WO2006061101A2 - Drug delivery peptides for crossing blood-brain barrier - Google Patents

Drug delivery peptides for crossing blood-brain barrier Download PDF

Info

Publication number
WO2006061101A2
WO2006061101A2 PCT/EP2005/012554 EP2005012554W WO2006061101A2 WO 2006061101 A2 WO2006061101 A2 WO 2006061101A2 EP 2005012554 W EP2005012554 W EP 2005012554W WO 2006061101 A2 WO2006061101 A2 WO 2006061101A2
Authority
WO
WIPO (PCT)
Prior art keywords
peptide
phe
gly
nps
peptides
Prior art date
Application number
PCT/EP2005/012554
Other languages
French (fr)
Other versions
WO2006061101A3 (en
Inventor
Flavio Forni
Maria Angela Vandelli
Luca Constantino
Original Assignee
Universita' Degli Studi Di Modena E Reggio Emilia
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 Universita' Degli Studi Di Modena E Reggio Emilia filed Critical Universita' Degli Studi Di Modena E Reggio Emilia
Priority to EP05817909A priority Critical patent/EP1819723A2/en
Publication of WO2006061101A2 publication Critical patent/WO2006061101A2/en
Publication of WO2006061101A3 publication Critical patent/WO2006061101A3/en

Links

Classifications

    • 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
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B82NANOTECHNOLOGY
    • B82YSPECIFIC USES OR APPLICATIONS OF NANOSTRUCTURES; MEASUREMENT OR ANALYSIS OF NANOSTRUCTURES; MANUFACTURE OR TREATMENT OF NANOSTRUCTURES
    • B82Y30/00Nanotechnology for materials or surface science, e.g. nanocomposites
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K9/00Peptides having up to 20 amino acids, containing saccharide radicals and having a fully defined sequence; Derivatives thereof
    • C07K9/001Peptides having up to 20 amino acids, containing saccharide radicals and having a fully defined sequence; Derivatives thereof the peptide sequence having less than 12 amino acids and not being part of a ring structure

