiety; and
(ii) if, in a controlled study, the nanoparticle is administered to a subject, then the nanoparticle exhibits selective targeting to a targeted tissue of the subject preferentially over a non-targeted tissue of the subject.
86. The composition of claim 85, wherein, in (ii), the nanoparticle is administered intravenously.
87. The composition of claim 85, wherein, in (ii), the nanoparticle is not administered subcutaneously.
88. The composition of claim 85, wherein the targeted tissue is a kidney.
89. The composition of claim 85, wherein the nanoparticle comprises a population of copolymers.
90. The composition of any one of claims 79, 84, and 89, wherein the population of copolymers is a population of poly(lactic-co-glycolic acid)-poly(ethylene glycol).
91. The composition of any one of claims 75-90, wherein the deoxyribonucleic acid molecule is an antisense oligonucleotide.
92. The composition of any one of claims 75-91, wherein the deoxyribonucleic acid molecule is a cytosine-phosphate-guanosine oligodeoxynucleotide (CpG-ODN).
93. The composition of any one of claims 75-92, wherein the target moiety is a polynucleotide.
94. The composition of any one of claims 75-93, wherein the target moiety is a messenger ribonucleic acid.
95. A method of treating a condition, the method comprising administering to a subject in need thereof a therapeutically-effective amount of the composition of any one of claims 75-94.
96. A method comprising forming a plurality of polymeric nanoparticles, wherein:
(i) a polymeric nanoparticle of the plurality of polymeric nanoparticles contains a deoxyribonucleic acid molecule that binds to a target moiety; and
(ii) the plurality of polymeric nanoparticles is characterized by an average diameter of no greater than about 1 micrometer, as ascertained by dynamic light scattering.
97. The method of claim 96, wherein the average diameter is from about 100 nanometers (nm) to about 1000 nm
98. The method of claim 96, wherein the average diameter is from about 100 nanometers (nm) to about 600 nm.
99. The method of claim 96, wherein the average diameter is from about 200 nm to about 500 nm.
100. The method of claim 96, wherein the polymeric nanoparticle comprises a population of copolymers.
101. A method comprising forming a plurality of nanoparticles, wherein:
(i) a nanoparticle of the plurality of nanoparticles contains a deoxyribonucleic acid molecule that binds to a target moiety; and
(ii) each nanoparticle of the plurality of nanoparticles independently comprises a polymeric structure, wherein the polymeric structure comprises a population of polymers, wherein the population of polymers has an average molar mass that is at least about 10 kilodaltons (kDa).
102. The method of claim 101, wherein the average molar mass is at least about 20 kDa.
103. The method of claim 101, wherein the average molar mass is from about 10 kDa to about 100 kDa.
104. The method of claim 101, wherein the average molar mass is from about 30 kDa to about 70 kDa.
105. The method of claim 101, wherein the population of polymers is a population of copolymers.
106. A method comprising forming a plurality of polymeric nanoparticles, wherein:
(i) a nanoparticle of the plurality of nanoparticles contains a deoxyribonucleic acid molecule that binds to a target moiety; and
(ii) if, in a controlled study, the nanoparticle is administered to a subject, then the nanoparticle exhibits selective targeting to a targeted tissue of the subject preferentially over a non-targeted tissue of the subject.
107. The method of claim 106, wherein, in (ii), the nanoparticle is administered intravenously.
108. The method of claim 106, wherein, in (ii), the nanoparticle is not administered subcutaneously.
109. The method of claim 106, wherein the targeted tissue is a kidney.
110. The method of claim 106, wherein the nanoparticle comprises a population of copolymers.
111. The method of any one of claims 100, 105, and 110, wherein the forming comprises mixing (1) the population of copolymers and (2) a population of nucleic acid molecules comprising the nucleic acid molecule.
112. The method of claim 111, wherein the population of copolymers is in a first solution and the population of nucleic acid molecules is in a second solution, wherein the first solution and the second solution are different.
113. The method of claim 112, wherein one of the first solution and the second solution is an organic solvent, and the other of the first solution and the second solution is an aqueous solution.
114. The method of any one of claims 100, 105, and 110-113, wherein the population of copolymers is a population of poly(lactic-co-glycolic acid)-poly(ethylene glycol).
115. The method of any one of claims 96-114, wherein the deoxyribonucleic acid molecule is an antisense oligonucleotide.
116. The method of any one of claims 96-115, wherein the deoxyribonucleic acid molecule is a cytosine-phosphate-guanosine oligodeoxynucleotide (CpG-ODN).
