WO2019070645A1 - Méthodes et compositions pour une administration efficace à travers de multiples barrières biologiques - Google Patents

Méthodes et compositions pour une administration efficace à travers de multiples barrières biologiques Download PDF

Info

Publication number
WO2019070645A1
WO2019070645A1 PCT/US2018/053873 US2018053873W WO2019070645A1 WO 2019070645 A1 WO2019070645 A1 WO 2019070645A1 US 2018053873 W US2018053873 W US 2018053873W WO 2019070645 A1 WO2019070645 A1 WO 2019070645A1
Authority
WO
WIPO (PCT)
Prior art keywords
peptide
mini
nanodrug
lll
blood
Prior art date
Application number
PCT/US2018/053873
Other languages
English (en)
Inventor
Eggehard Holler
Julia Y. Ljubimova
Keith L. Black
Original Assignee
Cedars-Sinai Medical Center
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 Cedars-Sinai Medical Center filed Critical Cedars-Sinai Medical Center
Priority to EP18864985.9A priority Critical patent/EP3691670A4/fr
Priority to CN201880065055.7A priority patent/CN111182913A/zh
Priority to RU2020114744A priority patent/RU2020114744A/ru
Publication of WO2019070645A1 publication Critical patent/WO2019070645A1/fr
Priority to US16/815,760 priority patent/US20200206304A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • 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
    • A61K38/00Medicinal preparations containing peptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/04Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
    • A61K38/08Peptides having 5 to 11 amino acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/56Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule
    • A61K47/59Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyureas or polyurethanes
    • A61K47/593Polyesters, e.g. PLGA or polylactide-co-glycolide
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/62Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being a protein, peptide or polyamino acid
    • A61K47/64Drug-peptide, drug-protein or drug-polyamino acid conjugates, i.e. the modifying agent being a peptide, protein or polyamino acid which is covalently bonded or complexed to a therapeutically active agent
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/43504Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from invertebrates
    • C07K14/43563Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from invertebrates from insects
    • C07K14/43572Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from invertebrates from insects from bees
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • C07K14/4701Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
    • C07K14/4711Alzheimer's disease; Amyloid plaque core protein
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/81Protease inhibitors
    • C07K14/8107Endopeptidase (E.C. 3.4.21-99) inhibitors
    • C07K14/811Serine protease (E.C. 3.4.21) inhibitors
    • C07K14/8114Kunitz type inhibitors
    • C07K14/8117Bovine/basic pancreatic trypsin inhibitor (BPTI, aprotinin)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/81Protease inhibitors
    • C07K14/8107Endopeptidase (E.C. 3.4.21-99) inhibitors
    • C07K14/811Serine protease (E.C. 3.4.21) inhibitors
    • C07K14/8121Serpins
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K17/00Carrier-bound or immobilised peptides; Preparation thereof
    • C07K17/02Peptides being immobilised on, or in, an organic carrier
    • C07K17/08Peptides being immobilised on, or in, an organic carrier the carrier being a synthetic polymer
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K19/00Hybrid peptides, i.e. peptides covalently bound to nucleic acids, or non-covalently bound protein-protein complexes
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/08Tripeptides
    • C07K5/0802Tripeptides with the first amino acid being neutral
    • C07K5/0804Tripeptides with the first amino acid being neutral and aliphatic
    • C07K5/0808Tripeptides with the first amino acid being neutral and aliphatic the side chain containing 2 to 4 carbon atoms, e.g. Val, Ile, Leu
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/08Tripeptides
    • C07K5/0802Tripeptides with the first amino acid being neutral
    • C07K5/0812Tripeptides with the first amino acid being neutral and aromatic or cycloaliphatic
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K7/00Peptides having 5 to 20 amino acids in a fully defined sequence; Derivatives thereof
    • C07K7/04Linear peptides containing only normal peptide links
    • C07K7/06Linear peptides containing only normal peptide links having 5 to 11 amino acids
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/01Fusion polypeptide containing a localisation/targetting motif
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/01Fusion polypeptide containing a localisation/targetting motif
    • C07K2319/03Fusion polypeptide containing a localisation/targetting motif containing a transmembrane segment
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/01Fusion polypeptide containing a localisation/targetting motif
    • C07K2319/06Fusion polypeptide containing a localisation/targetting motif containing a lysosomal/endosomal localisation signal