Definitions

  • This invention relates to new peptides which are useful for carrying drugs through the blood-brain barrier.
  • BBB blood-brain barrier
  • NPs nanoparticles
  • solid lipid nanoparticles into which the drug can be adsorbed or included (1,2).
  • NPs have the advantage that a large number of molecules of the drug can be carried by each NP, and that they mask the characteristics of the drug to enable it to cross the BBB and protect it against enzymatic degradation.
  • Different strategies have been proposed to enable NPs to cross the BBB, most of which are based on modifications to their hydrophobic surface.
  • NPs mainly consist of polycyanoacrylate (such as poly(butylcyanoacrylate, PBCA), and a hydrophilic polymer such as polysorbate 80 can be adsorbed onto their surface (2); another strategy involves the preparation of NPs from an amphiphilic copolymer wherein the hydrophobic component is able to form the solid phase (the body of the NP), while the hydrophilic component remains on its surface, facing outwards.
  • This copolymer can be represented by polyethylene glycol (PEG) and n- hexadecylcyanoacrylate (PEG-PHDCA) (3).
  • NPs constituted by PHDCA PEGylate are able to cross the BBB without modifying its permeability (3); PEGylation increases the plasma half-life of NPs, and it has been suggested that this may be the reason why they are able to penetrate into the brain to a greater extent than other formulations (3). If the NPs are not coated with surfactant, they remain inside the blood vessels (2).
  • Various mechanisms have been postulated to explain how NPs cross the BBB (1,4).
  • it is important for NPs to be able to cross the BBB it is also extremely important for them to have the brain as their priority target, so that they can be used with drugs aimed at the CNS, not at other organs or apparatus.
  • Receptor-mediated endocytosis is responsible for the fact that the BBB is crossed not only by molecules with a low molecular weight such as thiamine, but also by aggregates with a high molecular weight such as low-density lipoproteins (LDL), and by other macromolecules.
  • Specific receptors for insulin, insulin-like growth factors (IGF-I, IGF-2), angiotensin II, atrial natriuretic peptide (ANP), brain natriuretic peptide (BNP), interleukin-1 (IL-I) and transferrin have been identified in the brain capillaries. The transcytosis of these molecules through the BBB in vivo has so far only been demonstrated for insulin and transferrin (1).
  • NPs consisting of wax and a surfactant (Brji 78®) conjugated with thiamine (5)
  • nanogels consisting of flexible hydrophilic polymers of nanometric dimensions (nanogels) composed of polyethyleneimine crosslinked to poly(ethyleneglycol), which in turn are derivatised with transferrin or insulin, have been created specifically to reach the CNS (6,7).
  • Peptides are polar compounds, but various proteins are able to cross the lipid cell membranes; there is great interest in studying the possibility of increasing the cell permeability of liposomes, proteins and NPs by using membrane-permeable peptides and proteins as carriers (8). Although the primary structure of the peptides able to cross the cell membranes is highly variable (9), some of their general characteristics are known. It has been postulated that a certain abundance of positive charges due to the presence of
  • Arg (10-12) and the presence of aminoacids with large, hydrophobic side chains (13) inserted in the peptide sequence, play a crucial role in effective cell uptake.
  • opioid peptides have proved able to cross the BBB, and this permeability can be increased by the presence of sugar residues (glucose, lactose, etc.); however, the mechanism whereby these substances cross the BBB is not known.
  • an opioid peptide able to cross the BBE known as MMP-2200, with the formula H 2 N-Tyr-D- Thr-Gly-Phe-Leu-Ser-O- ⁇ -D-lactose-CONH 2 , has been described (14).
  • H 2 N-Gly-Phe-D-Thr-Gly-Phe-Leu-Ser-CONH 2 can be opportunely used to carry through the blood-brain barrier drugs conjugated to said sequence either directly or via a linear or branched polyvalent spacer, wherein other aminoacids can replace the first two aminoacids of the N-terminal portion, the order of which can be reversed, and their number may be different from two.
  • a first aspect of the invention therefore relates to a peptide having the sequence reported above, wherein the serine residue may be functionalised with sugar residues, with a C- or O-glycoside bond.
  • the sugar residue is preferably a residue of glucose, galactose, lactose, mannose or xylose, and more preferably an O- ⁇ -D-glucoside residue.
  • the invention also relates to a conjugate of said peptides with a pharmaceutically acceptable polyester or polyamide polymer, nanoparticle systems comprising said conjugates, and pharmaceutical compositions comprising said nanoparticle systems.
  • Fig. 3 ESCA spectrum of NPs prepared with PLGA-HN-Gly-Phe-D- Thr-Gly-Phe-Leu-Ser-CONH 2 .
  • Fig. 4 ESCA spectrum of NPs prepared with PLGA-HN-Gly-Phe-D- Thr-Gly-Phe-Leu-SerO- ⁇ -D-glucose-CONH 2 .
  • polymers which can be advantageously used to prepare the conjugates according to the invention include polymers or copolymers of biodegradable aliphatic hy ⁇ "roxyacids, preferably lactic acid and/or glycolic acid.
  • the copolymer poly(D 5 L-lactide-c ⁇ -glycolide) (PLGA) or poly(D,L- lactic) acid is particularly preferred.
  • the copolymer PLGA is a biodegradable polyester, approved by the American FDA, which breaks down without inducing inflammation or immune reactions, and is therefore particularly suitable for the purposes of this invention.
  • the peptides according to the invention can be conjugated to PLGA or other polymers containing reactive groups (typically carboxyl groups) with the amino-terminal groups of the peptide according to methods already known in themselves, such as by activation of the carboxyl group with N-hydroxysuccinimide in the presence of dicyclohexylcarbodiimide, and subsequent formation of an amide bond with the N-terminal (unprotected) portion of the peptide.
  • the peptide can be conjugated through a suitable all-purpose spacer, such as the aminoacid "Behera's amine".
  • the molecular weight of the polymer used for the conjugation to the peptide is not critical, but typically falls into the interval between 1000 and 50,000 Da; the degree of functionalisation of the polymer with the peptide is between 30 and 80% of the theoretical value.
  • the conjugates thus obtainable can be used to prepare nanoparticle systems according to known techniques, for example as described in (18) and in T. Niwa et al., J. Pharm. Sci. (1994) 83, 5, 727-732; C. S. Cho et al., Biomaterials (1997) 18, 323-326; T. Govender et al., J. Control. ReI. (1999) 57, 171-185; and M. F. Zambaux et al., J. Control. ReI.
  • the nanoparticles (NP) according to the invention will contain a drug, a suitable surfactant, and a pharmaceutical carrier such as water or aqueous saline solutions.
  • micro- or nanoparticle systems could be considered as an alternative to NPs.
  • the drugs which can be advantageously used according to this invention are obviously those which are required to perform their pharmacological action in the brain.
  • these drugs include antibacterial, antiviral, psychotropic, antidepressant, anti-tumoral and antimigraine agents, analgesics, antagonists or agonists of receptors present in the central nervous system, antibodies, antisense oligonucleotides, hormones, narcotic analgesics, and diagnostic agents (paramagnetic complexes for MRI, radionuclides for nuclear medicine or diagnosis, etc.).
