WO2022066636A1 - Nanoparticules et procédés d'utilisation associés - Google Patents

Nanoparticules et procédés d'utilisation associés Download PDF

Info

Publication number
WO2022066636A1
WO2022066636A1 PCT/US2021/051299 US2021051299W WO2022066636A1 WO 2022066636 A1 WO2022066636 A1 WO 2022066636A1 US 2021051299 W US2021051299 W US 2021051299W WO 2022066636 A1 WO2022066636 A1 WO 2022066636A1
Authority
WO
WIPO (PCT)
Prior art keywords
fold
composition
mixture
nanoparticle
medical agent
Prior art date
Application number
PCT/US2021/051299
Other languages
English (en)
Inventor
Greg Troiano
Chintan Hareshbhai KAPADIA
Ryan Martin WILLIAMS
Daniel Heller
Original Assignee
Goldilocks Therapeutics, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Goldilocks Therapeutics, Inc. filed Critical Goldilocks Therapeutics, Inc.
Publication of WO2022066636A1 publication Critical patent/WO2022066636A1/fr

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/14Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles
    • A61K9/19Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles lyophilised, i.e. freeze-dried, solutions or dispersions
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/4151,2-Diazoles
    • A61K31/41521,2-Diazoles having oxo groups directly attached to the heterocyclic ring, e.g. antipyrine, phenylbutazone, sulfinpyrazone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/4353Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/436Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems the heterocyclic ring system containing a six-membered ring having oxygen as a ring hetero atom, e.g. rapamycin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/4353Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/4375Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems the heterocyclic ring system containing a six-membered ring having nitrogen as a ring heteroatom, e.g. quinolizines, naphthyridines, berberine, vincamine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • A61K31/713Double-stranded nucleic acids or oligonucleotides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/50Microcapsules 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
    • A61K9/51Nanocapsules; Nanoparticles
    • A61K9/5107Excipients; Inactive ingredients
    • A61K9/513Organic macromolecular compounds; Dendrimers
    • A61K9/5161Polysaccharides, e.g. alginate, chitosan, cellulose derivatives; Cyclodextrin

