WO2016038534A1 - Complexes de fenrétinide - Google Patents

Complexes de fenrétinide Download PDF

Info

Publication number
WO2016038534A1
WO2016038534A1 PCT/IB2015/056862 IB2015056862W WO2016038534A1 WO 2016038534 A1 WO2016038534 A1 WO 2016038534A1 IB 2015056862 W IB2015056862 W IB 2015056862W WO 2016038534 A1 WO2016038534 A1 WO 2016038534A1
Authority
WO
WIPO (PCT)
Prior art keywords
fenretinide
complexes
cancers
salt
carrier
Prior art date
Application number
PCT/IB2015/056862
Other languages
English (en)
Inventor
Isabella Orienti
Ruggero De Maria Marchiano
Ann Pegna Zeuner
Original Assignee
Istituti Fisioterapici Ospitalieri (Ifo) - Istituto Regina Elena Per Lo Studio E La Cura Dei Tumori
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 Istituti Fisioterapici Ospitalieri (Ifo) - Istituto Regina Elena Per Lo Studio E La Cura Dei Tumori filed Critical Istituti Fisioterapici Ospitalieri (Ifo) - Istituto Regina Elena Per Lo Studio E La Cura Dei Tumori
Publication of WO2016038534A1 publication Critical patent/WO2016038534A1/fr

Links

Classifications

    • 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
    • A61K47/69Medicinal 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 conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit
    • A61K47/6949Medicinal 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 conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit inclusion complexes, e.g. clathrates, cavitates or fullerenes
    • A61K47/6951Medicinal 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 conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit inclusion complexes, e.g. clathrates, cavitates or fullerenes using cyclodextrin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents

Definitions

  • the present invention relates to new fenretinide-carrier molecules soluble-in- water complexes, their medical use, pharmaceutical compositions comprising the same, processes for the preparation of said complexes and uses of said complexes.
  • Fenretinide also known as N-(4-hydroxyphenil)retinamide or 4-HPR is a synthetic retinoid that induces apoptosis of cancer cells and acts synergically with chemotherapeutic drugs.
  • This molecule has emerged as a promising anticancer agent as well as a chemo-preventive agent based on numerous in vitro and animal studies and clinical trials that showed its high anticancer activity and low toxicity profile.
  • Fenretinide has exceptional anticancer activity on a wide variety of experimental models belonging to different types of cancer in both the experimental and clinical settings.
  • Fenretinide destroys cancer cells via an efficient induction of apoptosis and its effect is selective to tumour cells.
  • a major limitation to its clinical use is represented by its low bioavailability due to its very poor solubility in water and consequently in the aqueous body fluids. To date this limitation has prevented its clinical use because appropriate plasma levels of the drug cannot be attained in patients by the available drug formulations.
  • Many attempts have been made to prepare fenretinide formulations suitable to raise its bioavailability at least to the minimum levels suitable to elicit a therapeutic response.
  • fenretinide consisting of soft gelatine capsules containing fenretinide (100 mg) in corn oil and polysorbate, available at the National Cancer Institute (NCI).
  • NCI National Cancer Institute
  • Clinical l-lil evaluations of this formulation have shown good tolerability even at high doses (2,450 mg/m 2 /d) and only minimal systemic toxicity limited to reduced night vision which was reversed by discontinuation of the drug.
  • the therapeutic efficacy remained inadequate even after multiple and protracted administrations as the plasma concentrations of fenretinide continued to lay below the minimum threshold necessary for the onset of the antitumor activity.
  • amphiphilic branched PEGs linked to alkyl chains have been prepared and used as micelling agents for fenretinide.
  • the micelles improved the aqueous solubility of Fenretinide thus enhancing its cytotoxicity towards tumour cells(Orienti I et al Nanomedicine 2012;8:880-90 ).
  • amphiphilic dextrins linked to alkyl chains have been prepared and used as micelling agents for fenretinide.
  • the micelles improved the aqueous solubility of fenretinide thus improving its antitumor activity both in vitro (Carosio Ret al J Pharm Pharmacol. 2012 Feb;64(2):228-36 ) and in vivo (Orienti I et al 2009 Oct;16(7):389-98).
  • Another formulation based on the conjugation of fenretinide with hydrophilic polyvinyl alcohol provides micelles increasing the aqueous solubility of fenretinide thus improving its antitumor activity both in vitro and in vivo (Orienti I et al Biomacromolecules 2007 Oct;8(10):3258-62)
  • Described in the art is also the preparation of amphiphilic polyvinyl alcohol by substitution of the polymer with o!eyl and PEG chains to obtain micelles for the encapsulation of fenretinide (Orienti et al Biomacromolecules. 2006 Nov;7(11 ):3157-63).
  • albumin as a carrier for fenretinide
  • Another formulation using albumin as a carrier for fenretinide has been described in the art to improve the drug bioavailability limitedly to tumours expressing a receptor binding albumin (Durante et al Oncotarget. 2014 Jul 15:5(13):4811-20.)Also liposomes have been tried as a route to improve the bioavailability of fenretinide.
  • the methods currently used to raise the aqueous solubility of hydrophobic drugs mainly rely on the use of solubilizing excipients such as non-polymeric or polymeric amphiphiies , organic solvents and cyclodextrins.
  • Cyclodextrins are cyclic oligosaccharides prepared by the enzymatic conversion of starch. They are composed of 6, 7 or 8a(1-4)-Sinked glucose units which form a hydrophobic cavity and a hydrophilic shell. The hydrophi!ic shell makes them highly soluble in water.
  • the ⁇ -, ⁇ - or ⁇ -cyclodextrins differ in the diameter of their hydrophobic cavity. They have the ability to interact with poorly water-soluble drugs increasing the drug water solubility. The mechanism for this solubilisation is based on the formation of non-covalent dynamic inclusion complexes between the hydrophobic cavity of the cyclodextrins and the hydrophobic portions of the drug molecule.
  • the hydrophilic external part of the cyclodextrin molecule functions as a shield to the hydrophobic portions of the complex.
  • the self- association of cyclodextrin molecules not involved in the inclusion complexation also contribute to the final drug solubilisation by a surfactant-like effect also called 'supramolecular complexation'(Loftsson T., Masson M., Brewster M.E. J. Pharm. Sci. 2004;93:1091 -1099).
  • Cyclodextrins are suitable for complexation of many hydrophobic drugs with appropriate size to fit the hydrophobic cavity and are used as water-soluble drug carriers in pharmaceutical formulations as they improve the drug's aqueous solubility and the drug chemical stability (Brewster ME, Loftsson T. Adv Drug Deliv Rev. 2007;59:645-66)
  • the complex in solution undergoes a dynamic equilibrium of dissociation which allows the free drug to be released from the complex and absorbed into the cells when an appropriate concentration gradient is established through the cell membrane due to the drug partition to the membrane bilayer (Rajewski RA, Stella VJ. Pharmaceutical applications of cyclodextrins. 2. In vivo drug delivery. J Pharm Sci. 1996;85: 1 142-69).