  • the drugs can be included, adsorbed or absorbed into the nanoparticles or conjugated directly with the peptide or copolymer by means of a covalent bond which is labile in vivo, such as an ester or amide bond.
  • the invention therefore also relates to pharmaceutical or diagnostic compositions comprising a peptide or one of its conjugates as defined above, together with any excipients.
  • the dose and administration route of the nanometric systems according to the invention will obviously depend on a number of factors, such as the toxicological and pharmacokinetic characteristics of the drug, the severity of the disorder to be treated, and the patient's condition, especially his/her age, weight and sex.
  • the dose may in any event be determined on the basis of the results of pre-clinical trials and phase I clinical trials, as is usual in drug development.
  • EXAMPLE 1 - Peptide synthesis The peptides were synthesised by solid-phase peptide synthesis (SPPS) using a Rink amide resin and the FMOC method. All the peptides were synthesised manually using a suitable reactor (scale of 0.5 g of resin, 5.00 ml washing volume) and agitation by anhydrous nitrogen flow.
  • SPPS solid-phase peptide synthesis
  • All the peptides were synthesised manually using a suitable reactor (scale of 0.5 g of resin, 5.00 ml washing volume) and agitation by anhydrous nitrogen flow.
  • Rink amide resin (4-[[(2',4'-dimethoxy-phenyl)(9-fluorenylmethoxycarbonyl)amino]methyl] phenoxyl resin) with a degree of functionalisation of 0.63 mmol/g was used.
  • the Ser used to prepare the peptide H 2 N-Gly-Phe-D-Thr-Gry-Phe-Leu-Ser- CONH 2 (Peptide 1) was protected as tert-butyl ether; the hydroxyl groups of FMOC-SerO- ⁇ -D-glucoside, used for synthesis of the peptide H 2 N-Gly-Phe- D-Thr-Gly-Phe-Leu-SerO- ⁇ -D-glucose-CONH 2 (peptide 2), were protected by formation of esters with acetic acid.
  • the FMOC was removed with the piperidine/DMF (9:1, 2 x 5 min), followed by washing with DMF (5 x 1 min).
  • the couplings were performed by adding a solution in DMF (4ml) consisting of FMOC aminoacid (3 equiv.), TBTU (3 equiv.), HOBt (3 equiv.) and *Pr 2 Net (6 equiv.) to the peptide present on the resin.
  • the resin suspension was agitated by bubbling anhydrous nitrogen for 3 h, after which the degree of completeness of coupling was determined with the Kaiser test.
  • the FMOC group is removed as reported above; the acetic groups present on the sugar residue are removed by treatment with H 2 N-NH 2 .H 2 O/MeOH 4/1 (2 x 5 mL x 30 min, followed by 1 x 5 mL x 1 h), while the peptide remains bonded to the resin. The excess H 2 N-NH 2 -H 2 O is then removed, and the resin is washed with MeOH (5 x 1 min) followed by CH 2 Cl 2 (5 x 1 min).
  • the peptide is released from the dried resin by treatment with CF 3 COOH (TFA) (84%), anisole (4%) thioanisole (4%), ethanedithiol (4%) and H 2 O (4%) (4 ml x 2 h); the solution is filtered to remove the depleted resin, and the resulting solution is cooled and precipitated by adding anhydrous ether. The solid thus obtained is filtered and washed several times with anhydrous ether; the crude peptide is then purified by dissolving in MeOH and precipitating with anhydrous ether. The peptides thus obtained are then analysed by mass spectroscopy (Q-TOF Global Ultima, Micromass, MALDI) (m/z): Peptide 1, est.
  • EXAMPLE 2 Conjugation of PLGA with peptides 1 and 2 Dicyclohexylcarbodiimide (19.0 mg, 93 ⁇ mol) and N-hydroxy- succinimide (11.0 mg, 93 ⁇ mol) is added to a solution of 1.00 g (88 ⁇ mol) PLGA RG503H (Boehringer Ingelheim) with a molecular weight of 11,000, determined by titration of the carboxyl groups at its ends (4.94 mg KOH/g of polymer) in anhydrous dioxane (5 ml); the solution is then agitated for 4 hours at a temperature ranging from the initial 4 0 C to 20 0 C.
  • the dicyclohexylurea is then filtered, and the solution poured into ethyl ether.
  • the insoluble polymer is separated by decanting and purified by dissolving in CH 2 Cl 2 and precipitating with anhydrous ether (twice).
  • the polymer thus obtained is then dissolved in anhydrous DMSO (5 ml), and maintained under agitation at ambient temperature after addition of a solution of the peptide (80 ⁇ mol) in DMSO (1 ml) and triethylamine (17 ⁇ l, 120 ⁇ mol). After 4 hours at ambient temperature, the reaction solution is poured into ethyl ether; the modified polymer is separated by decanting and purified by solubilisation in CH 2 Cl 2 and precipitation with methanol (three times).
  • the nanoparticles were prepared by the nanoprecipitation technique (18).
  • the solution thus obtained was poured slowly into deionised water (25 ml) containing Pluronic F68 (100 mg).
  • NPs were then purified by gel-filtration chromatography (Sepharose CL4B gel (160 ml), column 50 x 2 cm), using water as mobile phase, and freeze-dried without the presence of a cryoprotector. Analysis of NPs.
  • a scanning electron microscope (SEM) (XL-40 Philips, Eindhoven, Netherlands) was used to evaluate the diameter and morphology of the NPs.
  • the samples were coated in an argon atmosphere with a 10 nm thickness of gold and palladium (Emitech K550 Sputter Coated, Emitech Ltd., Ashford, Kent, UK).
  • Electron microphotographs of at least 500 NPs per preparation were evaluated with image analysis (Image Proplus, Media Cybernetics, Silver Spring, MD, USA) to determine the percentage dimensional distribution.
  • NP polymer-modifying hydrophilic groups (peptide chains)
  • the spectra were recorded in FAT (fixed retard ratio) mode with 190 eV of pass energy.
  • the pressure in the chamber containing the samples was evaluated at approx. 10 ⁇ 9 mbars.
  • Data acquisition and processing was conducted with an RBD AugerScan 2.
  • the spectra (reported in Figs. 3 and 4) show the presence of N on the surface of the NPs.
  • NPs to cross the BBB was determined by an in vivo test (the in situ brain perfusion technique). The actual crossing of the BBB is usually determined by evaluating the pharmacological effect of a substance present in nanoparticle systems (dalargin, etc.) or using labelled products or polymers, followed by evaluation of the radioactivity present in brain homogenate; however, these methods do not demonstrate that the NPs have actually crossed the BBB. Fluorescent NPs have only been used in two cases (NP of PBCA-polysorbate 80 and NP of PHDCA PEGylate (3,16), which allows their subsequent determination in brain parenchyma by fluorescence microscopy.
  • This detection system has the advantage of allowing effective evaluation of whether the BBB has been crossed, and the method was used in the present study because of this characteristic.
  • the NPs according to the invention were rendered fluorescent by inclusion of 20% in weight of PLGA derivatised with fluorescein (17).
  • mice Male albino Wistar Hannover rats weighing 250 ⁇ 30 g (Harlan, San Pietro Natisone) were used; the animals were housed for a period of 15 days at 25°C before the experiments, and fed on a standard diet with water on demand.
  • NP neuropeptide
  • a suspension of NP in saline (1 mL, containing 5 mg of Np each animal weighting 250 g) was then administered without interrupting the perfusion of plasma-like fluid (19) with a 3 -way valve.
  • the NP suspension (10 mg/0.5 mL of saline solution) was obtained by sonication and agitation in a vortex; the integrity of the NPs and the absence of aggregates were evaluated in advance by SEM analysis.
  • the brain was removed, subjected to preservation procedures by washing with saline solution (0.9% w/v), and frozen with liquid nitrogen. A number of sections were cut with a 5 ⁇ m thick cryotome.
  • DAPI DAPI-4'-6-diamidino-2- phenylindole