Definitions

  • Medical agents can be administered to a subject for, e.g., diagnostics or therapeutic purposes.
  • therapeutic agents e.g., small molecules, DNA, siRNA, mRNA, plasmid DNA, miRNA, proteins, or antibodies
  • imaging agents e.g., dyes
  • imaging agents 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.
  • the present disclosure provides a method of manufacturing a nanoparticle composition, comprising: contacting a population of nanoparticles with a solution, wherein the solution comprises a polysaccharide, wherein the polysaccharide is present in the solution in an amount greater than 2 % by mass of the solution.
  • the polysaccharide is present in the solution in an amount of at least about 5 % by mass of the solution.
  • the polysaccharide is sucrose.
  • the polysaccharide is trehalose.
  • a nanoparticle of the population of nanoparticles comprises a therapeutic agent.
  • the therapeutic agent comprises a small molecule drug.
  • the method further comprises lyophilizing a mixture of (i) the population of nanoparticles and (ii) the solution.
  • the present disclosure provides a method of manufacturing a nanoparticle composition, the method comprising: (a) contacting a plurality of nanoparticles with a mixture, thereby forming the nanoparticle composition, wherein a medical agent is encapsulated by a nanoparticle of the plurality of nanoparticles, and wherein the mixture comprises a polysaccharide, wherein the polysaccharide is present in the mixture in an amount that is greater than 2 % and lesser than 10 % by mass of the mixture; and (b) reducing an average temperature of the nanoparticle composition.
  • the present disclosure provides a method of manufacturing a nanoparticle composition, the method comprising: contacting a plurality of nanoparticles with a mixture, thereby forming the nanoparticle composition, wherein a nanoparticle of the plurality of nanoparticles comprises a medical agent, and wherein the mixture comprises a first polysaccharide and a second polysaccharide.
  • the present disclosure provides a method of manufacturing a plurality of nanoparticles, the method comprising: (a) incubating a mixture for at least about 1 hour, wherein the mixture comprises (i) a medical agent, (ii) an excipient that is oppositely charged from the medical agent, and (iii) an encapsulation agent; (b) subsequent to (a), forming, from the mixture, the plurality of nanoparticles.
  • the present disclosure provides a method of manufacturing a nanoparticle composition, the method comprising: using a mixture to form a plurality of nanoparticles, wherein the mixture comprises (i) a medical agent that is not a nucleic acid molecule, (ii) an encapsulation agent, and (iii) an additional excipient, and wherein a nanoparticle of the plurality of nanoparticles comprises the medical agent, the additional excipient, and the encapsulation agent.
  • the present disclosure provides a method of manufacturing a plurality of nanoparticles, comprising: forming the plurality of nanoparticles, wherein a nanoparticle of the plurality of nanoparticles comprises a compound, wherein the compound is MK2206 or a pharmaceutically acceptable salt thereof.
  • the present disclosure provides a method of manufacturing a plurality of nanoparticles, comprising: using a mixture to form the plurality of nanoparticles, wherein the mixture comprises (i) a compound, wherein the compound is Edaravone or a pharmaceutically acceptable salt thereof and (ii) a protic solvent, wherein a nanoparticle of the plurality of nanoparticles comprises the compound.
  • FIGURES 1 and 2 show the effect of dehydrating agents to a change in an average diameter of a population of nanoparticles upon lyophilization.
  • FIGURE 3 shows a standard curve of rapamycin concentration in saline as a function of high-performance liquid chromatography (HPLC) peak area.
  • FIGURE 4 schematically illustrates a timeline of in vivo efficacy study using a rat model of polycystic kidney disease (PKD).
  • PPD polycystic kidney disease
  • FIGURE 5 illustrates an in vitro process of treating cells with siRNA-excipient loaded particles.
  • FIGURE 6 illustrates an in vivo process of treating a subject (e.g., an animal) with siRNA-excipient loaded particles.
  • FIGURE 7 schematically illustrates a rotor-stator configuration.
  • FIGURE 8 shows chemical structure of MK2206.
  • FIGURE 9 shows chemical structure of Oleic acid.
  • compositions and methods of manufacturing and using the same for particles for example, nanoparticles, such as mesoscale nanoparticles (MNPs), for delivering medical agents, for example, therapeutic agents or imaging agents.
  • MNPs mesoscale nanoparticles
  • the compositions of interest can be administered to a subject to deliver the medical agents for disease therapy or diagnosis.
  • the invention provides a composition comprising a population of particles.
  • a particle of the population of particles comprises a medical agent.
  • the population of particles comprises a population of nanoparticles.
  • the population of nanoparticles comprises a population of mesoscale nanoparticles.
  • 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.
  • the population of nanoparticles comprising (e.g., encapsulating) the medical agent is administered to a subject in need thereof.
  • 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.
  • a control medical agent e.g., a non-encapsulated medical agent, encapsulated by a different population of nanoparticles, etc.
  • the population of nanoparticles 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.).
  • the medical agent comprises a macrolide.
  • the macrolide comprises rapamycin (i.e., Sirolimus).
  • compositions and methods of the invention are provided.
  • 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.
  • the population of particles of the disclosure can comprise a metallic material.
  • 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.
  • the particles comprise a rare earth element.
  • the population of particles of the present invention can comprise a ceramic material.
  • the ceramic material include an aluminide, boride, beryllia, carbide, chromium oxide, hydroxide, iron oxide, sulfide, nitride, mullite, kyanite, ferrite, titania zirconia, yttria, and magnesia.
  • the population of particles of the disclosure can comprise an encapsulation agent, e.g., an agent that is used to form a plurality of nanoparticles, wherein a nanoparticle of the plurality of nanoparticles encapsulate a medical agent as disclosed herein.
  • the encapsulation agent can be an emulsifier.
  • the encapsulation agent can be a polymer (e.g., a polymeric emulsifier).
  • 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 polyfl, 6-bis(p-carboxyphenoxy)hexane] (pCPH), poly(hydroxbutyric acid-cohydroxyvaleric acid) (PHB-PVA), polyethylene glycol (PEG)-poly (lactic acid) (PLA) copolymer (PEG-PLA), poly-s-caprolactone (PCL), poly-alkyl-cyano- acrylate (PAC), poly(ethyl)cyanoacrylate (PEC), polyisobutyl cyanoacrylate, poly-N-(2- hydroxypropyl)methacryl
  • the polymers of the disclosure can comprise copolymers, for example, bipolymers, terpolymers, and quaterpolymers.
  • 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.
  • 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
  • a particle of the population of particles can comprise a single material (e.g., a single polymer, such as PLGA or PLGA-PEG).
  • 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.
  • 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.
  • the particle of the population of particles can be a core-shell particle.
  • a copolymer e.g., PLGA-PEG
  • a first polymer e.g., PLGA
  • a second polymer e.g., PEG
  • the core can be substantially made of the first polymer (e.g., PLGA)
  • the shell can be substantially made of the second polymer (e.g., PEG).
  • the population of particles of the present invention are polymeric particles (e.g., particles comprising a polymeric core).
  • 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.
  • the population of particles in a composition 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.
  • 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 1 nanometer (nm) to about 1000 nm, between about 10 nm to about 1000 nm, or between about 100 nm to about 1000 nm.
  • DLS dynamic light scattering
  • the cross-sectional dimension of the particle can be at least or up to about 1 nm, at least or up to about 5 nm, at least or up to about 10 nm, at least or up to about 50 nm, 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.
  • 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.
  • MNP mesoscale nanoparticle
  • 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 1 nanometer (nm) to about 1000 nm, between about 10 nm to about 1000 nm, or between about 100 nm to about 1000 nm.
  • the mean cross-sectional dimension of the population of particles can be at least or up to about 1 nm, at least or up to about 5 nm, at least or up to about 10 nm, at least or up to about 50 nm, 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.
  • the mean cross- sectional dimension of the population of particles 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.
  • 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 +
  • +40 mV at least or up to about +45 mV, at least or up to about +50 mV, at least or up to about
  • +90 mV or at least or up to about +100 mV.
  • 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.
  • 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 (pg), at least or up to about 5 pg, at least or up to about 10 pg, at least or up to about 50 pg, at least or up to about 100 pg, at least or up to about 500 pg, or at least or up to about 1,000 pg.
  • ng nanogram
  • 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
  • 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 pg, at least or up to about 5 pg, at least or up to about 10 pg, at least or up to about 50 pg, at least or up to about 100 pg, at least or up to about 500 pg, or at least or up to about 1,000 pg 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
  • encapsulated medical agent e.g., encapsulated in a particle of the population of particles
  • encapsulated medical agent 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 pg, at least or up to about 5 pg, at least or up to about 10 pg, at least or up to about 50 pg, at least or up to about 100 pg, at least or up to about 500 pg, or at least or up to about 1,000 pg per particle or per particle in a population of particles.
  • encapsulated medical agents e.g., encapsulated in a particle of the population of particles
  • 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 pg, at least or up to about 5 pg, at least or up to about 10 pg, at least or up to about 50 pg, at least or up to about 100 pg, at least or up to about 500 pg, or at least or up to about 1,000 pg per particle or per particle in a population of particles.
  • 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
  • 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 pg, at least or up to about 5 pg, at least or up to about 10 pg, at least or up to about 50 pg, at least or up to about 100 pg, at least or up to about 500 pg, 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.
  • mg milligram
  • 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
  • encapsulated medical agents e.g., encapsulated in a particle of the population of particles of the composition
  • milligram milligram
  • encapsulated medical agents e.g., encapsulated in a particle of the population of particles of the composition
  • 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 pg, at least or up to about 5 pg, at least or up to about 10 pg, at least or up to about 50 pg, at least or up to about 100 pg, at least or up to about 500 pg, 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.
  • milligram milligram
  • 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
  • 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.
  • 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, antiandrogens, 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,
  • a therapeutic agent can be an AKT inhibitor (e.g., MK2206), a ROS inhibitor (e.g., Edaravone), a statin, mTOR inhibitor (e.g., RAD001 (everolimus), rapamycin, etc.), TGF-P signaling agonist, TGF-P 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, phosphodie
  • AKT inhibitor e.
  • therapeutic agent can comprise a macrolide.
  • a macrolide include azithromycin, clarithromycin, dirithromycin, erythromycin, erythromycin A, erythromycin B, erythromycin C, erythromycin D, erythromycin E, erythromycin estolate, roxithromycin, troleandomycin, telithromycin, spectinomycin, methymycin, neomethymycin, erythronolid, megalomycin, picromycin, narbomycin, oleandomycin, rapamycin, triacetyl-oleandomycin, laukamycin, kujimycin A, albocyclin, and cineromycin B, and pharmaceutically-acceptable salts of any of the foregoing.
  • the therapeutic agent is rapamycin, a functional variant thereof, or a modification thereof. In some embodiments, the therapeutic agent is not a linear polysaccharide. In some embodiments, the therapeutic agent is not a monosaccharide. In some embodiments, the therapeutic agent is not streptomycin.
  • a therapeutic agent can comprise a chemotherapeutic agent.
  • 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, t
  • 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.
  • Radiolabels include " m Tc, ni In, 64 Cu, 67 Ga, 186 Re, 188 Re, 153 Sm, 177 LU, 67 CU, 123 I, 124 I, 125 I, U C, 13 N, 15 O, 18 F, 186 Re, 188 Re, 153 Sm, 166 Ho, 177 Lu, 149 Pm, 90 Y, 212 Bi, 103 Pd, 109 Pd, 159 Gd, 140 La, 198 Au, 199 Au, 169 Yb, 175 Yb, 165 Dy, 166 Dy, 67 Cu, 105 Rh, m Ag, 89 Zr, and 192 Ir.
  • Non-limiting examples of paramagnetic metal ions include Gd(III), Dy(III), Fe(III), and Mn(II).
  • 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.
  • fluorophores 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
  • 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 sugar include mannose, sucrose, dextrose, trehalose, lactose, raffinose, and cyclodextrin (e.g., beta-cyclodextrin, such as (2-Hydroxypropyl)-beta-cyclodextrin).
  • the dehydrating agent is a polysaccharide, such as 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.
  • 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.
  • the dehydrating agent e.g., a polysaccharide, such as sucrose
  • the dehydrating agent 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 pg, at least or up to about 5 pg, at least or up to about 10 pg, at least or up to about 50 pg, at least or up to about 100 pg, at least or up to about 500 pg, 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, at least or up to about 100 mg, at least or up to about 500 mg, or at least or up to about 1 mill
  • the dehydrating agent e.g., a polysaccharide, such as sucrose
  • the dehydrating agent 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
  • the nanoparticles of the present invention are suspended in a solution comprising the dehydrating agent (e.g., a polysaccharide, such as sucrose), and the resulting mixture is lyophilized and stored prior to use.
  • the dehydrating agent e.g., a polysaccharide, such as sucrose
  • 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
  • nanoparticles of the present invention are suspended in a solution comprising the dehydrating agent (e.g., a polysaccharide, such as sucrose), and the resulting mixture is lyophilized and stored prior to use.
  • the dehydrating agent e.g., a polysaccharide, such as sucrose
  • the plurality of nanoparticles (NP) and the dehydrating agent (DA) can be present in a weight-to- weight ratio (NP:DA) that is between about 20: 1 and about 1 : 100, about 20: 1 and about 1 :50, about 10: 1 and about 1 :50, about 1 : 1 and about 1 :50, about 1 : 1 and about 1 :40, about 1 : 1 and about 1 :30, about 1 : 1 and about 1 :25, about 1 : 1 and about 1 :20, about 1 : 1 and about 1 : 15, about 1 : 1 and about 1 : 10, or about 1 : 1 and about 1 :5.
  • NP:DA weight-to- weight ratio
  • the NP:DA ratio can be at least or up to about 10: 1, at least or up to about 5: 1, at least or up to about 4: 1, at least or up to about 3 : 1, at least or up to about 2: 1, at least or up to about 1 : 1, at least or up to about 1 :2, at least or up to about 1 :3, at least or up to about 1 :4, at least or up to about 1 :5, at least or up to about 1 :6, at least or up to about 1 :7, at least or up to about 1 :8, at least or up to about 1 :9, at least or up to about 1 : 10, at least or up to about 1 : 15, at least or up to about 1 :20, at least or up to about 1 :30, at least or up to about 1 :40, at least or up to about 1 :50, or at least or up to about 1 : 100.
  • a presence of the hydration agent as disclosed herein 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
  • the hydration agent as disclosed herein e.g., a polysaccharide, such as sucrose or trehalose
  • a composition in an amount that is greater than 1% and lesser than 20%, greater than 2% and lesser than 10%, greater than 2% and lesser than 9%, greater than 2% and lesser than 8%, greater than 2% and lesser than 7%, greater than 2% and lesser than 6%, greater than 2% and lesser than 5%, greater than 2% and lesser than 4%, greater than 2% and lesser than 2%, greater than 3% and lesser than 10%, greater than 4% and lesser than 10%, greater than 5% and lesser than 10%, greater than 6% and lesser than 10%, greater than 7% and lesser than 10%, greater than 8% and lesser than 10%, or greater than 9% and lesser than 10%.
  • the composition can comprise a plurality of nanoparticles, as disclosed herein.
  • the composition can be a mixture comprising the hydration agent but without any nanoparticle (e.g., a mixture comprising a solvent and the hydration agent prior to being in contact with a plurality of nanoparticles).
  • the hydration agent as disclosed herein can comprise at least two different polysaccharides (e.g., two or more members selected from the group consisting of sucrose, trehalose, and 2 hydroxy propyl beta-cyclodextrin).
  • an amount of a first polysaccharide and an amount of a second polysaccharide (which is different than the first polysaccharide) in a composition as disclosed herein may be the same.
  • the amount of the first polysaccharide and the amount of the second polysaccharide in the composition may be different.