  • the cyclodextrins are endowed with favourable biodistribution and reduced toxicity also after repeated administrations. Moreover, opposite to amphiphiies and organic solvents the cyclodextrins do not allow drug precipitation following dilution in the body fluids as both the inclusion complexation and the supramolecular complexation shield the drug molecules from aggregation.
  • inclusion complexation may be studied by 1 H-NMR spectroscopy by measuring the difference in the proton chemical shifts between the free drug and the drug inserted into the cyciodextrin cavity.
  • Proton chemical shift changes have been identified as a marker for the formation of inclusion complexes.
  • the chemical shift changes of the protons from inclusion complexes drug-cyclodextrins may be either downfield or up field changes depending on the molecular structure of the drug, the cyciodextrin type and the steric conformation of the drug molecule inside the cyciodextrin cavity(Francisco et al 2012: Ventura CA et al Mol Biomol Spectrosc. 2014).
  • Cyclodextrins hence, can be considered as good excipients to improve the bioavailability of hydrophobic drugs because their stability and safety has yet been assessed by their protracted use in several marketed products for parenteral, topical and oral administration.
  • both drug and cyciodextrin have to be moleculariy dispersed in water (i.e. solubifized in water) at least at a minimum extent to allow the spontaneous inclusion of the drug molecule into the hydrophobic cavity of the cyciodextrin. This is a thermodynamically driven process due to the mutual tendency of both the drug and the hydrophobic cavity of cyciodextrin to escape water.
  • the inventors have raised the aqueous solubility of fenretinide and have subsequently accomplished its complexation with various carrier molecules.
  • the inventors have raised the solubility to suitable levels for complexation with various known carrier molecules. This result was obtained by submitting for the first time in the art the fenretinide molecule to salification and by complexing the fenretinide salts with various carrier molecules, thereby obtaining complexes with higher water solubility that provide an improved bioavailability of the fenretinide drug.
  • the inventors have also provided inclusion complexes between fenretinide and cyclodextrins that allow for a very high solubility of fenretinide in water and release of the complexed drug only in the presence of an absorbing phase (such as, by way of example, a tumour in vivo) and processes for the preparation of said complexes.
  • inclusion complexes are evidenced by the presence of chemical shift changes in the 1 H-NMR spectrum of the complex corresponding to protons H-8, H-3and H-5 of the complexed fenretinide with respect to the 1 H-NMR spectrum of fenretinide salts.
  • Chemical shift changes in the 1 H-NMR spectrum of drug-cyclodextrins complexes vs. the free drug 1 H-NMR spectrum is reported in the art as a characterising feature of drug-cyclodextrins inclusion complexes.
  • a1 represents complexes characterised by the presence of chemical shift changes in the 1 H-NMR spectrum corresponding to protons H-8, H-5 andH-3,of the compiexed fenretinide with respect to the 1 H-NMR spectrum of fenretinide salts (the shift indicating the presence of inclusion complexes),whereas b c and d represent complexes that are characterised by the absence of said chemical shift changes i.e. by supermolecular complexes in absence of detectable inclusion complexes.
  • Figure 4 of the invention also shows that the release of fenretinide from the complexes of the invention (i.e. the complexes characterised by the presence of the chemical shift changes in the ⁇ -N R spectrum as described above) takes place only in the presence of an adsorbing phase while it is suppressed in a simple aqueous environment. This is clearly advantageous when for a targeted use of the drug.
  • Fenretinide-carrier molecules also Fenrefinide-CM herein
  • complexes characterised by the fact that said complexes comprise fenretinide salts as such and uses thereof as medicaments;
  • a pharmaceutical composition comprising the fenretinide-carrier molecules complexes of the invention and a pharmaceutically acceptable carrier
  • CM carrier molecule
  • CM carrier molecule
  • a medical treatment comprising the administration of the complexes of the invention to patients in need thereof wherein said treatment can be a treatment of a tumour or a prevention treatment of a tumour.
  • Figure 1 represents the 1 H-NMR spectrum of fenretinide (600 MHz; DMSO-d 6 )
  • Figure 2 represents the 1 H-N R spectrum of fenretinide sodium salt (600 MHz; DMSO-d 6 ).
  • Figure 3 represents the ⁇ H-NMR spectrum of the fenretinide-(2-Hydroxypropyl)- ⁇ -cyclodextrin complex (HPBCD) obtained by the physical mixture method of the invention (600 MHz; D 2 0).
  • Figure 4 represents the release of fenretinide from the fenretinide-HPBCD complex obtained by the physical mixture method in an aqueous environment (PBS) and in the presence of an absorbing phase.
  • PBS aqueous environment
  • Figure 5 shows the in vitro effect of pure fenretinide (dissolved in EtOH) and fenretinide-HPBCD compiex on the viability of cancer cells and CSC.
  • Figure 6 shows the effect of the fenretinide-HPBCD complex on commercial cancer celi lines.
  • Cells were treated with doses of fenretinide:HPBCD ranging from 1 to 50 mM and cell death was assessed after 48 hours with the Ceil Titer GLO assay (Promega).
  • Figure 7 shows the effect of the fenretinide-HPBCD complex on CSC lines derived from solid tumours.
  • CSC cultures from 9 lines of colorectal cancer (colon), 4 lines of glioblastoma, 4 lines of sarcoma, 4 lines of breast cancer (breast), 3 lines of melanoma and 2 !ines of ovarian cancer (ovarian) were treated for 48 hours with the fenretinide-HPBCD complex at the indicated doses and celi viability was assessed with the Ceii Titer GLO assay (Promega).
  • Figure 8 shows the effect of the fenretinide-HPBCD complex on CSC Sines derived from non-small eel! lung cancer
  • Panel A Spheroid cultures from two lines of adenocarcinoma (AC), Panel B three lines of squamous ceil carcinoma (SCC) and Panel C one line of large cell neuroendocrine carcinoma (LCNEC) were treated for 48 hours with the fenretinide-HPBCD compiex at the indicated doses and DCi viability was assessed with the Cell Titer GLO assay (Promega).
  • Figure 9 shows the effect of the fenretinide-HPBCD compiex on the lung cancer stem cell line LCSC 136 at different times of incubation (A) and in combination with chemotherapeutic drugs (B).
  • Figure 10 shows that the fenretinide-HPBCD complex significantly inhibits tumour growth in mice bearing lung CSC-derived tumour xenografts.
  • Figure 12 shows that the liposomal fenretinide formulation administered by oral route significantly reduces tumour growth in mice bearing colon CSC-derived tumour xenografts.
  • the present invention hence provides complexes between fenretinide salts and carrier molecules (herein also fenretinide-CM complexes), wherein said complexes comprise fenretinide salts.
  • fenretinide-CM complexes includes also complexes wherein the fenretinide salt is loaded into a carrier system such as liposomes, micelles, nanoparticles, supramolecular assemblies and ion-pairs.
  • the fenretinide-CM complexes can be in the form of fenretinide- cyclodextrins complexes, liposomes comprising fenretinide salts, micelles comprising fenretinide salts, nanoparticles comprising fenretinide salts, supramolecular assemlblies comprising fenretinide salts, ion pairs comprising fenretinide salts.