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Medicinal Chemistry (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biochemistry (AREA)
  • Nanotechnology (AREA)
  • Biophysics (AREA)
  • Composite Materials (AREA)
  • Condensed Matter Physics & Semiconductors (AREA)
  • General Physics & Mathematics (AREA)
  • Materials Engineering (AREA)
  • Crystallography & Structural Chemistry (AREA)
  • Physics & Mathematics (AREA)
  • Peptides Or Proteins (AREA)
  • Medicinal Preparation (AREA)

Abstract

Disclosed is a peptide comprising the sequence: H2N-Gly-Phe-D-Thr-Gly-Phe-Leu-Ser-CONH2, wherein the serine residue may be functionalised with sugar residues; other aminoacids can replace the first two aminoacids of the N-terminal portion, the order of which can be reversed, and their number may be different from two.

Description

DRUG DELIVERY PEPTIDES
This invention relates to new peptides which are useful for carrying drugs through the blood-brain barrier. PRIOR ART
There is considerable interest in the development of new systems able to carry drugs efficiently and specifically to the target tissues in which the substances are to perform their therapeutic action. One of the most difficult challenges is represented by the blood-brain barrier (BBB), which regulates access to the brain by the substances present in the bloodstream in a highly selective manner. The BBB is formed by special endothelial cells joined by tight junctions. This membrane selectively regulates the entry of drugs which could have therapeutic value in the treatment of psychiatric and other disorders if they reached the neurones. One approach to crossing the BBB is represented by the synthesis of suitable prodrugs; recent studies have also investigated the use of liposomes, nanoparticles (NPs), small polymeric colloidal particles (1-1000 nm), or solid lipid nanoparticles into which the drug can be adsorbed or included (1,2).
The use of NPs has the advantage that a large number of molecules of the drug can be carried by each NP, and that they mask the characteristics of the drug to enable it to cross the BBB and protect it against enzymatic degradation. Different strategies have been proposed to enable NPs to cross the BBB, most of which are based on modifications to their hydrophobic surface. NPs mainly consist of polycyanoacrylate (such as poly(butylcyanoacrylate, PBCA), and a hydrophilic polymer such as polysorbate 80 can be adsorbed onto their surface (2); another strategy involves the preparation of NPs from an amphiphilic copolymer wherein the hydrophobic component is able to form the solid phase (the body of the NP), while the hydrophilic component remains on its surface, facing outwards. This copolymer can be represented by polyethylene glycol (PEG) and n- hexadecylcyanoacrylate (PEG-PHDCA) (3). While it has been postulated on the basis of experimental data that the presence of the surfactant polysorbate 80 may act on the BBB, causing an increase in its permeability, NPs constituted by PHDCA PEGylate are able to cross the BBB without modifying its permeability (3); PEGylation increases the plasma half-life of NPs, and it has been suggested that this may be the reason why they are able to penetrate into the brain to a greater extent than other formulations (3). If the NPs are not coated with surfactant, they remain inside the blood vessels (2). Various mechanisms have been postulated to explain how NPs cross the BBB (1,4). However, although it is important for NPs to be able to cross the BBB, it is also extremely important for them to have the brain as their priority target, so that they can be used with drugs aimed at the CNS, not at other organs or apparatus.
Receptor-mediated endocytosis is responsible for the fact that the BBB is crossed not only by molecules with a low molecular weight such as thiamine, but also by aggregates with a high molecular weight such as low-density lipoproteins (LDL), and by other macromolecules. Specific receptors for insulin, insulin-like growth factors (IGF-I, IGF-2), angiotensin II, atrial natriuretic peptide (ANP), brain natriuretic peptide (BNP), interleukin-1 (IL-I) and transferrin have been identified in the brain capillaries. The transcytosis of these molecules through the BBB in vivo has so far only been demonstrated for insulin and transferrin (1). NPs consisting of wax and a surfactant (Brji 78®) conjugated with thiamine (5), or nanogels consisting of flexible hydrophilic polymers of nanometric dimensions (nanogels) composed of polyethyleneimine crosslinked to poly(ethyleneglycol), which in turn are derivatised with transferrin or insulin, have been created specifically to reach the CNS (6,7).
Peptides are polar compounds, but various proteins are able to cross the lipid cell membranes; there is great interest in studying the possibility of increasing the cell permeability of liposomes, proteins and NPs by using membrane-permeable peptides and proteins as carriers (8). Although the primary structure of the peptides able to cross the cell membranes is highly variable (9), some of their general characteristics are known. It has been postulated that a certain abundance of positive charges due to the presence of
Arg (10-12), and the presence of aminoacids with large, hydrophobic side chains (13) inserted in the peptide sequence, play a crucial role in effective cell uptake.
Recently, various synthetic analogues of opioid peptides have proved able to cross the BBB, and this permeability can be increased by the presence of sugar residues (glucose, lactose, etc.); however, the mechanism whereby these substances cross the BBB is not known. In particular, an opioid peptide able to cross the BBE, known as MMP-2200, with the formula H2N-Tyr-D- Thr-Gly-Phe-Leu-Ser-O-β-D-lactose-CONH2, has been described (14).
The derivatisation of PLGA with short membrane-permeable peptide chains was recently successfully used to prepare NPs able to penetrate into cultured HaCat cells. In this case, PLGA was derivatised with the peptide Tat 49-57, which is the transduction domain of the Tat protein of HIV virus type 1; the NPs thus obtained were carried to the nucleus of the cultured cells without causing cytotoxicity (15). However the need remains for alternative, more effective and/or safer delivery systems. DESCRIPTION OF THE INVENTION
It has now been discovered that the peptide sequence:
H2N-Gly-Phe-D-Thr-Gly-Phe-Leu-Ser-CONH2 can be opportunely used to carry through the blood-brain barrier drugs conjugated to said sequence either directly or via a linear or branched polyvalent spacer, wherein other aminoacids can replace the first two aminoacids of the N-terminal portion, the order of which can be reversed, and their number may be different from two. A first aspect of the invention therefore relates to a peptide having the sequence reported above, wherein the serine residue may be functionalised with sugar residues, with a C- or O-glycoside bond.
The sugar residue is preferably a residue of glucose, galactose, lactose, mannose or xylose, and more preferably an O-β-D-glucoside residue. The invention also relates to a conjugate of said peptides with a pharmaceutically acceptable polyester or polyamide polymer, nanoparticle systems comprising said conjugates, and pharmaceutical compositions comprising said nanoparticle systems.
DESCRIPTION OF FIGURES Fig. 1. 1H-NMR spectrum of conjugate PLGA-HN-Gly-Phe-D-Thr-Gly-
Phe-Leu-Ser-CONH2.
Fig. 2. 1H-NMR spectrum of conjugate PLGA-HN-Gly-Phe-D-Thr-Gly- Phe-Leu-SerO-β-D-glucose-CONH2.
Fig. 3. ESCA spectrum of NPs prepared with PLGA-HN-Gly-Phe-D- Thr-Gly-Phe-Leu-Ser-CONH2.
Fig. 4. ESCA spectrum of NPs prepared with PLGA-HN-Gly-Phe-D- Thr-Gly-Phe-Leu-SerO-β-D-glucose-CONH2.
Fig. 5. SEM image of NPs prepared with PLGA-HN-Gly-Phe-D-Thr- Gly-Phe-Leu-Ser-CONH2. Fig. 6. SEM image of NPs prepared with PLGA-HN-Gly-Phe-D-Thr-
Gly-Phe-Leu-SerO-β-D-glucose-CONH2.
DETAILED DESCRIPTION OF THE INVENTION
Examples of polymers which can be advantageously used to prepare the conjugates according to the invention include polymers or copolymers of biodegradable aliphatic hyα"roxyacids, preferably lactic acid and/or glycolic acid. The copolymer poly(D5L-lactide-cø-glycolide) (PLGA) or poly(D,L- lactic) acid is particularly preferred. The copolymer PLGA is a biodegradable polyester, approved by the American FDA, which breaks down without inducing inflammation or immune reactions, and is therefore particularly suitable for the purposes of this invention.