  • the first polysaccharide and the second polysaccharide can be present in the composition in a weight-to- weight ratio of at least or up to about 1 : 1, at least or up to about 1 :2, at least or up to about 1 :3, at least or up to about 1 :4, at least or up to about 1 :5, at least or up to about 1 :6, at least or up to about 1 :7, at least or up to about 1 :8, at least or up to about 1 :9, at least or up to about 1 : 10, at least or up to about 1 : 15, at least or up to about 1 :20, at least or up to about 1 :25, at least or up to about 1 :30, at least or up to about 1 :35, at least or up to about 1 :40, at least or up to about 1 :45, at least or up to about 1 :50, at least or up to about 1 :60, at least or up to about 1 :70, at least or up to about 1 :50, at
  • a medical agent of the disclosure can be a nucleic acid molecule (e.g., RNA).
  • a particle of the population of particles (e.g., MNPs) as disclosed herein comprises (i) a particle forming agent, (i) a medical agent (e.g., a nucleic acid molecule, such as small interfering RNA (siRNA)), and (iii) an excipient.
  • the particle can be further treated to form an additional outer layer (e.g., via using a surfactant, such as F-68 for double emulsion).
  • the particle forming agent as disclosed herein can be a polymer.
  • the polymer can comprise a first portion for forming an inner portion (e.g., core) of the particle and a second portion for forming an outer portion (e.g., an outer later or a shell) of the particle.
  • the particle forming agent is poly(lactic-co-glycolic acid) (PLGA) that is coupled to one or more of the following: polyethylene glycol (PEG), PEG-carboxylic acid, PEG-carboxylic acid- didodecyldimethylammonium bromide (DMAB), and methoxy PEG.
  • the particle forming agent is PLGA-PEG.
  • the medical agent disclosed herein comprises a therapeutic agent, such as a small molecule, polypeptide (e.g., a peptide, a protein, an antibody, etc.), polynucleotide (e.g., deoxyribonucleic acid (DNA), ribonucleic acid (RNA), variants thereof, combinations thereof, etc.
  • a therapeutic agent such as a small molecule, polypeptide (e.g., a peptide, a protein, an antibody, etc.), polynucleotide (e.g., deoxyribonucleic acid (DNA), ribonucleic acid (RNA), variants thereof, combinations thereof, etc.
  • the therapeutic agent can comprise siRNA, antisense oligonucleotide (ASO), microRNA, mRNA (messenger RNA) for expression of one or more proteins (e.g., CRISPR/Cas activity of mutated protein), guide RNA (e.g., single guide RNA) for endonuclease activity (e.g., CRISPR/Cas activity), etc.
  • the therapeutic agent is siRNA.
  • the medical agent can as disclosed herein be a polypeptide that is not a renal proximal tubular (PT) NFKB essential modulator (NEMO)-binding peptide.
  • PT renal proximal tubular
  • NEMO essential modulator
  • the medical agent can as disclosed herein can have a number average molar mass that is less than or equal to about 20 kilodalton (kDa), less than or equal to about 19 kDa, less than or equal to about 18 kDa, less than or equal to about 17 kDa, less than or equal to about 16 kDa, less than or equal to about 15 kDa, less than or equal to about 14 kDa, less than or equal to about 13 kDa, less than or equal to about 12 kDa, less than or equal to about 11 kDa, less than or equal to about 10 kDa, less than or equal to about 9 kDa, less than or equal to about 8 kDa, less than or equal to about 7 kDa, less than or equal to about 6 kDa, less than or equal to about 5 kDa, less than or equal to about 4 kDa, less than or equal to about 3 kDa, less than or
  • the medical agent does not consist of a siRNA molecule. In some embodiments, the medical agent is not a siRNA molecule. In some embodiments, the medical agent does not consist of a nucleic acid molecule. In some embodiments, the medical agent is not a nucleic acid molecule.
  • the excipient as disclosed herein can comprise a polymer excipient or a lipid molecule excipient (e.g., a fatty acid excipient).
  • the polymer excipient can be a homopolymer or a heteropolymer.
  • Non-limiting examples of the polymer excipient include a poly(amino acid) excipient, polyethyleneimine (PEI, linear or branched) (e.g., an average molecular weight or a number average molar mass ranging between 600 Dalton (Da) and 270,000 Da), poly 2-dimethyl amino ethyl methacylate (PDMAEMA), and chitosan (e.g., high molecular weight or high number average molar mass chitosan).
  • PEI polyethyleneimine
  • PDMAEMA poly 2-dimethyl amino ethyl methacylate
  • chitosan e.g., high molecular weight or high number average molar mass chitosan.
  • Non-limiting examples of the poly(amino acid) excipient include poly-l-lysine; poly-l-arginine; and poly-l-histidine.
  • Non-limiting examples of the lipid excipient include l,2-di-O-octadecenyl-3 -trimethylammonium propane (DOTMA); 1,2-dioleoyl- 3 -trimethylammonium -propane (DOTAP); DC-Cholesterol-HCl (3B-[N-(N',N'- dimethylaminoethane)-carbamoyl]cholesterol hydrochloride); l,2-dioleyloxy-3- dimethylaminopropane (DODMA); l,2-distearoyl-sn-glycero-3-phosphoethanolamine-N- [amino(polyethylene glycol)-5000] (ammonium salt) (DSPE PEG); and 1,2-distearoyl-sn- glycero-3 -phosphocholine (DSPC or DSPC PEG).
  • DOTMA 1,2-dioleoyl- 3 -trimethylammonium -prop
  • the excipient is poly-l-lysine (e.g., having a number average molar mass between about 1.6 kDa to about 132 kDa). In some embodiments, the excipient is poly-l-arginine (e.g., having a number average molar mass between about 1.9 kDa to about 40 kDa). In some embodiments, the excipient is poly-l-histidine (e.g., having a number average molar mass between about 5 kDa to about 70 kDa).
  • the excipient is a protamine or a protamine sulfate (e.g., having a number average molar mass of about 4 or 5 kDa.
  • the medical agent and the excipient can be mixed, prior to further combining such mixture with a particle forming agent, to form one or more particles (e.g., one or more MNPs).
  • the particle forming agent and the excipient can be mixed, prior to further combining such mixture with a medical agent, to form one or more particles (e.g., one or more MNPs).
  • the particle forming agent, the medical agent, and the excipient can be mixture together (e.g., in a single batch) to form the one or more particles.
  • the medical agent (MA) and the excipient (EX) can be mixed in a weight-to-weight ratio (MA:EX) of at least or up to about 1 : 100, at least or up to about 2: 100, at least or up to about 3: 100, at least or up to about 4: 100, at least or up to about 5: 100, at least or up to about 10: 100, at least or up to about 20: 100, at least or up to about 30: 100, at least or up to about 40: 100, at least or up to about 50: 100, at least or up to about 100: 100, at least or up to about 100:50, at least or up to about 100:40, at least or up to about 100:30, at least or up to about 100:20, at least or up to about 100: 10, at least or up to about 100:5, at least or up to about 100:4, at least or up to about 100:3, at least or up to about 100:2, or at least or up to about 100: 1.
  • MA:EX weight-to-weight ratio
  • the medical agent (MA) and the excipient (EX) can be mixed in a molar ratio (MA:EX) of at least or up to about 1 : 100, at least or up to about 2: 100, at least or up to about 3: 100, at least or up to about 4: 100, at least or up to about 5: 100, at least or up to about 10: 100, at least or up to about 20: 100, at least or up to about 30: 100, at least or up to about 40: 100, at least or up to about 50: 100, at least or up to about 100: 100, at least or up to about
  • 100:20 at least or up to about 100: 10, at least or up to about 100:5, at least or up to about 100:4, at least or up to about 100:3, at least or up to about 100:2, or at least or up to about 100: 1.
  • the medical agent can be a polynucleotide (e.g., siRNA), and the medical agent and the excipient can be a poly(amino acid).
  • the polynucleotide medical agent and the poly(amino acid) excipient can be mixed in a ratio between (i) a number of phosphates in the backbone of the polynucleotide (e.g., a total number of phosphates in a total amount of polynucleotides) (MA-p) and (ii) a number of amine groups in the backbone of the poly(amino acid) excipient (e.g., a total number of amines in a total amount of poly(amino acid)) (EX-a) (MA-p:EX-a) of at least or up to about 1 : 100, at least or up to about 2: 100, at least or up to about 3: 100, at least or up to about
  • the medical agent and the excipient can be mixed (prior to forming the particle as disclosed herein) 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 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 12 hours, or at least or up to about 24 hours.
  • a population of particles comprising the medical agent and the excipient can exhibit enhance functionality of the medical agent (e.g., bioactivity of a therapeutic agent, such as gene silencing activity of siRNA) as compared to a control population of nanoparticles with the medical agent but without the excipient by 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 10%, at least or up to about 15%, at least or up to about 20%, at least or up to about 30%, at least or up to about 40%, at least or up to about 50%, at least or up to about 60%, at least or up to about 70%, at least or up to about 80%, at least or up to about 90%, at least or up to about 100%, at least or up to about 150%, at least or up to about 200%, at least or up to about 300%, at least or up to about 400%, or at least or at least or
  • the excipient as disclosed herein is coupled to (e.g., covalently conjugated to) the particle forming agent. In some embodiments, the excipient as disclosed herein is not and need not be coupled to (e.g., covalently conjugated to) the particle forming agent.
  • the medical agent (e.g., siRNA) and the excipient are mixed prior to mixing with the particle forming agent to form one or more particles (e.g., MNPs), such that the medical agent and the excipient are substantially within an inner portion of the particle(s) (e.g., encapsulated within the inner portion of the particle(s)). In some examples, the medical agent and the excipient is not and need not be coated on an outer surface of the particle(s).
  • a composition as disclosed herein can be a mixture comprising a medical agent and an excipient.
  • the excipient can be oppositely charged from the medical agent (e.g., in a solvent, such as an aqueous solvent or an organic solvent).
  • the excipient can be oppositely charged from the medical agent in a protic solvent.
  • the excipient can be an anionic excipient (e.g., having an overall negative charge in a solvent for complexing with an overall positively charged medical agent).
  • the excipient can be a cationic excipient (e.g., having an overall positive charge in a solvent for complexing with an overall negatively charged medical agent).
  • the mixture can further comprise an encapsulation agent, such as a polymer as disclosed herein (e.g., a PLA, PGA, PLGA, etc.).
  • an encapsulation agent such as a polymer as disclosed herein (e.g., a PLA, PGA, PLGA, etc.).
  • Such mixture can be incubated (e.g., prior to using the mixture to form a plurality of nanoparticles) 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 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
  • Such incubation can occur at a temperature of at least or up to about -20 degree Celsius (°C), at least or up to about -18 °C, at least or up to about -15 °C, at least or up to about -10 °C, at least or up to about -5 °C, at least or up to about 0 °C, at least or up to about 1 °C, at least or up to about 2 °C, at least or up to about 3 °C, at least or up to about 4 °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 24 °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 40 °C, or at least or up to about 50 °C.
  • °C degree Celsius
  • a nanoparticle e.g., each nanoparticle of the plurality of nanoparticles disclosed herein may comprise (e.g., encapsulate) a medical agent and an excipient that is oppositely charged from the medical agent.
  • An average amount of the medical agent loaded to the nanoparticle of the plurality of nanoparticles may be higher than that in a particle of a control plurality of nanoparticles lacking the excipient by 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 10%, at least or up to about 15%, at least or up to about 20%, at least or up to about 30%, at least or up to about 40%, at least or up to about 50%, at least or up to about 60%, at least or up to about 70%, at least or up to about 80%, at least or up to about 90%, at least or up to about 100%, at least or up to about 150%, at least or up to about 200%, at least or up to about 300%, at least or up to about 400%, or at least or up to about 500%.
  • a nanoparticle e.g., each nanoparticle of the plurality of nanoparticles disclosed herein may comprise (e.g., encapsulate) a medical agent and an excipient that is oppositely charged from the medical agent.
  • a poly dispersity index (PDI) of the plurality of nanoparticles can be (e.g., as ascertained by dynamic light scattering (DLS)) less than or equal to about 20, 19, 18, 17, 16, 16, 15, 14, 13, 12, 11, 10, 9, 8, 7, 6, 5, 4.9, 4.8, 4.7, 4.6, 4.5, 4.4, 4.3, 4.2, 4.1, 4.0, 3.9, 3.8, 3.7, 3.6, 3.5, 3.4, 3.3, 3.2, 3.1, 3.0, 2.9, 2.8, 2.7, 2.6, 2.5, 2.4, 2.3, 2.2, 2.1, 2.0, 1.9, 1.8, 1.7, 1.6, 1.5, 1.45, 1.40, 1.35, 1.30, 1.25, 1.20, 1.15, 1.14, 1.13, 1.12, 1.11, 1.10, 1.09, 1.08, 1.07, 1.06, 1.05, 1.04, 1.03, 1.02, 1.01, or 1.00.
  • DLS dynamic light scattering
  • the PDI of the plurality of nanoparticles can be lower than that a control plurality of nanoparticles lacking the excipient by at least about 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1, 1.5, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, or more.
  • the medical agent can have a net positive charge (e.g., in a solvent), and the oppositely charged excipient can have a net negative charge. In some embodiments, the medical agent can have a net negative charge (e.g., in a solvent), and the oppositely charged excipient can have a net positive charge.
  • Non-limiting examples of an excipient that can be negatively charged can include a lipid molecule (e.g., a fatty acid molecule), such as lauric acid, lauroleic acid, tetradeadienoic acid, octanoic acid, myristic acid, myristoleic acid, decenoic acid, decanoic acid, hexadecenoic acid, palmitoleic acid, palmitic acid, linolenic acid, linoleic acid, pamoic acid, oleic acid, cholic acid, vaccenic acid, stearic acid, eicosapentaenoic acid, arachadonic acid, mead acid, arachidic acid, docosahexaenoic acid, docosapentaenoic acid, docosatetraenoic acid, docosenoic acid, tetracosanoic acid, hexacosenoic acid
  • Non-limiting examples of an excipient that can be positively charged can include a lipid molecule (e.g., a fatty acid molecule), such as N,N-dimethyl-(2,3-dioleyloxy) propylamine (DODMA), l,2-di-O-octadecenyl-3 -trimethylammonium propane (DOTMA), 1,2-dipalmitoyl- sn-glycero-O-ethyl-3 -phosphocholine (DPePC), l,2-dioleoyl-3 -dimethylammonium propane (DODAP), and l,2-dioleoyl-3 -trimethylammonium -propane (DOTAP); and the neutral lipid is selected from the group consisting of l,2-distearoyl-sn-glycero-3 -phosphocholine (DSPC), 1- palmitoyl-2-oleoyl-sn-glycero-3 -phosphocho
  • an excipient as disclosed herein can be a neutral lipid molecule that exists either in an uncharged or neutral zwitterionic form at physiological pH.
  • the present disclosure provides methods of making the population of particles as disclosed herein.
  • the present disclosure provides methods of use of the population of particles as disclosed herein, e.g., for diagnostics or therapeutic applications. Uses
  • 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).
  • 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.).
  • control population of particles may be a population of emulsions or micelles.
  • 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).
  • 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.
  • 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.
  • 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.
  • 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.
  • 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.
  • 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.
  • 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.
  • 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.
  • certain nanoparticles e.g., mesoscale nanoparticles, e.g., having a cross-sectional diameter between about 300 nm to about 700 nm
  • 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.
  • 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).
  • the population of particles does not comprise any such targeting agent for selective targeting or localization to a specific tissue.
  • 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.
  • 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.
  • optical signal e.g., fluorescence
  • 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
  • the medical agents of the present invention e.g., therapeutic agents, such as rapamycin
  • the population of particles e.g., encapsulated therein or coupled thereto
  • 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.
  • HPLC high-performance liquid chromatography
  • the medical agents 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
  • the medical agents e.g., therapeutic agents, such as rapamycin
  • the medical agents 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).
  • the medical agents e.g., therapeutic agents, such as rapamycin
  • the medical agents 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,
  • tissue e.g., the kidneys
  • the composition as disclosed herein e.g., a composition comprising a plurality of nanoparticles and optionally a polysaccharide dehydration agent
  • the thermal treatment can comprise reducing an average temperature of the composition, e.g., to freeze the composition.
  • the average temperature of the composition can reduced by at least or up to about 0.1 degree Celsius (°C), at least or up to about 0.2 °C, at least or up to about 0.5 °C, at least or up to about 1 °C, at least or up to about 2 °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 21 °C, at least or up to about 22 °C, at least or up to about 23 °C, at least or up to about 24 °C, at least or up to about 25 °C, at least or up to about 26 °C, at least or up to about 27 °C, at least or up to about 28 °C, at least or up to about 29 °C, at least or up to about 30 °C, at least or up to about 0.1 degree Celsius (°C), at least or up to about 0.2 °C, at least or up to