Definitions

  • the disclosure generally relates to mini nanodrugs that include peptides capable of crossing blood-brain barrier, plaque-binding peptides and/or therapeutic agents conjugated to the polymalic acid-based scaffold.
  • The also disclosure relates to methods for treating brain diseases, including neurological disorders, reducing formation of amyloid plaques in the brains of patients suffering from Alzheimer's disease, and/or imaging the same by administering the mini nanodrugs described herein.
  • the A61-42 targeting D-peptide has been screened using a mirror imaging display selection and has a binding affinity in the sub-micro molar concentration (Wiesehan et al. (2003), which is incorporated herein by reference as if fully set forth).
  • nanoparticles deliver drugs by encapsulation, but they have unfavorable hydrodynamic diameters in the range 30-300 nm and limited BBB penetration. Such particles are also not biodegradable and can result in toxic, insoluble depositions. In addition, nonspecific drug effects may arise due to spontaneous release of drug cargo, via drug diffusion, or via nanoparticle dissolution (Elnegaard et al. (2017), which is incorporated by reference as if fully set forth).
  • antibody-based therapeutics even when humanized, can trigger systemic immune-responses, which comphcate long-term treatment perspectives (Borlak et al. (2016), which is incorporated by reference as if fully set forth).
  • antibody molecules are large and limit cargo capacity and hence the delivery of multiple drug cargoes to recipient cells.
  • the invention relates to a mini nanodrug comprising a polymalic acid-based molecular scaffold, at least one peptide capable of crossing the blood-brain barrier, at least one plaque-binding peptide and an endosomolytic ligand.
  • the at least one peptide capable of crossing the blood- brain barrier, the at least one plaque-binding peptide and the endosomolytic ligand are covalently linked to the polymalic acid-based molecular scaffold.
  • the mini nanodrug ranges in size from 1 nm to 10 nm.
  • the invention relates to a mini nanodrug comprising a polymalic acid-based molecular scaffold, at least one peptide capable of crossing the blood-brain barrier, an endosomolytic ligand and a therapeutic agent.
  • a mini nanodrug ranges in size from 1 nm to 10 nm.
  • the invention relates to a pharmaceutically acceptable composition
  • a pharmaceutically acceptable composition comprising any one of the mini nanodrugs described herein and a pharmaceutically acceptable carrier or excipient.
  • the invention relates to a method for treating a disease or abnormal condition in a subject.
  • the method comprises administering a therapeutically effective amount of any one of the mini nanodrugs described herein or any one of the pharmaceutically acceptable compositions described herein to a subject in need thereof.
  • the invention relates to a method for reducing formation of amyloid plaques in the brain of a subject.
  • the method comprises administering any one of the mini nanodrugs described herein, or any one of the compositions described herein to a subject in need thereof.
  • the invention relates to a method for treating a proliferative disease in a subject.
  • the method comprises administering a therapeutically effective amount of a mini nanodrug comprising a polymalic acid-based molecular scaffold, at least one peptide capable of crossing the blood-brain barrier, an endosomolytic ligand and an therapeutic agent to a subject in need thereof.
  • a mini nanodrug comprising a polymalic acid-based molecular scaffold, at least one peptide capable of crossing the blood-brain barrier, an endosomolytic ligand and an therapeutic agent to a subject in need thereof.
  • Each of the at least peptide, the endosomolytic ligand and the therapeutic agent are covalently linked to the polymalic acid-based molecular scaffold.
  • the mini nanodrug ranges in size from 1 nm to 10 nm.
  • FIG. 1 is a schematic drawing illustrating overview of molecular pathway for the delivery of the mini nanodrugs of the embodiments described herein.
  • FIG. 2 is a schematic drawing illustrating mini nanodrugs that permeate through multiple bio barriers into targeted tumors.
  • FIGS. 3A - 3B are schematic drawings illustrating advantages of mini nanodrugs for crossing the blood-brain barrier and entering brain parenchima.
  • FIG. 3 A is a schematic drawing illustrating mini nanodrugs carrying AP-2 peptides and tri-leucins (endosomic escape units) entering brain parenchima.
  • FIG. 3B is a schematic drawing comparing the efficiency of crossing the blood-brain barrier of a mini nanodrug carrying peptides and nanodrugs that carry antibodies.
  • FIG. 4 illustrates an example of the mini nanodrugs containing a single peptide.
  • FIG. 5 illustrates an example of the mini nanodrugs containing three peptides.
  • FIGS. 6A - 6D illustrate synthetic route for PMLA/LLL/Angiopep- 2/rhodamine (P/LLL/AP2) mini nanodrug.
  • FIG. 6A illustrates activation of biosynthesized polymalic acid (PMLA or P) by using a DCC/NHS chemistry to create the activated PMLA.
  • FIG. 6B illustrates conjugation of the activated PMLA with tri-leucine (LLL) and 2-mercaptoethylamine (ME A).
  • FIG. 6C illustrates conjugation of PMLA/LLL to Angiopep-2 (AP-2) and rhodamine dye.
  • FIG. 6D illustrates that MEA moiety was used to bind AP-2 peptide conjugated to a PEG linker via a Maleimide-thiol reaction. Rhodamine was attached in the same manner.
  • FIGS. 7A - 7G illustrate examples of product verification by HPLC.
  • FIG. 7A illustrates verification of PMLA/LLL/ Angiopep-2-PEG3400-MAL /rhodamine.
  • FIG. 7B illustrates verification of PMLA/ LLL/"Fe mimetic peptide" (SEQ ID NO: 2) CRTIGPSVC(cyclic)-peptide-PEG2000- Mal/rhodamine.
  • FIG. 7C illustrates verification PMLA/LLL/Miniap -4- PEG2000-Mal/cy 5.5.
  • FIG. 7D illustrates control: PMLA/LLL/rhodamine.
  • FIGS. 8A - 8C illustrate characterization of synthesized P/LLL/AP2.
  • FIG. 8A illustrates SEC-HPLC 3D view of A200-A700 nm vs. retention time and absorbances of the P/LLL/AP2 nanoconjugate constituents.
  • FIG. 8B illustrates SEC-HPLC chromatogram of P/LLL/AP2 recorded at 220 nm wavelength.
  • FIG. 8C illustrates the FTIR (Fourier-transform infrared) spectrum of P/LLL/AP2 nanoconjugate (dashed line), AP2 free peptide (solid line) and pre-conjugate (dashed-dotted line).
  • FTIR Fastier-transform infrared
  • FIG. 9 iUustrates PK for P/AP-2 (2%)/rhodamine (1%) conjugate measured by fluorescence intensity of the attached dye as a function of time from IV injection into tail vain until blood samples were taken.
  • FIGS. 10A - IOC illustrate characterization of synthesized P/LLL/AP-2/ACI-89/rhodamine
  • FIG. 10A illustrates SEC-HPLC top view of scanning A200-A700 nm vs. retention time displaying absorbances of the complete nanoconjugate
  • FIG. 10B illustrates the scanning profile of the same conjugate as shown on FIG. 10A at 572 nm wavelength indicating the rhodamine component.
  • FIG. IOC illustrates the scanning profile of the same conjugate as shown on FIG. 10A at 220 nm wavelength indicating the P/ LLL/ AP-2/ACI-89 component.
  • FIGS. 11A - l lC illustrates SEC-HPLC chromatogram of P/LLL/AP- 2/D1- peptide/rhodamine at A200-A700 nm vs. retention time displaying absorbancies of PMLA/ LLL/AP-2/D-peptide/rhodamine complete nanoconjugate.
  • FIG. 1 IB is a scanning profile of the same nanoconjugate as shown on FIG. 11A at 572 nm indicating the rhodamine component.
  • FIG. 11C is a scanning profile of the same nanoconjugate as shown on FIG. 11A at 220 nm indicating the PMLA/ LLL/AP-2/D1- peptide component.
  • FIGS. 11A - l lC illustrates SEC-HPLC chromatogram of P/LLL/AP- 2/D1- peptide/rhodamine at A200-A700 nm vs. retention time displaying absorbancies of PMLA/ LLL
  • FIG. 12A - 12C illustrate characterization of synthesized P/LLL/AP-2/ D3-peptide/rhodamine.
  • FIG. 12A illustrates SEC-HPLC top view displaying A200-A700 nm vs. retention time and absorbances of the P/LLL/AP-2/D3-peptide/rhodamine complete nanoconjugate.
  • FIG. 12B is the scanning profile of the same nanoconjugate as shown on FIG. 12A at 572 nm absorbance of rhodamine.
  • FIG. 12C is the scanning profile of the nanoconjugate shown on FIG. 12A recorded at 220 nm wavelength for the P/ LLL/ AP-2/ D3-peptide component.
  • FIG. 13 is a photograph of the left hippocampus CAl examined under fluorescence 2 hours following IV injection of PBS buffer into the tail vain of a mouse
  • FIG. 14 is a schematic drawing of the brain showing main blood vessels including the superior sagittal sinus (SSS), a large blood vessel that runs along the midline of the brain.
  • SSS superior sagittal sinus
  • FIGS. 15A - 15C illustrate concentration dependent BBB penetration of P/LLL/AP-2/rhodamine.
  • FIG. 15A is a set of photographs illustrating optical imaging data acquired at 120 min after i.v. injection of P/LLL/AP-2/rhodamine. at the following concentrations: photograph 1 - 29.5 ⁇ /kg; photograph 2 - 59 ⁇ /kg; photograph 3 - 118 ⁇ /kg; and photograph 4 - 236 ⁇ /kg.
  • FIG. 15B is a chart illustrating nanoconjugate fluorescence intensity vs. "distance from vasculature" measurements in brain parenchyma of mice injected with three different concentrations.
  • FIG. 15A is a set of photographs illustrating optical imaging data acquired at 120 min after i.v. injection of P/LLL/AP-2/rhodamine. at the following concentrations: photograph 1 - 29.5 ⁇ /kg; photograph 2 - 59 ⁇ /kg; photograph 3 - 118
  • 15C is set of charts: chart 1 - Cortex; chart 2 - Midbrain and chart 3 Hippocampus, illustrating average nanoconjugate fluorescence in the brain parenchyma measured following injections at four different drug concentrations.
  • the terms "P/LLL/AP-2” and" P/LLL/AP-2/rhodamine” are used interchangeably herein in reference to the mini nanodrugs.
  • FIGS. 16A - 16D illustrate blood vessel diameters, vascular coverage and inter-vessel distances in different brain regions.
  • FIG. 16A is a set of photographs illustrating blood vessels in the cortex, midbrain and hippocampal CAl cellular layer (outlined).
  • FIG. 16B is a bar graph illustrating vessel diameters.
  • FIG. 16C are bar graphs illustrating vascular coverage.
  • FIG. 16D illustrates the inter vessel distance defined as the shortest (Euclidian) distance between two adjacent blood vessels, comprehensively sampled for all vessels in each image.
  • FIGS. 17A - 17B illustrate that the nanoconjugate composition determines degree and locus of BBB penetration.
  • FIG. 17A is set of photographs illustrating nanoconjugate permeation of the cerebral cortex: photograph l-P/LLL/AP-2; photograph 2 - P/AP-2 and photograph 3 -P/LLL at constant injected dose (118 ⁇ /kg).
  • FIG. 17B is a set of bar graphs showing average nanoconjugate fluorescence in the cerebral cortex (1), the midbrain (2) and the hippocampus (2) as a function of nanoconjugate composition and concentration: P/LLL/AP-2 is shown in black, P/AP-2 in grey and P/LLL in white. All nanoconjugates referenced in FIGS. 17A - 17B contain rhodamine.
  • FIGS. 18A - 18B illustrate the effect of conjugated LLL residues on nanoconjugate conformation.
  • FIG. 18A is a chemical structure of the conjugate. LLL is indicated with black arrows in the structural scheme.
  • FIG. 18B is a three-dimensional image of short PMLA (16 malic acid residues) with PEG (2 chains of ethylene glycol-hexamer conjugated via maleimide to PMLA), capped sulfhydryl (two moieties) and LLL (4 moieties).
  • FIGS. 19A - 19B illustrate nanoconjugate conformation in the absence of LLL.
  • FIG. 19A illustrates the structural model, and is similar as the one shown in FIG. 18A but lacking LLL.
  • FIG. 19B is a three-dimensional image of the structure shown in FIG. 19A.
  • FIGS. 20A - 20E illustrate nanoconjugate peptide moiety screen.
  • FIG. 20A is a set of photographs illustrating the P/LLL nanoconjugates equipped with different peptides (1- P/LLL/AP-2; 2- P/LLL/M4; and 3 - P/LLL/B6) to assess their role in BBB penetration following the injection into mice at the concentration of 118 ⁇ /kg (i.e., at a constant injected dose).
  • FIGS. 20B - 20D is a set of bar graphs showing average nanoconjugate fluorescence in the cerebral cortex (FIG. 20B), midbrain (FIG. 20C) and hippocampus (FIG. 20D) as a function of injected concentration.
  • FIG. 20B is a set of bars graphs showing average nanoconjugate fluorescence in the cerebral cortex (FIG. 20B), midbrain (FIG. 20C) and hippocampus (FIG. 20D) as a function of injected concentration.
  • FIGS. 20E illustrates fluorescence measurements in the cerebral cortex for nanoconjugates P/LLL/AP-2 (2%), P/LLL/AP-2/M4, P/LLL/AP-2 (4%) and P/LLL/AP-7 injected into mice at the concentrations of 59 ⁇ /kg or 118 ⁇ /kg (i.e., two doses were assessed). All nanoconjugates referenced in FIGS. 20A - 20E contain rhodamine.
  • FIGS. 21A - 2 ID illustrates pharmacokinetics of nanoconjugate fluorescence in serum and brain tissue.
  • FIG. 21A is a chart illustrating serum clearance analysis was conducted for P/LLL/AP-2 (black) and P/LLL (grey), and optically via imaging of the cerebral vasculature content (black, triangles).
  • FIG. 2 IB is a set of photographs illustrating optical imaging data of and around the saggital sinus showing drug clearance and parenchyma accumulation over 240 minutes.
  • FIG. 21C illustrates vascular fluorescence intensity profile for the saggital sinus as indicated along the white hne in the utmost left panel of FIG. 2 IB.
  • FIG. 21A is a chart illustrating serum clearance analysis was conducted for P/LLL/AP-2 (black) and P/LLL (grey), and optically via imaging of the cerebral vasculature content (black, triangles).
  • FIG. 2 IB is a set of photographs illustrating optical imaging data of and around the
  • 2 ID is a bar graph illustrating time dependence of nanoconjugate fluorescence intensity in brain tissue for P/LLL/AP-2 (black), P/LLL (grey) and P/AP-2 (white) that are different from the serum PK kinetics. All nanoconjugates referenced in FIGS. 21A - 2 ID contain rhodamine.
  • FIGS. 22A - 22C illustrate concentrations indicated by clouds in different shades of grey of the nanoconjugate (A1-A2) and quantitative in ⁇ g/mL in FIG, 22B and FIG. 22C after i.v. injection of P/LLL/AP-2 in the parenchyma of the cerebral cortex.
  • FIG. 22A is set of photographs illustrating optical imaging data showing cortical tissue from mice injected with P/LLL/AP-2 at 29.5 ⁇ /kg (Al) and 118 ⁇ /kg (A2) and regions (dotted) of interest for comparison of fluorescence intensities in vascular tissue and parenchyma.
  • FIG. 22B illustrates fluorescence ratios in vasculature / cortical brain parenchyma.
  • FIGS. 22A - 22C illustrates estimated P/LLL/AP-2 concentration in the cortical brain parenchyma as a function of injected dose, based on known concentrations from PK measurements in the vascular and the measured intensity ratios of fluorescence in the vascular to the regions of interest. All nanoconjugates referenced in FIGS. 22A - 22C contain rhodamine.
  • FIGS. 23A - 23C illustrate peptide-dependent labeling of plaques by injected nanoconjugates labeled with rhodamine.
  • FIG. 23A is a photograph illustrating optical imaging data following mice injected with P/LLL/M4.
  • FIG. 23B is a photograph illustrating optical imaging data following mice injected with P/LLL/M4/D1.
  • FIG. 23A is a photograph illustrating optical imaging data following mice injected with P/LLL/M4/D1.
  • 23C is a bar graph showing fluorescence intensities of A6 binding of nanoconjugates PMLA, P/cTfRL, P/M4, P/LLL, P/LLL/AP-2, P/LLL/M4, P/AP-2/ACI-89, P/LLL/AP-2/D3, P/LLL/AP-2/D 1 and P/LLL/M4/D 1 labeled with rhodamine. Plaque vs. background labeling (signal noise) is indicated.
  • peptide refers to a contiguous and relatively short sequence of amino acids linked by peptidyl bonds.
  • peptide and polypeptide are is used interchangeably.”
  • the peptide may have a length of about 2 to 10 amino acids, 8 to 20 amino acids or 6 to 25 amino acids.
  • amino acid and “amino acid residue” are used interchangeably herein.
  • An abnormal condition refers to a function in the cells and tissues in a body of a patient that deviates from the normal function in the body.
  • An abnormal condition may refer to a disease.
  • Abnormal condition may include brain disorders. Brain disorders may be but are not limited to Alzheimer's disease, Multiple sclerosis, Parkinson's disease, Huntington's disease, schizophrenia, anxiety, dementia, mental retardation, and anxiety.
  • Abnormal condition may include proliferative disorders.
  • the terms "proliferative disorder” and “proliferative disease” refer to disorders associated with abnormal cell proliferation. Proliferative disorders may be, but are not limited to, cancer, vasculogenesis, psoriasis, and fibrotic disorders.
  • An embodiment provides a mini nanodrug comprising a polymalic acid-based molecular scaffold, one or more peptides capable of crossing the blood-brain barrier, an endosomolytic ligand and a therapeutic agent.
  • a mini nanodrug comprising a polymalic acid-based molecular scaffold, one or more peptides capable of crossing the blood-brain barrier, an endosomolytic ligand and a therapeutic agent.
  • Each of the peptides capable of crossing the blood-brain barrier, endosomolyitic hgand and therapeutic agent may be covalently linked to the polymalic acid-based molecular scaffold.
  • peptide capable of crossing blood-brain barrier refers to any peptide that can bind to receptors responsible for maintaining the integrity of the brain-blood barrier and brain homeostasis.
  • One or more peptides capable of crossing blood-brain barrier may be an LRP-1 ligand, or a transferrin receptor ligand.
  • One or more peptides capable of crossing blood-brain barrier may be a peptide that may bind the low density lipoprotein (LDL) receptor-related protein (LPR), which possesses the ability to mediate transport of ligands across endothelial cells of the brain-blood barrier.
  • LDL low density lipoprotein
  • LPR low density lipoprotein
  • One or more peptides capable of crossing blood-brain barrier may be Angiopep-2, an aprotinine- derived peptide, capable of binding lipoprotein receptor-related protein- 1 (LRP-1) and promoting drug delivery in the CNS (Demeule et al., 2008, which is incorporated herein by reference as if fully set forth).
  • the terms "Angiopep-2" and "AP-2” are used herein interchangeably.
  • the Angiopep-2 may be the cysteine-modified Angiopep-2.
  • the cysteine- modified Angiopep-2 peptide may be a peptide comprising the amino acid sequence TFFYGGSRGKRNNFKTEEYC (SEQ ID NO: 1).
  • the Angiopep-2 peptide may be a variant of Angiopep-2 peptide.
  • the variant of the Angiopep-2 peptide may be a peptide comprising an amino acid with at least 70, 72, 75, 80, 85, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99 or 100% sequence identity to a sequence of SEQ ID NO: 1.
  • the variant of the Angiopep-2 peptide may be any variant of the sequence of SEQ ID NO: 1, in which lysine residue at the positions 10 and/or 15 remain invariant.
  • One or more peptides may be any other peptide capable of binding LPR, crossing blood-brain barrier, and promoting delivery of the mini nanodrug in the CNS.
  • one or more peptides may be a peptide that enhances penetration of any one of the mini nanodrugs described herein across the blood-brain barrier via the transferrin receptor (TfR) pathway.
  • TfR transferrin receptor
  • the TfR pathway imports iron (complexed to transferrin, Tf) into the brain and is involved in cerebral iron homeostasis.
  • One or more peptides capable of crossing the blood-brain barrier may be a ligand binding to TfR or a ligand binding to transferrin (Tf).
  • the transferrin ligand may be a Fe mimetic peptide, also referred to herein as cTfRL.
  • the Fe mimetic peptide may be a peptide comprising the amino acid sequence CRTIGPSVC (SEQ ID NO: 2).
  • the variant of the Fe mimetic peptide may be a peptide comprising an amino acid with at least 70, 72, 75, 80, 85, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99 or 100% sequence identity to a sequence of SEQ ID NO: 2.
  • the variant of the Fe mimetic peptide may be any variant of the sequence of SEQ ID NO: 2, which is capable to bind its target and penetrate the blood-brain barrier.
  • the variant binding to the immobilized transferrin (Tf) which further binds the transferrin receptor (TfR) may be tested by the surface plasmon resonance (SPR) method (Ding et al. (2016), which is incorporated herein by reference as if fully set forth).
  • the Fe mimetic peptide or a variant thereof may be cyclic, may comprise disulfide bonds (-S-S-), or may comprise any other modifications known in the art.
  • the Fe mimetic peptide or a variant thereof may be linked to PMLA via an appropriate linker at its terminal -NH2 group when the sulfhydryls forms a disulfide (-SS-)-cyclic variant, or in the linear version at one of the thio groups as thioether.
  • the transferrin receptor ligand may be a B6 peptide.
  • the B6 peptide may be a peptide comprising the amino acid sequence CGHKAKGPRK (SEQ ID NO: 8).
  • the B6 peptide may be a peptide comprising an amino acid with at least 70, 72, 75, 80, 85, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99 or 100% sequence identity to an amino acid sequence of SEQ ID NO: 8.
  • the variant of the B6 peptide may be any variant of the amino acid sequence of SEQ ID NO: 8, which is capable to bind its target TfR and penetrate the blood- brain barrier.
  • Binding of the variant of the B6 peptide to a transferrin receptor (TfR) can be tested, for example, by the surface plasmon resonance (SPR) method (Ding et al. (2016), which is incorporated herein by reference as if fully set forth).
  • SPR surface plasmon resonance
  • MiniAp-4 is a peptide derived from the bee venom, and is capable of penetrating the blood-brain barrier (Oller-Salvia et al. 2010, which is incorporated herein by reference as if fully set forth).
  • the MiniAp-4 peptide may be a peptide comprising the amino acid sequence KAPETAL D (SEQ ID NO: 3).
  • the MiniAp-4 peptide may comprise an amino acid sequence with at least 70, 72, 75, 80, 85, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99 or 100% identity to a sequence of SEQ ID NO: 3.
  • the variant of the MiniAp-4 peptide may be any variant of the sequence of SEQ ID NO: 3, which is capable of penetrating the blood-brain barrier (BBB).
  • BBB permeation of mini nanodrugs can be assayed ex vivo using fluorescence imaging as described in Example 4 herein.
  • one or more peptides capable of crossing the blood -brain barrier may be two or more peptides. Two or more peptides may be similar peptides. Two or more peptides may be selected independently.
  • the mini nanodrug may comprise Angiopep-2, Fe mimetic peptide, B6 peptide, and Miniap-4 peptide in any combination.
  • the mini nanodrug may comprise any other peptides capable of crossing the blood-brain barrier.
  • the mini nanodrug may comprise a therapeutic agent.
  • the therapeutic agent may be an antisense oligonucleotide, an siRNA oligonucleotide, a peptide, or a low molecular weight drug.
  • the therapeutic agent may be a combination of two or more therapeutic agents.
  • the therapeutic agent may be an antisense oligonucleotide or an siRNA.
  • the antisense oligonucleotide may be a Morpholino antisense oligonucleotide.
  • the therapeutic agent may inhibit the synthesis or activity of the 6-secretase or ⁇ -secretase for amyloid 6 (A6) production.
  • the antisense oligonucleotide or the siRNA may comprise a sequence complementary to a sequence contained in an mRNA transcript of 6-secretase or ⁇ -secretase.
  • the antisense oligonucleotide may include a nucleic acid sequence with at least 70, 72, 75, 80, 85, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99 or 100% identity to a sequence of SEQ ID NO: 4.