The peptides according to the invention can be conjugated to PLGA or other polymers containing reactive groups (typically carboxyl groups) with the amino-terminal groups of the peptide according to methods already known in themselves, such as by activation of the carboxyl group with N-hydroxysuccinimide in the presence of dicyclohexylcarbodiimide, and subsequent formation of an amide bond with the N-terminal (unprotected) portion of the peptide. Alternatively, the peptide can be conjugated through a suitable all-purpose spacer, such as the aminoacid "Behera's amine". The molecular weight of the polymer used for the conjugation to the peptide is not critical, but typically falls into the interval between 1000 and 50,000 Da; the degree of functionalisation of the polymer with the peptide is between 30 and 80% of the theoretical value. The conjugates thus obtainable can be used to prepare nanoparticle systems according to known techniques, for example as described in (18) and in T. Niwa et al., J. Pharm. Sci. (1994) 83, 5, 727-732; C. S. Cho et al., Biomaterials (1997) 18, 323-326; T. Govender et al., J. Control. ReI. (1999) 57, 171-185; and M. F. Zambaux et al., J. Control. ReI. (1998) 50, 31-40. In addition to the PLGA-peptide conjugate, the nanoparticles (NP) according to the invention will contain a drug, a suitable surfactant, and a pharmaceutical carrier such as water or aqueous saline solutions.
Other micro- or nanoparticle systems (liposomes, microemulsions etc.) could be considered as an alternative to NPs.
The drugs which can be advantageously used according to this invention are obviously those which are required to perform their pharmacological action in the brain. Examples of these drugs include antibacterial, antiviral, psychotropic, antidepressant, anti-tumoral and antimigraine agents, analgesics, antagonists or agonists of receptors present in the central nervous system, antibodies, antisense oligonucleotides, hormones, narcotic analgesics, and diagnostic agents (paramagnetic complexes for MRI, radionuclides for nuclear medicine or diagnosis, etc.). The drugs can be included, adsorbed or absorbed into the nanoparticles or conjugated directly with the peptide or copolymer by means of a covalent bond which is labile in vivo, such as an ester or amide bond.
The invention therefore also relates to pharmaceutical or diagnostic compositions comprising a peptide or one of its conjugates as defined above, together with any excipients. The dose and administration route of the nanometric systems according to the invention will obviously depend on a number of factors, such as the toxicological and pharmacokinetic characteristics of the drug, the severity of the disorder to be treated, and the patient's condition, especially his/her age, weight and sex. The dose may in any event be determined on the basis of the results of pre-clinical trials and phase I clinical trials, as is usual in drug development.
The invention will be now be described in greater detail by means of the following Examples.
EXAMPLE 1 - Peptide synthesis The peptides were synthesised by solid-phase peptide synthesis (SPPS) using a Rink amide resin and the FMOC method. All the peptides were synthesised manually using a suitable reactor (scale of 0.5 g of resin, 5.00 ml washing volume) and agitation by anhydrous nitrogen flow. Rink amide resin (4-[[(2',4'-dimethoxy-phenyl)(9-fluorenylmethoxycarbonyl)amino]methyl] phenoxyl resin) with a degree of functionalisation of 0.63 mmol/g was used. The Ser used to prepare the peptide H2N-Gly-Phe-D-Thr-Gry-Phe-Leu-Ser- CONH2 (Peptide 1) was protected as tert-butyl ether; the hydroxyl groups of FMOC-SerO-β-D-glucoside, used for synthesis of the peptide H2N-Gly-Phe- D-Thr-Gly-Phe-Leu-SerO-β-D-glucose-CONH2 (peptide 2), were protected by formation of esters with acetic acid. The FMOC was removed with the piperidine/DMF (9:1, 2 x 5 min), followed by washing with DMF (5 x 1 min). FMOC release was monitored by UV spectrophotometry (I = 290nm). The couplings were performed by adding a solution in DMF (4ml) consisting of FMOC aminoacid (3 equiv.), TBTU (3 equiv.), HOBt (3 equiv.) and *Pr2Net (6 equiv.) to the peptide present on the resin. The resin suspension was agitated by bubbling anhydrous nitrogen for 3 h, after which the degree of completeness of coupling was determined with the Kaiser test. When the peptide is entirely synthesised, the FMOC group is removed as reported above; the acetic groups present on the sugar residue are removed by treatment with H2N-NH2.H2O/MeOH 4/1 (2 x 5 mL x 30 min, followed by 1 x 5 mL x 1 h), while the peptide remains bonded to the resin. The excess H2N-NH2-H2O is then removed, and the resin is washed with MeOH (5 x 1 min) followed by CH2Cl2 (5 x 1 min). Next, the peptide is released from the dried resin by treatment with CF3COOH (TFA) (84%), anisole (4%) thioanisole (4%), ethanedithiol (4%) and H2O (4%) (4 ml x 2 h); the solution is filtered to remove the depleted resin, and the resulting solution is cooled and precipitated by adding anhydrous ether. The solid thus obtained is filtered and washed several times with anhydrous ether; the crude peptide is then purified by dissolving in MeOH and precipitating with anhydrous ether. The peptides thus obtained are then analysed by mass spectroscopy (Q-TOF Global Ultima, Micromass, MALDI) (m/z): Peptide 1, est. 726.8, found 727.4 (M+); peptide 2, est. 889.0, found 889.4 (M+), and their purity is determined by HPLC (UV detector, I 210 nm; column Vydac 218, 4.6 x 250 mm, Cl 8, pore diameter 300 A, packing particle size 5 μm; mobile phase Solvent A: H2O/0.1% TFA; solvent B: CH3CNA).1%TFA; elution conditions: linear gradient from 95%A to 35%A in 35 min; flow rate 0.8 ml/min. Rt: peptide 1, 20.5 min, peptide 2, 19.11 min.; purity >90% in both cases).
EXAMPLE 2 - Conjugation of PLGA with peptides 1 and 2 Dicyclohexylcarbodiimide (19.0 mg, 93 μmol) and N-hydroxy- succinimide (11.0 mg, 93 μmol) is added to a solution of 1.00 g (88 μmol) PLGA RG503H (Boehringer Ingelheim) with a molecular weight of 11,000, determined by titration of the carboxyl groups at its ends (4.94 mg KOH/g of polymer) in anhydrous dioxane (5 ml); the solution is then agitated for 4 hours at a temperature ranging from the initial 40C to 200C. The dicyclohexylurea is then filtered, and the solution poured into ethyl ether. The insoluble polymer is separated by decanting and purified by dissolving in CH2Cl2 and precipitating with anhydrous ether (twice). The polymer thus obtained is then dissolved in anhydrous DMSO (5 ml), and maintained under agitation at ambient temperature after addition of a solution of the peptide (80 μmol) in DMSO (1 ml) and triethylamine (17 μl, 120 μmol). After 4 hours at ambient temperature, the reaction solution is poured into ethyl ether; the modified polymer is separated by decanting and purified by solubilisation in CH2Cl2 and precipitation with methanol (three times).
Confirmation of conjugation of PLGA with the peptide.
Conjugation was confirmed by 1H-NMR spectroscopy. Examination of the spectra shown in Figs. 1 and 2 demonstrates the presence of the signal at 7-8 ppm attributable to the presence of aromatic protons of the Phe's present in the peptide. The derivatisation yield of the PLGA polymer was determined by the ratio between the areas of the signals at 7.2-7.5 ppm, corresponding to the aromatic protons of the Phe's, and the areas of the multiplet at 1.80-1.60 ppm, corresponding to the protons of the methyl groups of the polymer; in both cases (PLGA-peptide 1 and PLGA-peptide 2), the degree of functionalisation was 30 micromoles of peptide/g of polymer. EXAMPLE 3 - preparation of nanoparticles using the conjugates
PLGA-peptide 1 and PLGA-peptide 2
The nanoparticles were prepared by the nanoprecipitation technique (18). The conjugate PLGA-peptide 1 or PLGA-peptide 2, prepared as above (100 mg), and PLGA derivatised with fluorescein (17) (25 mg), were solubilised in acetone (8 ml). The solution thus obtained was poured slowly into deionised water (25 ml) containing Pluronic F68 (100 mg). After agitation at ambient temperature for 10 min, the organic solvent was removed at 300C at low pressure, and the final volume of the suspension adjusted to 10 mL with deionised water; the NPs were then purified by gel-filtration chromatography (Sepharose CL4B gel (160 ml), column 50 x 2 cm), using water as mobile phase, and freeze-dried without the presence of a cryoprotector. Analysis of NPs.
A scanning electron microscope (SEM) (XL-40 Philips, Eindhoven, Netherlands) was used to evaluate the diameter and morphology of the NPs. The samples were coated in an argon atmosphere with a 10 nm thickness of gold and palladium (Emitech K550 Sputter Coated, Emitech Ltd., Ashford, Kent, UK). Electron microphotographs of at least 500 NPs per preparation were evaluated with image analysis (Image Proplus, Media Cybernetics, Silver Spring, MD, USA) to determine the percentage dimensional distribution. Each sample of NP has an average diameter of 120 ± 45 nm, and presents a uniform distribution and an intact surface, both before and after freeze-drying and resuspension, thus confirming that the preparation method and resuspension do not alter the morphological characteristics of NPs. In order to determine the position of the polymer-modifying hydrophilic groups (peptide chains), X-ray Induced Photoelectronic Spectrophotometry analysis (ESCA) was conducted with an 04-153 X-ray source instrument (PHI, Uvalca-PHI, Tokyo, Japan) and an EAIl hemispheric electron energy analyser (Leybold Optics, Germany), using MgK radiation αl,2 (£=1253.6 eV). The spectra were recorded in FAT (fixed retard ratio) mode with 190 eV of pass energy. The pressure in the chamber containing the samples was evaluated at approx. 10~9 mbars. Data acquisition and processing was conducted with an RBD AugerScan 2. The spectra (reported in Figs. 3 and 4) show the presence of N on the surface of the NPs.