  • the complexes are fenretinide-cyclodextrins complexes, they comprise also inclusion complexes fenretinide-cyclodextrins. It is to be understood that in all part of this description, the term fenretinide-cyclodextrins complexes implies complexes formed between cyclodextrins and a fenretinide salt.
  • the fenretinide-cyclodextrins complexes of the invention are characterised by the fact that fenretinide is complexed in the form of a salt and by chemical shift changes in the 1 H-NMR spectrum (600 MHz; D20) of said complexes corresponding to protons H-8 H-3 and H-5 of the complexed fenretinide with respect to the 1 H-NMR spectrum of fenretinide salts as depicted in figures 2 and 3.
  • the fenretinide-cyclodextrins complexes of the invention are characterised by the fact that fenretinide is complexed in the form of a salt and by chemical shifts in the 1 H-NMR spectrum corresponding to protons H-8 , H-3 and H-5 of the complexed fenretinide that are different from the chemical shifts in the 1 H-NMR spectrum corresponding to protons H-8 , H-3 and H-5 of fenretinide salts as such.
  • the chemical shift changes according to the invention are significant shift changes, wherein the term significant, in the present description, when referring to the 1 H-NMR spectrum implies a shift change from at least 0.1 ppm to more than 1.0 ppm.
  • the aqueous solubility of fenretinide from the fenretinide-cyciodextrins complexes of the invention ranges from 0.42 to 3.27 mg/ml at 25°C as reported in table 1 above and further discussed in other parts of this description.
  • the release of fenretinide from the fenretinide- cyciodextrins complexes of the invention is very efficient in an aqueous environment in the presence of an absorbing phase being 56% the fractional amount of drug released at 12 h and 70% at 24 h while no release takes place in a simple aqueous environment as reported in fig 4 and further discussed in other parts of this description.
  • the cyclodextrins forming complexes with the fenretinide salts according to the present invention are selected from monomers, dimers or polymers of substituted or not substituted ⁇ -, ⁇ - and/or ⁇ -cyclodextrins or pharmaceutically acceptable salts thereof, in particular, said substituted or not substituted ⁇ -, ⁇ - and/or y-cyclodextrins or pharmaceutically acceptable salts thereof are selected from: (2-Hydroxypropyl)- -cyclodextrin; Heptakss(6-0-sulfo)-P-cyclodextrin; (2- Hydroxyethyl)-p-cyclodextrin; Methyl- -cyclodextrin; (2-Hydroxypropyl) ⁇ - cyclodextrin; (2-Hydroxypropyl)-Y-cyclodextrin; ( 2-Hyd roxypropyl )-ct-cyclo
  • said dimers or polymers of substituted or not substituted ⁇ -, ⁇ - and/or ⁇ -cyclodextrins can be selected from Carboxymethyl-a-cyclodextrin polymer; ⁇ -Cyclodextrin polymer, ⁇ -Cyclodextrin polymer.
  • the cyclodextrin used in the complexes of the present invention is (2-Hydroxypropyl)- -cyclodextrin dextrin (HPBCD).
  • the fenretinide-CM complexes are complexes formed by a fenretinide salt with one or more of the following molecules: phospholipids, sphingolipids, cationic lipids, amphiphilic polymers, amphiphilic macromolecules, surfactants.
  • the fenretinide-CM is a liposome comprising fenretinide salts.
  • suitable phospholipids is represented by Soybean phosphatidylcholine (SPC), Hydrogenated soybean phosphatidylcholine
  • HSPC Egg phosphatidylcholine
  • EPC Dimyristoyl phosphatidylcholine
  • DPPC Dipalmitoyi phosphatidylcholine
  • DOPC Dioleoyl phosphatidylcholine
  • DSPE Distearoyl phosphatidylcholine
  • DSPC Distearoyl phosphatidylethanolamine
  • DMPG Dimyristoyl phosphatidylglycerol
  • DPPG Dipalmitoyi phosphatidylglycerol
  • DOPG Dioleoyl phosphatidylglycerol
  • DOPG Distearoyl phosphatidylglycerol
  • DSPG Distearoyl phosphatidylserine
  • DMPE Dimyristoyl phosphatidylethanolamine
  • DPPE Dipalmitoyi phosphatidylethanolamine
  • DOPE Dioleoyl phosphatidyl
  • phospholipids carrying cationic residues such as: Soybean phosphatidylcholine (SPC), Hydrogenated soybean phosphatidylcholine (HSPC), Egg phosphatidylcholine (EPC), Dimyristoyl phosphatidylcholine (DMPC), Dipalmitoyi phosphatidylcholine (DPPC), Dioleoyl phosphatidylcholine (DOPC), Distearoyl phosphatidylcholine (DSPE), Distearoyl phosphatidylethanolarnine (DSPC), Dimyristoyl phosphatidylethanolamine (DMPE), Dipalmitoyi phosphatidylethanolamine (DPPE), Dioleoyl phosphatidylethanolamine (DOPE) or mixtures thereof may be used and they may form ion pairs with the fenretinide salt.
  • SPC Soybean phosphatidylcholine
  • HPC Hydrogenated soybean phosphatidy
  • phospholipids may be used also in their pegylated form preferred examples are Distearoyl phosphatidylethanoiamine- Polyethyleneglycol 2000 (DSPE-PEG2000), Dimyristoyl phosphatidylethanolamine-N-poly(ethylene glycol)-5000 (DMPE-PEG5000).
  • DSPE-PEG2000 Distearoyl phosphatidylethanoiamine- Polyethyleneglycol 2000
  • DMPE-PEG5000 Dimyristoyl phosphatidylethanolamine-N-poly(ethylene glycol)-5000
  • sphingolipids according to the invention is represented by Sphingomyelin, Sphingosyl Phosphoethanolamine, Sphingosyl Phosphoinositol, Sphingosylphosphorylchoiine, Sphingosine, Ceramides, Glycosphingolipids, Gangliosides or mixtures thereof, in an embodiment Sphingolipids carrying cationic residues such as: Sphingomyelin, Sphingosyl Phosphoethanolamine, Sphingosylphosphorylchoiine, may be used, and they may form ion pairs with the fenretinide salt
  • a non-limiting example of suitable cationic lipids that may be used in the preparation of fenretinide-carrier systems of the invention is represented by: (DC-Choi) 3b[N-(N',N'-dimethylaminoethane)-carbamoyl] cholesterol; (DOPE) dioleoyl phosphatidylethanolamine; (DOTAP), 1 ,2-bis(oleoyloxy)-3- (trimethylammonio)propane; (DOTMA), N-[1-(2,3-dio!eyloxy)propyl]-N,N,N,- trimethylammonium chloride; (EMPDC), 1 ,2-dimyristoyl-P-O- ethylphosphatidylcholine or mixtures thereof.
  • DC-Choi 3b[N-(N',N'-dimethylaminoethane)-carbamoyl] cholesterol
  • DOPE dioleoyl
  • the fenretinide-CM complexes in the form of lyposomes are complexes with phosphatidylcholine.