  • ⁇ -secretase and ⁇ -secretase are proteolytic enzymes that cleave the amyloid precursor protein (APP) at substrate specific amino acid sites and generate the amyloid-6 (A6) peptide that accumulates in brain tissue and causes Alzheimer's disease (AD). Inhibition 6- or ⁇ -secretase activity may have therapeutic potential in the treatment of AD.
  • APP amyloid precursor protein
  • A6 amyloid-6
  • the mini nanodrug may comprise Angiopep-2, Fe mimetic peptide, B6 peptide, or Miniap-4 peptide, or any combination thereof, and the antisense oligonucleotide or the siRNA comprising a nucleic acid sequence complementary to the sequence contained in an mRNA transcript of 6-secretase or ⁇ -secretase.
  • the therapeutic agent may be a therapeutic peptide, for example, for AD treatment.
  • the therapeutic peptides may be a peptide that may target the amyloid plagues and induce the degradation activity of the mini nanodrugs to the Alzheimer disease (AD) lesions.
  • the therapeutic peptide may be a 6- sheet breaker peptide.
  • ⁇ -sheet breaker peptide refers to a peptide that disrupts 6-sheet elements and the self-recognition motif of A6 by inhibiting the interconnection of 6-sheet A61-42, so as to prevent misfolding and aggregation of A6 (Lin et al. (2014), which is incorporated herein by reference as if fully set forth).
  • the 6-sheet breaker peptide may be H102 peptide.
  • the 102 peptide may be a peptide comprising the amino acid sequence HKQLPFFEED (SEQ ID NO: 6).
  • the 102 peptide may be a peptide comprising an amino acid with at least 70, 72, 75, 80, 85, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99 or 100% sequence identity to an amino acid sequence of SEQ ID NO: 6.
  • the variant of the H102 peptide may be any variant of the sequence of SEQ ID NO: 6, which is capable of inhibiting formation of 6-sheet A61-4 and by "misfolding" and aggregation of A6.
  • the variant of the H102 peptide may be any variant that is capable of solubilizing plaques.
  • the ability to solubilize plaques may be measured.
  • the number and the size of plaques in treated and referenced animals can be measured ex vivo by optical imaging as described in Example 4 herein.
  • In vivo asssays for example, positron emission tomography (PET), near -infrared spectroscopy (NIR), or infra-red (IR) imaging are known in the art, and can be used for imaging amyloid plaques (Nordberg (2008), Kung et al. (2012), and Cheng et al. (2016), all of which are incorporated herein by reference as if fully set forth).
  • the mini nanodrug may comprise one or more peptides capable of crossing the blood-brain barrier, and a 6-sheet breaker peptide.
  • the mini nanodrug may comprise Angiopep-2, Fe mimetic peptide, B6 peptide, or Miniap-4 peptide, or any combination thereof, and the H102 peptide.
  • the mini nanodrug may further carry any of the antisense oligonucleotides described herein.
  • the therapeutic peptide for AD treatment may be a plaque-binding peptide.
  • plaque-binding peptide refers to a peptide that binds to or labels neuritic plaques that consists of amyloid peptide 6 (A6), the characteristic pathological hallmark of AD.
  • the plaque-binding peptide may be a ⁇ -sheet breaker peptide(s) described herein.
  • the plaque-binding peptide may be a D-enantiomeric peptide that specifically binds to amyloid 61-42 (A642).
  • the D-enantiomeric peptide may bind to or label plaques that contain A642 in the brain.
  • the D-enantiomeric peptide may be one or more of a Dl-peptide, a D3-peptide and an ACI-89-peptide, or variants thereof.
  • the D-enantiomeric peptide may be the Dl-peptide comprising an amino acid sequence QSHYKHISPAQVC (SEQ ID NO: 9).
  • the Dl- peptide may be a peptide comprising an amino acid with at least 70, 72, 75, 80, 85, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99 or 100% sequence identity to an amino acid sequence of SEQ ID NO: 9.
  • the variant of the Dl-peptide may be any variant of the sequence of SEQ ID NO: 9, which is capable to of binding or labeling plaques that contain A642.
  • assaying plaques ex vivo may include binding of reagent molecules to structural units (amino acid domains) of the amyloids, and measuring changes in fluorescence properties of the reagent-amyloid formations, e.g., by solubilization of the plaque material in these formations.
  • Different D-peptides may recognize different amino acid sequences in 6- amyloids as they are exposed in plaques.
  • these reagents may destabilize amyloid interactions forming free amyloid species, which can involve further binding to the reagent.
  • the overall efficacy of the reagents may depend on the strength of binding to plaque domains. In case of plaque dissolution, morphometriuc analysis can be used to compare treated and referenced mice of similar stage of disease.
  • the D-enantiomeric peptide may be a D3-peptide comprising an amino acid sequence RPRTRLHTHRNRC (SEQ ID NO: 10).
  • the D3- peptide may be a peptide comprising an amino acid with at least 70, 72, 75, 80, 85, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99 or 100% sequence identity to an amino acid sequence of SEQ ID NO: 10.
  • the variant of the D3-peptide may be any variant of the sequence of SEQ ID NO: 10, which is capable of binding or labeling plaques that contain A642.
  • the D-enantiomeric peptide may be ACI-89-peptide comprising an amino acid sequence PSHYKHISPAQKC (SEQ ID NO: 11).
  • the ACI-89 peptide may be a peptide comprising an amino acid with at least 70, 72, 75, 80, 85, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99 or 100% sequence identity to an amino acid sequence of SEQ ID NO: 11.
  • the variant of the ACI-89-peptide may be any variant of the sequence of SEQ ID NO: 11, which is capable of binding or labeling plaques that contain A642.
  • the mini nanodrug may comprise one or more peptides capable of crossing the blood-brain barrier, and one or more plaque- binding peptides.
  • the mini nanodrug may comprise Angiopep-2, Fe mimetic peptide, B6 peptide, or Miniap-4 peptide, or any combination thereof, and the Dl-peptide, D3-peptides or ACI-89 peptide, or any combination thereof.
  • the mini nanodrug may further comprise a ⁇ -sheet breaker peptide.
  • the mini nanodrug may further carry any of the antisense oligonucleotides.
  • the mini nanodrug may comprise peptides described herein and thereapeutic agents in any combination.
  • Determining percent identity of two amino acid sequences or two nucleic acid sequences may include ahgning and comparing the amino acid residues or nucleotides at corresponding positions in the two sequences. If all positions in two sequences are occupied by identical amino acid residues or nucleotides then the sequences are said to be 100% identical. Percent identity is measured by the Smith Waterman algorithm (Smith TF, Waterman MS 1981 "Identification of Common Molecular Subsequences," J Mol Biol 147: 195 -197, which is incorporated herein by reference as if fully set forth).
  • variant refers to a peptide that retains a biological activity that is the same or substantially similar to that of the original sequence.
  • the variant may have a sequence that is similar to, but not identical to, the original sequence of the peptide or a fragment thereof.
  • the variant may include one or more amino acid substitutions, deletions, insertions of amino acid residues, or any combination thereof.
  • the variant may be from the same or different species or be a synthetic sequence based on a natural or prior sequence.
  • the variant peptide may have the same length as the specified sequence of the peptide or may have additional amino acid residues at either or both termini of the peptide.
  • the variant may be a fragment of the peptide.
  • a fragment of the original sequence is a continuous or contiguous portion of the original sequences.
  • the length of the fragment of the original peptide 20 amino acid-long may vary in be any 2 to 19 contiguous amino acids within the original peptide.
  • An embodiment comprises amino acid sequences, peptides or polypeptides having a portion of the sequence as set forth in any one of the amino acids listed herein or the complement thereof. These amino acid sequences, peptides or polypeptides may have a length in the range from 2 to full length, 4 to 6, 6 to 8, 8 to 10, 10 to 12, 12 to 14, 14 to 16, or 7 to 13, or 7, 8, 9, 10, 13, 20 or 21 amino acids. An amino acid sequence, peptide or polypeptide having a length within one of the above ranges may have any specific length within the range recited, endpoints inclusive.
  • the recited length of amino acids may start at any single position within a reference sequence (i.e., any one of the amino acids herein) where enough amino acids follow the single position to accommodate the recited length.
  • the recited length may be full length of a reference sequence.
  • the variant or fragment of any one the peptides described herein capable of crossing the BBB are biologically active when the variant or fragment retains some or all activity of the original peptide, and is capable of transporting the mini nanodrug to which it is attached across the BBB.
  • the variant or fragment of any one the plaque-binding peptides described herein are biologically active when the variant or fragment retains some or all activity of the original peptide, and is capable of binding or labeling neuritic plaques that consists of amyloid peptide 6 (A6).
  • the activity of the variants and fragments may be determined in an assay.
  • the assay may involve testing variant's ability to bind to a receptor, or traverse BBB.
  • the assay may test binding or labeling neuritic plaques that consists of amyloid peptide 6 (A6).
  • the variants and fragments of the original peptide may be more or less active compared to the original peptide.
  • the variants of fragments may have lower activity compared to the original peptide as long as they are capable of achieving the desirable result.
  • the peptide or a variant thereof may have additional components or groups.
  • the sequence of the peptide or its variant may be linked to -NH2 group at the C-terminus.
  • the sequence of the peptide or a variant thereof may be linked to diaminopimehc acid (DAP) or hydroxy! diaminopimelic acid (H-DAP) at the N-terminus.
  • DAP diaminopimehc acid
  • H-DAP hydroxy! diaminopimelic acid
  • the peptide or a variant thereof may contain bonds to increase stability and folding of the peptide.
  • the peptide or a variant thereof may comprise disulfide bonds (-S-S-) forming an exocyclic structure that improves resistance to cleavage by peptidases.
  • the sequence of the peptide or a variant thereof may be linked to any other moiety or group.
  • the peptide may be of any desired molecular weight.
  • the peptide may have a molecular weight of about 1,000 kDa, about 1,500 kDa, about 2,000 kDa, about 2,500 kDa, about 3,000 kDa, about 3,500 kDa, about 4,000 kDa, about 4,500 kDa, about 5,000 kDa, about 10,000 kDa, or about 15,000 Da.
  • the peptide may have a molecular weight of about 1 kDa to about 15kDa. In an embodiment the peptide may have a molecular weight of 15kDa, or less.
  • each of peptides described herein may be conjugated to the polymalic acid-based molecular scaffold by a linker.
  • linker means an organic moiety that connects two parts of a compound.
  • the linker may comprise a polyethylene glycol (PEG).
  • the PEG may be of any desired molecular weight.
  • the PEG may have a molecular weight of about 1,000 Da, about 1,500 Da, about 1,000 Da, about 2,500 Da, about 3,000 Da, about 3,500 Da, about 4,000 Da, about 4,500 Da, about 5,000 Da, about 10,000 Da, about 15,000 Da, about 20,000 Da, about 25,000 Da, or about 30,000 Da.
  • the PEG may have a molecular weight of about 3,400 Da.
  • the mini nanodrug may include an endosomolytic ligand.
  • the endosomolytic ligand may be covalently linked with the polymalic acid-based molecular scaffold.
  • the term "endosomolytic ligand” refers to molecules having endosomolytic properties. Endosomolytic ligands promote the lysis of and/or transport of the composition of the invention, or its components, from the cellular compartments such as the endosome, lysosome, endoplasmic reticulum (ER), golgi apparatus, microtubule, peroxisome, or other vesicular bodies within the cell, to the cytoplasm of the cell.
  • the endosomolytic ligands may be, but are not limited to, imidazoles, poly or oligoimidazoles, linear or branched polyethyleneimines (PEIs), linear or branched polyamines, e.g. spermine, cationic linear or branched polyamines, polycarboxylates, polycations, masked oligo or poly cations or anions, acetals, polyacetals, ketals/polyketals, orthoesters, linear or branched polymers with masked or unmasked cationic or anionic charges, dendrimers with masked or unmasked cationic or anionic charges, polyanionic peptides, polyanionic peptidomimetics, pH-sensitive peptides, natural or synthetic fusogenic lipids, natural or synthetic cationic lipids.
  • PEIs polyethyleneimines
  • linear or branched polyamines e.g. spermine, cationic linear or branched polyamine
  • the endosomolytic ligand may include a plurality of leucine, isoleucine, valine, tryptophan, or phenylalanine residues.
  • the endosomolytic ligand may be Trp-Trp-Trp (WWW), Phe-Phe-Phe (FFF), Leu- Leu-Leu (LLL), or Ile-Ile-Ile (I-I-I).
  • WWW Trp-Trp-Trp
  • FFF Phe-Phe-Phe
  • LLL Leu- Leu-Leu
  • I-Ile-Ile I-I-Ile
  • the WWW, FFF, LLL or III may enhance the ability of the mini nanodrug to cross the blood-brain barrier.
  • the polymalic acid-based molecular scaffold may be polymalic acid.
  • polymalic acid refers to a polymer, e.g., a homopolymer, a copolymer or a blockpolymer that contains a main chain ester linkage.
  • the polymalic acid may be at least one of biodegradable and of a high molecular flexibility, soluble in water (when ionized) and organic solvents (in its acid form), non-toxic, or non-immunogenic (Lee B et al., Water-soluble aliphatic polyesters: poly(malic acid)s, in: Biopolymers, vol.
  • the polymalic acid may be poly(6-L-malic acid), herein referred to as poly-6-L-malic acid or PMLA.
  • the polymalic acid may be of any length and of any molecular mass.
  • the polymalic acid may have a molecular mass of 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, or 200 kDa.
  • the polymalic acid may have a molecular mass of 10, 20, 30, 40, 50, or 60 kDa.
  • the polymalic acid may have a molecular mass in a range between any two of the following molecular masses: 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, or 200 kDa. In an embodiment, the polymalic acid may have a molecular mass in a range between any two of the following masses: 40, 45, 50, 55, or 60 kDa.
  • Exemplary polymalic acid-based molecular scaffolds amenable to the imaging nanoagents disclosed herein are described, for example, in PCT Appl. Nos. PCT/US04/40660, filed December 3, 2004, PCT/US09/40252, filed April 10, 2009, and PCT/US 10/59919, filed December 10, 2010, PCT/US 10/62515, filed December 30, 2010; and US patent application Ser. No. 10/580,999, filed March 12, 2007, and Ser. No. 12/935, 110, filed September 28, 2010, contents of all which are incorporated herein by reference as if fully set forth.
  • the mini nanodrug may be linked to an additional therapeutic agent.
  • the additional therapeutic agent may be a drug for treatment of AD.
  • Additional exemplary drugs for treatment of AD may be but are not limited to cholinesterase inhibitors, muscarinic agonists, anti-oxidants or antiinflammatories. Galantamine (Reminyl), tacrine (Cognex), selegiline, donepezil, (Aricept), saeluzole, acetyl-L-carnitine, idebenone, ENA-713, memric, quetiapine, or verubecestat (3-imino-l,2,4-thiadiazinane 1, 1- dioxidederivative) may be used.
  • the additional therapeutic agent may be an anti-cancer agent.
  • Additional exemplary anti-cancer agents amenable to the present invention may be, but are not limited to, paclitaxel (taxol); docetaxel; germicitibine; alitretinoin; amifostine; bexarotene bleomycin; calusterone; capecitabine; platinate; chlorambucil; cytarabine; daunorubicin, daunomycin; docetaxel; doxorubicin; dromostanolone propionate; fluorouracil (5-FU); leucovorin; methotrexate; mitomycin C; mitoxantrone; nandrolone pamidronate; mithramycin; porfimer sodium; procarbazine; quinacrine; temozolomide; or topotecan.
  • paclitaxel taxol
  • docetaxel germicitibine
  • alitretinoin am
  • the mini nanodrug may further comprise an imaging agent.
  • the imaging agent may be any fluorescent reporter dye.
  • fluorescent reporter dyes e.g., fluorophores
  • the fluorophore is an aromatic or heteroaromatic compound and can be a pyrene, anthracene, naphthalene, acridine, stilbene, indole, benzindole, oxazole, thiazole, benzothiazole, cyanine, carbocyanine, salicylate, anthranilate, coumarin, fluorescein, rhodamine or other like compound.
  • Suitable fluorescent reporters may include xanthene dyes, such as fluorescein or rhodamine dyes.
  • Fluorophores may be, but are not limited to, 1,5 IAEDANS; 1,8-ANS; 4-Methylumbelliferone; 5-carboxy-2,7- dichlorofluorescein; 5-Carboxy fluorescein (5-FAM); 5-
  • Carboxynapthofluorescein (pH 10); 5-Carboxytetramethyl rhodamine (5- TAMRA); 5-FAM (5-Carboxyfluorescein); 5-Hydroxy Tryptamine (HAT); 5- ROX (carboxy-X-rhodamine); 5-TAMRA (5-Carboxytetramethyl rhodamine); 6- Carboxyrhodamine 6G; 6-CR 6G; 6-JOE; 7-Amino-4-methylcoumarin; 7- Aminoactinomycin D (7-AAD); 7-Hydroxy-4-methylcoumarin; 9-Amino-6- chloro-2-methoxy acridine; ABQ; Acid Fuchsin; ACMA (9-Amino-6-chloro-2- methoxyacridine); Acridine Orange; Acridine Red; Acridine Yellow; Acriflavin; Acriflavin Feulgen SITSA; Aequorin (Photoprotein); Alex
  • fluorescent proteins suitable for use as imaging agents include, but are not limited to, green fluorescent protein, red fluorescent protein (e.g., DsRed), yellow fluorescent protein, cyan fluorescent protein, blue fluorescent protein, and variants thereof (see, e.g., U.S. Pat. Nos. 6,403, 374, 6,800,733, and 7, 157,566, contents of which are incorporated herein by reference as if fully set forth).
  • GFP variants include, but are not limited to, enhanced GFP (EGFP), destabilized EGFP, the GFP variants described in Doan et al, Mol. Microbiol, 55: 1767-1781 (2005), the GFP variant described in Crameri et al, Nat.
  • DsRed variants are described in, e.g., Wang et al, Proc. Natl. Acad. Sci. U.S.A., 101: 16745-16749 (2004) and include mRaspberry and mPlum. Further examples of DsRed variants include mRFPmars described in Fischer et al, FEBS Lett., 577:227-232 (2004) and mRFPruby described in Fischer et al, FEBS Lett, 580:2495-2502 (2006).
  • the imaging agent may be one or more cyanine dyes.
  • the cyanine dye may be but is not limited to indocyanine green (ICG), Cy5, Cy5.5, Cy5.18, Cy7 and Cy7.18, IRDye 78, IRDye 680, IRDye 750, IRDye 800 phosphoramidite, DY-681, DY-731, and DY-781.
  • the imaging agent may be a fluorescent dye suitable for near- infrared (NIR) fluorescence.
  • NIR imaging may be used for intraoperative visualization and non-invasive imaging of cells and tissues in a subject.
  • the NIR fluorescence imaging involves administration of a fluorescent contrast agent that can be excited at wavelengths of 780 nm or greater, and has a significant Stoke's shift emitting fluorescence at wavelengths of 800 nm or greater.
  • the imaging agent may be an agent suitable for imaging by magnetic resonance (MRI).
  • the imaging agents may comprise paramagnetic metal ions such as manganese (Mnll), iron (Felll), or gadolinium (Gd-III).
  • the imaging agent may be DOTA-Gd(ffl).
  • the molecular scaffold and the components covalently linked with the polymalic acid-based molecular scaffold may be linked to each other via a linker.
  • Linkers typically comprise a direct bond or an atom such as oxygen or sulfur, a unit such as NR 1 , C(O), C(0)OC, C(0)NH, SO, SO 2 , SO 2 NH, -SS- or a chain of atoms, such as substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, arylalkyl, arylalkenyl, arylalkynyl, heteroarylalkyl, heteroarylalkenyl, heteroarylalkynyl, heterocyclylalkyl, heterocyclylalkenyl, heterocyclylalkynyl, aryl, heteroaryl, heterocyclyl, cycloalkyl, cycloalkenyl, alkylaryl
  • the mini nanodrug may further comprise a PK modulating ligand covalently linked with the polymalic acid-based molecular scaffold.
  • PK modulating ligand and “PK modulator” refers to molecules which can modulate the pharmacokinetics of the imaging nanoagent.
  • the PK modulator can inhibit or reduce resorption of the imaging nanoagent by the reticuloendothelial system (RES) and/or enzyme degradation.
  • RES reticuloendothelial system
  • the PK modulator may be a PEG.
  • the PEG may be of any desired molecular weight.
  • the PEG may have a molecular weight of about 1,000 Da, about 1,500 Da, about 1,000 Da, about 2,500 Da, about 3,000 Da, about 3,500 Da, about 4,000 Da, about 4,500 Da, about 5,000 Da, about 10,000 Da, about 15,000 Da, about 20,000 Da, about 25,000 Da, or about 30,000 Da.
  • the PK modulator may be PEG of about 2,000 Da. Other molecules known to increase half-life may also be used as PK modulators.
  • the mini nanodrug may be of any desired size.
  • the mini nanodrug may be of a size that allows the mini nanodrug to cross the blood brain barrier via targeting or via transcytosis.
  • the mini nanodrug may range in size from about 1 nm to about 10 nm; from about 1 nm to about 2 nm; from about 2 nm to about 3 nm; from about 3 nm to about 4 nm; from about 4 nm to about 5 nm; from about 5 nm to about 6 nm; from about 6 nm to about 7 nm; from about 7 nm to about 8 nm; from about 8 nm to about 9 nm; from about 9 nm to about 10 nm.
  • the mini nanodrug may be about 5 nm to about 10 nm in size.
  • the mini nanodrug may be 10 nm or less in size.
  • the mini nanodrug may exhibit a distribution of sizes around the indicated "size.”
  • size refers to the mode of a size distribution of mini nanodrugs, i.e., the value that occurs most frequently in the size distribution.
  • Methods for measuring the size are known to a skilled artisan, e.g., by dynamic light scattering (such as photocorrelation spectroscopy, laser diffraction, low-angle laser light scattering (LALLS), and medium-angle laser light scattering (MALLS)), light obscuration methods (such as Coulter analysis method), or other techniques (such as rheology, and light or electron microscopy).
  • a pharmaceutically acceptable composition comprising any one the mini nanodrugs disclosed herein and a pharmaceutically acceptable carrier or excipient is provided.
  • An embodiment provides a method for treating a brain disease or abnormal condition.
  • the method may comprise administering a therapeutically effective amount of a composition comprising any one of the mini nanodrugs described herein to a subject in need thereof.
  • the method may further comprise analyzing the plaque formation in the subject affected or suffering from AD.
  • the step of analyzing may include observing more than about 50%, 60%, 70%, 80% or about 90% decrease in the formation of AD plaques in the subject.
  • the step of analyzing may include observing of the dissolution of AD plaques in the subject.
  • the step of analyzing may include observing stabilizing growth of the AD plaques in the subject.
  • the method may further comprise analyzing inhibition of tumor growth.
  • the step of analyzing may include observing more than about 60%, 70%, 80% or about 90% inhibition of tumor growth in the subject.
  • the step of analyzing may include observing the inhibition of HER2/neu receptor signaling by suppression of Akt phosphorylation.
  • Domestic and game animals include cows, horses, pigs, deer, bison, buffalo, feline species, e.g., domestic cat, canine species, e.g., dog, fox, wolf, avian species, e.g., chicken, emu, ostrich, and fish, e.g., trout, catfish and salmon.
  • Patient or subject includes any subset of the foregoing, e.g., all of the above, but excluding one or more groups or species such as humans, primates or rodents.
  • the subject may be a mammal, e.g., a primate, e.g., a human.