EXAMPLE 4 - Experiments in vivo: in situ brain perfusion technique.
The ability of NPs to cross the BBB was determined by an in vivo test (the in situ brain perfusion technique). The actual crossing of the BBB is usually determined by evaluating the pharmacological effect of a substance present in nanoparticle systems (dalargin, etc.) or using labelled products or polymers, followed by evaluation of the radioactivity present in brain homogenate; however, these methods do not demonstrate that the NPs have actually crossed the BBB. Fluorescent NPs have only been used in two cases (NP of PBCA-polysorbate 80 and NP of PHDCA PEGylate (3,16), which allows their subsequent determination in brain parenchyma by fluorescence microscopy. This detection system has the advantage of allowing effective evaluation of whether the BBB has been crossed, and the method was used in the present study because of this characteristic. The NPs according to the invention were rendered fluorescent by inclusion of 20% in weight of PLGA derivatised with fluorescein (17).
Male albino Wistar Hannover rats weighing 250 ± 30 g (Harlan, San Pietro Natisone) were used; the animals were housed for a period of 15 days at 25°C before the experiments, and fed on a standard diet with water on demand.
Each sample of NP was perfused into the rats after anaesthesia induced with an i.p. injection of an aqueous solution (2 mL/Kg) of xylazine (0.001 mg/Kg) and ketamine (0.125 mg/Kg). The behaviour of the NPs was determined by the in situ brain perfusion technique (5,19,20). The rectal temperature of the rats during the experiment was monitored and maintained at 37°C by a heating device connected to a feedback mechanism.
A suspension of NP in saline (1 mL, containing 5 mg of Np each animal weighting 250 g) was then administered without interrupting the perfusion of plasma-like fluid (19) with a 3 -way valve. The NP suspension (10 mg/0.5 mL of saline solution) was obtained by sonication and agitation in a vortex; the integrity of the NPs and the absence of aggregates were evaluated in advance by SEM analysis. After the rats has been killed, the brain was removed, subjected to preservation procedures by washing with saline solution (0.9% w/v), and frozen with liquid nitrogen. A number of sections were cut with a 5 μm thick cryotome. Some sections were stained with haematoxylin-eosin for observation of the histological morphology of the organ section. Some tissue samples were treated with DAPI (4'-6-diamidino-2- phenylindole), a compound able to form fluorescent blue complexes with double-chain DNA (21), to highlight the presence of the cell nuclei.
After treatment of the sections with DAPI, the samples were observed with a fluorescence microscope with dual-band excitation relating to DAPI and fluorescein isothiocyanate (FITC) and an emission filter for fluorescence images (enlargement 40-10Ox), and with a confocal microscope (SP2-AOBS, Leika, Bannockburn, IL, USA). Green fluorescence, due to the fluorescein- PLGA polymer, constitutes the visual marker for NPs. Results
Although it is impossible to obtain quantitative data from the experiments conducted, the data analysis obtained from the in vivo study with fluorescence microscopy clearly shows that the NPs obtained with PLGA modified with the peptide H2N-Gly-Phe-D-Thr-Gly-Phe-Leu-Ser-CONH2 (peptide 1), and to a greater extent with the peptide H2N-Gly-Phe-D-Thr-Gly- Phe-Leu-SerO-β-D-glucose-CONH2 (peptide 2), are able to cross the BBB and penetrate into the brain parenchyma cells. However, NPs only constituted by PLGA are unable to cross the BBB, and remain in the blood vessels (capillaries).
In order to obtain a precise determination of the location of the NPs, studies were conducted with a confocal microscope. This detailed information cannot be obtained with fluorescence microscopy alone, which only provides information in a two-dimensional (2D) mode. The three-dimensional images obtained with confocal fluorescence microscopy enable the position of the NPs to be located (green fluorescence due to the presence of fluorescein) in relation to the nuclei of neuronal cells (blue fluorescence due to staining with DAPI).
There is no doubt that fluorescent NPs consisting only of PLGA are unable to cross the BBB. This result confirms that no damage to the BBB took place during the experiments. Conversely, NPs prepared with PLGA-peptide 1 and PLGA-peptide 2 were observed in the brain parenchyma, in close contact with the nuclear structures, and in some cases in the cytoplasm of nonvascular cells. References
1. Reddy, J. S.; Venkateswarlu, V. Novel delivery systems for drug targeting to the brain. Drugs Fut. 29, 63 (2004). 2. Lockman, P.R.; Mumper, RJ.; Khan, M.A.; Allen, D.D. Nanoparticle technology for drug delivery across the blood brain barrier. Drug Dev. Ind. Pharm. 28, 1-13 (2002).
3. Calvo, P., Gouritin, B.; Chacun, H.; Desmaele, D.; D'Angelo, J.; Noel, J.P.; Georgin, D.; Fattal, E.; Andreux, J.P.; Couvreur, P. Long-circulating PEGylated polycyanoacrylate nanoparticles as new drug carrier for brain delivery. Pharm. Res. 18, 1157-1166 (2001).
4. Kreuter, J. Nanoparticulate systems for brain delivery of drugs. Adv Drug del Rev. 47, 65-81 (2001).
5. Lockman, P.R.; Oyewumi, M.O.; Koziara, J.M.; Roder, K.E.; Mumper, RJ.; Allen, D.D. Brain uptake of thiamine coated nanoparticles. J. Contr. ReI.
93, 271-282 (2003).
6. A.V. Kabanov, E.V. Batrakova, New technologies for drug delivery across the blood brain barrier. Curr. Pharm. Des. 10, 1355-1363 (2004).
7. Vinogradov, S.V.; Batrakova, E.V.; Kabanov, A.V. Nanogels for oligonucleotide delivery to the brain. Bioconj. Chem. 15, 50-60 (2004).
8. Lindgren, M.; Hallbrink, M.; Prochiantz, A.; Laugel, U. Cell penetrating peptides. Trends in Pharmacol. Sci. 21, 99-102 (2000).
9. Langel, U. Cell penetrating peptides, Processes and applications. CRC Press, Boca Raton (2002). 10. Futaki, S.; Suzuki, T.; Ohashi, W.; Yagami, T.; Tanaka, S.; Ueda, K.; Sogiura, Y. Arginine-rich peptides. An abundant source of membrane- permeable peptides having potential as carrier for intracellular protein delivery. J. Biol. Chem. 276, 5836-5840 (2001). 11. Mitchell, DJ.; Kim, D.T.; Steinman, L.; Fathman, C.G.; Rothbard, J.B. Polyarginine enters cells more efficiently than other polycationic homopolymers. J. Pept. Res. 56, 318-325 (2000).
12. Wender, P.A.; Mitchell, DJ.; Pattabiraman, K.; Pelkey, E.T.; Steinman, L.; Rothbard, J.B. The design, synthesis, and evaluation of molecules that enable or enhance cellular uptake: peptoid molecular transporters. Proc. Nat. Acad. Sci. USA 97, 13003 (2000).
13. Derossi, D.; Chassaing, G.; Prochiantz, A. Trojan peptides: the penetration system for intracellular delivery. Trends Cell Biol. 8, 84-87 (1998).
14. PoIt, R.; Palian, M.M.; Glycopeptide analgesics. Drugs Fut. 26, 561- 576 (2001).
15. Nam, Y.S.; Park, Y.; Han, S.H.; Chang, LS. Intracellular drug delivery using poly(D,L-lactide-co-glycolide) nanoparticles derivatized with a peptide from a transcriptional activator protein of HIV-I. Biotechnol. Lett. 24, 2093-2098 (2003).
16. Kreuter, J.; Alyantdin, R.N.; Kharkevich, D.A.; Ivanov, A.A. Passage of peptides through the blood-brain barrier with colloidal polymer particles (nanoparticles). Brain Res 674, 171-174 (1995). 17. Tosi, G.; Rivasi, F.; Gandolfi, F.; Costantino, L.; Vandelli, M.A.; Forni, F. Conjugated poly-(D,L-lactide-co-glycolide) for the preparation of in-vivo detectable nanoparticles. Biomaterials, in press.
18. Fessi, H.; Puisieux, F.; Devissaguet, J.P.; Ammoury, N.; Benita, S. Nanocapsule formation by interfacial polymer deposition following solvent displacement. Int. J. Pharm. 55, R1-R4 (1989).
19. Takasato, Y.; Rapaport, S.I.; Smith, Q.R. An in situ brain perfusion technique to study cerebrovascular transport in the rat. Am. J. Physiol. 247, H484-93 (1984). 20. Lockman, P.R.; Mumper, R.J.; Allen, D.D. Evaluation of blood-brain barrier thiamine efflux using the in situ rat brain perfusion method. J. Neurochem. 86, 627-634 (2003).
21. Xu, Z.; Pilch, D.S.; Srinivasan, A.R.; Olson, W.K.; Geacintov, N.E.; Breslauer, KJ. Modulation of nucleic acid structure by ligand binding: induction of a DNA-RNA-DNA hybrid triplex by DAPI intercalation. Bioorg. Med. Chem. 5, 1137-1147 (1997).