  • the average temperature of the composition can be at least or up to about 30 °C, at least or up to about 25 °C, at least or up to about 20 °C, at least or up to about 15 °C, at least or up to about 10 °C, at least or up to about 5 °C, at least or up to about 4 °C, at least or up to about 3 °C, at least or up to about 2 °C, at least or up to about 1 °C, at least or up to about 0 °C, at least or up to about -5 °C, at least or up to about - 10 °C, at least or up to about -20 °C, at least or up to about -30 °C, at least or up to about -40 °C, at least or up to about -50 °C, at least or up to about -60 °C, at least or up to about -70 °C, at least or up to about -80 °C, at least or up to about
  • the composition as disclosed herein can comprise a solvent (e.g., an aqueous medium), and at least a portion of the solvent may be removed from the composition.
  • a solvent e.g., an aqueous medium
  • at least a portion of the solvent may be removed from the composition after the composition is subjected to the thermal treatment (e.g., substantially frozen) as disclosed herein.
  • At least or up to about 1 %, at least or up to about 2 %, at least or up to about 5 %, at least or up to about 10 %, at least or up to about 15 %, at least or up to about 20 %, at least or up to about 30 %, at least or up to about 40 %, at least or up to about 50 %, at least or up to about 60 %, at least or up to about 70 %, 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 substantially all of the solvent may be removed from the composition.
  • Bioavailability of the medical agents of the present disclosure in an animal 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 pg-hour/ml).
  • 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
  • AUCo-24 area under the curve
  • 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.
  • 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 can be any parameters suitable for describing pharmaceutical compositions of the disclosure.
  • the medical agents e.g., therapeutic agents, such as rapamycin
  • 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
  • the medical agents e.g., therapeutic agents, such as rapamycin
  • administered to the subject via the population of particles of the present invention can exhibit a mean AUC0-24 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
  • the medical agents e.g., therapeutic agents, such as rapamycin
  • 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
  • the medical agents e.g., therapeutic agents, such as rapamycin
  • 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 -
  • the medical agents e.g., therapeutic agents, such as rapamycin
  • 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
  • the medical agents e.g., therapeutic agents, such as rapamycin
  • 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 -
  • the medical agents e.g., therapeutic agents, such as rapamycin
  • 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 -
  • the medical agents e.g., therapeutic agents, such as rapamycin
  • 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
  • the medical agents e.g., therapeutic agents, such as rapamycin
  • 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
  • the medical agents e.g., therapeutic agents, such as rapamycin
  • 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
  • the medical agents e.g., therapeutic agents, such as rapamycin
  • 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
  • a degradation rate of the medical agents e.g., therapeutic agents, such as rapamycin
  • the degradation rate can be measured at an ambient temperature of at least or up to about 3 degrees Celsius (°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.
  • °C degrees Celsius
  • the degradation rate at an ambient temperature (e.g., at least about 25 °C) of the medical agents (e.g., therapeutic agents, such as rapamycin) 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
  • the population of particles of the disclosure can exhibit a different release rate of the medical agents (e.g., therapeutic agents, such as rapamycin) as compared to that from a control population of particles.
  • the medical agents e.g., therapeutic agents, such as rapamycin
  • the release rate of medical agents 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
  • the release rate of medical agents 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
  • the population of particles of the disclosure can release the medical agents (e.g., therapeutic agents, such as rapamycin) 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
  • the medical agents e.g., therapeutic agents, such as rapamycin
  • the population of particles comprising the therapeutic agents of the disclosure can modulate (e.g., enhance, reduce, or prolong) a signaling activity of a cell of the body of the subject, e.g., to a greater degree than that by control therapeutic agents.
  • administration of the therapeutic agents via the population of particles of the disclosure can enhance the signaling activity of the cell of the subject’s body, as compared to the control therapeutic agents.
  • administration of the therapeutic agents via the population of particles of the disclosure can reduce (e.g., inhibit) the signaling activity of the cell of the subject’s body, as compared to the control therapeutic agents.
  • administration of the therapeutic agents via the population of particles of the disclosure can prolong the signaling activity of the cell of the subject’s body, as compared to the control therapeutic agents.
  • the signaling activity of the cell can be an intracellular activity of the cell.
  • the intracellular signaling activity involves pS6 tyrosine kinase activity, and a change in the pS6 tyrosine kinase activity can be ascertained by measuring a change in the degree of phosphorylation of pS6 in the cell (e.g., via Western blot analysis using Phospho-S6 Ribosomal Protein (Ser235/236) Antibody).
  • the intracellular signaling activity involves mTOR activity, and a change in the mTOR activity can be ascertained by measuring a change in the degree of phosphorylation of mTOR in the cell (e.g., via Western blot analysis using Anti-Phospho-mTOR (Ser2481) antibody).
  • administration of the therapeutic agents via the population of particles of the disclosure can reduce the signaling activity (e.g., pS6 tyrosine kinase activity, mTOR activity) of the cell to a greater degree than the control 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
  • the signaling activity e.g., pS6 tyrosine kinase activity
  • administration of the therapeutic agents via the population of particles of the disclosure can reduce (e.g., inhibit) the signaling activity (e.g., pS6 tyrosine kinase activity, mTOR activity) of the cell for a longer duration of time than the control therapeutic agents do 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
  • administration of the therapeutic agents via the population of particles of the disclosure can reduce (e.g., inhibit) the signaling activity (e.g., pS6 tyrosine kinase activity, mTOR activity) of the cell for a longer duration of time than the control therapeutic agents do by 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
  • the signaling activity e.g., pS6 tyrosine kinase activity
  • administration of the therapeutic agents via the population of particles of the disclosure can reduce the signaling activity (e.g., pS6 tyrosine kinase activity, mTOR activity) of a cell in a targeted or localized tissue (e.g., the kidneys) to a greater degree than that 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
  • administration of the therapeutic agents via the population of particles of the disclosure can reduce the signaling activity (e.g., pS6 tyrosine kinase activity, mTOR activity) of a cell in a targeted or localized tissue (e.g., the kidneys) for a longer duration of time than that in other types of tissue (e.g., heart, lung, spleen, liver) by 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 1 minute, at least or up to about 2 minutes, at least or up to about 3 minutes, at
  • a subject with polycystic kidney disease can be administered with the therapeutic agents (e.g., a macrolide, such as rapamycin) via the population of particles of the disclosure, to reduce or eliminate the presence (e.g., amount, volume, etc.) of cysts in the kidneys.
  • the therapeutic agents e.g., a macrolide, such as rapamycin
  • Administration of the therapeutic agents via the population of nanoparticles of the disclosure can reduce the presence of cysts in the kidneys of the subject by at least or up to about 1 %, at least or up to about 5 %, at least or up to about 10 %, at least or up to about 15 %, 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 %, at least or up to about at least or up to about 99 %, or about 100%, as ascer
  • administration of the therapeutic agents via the population of nanoparticles of the disclosure can reduce cyst growth in the kidneys of the subject to a greater degree than the control therapeutic agents do 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
  • administration of the therapeutic agents via the population of nanoparticles of the disclosure can enhance renal functional of the subject to a greater degree than the control therapeutic agents do 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-
  • the therapeutic agents e.g., a macrolide, such as rapamycin
  • administration of the therapeutic agents via the population of nanoparticles of the disclosure can preserve or prolong renal functional of the subject for a longer duration than the control therapeutic agents do 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-
  • the therapeutic agents e.g., a macrolide, such as rapamycin
  • administration of the therapeutic agents via the population of nanoparticles of the disclosure can reduce epithelial cell proliferation in the kidneys of the subject to a greater degree than the control therapeutic agents do 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
  • administration of the therapeutic agents via the population of nanoparticles of the disclosure can increase apoptosis of cyst-lining cells in the kidneys of the subject to a greater degree than the control therapeutic agents do 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
  • administration of the therapeutic agents 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 sideeffects related to the therapeutic agents, or (iii) reduce a number of administrations of the therapeutic agents that is sufficient to elicit a therapeutic effect.
  • administration of the therapeutic agents via the population of particles of the present invention can reduce a therapeutic dose of the therapeutic agents (e.g., a macrolide, such as rapamycin) 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
  • the therapeutic agents e.g.,
  • 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 macrolide, such as rapamycin) 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
  • the therapeutic agents e.g.,
  • 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,
  • 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.
  • kidney cells i.e., renal cells
  • 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.
  • kidney diseases include lupus, glomerulonephritis, and renal cell carcinoma (RCC), each of which can arise in the proximal tubules of the kidneys.
  • RRC renal cell carcinoma
  • therapeutics utilizing the population of particles of the disclosure can be utilized to treat or ameliorate symptoms of other tissue-specific diseases.
  • therapeutics utilizing the population of particles of the disclosure can be utilized to treat cancer.
  • 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 lympho
  • 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.
  • 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.
  • 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.
  • 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, Bo
  • 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), Gas
  • 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).
  • a population of nanoparticles can be prepared for use as delivery carriers for medical agents, such as therapeutics agents (e.g., drugs, such as rapamycin) or imaging agents (e.g., dyes).
  • the population of nanoparticles can comprise polymeric nanoparticles, such as PLGA-PEG nanoparticles.
  • PLGA-COOH Poly (lactic-co-glycolic acid) (e.g., about 5 grams (g)) was gradually added into di chloromethane (DCM) (e.g., about 20 milliliters (mL)) under constant stirring on a stirrer plate. The mixture was subjected to stirring until PLGA-COOH was substantially dissolved in DCM.
  • DCM di chloromethane
  • N-hydroxy succinimide (e.g., about 135 milligrams (mg)) and l-ethyl-3-(3- dimethylaminopropyl)-carbodiimide (EDC) (e.g., about 230 mg) were added to of the mixture of PLGA-COOH in DCM. This mixture was stirred (e.g., overnight for about 16 hours) at, e.g., 1100 rotations per minute (RPM).
  • N-hydroxy succinimide (e.g., about 135 milligrams (mg)
  • EDC l-ethyl-3-(3- dimethylaminopropyl)-carbodiimide
  • polymer e.g., NHS-conjugated PLGA
  • cold ethyl ether e.g., about 10 mL
  • the precipitated polymer was washed (e.g., 3 times) in a mixture of cold ethanol and diethyl ether (e.g., a total volume of about 10 mL, at a 50:50 volume ratio of ethanol to diethyl ether) in a slow stream. Waste was discarded and the polymer was vacuum dried, e.g., for around 4 hours. After drying, PLGA-NHS was stored at -20 °C.
  • PLGA-NHS e.g., about 1 g
  • chloroform e.g., about 4 mL
  • N,N-diisopropylethylamine e.g., 37.7 microliters or 28 mg
  • NH2-PEG-COOH e.g., about 250 mg
  • the mixture was stirred (e.g., overnight for about 16 hours) at, e.g., 1100 RPM.
  • the polymer e.g., PLGA-PEG
  • cold methanol e.g., about 10 mL
  • Methanol was poured off, waste was discarded, and the resulting polymer product was washed (e.g., 3 times) with cold methanol by gently pouring methanol into the scintillation vial.
  • the polymer was vacuum dried (e.g., for about 4 hours) and stored at -20 °C.
  • PLGA-PEG e.g., about 100 mg
  • CAN acetonitrile
  • Rapamycin e.g., about 10 mg
  • medium stirring e.g., for about 10-15 minutes
  • deionized water e.g., about 4mL
  • Pluronic F-68 poloxamer e.g., about 75 pL
  • a Hamilton glass syringe (e.g., a 10 mL Hamilton glass syringe) was filled with a solution (e.g., about 2 mL) comprising PLGA-PEG and rapamycin.
  • the syringe was secured on a syringe pump.
  • the solution comprising PLGA-PEG and rapamycin was added into the solution comprising deionized water and F-68 at a speed of 0.1 mL per minute.
  • polymeric nanoparticles comprising rapamycin were collected in a container (e.g., a 50 mL conical tube) and spun down by using ultracentrifugation at, e.g., 7356 relative centrifugal force (RCF) for about 15 minutes at 4 °C. Subsequently, supernatant was discarded, and the polymeric nanoparticles were resuspended in deionized water (e.g., about 10 mL) and spun down, e.g., via ultra-centrifugation as abovementioned. After about 15 minutes, supernatant was discarded, and the polymeric nanoparticles were resuspended in a solution comprising sucrose (e.g., about 5% sucrose by weight).
  • sucrose e.g., about 5% sucrose by weight
  • the polymeric nanoparticles resuspended in 5% sucrose were frozen by dipping the container into a liquid nitrogen-filled dewar for, e.g., about 3 minutes. Following, the container was removed from the liquid nitrogen dewar, and the lid of the container was opened. The tube was then covered with paper (e.g., a lint-free paper) and tightened, e.g., with a rubber band. The resulting tube of frozen polymeric nanoparticles was transferred to a Fast Freeze Flask and attached to the lyophilizer (e.g., Labconco, 2.5 L, -50 °C, bench top).
  • lyophilizer e.g., Labconco, 2.5 L, -50 °C, bench top.
  • the lyophilizer prior to the lyophilization step, was pre-set to be equilibrated to a temperature of about -50 °C at a pressure of about 0.040 mBar.
  • the polymeric nanoparticles comprising rapamycin were kept coupled to the lyophilizer (e.g., for about 66 hours) before measuring their cross-sectional dimensions (e.g., diameter).
  • saline e.g., about ImL of lx PBS
  • DSL dynamic light scattering
  • the nanoparticles e.g., about 1 mg
  • deionized water e.g., about 1 mL
  • Lyophilization can change the average cross-sectional dimension of the population of nanoparticles of the disclosure. For example, once lyophilized and resuspended (e.g., in aqueous solution), the average cross-sectional dimension of the population of nanoparticles can increase. In another example, once lyophilized and resuspended (e.g., in aqueous solution), the average cross-sectional dimension of the population of nanoparticles can decrease. In some cases, a presence of a 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. In some cases, therapeutic efficacy of the population of nanoparticles can be highly dependent on the average cross-sectional dimension of the population of nanoparticles, and thus substantially maintain the average cross-sectional dimension throughout storage or preparation steps is desirable.
  • a hydration agent as disclosed herein (
  • PLGA-PEG nanoparticles were synthesized as described herein.
  • the nanoparticles were synthesized as (i) blank mesoscale nanoparticles (e.g., blank MNPs without any medical agent encapsulated), (ii) MNPs with encapsulated 3,3 '-di ethylthiadicarbocyanine iodide (DEDC) dye, or (iii) MNPs with encapsulated rapamycin (e.g., for regulating mTOR signaling in a cell).
  • DEDC 3,3 '-di ethylthiadicarbocyanine iodide
  • MNPs with encapsulated rapamycin e.g., for regulating mTOR signaling in a cell.
  • Each of the population of MNPs were collected in a container and spun down.
  • the population of MNPs resuspended in the solution with or without the dehydrating agent were frozen by dipping the container into liquid nitrogen filled dewar for, e.g., about 3 minutes.
  • the container was removed from the liquid nitrogen dewar, and the lid of the container was opened.
  • the tube as then covered with paper and tightened, e.g., with a rubber band.
  • the resulting tube of frozen polymeric nanoparticles was transferred to a Fast Freeze Flask and attached to the lyophilizer.
  • the populations of MNPs were stored before being re-dissolved in aqueous solution (e.g., water or saline) for measuring their cross-sectional dimensions (e.g., diameter).