  • the terms, "patient” and “subject” are used interchangeably herein.
  • patient and “subject” are used interchangeably herein.
  • the subject is a mammal.
  • the mammal may be a human, non-human primate, mouse, rat, dog, cat, horse, or cow, but are not limited to these examples.
  • Mammals other than humans may be advantageously used as subjects that represent animal models for Alzheimer's disease.
  • Double or Triple Transgenic Alzheimer's mouse may be used.
  • Mammals other than humans may be advantageously used as subjects that represent animal models of cancer.
  • the methods described herein may be used to treat domesticated animals and/or pets.
  • a subject may be male or female.
  • a subject may be one who has been previously diagnosed with or identified a suffering from Alzheimer's disease, but need not have already undergone treatment.
  • a subject may be one who has been previously diagnosed with or identified as suffering from cancer, but need not have already undergone treatment.
  • a therapeutically effective amount is generally well within the capability of those skilled in the art. Generally, a therapeutically effective amount can vary with the subject's history, age, condition, sex, as well as the severity and type of the medical condition in the subject, and administration of other agents alleviate the disease or disorder to be treated.
  • the data obtained from the cell culture assays and animal studies may be used in formulating a range of dosage for use in humans.
  • the dosage of such compounds lies preferably within a range of circulating concentrations that include the ED50 with little or no toxicity.
  • the dosage may vary within this range depending upon the dosage form employed and the route of administration utilized.
  • the therapeutically effective dose may be estimated initially from cell culture assays.
  • a dose may be formulated in animal models to achieve a circulating plasma concentration range that includes the IC50 (i.e., the concentration of the therapeutic which achieves a half-maximal inhibition of symptoms) as determined in cell culture.
  • Levels in plasma may be measured, for example, by high performance liquid chromatography.
  • the effects of any particular dosage may be monitored by a suitable bioassay.
  • the dosage may be determined by a physician and adjusted, as necessary, to suit observed effects of the treatment.
  • the compositions may be administered so that the active agent is given at a dose from 1 ⁇ g/kg to 150 mg/kg, 1 ⁇ g/kg to 100 mg/kg, 1 ⁇ g/kg to 50 mg/kg, 1 ⁇ g/kg to 20 mg/kg, 1 ⁇ g/kg to 10 mg/kg, ⁇ g/kg to lmg/kg, 100 ⁇ g/kg to 100 mg/kg, 100 ⁇ g/kg to 50 mg/kg, 100 ⁇ g/kg to 20 mg/kg, 100 ⁇ g/kg to 10 mg/kg, 100 ⁇ g/kg to lmg/kg, 1 mg/kg to 100 mg/kg, 1 mg/kg to 50 mg/kg, 1 mg/kg to 20 mg/kg, 1 mg/kg to 10 mg/kg, 10 mg/kg to 100 mg/kg, 10 mg/kg to 50 mg/kg, or 10 mg/kg to 20 mg/kg.
  • ranges given here include all intermediate ranges, for example, the range 1 tmg/kg to 10 mg/kg includes lmg/kg to 2 mg/kg, lmg/kg to 3 mg/kg, lmg/kg to 4 mg/kg, lmg/kg to 5 mg/kg, lmg/kg to 6 mg/kg, lmg/kg to 7 mg/kg, lmg/kg to 8 mg/kg, lmg/kg to 9 mg/kg, 2mg/kg to lOmg/kg, 3mg/kg to lOmg/kg, 4mg/kg to lOmg/kg, 5mg/kg to lOmg/kg, 6mg/kg to lOmg/kg, 7mg/kg to lOmg/kg, 8mg/kg to lOmg/kg, 9mg/kg to lOmg/kg, and the like.
  • the compositions may be administered at a dosage so that the active agent has an in vivo concentration of less than 500 nM, less than 400 nM, less than 300 nM, less than 250 nM, less than 200 nM, less than 150 nM, less than 100 nM, less than 50 nM, less than 25 nM, less than 20, nM, less than 10 nM, less than 5 nM, less than 1 nM, less than 0.5 nM, less than 0.1 nM, less than 0.05, less than 0.01, nM, less than 0.005 nM, less than 0.001 nM after 15 mins, 30 mins, 1 hr, 1.5 hrs, 2 hrs, 2.5 hrs, 3 hrs, 4 hrs, 5 hrs, 6 hrs, 7 hrs, 8 hrs, 9 hrs, 10 hrs, 11 hrs, 12 hrs or more of time of administration.
  • the dosing schedule may vary from once a week to daily depending on a number of clinical factors, such as the subject's sensitivity to the peptides.
  • the desired dose may be administered every day or every third, fourth, fifth, or sixth day.
  • the desired dose may be administered at one time or divided into subdoses, e.g., 2-4 subdoses and administered over a period of time, e.g., at appropriate intervals through the day or other appropriate schedule.
  • Such sub-doses may be administered as unit dosage forms.
  • administration may be chronic, e.g., one or more doses daily over a period of weeks or months.
  • dosing schedules may include administration daily, twice daily, three times daily or four or more times daily over a period of 1 week, 2 weeks, 3 weeks, 4 weeks, 1 month, 2 months, 3 months, 4 months, 5 months, or 6 months or more.
  • administer refers to the placement of a composition into a subject by a method or route which results in at least partial localization of the composition at a desired site such that desired effect is produced.
  • a compound or composition described herein may be administered by any appropriate route known in the art including, but not limited to, oral or parenteral routes, including intravenous, intramuscular, subcutaneous, transdermal, airway (aerosol), pulmonary, nasal, rectal, or topical (including buccal and subhngual) administration.
  • Exemplary modes of administration include, but are not limited to, injection, infusion, instillation, inhalation, or ingestion.
  • injection include, without limitation, intravenous, intramuscular, intraarterial, intrathecal, intraventricular, intracapsular, intraorbital, intracardiac, intradermal, intraperitoneal, trans tracheal, subcutaneous, subcuticular, intraarticular, sub capsular, subarachnoid, intraspinal, intracerebro spinal, and intrastemal injection and infusion.
  • the compositions may be administered by intravenous infusion or injection.
  • the mini nanodrug may be provided in pharmaceutically acceptable compositions.
  • an embodiment also provides pharmaceutical compositions comprising the mini nanodrugs as disclosed herein.
  • These pharmaceutically acceptable compositions may comprise a therapeutically-effective amount of one or more of the mini nanodrugs, formulated together with one or more pharmaceutically acceptable carriers (additives) and/or diluents.
  • compositions may be specially formulated for administration in solid or liquid form, including those adapted for the following: (1) oral administration, for example, drenches (aqueous or non-aqueous solutions or suspensions), lozenges, dragees, capsules, pills, tablets (e.g., those targeted for buccal, subhngual, and systemic absorption), boluses, powders, granules, pastes for application to the tongue; (2) parenteral administration, for example, by subcutaneous, intramuscular, intravenous or epidural injection as, for example, a sterile solution or suspension, or sustained-release formulation; (3) topical application, for example, as a cream, ointment, or a controlled-release patch or spray applied to the skin; (4) intravaginally or intrarectally, for example, as a pessary, cream or foam; (5) sublingually; (6) ocularly; (7) trans derm ally; (8) transmucosally; or (9) nasally. Additionally, the mini oral
  • a variety of known controlled- or extended-release dosage forms, formulations, and devices may be adapted for use with the mini nanodrugs and compositions of the disclosure. Examples include, but are not limited to, those described in U.S. Pat. Nos.: 3,845,770; 3,916,899; 3,536,809; 3,598, 123; 4,008,719; 5674,533; 5,059,595; 5,591,767; 5, 120,548; 5,073,543; 5,639,476; 5,354,556; 5,733,566; and 6,365, 185 Bl, all of which are incorporated herein by reference as if fully set forth.
  • dosage forms may be used to provide slow or controlled-release of one or more active ingredients using, for example, hydroxypropylm ethyl cellulose, other polymer matrices, gels, permeable membranes, osmotic systems (such as OROS ® (Alza Corporation, Mountain View, Calif. USA)), or a combination thereof to provide the desired release profile in varying proportions.
  • active ingredients for example, hydroxypropylm ethyl cellulose, other polymer matrices, gels, permeable membranes, osmotic systems (such as OROS ® (Alza Corporation, Mountain View, Calif. USA)), or a combination thereof to provide the desired release profile in varying proportions.
  • OROS ® Alza Corporation, Mountain View, Calif. USA
  • the pharmaceutically acceptable composition may be formulated in dosage unit form for ease of administration and uniformity of dosage.
  • dosage unit form refers to a physically discrete unit of active agent appropriate for the subject to be treated.
  • the term "pharmaceutically acceptable” refers to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
  • the term "pharmaceutically-acceptable carrier” means a pharmaceutically-acceptable material, composition or vehicle, such as a liquid or solid filler, diluent, excipient, manufacturing aid (e.g., lubricant, talc magnesium, calcium or zincstearate, or steric acid), or solvent encapsulating material, involved in carrying or transporting the subject compound from one organ, or portion of the body, to another organ, or portion of the body.
  • manufacturing aid e.g., lubricant, talc magnesium, calcium or zincstearate, or steric acid
  • solvent encapsulating material involved in carrying or transporting the subject compound from one organ, or portion of the body, to another organ, or portion of the body.
  • Each carrier must be “acceptable” in the sense of being compatible with the other ingredients of the formulation and not injurious to the patient.
  • materials which may serve as pharmaceutically-acceptable carriers include: (1) sugars, such as lactose, glucose and sucrose; (2) starches, such as com starch and potato starch; (3) cellulose, and its derivatives, such as sodium carboxymethyl cellulose, methylcellulose, ethyl cellulose, microcrystalline cellulose and cellulose acetate; (4) powdered tragacanth; (5) malt; (6) gelatin; (7) lubricating agents, such as magnesium stearate, sodium lauryl sulfate and talc; (S) excipients, such as cocoa butter and suppository waxes; (9) oils, such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, com oil and soybean oil; (10) glycols, such as propylene glycol; (11) polyols, such as glycerin, sorbitol, mannitol and polyethylene glycol (PEG); (12) esters, such as e
  • wetting agents coloring agents, release agents, coating agents, sweetening agents, flavoring agents, perfuming agents, preservative and antioxidants may also be present in the formulation.
  • excipient carrier
  • pharmaceutically acceptable carrier or the likes are used interchangeably herein.
  • a mini nanodrug comprising a polymalic acid-based molecular scaffold
  • the mini nanodrug of embodiment 1, wherein the at least one peptide capable of crossing the blood-brain barrier is an LRP-1 hgand, or a transferrin receptor ligand.
  • mini nanodrug of any one or more of embodiments 1 - 5 wherein the at least one peptide crossing the blood-brain barrier is B6 peptide comprising an amino acid sequence of SEQ ID NO: 8, or a variant thereof.
  • the endosomolytic ligand comprises Trp-Trp-Trp (WWW), Phe-Phe- Phe (FFF), Leu-Leu-Leu (LLL), or Ile-Ile-Ile (I-I-I).
  • a mini nanodrug comprising a polymalic acid-based molecular scaffold
  • each of the at least peptide capable of crossing the blood-brain barrier, the endosomolytic ligand and the therapeutic agent are covalently linked to the polymalic acid-based molecular scaffold, and the nanodrug ranges in size from 1 nm to 10 nm.
  • mini nanodrug of embodiment 29, wherein the at least one peptide capable of crossing the blood-brain barrier is a peptide selected from the group consisting of Angiopep-2, Fe mimetic peptide, B6 peptide, and Miniap-4 peptide, or variants thereof.
  • mini nanodrug of any one or more of embodiments 29 - 32, wherein the at least one peptide capable of crossing the blood-brain barrier is B6 peptide comprising an amino acid sequence of SEQ ID NO: 8, or a variant thereof.
  • the at least one peptide capable of crossing the blood-brain barrier comprises at least two peptides, wherein each of the at least two peptides are independently selected peptides or similar peptides.
  • the therapeutic agent is selected from the group consisting of: an antisense oligonucleotide, an siRNA oligonucleotide, a peptide, and a low molecular weight drug.
  • mini nanodrug of embodiment 39 wherein the antisense oligonucleotide comprises a nucleic acid sequence with at least 90% identity to SEQ ID NO: 4.
  • plaque-binding peptide is a D-enantiomeric peptide selected from the group consisting of: a Dl-peptide, a D3-peptide and an ACI-89-peptide, or variants thereof.
  • plaque-binding peptide comprises two or more plaque-binding peptides independently selected from the group consisting of: a Dl-peptide, a D3-peptide and an ACI-89-peptide, or variants thereof.
  • plaque-binding peptide comprises two or more plaque-binding peptides selected from the group consisting of: a Dl-peptide, a D3-peptide and an ACI-89-peptide, or variants thereof, and the selected peptides are similar.
  • the mini nanodrug of embodiment 48 wherein the imaging agent comprises a fluorescence moiety, a radioisotope moiety, or a magnetic resonance imaging moiety.
  • the mini nanodrug of embodiment 28 or 51, wherein the poly(6-L- malic acid) has a molecular mass between 40 kDa and 60 kDa.
  • a pharmaceutically acceptable composition comprising a mini nanodrug of any one or more of embodiments 1 - 53 and a pharmaceutically acceptable carrier or excipient.
  • a method for reducing formation of amyloid plaques in the brain of a subject comprising administering the mini nanodrug of any one or more of embodiments 1 - 53, or composition of embodiment 54 to a subject in need thereof.
  • a method of detecting amyloid plaques in the brain of a subject comprising administering the mini nanodrug of any one or more of embodiments 1 - 25, 28 - 47, and 50 - 53, wherein the mini nanodrug further comprises an imaging agent comprising a fluorescence moiety, a radioisotope moiety, or a magnetic resonance imaging moiety; and visualizing the mini nanodrug.
  • a method for treating a proliferative disease in a subject comprising: administering a therapeutically effective amount of a mini nanodrug of any one or more of embodiments 29 - 38 or the composition comprising the mini nanodrugs of any one of embodiments 29 - 38 and a pharmaceutically acceptable carrier or excipient to the subject in need thereof.
  • 69 The method of embodiment 68, wherein the mammal is selected from the group consisting of: a rodent, a canine, a primate, an equine, an experimental human-breast tumor-bearing nude mouse, and a human.
  • Example 1 Design of mini nanodrugs for efficient crossing blood-brain barrier
  • the nanoconjugate drug delivery system is also referred to herein as a mini nanodrug.
  • the designed mini nanodrugs are characterized by hydrodynamic diameter 5-8 nm, elongated shape and ability of chemical attachment of drugs and operational groups, e.g. receptor targeting, to a polymer platform.
  • the elongated shape enables the mini nanodrug for rapid diffusion compared to spherical nanodrugs of the same mass, and to pass through pores of narrow diameter.
  • the platform also provided chemical anchorage for various modules designed for endosome disruption, MRI and fluorescence imaging or protection. Cleavable linkers can be used that enable drug activation in response to chemistry in the targeted compartment.
  • the mini nanodrugs may be developed for highly efficient treatment of preclinical HER2-positive human breast cancer by replacement of targeting antibodies with HER2-affine peptide.
  • the mini nanodrug may be designed to deliver multiple copies of antisense oligonucleotide or docetaxel to the cytoplasm and arrested tumor growth. Delivery of imaging agents may be achieved across the blood-brain barrier (BBB) with peptides targeting different delivery routes when attached separately or combination of routes when attached simultaneously.
  • BBB blood-brain barrier
  • Another design may be a targeted mini nanodrug carrying the NIR fluorescent dye ICG that brightly lights up glioblastoma in mice for imaging guided tumor resection. In all the designs, mini nanodrugs are cleared with half -lives of one hour and residing times of several hours inside tumors or other targeted regions.
  • mini nanodrugs to treat Alzheimer disease is described herein. Despite multiple attempts to persistently treat Alzheimer disease, a satisfactory prevention of toxic ⁇ production is still not in sight. Treatment with a nanosized multi drug delivery platform is described herein that was designed for efficious targeted multi-prone inhibition of soluble A6 production. In applying nanocarrier cascade targeting of multiple BBB crossing transcytosis pathways and of agents/cells in the brain, the treatment exceeds the outcome of existing attempts in efficacy, absence of side effects and improved image guided control.
  • PMLA polymalic acid or P
  • PMLA was selected as platform for mini nanodrug development because PMLA is completely biodegradable to carbon dioxide and water, biologically inert, nontoxic and nonimmunogenic. PMLA also carries abundant carboxyl groups that can be conjugated with multiple targeting and therapeutic moieties, ultimately constituting a mini nanodrug platform that can carry any number and type of functional moieties.
  • LRP-1 low-density lipoprotein receptor pathway
  • LRP-1 mediated blood-to-brain transport occurs when suitable ligands bind to and become internalized by LRP-1 in the vascular endothelium. After internalization, LRP-1 bound ligands are transcytosed into the brain parenchyma.
  • a synthetic LRP-1 peptide ligand, Angiopep-2 (AP-2; TFFYGGSRGKRNNFKTEEY (SEQ ID NO: 1)) was identified by Demeule et al. (Demeule et al. (2008), which is incorporated herein by reference as if fully set forth). It was reported that the transport of AP-2 saturates at high concentrations and is inhibited by other LRP-1 ligands, confirming AP-2 transcytosis. AP-2 was selected for initial screening.
  • TfR transferrin receptor
  • Fe mimetic peptide ((SEQ ID NO: 2), CRTIGPSVC -NH2, cyclic, S-S bonded) was isolated via phage display and has been shown to deliver cargo into brain tumors (Staquicini et al. (2011), which is incorporated herein by reference as if fully set forth). Fe mimetic peptide, or cTfRL was also selected for the design.
  • TfR ligand B6 (CGHKAKGPRK (SEQ ID NO: 9)
  • CGHKAKGPRK SEQ ID NO: 9
  • Miniap-4 also referred to herein as M4; H-[Dap] KAPETAL D-NH 2 (SEQ ID NO: 3), a cyclic peptide that was derived from bee venom. This peptide was reported to be capable of translocating proteins and nanoparticles across a human cell-based BBB model, (Oller-Salvia et al. (2016), which is incorporated herein by reference as if fully set forth).
  • BBB penetrating peptides have inherent therapeutic value(s) and they have not been designed to carry reversibly bound cargoes by themselves. These peptides were selected as components of cargo delivery molecules and were examined to determine how conjugation with other peptide or non-peptide moieties influences their BBB penetration abilities.
  • the mini nanodrugs based on the PMLA backbone conjugated to synthetic peptides that enable BBB penetration were additionally designed to carry tri-leucine (LLL).
  • LLL displays pH-responsive lipophilicity and promotes endosomal escape of PMLA bound agents once they are internalized and part of the endosomal pathway. Endosomal escape for cytoplasmic drug delivery was described for intracellular drug treatment (Ding et al. (2011), which is incorporated herein by reference as if fully set forth).
  • the mini nanodrugs were also conjugated to rhodamine in order to visualize the compound in brain tissues.
  • mini nanodrugs were initially designed to be neutral to test their ability to penetrate BBB and be distributed over all brain regions which could potentially be affected by neurological disorders.
  • mini nanodrugs were designed for multi targeted systemic delivery of antisense oligo nucleotides (AONs) and chemotherapeutics across blood brain barrier (BBB) to silence AB production.
  • AONs antisense oligo nucleotides
  • BBB blood brain barrier
  • the mini nanodrugs can be conjugated to ⁇ -sheet breaker peptides.
  • ⁇ -sheet breaker peptides are o designed to specifically interfere with 6-sheets within A6 preventing the misfolding and deposition of A6 and decreasing toxicity e.g. H102 (HKQLPFFEED; SEQ ID NO: 7) peptide (Zhang et al. (2014), which is incorporated herein by reference as if fully set forth).
  • H102 HKQLPFFEED; SEQ ID NO: 7
  • peptide Zhang et al. (2014), which is incorporated herein by reference as if fully set forth.
  • FIG. 1 is a schematic drawing illustrating overview of molecular pathway of mini nanodrugs.
  • the mini nanodrugs are i.v. injected into a subject.
  • the massive flux (flux 1) is maintained by binding of different attached peptides that target specific barriers, such as endosomal membrane, cellular membrane, intracellular matrix, extravasion, along this mini nanodrug pathway. Multiple peptides targeting different pathways to same barriers would increase the overall flux of drug delivery through barriers.
  • the covalent attached drug(s) are cleaved from the nanocarrier by enzymatic reaction or spontaneous reaction with reactant contained only in the targeted site of treatment (i.e. hydrogen ions (pH), or Glutathion-SH for reductive cleavage of disulfide linkers of drug with carrier).
  • reactant contained only in the targeted site of treatment i.e. hydrogen ions (pH), or Glutathion-SH for reductive cleavage of disulfide linkers of drug with carrier.
  • Another flux is directed to renal clearance.
  • the mini nanodrugs were designed to carry peptides and specifically target to neuron cells which overproduce the A6 precursor peptides (APPs).
  • the mini nanodrugs were designed to carry antisense oligonucleotides (AONs) to silence mRNAs, and thus, biosynthesis of ⁇ -secretase and/or ⁇ -secretase for A6 production.
  • FIG. 2 is a schematic drawing illustrating mini nanodrugs carrying peptides that permeate through multiple bio barriers into targeted neurons, chemo, AONs, and peptides targeting APP and A6.
  • AON inhibiting the syntheses of 6-secretase
  • another kind of AON is an AON inhibiting the synthesis of ⁇ -secretase presenilin 1 (the enzyme active) subunit.
  • the mini nanodrug further carries drugs (marked as "chemo" on FIG. 2) to inhibit the secretase activities.
  • the mini nanodrug further carries trileucine for release of the delivery system across the endosome membrane into the cytoplasm.
  • the mini nanodrug further carries optionally Cy 5.5 (NIR fluorescence), Phalloidin (red fluorescence) or DOTAGd(III) for fluorescence imaging or imaging by magnetic resonance (MRI).
  • the mini nanodrug permeates BBB and unfolds inhibition of A6 by blocking 6- and ⁇ -secretase protein syntheses and enzyme activities (contained in cytoplasma and/or organelles).
  • the peptides angiopep-2, cyclic MiniAp-4, cyclic CRTIGPSVC (SEQ ID NO: 2)- peptide target the delivery across BBB on parallel routes of transcytosis. Transcytosis of high flux competes successfully with vascular clearance.
  • An amyloid targeting peptide specifically adheres the mini nanodrug to amyloid precursor peptides (APP) on the surface of A6 overproducing neurons.
  • APP and adhering mini nanodrug are internalized into the endosomal system for cleavage by ⁇ -secretase and release of AONs and secretase inhibitory drugs.
  • AONs released into the cytoplasm specifically inhibit the biosynthesis of 6- secretase and ⁇ -secretase.
  • the membrane permeable drugs inhibit secretase cleavage of APP and release of A6 into extracellular space. Absence of A6 production stops ⁇ aggregation, fibril formation and toxic reactions. Dissolution of existing plaques occurs with time or may be accelerated by treatment with aggregate disrupting reagents (e.g., peptides and synthetics).