117. The method of any one of claims 96-116, wherein the target moiety is a polynucleotide.
118. The method of any one of claims 96-117, wherein the target moiety is a messenger ribonucleic acid.
Applications Claiming Priority (2)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US202062969528P | 2020-02-03 | 2020-02-03 | |
US62/969,528 | 2020-02-03 |
Publications (1)
Publication Number | Publication Date |
---|---|
WO2021158627A1 true WO2021158627A1 (en) | 2021-08-12 |
Family
ID=77200528
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/US2021/016368 WO2021158627A1 (en) | 2020-02-03 | 2021-02-03 | Compositions comprising nanoparticles and methods thereof |
Country Status (1)
Country | Link |
---|---|
WO (1) | WO2021158627A1 (en) |
Citations (4)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US20060153923A1 (en) * | 2003-07-04 | 2006-07-13 | Fernandez Maria Jose A | Nanoparticles of polypoxyethylenated derivatives |
US20110014473A1 (en) * | 2007-01-31 | 2011-01-20 | Ying Jackie Y | Polymer-coated nanoparticles |
US20140220143A1 (en) * | 2011-07-15 | 2014-08-07 | Shanta Dhar | Immune-stimulating photoactive hybrid nanoparticles |
US20180243227A1 (en) * | 2015-03-20 | 2018-08-30 | Memorial Sloan Kettering Cancer Center | Mesoscale nanoparticles for selective targeting to the kidney and methods of their therapeutic use |
-
2021
- 2021-02-03 WO PCT/US2021/016368 patent/WO2021158627A1/en active Application Filing
Patent Citations (4)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US20060153923A1 (en) * | 2003-07-04 | 2006-07-13 | Fernandez Maria Jose A | Nanoparticles of polypoxyethylenated derivatives |
US20110014473A1 (en) * | 2007-01-31 | 2011-01-20 | Ying Jackie Y | Polymer-coated nanoparticles |
US20140220143A1 (en) * | 2011-07-15 | 2014-08-07 | Shanta Dhar | Immune-stimulating photoactive hybrid nanoparticles |
US20180243227A1 (en) * | 2015-03-20 | 2018-08-30 | Memorial Sloan Kettering Cancer Center | Mesoscale nanoparticles for selective targeting to the kidney and methods of their therapeutic use |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
CN102666879B (en) | Templated nanometer conjugate | |
US20220175953A1 (en) | Biomimetic anisotropic polymeric particles with naturally derived cell membranes for enhanced drug delivery | |
CN107427466B (en) | Nanobubsomes derived from cell membranes and uses thereof | |
US20040023855A1 (en) | Biologic modulations with nanoparticles | |
US20040038303A1 (en) | Biologic modulations with nanoparticles | |
Yang et al. | Enhanced nose-to-brain delivery of siRNA using hyaluronan-enveloped nanomicelles for glioma therapy | |
EP2029119A2 (en) | Functionalized solid polymer nanoparticles for diagnostic and therapeutic applications | |
ES2745528T3 (en) | Magnetic nanoparticles functionalized with catechol, production and use thereof | |
CN104023749A (en) | Nanoconjugates able to cross the blood-brain barrier | |
US20140242180A1 (en) | Nanoparticles for controlling bleeding and drug delivery | |
US20130011333A1 (en) | Methods and Compositions for Nanoparticle-Mediated Cancer Cell-Targeted Delivery | |
US10695427B2 (en) | Shape memory particles for biomedical uses | |
EP2790739A1 (en) | Nanoparticles and uses thereof | |
Shi et al. | The delivery of thrombi-specific nanoparticles incorporating oligonucleotides into injured cerebrovascular endothelium | |
WO2021158627A1 (en) | Compositions comprising nanoparticles and methods thereof | |
KR101755680B1 (en) | Polysaccharidic nanogel for protein drug delivery and preparation method thereof | |
DE60208400T2 (en) | THERAPEUTICALLY USE TRIETHYLENE GLYCOL CHOLESTERYL OLIGONUCLEOTIDES | |
EP4410825A1 (en) | Fragments of the n-terminal domain of gsdmb for the treatment of cancer | |
US9782494B2 (en) | Methods of using multilayer magnetic micelle compositions | |
Haque et al. | Nanomedicines for brain targeting: A patent review | |
Kommineni et al. | Special Issue “Latest Advances in Nanomedicine Strategies for Different Diseases” | |
Yan et al. | SYNTHESIS AND VERIFICATION OF A NOVEL ATTAINABLE TUMOR TARGETING MAGNETIC NANOPARTICLE. | |
Yang et al. | Paclitaxel Loaded Hyaluronic Acid Polymerized Nanoparticles Designed for Ovarian Cancer Therapy | |
Sorge | Targeting the CD44 Receptor by Conjugated Polymer Nanoparticles in Glioma: Exploring Functional and Molecular Mechanism of HA-CPNs | |
CN118450890A (en) | Treatment of cancer and autoimmune disorders using nano-polymers of histone deacetylase inhibitors |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
121 | Ep: the epo has been informed by wipo that ep was designated in this application |
Ref document number: 21750022 Country of ref document: EP Kind code of ref document: A1 |
|
NENP | Non-entry into the national phase |
Ref country code: DE |
|
122 | Ep: pct application non-entry in european phase |
Ref document number: 21750022 Country of ref document: EP Kind code of ref document: A1 |