  • aqueous solution e.g., water or saline
  • FIG. 1 shows the effect of the dehydrating agent (e.g., sucrose, trehalose) to the change of the average cross-sectional dimension (i.e., size (nm)) of the population of MNPs upon lyophilization.
  • Lyophilization of the population of MNPs in 5 wt.% sucrose reduced aggregation of the MNPs, as compared to using less than 5 wt.% sucrose.
  • Lyophilization of the population of MNPs in 5 wt.% sucrose resulted in more consistent (or uniform) particle size, as compared to using less than 5 wt.% sucrose.
  • the presence of sucrose at 5 wt.% or more can provide more hydrogen bonds during lyophilization, thereby reducing or preventing the aggregation of the MNPs.
  • FIG. 2 shows additional data demonstrating that the average cross-sectional dimension remained substantially the same when lyophilized in the presence of 5% sucrose as the hydrating agent.
  • mTOR indicates a presence of encapsulated rapamycin in the population of MSPs.
  • a population of nanoparticles can be prepared for use as delivery carriers for medical agents, such as therapeutics agents (e.g., drugs, such as rapamycin) or imaging agents (e.g., dyes).
  • the population of nanoparticles can comprise polymeric nanoparticles, such as PLGA-PEG nanoparticles.
  • the population of nanoparticles, either with or without loaded medical agents can be administered to animals (e.g., mice, rats, rabbits, sheep, pigs, non-human primates, etc.), and different organs of the animals can be analyzed to determine differential biodistribution of the population of nanoparticles in specific organs or cell types.
  • the biodistribution study of the population of nanoparticles disclosed herein can be performed in in healthy or diseased mice (e.g., from about 2-month-old to about 3-month-old C57BL/6 mice) or healthy or diseased rats (e.g., from about 3-week-old to about 7-week-old rats).
  • the diseased mice model is polycystic kidney disease (PKD) mouse model.
  • PPD polycystic kidney disease
  • the population of nanoparticles can be PLGA-PEG nanoparticles encapsulating imaging agents.
  • the imaging agents can be fluorescent dyes, in which case biodistribution of the population of nanoparticles can be assessed via fluoresce imaging of the dyes.
  • the imaging agents can be radioactive labels, in which case biodistribution of the population of nanoparticles can be accessed via positron emission tomography (PET) scanning.
  • PET positron emission tomography
  • the nanoparticles are modified with chelators (e.g., deferoxamine (DFO)) capable of chelating the radioactive labels.
  • DFO deferoxamine
  • Groups of animals e.g., mice
  • a composition comprising a population of PLGA-PEG nanoparticles with encapsulated dyes (e.g., 50 mg/kg of nanoparticles per weight of the animal).
  • Control group animals are each injected with the same dyes in PBS with 0.5% DMSO (equal to the amount of the dyes in particle-injected animal).
  • the animals are imaged dorsally with an IVIS Spectrum Pre-clinical In vivo Imaging System (Perkin Elmer; Waltham, Mass.) using recommended excitation and emission filters for the dyes to determine fluorescence biodistribution at the following times post-injection: 30 minutes, 1 hour, 4 hours, 8 hours, 24 hours, 48 hours, 72 hours, and 120 hours.
  • IVIS Spectrum Pre-clinical In vivo Imaging System Perkin Elmer; Waltham, Mass.
  • ROIs regions of interest
  • Groups of animals are each injected intravenously via the tail vein with a composition comprising a population of PLGA-PEG-DFO nanoparticles with radiolabels chelated by the DFO of each nanoparticle (e.g., 50 mg/kg of nanoparticles per weight of the animal).
  • Control group animals are each injected with the same radiolabel (e.g., chelated by non- nanoparticle bound DFO) in PBS with 0.5% DMSO (equal to the amount of the dyes in particle- injected animal).
  • the animals are imaged dorsally with a PET scanner to determine radiolabel biodistribution at the following times post-injection: 1 hour, 4 hours, 8 hours, 24 hours, 48 hours, 72 hours, and 120 hours.
  • In vivo PET images are analyzed with regions of interest (ROIs) selected around each kidney and the central lung region.
  • ROIs regions of interest
  • a population of nanoparticles can be prepared for use as delivery carriers for medical agents, such as therapeutics agents (e.g., drugs, such as rapamycin) or imaging agents (e.g., dyes).
  • the population of nanoparticles can comprise polymeric nanoparticles, such as PLGA-PEG nanoparticles.
  • Medical agents such as therapeutics agents (e.g., drugs, such as rapamycin) or imaging agents (e.g., dyes) can be administered to a subject in either (i) the population of nanoparticles of the disclosure or (ii) aqueous vehicle (e.g., saline). Subsequently, systemic concentrations of the therapeutic agents over time can be measured and utilized to determine mean pharmacokinetic (PK) parameters (e.g., AUCo -24, Cmax, Tmax, TI/2, skewness, bioavailability, etc.).
  • PK pharmacokinetic
  • Rapamycin standard curve A standard curve of soluble rapamycin was measured to generate a standard curve of rapamycin concentration. Rapamycin was diluted to 20 mg/mL in acetonitrile (ACN), agitated by vortex for a few minutes, and spun down at 33000 RCF for 5 minutes. A supernatant was removed, then analyzed via reversed phase (RP) HPLC. An area of a peak indicative of the molecular weight of rapamycin was measured and plotted as a function of the rapamycin concentrations.
  • ACN acetonitrile
  • RP reversed phase
  • concentrations of rapamycin used to generate the standard curve were about 0.15625 pg/mL, about 0.3125 pg/mL, about 0.625 pg/mL, about 1.25 pg/mL, about 2.5 pg/mL, about 5 pg/mL, about 10 pg/mL, and about 20 pg/mL.
  • FIG, 3 shows the standard curve of rapamycin.
  • Rapamycin is administered to animals (e.g., mice or rats) either as (i) encapsulated in the population of nanoparticles of the present disclosure or (ii) non-encapsulated (e.g., in saline).
  • systemic concentrations of rapamycin over time are obtained by: (1) obtaining plasma samples from the subject at multiple time points (e.g., 1 minutes, 30 minutes, 60 minutes, 2 hours, 3 hours, 4 hours, 5 hours, 6 hours, 7 hours, 8 hours, 9 hours, 10 hours, 11 hours, 12 hours, 14 hours, 16 hours, 18 hours, 20 hours, 22 hours, 24 hours, etc.); and (2) measuring the concentration of rapamycin of interest in the obtained plasma samples (e.g., via HPLC detection and calculation via the working curve, as abovementioned).
  • time points e.g., 1 minutes, 30 minutes, 60 minutes, 2 hours, 3 hours, 4 hours, 5 hours, 6 hours, 7 hours, 8 hours, 9 hours, 10 hours, 11 hours, 12 hours, 14 hours, 16 hours, 18 hours, 20 hours, 22 hours, 24 hours, etc.
  • a population of nanoparticles encapsulating therapeutic agents can be administered to a diseased animal model to study efficacy of therapeutic agents delivered with or without the population of nanoparticles.
  • the purpose is to evaluate the effects of rapamycin delivered by the nanoparticles (e.g., PLGA-PEG nanoparticles) on the rate of renal cystic disease progression in the outbred PkhdlPCK (PCK) rat model of PKD.
  • Rapamycin e.g., encapsulated in the nanoparticles disclosed herein, or nonencapsulated
  • the rats are sacrificed at multiple time points (e.g., 6 hours, 24 hours, 48 hours, and 72 hours) to assess the extent to which mTOR signaling in cells of the rats (e.g., renal cells) is reduced or inhibited.
  • expression of protein phosphorylated ribosomal protein s6 (pS6) can be measured by western blot analysis of extracted kidney tissue samples from treated animals.
  • Rapamycin e.g., encapsulated in the nanoparticles disclosed herein, or nonencapsulated
  • PKD model rats are administered to the PKD model rats for eight weeks until p77 (11 weeks of age).
  • the rapamycin can be administered to the PKD model rats four times per week.
  • One or two control arms are monitored. All animals are imaged by MRI again at the end of the study. Immediately after the final MRI, terminal collections of blood and various organs are performed. At termination, body weight and length, kidney weights and lengths, and brain weight are recorded. Whole blood is collected for serum separation.
  • Liver and kidney samples are be collected in formaldehyde for histomorphometry and a separate set of frozen liver and kidney samples can be used for protein, RNA, DNA, tissue exposures and other assays. Serum are be analyzed for creatinine via mass spectrometry. The timeline of the in vivo efficacy study is schematically illustrated in FIG. 4.
  • the treatment with rapamycin via the population of nanoparticles as disclosed herein can inhibit a pharmacological target (e.g., pS6 protein) in the kidneys of the rats.
  • a pharmacological target e.g., pS6 protein
  • inhibition of the pharmacological target can be significantly higher in the animals receiving nanoparticle-encapsulated rapamycin as compared to the animals receiving rapamycin in solution.
  • inhibition of the pharmacological target can last longer in the animals receiving nanoparticle-encapsulated rapamycin as compared to the animals receiving rapamycin in solution.
  • a significantly higher dose of rapamycin in solution can be required as compared to a therapeutic dose of rapamycin required when encapsulated in the nanoparticles.
  • siRNA Small interfering ribonucleic acid
  • poly(amino acid) excipient such as poly-l-lysine, poly-l-arginine, or poly-l-histidine
  • siRNAs can be mixed with the poly(amino acid) excipient(s) at a specific ratio of (i) a phosphates in the backbone of siRNA to (ii) amine groups in the backbone of poly(amino acid) excipient.
  • DI deionized
  • the siRNA and the poly(amino acid) excipient can be mixed at pH ranging between 3.5 and 8.
  • the siRNA and the poly(amino acid) excipient can be allowed to mix (e.g., complex) for between 5 mins to 30 mins, by adding either a siRNA solution to a poly(amino acid) excipient solution or by adding a poly(amino acid) excipient solution to a siRNA solution.
  • lipid excipients e.g., DC cholesterol, DOTAP, DODAP, DSPC, DSPE PEG
  • polymeric excipients e.g., PEI, PDMAEMA, chitosan
  • excipients can be added to or mixed with PLGA or PLGA-PEG in a solvent (e.g., acetonitrile, acetone, ethanol, etc.) at a different ratio of PLGA or PLGA PEG, and then siRNA in RNAse free water or a buffer as disclosed herein can be added to the resulting solution comprising PLGA or PLGA PEG and the excipient.
  • a solvent e.g., acetonitrile, acetone, ethanol, etc.
  • siRNA in RNAse free water or a buffer as disclosed herein can be added to the resulting solution comprising PLGA or PLGA PEG and the excipient.
  • the mixture comprising the siRNA and the poly(amino acid) excipient can be added dropwise to already prepared solution of PLGA-PEG (e.g., 50 mg/mL) in acetonitrile (e.g., 2 mL).
  • PLGA-PEG e.g., 50 mg/mL
  • acetonitrile e.g. 2 mL
  • the resultant primary emulsion can be sonicated, e.g., for between 2 and 5 minutes.
  • a Hamilton glass syringe e.g., a 10 mL Hamilton glass syringe
  • a solution e.g., about 2 mL
  • the syringe can be secured on a syringe pump.
  • the solution comprising primary emulsion can be added into a solution comprising deionized water (DEPC treated water or DI water or RNAse/ DNAse free) and F-68 at a speed of 0.1 mL per minute (e.g., for double emulsion).
  • polymeric nanoparticles comprising siRNA can be collected in a container (e.g., a 50 mL conical tube) and spun down by using ultra-centrifugation at, e.g., 7356 relative centrifugal force (RCF) for about 15 minutes at 4 °C. Subsequently, supernatant can be discarded, and the polymeric nanoparticles can be resuspended in deionized water (e.g., about 10 mL) and spun down, e.g., via ultra-centrifugation as above mentioned.
  • a container e.g., a 50 mL conical tube
  • RCF relative centrifugal force
  • sucrose e.g., about 5% sucrose by weight or in a range of 2% sucrose to 10% sucrose by weight of other cryoprotectant such as trehalose, mannitol, hydrodypropyl betacyclodextrin in a same weight percentage (2%-10%)).
  • nanoprecipitation of the mixture of PLGA-PEG and siRNA/excipient can be performed using a homogenizer (e.g., a high shear homogenizer, such as Microfluidizer®).
  • a homogenizer e.g., a high shear homogenizer, such as Microfluidizer®
  • the mixture can enter into a high shear homogenizer via an inlet reservoir and can be pushed through a fixed geometry interaction chamber at up to, for example, 30,000 psi (2068 bar), via a constant pressure pumping system.
  • the mixture can experience consistent, high shear rates, and impact forces.
  • Such treatment of the mixture can generate the plurality of nanoparticles (e.g., MNPs).
  • the plurality of nanoparticles as disclosed herein can be temperature controlled during its manufacture, e.g., via passing through a temperature controlled interaction chamber.
  • precipitation of the mixture of PLGA-PEG and siRNA/excipient can be performed using a microfluid channel system (e.g., NanoAssemblr®).
  • a solution comprising PLGA-PEG can be mixed with another solution comprising siRNA and the excipient in a microfluidic cartridge. Rapid, homogeneous mixing of the two solutions can ensure that particles are formed under consistent conditions. Computer controlled independent injection of both solutions can allow mixing speed and/or mixing ratio to be controlled, thereby to systematically optimize particle formation parameters.
  • the polymeric nanoparticles resuspended in a solution e.g., having sucrose in an amount ranging between 2% and 10%, and optionally further comprising one or more cryoprotectants, such as trehalose, mannitol, hydrodypropyl betacyclodextrin, etc., in an amount ranging between 2% and 10%
  • a solution e.g., having sucrose in an amount ranging between 2% and 10%, and optionally further comprising one or more cryoprotectants, such as trehalose, mannitol, hydrodypropyl betacyclodextrin, etc., in an amount ranging between 2% and 10%
  • cryoprotectants such as trehalose, mannitol, hydrodypropyl betacyclodextrin, etc.
  • the tube can be then covered with paper (e.g., a lint-free paper) and tightened, e.g., with a rubber band.
  • the resulting tube of frozen polymeric nanoparticles can be transferred to a Fast Freeze Flask and attached to the lyophilizer (e.g., Labconco, 2.5 L, -50 °C, bench top).
  • the lyophilizer e.g., Labconco, 2.5 L, -50 °C, bench top.
  • the lyophilizer prior to the lyophilization step, can be pre-set to be equilibrated to a temperature of about -50 °C at a pressure of about 0.040 mBar.
  • the polymeric nanoparticles comprising siRNA and excipient can be kept coupled to the lyophilizer (e.g., for about 42 hours) before measuring their cross-sectional dimensions (e.g., diameter).
  • dry nanoparticles e.g., about 10 mg
  • phosphate buffer saline e.g., about ImL of lx PBS
  • the saline solution comprising the nanoparticles can be transferred to an appropriate cuvette (e.g., a size measurement cuvette) and hydrodynamic diameter of the siRNA-loaded nanoparticles can be measured by a dynamic light scattering (DSL) instruction (e.g., Malvern Zetasizer).
  • DSL dynamic light scattering
  • the nanoparticles e.g., about 1 mg
  • deionized water e.g., about 1 mL
  • the resulting solution of polymer excipient can be bath sonicated (e.g., for 10 minutes).
  • 125 microliters (pL) or 100 pL of the polymer solution can be slowly added to an equal volume of siRNA solution (e.g., 125 pL comprising 250 pg of siRNA or 100 pL comprising 200 pg of siRNA).
  • the resulting solution can be mixed (e.g., by pipetting up and down), then incubated (e.g., at room temperature for 5 minutes) for complexation.
  • the mixture of siRNA and the polymer excipient can be added dropwise to the vial containing PLGA-PEG (e.g., 110 mg or 100 mg in 2 mL of acetonitrile).
  • the vial can be exposed to a batch sonication process (e.g., for 2 minutes) before synthesizing secondary emulsion.
  • HK-2 cells human kidney cortex/medulla cells
  • WT 9-12 cells kidney; cyst from a distal and proximal cortical tubule
  • RK3E cells rat kidney cells
  • nanoparticles e.g., siRNA-excipient nanoparticles as disclosed herein
  • RNA e.g., siRNA concentration
  • nM nanomolar
  • pM micromolar
  • RNA from the cells can be isolated, e.g., by using Invitrogen PureLink RNA Mini Kit and qRT-PCR was performed, e.g., by using iTaq Univer SYBR Green 1-Step Kt 100 Rx.
  • siRNA-excipient loaded nanoparticles can induce gene knockdown of a target gene in a range between about 5% and about 100%.
  • the siRNA-excipient nanoparticles can exhibit enhanced gene knockdown as compared to siRNA nanoparticles without the excipient.
  • cells treated with the siRNA-excipient loaded nanoparticles and controls can be isolated and expression of one or more target proteins can be evaluated, e.g., by Western blot analysis as compared to a control protein (e.g., GAPDH, actin, alpha tubulin, etc.).
  • a control protein e.g., GAPDH, actin, alpha tubulin, etc.
  • Treatment of the siRNA-excipient loaded nanoparticles can induce reduction of the target protein expression by a level ranging between about 10% to about 100%.
  • excipients e.g., cationic poly(amino acid) polymers or lipids
  • excipients can be utilized for delivery of various nucleic acid molecules, such as, e.g., siRNA delivery, micro RNA (miRNA) delivery, plasmid DNA delivery, or messenger RNA (mRNA) delivery.
  • miRNA micro RNA
  • mRNA messenger RNA
  • similar in vitro assays as abovementioned in Example 2 can be performed to evaluate knockdown of one or more genes, as evaluated by assessing expression of target mRNAs or proteins, imaging (e.g., microscopy), flow cytometry, or measuring relative luminescence assay of reporter gene and/or protein.
  • a population of nanoparticles encapsulating above-mentioned nucleic acid therapeutics agents and excipients can be administered to a diseased animal model or nondiseased animal model to study efficacy of the therapeutic agents delivered with or without the population of nanoparticles or excipients.
  • the effects of nucleic acid delivery by the nanoparticles e.g., PLGA-PEG nanoparticles with excipients
  • a rate of renal cystic disease progression e.g., PLGA-PEG nanoparticles with excipients
  • a level of renal clearance biomarkers e.g., a level of renal clearance biomarkers
  • a modified expression level of a target protein or gene e.g., a modified expression level of a target protein or gene.