  • the mini nanodrugs consisting of degradable non-immunogenic systemic IV-injectable nanoagent is suitable for imaging and treatment of Alzheimer disease.
  • the mini nanodrug can be applied for treatment of other neurological disorder by use of appropriate peptides, chemotherapeutics and antisense oligonucleotides. Because of the multiplicity of attachment sites on the PMLA carrier, the mini nanodrug can be equipped with multiple chemotherapeutics and DNA-antisense drugs for blockage of Alzheimer specific markers. Attachment of chemotherapeutics and oligonucleotides to the mini nanodrug is reversible when responding to local pH or glutathion and suits drug activation inside targeted cells. Reagents carry dyes for NIR or IR image guided space and time resolved analysis.
  • FIGS. 3A - 3B are schematic drawings illustrating advantages of mini nanodrugs for crossing the blood-brain barrier and entering brain parenchima.
  • FIG. 3 A is a schematic drawing illustrating mini nanodrugs carrying AP-2 peptides and tri-leucins (endosomic escape units) entering brain parenchima.
  • the mini nanodrugs for fast delivery and deep penetration were designed to be 6-10 nm size and have an elongated architecture. This was achieved by attaching low molecular targeting peptides to PMLA instead of antibodies.
  • FIG. 3B is a schematic drawing comparing the efficiency of crossing the blood-brain barrier of a mini nanodrug carrying peptides and nanodrugs that carry antibodies.
  • Polymalic acid is an unbranched polymer and macromolecule with multiple pendant carboxylic groups for attachment of a diversity of pharmaceutical functional modules.
  • the linear organization of structurally highly flexible polymalic acid allows enhanced diffusion through interstitial space and optimal accessibility of multiple peptides with interacting sites.
  • the small molecular size on the lower nanoscale and the molecular flexibility provide an optimal penetration in brain.
  • Favorable high influx from circulating vasculature into brain is obtained by attachment of several different affinity peptides that engage simultaneously in binding to multiple sites and BBB crossing pathways of different specificity.
  • second peptides target specific markers of Alzheimer or of other neurodegenerative diseases.
  • NIR fluorescent dyes are attached for imaging, and chemotherapeutic drugs and antisense oligo nucleotides for treatment.
  • Peptides have low immunogenicity, are robust against denaturation and in an exocyclic form less vulnerable by enzymatic cleavage. Peptides have less affinity and hence favorable release kinetics after receptor binding. Conjugation of targeting peptides with multi attachment sites carried by polymalic acid increases influx of functional groups for inside targeting, imaging and treatment.
  • the mini nanodrugs can be useful in addressing the problem of poor availability of delivery pathways across BBB and their inefficacy to manage large nan op articles, instability and long circulation times prone for loss of cargo and induction of systemic side effects.
  • the mini nanodrugs can be used for solving additional problems such as expensive production (antibodies), limited shelf life, difficult to manage shipment in solution, and the necessity to apply large volumes for patient application.
  • the mini nanodrugs can be used for solving the problem of incomplete inhibition of secretases and high degree of side effects caused by lack of targeting producer cells, and the need of imaging to control progress of treatment.
  • the nanocarrier's structure is designed for fast diffusion and easy barrier penetration, excellent access of interaction sites, attachment of agents for optical (fluorescence) and magnetic imaging (MRI). Manageable costs by simplified production, storage, shipping, and patient application.
  • ⁇ peptide overproducing cells are peptide targeted. Targeting was also addressed to silence over production of proteins and peptides. Silencing employs antisense oligonucleotides in a multi-pronged initiative and includes inhibition by chemo therapeutics.
  • FIG. 4 illustrates synthesis of the mini nanodrug with a single peptide.
  • the mini nanodrug has capability for the extension to specific cascade targeting across BBB to addressed brain cells.
  • the flow of synthesis starts on the upper left corner with NHS activation of polymalic acid (PMLA).
  • PMLA polymalic acid
  • Activation is followed by amide forming substitution with tricleucine (LLL) consuming 40% of pendant activated carboxylates, then by amide forming substitution with 2-mercapto ethylamine (MEA) (10% of available carboxylic groups or consuming an optional amount of activated carboxylates) to achieve the intermediate product termed "preconjugate".
  • LLL polymalic acid
  • MEA 2-mercapto ethylamine
  • the sulfhydryls on the PMLA scaffold react with maleimide tagged peptides and imaging groups forming the corresponding thioether conjugates.
  • the conjugation of peptides to present the maleimide reactive groups employs commercially available bifunctional PEG2000/3400-linkers attached to reactive groups on peptides (and dyes, if required) (see scheme in the upper right corner of the Scheme).
  • Morpholino oligonucleotides (AONs) are loaded by disulfide exchange of preconjugate-SH with 3-pyridyldithiopropionyl-3'-amido-AON (Ljubimova et al. (2014), which is incorporated herein by reference as if fully set forth).
  • FIG. 5 illustrates an example of the nanoconjugate with three peptides.
  • the peptides Angiopep-2-cys (containing an additional C-terminal cysteine group; TFFYGGSRGKRNNFKTEEYCNH 2 (SEQ ID NO: 1)), Angiopep-7-cys (TFFYGGSRGRRNNFRTEEYCNH 2 (SEQ ID NO: 7)), B6 (CGHKAKGPRK (SEQ ID NO: 9)), M4 (H-[Dap] KAPETAL D-NH 2 (SEQ ID NO: 3)), and cTfRL, also referred herein as the Fe mimetic peptide, (CRTIGPSVC-NH2, (SEQ ID NO: 2), S-S bonded) were custom synthesized by AnaSpec (Fremont, CA, USA).
  • Rhodamine-maleimide was purchased from ThermoFisher Scientific (Canoga Park, CA, USA). Mal-PEG3400-Mal or Mal- PEG2000-Mal was purchased from Creative PEGWorks (Durham, NC, USA). Tri-Leucine was ordered from Bachem (Torrance, CA, USA) while the reagents DCC, NHS, TFA, MEA and DTT were obtained from Sigma (St. Louis, MO, USA).
  • Products peptides were stored a -20 °C or lyophilized.
  • N-hydroxy succinimide (NHS, 115 g/mol, 9.6mg, 0.083 ⁇ , 50 mole% of PMLA COOH) and N,N'-Dicyclohexylcarbodiimide (DCC, 206 g/mol, 17.7 mg, 0.086 ⁇ , 50 mol% of PMLA COOH) were dissolved in 500 ⁇ L of DMF and added drop wise to the reaction mixture, followed by 15 mg of dithiothreitol (DTT, 154.25 g/mol, 0.097 ⁇ ) in 38 ⁇ L of DMF and then cysteamine (MEA, 113.61 g/mol, 1.9 mg, 0.017 ⁇ , in 7.8 ⁇ L DMF) and Et 3 N (2.3 ⁇ L, 1 eq to MEA).
  • DTT dithiothreitol
  • MEA cysteamine
  • reaction extent was monitored using TLC (n-BuOH:H20:AcOH 5: 1: 1) and ninhydrin reaction.
  • DTT dithiothreitol
  • MEA cysteamine
  • Et 3 N Et 3 N
  • the lyophihzed product (10 mg/mL in phosphate buffer with pH 6.3) was used for the reaction with PMLA preconjugate (SEC-HPLC analysis: retention time 8.2 min at 220 nm wavelength).
  • Angiopep-7-PEG3400-Mal (SEC-HPLC retention 8.25 min at 220 nm)
  • B6-PEG-Mal (SEC-HPLC retention 7.92 min at 220 nm) were synthesized in the same manner.
  • HPLC pump Hitachi L-2130; detector, Hitachi L-2455; software, EZChrome; Column, Polysep 4000; flow rate: lml/min; buffer, PBS.
  • Miniap-4-PEG2000-Mal In a glass vial with magnetic stirrer (ambient temperature), Mal-PEG-SCM 2000 (2000 g/mol, 5.5 mg, 2.76 ⁇ , 1.2 eq) was dissolved in 200 ⁇ , of DMF.
  • the reaction was monitored using HPLC (same conditions mentioned above), and 0.3 eq of Mal-PEG2000-SCM (1.32 mg in DMF) and O. ⁇ L of Et 3 N were added in case the reaction was not progressing. Much excess of Mal-PEG2000-SCM and an overnight reaction were avoided to keep side reactions with lysine at a minimum.
  • the reaction was purified using PD- 10 column, analyzed using HPLC and lyophilized. A solution of 10 mg/mL product in phosphate buffer 6.3 was used for the reaction with PMLA preconjugate.
  • the reaction was monitored using HPLC (usually overnight), and 0.1 ⁇ ⁇ of Et3N were added in case the reaction was not progressing.
  • the reaction was purified using a PD-10 column, analyzed using HPLC, and lyophilized.
  • Miniap-4-PEG2000-Mal was synthesized in the same manner, using the N-terminus and the succinimidyl carboxyl methyl ester reaction for attachment.
  • peptide-PEG-MAL 2% (0.314 ⁇ ) of peptide-PEG-MAL were added dissolved in phosphate buffer pH 6.3 to 10 mg/mL of optional peptide-linker-Mal: optionally 1.82 mg of angiopep-2-PEG3400-MAL (5802.7 g/mol) or 0.88 mg of "Fe mimetic peptide" (SEQ ID NO: 2): CRTIGPSVC (cyclic)-peptide-PEG2000- Mal (2817 g/mol) or 0.88 mg of Miniap-4-PEG2000-Mal (2796 g/mol), or buffer without peptide (control).
  • optional peptide-linker-Mal optionally 1.82 mg of angiopep-2-PEG3400-MAL (5802.7 g/mol) or 0.88 mg of "Fe mimetic peptide” (SEQ ID NO: 2): CRTIGPSVC (cyclic)-peptide-PEG2000- Mal (2817 g/mol) or 0.
  • the reaction mixture was monitored at 220 nm by HPLC (typically 1 h reaction) and, once completed, Rhodamine C2*) was loaded by thioether formation with the PMLA platform -SH (0.107 mg for 1% loading, 680.79 g/mol, 0.153 ⁇ , 52.2 ⁇ L of 2 mg/mL solution in DMF).
  • the reaction under exclusion of light was monitored using HPLC. Absorbance spectra were recorded to detect dye absorbance in the PMLA conjugate elution peak.
  • 15 ⁇ L of 3-(2- pyridyldithiopropionic acid) or PDP (10 mg/mL solution in DMF) was added to cap the free SH groups.
  • the reaction was stirred for an additional hour and purified over PD- 10 column, analysed by HPLC, lyophilized and stored at -20 °C. *)
  • NIR dye Cy5,5 was also used for fluorescence labeling.
  • peptide-PEG-MAL were added dissolved in phosphate buffer pH 6.3 to 10 mg/mL concentration: optionally 1.78 mg of angiopep-2-PEG3400-MAL (5802.7 g/mol) or 0.86 mg of "Fe mimetic peptide" (SEQ ID NO: 2): CRTIGPSVC (cyclic)-peptide-PEG2000-Mal (2817 g/mol) or 0.88 mg of Miniap- 4-PEG2000-Mal (2796 g/mol), or buffer without peptide (control). The reaction was continued as under S7.
  • rhodamine- maleimide (0.104 mg for 1% loading, 680.79 g/mol, 0.149 ⁇ , 52 ⁇ L of 2 mg/mL solution in DMF) was loaded onto the conjugates forming thioethers with the PMLA platform at pendant MEA-SH.
  • the reaction was conducted in the dark and was monitored using HPLC. Success of the conjugation was indicated by the rhodamine absorbance in the PMLA conjugate elution peak.
  • 15 ⁇ ⁇ of 3-(2- pyridyldithio)propionic acid (10 mg/mL solution in DMF) was added to cap the free SH groups.
  • the product was purified over a PD- 10 column, analyzed, lyophilized and stored at -20 °C.
  • peptide-PEG-MAL For 2% loading, (0.077 ⁇ ) of peptide-PEG-MAL were added dissolved in phosphate buffer pH 6.3 to 10 mg/mL concentration: optionally 21.5 ⁇ L "Fe mimetic peptide" (SEQ ID NO: 2): CRTIGPSVC (cyclic)-peptide-PEG2000-Mal (2817 g/mol) or 0.215 mg (21.5 ⁇ ) of Miniap-4-PEG2000-Mal (2796 g/mol). The reaction is monitored using HPLC.
  • the second peptide is added: optionally 0.445 mg of angiopep-2-PEG-MAL 3400 (5802.7 g/mol) or 0.215 mg of "Fe mimetic peptide" (SEQ ID NO: 2): CRTIGPSVC(cyclic)-peptide- PEG2000-Mal (2817 g/mol, in case of miniap-4 was the first peptide).
  • the reaction mixture was monitored at 220 nm using HPLC (typically 1 h reaction) and once completed, Rhodamine C2 was added (0.026 mg for 1% loading, 680.79 g/mol, 0.38 ⁇ , 13.05 ⁇ L of 2 mg/mL solution in DMF) and the reaction under exclusion of light was monitored using HPLC.
  • the reaction is monitored at 220 nm and dye absorbance using HPLC, and is typically complete after lh.
  • the second peptide is added: optionally 21.5 ⁇ ⁇ of "Fe mimetic peptide” (SEQ ID NO: 2): CRTIGPSVC (cyclic)-peptide-PEG2000-Mal (2817 g/mol) or 0.215 mg (21.5 ⁇ _) of Miniap-4-PEG2000-Mal (2796 g/mol).
  • Peptide sequences a TFFYGGSRGKRNNFKTEEY (SEQ ID NO: 1); b TFFYGGSRGRRNNFRTEEYCNH2 (SEQ ID NO: 7); ° H-[Dap] KAPETAL D- NH2 (SEQ ID NO: 3), cyclic; d CRTIGPSVC-NH2 (SEQ ID NO: 2), cyclic, S-S bonded; * CGHKAKGPRK (SEQ ID NO: 9).
  • FIGS. 6A - 6D illustrate synthetic route for PMLA/LLL/Angiopep- 2/rhodamine (P/LLL/AP2) nanoconjugate.
  • FIG. 6A illustrates activation of biosynthesized PMLA was using a DCC/NHS chemistry to create the activated PMLA.
  • FIG. 6B illustrates conjugation of the activated PMLA with LLL and MEA
  • FIG. 6C illustrates conjugation of PMLA/LLL to Angiopep-2 (AP2) and rhodamine dye.
  • FIG. 6D illustrates that MEA moiety was used to bind AP2 peptide conjugated to a PEG linker via a Maleimide-thiol reaction. Rhodamine was attached in the same manner.
  • FIGS. 7A-7G Examples of product verification by HPLC are illustrated on FIGS. 7A-7G.
  • FIG. 7A illustrates verification of PMLA/LLL/ angiopep-2-PEG3400- MAL /rhodamine.
  • FIG. 7B illustrates verification of PMLA/ LLL/"Fe mimetic peptide" CRTIGPSVC (SEQ ID NO: 2)(cyclic)-peptide-PEG2000- Mal/rhodamine.
  • FIG. 7C illustrates verification PMLA/LLL/Miniap -4- PEG2000-Mal/cy 5.5.
  • FIG. 7D iUustrates control: PMLA LLL/rhodamine.
  • FIG. 7E illustrates PMLA LLL/angiopep2(2%)/"Fe Mimetic
  • FIG. 7F illustrates PMLA/ LLL/ angiopep-2(2%)/miniap-4(2%)/rhodamine (1%) dipeptide for targeting.
  • FIG. 7G illustrates PMLA LLL/miniap-4 (2%)/angiopep-2 (2%)/"Fe mimetic Peptide” (2%)/rhodamine (1%) tripeptide for targeting.
  • FIGS. 8A - 8C illustrate characterization of synthesized P/LLL/AP2.
  • FIG. 8A illustrates SEC-HPLC 3D view of A200-A700 nm vs. retention time and absorbances of the P/LLL/AP2 nanoconjugate constituents.
  • FIG. 8B illustrates SEC-HPLC chromatogram of P/LLL/AP2 recorded at 220 nm wavelength.
  • FIG. 8C illustrates FTIR spectrum of P/LLL/AP2 nanoconjugate (dashed ine), AP2 free peptide (solid lined) and pre-conjugate (dashed-dotted line). Arrows in FIG. 8C indicate peak shifts in the P/LLL/AP2 conjugate compared with AP2 peptide and preconjugate.
  • the FTIR spectrum of P/LLL/AP2 contains several distinctive peaks that can be attributed to both the pre-conjugate and the pristine AP2 peptide, while some peaks were shifted or decreased in intensity.
  • a prominent peak shift is visible from 3050 cm 1 in the pre- conjugate spectrum to 3057cm- 1 in the P/LLL/AP2 spectrum as well as other changes in peaks at the lower frequencies of 1040, 1104 and 950 cm 1 .
  • the SEC-HPLC analysis of all conjugates above was performed using a Hitachi L-2130 pump with a Hitachi L-2455 detector with EZChrome Software.
  • the column that was used was a Polysep 4000, and the flow rate lml/min; the buffer was PBS (pH 7.4).
  • Dl-peptide (QSHYRHISPAQVC (SEQ ID NO: 10)), all D-amino acids;
  • D3-peptide RPR TRL HTH RNRC(SEQ ID NO: 11)
  • aU D-amino acids and ACI-89 (PSHYRHISPAQKC (SEQ ID NO: 12)), all D-amino acids.
  • the glass vial was covered with aluminum foil and Rhodamine C2 was added (0.0516 mg for 1% loading, 680.79 g/mol, 25.8 ⁇ ⁇ of 2 mg/mL solution in DMF) and reaction was monitored again using HPLC. Mixed view required to see dye absorbance in the PMLA peak. Typically, the reaction should be stirred for lh. Then, 15 ⁇ L of 3-(2-pyridyldithiopropionic acid) or PDP (10 mg/mL solution in DMF) was added to cap the free SH groups. The reaction was stirred for an additional hour before purification using PD-10 column, HPLC analysis and freeze drying.
  • FIGS. 10A - IOC illustrate characterization of synthesized P/LLL/AP-2/ACI-89/rhodamine
  • FIG. 10A illustrates SEC-HPLC top view of scanning A200-A700 nm vs. retention time displaying absorbances of the complete nanoconjugate.
  • FIG. 10B illustrates the scanning profile of the same conjugate as shown on FIG. 10A at 572 nm wavelength indicating the rhodamine is part of the physical entity.
  • FIG. IOC illustrates the scanning profile of the same conjugate as shown on FIG. 10A at 220 nm wavelength indicating the P/LLL/ AP-2/ACI-89 is part of the physical identity.
  • FIGS. 11A - 11C illustrates SEC-HPLC chromatogram of P/LLL/AP- 2/D1- peptide/rhodamine at A200-A700 nm vs. retention time displaying absorbancies of PMLA/LLL/AP-2/Dl-peptide/rhodamine complete nanoconjugate.
  • FIG. 1 IB is a scanning profile of the same nanoconjugate as shown on FIG. 11A at 572 nm indicating the rhodamine component.
  • FIG. 11C is a scanning profile of the same nanoconjugate as shown on FIG. 11A at 220 nm indicating the PMLA/LLL/AP-2/D1- peptide component.
  • Copolymers were subjected to hydrolytic cleavage in sealed ampoules containing 2 M HC1 for 12 h at 100 °C. Malic acid in the hydrolysate was quantified by a colorimetric method based on an enzymatic reaction using malate dehydrogenase (Rozemaet al. (2003) Bioconjugate Chemistry, 14, 51-57, which is incorporated herein by reference as if fully set forth).
  • FTIR measurements A dry sample of the materials tested was added to KBr powder and scanned using a Bruker Alpha instrument with a DRIFT module (Bruker, Billerica, Ma, USA). KBr alone was used for the background scan.
  • mice Eight to nine week old BALB/C mice were obtained from Charles River Laboratories (Wilmington, MA, USA). Mouse maintenance and experimental procedures followed the guidelines established by the Cedars Sinai Institutional Animal Care and Use Committee (IACUC Protocol #7416). Three to four mice of each sex were used for each experiment. A total of 110 mice were used to produce the data described herein.
  • Nanoconjugates were dissolved freshly in PBS (pH 7.4) prior to each experiment and injected intravenously (i.v.) into the lateral tail vein. Mice were anesthetized with isoflurane beforehand and their tails were briefly warmed to allow access to the tail vein. All conjugates were administered as a single dose. Conjugates were injected at a final concentration of 29.5 to 236 ⁇ of total nanoconjugate per Kg bodyweight, as indicated for each experiment. The drug injection volume was kept constant at 150 ⁇ After each injection, mice were promptly returned to their cages.
  • Retroorbital blood collection & tissue collection Blood was drawn from the retroorbital sinus at multiple timepoints to measure the concentration of drug in the serum. Time points ranged from 30 to 480 minutes and are indicated separately for each experiment. Blood was collected with a microhematocrit capillary tube (I.D. 1.1mm; Chase Scientific Glass, Rockwood, TN, USA) and 150 ⁇ blood was collected per mouse into a BD Microtainer SST and stored at room temperature for 45 min, and then centrifuged at 6000 rpm for 5 min. The serum was then transferred into fresh tubes and stored at -80°C until further use.
  • a microhematocrit capillary tube I.D. 1.1mm; Chase Scientific Glass, Rockwood, TN, USA
  • mice were euthanized at predetermined timepoints. Euthanasia was conducted by spinal dislocation of deeply anesthetized animals; the brain, spleen, liver, heart, lungs and kidneys were promptly removed, flash frozen, and placed into -80°C storage. AH tissue used for microscopic analysis was embedded in optimal cutting temperature compound (OCT; Sakura, Torrance, CA, USA) and placed on dry ice for freezing.
  • OCT optimal cutting temperature compound
  • PK measurements using serum Fluorescently -labeled nanoconjugates with known concentrations (in ⁇ /mL) were used to obtain standard fluorescence calibration curves, which were used to convert raw fluorescent measurements in collected serum to ⁇ /mL units shown in this paper. Amounts of 20 ⁇ ⁇ of the processed blood serum containing injected conjugates were placed in 96-well white opaque plates and the fluorescence was measured using a fluorimeter at 570 / 600nm excitation / emission with a 590nm cutoff (Flexstation, Molecular Devices, Sunnyvale, CA, USA). Results were converted to ⁇ g / mL using the calibration curve and plotted as a function of time.
  • PK half-life ti/2 values were calculated using Prism (Graphpad, LaJolla, CA, USA).
  • Optical drug clearance measurements e.g., FIG. 21A, 21C
  • Vascular fluorescence was defined as the difference between fluorescent peaks and shoulders in a linear profile that was drawn perpendicularly across the blood vessel (see FIG. 21C). The sequential decrease in fluorescence was then converted to ⁇ /mL via calculation with a fluorescent standard with a known concentration, and plotted alongside serum measurements in FIG. 21 A.
  • Tissue processing & staining The cerebral vasculature was stained in every experiment in order to differentiate blood vessels from brain parenchyma. In most experiments (FIGS. 15A - 15C, 16A-16B, 17A-17B, 20A- 20E, 22A-22C) DyLight 488 tomato-lectin (DL-1174; Vector Laboratories, Burlingame, CA) was injected as a 150 ⁇ bolus at a 1:2 dilution in saline, 15 minutes prior to euthanasia. This led to widespread and optimal staining of the vasculature. Immunohistochemical staining of the vasculature was performed for tissue shown in FIGS. 21A-21D.
  • Imaging was performed with a Lecia DM 6000B epifluorescence microscope (Leica Microsystems, Wetzlar, Germany). Rhodamine-labeled nanoconjugates were visualized with a 534-558 nm excitation and 560-640 nm emission filter set, viewed with a 20X Leica HC Plan Apo 0.70 N.A. and a 40X Leica HCX Plan Apo 0.85 N.A. lens, and recorded with a Leica DFC 360 FX camera. The camera was controlled with Leica LAS X software and images were acquired with 4.5 sec + 2.0 gain exposures for the 20X lens and 3.5 sec + 2.0 gain exposures for the 40X lens. These parameters were held constant throughout the imaging experiment to enable accurate image-to-image comparisons across trials and experiments. Other fluorophores (DAPI, tomato- lectin, antibodies) were viewed using complementary standard filter sets and their imaging parameters were also held consistent across experimental trials.
  • DAPI DAPI, tomato- lectin, antibodies
  • FIG. 9 iUustrates PK for PMLA/angiopep-2 (2%)/rhodamine (1%) conjugate measured by fluorescence intensity of the attached dye as a function of time from IV injection into tail vain until blood samples were taken.
  • the sample fluorescence intensity was converted to mg injected nanoconjugate on the basis of standard curves obtained by spiking blood samples with known mg-amounts of conjugate and botting fluorescence intensity as function of mg nanoconjugate.
  • the drawn curve in FIG. 9 was calculated for the obtained best fit to the experimental points. Parameters shown in Table 3 below were calculated on the basis of the curve.
  • the second phase is considered and follows the half life of 1.31 h. Residual amount of nanoconjugate after 4 h from injection is less than 6 %.
  • PMLA/LLL(40%)/Angiopep-2(2%)/rhodamine(l%) nanoconjugate was IV (tail) injected into healthy nude mice. Ex vivo brain slices were examined at 0.5 hours, 1 hour, 2 hours and 4 hours after injection. It was observed that the nanoconjugate was visible around blood vessels for two hours and almost disappeared at 4 hours after injection of the nanoconjugate.
  • nanoconjugates that do not carry A6 binding peptide do not show depositions at AD plaques in Alzheimer diseased mice. It was also observed that deposition of dye fluorescence was independent of type of dye at characteristic fluorescence wave lengths.
  • FIG. 13 is an image of the left hippocampus CAl 2 hours after (IV) injection of buffer into the tail vain of a healthy mouse.
  • the location of the fluorescent spots was observed to be next to nuclei, have excitable fluorescence in the green and red wavelength region and have been reported to represent disposed lipophilic material called lipofuscine.
  • These are different from the nanoconjugates, which appear as red "haze,” and are only excitable in the red light range.
  • the clouds are translated in clouds of shades of white and grey.
  • FIG. 14 is a schematic drawing of the brain showing main blood vessels including the Superior Sagittal Sinus (SSS), a large blood vessel that runs along the midline of the brain.