  • the fenretinide-CM is a micelle comprising fenretinide salts, or, when prepared by suitable standard methods known to the skilled person, a nanoparticle comprising fenretinide salts.
  • amphiphilic polymers and amphiphilic macromolecuies suitable for the fenretinide-carrier systems in the form of micelles or nanoparticles according to the invention are represented by: -Amphiphilic proteins or peptides, preferably serum albumin or lysozyme either pure or partially substituted with hydrophobic moieties; Hydophilic polymers/macromolecules, preferably polylysine, polyethyleneimine, chitosan partially substituted with hydrophobic moieties; -Hydrophobic polymers substituted with PEG preferably poly(ethylene glycol)-b-poly(lactic acid), poly(ethylene glycol)-b-poly(d,l-lactide); poly(ethylene glycol)-b-poly(amino acid) ; poly(e-caprolactone)-b-poly(ethylene glycol) or mixtures thereof.
  • -Amphiphilic proteins or peptides preferably serum albumin or lysozyme either pure or partially substitute
  • Suitable surfactants are represented by Alkyl ammonium type, alkyl PEG type and the like, and the example of suitably cationic lipids is the one already provided above, i.e. DC-Choi) 3b[N-(N',N'-dimethylaminoethane)-carbamoyl] cholesterol; (DOPE) dioleoyl phosphatidylethanolamine; (DOTAP), 1 ,2- bis(oleoyloxy)-3-(trimethylammonio)propane; (DOTMA), N-[1-(2,3- dioleyloxy)propyl]-N,N,N,-trimethylammonium chloride; (EMPDC), 1 ,2- dimyristoyl-P-O-ethylphosphatidylcholine or mixtures thereof.
  • the fenretinide salt when the fenretinide salt is complexed with any of the cyclodextrins, phospholipids, sphingolipids, cationic lipids, amphipilic polymers, amphiphilic macromolecules, surfactants or a mixture thereof, the fenretinide-CM is a supramolecular assembly comprising fenretinide salts.
  • the fenretinide-CM when the fenretinide salt is complexed with any one of the above mentioned carrier molecule in cationic form, the fenretinide-CM is in the form of ion pairs comprising fenretinide salts where the counter ion of the salt is represented by the carrier molecule in its cationic form
  • Suitable carrier molecules in cationic form are hence any of the above mentioned carrier molecules in cationic form such as, by way of example, cationic phospholipids, cationic sphingolipids, cationic lipids, cationic surfactants, cationic amhiphilic polymers, cationic amphiphilic macromolecules
  • Non-limiting examples of said cationic carrier molecules are represented by: Soybean phosphatidylcholine (SPC), Hydrogenated soybean phosphatidylcholine (HSPC), Egg phosphatidylcholine (EPC), Dimyristoyl phosphatidylcholine (DMPC), Dipalmitoyl phosphatidylcholine (DPPC), Dioleoyl phosphatidylcholine (DOPC), DistearoyI phosphatidylcholine (DSPE), DistearoyI phosphatidylethanolamine (DSPC), Dimyristoyl phosphatidylethanolamine (DMPE), Dipalmitoyl phosphatidylethanolamine (DPPE), Dioleoyl phosphatidylethanolamine (DOPE), Sphingomyelin, Sphingosyl Phosphoethanolamine, Sphingosylphosphorylcholine,
  • DC-Choi 3b[N-(N',N'-dimethylaminoethane)-carbamoyl] cholesterol;
  • DOPE dioleoyl phosphatidylethanolamine;
  • DOTAP 1,2-bis(oleoyloxy)-3- (trimethylammonio)propane;
  • DOTMA 1,2-bis(oleoyloxy)-3- (trimethylammonio)propane;
  • DOTMA 1, N-[1-(2,3-dioleyloxy)propyl]-N,N,N,- trimetby!ammonium chloride;
  • E PDC 1 ,2-dimyristoyi-P-O- eihylphosphatidy!choline or mixtures thereof.
  • any pharmaceutically acceptable fenretinide salt can be used.
  • the salt can be a fenretinide K salt, a fenretinide Na salt, a fenretinide ammonium salt.
  • said ammonium can be selected from R-i RaRsFijN wherein each of R t R 2l R3 and R 4 is independently selected from the group consisting of hydrogen, a C C 2 o aikyl group, a C 3 -C 8 cyc!oalkyl group, a phenyl, a morpho!ino, and a pyridine group.
  • ammonium according to the invention can also be cyclic such as aziridines, azetidines, pyrrolidines, piperidines, piperazines, morpholines or derivatives thereof.
  • ammonium may be phosphatidylcholine, sphingomyelin or a derivative thereof, thiamine, carnitine, choline and quaternized aminoacids or derivatives thereof.
  • the complex is a complex between (2-Hydroxypropyl) ⁇ -cyclodextrin dextrin (HPBCD) and a fenretinide K or Na salt.
  • HPBCD (2-Hydroxypropyl) ⁇ -cyclodextrin dextrin
  • fenretinide K or Na salt a complex between (2-Hydroxypropyl) ⁇ -cyclodextrin dextrin (HPBCD) and a fenretinide K or Na salt.
  • the complexes of the present invention are of particular interest for the therapeutic targeting of cancer ceils, including cancer stem cells, and hence for the treatment or prevention of solid and/or haematological tumours.
  • tumours A non-limiting example of said tumours is represented by:
  • gynaecological cancers skin cancers, brain and neurological cancers, urologic cancers, hepatobiliopancreatic cancers, neuroendocrine cancers, endocrine cancers, adenocarcinomas, digestive-tract cancers, respiratory-tract cancers, hematologic cancers, mesothelioma, embryonic carcinoma, head-and-neck cancer, sarcomas.
  • the term urologic cancer include but are not limited to cancers of the bladder, kidney, prostate and testicles; the term hepatobiliopancreatic cancers include liver, gallbladder, bile ducts, and pancreas.
  • the respiratory-tract cancers include but are not limited to lung cancers, laryngeal cancers.
  • digestive-tract cancers include but are not limited to pharynx and oesophagus cancers and gastrointestinal cancers such as, by way of example, stomach, pancreas, small intestine, large intestine, colorectal, rectum, gastric, and anus cancers and gastrointestinal stromal tumour.
  • gynaecological cancers include but are not limited to breast, ovarian, uterine, cervical, vaginal and vulvar cancers.
  • cancers include but are not limited to adult and paediatric non limiting examples thereof are leukaemias such as acute and chronic leukaemias, hairy ceil leukaemia, lymphomas including Hodgkin and non-Hodgkin lymphomas, mycosis fungoide, CNS lymphoma, Sezary syndrome, cutaneous lymphomas, myelomas, mye!odysp!astic and myeloproliferative neoplasms, Waldenstrom macrogiobulinemia.