  • RNA can be isolated from cells (e.g., using commercially available kits).
  • the expression level of target gene mRNA or protein can be evaluated by qRT-PCR and/or by Western blot to evaluate bioluminescence of protein via live or ex vivo imaging at a different time point after injection (intravenous or intraperitoneal) of nucleic acid loaded nanoparticles.
  • nucleic acid-excipient e.g., siRNA-excipient
  • Example 10 Emulsion process for producing a plurality of particles
  • a polymer e.g., PLGA-PEG polymer
  • a homogenizer to synthesize particles (e.g., in the range between 100 nanometers and about 6 micrometers in diameter) (i.e., an emulsion process).
  • the homogenization can yield, for example, primary emulsion (e.g., oil in water) to produce a plurality of particles that encapsulate medical agents (e.g., small molecule therapeutic payload such as Rapamycin, Edaravone, MK2206 and Dye such as DEDC, Rhodamine, Cy5 etc.).
  • medical agents e.g., small molecule therapeutic payload such as Rapamycin, Edaravone, MK2206 and Dye such as DEDC, Rhodamine, Cy5 etc.
  • the emulsion process can utilize hydrophobic ion paring method to pair cationic charged medical agent (e.g., MK2206) to anionic charged moiety (e.g., excipient), such as pamoic acid, oleic acid and cholic acid, in hydrophobic solution of benzyl alcohol and ethyl acetate (BA:EA) or other organic solvent such as DMF, acetonitrile, acetone etc.
  • cationic charged medical agent e.g., MK2206
  • anionic charged moiety e.g., excipient
  • BA:EA ethyl acetate
  • other organic solvent such as DMF, acetonitrile, acetone etc.
  • the emulsion process can utilize stabilizers/emulsifier such as poly vinyl alcohol, F68 (pluronics), sodium cholate in aqueous core (percentage from 0.1% to 5%) to stabilize nanoparticles to finally formed Oil in water emulsion.
  • stabilizers/emulsifier such as poly vinyl alcohol, F68 (pluronics), sodium cholate in aqueous core (percentage from 0.1% to 5%) to stabilize nanoparticles to finally formed Oil in water emulsion.
  • PLGA-PEG polymer e.g., PLGA (40kDA) and PEG (5kDA)
  • PLGA (40kDA) and PEG (5kDA) can be used from commercial suppliers, or can be synthesized as mentioned below.
  • Precipitated polymer was washed about 3 times in total 10 mL (50:50) mixture of cold methanol (Sigma Aldrich, Cat. No. 34860-1L-R, Lot No. SHBL4287) and diethyl ether in a slow stream.
  • Waste was discarded and polymer was vacuum dried for around 4 hours.
  • Hydrophobic polymer PLGA-PEG (e.g., 30mg, 50mg, 80 mg or 100 mg) was dissolved in about 1 mL to 3 mL of solution of ethyl acetate (EA) and benzyl alcohol (BA) (e.g., at a volume to volume ratio of EA to BA (EA:BA) from 0 to 100, 10 to 90, 20 to 80, 30 to 70, 70 to 30, 20 to 80, etc.)
  • Free base of MK2206 small molecule (purchased from Wuxi App tech) in quantities of 10 mg, 20 mg, 30 mg, or more was dissolved in above solution in presence of oleic acid, cholic acid or pamoic acid (e.g., at a molar ratio of oleic acid to MK2206 at about 1 : 1, 1 :5, 1 : 10, 1 :20, 1 :50, etc.)
  • aqueous solution e.g., 3 mL or 5mL or 10 mL
  • deionized water e.g., 2% or 5% or 0.1% of sodium cholate, poly vinyl alcohol, or F68
  • flow rate was 6 mL per min to 9 mL per min or just by hand mixing.
  • probe sonicator was also used (pulse or continuous) from 5 seconds to 2 minutes.
  • batch sonication was used for 1 min to 5 mins.
  • Rhodamine Rhodamine PLGA, Rapamycin, or Edaravone was used in place of MK2206.
  • Emulsion was quenched by adding cold solution of 0.1% to 2% of PVA, or 0.1% to 10% of Tween.
  • a volume of aqueous solution used for quenching was from 10 mL to 300 mL (e.g., such that the concentration of organic was from 1% to 20%).
  • Rotor-stator type homogenizer comprises a rotor with two or more blade knife at the bottom and a stator with a vertical slot around the wall of the homogenizer cell.
  • VWR micro homogenizer model 200 comprises a rotor that rotates at a speed between 5000 RPM and 35000 RPM. As rotor rotates, it generates a vacuum to draw liquid in an out of the assembly, thereby resulting in circulation.
  • FIG. 7 schematically illustrates a rotor-stator configuration.
  • the emulsion process may have advantages as compared to precipitation process (e.g., nanoprecipitation).
  • various parameters involved in the emulsion process e.g., speed of the rotor knife, homogenization time, the types of organic solvents, the concentration of polymer and/or therapeutics, ratio of organic and aqueous phase
  • particles with a range of hydrodynamic diameter e.g., between aboutlOO nanometers to about 5 micrometers
  • a higher drug loading to provide a greater flexibility and a superior robustness that may not be achievable to the same degree by the nanoprecipitation process.
  • the nanoprecipitation process can be limited by the concentration of polymer in which 50 mg/ mL polymer and therapeutics in 2 mL of acetonitrile is added dropwise to the 4 mL aqueous mixture of stabilizer/ emulsifier were polymer and hydrophobic therapeutics self-assembled into tiny droplets of mesoscale nanoparticles.
  • size of nanoparticles can be completely dependent on the concentration of polymer.
  • the nanoprecipitation process can provide a limited bandwidth with respect to the organic solvents where one must use organic solvents which are miscible with water or aqueous media.
  • Drug loading in the mesoscale nanoparticles can be assessed, e.g., via High- performance liquid chromatography (HPLC), to evaluate drug loading on this batch.
  • HPLC High- performance liquid chromatography
  • the emulsion process as disclosed herein can enhance the drug loading of Edaravone per mg of nanoparticles, e.g., by at least about 1%, 2%, 5%, 10%, 15%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or more.
  • Free base of MK2206 was incubated with fatty acid molecule such as oleic acid, as described in Section 2 of Example 10, in the mixture of ethyl acetate and benzyl alcohol (EA:BA ratio from 10:90 to 90: 10) at room temperature (RT).
  • MK22006 has primary and secondary amine nitrogen from pyridine ring could electrostatically forms ion pair with -COOH of oleic acid (as shown in FIG. 9).
  • Mixture of MK2206 and Oleic acid e.g., in presence of PLGA PEG polymer
  • MK2206 with oleic acid was encapsulated in emulsion particles as abovementioned.
  • MK2206 loaded particles with a desired size range, as shown in Table 5, were produced.
  • Particles as disclosed herein were washed using three different methods: [0244] (1) Dialysis method: particles were dialyzed against 5% sucrose, 0.1% PVA or IX
  • Centrifugal filtration units particles were filtered using lOkDa or 30kDa or 100 kDa ultra centrifugal filtration units Pall Laboratories or Millipore Sigma at 1500 RCF to 4000 RCF; and/or
  • Resuspended nanoparticles in 5% sucrose and or 5% hydroxy propyl beta cyclodextrin, 10 mL were frozen by dipping 50 mL conical tube into liquid nitrogen filled dewar for 3 minutes.
  • the tube of frozen nanoparticles was transferred to 600 mL Fast Freeze Flask and attached to the lyophilizer (Labconco, 2.5 L, -50 °C, bench top).
  • the lyophilizer Prior to above step, the lyophilizer was allowed to equilibrate to a temperature of -50 °C and pressure at -0.040 mBar.
  • the nanoparticles were kept in the lyophilizer (e.g., for around 66 hours) before measuring their average size.
  • Nanoparticles were spun down at high speed (e.g., 14000 RCF) to collect polymer debris and supernatant was collected for further analysis.
  • Standard solution of Edaravone or MK2206 or Rhodamine was prepared in same organic solvent and quantities of small molecule was determined using UV-Vis spectrophotometer.
  • Embodiment AL A method of manufacturing a nanoparticle composition, the method comprising:
  • Embodiment A2 The method of embodiment Al, wherein the polysaccharide is present in the mixture in an amount that is at least about 5 % by mass of the mixture.
  • Embodiment A3 The method of any one of the embodiments A1-A2, wherein the polysaccharide comprises sucrose.
  • Embodiment A4 The method of any one of the embodiments A1-A2, wherein the polysaccharide comprises trehalose.
  • Embodiment A5 The method of any one of the embodiments A1-A2, wherein the polysaccharide comprises 2 hydroxy propyl beta-cyclodextrin.
  • Embodiment A6 The method of any one of the embodiments A1-A5, wherein the average temperature is reduced by at least about 20 °C.
  • Embodiment A7 The method of any one of the embodiments A1-A5, wherein the average temperature is reduced by at least about 40 °C.
  • Embodiment A8 The method of any one of the embodiments A1-A7, wherein (b) comprises substantially freezing the nanoparticle composition.
  • Embodiment A9 The method of any one of the embodiments A1-A8, wherein the mixture further comprises an aqueous medium, and wherein the method further comprises, subsequent to (b), removing at least 50% of the aqueous medium by volume from the nanoparticle composition via sublimation.
  • Embodiment A10 The method of any one of the embodiments A1-A9, further comprising, subsequent to (b), substantially lyophilizing the nanoparticle composition.
  • Embodiment Al l The method of any one of the embodiments A1-A10, wherein the medical agent comprises a small molecule drug.
  • Embodiment A12 The method of any one of the embodiments A1-A10, wherein the medical agent comprises Rapamycin.
  • Embodiment Al 3. The method of any one of the embodiments A1-A10, wherein the medical agent comprises MK2206 or a pharmaceutically acceptable salt thereof.
  • Embodiment A14 The method of any one of the embodiments A1-A10, wherein the medical agent comprises Edaravone or a pharmaceutically acceptable salt thereof.
  • Embodiment Al 5 The method of any one of the embodiments A1-A10, wherein the medical agent comprises a nucleic acid molecule.
  • Embodiment Al 6 The method of any one of the embodiments A1-A10, wherein the medical agent comprises a siRNA.
  • Embodiment Al 7 The method of any one of the embodiments Al -Al 6, wherein an average nanoparticle size of the plurality of nanoparticles is between about 100 nanometers and 600 nanometers.
  • Embodiment Bl A nanoparticle composition comprising a plurality of nanoparticles and a mixture, wherein a medical agent is encapsulated by a nanoparticle of the plurality of nanoparticles, wherein the mixture comprises a polysaccharide, wherein the polysaccharide is present in the mixture in an amount that is greater than 2 % and lesser than 10 % by mass of the mixture, and wherein an average temperature of the nanoparticle composition is less than about 5 °C.
  • Embodiment B2 The composition of embodiment Bl, wherein the nanoparticle composition is substantially frozen.
  • Embodiment B3 The composition of any one of the embodiments B1-B2, wherein the polysaccharide is present in the mixture in an amount that is at least about 5 % by mass of the mixture.
  • Embodiment B4 The composition of any one of the embodiments B1-B3, wherein the polysaccharide comprises sucrose.
  • Embodiment B5. The composition of any one of the embodiments B1-B3, wherein the polysaccharide comprises trehalose.
  • Embodiment B6 The composition of any one of the embodiments B1-B3, wherein the polysaccharide comprises 2 hydroxy propyl beta-cyclodextrin.
  • Embodiment B7 The composition of any one of the embodiments B1-B6, wherein the medical agent comprises a small molecule drug.
  • Embodiment B8 The composition of any one of the embodiments B1-B6, wherein the medical agent comprises Rapamycin.
  • Embodiment B9 The composition of any one of the embodiments B1-B6, wherein the medical agent comprises MK2206 or a pharmaceutically acceptable salt thereof.
  • Embodiment B10 The composition of any one of the embodiments B1-B6, wherein the medical agent comprises Edaravone or a pharmaceutically acceptable salt thereof.
  • Embodiment Bl 1. The composition of any one of the embodiments B1-B6, wherein the medical agent comprises a nucleic acid molecule.
  • Embodiment B 12 The composition of any one of the embodiments B1-B6, wherein the medical agent comprises a siRNA.
  • Embodiment B 13 The composition of any one of the embodiments Bl -Bl 2, wherein an average nanoparticle size of the plurality of nanoparticles is between about 100 nanometers and 600 nanometers.
  • Embodiment Cl A method of manufacturing a nanoparticle composition, the method comprising: contacting a plurality of nanoparticles with a mixture, thereby forming the nanoparticle composition, wherein a nanoparticle of the plurality of nanoparticles comprises a medical agent, and wherein the mixture comprises a first polysaccharide and a second polysaccharide [0293] Embodiment C2. The method of embodiment Cl, wherein an amount of a combination of the first polysaccharide and the second polysaccharide in the mixture is between about 1% and about 30% by mass of the mixture.
  • Embodiment C3 The method of embodiment Cl, wherein an amount of a combination of the first polysaccharide and the second polysaccharide in the mixture is between about 2% and about 20% by mass of the mixture.
  • Embodiment C The method of any one of the embodiments C1-C3, wherein one of the first polysaccharide and the second polysaccharide is sucrose.
  • Embodiment C5. The method of any one of the embodiments C1-C3, wherein one of the first polysaccharide and the second polysaccharide is trehalose.
  • Embodiment C6 The method of any one of the embodiments C1-C3, wherein one of the first polysaccharide and the second polysaccharide is 2 hydroxy propyl beta-cyclodextrin.
  • Embodiment C7 The method of any one of the embodiments C1-C6, further comprising reducing an average temperature of the nanoparticle composition.
  • Embodiment C8 The method of any one of the embodiments C1-C7, wherein the average temperature is reduced by at least about 20 °C.
  • Embodiment C9 The method of any one of the embodiments C1-C7, wherein the average temperature is reduced by at least about 40 °C.
  • Embodiment CIO The method of any one of the embodiments C1-C9, further comprising substantially freezing the nanoparticle composition.
  • Embodiment Cl 1. The method of any one of the embodiments Cl -CIO, wherein the mixture further comprises an aqueous medium, and wherein the method further comprises removing at least 50% of the aqueous medium by volume from the nanoparticle composition via sublimation.
  • Embodiment C12 The method of any one of the embodiments Cl-Cl 1, further comprising substantially lyophilizing the nanoparticle composition.
  • Embodiment C13 The method of any one of the embodiments Cl -Cl 2, wherein the medical agent comprises a small molecule drug.
  • Embodiment C14 The method of any one of the embodiments C1-C12, wherein the medical agent comprises Rapamycin.
  • Embodiment Cl 5 The method of any one of the embodiments Cl -Cl 2, wherein the medical agent comprises MK2206 or a pharmaceutically acceptable salt thereof.
  • Embodiment C16 The method of any one of the embodiments C1-C12, wherein the medical agent comprises Edaravone or a pharmaceutically acceptable salt thereof.
  • Embodiment C17 The method of any one of the embodiments C1-C12, wherein the medical agent comprises a nucleic acid molecule.
  • Embodiment Cl 8 The method of any one of the embodiments Cl -Cl 2, wherein the medical agent comprises a siRNA.
  • Embodiment Cl 9 The method of any one of the embodiments Cl -Cl 8, wherein an average nanoparticle size of the plurality of nanoparticles is between about 100 nanometers and 600 nanometers.
  • Embodiment DI A nanoparticle composition comprising a plurality nanoparticles in a mixture, wherein a nanoparticle of the plurality of nanoparticles comprises a medical agent, and wherein the mixture comprises a first polysaccharide and a second polysaccharide.
  • Embodiment D2 The composition of embodiment DI, wherein an amount of a combination of the first polysaccharide and the second polysaccharide in the mixture is between about 1% and about 30% by mass of the mixture.
  • Embodiment D3 The composition of embodiment DI, wherein an amount of a combination of the first polysaccharide and the second polysaccharide in the mixture is between about 2% and about 20% by mass of the mixture.
  • Embodiment D4 The composition of any one of the embodiments D1-D3, wherein one of the first polysaccharide and the second polysaccharide is sucrose.
  • Embodiment D5 The composition of any one of the embodiments D1-D3, wherein one of the first polysaccharide and the second polysaccharide is trehalose.
  • Embodiment D6 The composition of any one of the embodiments D1-D3, wherein one of the first polysaccharide and the second polysaccharide is 2 hydroxy propyl betacyclodextrin.
  • Embodiment D7 The composition of any one of the embodiments D1-D6, wherein an average temperature of the nanoparticle composition is less than about 5 °C.
  • Embodiment D8 The composition of any one of the embodiments D1-D7, wherein the nanoparticle composition is substantially frozen.
  • Embodiment D9 The composition of any one of the embodiments D1-D8, wherein the medical agent comprises a small molecule drug.
  • Embodiment D10 The composition of any one of the embodiments D1-D8, wherein the medical agent comprises Rapamycin.
  • Embodiment Dl l The composition of any one of the embodiments D1-D8, wherein the medical agent comprises MK2206 or a pharmaceutically acceptable salt thereof.
  • Embodiment D12 The composition of any one of the embodiments D1-D8, wherein the medical agent comprises Edaravone or a pharmaceutically acceptable salt thereof.
  • Embodiment D13 The composition of any one of the embodiments D1-D8, wherein the medical agent comprises a nucleic acid molecule.
  • Embodiment D14 The composition of any one of the embodiments D1-D8, wherein the medical agent comprises a siRNA.
  • Embodiment DI 5 The composition of any one of the embodiments DI -DI 4, wherein an average nanoparticle size of the plurality of nanoparticles is between about 100 nanometers and 600 nanometers.
  • Embodiment El A method of manufacturing a plurality of nanoparticles, the method comprising: (a) incubating a mixture for at least about 1 hour, wherein the mixture comprises (i) a medical agent, (ii) an excipient that is oppositely charged from the medical agent, and (iii) an encapsulation agent; and
  • Embodiment E2 The method of embodiment El, further comprising, prior to (a), mixing the medical agent, the excipient, and the encapsulation agent to form the mixture.