  • SSS Superior Sagittal Sinus
  • FIGS. 2 IB - 21C provides information about the transfer of the drug from the vasculature into the brain parenchyma and its disappearance after 2-4 hours. This is a qualitative observation (FIG.
  • Example 7 Characterization of nanoconjugate fluorescence in brain parenchyma
  • nanoconjugate fluorescence may contribute to the lipofuscin signal ⁇ i.e., via degradation and accumulation of rhodamine in intracellular organelles), but this type of fluorescence was excluded from the spectral analysis.
  • a distinction between diffuse nanoconjugate fluorescence and lipofuscin has not been reported, even though several studies have shown lipofuscin-like particulate staining patterns.
  • Table 1 lists 12 nanoconjugates that were examined for their ability to penetrate the BBB and distribute in the brain parenchyma. The results indicate that P/LLL/AP2 has the best BBB penetration ability.
  • FIGS. 15A - 15C illustrate concentration dependent BBB penetration of P/LLL/AP-2/rhodamine.
  • FIG. 15A is a set of photographs illustrating optical imaging data acquired at 120 min after i.v. injection of P/LLL/AP-2/rhodamine at the following concentrations: photograph 1 - 29.5 ⁇ /kg; photograph 2 - 59 ⁇ /kg;. photograph 3 - 118 ⁇ /kg; and photograph 4 - 236 ⁇ /kg.
  • Drug concentrations are listed with regard to total nanoconjugate content systemically injected. Referring to this figure, the vasculature is shown in light grey, and the nanoconjugate as whitish diffused clouds.
  • FIG. 15A is a set of photographs illustrating optical imaging data acquired at 120 min after i.v. injection of P/LLL/AP-2/rhodamine at the following concentrations: photograph 1 - 29.5 ⁇ /kg; photograph 2 - 59 ⁇ /kg;. photograph 3 -
  • 15B is a chart illustrating nanoconjugate fluorescence intensity vs. "distance from vasculature" measurements in brain parenchyma of mice injected with three different concentrations.
  • fluorescence measurements were obtained from 10 ⁇ 2 -8 ⁇ regions of interest (ROI) that were randomly overlaid on regions devoid of vasculature shown as white squares on photograph 1 of FIG. 15A. Intensity measurements and positions were then obtained for each ROI and plotted against the location of the nearest blood vessel wall.
  • ROI regions of interest
  • chart 15C is set of charts: chart 1 - Cortex; chart 2 - Midbrain and chart 3 Hippocampus, illustrating average nanoconjugate fluorescence in the brain parenchyma measured following injections at four different drug concentrations.
  • FIG. 15 A presented are the optical imaging data of mice i.v. tail-injected with different concentrations of P/LLL/AP-2/rhodamine and sacrificed 120 minutes post-injection.
  • the drug concentration is listed as the total concentration of each injected nanoconjugate, where the conjugates contained 40% LLL, 2% peptide and 1% rhodamine, unless indicated otherwise.
  • the tissue shown in FIG. 15A was counter stained with tomato- lectin to show the vasculature (light grey), while the nanoconjugate is shown in grey.
  • FIG. 15A this is visible in Photograph 4, as strong nanoconjugate fluorescence (grey "haze”) near the blood vessels, but diminished fluorescence further away from the blood vessels.
  • FIG. 15B explores this relationship in a plot from all of the measurements (for each condition: 4 mice, 3-4 sections with 20 random measurements each). All fluorescence intensity measurements were conducted with 10 ⁇ 2 -8 ⁇ regions of interest placed outside of tomato-lectin stained blood vessels (ROI as in Photograph 1 of FIG. 15A); the positions of these ROIs were then measured against the location of the nearest blood vessel wall to produce the scatterplot in FIG. 15B.
  • ROI tomato-lectin stained blood vessels
  • the y-intercept for the 236 ⁇ /kg drug injection condition was 34.07 ⁇ 2.3; 17.49 ⁇ 0.8 for the 118 ⁇ /kg drug injection, and 6.342 ⁇ 0.34 for drug injected at 29.5 ⁇ /kg.
  • FIG. 15C the results described above are applicable to the cerebral cortex (Chart 1), the midbrain (Chart 2) and the hippocampus (Chart 3).
  • the data shown in on charts 1 - 3 of FIG. 15C are average nanoconjugate fluorescence intensity values and their standard errors: these were obtained from randomly sampled ROIs, irrespective of their location and distance from the vasculature (4 mice in each condition).
  • Chart 3 of FIG. 15C shows that fluorescence measurements in the hippocampus were consistently lower than those in the cortex or midbrain.
  • FIG. 13 shows that the background fluorescence in the hippocampus area was attributed to lipofuscin, which is preexisting autofluorescence and not dependent on injection of the buffer or peptide nanoconjugates. The background fluorescence has been subtracted from the fluorescence intensities illustrated on FIG. 15C.
  • FIGS. 16A - 16D illustrate blood vessel diameters, vascular coverage and inter-vessel distances in different brain regions.
  • FIG. 16A is a set of photographs illustrating blood vessels in the cortex, midbrain and hippocampal CAl cellular layer (outlined). The vessels were stained with tomato -lectin (shown here as white stretches) and nuclei were counterstained with DAPI (grey dots).
  • FIG. 16B are bar graphs illustrating vessel diameters. Referring to FIG. 16B, the vessel diameters were measured as the shortest distance between the vessel walls and were on average 4-5 ⁇ in every brain region. Blood vessels of this diameter were within the range of the cerebral microvasculature.
  • FIG. 16C is a bar graph illustrating vascular coverage. Referring to FIG.
  • FIGS. 16B - 16C similar-sized blood vessels were observed in the cortex, midbrain and hippocampus (FIG. 16B), but the area covered by these blood vessels is less in the hippocampus than the cortex or midbrain (FIG. 16C).
  • FIG. 16D these results in an inter-vessel distance in the hippocampus of 59 ⁇ , which is almost twice that of the cortex (32 ⁇ ) and midbrain (30 ⁇ ).
  • P/LLL/AP- 2/rhodamine distributes preferentially within ⁇ 30 ⁇ from the microvasculature (i.e., FIG.
  • Example 9 - BBB penetration depends on nanoconjugate composition
  • FIGS. 17A - 17B illustrate that the nanoconjugate composition determines degree and locus of BBB penetration.
  • FIG. 17A is set of photographs illustrating nanoconjugate permeation of the cerebral cortex: photograph 1-P/LLL/AP2; photograph 2 - P/AP-2 and photograph 3 -P/LLL.
  • optical imaging data showing nanoconjugate permeation of the cerebral cortex nanconjugate fluorescence is grey "haze” and the vasculature is indicated by white stretches. The most intense "haze” fluorescence was observed for P/LLL/ AP-2 as shown on photograph 1.
  • FIG. 17A is set of photographs illustrating nanoconjugate permeation of the cerebral cortex: photograph 1-P/LLL/AP2; photograph 2 - P/AP-2 and photograph 3 -P/LLL.
  • FIG. 17A data shown on photograph 1 vs. photograph 2 show that P/LLL/AP-2 penetrated the brain parenchyma better than P/AP2. This is especially apparent in the perivascular space where much of the diffuse grey nanoconjugate fluorescence "haze" can be seen in the P/LLL/AP-2 but not the P/AP-2 condition.
  • Corresponding fluorescence measurements from the cortex are summarized in FIG. 17B, chart 1, (black vs. grey data) and were significantly larger for P/LLL/AP-2 vs. P/AP-2 injected at 29.5 ⁇ /kg (Tukey: p ⁇ 0.0001), 59 ⁇ /kg (Tukey: p ⁇ 0.0001), and 118 ⁇ /kg (Tukey: p ⁇
  • P/AP-2 at 29.5 ⁇ /kg (Tukey: p ⁇ 0.01), 59 ⁇ /kg (Tukey: p ⁇ 0.0001), and 118 ⁇ /kg (Tukey: p ⁇ 0.0001). This observation was also made in the midbrain (FIG. 17B, chart 2), and in the hippocampus (FIG. 17B, chart 3). Thus, P/LLL penetrates the BBB even without a peptide moiety. The addition of the AP-2 peptide significantly increases BBB penetration, and in combination with LLL, produces the optimal nanoconjugate formula, P/LLL/AP2.
  • FIGS. 18A - 18B illustrate the effect of conjugated LLL residues on nanoconjugate conformation.
  • FIG. 18A is a schematic drawing of a chemical structure of the representative conjugate containing LLL and part of the conjugated peptide linker (PEG). LLL is indicated with black arrows in the structural scheme.
  • FIG. 18B is a three-dimensional image of the short representative PMLA structure illustrated in FIG. 18A (16 malic acid residues) with PEG (2 chains of ethylene glycol-hexamer conjugated via maleimide to PMLA), capped sulfhydryl (two moieties) and LLL (4 moieties).
  • FIG. 18B The structure shown on FIG. 18B is the result of total energy minimization calculated in vacuum indicated 226 kcal/mol for the analogue with LLL (Chem3D Pro 11.0).
  • FIGS. 19A - 19B illustrate nanoconjugate conformation in the absence of LLL.
  • FIG. 19A illustrate the structural model and is similar as the one shown in FIG. 18A. Because the structure is lacking LLL, the 3- dimensional conformation of the conjugate appears extended in comparison with the one in FIG. 18B.
  • FIG. 19B is a three-dimensional image of the structure shown in FIG. 19A obtained after energy minimum calculation. The total energy is 1194 kcal/mol according to energy minimization calculated for vacuum (Chem3D Pro 11.0).
  • LLL sterically prevents this interaction so that the AP-2 peptide becomes biologically active by interacting with LRP-1 (or other receptor molecules).
  • LDS dynamic light scattering
  • PDI polydispersity index measurements
  • Table 2 Energy calculations as shown on FIGS. 18A - 18B and 19A- 19B indicate that LLL can induce folding of nanoconjugates via LLL-LLL interactions, which ultimately decreases conformations of the free polymer and hence reduces numbers and diameters of conformational variants.
  • conjugation with LLL is favorable for BBB permeation by (i) optimizing the interactions of targeting peptides with receptors of a particular transcytosis pathway, (ii) reducing the diameter of the permeating nanoconjugate, and (iii) increasing the rigidity of the nanoconjugate.
  • BBB-penetrating peptides namely AP-2, M4, B6, and cTfRL were conjugated to P/LLL and screened for their ability to permit or enhance BBB -penetration of the nanoconjugate (Demeule et al. (2008); Staquicini et al. (2011); Yin et al. (2015); Liu et al. (2013); and Oller-Salvia et al. (2016), all of which are incorporated by reference as if fully set forth).
  • FIGS. 20A - 20E illustrate nanoconjugate peptide moiety screen.
  • FIG. 20A is a set of photographs illustrating P/LLL equipped with different peptides (1- P/LLL/AP-2; 2- P/LLL/M4; and 3 - P/LLL/B6) to assess their role in BBB penetration.
  • optical imaging data of the rhodamine labeled peptide conjugates show permeation of the cerebral cortex by P/LLL conjugated to AP-2 (1), M4 (2) and B6 (3). Nanoconjugate fluorescence is grey and the vasculature is white.
  • FIG. 20B - 20D is a set of bar graphs showing average nanoconjugate fluorescence in the cerebral cortex (FIG. 20B), midbrain (FIG. 20C) and hippocampus (FIG. 20D) injected at concentrations of 29.5 ⁇ /kg or 118 ⁇ /kg.
  • FIG. 20E illustrates nanoconjugate fluorescence measurements in the cerebral cortex for peptide combinations P/LLL/AP-7 (light grey bar), P/LLL/AP-2 (4%) (white bar), P/LLL/AP-2/M4 (dark grey bar) and P/LLL/AP-2 (2%) (black bar) injected at concentrations of 59 ⁇ /kg or 118 ⁇ /kg.
  • the nanoconjugate with high BBB penetration had the formula P/LLL/AP-2/rhodamine.
  • the conjugate P/LLL/M4 injected at 118 ⁇ /kg produced significantly less fluorescence in the cortex than P/LLL/AP-2 (FIG. 20B; Sidak: p ⁇ 0.0001).
  • P/LLL/M4 and P/LLL/AP-2 fluorescence were measured in both, the midbrain and the hippocampus, regardless of the injected drug concentrations (black vs. dark grey in FIGS. 20C and 20D).
  • P/LLL/M4 and P/LLL/AP-2 appear to permeate the brain tissue with similar efficacies, but P/LLL/M4 shows regional selectivity and poor permeation of the cerebral cortex.
  • P/LLL/AP-2/M4 failed to display a significant sum of effects by each peptide. Moreover, P/LLL/AP-2/M4 has a reduced cargo capacity due to higher occupancy of the polymer platform and thus a reduced number of free ligand attachment sites.
  • AP- 7 differs from AP-2 by the replacement of two lysine residues in positions 10 and 15 with arginine residues (TFFYGGSRGRRNNFRTEEYCNH 2 (SEQ ID NO: 7)), which reportedly impairs peptide interactions with endothelial LRP-1 receptors (Demeule et al. (2008), which is incorporated by reference herein as if fully set forth).
  • the results apply to the brain of healthy mice. It is instructive to consider that the performance of certain peptides may differ in pathological conditions in which the BBB is impaired, or trans-BBB receptor expression is altered.
  • the TfR route may be effective for drug delivery into brain tumors. Gliomas overexpress TfR in their vascular endothelium, and this may aid drug-tumor penetration and delivery via enhanced TfR transport (Meng et al. (2017), which is incorporated herein by reference as if fully set forth).
  • the LRP-1 route is hnked to less active amyloid 6 protein clearance and effects homeostasis in Alzheimer's disease (Grimmer et al. (2014), which is incorporated herein by reference as if fully set forth).
  • Example 11 Nanoconjugate pharmacokinetics in blood and brain
  • FIGS. 21A - 2 ID illustrate pharmacokinetics of nanoconjugate fluorescence in serum and brain tissue.
  • FIG. 21A is a chart illustrating serum clearance analysis that was conducted for P/LLL/AP-2 (black) and P/LLL (grey), and optically via imaging of the cerebral vasculature content (black triangle).
  • FIG. 2 IB is a set of photographs illustrating optical imaging data showing drug clearance vascular and parenchyma accumulation over 240 minutes. These images show the nanoconjugate P/LLL/AP-2 in whitish "haze” and the vasculature in grey.
  • FIG. 21C illustrates vascular fluorescence intensity profile for the saggital sinus as indicated with a white line in FIG. 2 IB. Timepoints are indicated in the top right corner of this plot.
  • FIG. 2 ID is a bar graph illustrating time dependence of nanoconjugate fluorescence intensity in brain tissue for rhodamineP/LLL/AP2 (black), P/LLL (grey) and P/AP2 (white) is different from the serum PK kinetics. Fluorescence has a rapid onset and remains quasi-stable for 120 minutes. Clearance occurs at 240-480 minutes. All data shown are from the cerebral cortex and are relative fluorescence values that were subtracted from background image intensities of representative tissues of PBS injected mice.
  • the nanoconjugate serum concentrations were calculated from calibration curves that was previously derived from fluorescence measurements of nanoconjugates with known concentrations.
  • the conjugates P/LLL/AP-2 and P/LLL had serum half-lives of 76.7 min and 119 min, respectively.
  • FIG. 2 IB shows imaging data from a large central blood vessel, the sagittal sinus, from 30 to 240 minutes after i.v. injection. The images show the mini nanodrug (whitish "haze") and the sinus vasculature (grey).
  • FIG. 21C shows the fluorescence intensity profile for this blood vessel and adjacent brain tissues, as indicated with a white line in FIG. 2 IB.
  • the fluorescent nanoconjugate is clearly concentrated in the vasculature at 30 minutes post i.v. injection, while subsequent timepoints show a progressive loss of vascular fluorescence (FIG. 21C).
  • the "optical vascular fluorescence” was calculated by measuring the difference between fluorescence peaks and the fluorescence intensity in the surrounding parenchyma (see dashed lines in FIG. 21C) and then plotted the vascular fluorescence over multiple timepoints alongside the actual serum measurements in FIG. 21A (black triangle).
  • optical vascular fluorescence measurements were converted to ⁇ /mL units via normalization to one time point of serum P/LLL/AP-2 (120 min); the remaining timepoints were then converted using the same ratio (0.115 ⁇ /mL serum concentration for the 29.5 ⁇ /kg injection at 120 minutes).
  • optical half-life 73.2 min
  • the imaging results were to estimate the actual concentration of P/LLL/AP-2 conjugates in cortical brain parenchyma at 120 minutes after the drug injection. This was accomplished by first measuring P/LLL/AP-2/rhodamine fluorescence in the cerebral vasculature and then the surrounding parenchyma with identical regions of interest, followed by a calculation of the vessel / parenchyma fluorescence ratio
  • FIGS. 22A - 22C illustrate estimation of the nanoconjugate concentration in ⁇ g/mL of i.v. injected P/LLL/AP-2 in the parenchyma of the cerebral cortex.
  • FIG. 22A is set of photographs illustrating optical imaging data showing cortical tissue from mice injected with P/LLL/AP- 2/rhodamine at 29.5 ⁇ /kg (Al) and 118 ⁇ /kg (A2).
  • the top images show cell nuclei (grey), vasculature (light grey stretches) and P/LLL/AP-2 conjugate (grey).
  • the lower panels show only P/LLL/AP-2 conjugate-associated fluorescence.
  • FIG. 22C illustrates estimated P/LLL/AP-2 concentration in the cortical brain parenchyma.
  • FIG. 22A data was summarized for 4 mice, 4 sections with 10 measurements for each condition.
  • the images in FIG. 22A (Al and A2, bottom panel) demonstrate this procedure in two samples from mice injected with 29.5 ⁇ /kg and 118 ⁇ /kg of P/LLL/AP-2 conjugate, respectively.
  • the fluorescence ratios that resulted from the measurements are summarized in FIG. 22B.
  • the lowest P/LLL/AP-2 parenchyma concentration is estimated at 0.049 ⁇ 0.001 ⁇ /ml for the 29.5 ⁇ /kg injection; the highest parenchyma concentration is 0.32 ⁇ 0.01 ⁇ /ml for the 236 ⁇ /kg injection. Based on these estimates, the conclusion was made that P/LLL/AP-2 traverses the BBB efficiently and that 40% or higher percentage of free serum drug in the vascular tissue can be detected in the brain within 120 minutes after i.v. administration (% depending on the distance from the vascular tissue).
  • Example 13 - Mini nanodrugs targeting amyloid plaques [00293] The peptide nanodrugs targeting the carrier to a brain-intern cell or structure were designed. Towards this goal, the nanoconjugates including the D-enantiomeric peptides targeting amyloid and amyloid plaques were used. The efficacy of amyloid targeting peptides Dl, D3, ACI-89 was evaluated.
  • FIGS. 23A - 23C illustrate peptide-dependent labeling of plagues.
  • FIG. 23A is a photograph illustrating optical imaging data following mice injected with P/LLL/M4.
  • FIG. 23B is a photograph illustrating optical imaging data following mice injected with P/LLL/M4/Dl/rhodamine.
  • plaques staining with the conjugates was observed as "whitish grey cloud" in the center of the photographs. Staining by P/LLL/M4/rhodamine (FIG. 23A) was observed to be less intensive than by P/LLL/M4/D1- peptide/rhodamine as was revealed by optical measurement.
  • nanodrugs carrying the peptides were iv injected into the mouse tail at doses of 236 ⁇ /kg of P/LLL/M4/rhodamine orP/LLL/M4/Dl.
  • FIG. 23C is a bar graph showing A6 plaque vs.
  • Plaques have a unique structural appearance like a hairy star of the size of approximately 3 microns or more.
  • the reagents can be also applied applied in vitro to mounted slides after fixation, incubated for 20-30 minutes in the plaque reagent and then washed exhaustively.
  • FIGS. 23A - 23B this is shown by the figure showing that plaques are more intensively stained after iv injection of P/LLL/M4/D1- peptide/rhodamine mini nondrug (FIG. 23B; referred to in the figure as P/LLL/M4/Dl-peptide) in comparison with staining after iv injection with P/LLL/M4/rhodamine (FIG. 23A; referred to in the figure as P/LLL/M4).
  • the result shows that the nanodrugs, such as P/LLL/M4/rhodamine, can be used for further targeting inside brain by carrying additional peptides, such as Dl.
  • the bar-panels of FIG. 23C show quantitatively the effect of increased staining plaques in the presence of conjugated Dl compared to staining in the absence of Dl.
  • Example 14 Advantages of the Mini Nanodrugs for Trans- BBB Delivery
  • a biodegradable non-toxic 6-poly(L-malic acid) (PMLA or P) was synthesized as a scaffold to chemically bind the BBB crossing peptides Angiopep-2 (AP2), Miniap-4 (M4), and the transferrin receptor directed ligands cTfRL and B6.
  • AP2 Angiopep-2
  • M4 Miniap-4
  • cTfRL transferrin receptor directed ligands
  • a tri-leucine endosome escape unit LLL
  • rhodamine fluorescent marker
  • the mini nanodrug containing P/LLL/AP-2 produced significant fluorescence in the parenchyma of the cortex, midbrain and hippocampus 30 minutes after a single intravenous injection; clearance was observed after four hours.
  • the mini nanodrug variant P/LLL lacking AP-2, or the variant P/AP-2 lacking LLL, showed significantly less BBB penetration.
  • the LLL moiety appeared to stabilize the nanoconjugate, while AP-2 enhanced BBB penetration.
  • the mini nanodrug containing the peptide cTfRL displayed comparably little and / or inconsistent infiltration of brain parenchyma, likely due to reduced trans-BBB transport.
  • P/LLL/AP-2 or the other peptides can now be functionalized with intra-brain targeting and drug treatment moieties that are aimed at molecular pathways imp heated in neurological disorders.
  • a nanodrug platform for trans-BBB drug delivery was presented.
  • the strategy builds on previously published peptides to shuttle a PMLA- based drug platform across the BBB.
  • PMLA/LLL/peptide interactions were observed to determine the BBB passage, and detailed investigation was performed to determine how the mini nanodrug was distributed in the brain.
  • moieties of inherent hydrophobic structure, such as LLL influence and enhance brain delivery, especially in areas with high blood vessel density such as the midbrain. This effect may be due to inherent drug properties.
  • the BBB for the nanodrug s (P/LLL/AP-2, P/LLL/M4 or P/LLL/B6-conjugates) and under applied conditions, may not constitute an efficient barrier and that it can be open to dehver high amounts of covalently bound drug for pharmaceutical treatment.
  • Sequences and conformation of targeting and functioning peptides provide high resistance to in vivo degradation (exocyclic or D -conformation). Values of dissociation constants at micro molar or below. Except for tau, nucleic acids sequences of genes/amino acid sequences for targeting malignant disease marker proteins 6-secretase 1 (BACE1), presenilin 1, are available for targeting and the design of antisense oligo nucleotides.
  • BACE1 malignant disease marker proteins 6-secretase 1
  • presenilin 1 presenilin
  • a mini nanodrug provides sufficient activity against homeostasis unbalancing body constituents during treatment of the recipients.
  • Mini nanodrugs do not oversupply the recipient organism with drugs and delivering vehicles and the components they are built from.
  • a mini drug eludes principles of carrying a close to minimal supply at maximum effective drug doses in the best efficious physical make up for deep tissue penetration.
  • the mini nanodrug is a receptor targeting construct of minimum surface, elongated form and moderately strong binding affinities in order to maximise receptor releasing kinetics and fast biobarrier penetration, minimum antigenetic content to minimise immune reaction and biodegradability to avoid long lasting in vivo depositions.
  • Fiji an open-source platform for biological-image analysis. Nature methods 2012, 9, 676-82.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Molecular Biology (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Epidemiology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Zoology (AREA)
  • Insects & Arthropods (AREA)
  • Biomedical Technology (AREA)
  • Immunology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Toxicology (AREA)
  • Neurology (AREA)
  • Neurosurgery (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Peptides Or Proteins (AREA)
  • Medicinal Preparation (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