  • leukaemias such as acute and chronic leukaemias, hairy ceil leukaemia, lymphomas including Hodgkin and non-Hodgkin lymphomas, mycosis fungoide, CNS lymphoma, Sezary syndrome, cutaneous lymphomas, myelomas, mye!odysp!astic and myeloproliferative neoplasms, Waldenstrom macrogiobulinemia.
  • brain and neurological cancers include but are not limited to adult and paediatric neurological tumours, non-limiting examples thereof are gliomas including glioblastoma, medullob!astomas, oligodendroglioma, neurinomas, chordomas, ependymoma, neuroblastoma and peripheral nerves cancers.
  • skin cancers include but are not limited to, basal cell cancer (BCC), squamous cell cancer (SCC), melanoma and Merkel's cancer.
  • endocrine cancers include but are not limited to andenocortical carcinoma, thyroid cancer, parathyroid cancer, pituitary tumours.
  • neuroendocrine cancers are cancers that affect the neuroendocrine system.
  • the neuroendocrine system makes chemical messengers called hormones, which regulate the workings of different organs in the body.
  • Neuroendocrine cells are spread throughout the body in organs such as the stomach, bowels and lungs.
  • Cancerous NETs are classified according to where the cancer started (where the primary tumour is) in the body.
  • Non limiting examples of neuroendocrine cancers according to the present description include: small bowel NETs, large bowel NETs, appendiceal NETs , pancreatic ETs, gastric NETs, lung NETs.
  • NETs are found in other areas, including the liver, gallbladder, bile ducts, kidneys, ovaries or testicles.
  • adenocarcinoma is a type of cancer that forms in mucus-secreting glands throughout the body.
  • sarcomas arise from transformed cells of mesenchymal origin thus malignant tumours made of cancerous bone, cartilage, fat, muscle, vascular, or hematopoietic tissues are, by definition, considered sarcomas.
  • Non limiting examples include: osteosarcomas, chondrosarcomas, liposarcomas, leiomyosarcomas and rabdomyiosaromas.
  • the fenretinide-cyclodextrins complexes herein described and claimed can be used in cancer patients either alone or in alternation or in combination with chemotherapeutic agents commonly used for each tumour type, and/or small molecules commonly used for each tumour type and/or biological agents commonly used in biological therapies for each tumour type, i.e. in alternation or in combination with further chemotherapeutic agents and/or anti-tumour biological agents and/or anti-tumour small molecules.
  • Non limiting examples of said chemotherapeutic agents are selected in the group comprising or consisting of cisplatin, gemcitabine, docetaxel, cytosine arabinoside, bleomycin, busulfan, capecitabine, carboplatin cyclophosphamide, daunorubicin, doxorubicin, epirubicin, etoposide, fludarabine 5-fluorouracil, irinotecan, idarubicin, ifosfamide, hydroxyurea, lenalidomide lomustine, melphalan, mitomycin C, mitoxantrone, oxaliplatin, paclitaxel, temozolomide, thalidomide, topotecan, vinblastine, vincristine, vinorelbine, vindesine.
  • the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising the fenretinide-cyclodextrins complexes according to the present description and a pharmaceutically acceptable carrier.
  • small molecules for cancer treatment are molecules commonly used for the treatment of cancer such as targeted pathway inhibitors (including kinase inhibitors, cell cycle inhibitors/checkpoint inhibitors, Bcl2 family inhibitors, mitochondrial inhibitors, metabolic inhibitors, apoptosis and/or autophagy inducers, histone deacetylase (HDAC) inhibitors, gamma-secretase inhibitors, differentiative agents and multipurpose inhibitors); examples include bortezomib (small molecule proteasome inhibitor), imatinib (small molecule tyrosine kinase inhibitor), seiiciclib (small molecule cyclin-dependent kinase inhibitor), palbociclib (CD-4/6 inhibitor), AG-221 (isocitrate dehydrogenase 2 inhibitor), ABT-263/ABT-199 (Bcl2 family proteins inhibitors), valproic acid (HDAC inhibitor), thalidomide and derivatives (lenalidomide, pomalidomide
  • targeted pathway inhibitors including kin
  • biological therapy involves the use of biological agents such as: living organisms, substances derived from living organisms, or laboratory-produced versions of such substances to treat disease.
  • Some biological therapies for cancer use vaccines or bacteria to stimulate the body's immune system to act against cancer cells.
  • biological therapies include “immunotherapy", “biological response modifier therapy” or anti-angiogenic therapy that do not target cancer cells directly or other therapies, such as antibodies or segments of genetic material (RNA or DNA) that do target cancer cells directly.
  • Biological therapies that interfere with specific molecules involved in tumour growth and progression are also referred to as targeted biological therapies.
  • Non limiting examples of biological agents for cancer therapy are: nucleic acids, miRNAs, antibodies (or antibody fragments) such as cetuximab, trastuzumab, bevacizumab, cytokines, proteins or peptides.
  • the fenretinide-cyclodextrins complexes can be used in alternation or in combination with anti-tumour biological agents known in the art and/or chemotherapeutic agents known in the art and/or anti- tumour small-molecules commonly known in the art.
  • the pharmaceutical composition comprising the fenretinide- cyclodextrins complexes herein described are in a form for oral, intravenous, intramuscular, intradermal, subcutaneous, intraperitoneal, intrapulmonary, intranasal, rectal, transdermal, topical administration
  • the pharmaceutical composition of the invention can be formulated in the form of capsule; tablet; lozenge; granule; powder; solution; suspension; emulsion; syrup; elixir; hard gelatine; soft gelatine; injectable suspension, emulsion, solution; suppository; cream; gel; spray; ointment; pomade; paste; emulsion oil-in-water; water-in-oil emulsion; oil-in-gel emulsion; gel-in-oil emulsion.
  • compositions of the present invention are suitable for use in the treatment or prevention of tumours as described above with reference to the complexes.
  • the pharmaceutical composition of the present invention is of particular interest for the therapeutic targeting of cancer cells, including cancer stem cells, and hence for the treatment or prevention of solid and/or hematological tumours.
  • tumours A non-limiting example of said tumours is represented by
  • gynaecological cancers skin cancers, brain and neurological cancers, urologic cancers, hepatobiliopancreatic cancers, neuroendocrine cancers, endocrine cancers, adenocarcinomas, digestive-tract cancers, respiratory-tract cancers, hematologic cancers, mesothelioma, embryonic carcinoma, head-and-neck cancer, sarcomas.
  • compositions herein described and claimed can be used in cancer patients either alone or in alternation or in combination with chemotherapeutic agents commonly used for each tumour type, and/or small molecules commonly used for each tumour type and/or biological agents commonly used in biological therapies for each tumour type.
  • chemotherapeutic agents, biological agents and small molecules are as defined above.
  • the present invention provides a process for the preparation of fenretinide-carrier molecule complexes comprising the following steps:
  • CM carrier molecule
  • each of the following complexes comprising fenretinide salts may be obtained: fenretinide-cyclodextrins complexes, liposomes, micelles, nanoparticles, supramolecular assemblies, ion pairs.
  • cyclodextrin-fenretinide complex when a cyclodextrin-fenretinide complex is desired, one or more of the exemplified cyclodextrins are used as carrier molecule in step a. of the method above.