  • Embodiment E3 The method of any of embodiments E1-E2, wherein a nanoparticle of the plurality of nanoparticles comprises the medical agent and the encapsulation agent.
  • Embodiment E4 The method of any of embodiments E1-E3, wherein the mixture is incubated for at least about 6 hours.
  • Embodiment E5. The method of any of embodiments E1-E3, wherein the mixture is incubated for at least about 12 hours.
  • Embodiment E6 The method of any of embodiments E1-E5, wherein the incubation is at a temperature of at least about 3 °C.
  • Embodiment E7 The method of any of embodiments E1-E5, wherein the incubation is at a temperature of at least about 20 °C.
  • Embodiment E8 The method of any of embodiments E1-E7, wherein the mixture further comprises a protic solvent.
  • Embodiment E9 The method of any of embodiments E1-E7, wherein the mixture further comprises an organic solvent.
  • Embodiment E10 The method of any of embodiments E1-E7, wherein the mixture further comprises an aqueous solvent.
  • Embodiment El l The method of any of embodiments E1-E10, wherein the medical agent is cationic in a solvent, and wherein the nucleic acid molecule is anionic in the solvent.
  • Embodiment E12 The method of any of embodiments E1-E10, wherein the medical agent is anionic in a solvent, and wherein the nucleic acid molecule is cationic in the solvent.
  • Embodiment E13 The method of any of embodiments E1-E12, wherein in (a), the excipient (EX) and the medical agent (MA) are present in the mixture in a molar ratio of between about 2: 1 and about 1 : 100 (EX:MA).
  • Embodiment E14 The method of any of embodiments E1-E12, wherein the molar ratio of EX:MA is between about 1 : 1 and about 1 :50.
  • Embodiment E15 The method of any of embodiments E1-E14, wherein the excipient is a polymer.
  • Embodiment E16 The method of any of embodiments E1-E14, wherein the excipient is a lipid molecule.
  • Embodiment El 7 The method of any of embodiments El -El 6, wherein the medical agent comprises a small molecule drug.
  • Embodiment E18 The method of any of embodiments E1-E16, wherein the medical agent comprises Rapamycin.
  • Embodiment El 9. The method of any of embodiments El -El 6, wherein the medical agent comprises MK2206 or a pharmaceutically acceptable salt thereof.
  • Embodiment E20 The method of any of embodiments El -El 6, wherein the medical agent comprises Edaravone or a pharmaceutically acceptable salt thereof.
  • Embodiment E21 The method of any of embodiments E1-E16, wherein the medical agent comprises a nucleic acid molecule.
  • Embodiment E22 The method of any of embodiments El -El 6, wherein the medical agent comprises a siRNA.
  • Embodiment E23 The method of any of embodiments E1-E22, wherein a number average molar mass of the medical agent is less than about 13 kilodalton (kDa).
  • Embodiment E24 The method of any of embodiments E1-E22, wherein the number average molar mass of the medical agent is less than about 5 kDa.
  • Embodiment E25 The method of any of embodiments E1-E22, wherein the number average molar mass of the medical agent is less than about 1 kDa.
  • Embodiment E26 The method of any of embodiments E1-E25, wherein the encapsulation agent is a polymeric emulsifier.
  • Embodiment E27 The method of any of embodiments E1-E26, wherein an average nanoparticle size of the plurality of nanoparticles is between about 100 nanometers and 600 nanometers.
  • Embodiment Fl A method of manufacturing a nanoparticle composition, the method comprising: using a mixture to form a plurality of nanoparticles, wherein the mixture comprises (i) a medical agent that is not a nucleic acid molecule, (ii) an encapsulation agent, and (iii) an additional excipient, and wherein a nanoparticle of the plurality of nanoparticles comprises the medical agent, the additional excipient, and the encapsulation agent.
  • Embodiment F2 The method of embodiment Fl, further comprising mixing the medical agent, the additional excipient, and the encapsulation agent to form the mixture.
  • Embodiment F3 The method of any one of the embodiments F1-F2, further comprising incubating the mixture prior to forming the plurality of nanoparticles.
  • Embodiment F4 The method of any one of the embodiments F1-F3, wherein the mixture is incubated for at least about 6 hours.
  • Embodiment F5. The method of any one of the embodiments F1-F3, wherein the mixture is incubated for at least about 12 hours.
  • Embodiment F6 The method of any one of the embodiments F1-F5, wherein the incubation is at a temperature of at least about 3 °C.
  • Embodiment F7 The method of any one of the embodiments F1-F5, wherein the incubation is at a temperature of at least about 20 °C.
  • Embodiment F8 The method of any one of the embodiments F1-F7, wherein the mixture further comprises a protic solvent.
  • Embodiment F9 The method of any one of the embodiments F1-F7, wherein the mixture further comprises an organic solvent.
  • Embodiment F10 The method of any one of the embodiments F1-F7, wherein the mixture further comprises an aqueous solvent.
  • Embodiment Fl 1. The method of any one of the embodiments F1-F10, wherein the medical agent is cationic in a solvent, and wherein the nucleic acid molecule is anionic in the solvent.
  • Embodiment F12 The method of any one of the embodiments F1-F10, wherein the medical agent is anionic in a solvent, and wherein the nucleic acid molecule is cationic in the solvent.
  • Embodiment F13 The method of any one of the embodiments Fl -Fl 2, wherein the additional excipient (EX) and the medical agent (MA) are present in the mixture in a molar ratio of between about 2: 1 and about 1 : 100 (EX:MA).
  • Embodiment F14 The method of any one of the embodiments F1-F13, wherein the additional excipient (EX) and the medical agent (MA) are present in the mixture in a molar ratio of between about 2: 1 and about 1 : 100 (EX:MA).
  • Embodiment Fl 5 The method of any one of the embodiments Fl -Fl 3, wherein the molar ratio of EX:MA is between about 1 : 1 and about 1 :50.
  • Embodiment Fl 6 The method of any one of the embodiments Fl -Fl 5, wherein the additional excipient (EX) and the medical agent (MA) are present in the nanoparticle in a molar ratio of between about 2: 1 and about 1 : 100 (EX:MA).
  • Embodiment Fl 7 The method of any one of the embodiments Fl -Fl 5, wherein the molar ratio of EX:MA is between about 1 : 1 and about 1 :50.
  • Embodiment Fl 8 The method of any one of the embodiments Fl -Fl 7, wherein the additional excipient is a polymer.
  • Embodiment Fl 9 The method of any one of the embodiments Fl -Fl 7, wherein the additional excipient is a lipid molecule.
  • Embodiment F20 The method of any one of the embodiments Fl -Fl 9, wherein the medical agent comprises a small molecule drug.
  • Embodiment F21 The method of any one of the embodiments Fl -Fl 9, wherein the medical agent comprises Rapamycin.
  • Embodiment F22 The method of any one of the embodiments Fl -Fl 9, wherein the medical agent comprises MK2206 or a pharmaceutically acceptable salt thereof.
  • Embodiment F23 The method of any one of the embodiments Fl -Fl 9, wherein the medical agent comprises Edaravone or a pharmaceutically acceptable salt thereof.
  • Embodiment F24 The method of any one of the embodiments F1-F23, wherein a number average molar mass of the medical agent is less than about 13 kilodalton (kDa).
  • Embodiment F25 The method of any one of the embodiments F1-F23, wherein the number average molar mass of the medical agent is less than about 5 kDa.
  • Embodiment F26 The method of any one of the embodiments F1-F23, wherein the number average molar mass of the medical agent is less than about 1 kDa.
  • Embodiment F27 The method of any one of the embodiments F1-F26, wherein the encapsulation agent is a polymeric emulsifier.
  • Embodiment F28 The method of any one of the embodiments F1-F27, wherein an average nanoparticle size of the plurality of nanoparticles is between about 100 nanometers and 600 nanometers.
  • Embodiment Gl A nanoparticle composition comprising a plurality of nanoparticles, wherein a nanoparticle of the plurality of nanoparticles comprises a medical agent that is not a nucleic acid molecule, (ii) an encapsulation agent, and (iii) an additional excipient, wherein the additional excipient is oppositely charged from the medical agent.
  • Embodiment G2 The composition of embodiment Gl, wherein the additional excipient (EX) and the medical agent (MA) are present in the nanoparticle composition in a molar ratio of between about 2: 1 and about 1 : 100 (EX:MA).
  • Embodiment G3 The composition of embodiment Gl, wherein the molar ratio of EX:MA is between about 1 : 1 and about 1 :50.
  • Embodiment G4 The composition of any one of the embodiments G1-G3, wherein the additional excipient (EX) and the medical agent (MA) are present in the nanoparticle in a molar ratio of between about 2: 1 and about 1 : 100 (EX:MA).
  • Embodiment G5. The composition of any one of the embodiments G1-G3, wherein the molar ratio of EX:MA is between about 1 : 1 and about 1 :50.
  • Embodiment G6 The composition of any one of the embodiments G1-G5, wherein the additional excipient is a polymer.
  • Embodiment G7 The composition of any one of the embodiments G1-G5, wherein the additional excipient is a lipid molecule.
  • Embodiment G8 The composition of any one of the embodiments G1-G7, wherein the medical agent comprises a small molecule drug.
  • Embodiment G9 The composition of any one of the embodiments G1-G7, wherein the medical agent comprises Rapamycin.
  • Embodiment G10 The composition of any one of the embodiments G1-G7, wherein the medical agent comprises MK2206 or a pharmaceutically acceptable salt thereof.
  • Embodiment Gl 1. The composition of any one of the embodiments G1-G7, wherein the medical agent comprises Edaravone or a pharmaceutically acceptable salt thereof.
  • Embodiment G12 The composition of any one of the embodiments G1-G7, wherein the medical agent comprises a nucleic acid molecule.
  • Embodiment G13 The composition of any one of the embodiments G1-G7, wherein the medical agent comprises a siRNA.
  • Embodiment G14 The composition of any one of the embodiments G1-G13, wherein a number average molar mass of the medical agent is less than about 13 kilodalton (kDa).
  • Embodiment G15 The composition of any one of the embodiments G1-G13, wherein the number average molar mass of the medical agent is less than about 5 kDa.
  • Embodiment G16 The composition of any one of the embodiments G1-G13, wherein the number average molar mass of the medical agent is less than about 1 kDa.
  • Embodiment G17 The composition of any one of the embodiments G1-G16, wherein the encapsulation agent is a polymeric emulsifier.
  • Embodiment G18 The composition of any one of the embodiments G1-G17, wherein an average nanoparticle size of the plurality of nanoparticles is between about 100 nanometers and 600 nanometers.
  • Embodiment Hl A method of manufacturing a plurality of nanoparticles, comprising: forming the plurality of nanoparticles, wherein a nanoparticle of the plurality of nanoparticles comprises a compound, wherein the compound is MK2206 or a pharmaceutically acceptable salt thereof.
  • Embodiment H2 The method of embodiment Hl, wherein the forming comprises using a mixture to generate the plurality of nanoparticles, wherein the mixture comprises (i) the compound and (ii) a solvent.
  • Embodiment H3 The method of any one of the embodiments H1-H2, wherein the compound is present in the mixture at an amount that is at least about 0.1 % by mass of the mixture.
  • Embodiment H4 The method of any one of the embodiments H1-H2, wherein the compound is present in the mixture in an amount that is at least about 1 % by mass of the mixture.
  • Embodiment H5. The method of any one of the embodiments H1-H2, wherein the compound is present in the mixture in an amount that is at least about 5 % by mass of the mixture.
  • Embodiment H6 The method of any one of the embodiments H1-H5, wherein the solvent is a protic solvent.
  • Embodiment H7 The method of any one of the embodiments H1-H5, wherein the solvent is an organic solvent.
  • Embodiment H8 The method of any one of the embodiments H1-H7, wherein the mixture further comprises an anionic excipient.
  • Embodiment H9 The method of any one of the embodiments H1-H8, wherein the anionic excipient is a polymer.
  • Embodiment H10 The method of any one of the embodiments H1-H8, wherein the anionic excipient is a lipid molecule.
  • Embodiment Hl 1. The method of any one of the embodiments H1-H10, wherein the anionic excipient is encapsulated within the nanoparticle.
  • Embodiment H12 The method of any one of the embodiments Hl-Hl 1, wherein the mixture further comprises an encapsulation agent.
  • Embodiment H13 The method of any one of the embodiments H1-H12, wherein the mixture is incubated for at least about 1 hour prior to generation of the plurality of nanoparticles.
  • Embodiment H14 The method of any one of the embodiments H1-H13, wherein the compound is present in the nanoparticle composition in an amount that is at least about 0.1 % by mass of the nanoparticle.
  • Embodiment H15 The method of any one of the embodiments H1-H13, wherein the compound is present in the nanoparticle composition in an amount that is at least about 1% by mass of the nanoparticle.
  • Embodiment H16 The method of any one of the embodiments H1-H13, wherein the compound is present in the nanoparticle composition in an amount that is at least about 5% by mass of the nanoparticle.
  • Embodiment Hl 7 The method of any one of the embodiments Hl -Hl 6, wherein the compound is encapsulated within the nanoparticle.
  • Embodiment H18 The method of any one of the embodiments H1-H17, wherein an average nanoparticle size of the plurality of nanoparticles is between about 100 nanometers and 600 nanometers.
  • Embodiment 11 A composition comprising (i) a compound, wherein the compound is MK2206 or a pharmaceutically acceptable salt thereof and (ii) a nanoparticle vehicle.
  • Embodiment 12 The composition of embodiment 11, wherein the nanoparticle vehicle comprises a plurality of nanoparticles, wherein the compound is encapsulated by a nanoparticle of the plurality of nanoparticles.
  • Embodiment 13 The composition of any one of the embodiments 11-12, wherein the compound is present in the mixture in an amount that is at least about 0.1 % by mass of the nanoparticle.
  • Embodiment 14 The composition of any one of the embodiments 11-12, wherein the compound is present in the mixture in an amount that is at least about 1 % by mass of the nanoparticle.
  • Embodiment 15 The composition of any one of the embodiments 11-12, wherein the compound is present in the mixture in an amount that is at least about 5 % by mass of the nanoparticle.
  • Embodiment 16 The composition of any one of the embodiments 11-15, wherein an average nanoparticle size of the plurality of nanoparticles is between about 100 nanometers and 600 nanometers.
  • Embodiment 17 The composition of any one of the embodiments 11-16, wherein the compound is present in the mixture in an amount that is at least about 0.1 % by mass of the composition.
  • Embodiment 18 The composition of any one of the embodiments 11-16, wherein the compound is present in the mixture in an amount that is at least about 1 % by mass of the composition.
  • Embodiment 19 The composition of any one of the embodiments 11-16, wherein the compound is present in the mixture in an amount that is at least about 5 % by mass of the composition.
  • Embodiment 110 The composition of any one of the embodiments 11-19, wherein the composition further comprises an anionic excipient.
  • Embodiment Il l The composition of any one of the embodiments II -Il 0, wherein the anionic excipient is a polymer.
  • Embodiment 112. The composition of any one of the embodiments II -Il 0, wherein the anionic excipient is a lipid molecule.
  • Embodiment 113 The composition of any one of the embodiments 11-112, wherein the anionic excipient is encapsulated within the nanoparticle vehicle.
  • Embodiment 114 The composition of any one of the embodiments 11-113, wherein an average nanoparticle size of the plurality of nanoparticles is between about 100 nanometers and 600 nanometers.
  • Embodiment JI A method of manufacturing a plurality of nanoparticles, comprising: using a mixture to form the plurality of nanoparticles, wherein the mixture comprises
  • a compound wherein the compound is Edaravone or a pharmaceutically acceptable salt thereof and (ii) a protic solvent, wherein a nanoparticle of the plurality of nanoparticles comprises the compound.
  • Embodiment J2 The method of embodiment JI, wherein the compound is present in the nanoparticle in an amount that is at least about 0.1 % by mass of the nanoparticle.
  • Embodiment J3 The method of embodiment JI, wherein the compound is present in the nanoparticle in an amount that is at least about 1 % by mass of the nanoparticle.
  • Embodiment J4 The method of embodiment JI, wherein the compound is present in the nanoparticle in an amount that is at least about 5 % by mass of the nanoparticle.
  • Embodiment J5. The method of any one of the embodiments J1-J4, wherein the compound is encapsulated within the nanoparticle.
  • Embodiment J6 The method of any one of the embodiments J1-J5, wherein the protic solvent is an organic solvent.
  • Embodiment J7 The method of any one of the embodiments J1-J6, wherein the mixture further comprises an anionic excipient.
  • Embodiment J8 The method of any one of the embodiments J1-J7, wherein the anionic excipient is a polymer.
  • Embodiment J9 The method of any one of the embodiments J1-J7, wherein the anionic excipient is a lipid molecule.
  • Embodiment JI 0. The method of any one of the embodiments J1-J9, wherein the anionic excipient is encapsulated within the nanoparticle.
  • Embodiment JI 1 The method of any one of the embodiments JI -JI 0, wherein the mixture further comprises an encapsulation agent.
  • Embodiment J12 The method of any one of the embodiments JI -JI 1 , wherein an average nanoparticle size of the plurality of nanoparticles is between about 100 nanometers and 600 nanometers.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Molecular Biology (AREA)
  • Physics & Mathematics (AREA)
  • Biochemistry (AREA)
  • Biomedical Technology (AREA)
  • Nanotechnology (AREA)
  • Optics & Photonics (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