L'invention concerne des mini-nanomédicaments qui contiennent un échafaudage moléculaire à base d'acide polymalique comprenant un ou plusieurs peptides capables de traverser la barrière hémato-encéphalique, un ou plusieurs peptides se liant aux plaques et un ou plusieurs agents thérapeutiques fixés audit échafaudage. L'invention concerne également des méthodes de traitement de maladies cérébrales ou autres états pathologiques anormaux, et l'imagerie de ceux-ci chez un sujet par administration des mini-nanomédicaments. Des méthodes destinées à réduire la formation de plaques amyloïdes dans le cerveau d'un sujet sont en outre décrites.
PCT/US2018/053873 2017-10-02 2018-10-02 Méthodes et compositions pour une administration efficace à travers de multiples barrières biologiques WO2019070645A1 (fr)

Priority Applications (4)

Application Number Priority Date Filing Date Title
EP18864985.9A EP3691670A4 (fr) 2017-10-02 2018-10-02 Méthodes et compositions pour une administration efficace à travers de multiples barrières biologiques
CN201880065055.7A CN111182913A (zh) 2017-10-02 2018-10-02 用于通过多个生物屏障进行有效递送的方法和组合物
RU2020114744A RU2020114744A (ru) 2017-10-02 2018-10-02 Способы и композиции для эффективной доставки через множество биобарьеров
US16/815,760 US20200206304A1 (en) 2017-10-02 2020-03-11 Methods and compositions for efficient delivery through multiple bio barriers