  • fenretinide-cyciodextrins complexes comprising fenretinide salts, as described above, i.e.
  • complexes characterised by the fact that fenretinide is complexed in the form of a salt and by chemical shift changes in the H-NMR spectrum corresponding to protonsH-8 , H-3 and H-5 of the complexed fenretinide with respect to the 1 H-NMR spectrum of fenretinide salts as such; further steps may be added to the method above that allow a higher purification and a better yield of fenretinide-cyciodextrins complexes.
  • the process as previously defined may comprise additional steps c, d. and, optionally e. as defined below: submitting the suspension thus obtained to stirring or sonication and incubating said suspension until the formation of a finely dispersed solid phase (indicative of the maximum conversion of the solubifized salt into the complex and saturation of the aqueous phase with the complex)
  • the process may further comprise step f. dissolving the lyophilized complexes obtained in e. in water or aqueous buffer or other suitable aqueous solvent and submitting the solution to column chromatography, collecting the eluate and optionally lyophilizing the eluate collected.
  • the molar ratio salt yclodextrin can be, by way of example, from 1 :10 to 1 :20 and the mixture can be suspended, e.g. at a concentration from 0.05 to 1g/ml.
  • Step g. can be carried out by dissolving the powder obtained in e. in water at a concentration ranging from 10 to 500 mg/ml, such as, e.g. from 50 to 200 mg/ml preferably about 100 mg/ml.
  • the solution obtained can then be optionally applied to a chromatographic column such as a sephadex G-50 column or an analogous column, known to the skilled person, previously equilibrated with water.
  • the column is eluted by water.
  • the fractions containing the complex are pooled and lyophilized to a solid powder.
  • the process sha!i comprise steps a, and b. as previously described herein and will preferably comprise also steps c. and d. as described above.
  • the process may further optionally comprise, in the foi!owing order, one or more of steps e., f, and g. as also described above.
  • steps a. and b. of the process of the invention may be followed, for a better yield of the desired complex, by subsequent step c 1 ., and optionally subsequent step d'. and optionally subsequent step e'., and optionally subsequent step f .
  • the fenretinide salt and the seiecied carrier molecule in b. may be further admixed with a dispersion promoter such as tributyrine, ethyl butyrate, butyric acid and/or ethanol.
  • a dispersion promoter such as tributyrine, ethyl butyrate, butyric acid and/or ethanol.
  • the chromatographic column can be a sephadex G-50 column or an analogous column known to the skilled person.
  • the carrier molecule, dispersion promoters and fenretinide salt can be in respective molar ratios ranging from 3 : 0.3 : 1 to 3 : 2.0 : 1 preferably to 3 : 1.5 : 1.
  • the dispersion promoter can be a mixture of tributyrine and ethanol, and the carrier molecule, tributyrine, ethanol and fenretinide salt can be in molar ratios ranging from 3 : 0.3 :10 : 1 to 3 : 2.Q : 20 : 1 preferably to 3 : 1.5 : 15 : 1 respectively.
  • step b. can be carried out as follows:
  • step b. a mixture of phospholipids or pegylated phospholipids, one or more dispersion promoters and a fenretinide salt (preferably phosphatidylcholine : tributyrine : ethanol: fenretinide potassium salt) in molar ratios ranging from 3 : 0.3 :10 : 1 to 3 : 2.0 : 20 : 1 preferably to 3 : 1.5 :15 : 1 respectively is dispersed in an aqueous buffer pH 7.0 and stirred to homogeneity at 25 °C in the dark until formation of the complex of the invention as detected by optical microscopy.
  • step b. can be carried out as follows:
  • a mixture of amphiphilic polymers or/and macromolecules or/and surfactants one or more dispersion promoters and a fenretinide salt preferably human serum albumin : tributyrine : ethanol: fenretinide potassium salt
  • a fenretinide salt preferably human serum albumin : tributyrine : ethanol: fenretinide potassium salt
  • 3 : 0.3 :10 : 1 to 3 : 2.0 : 20 : 1 preferably to 3 : 1.5 :15 : 1 respectively is dispersed in an aqueous buffer pH 7.0 and stirred to homogeneity at 25 °C in the dark until formation of the complex of the invention as detected by optica! microscopy.
  • a process for the preparation of fenretinide-carrier molecule complexes comprising the following steps:
  • fenretinide salt with one or more carrier molecule (CM) selected from: cyclodextrins, phospholipids, sphingolipids, cationic lipids, amphiphilic polymers, amphiphilic macromolecules, surfactants in a molar ratio salt:carrier molecule of from 1 :1 to 1 :100 to homogeneity and subsequently homogeneusly suspending the physical mixture thus obtained in ethanol, methanol, tetrahydrofuran, chloroform or a mixture of them at a concentration of from 0.05 to 1g/ml;
  • CM carrier molecule
  • the fenretinide salt and the selected carrier molecule in b. may be further admixed with a dispersion promoter such as tributyrine, ethyl butyrate, butyric acid and/or ethanol.
  • a dispersion promoter such as tributyrine, ethyl butyrate, butyric acid and/or ethanol.
  • the chromatographic column can be a sephadex G-50 column or an analogous column known to the skilled person.
  • the carrier molecule, dispersion promoters and fenretinide salt can be in respective molar ratios ranging from 3 : 0.3 : 1 to 3 : 2.0 : 1 preferably to 3 : 1.5 : 1.
  • step b for the preparation of liposomes and/or supramolecular aggregates, step b". can be carried out as follows:
  • a mixture of phospholipids or pegylated phospholipids, a dispersion promoter and a fenretinide salt (preferably phosphatidylcholine : tributyrine : fenretinide potassium salt) in molar ratios ranging from 3 : 0.3 : 1 to 3 : 2.0 : 1 preferably to 3 : 1.5 : 1 respectively is dissolved in an organic solvent preferably ethanol.
  • the organic solvent is evaporated by a rotary evaporator until the formation of a thin lipid film which is subsequently hyd rated by an aqueous buffer pH 7.0.
  • the mixture is then passed through a 200 nm polycarbonate membrane to obtain the fenretinide-CM complexes as detected by optical microscopy.
  • step b can be carried out as follows:
  • a mixture of amphiphilic polymers or/and macromolecules or/and surfactants , a dispersion promoter and a fenretinide salt preferably human serum albumin : tributyrine : fenretinide potassium salt
  • a fenretinide salt preferably human serum albumin : tributyrine : fenretinide potassium salt
  • an organic solvent preferably ethanol
  • step a i.e. the salification of fenretinide
  • step a can be carried out in several ways.
  • the invention here provides various examples for the preparation of fenretinide salts. Starting from these examples, the skilled person can adapt or modify according to general chemistry knowledge in order to provide further preparations of fenretinide salts without use of inventive skill.
  • step a. can be carried out by grinding a stoichiometric mixture of fenretinide with an alkali metal hydroxide until development of a red-orange colour indicative of formation of a fenretinide monohydrate salt.
  • the alkali metal hydroxide can be, by way of example, KOH or NaOH.