Un certain nombre d'agents médicaux sont administrés à un sujet pour des applications de diagnostic de maladie ou des applications thérapeutiques. La présente invention concerne des compositions et des procédés de fabrication et d'utilisation de celles-ci pour une population de particules qui peuvent être utilisées comme véhicule d'apport d'agents médicaux. La population de particules peut être des particules polymères. La population de particules peut, de manière sélective, cibler un tissu spécifique ou se positionner sur celui-ci, pour apporter à ce même tissu spécifique les agents médicaux ou positionner ces derniers sur celui-ci, de manière sélective. La population de particules peut encapsuler des agents thérapeutiques, tels qu'un macrolide (par exemple, la rapamycine).
PCT/US2021/051299 2020-09-22 2021-09-21 Nanoparticules et procédés d'utilisation associés WO2022066636A1 (fr)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US202063081682P 2020-09-22 2020-09-22
US63/081,682 2020-09-22
US202163144693P 2021-02-02 2021-02-02
US63/144,693 2021-02-02

Publications (1)

Publication Number Publication Date
WO2022066636A1 true WO2022066636A1 (fr) 2022-03-31

Family

ID=80844649

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2021/051299 WO2022066636A1 (fr) 2020-09-22 2021-09-21 Nanoparticules et procédés d'utilisation associés

Country Status (1)

Country Link
WO (1) WO2022066636A1 (fr)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN115137709A (zh) * 2022-06-30 2022-10-04 重庆医科大学 一种足细胞主动靶向的强化生物均质复合膜纳米药物递送系统及其制备方法和应用

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030162952A1 (en) * 2002-02-23 2003-08-28 Clariant Gmbh High-concentration aqueous solutions of betaines or amine oxides
US20100068286A1 (en) * 2008-06-16 2010-03-18 Greg Troiano Drug Loaded Polymeric Nanoparticles and Methods of Making and Using Same
US20140242115A1 (en) * 2005-08-08 2014-08-28 Oregon Health And Science University Inactivating pathogens with oxidizing agents for vaccine production
US20140328918A1 (en) * 2010-09-30 2014-11-06 Cerulean Pharma Inc. Methods of treating a subject and related particles, polymers and compositions
US20170087135A1 (en) * 2014-03-17 2017-03-30 Merck Sharp & Dohme Corp. Polymeric nanoparticles and methods of making and using same
US20190135873A1 (en) * 2016-12-19 2019-05-09 Morehouse School Of Medicine Compositions and methods for treating diseases by inhibiting exosome release

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030162952A1 (en) * 2002-02-23 2003-08-28 Clariant Gmbh High-concentration aqueous solutions of betaines or amine oxides
US20140242115A1 (en) * 2005-08-08 2014-08-28 Oregon Health And Science University Inactivating pathogens with oxidizing agents for vaccine production
US20100068286A1 (en) * 2008-06-16 2010-03-18 Greg Troiano Drug Loaded Polymeric Nanoparticles and Methods of Making and Using Same
US20140328918A1 (en) * 2010-09-30 2014-11-06 Cerulean Pharma Inc. Methods of treating a subject and related particles, polymers and compositions
US20170087135A1 (en) * 2014-03-17 2017-03-30 Merck Sharp & Dohme Corp. Polymeric nanoparticles and methods of making and using same
US20190135873A1 (en) * 2016-12-19 2019-05-09 Morehouse School Of Medicine Compositions and methods for treating diseases by inhibiting exosome release

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN115137709A (zh) * 2022-06-30 2022-10-04 重庆医科大学 一种足细胞主动靶向的强化生物均质复合膜纳米药物递送系统及其制备方法和应用
CN115137709B (zh) * 2022-06-30 2023-05-09 重庆医科大学 一种足细胞主动靶向的强化生物均质复合膜纳米药物递送系统及其制备方法和应用

Similar Documents

Publication Publication Date Title
Sur et al. Recent developments in functionalized polymer nanoparticles for efficient drug delivery system
Dave et al. Lipid-polymer hybrid nanoparticles: Synthesis strategies and biomedical applications
Bhatia et al. Nanoparticles types, classification, characterization, fabrication methods and drug delivery applications
JP6175237B2 (ja) コルチコステロイドを含む治療用ポリマーナノ粒およびそれを製造かつ使用する方法
Bose et al. Overview of nano-drugs characteristics for clinical application: the journey from the entry to the exit point
US20100196280A1 (en) Functionalized solid polymer nanoparticles for diagnostic and therapeutic applications
JP7164950B2 (ja) 胆汁酸粒子の組成物及び方法
JP2015098493A (ja) ビンカアルカロイド含有治療用ポリマーナノ粒子並びにその製造方法及び使用方法
US20230330273A1 (en) Magnetic nanoparticles functionalized with cathecol, production and use thereof
Li et al. Reduction breakable cholesteryl pullulan nanoparticles for targeted hepatocellular carcinoma chemotherapy
Janrao et al. Recent advances of polymer based nanosystems in cancer management
WO2022066636A1 (fr) Nanoparticules et procédés d'utilisation associés
Fan et al. Preparation and evaluation of doxorubicin‐loaded micelles based on glycyrrhetinic acid modified gelatin conjugates for targeting hepatocellular carcinoma
Sun et al. Pharmaceutical Nanotechnology
Ogier et al. Recent advances in the field of nanometric drug carriers
Naik Nano based drug delivery
KR101755680B1 (ko) 단백질 약물 전달용 다당류 나노겔 및 이의 제조방법
US20180271796A1 (en) Compositions and methods to improve nanoparticle distribution within the brain interstitium
Paulmurugan et al. Biodegradable polymer nanocarriers for therapeutic antisense microRNA delivery in living animals
Tomar Dendrimers as nanocarriers in cancer chemotherapy
Kumari et al. Biodegradable nanoparticles and their in vivo fate
Noe Rimondino et al. More than just size: Challenges and opportunities of hybrid dendritic nanocarriers
Hussein et al. Drug transport via Nanocarrier for liver cancer treatment
Raghava et al. Ophthalmic applications of nanotechnology
Certo Technological innovations in multimodal management of glioblastoma: from nano-drugs to imaging guided surgery and supra-maximal resection.

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: 21873277

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: 21873277

Country of ref document: EP

Kind code of ref document: A1