Claims

1. A peptide comprising the sequence:
H2N-Gly-Phe-D-Thr-Gly-Phe-Leu~Ser-CONH23 wherein the serine residue may be functionalised with sugar residues; other aminoacids can replace the first two aminoacids of the N-terminal portion, the order of which can be reversed, and their number may be different from two.
2. A peptide as claimed in claim 1, wherein the serine residue is not glycosylated.
3. A peptide as claimed in claim 1, wherein the serine residue is C- or O-glycosylated.
4. A peptide as claimed in claim 3, wherein the sugar residue is a residue of glucose, galactose, lactose, mannose or xylose.
5. A peptide as claimed in claim 4, wherein the sugar residue is an O-β-D- glucoside residue.
6. A peptide as claimed in any of claims 1 to 5, constituted by the sequence H2N-Gly-Phe-D-Thr-Gly-Phe-Leu-Ser-CONH2 as defined above.
7. A peptide as claimed in any of claims 1 to 6, selected from among the peptides in the sequence: H2N-Gly-Phe-D-Thr-Gly-Phe-Leu-Ser-CONH2
H2N-Gly-Phe-D-Thr-Gly-Phe-Leu-SerO-β-D-glucose-CONH2.
8. Conjugates of peptides as claimed in claims 1 to 7 with pharmaceutically acceptable polyester or polyamide polymers.
9. Conjugates as claimed in claim 8, wherein said polymer is a polymer or copolymer of lactic acid and/or glycolic acid.
10. Conjugates as claimed in claim 9, wherein the polymer is poly (D,L-lactide-co-glycolide) or poly(D,L-lactic) acid.
11. Nanoparticle systems comprising a conjugate as claimed in claims 8-10.
12. Pharmaceutical compositions comprising the nanoparticle systems claimed in claim 11.
13. Use of the peptides claimed in claims 1 to 7 or the conjugates claimed in claims 8 to 12 to prepare medicinal products designed to cross the blood-brain barrier.
14. Use as claimed in claim 13, wherein said medicinal products take the form of nanoparticle systems.
PCT/EP2005/012554 2004-12-10 2005-11-24 Drug delivery peptides for crossing blood-brain barrier WO2006061101A2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
EP05817909A EP1819723A2 (en) 2004-12-10 2005-11-24 Drug delivery peptides for crossing blood-brain barrier

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
ITMI2004A002353 2004-12-10
IT002353A ITMI20042353A1 (en) 2004-12-10 2004-12-10 PEPTIDES FOR DRUG VEHICLES

Publications (2)

Publication Number Publication Date
WO2006061101A2 true WO2006061101A2 (en) 2006-06-15
WO2006061101A3 WO2006061101A3 (en) 2006-09-14

Family

ID=36578268

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2005/012554 WO2006061101A2 (en) 2004-12-10 2005-11-24 Drug delivery peptides for crossing blood-brain barrier

Country Status (3)

Country Link
EP (1) EP1819723A2 (en)
IT (1) ITMI20042353A1 (en)
WO (1) WO2006061101A2 (en)

Cited By (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2010027108A1 (en) * 2008-09-08 2010-03-11 国立大学法人東京工業大学 Fluorescent sugar derivative compound and sensor using same
US20110229555A1 (en) * 2010-03-22 2011-09-22 Signpath Pharma Inc. Intravenous curcumin and derivatives for treatment of neurodegenerative and stress disorders
US8747890B2 (en) 2009-11-19 2014-06-10 Signpath Pharma Inc. Intravenous infusion of curcumin and a calcium channel blocker
US9138411B2 (en) 2012-08-31 2015-09-22 University Of North Texas Health Science Center At Fort Worth Curcumin-ER, a liposomal-PLGA sustained release nanocurcumin for minimizing QT prolongation for cancer therapy
US9682041B2 (en) 2011-06-03 2017-06-20 Signpath Pharma Inc. Liposomal mitigation of drug-induced long QT syndrome and potassium delayed-rectifier current
EP3252068A2 (en) 2009-10-12 2017-12-06 Larry J. Smith Methods and compositions for modulating gene expression using oligonucleotide based drugs administered in vivo or in vitro
US10117881B2 (en) 2011-06-03 2018-11-06 Signpath Pharma, Inc. Protective effect of DMPC, DMPG, DMPC/DMPG, LYSOPG and LYSOPC against drugs that cause channelopathies
US10238602B2 (en) 2011-06-03 2019-03-26 Signpath Pharma, Inc. Protective effect of DMPC, DMPG, DMPC/DMPG, LysoPG and LysoPC against drugs that cause channelopathies
US10349884B2 (en) 2011-06-03 2019-07-16 Sighpath Pharma Inc. Liposomal mitigation of drug-induced inhibition of the cardiac ikr channel
US10449193B2 (en) 2011-06-03 2019-10-22 Signpath Pharma Inc. Protective effect of DMPC, DMPG, DMPC/DMPG, lysoPG and lysoPC against drugs that cause channelopathies
US10532045B2 (en) 2013-12-18 2020-01-14 Signpath Pharma, Inc. Liposomal mitigation of drug-induced inhibition of the cardiac IKr channel
US11806401B2 (en) 2016-04-27 2023-11-07 Signpath Pharma, Inc. Prevention of drug-induced atrio-ventricular block

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002089776A1 (en) * 2001-05-05 2002-11-14 Lts Lohmann Therapie-Systeme Ag Nanoparticles made of protein with coupled apolipoprotein e for penetration of the blood-brain barrier and methods for the production thereof

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002089776A1 (en) * 2001-05-05 2002-11-14 Lts Lohmann Therapie-Systeme Ag Nanoparticles made of protein with coupled apolipoprotein e for penetration of the blood-brain barrier and methods for the production thereof