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201762566813P 2017-10-02 2017-10-02
US62/566,813 2017-10-02

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US16/815,760 Continuation-In-Part US20200206304A1 (en) 2017-10-02 2020-03-11 Methods and compositions for efficient delivery through multiple bio barriers

Publications (1)

Publication Number Publication Date
WO2019070645A1 true WO2019070645A1 (fr) 2019-04-11

Family

ID=65995287

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2018/053873 WO2019070645A1 (fr) 2017-10-02 2018-10-02 Méthodes et compositions pour une administration efficace à travers de multiples barrières biologiques

Country Status (4)

Country Link
EP (1) EP3691670A4 (fr)
CN (1) CN111182913A (fr)
RU (1) RU2020114744A (fr)
WO (1) WO2019070645A1 (fr)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN115025249A (zh) * 2022-05-12 2022-09-09 深圳市第二人民医院(深圳市转化医学研究院) 靶向探针及其制备方法和应用
WO2023094810A1 (fr) * 2021-11-24 2023-06-01 Ucl Business Ltd Polymersomes pour l'élimination de protéines amyloïdes bêta et/ou tau

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022156531A1 (fr) * 2021-01-19 2022-07-28 中国人民解放军军事科学院军事医学研究院 Peptide de liaison à la dynéine capable de traverser une barrière biologique, et son utilisation

Citations (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100297120A1 (en) * 2007-05-29 2010-11-25 Angiochem Inc. Aprotinin-like polypeptides for delivering agents conjugated thereto to tissues
US20110105404A1 (en) * 2008-03-13 2011-05-05 Tianjin Medical University Beta Sheet Inhibiting Peptides For Preventing And/Or Treating Alzheimer`s Disease
US20110281769A1 (en) * 2002-11-14 2011-11-17 Dharmacon, Inc. siRNA targeting beta secretase (BACE)
US20140017766A1 (en) * 2004-06-03 2014-01-16 The Regents Of The University Of California Targeting pseudotyped retroviral vectors
US20140147440A1 (en) * 2011-06-24 2014-05-29 Universite De Geneve Uses of nanog inhibitors and related methods
US20140193398A1 (en) * 2010-12-30 2014-07-10 Cedars-Sinai Medical Center Polymalic acid-based nanobiopolymer compositions
WO2015001015A1 (fr) * 2013-07-04 2015-01-08 Universitat De Barcelona Peptides transportés activement et résistant aux protéases en tant que navettes bbb et produits de construction navette-cargaison
US20150307552A1 (en) * 2012-09-14 2015-10-29 Forschungszentrum Jülich GmbH Novel d-enantiomeric peptides derived from d3 and use thereof
US20160175450A1 (en) * 2009-12-10 2016-06-23 Cedars-Sinai Medical Center Drug delivery of temozolomide for systemic based treatment of cancer
WO2017152054A1 (fr) * 2016-03-04 2017-09-08 Cedars-Sinai Medical Center Nanoimmunoconjugués à base d'acide polymalique et leurs utilisations

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10383958B2 (en) * 2011-04-06 2019-08-20 Cedars-Sinai Medical Center Polymalic acid based nanoconjugates for imaging
CN102716495B (zh) * 2012-06-20 2014-04-23 中国人民解放军第四军医大学 靶向肿瘤多功能聚苹果酸载体药物

Patent Citations (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20110281769A1 (en) * 2002-11-14 2011-11-17 Dharmacon, Inc. siRNA targeting beta secretase (BACE)
US20140017766A1 (en) * 2004-06-03 2014-01-16 The Regents Of The University Of California Targeting pseudotyped retroviral vectors
US20100297120A1 (en) * 2007-05-29 2010-11-25 Angiochem Inc. Aprotinin-like polypeptides for delivering agents conjugated thereto to tissues
US20110105404A1 (en) * 2008-03-13 2011-05-05 Tianjin Medical University Beta Sheet Inhibiting Peptides For Preventing And/Or Treating Alzheimer`s Disease
US20160175450A1 (en) * 2009-12-10 2016-06-23 Cedars-Sinai Medical Center Drug delivery of temozolomide for systemic based treatment of cancer
US20140193398A1 (en) * 2010-12-30 2014-07-10 Cedars-Sinai Medical Center Polymalic acid-based nanobiopolymer compositions
US20140147440A1 (en) * 2011-06-24 2014-05-29 Universite De Geneve Uses of nanog inhibitors and related methods
US20150307552A1 (en) * 2012-09-14 2015-10-29 Forschungszentrum Jülich GmbH Novel d-enantiomeric peptides derived from d3 and use thereof
WO2015001015A1 (fr) * 2013-07-04 2015-01-08 Universitat De Barcelona Peptides transportés activement et résistant aux protéases en tant que navettes bbb et produits de construction navette-cargaison
WO2017152054A1 (fr) * 2016-03-04 2017-09-08 Cedars-Sinai Medical Center Nanoimmunoconjugués à base d'acide polymalique et leurs utilisations

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
FUNKE ET AL.: "Development of a small D-enantiomeric Alzheimer's amyloid-P binding peptide ligand for future in vivo imaging applications", PLOS ONE, vol. 7, 24 July 2012 (2012-07-24), pages 1 - 10, XP055056284, DOI: 10.1371/journal.pone.0041457 *
See also references of EP3691670A4 *

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023094810A1 (fr) * 2021-11-24 2023-06-01 Ucl Business Ltd Polymersomes pour l'élimination de protéines amyloïdes bêta et/ou tau
CN115025249A (zh) * 2022-05-12 2022-09-09 深圳市第二人民医院(深圳市转化医学研究院) 靶向探针及其制备方法和应用
CN115025249B (zh) * 2022-05-12 2024-02-20 深圳市第二人民医院(深圳市转化医学研究院) 靶向探针及其制备方法和应用

Also Published As

Publication number Publication date
CN111182913A (zh) 2020-05-19
EP3691670A4 (fr) 2021-08-04
EP3691670A1 (fr) 2020-08-12
RU2020114744A (ru) 2021-11-09

Similar Documents

Publication Publication Date Title
Cai et al. A nanostrategy for efficient imaging‐guided antitumor therapy through a stimuli‐responsive branched polymeric prodrug
US20200206304A1 (en) Methods and compositions for efficient delivery through multiple bio barriers
Saad et al. Receptor targeted polymers, dendrimers, liposomes: which nanocarrier is the most efficient for tumor-specific treatment and imaging?
Wang et al. Nanoscale covalent organic polymers as a biodegradable nanomedicine for chemotherapy-enhanced photodynamic therapy of cancer
Li et al. Near infrared fluorescent imaging of brain tumor with IR780 dye incorporated phospholipid nanoparticles
Accardo et al. Receptor binding peptides for target-selective delivery of nanoparticles encapsulated drugs
Xu et al. Smart and hyper-fast responsive polyprodrug nanoplatform for targeted cancer therapy
Na et al. Real-time and non-invasive optical imaging of tumor-targeting glycol chitosan nanoparticles in various tumor models
Jenkins et al. Mini-review: fluorescence imaging in cancer cells using dye-doped nanoparticles
US9149544B2 (en) Bioconjugation of calcium phosphosilicate nanoparticles for selective targeting of cells in vivo
Li et al. Cathepsin B-responsive nanodrug delivery systems for precise diagnosis and targeted therapy of malignant tumors
US10442890B2 (en) Multifunctional degradable nanoparticles with control over size and functionalities
Zhang et al. Stepwise dual targeting and dual responsive polymer micelles for mitochondrion therapy
US20150258217A1 (en) Methods of Synthesizing and Using Peg-Like Fluorochromes
WO2019070645A1 (fr) Méthodes et compositions pour une administration efficace à travers de multiples barrières biologiques
Praça et al. A nanoformulation for the preferential accumulation in adult neurogenic niches
Patil et al. Polymalic acid chlorotoxin nanoconjugate for near-infrared fluorescence guided resection of glioblastoma multiforme
Cao et al. A triple modality BSA-coated dendritic nanoplatform for NIR imaging, enhanced tumor penetration and anticancer therapy
EP3610243A1 (fr) Procédés d'imagerie par fluorescence ratiométrique
Moorthy et al. Dendrimer architectonics to treat cancer and neurodegenerative diseases with implications in theranostics and personalized medicine
US20110274620A1 (en) Multifunctional degradable nanoparticles with control over size and functionalities
CA2713813A1 (fr) Vehicules de medicaments pour chimiotherapie intralymphatique
Lee et al. Supramolecular assembly based on host–guest interaction between beta-cyclodextrin and adamantane for specifically targeted cancer imaging
Hyun et al. Ischemic brain imaging using fluorescent gold nanoprobes sensitive to reactive oxygen species
Guo et al. Blood-brain-barrier penetrable thiolated paclitaxel-oligo (p-phenylene vinylene) nanomedicine with increased drug efficiency for glioblastoma treatment

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: 18864985

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2018864985

Country of ref document: EP

Effective date: 20200504