  • step a. can be carried out by adding an alkali metal hydroxide to an alcoholic solution containing fenretinide in stoichiometric amounts and removing the solvent by evaporation to obtain the solid salt.
  • the alkali metal hydroxide can be, by way of example, KOH or NaOH, and the alcoholic solution can be ethanol.
  • the salt obtained may be optionally washed with water to eliminate traces of the base used for salification and subsequently lyophilized.
  • the solid obtained may be optionally washed with a suitable solvent such as diethyl ether to eliminate traces of the unsalted drug..
  • step a. can be carried out by mixing fenretinide with alcohol, dimethyl sulphoxide or tetrahydrofuran or acetone or acetonitriie or N-methylpirrolidone or a mixture thereof in a 1 :1 w:v fenretinide solvent ratio to obtain a knead, adding an excess of an alkali metal hydroxide as a concentrated aqueous solution to form a precipitate, submitting the precipitate thus obtained to sonication until an homogeneous dispersion of the precipitate is obtained and centrifuging to separate the solid salt.
  • the salt obtained can then be washed with water to eliminate traces of the base used for salification and subsequently lyophilized.
  • the solid obtained can be further washed with a suitable solvent such as diethyl ether to eliminate traces of the unsalted drug.
  • the alkali meta! hydroxide can be, by way of example, KOH or NaOH.
  • the alcohol into which fenretinide is mixed for the preparation of the salts can be ethanol or methanol
  • the alcoholic solution can hence be a solution in ethanol or methanol.
  • step a. can be carried out by grinding and/or kneading (depending on the physical state) a stoichiometric mixture of fenretinide with an amine or a quaternary ammonium compound until formation of a fenretinide quaternary ammonium salt.
  • step a. can be carried out by solubilizing fenretinide in alcohol or dimethyl sulphoxide or tetrahydrofuran or acetone or acetonitrile or a mixture thereof at concentrations ranging from 0.1% to 10% w:v fenretinide : solvent, adding a stoichiometric amount of an amine or a quaternary ammonium compound, submitting the mixture to stirring and/or sonication and removing the solvent by evaporation to obtain the salt.
  • the amine can be selected from R ⁇ Ra wherein each of R s R 2 and R 3 is independently selected from the group consisting of hydrogen, a C C 2 o alky! group, a C 3 -C 3 cycloalkyl group, a phenyl, a morpho!ino, and a pyridino group.
  • the amine may be cyclic amines such as aziridines, azetidines, pyrrolidines, piperidines, piperazines, morpholines or derivatives thereof; the amine can be an aminoacid or a derivative threof.
  • the quaternary ammonium compound is selected from RiR 2 R 3 R 4 wherein each of Ri, R 2> R 3 and R 4 is independently selected from the group consisting of hydrogen, a C C 2 o alkyl group, a C 3 -C 8 cycioalkyl group, a phenyl, a morpholino, and a pyridine group .
  • the ammonium may be cyclic such as aziridines, azetidines, pyrrolidines, piperidines, piperazines, morpholines or derivatives thereof, the ammonium can be phosphatidylcholine, sphingomyelin or a derivative thereof, thiamine, carnitine, choline and quaternized aminoacis or derivatives thereof;
  • the salt obtained may be optionally washed with water to eliminate traces of the base used for salification and subsequently lyophilized.
  • the solid obtained may be optionally washed with a suitable solvent such as diethyl ether to eliminate traces of the unsalted drug.
  • step a. of the process as herein described are more soluble in water than fenretinide as summarised in table 2 below reporting the aqueous solubility of some fenretinide salts of the invention at 25°C
  • Salification of fenretinide has never been reported in the literature to date neither as an intermediate step for drug complexation with molecular carriers in the forms described herein, nor as a mean to improve its aqueous solubility or the solubility of pharmacologically active derivatives such as 4-oxo-fenretinide, keto fenretinide etc.
  • fenretinide salts represent an embodiment of the invention and are innovative tools to raise fenretinide aqueous solubility with the aim to obtain drug complexation with different complexing agents including complexes by entrapment of said fenretinide salts into solubilizing systems such as liposomes, micelles, nanoparticles, supramolecular assemblies, or ion pairs.
  • solubilizing systems such as liposomes, micelles, nanoparticles, supramolecular assemblies, or ion pairs.
  • salification of fenretinide may be used as a method to improve the solubility and therefore the bioavailability of the drug.
  • fenretinide salts are here used as intermediates in the preparation of the complexes described herein in the form of other drug delivery systems such as fenretinide-cyclodextrins complexes, fenretinide-salts-comprising liposomes, micelles, nanoparticles, supramolecular assemblies, and ion pairs.
  • fenretinide salts may be used in the place of fenretinide in pharmaceutical formulations to improve the drug bioavailability and thus its therapeutic efficacy.
  • Objects of the present invention are hence the fenretinide complexes with cyclodextrins or other carrier molecules obtainable by the process herein described, pharmaceutical compositions comprising said fenretinide- cyclodextrins complexes as well as fenretinide salts obtainable as intermediates of the process herein described.
  • fenretinide salts-cyclodextrins complexes herein defined as the "physical mixture process” also indicated as process a1 or a2 that corresponds, respectively, to the process described above and claimed (steps a-e) and to the process described above and claimed also including step g, the "kneading process” also indicated as process b (described only in the examples section below), the “co-precipitation process” also indicated as process c (described only in the examples section below) and the “ co-evaporation process” also indicated as process d (described in the examples section below)and have verified that only the "physical mixture process” provided the claimed complexes, i.e. complexes that do include inclusion complexes as they are characterised by chemical shift changes in the 1 H-NMR spectrum described and claimed and that provide the highest increase of fenretinide aqueous solubility.
  • the inventive mixture process also indicated as process a1 or a2 that corresponds, respectively, to the process described above
  • the different preparative methods also provided different drug: cyclodextrin molar ratios and different solubility of fenretinide in water.
  • Table 3 summarises the fenretinide: cyclodextrin molar ratios obtained from the different preparative processes: physical mixture process (a), kneading process
  • the complex obtained by steps a-e of the process claimed is formed by inclusion complexation + supramolecular complexation where inclusion complexation provides 1 :1 m:m (drug: cyclodextrin) ratio while supramolecular complexation provides 1 : >1 m:m (drug: cyclodextrin) ratio due to additional cyclodextrin molecules establishing electrostatic interactions with the inclusion 1 : 1 complex.
  • a final 1 :30 m:m (drug: cyclodextrin) ratio complex is obtained by the (a1 ) process.
  • step g provides only inclusion complexes.
  • Tables 1 and 2 refer in particular to fenretinide-(2-Hydroxypropyl)- -cyclodextrin complexes but similar results have been obtained with other cyclodextrins.
  • the complexes prepared by the a1 method are also endowed with the ability to release the complexed drug only in the presence of an absorbing phase (that in vivo may be a tumour mass) and not in a simple aqueous environment (that in vivo may be blood and body fluids).