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
COSTANTINO LUCA ET AL: "Peptide-derivatized biodegradable nanoparticles able to cross the blood-brain barrier." JOURNAL OF CONTROLLED RELEASE : OFFICIAL JOURNAL OF THE CONTROLLED RELEASE SOCIETY. 2 NOV 2005, vol. 108, no. 1, 2 November 2005 (2005-11-02), pages 84-96, XP002386303 ISSN: 0168-3659 *
ELMAGBARI NURA O ET AL: "Antinociceptive structure-activity studies with enkephalin-based opioid glycopeptides." THE JOURNAL OF PHARMACOLOGY AND EXPERIMENTAL THERAPEUTICS. OCT 2004, vol. 311, no. 1, October 2004 (2004-10), pages 290-297, XP002386302 ISSN: 0022-3565 *

Cited By (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2010027108A1 (en) * 2008-09-08 2010-03-11 国立大学法人東京工業大学 Fluorescent sugar derivative compound and sensor using same
EP3252068A2 (en) 2009-10-12 2017-12-06 Larry J. Smith Methods and compositions for modulating gene expression using oligonucleotide based drugs administered in vivo or in vitro
EP4089169A1 (en) 2009-10-12 2022-11-16 Larry J. Smith Methods and compositions for modulating gene expression using oligonucleotide based drugs administered in vivo or in vitro
US8747890B2 (en) 2009-11-19 2014-06-10 Signpath Pharma Inc. Intravenous infusion of curcumin and a calcium channel blocker
US20110229555A1 (en) * 2010-03-22 2011-09-22 Signpath Pharma Inc. Intravenous curcumin and derivatives for treatment of neurodegenerative and stress disorders
US9393198B2 (en) * 2010-03-22 2016-07-19 Signpath Pharma Inc. Intravenous curcumin and derivatives for treatment of neurodegenerative and stress disorders
US10238602B2 (en) 2011-06-03 2019-03-26 Signpath Pharma, Inc. Protective effect of DMPC, DMPG, DMPC/DMPG, LysoPG and LysoPC against drugs that cause channelopathies
US10117881B2 (en) 2011-06-03 2018-11-06 Signpath Pharma, Inc. Protective effect of DMPC, DMPG, DMPC/DMPG, LYSOPG and LYSOPC against drugs that cause channelopathies
US9682041B2 (en) 2011-06-03 2017-06-20 Signpath Pharma Inc. Liposomal mitigation of drug-induced long QT syndrome and potassium delayed-rectifier current
US10349884B2 (en) 2011-06-03 2019-07-16 Sighpath Pharma Inc. Liposomal mitigation of drug-induced inhibition of the cardiac ikr channel
US10357458B2 (en) 2011-06-03 2019-07-23 Signpath Pharma Inc. Liposomal mitigation of drug-induced long QT syndrome and potassium delayed-rectifier current
US10449193B2 (en) 2011-06-03 2019-10-22 Signpath Pharma Inc. Protective effect of DMPC, DMPG, DMPC/DMPG, lysoPG and lysoPC against drugs that cause channelopathies
US10617639B2 (en) 2011-06-03 2020-04-14 Signpath Pharma, Inc. Liposomal mitigation of drug-induced long QT syndrome and potassium delayed-rectifier current
US9138411B2 (en) 2012-08-31 2015-09-22 University Of North Texas Health Science Center At Fort Worth Curcumin-ER, a liposomal-PLGA sustained release nanocurcumin for minimizing QT prolongation for cancer therapy
US10532045B2 (en) 2013-12-18 2020-01-14 Signpath Pharma, Inc. Liposomal mitigation of drug-induced inhibition of the cardiac IKr channel
US11806401B2 (en) 2016-04-27 2023-11-07 Signpath Pharma, Inc. Prevention of drug-induced atrio-ventricular block

Also Published As

Publication number Publication date
EP1819723A2 (en) 2007-08-22
WO2006061101A3 (en) 2006-09-14
ITMI20042353A1 (en) 2005-03-10

Similar Documents

Publication Publication Date Title
EP1819723A2 (en) Drug delivery peptides for crossing blood-brain barrier
Costantino et al. Peptide-derivatized biodegradable nanoparticles able to cross the blood–brain barrier
JP5211109B2 (en) Composite of transmembrane agent and pharmaceutically active ingredient
She et al. The potential of self-assembled, pH-responsive nanoparticles of mPEGylated peptide dendron–doxorubicin conjugates for cancer therapy
Costantino et al. Nanoparticulate drug carriers based on hybrid poly (D, L-lactide-co-glycolide)-dendron structures
JP4936312B2 (en) Novel amphiphile, drug delivery system and molecular imaging system using the same
Martín et al. Design, synthesis and characterization of a new anionic cell‐penetrating peptide: SAP (E)
WO2009108822A1 (en) Engineered tunable nanoparticles for delivery of therapeutics, diagnostics, and experimental compounds and related compositions for therapeutic use
KR101740895B1 (en) Brain targeting nano-carrier containing pluoronic polymer comprising brain targeting peptide capable of specifice delivery to brain tissue and chitosan
US6780846B1 (en) Membrane translocating peptide drug delivery system
WO2017063542A1 (en) Stabilized a7r polypeptides, and use thereof in constructing tumor targeted therapeutic drug delivery system
Zhang et al. Novel solid lipid nanoparticles as carriers for oral administration of insulin
JP4799790B2 (en) Membrane translocation peptide, composition containing the peptide, pharmaceutical preparation containing the peptide, and use in preparation of the preparation of the peptide
EP3405429B1 (en) Formation of functionalized nanoparticles by supramolecular co-assembly
WO2018103660A1 (en) Vap polypeptide and use thereof in preparation of drug for targeted diagnosis and treatment of tumour
Wang et al. Luteinizing-hormone-releasing-hormone-containing biodegradable polymer micelles for enhanced intracellular drug delivery
Jabbari Targeted delivery with peptidomimetic conjugated self-assembled nanoparticles
Yang et al. The application progress of peptides in drug delivery systems in the past decade
EP4141020A1 (en) Novel cell-penetrating peptide and use thereof
JP4566406B2 (en) Retro-inverted peptides targeting GIT transport receptors and related methods
US20180066031A1 (en) Enzymatically-cleavable peptide amphiphiles
Ivanova et al. Complex systems for dru g transport across cell membranes
Nakase Arginine‐Rich Cell‐Penetrating Peptides: Recent Advances of Design and Applications for Intracellular Delivery
KR20230006405A (en) Novel cell penetrating peptides and use thereof

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BW BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE EG ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KM KN KP KR KZ LC LK LR LS LT LU LV LY MA MD MG MK MN MW MX MZ NA NG NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SM SY TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): BW GH GM KE LS MW MZ NA SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IS IT LT LU LV MC NL PL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

WWE Wipo information: entry into national phase

Ref document number: 2005817909

Country of ref document: EP

NENP Non-entry into the national phase in:

Ref country code: DE

WWP Wipo information: published in national office

Ref document number: 2005817909

Country of ref document: EP