  • Fig 4 reports the release of fenretinide from the a1 complex in phosphate buffered saline (PBS)and in the same buffer in the presence of an organic solvent acting as an absorbing phase for the drug. The release is described by the decrease of the UV absorbance at the maximum wavelength of fenretinide (340 nm) in the aqueous solution of the complex.
  • PBS phosphate buffered saline
  • Method a preparation of fenretinide potassium salt.
  • 4 mmoles of fenretinide and 4 mmoles of anhydrous_potassium hydroxide are mixed together and grinded by an end-runner mill or by a mortar-pestle under dry conditions at 30 °C in the dark. Mixing and grinding are carried out at homogeneity until the development of a red-orange colour indicating the formation of the fenretinide monohydrate salt.
  • Method b preparation of fenretinide potassium salt.
  • 4 mmoles of fenretinide and 4 mmoles of anhydrous_sodium hydroxide are mixed together and grinded by an end-runner mill or by a mortar-pestle under dry ccoonnddiittiioonnss aatt 3300 °°CC iinn tthhee ddaarrkk..
  • MMeetthhoodd dd :: pprreeppaarraattiioonn ooff aajjfeennrreettiinniiddee sodium.. ⁇ ppoottaassssiiuumm ssaalltt
  • the amine may be R 1 R 2 R 3 N wherein each of Ri , R 2 and R 3 is independently selected from the group consisting of hydrogen, a C C 2 o alkyl group, a C 3 -C 8 cycloalkyl group, a phenyl, a morpholino, and a pyridino group.
  • the amine may be cyclic amines such as aziridines, azetidines, pyrrolidines, piperidines, piperazines, morpholines and their derivatives.
  • the amine can be an amino acid.
  • the quaternary ammonium compound is selected from R R 2 R 3 R 4 N wherein each of R «, R 2 , R 3 and R 4 is independently selected from the group consisting of hydrogen, a Ci-C 2 o alkyl group, a C 3 -C 8 cycloalkyl group, a phenyl, a morpholino, and a pyridino group, .
  • the ammonium may be cyclic such as aziridines, azetidines, pyrrolidines, piperidines, piperazines, morpholines or derivatives thereof, the ammonium can be phosphatidylcholine, sphingomyelin or derivative thereof, thiamine, carnitine, choline and quaternized amino acids;
  • the solid or semisolid residue obtained may be optionally washed with water to eliminate traces of the base used for salification or the acid formed by the combination of the ammonium coupled anion with the phenolic hydrogen of fenretinide when salification is performed by quaternary ammonium compounds.
  • the washed residue is subsequently lyophilized and optionally washed with diethyl ether to eliminate traces of the unsalted drug.
  • Fenretinide is solubiiized in ethanol or methanol or dimethyl sulphoxide or tetrah drofuran or acetone or acetonitrile or a mixture of them at concentrations ranging from 0.1% to 10% (w:v fenretinide : solvent)). A stoichiometric amount of an amine or a quaternary ammonium compound is then added .
  • the amine may be R 1 R 2 RaN wherein each of Ri, R 2 and R 3 is independently selected from the group consisting of hydrogen, a C C 2 o alkyl group, a C 3 - C 8 cycioalkyl group, a phenyl, a morpholino, and a pyridino group.
  • the amine may be cyclic amines such as aziridines, azetidines, pyrrolidines, piperidines, piperazines, morpholines and their derivatives.
  • the amine can be an amino acid or a derivative thereof.
  • the quaternary ammonium compound is selected from R1 R2 3R4 wherein each of R-i, R 2 , R 3 and R 4 is independently selected from the group consisting of hydrogen, a C1-C20 alkyl group, a C 3 ⁇ C 8 cycioalkyl group, a phenyl, a morpholino, and a pyridino group, .
  • the ammonium may be cyclic such as aziridines, azetidines, pyrrolidines, piperidines, piperazines, morpholines or derivatives thereof, the ammonium can be phosphatidylcholine, sphingomyelin or derivatives thereof, thiamine, carnitine, choline and quaternized aminoacis;
  • the mixture is then stirred or sonicated in an ultrasound bath in the dark until homogeneity then the solvent is evaporated in a rotary evaporator.
  • the solid or semisolid residue obtained may be optionally washed with water to eliminate traces of the base used for salification or the acid formed by the combination of the ammonium coupled anion with the phenolic hydrogen of fenretinide when salification is performed by quaternary ammonium compounds.
  • the residue is subsequently lyophilized and may be optionally washed with diethyl ether to eliminate traces of the unsalted drug.
  • the aqueous solubility of the fenretinide salts has been evaluated by saturated solutions obtained by placing excess amounts of the salts in water and stirring 12 h in the dark at 25°C. After filtration through 0.2 pm the saturated solutions have been spectrophotometrically analysed at the maximum absorption wavelength obtained by scanning each solution in the range 190 nm to 400 nm. The extinction coefficient for each salt has been obtained from the absorbance, at the same maximum absorption wavelength, of unsaturated solutions prepared at known concentrations. The salts resulted more soluble than fenretinide (see Table 2 in the description above) whose solubility in water at 25 °C is 1.71 ⁇ 0.08 pg/ml.
  • solubility of the salts is suitable for complexation with cyclodextrins and the carrier molecules that provide liposomes, micelles, supramolecular assemblies, ion pairs. Indeed the solubility of the fenretinide salts is neither too high to prevent complexation cause the strong salt affinity for water, nor too low to hinder complexation due to the hydrophobicity of the salt molecules preventing their dissolution in water.
  • the cyclodextrin is physically mixed with fenretinide salt in a molar ratio varying from 1 :1 to 1 :100 (salt:cyclodextrin) to homogeneity.
  • the physical mixture obtained is subsequently dispersed in water at a concentration ranging from 0.05 to 1g/ml.
  • the suspension so obtained is stirred at room temperature for 1 h in the dark .then left at 4°C overnight in the dark. Subsequently the suspension is again stirred at room temperature for 1 h in the dark , then diluted with aqueous buffer pH 7.0 - 8.0 and filtered through a 0.2 ⁇ filter.
  • the filtered solution is finally lyophilized.
  • the cyclodextrin is physically mixed with a fenretinide salt in a molar ratio varying from 1 :1 to 1 :100 (salt: cyclodextrin) to homogeneity.
  • the physical mixture obtained is subsequently dispersed in water at a concentration ranging from 0.05 to 1g/ml.
  • the suspension so obtained is stirred at room temperature for 1 h in the dark then left at 4°C overnight in the dark. Subsequently the suspension is again stirred at room temperature for 1 h in the dark then diluted with aqueous buffer pH 7.0 - 8.0and filtered through a 0.2 pm filter.
  • the filtered solution is finally lyophilized.
  • the powder is dissolved in water at a concentration ranging from 50 to 150 mg/ml preferably 100 mg/ml.
  • the solution obtained is applied to a sephadex G-50 column previously equilibrated with water.
  • the column is eluted by water.
  • the fractions containing the complex are pooled and lyophilized .
  • the cyclodextrin is dispersed in the minimum volume of N-methylpyrrolidone or dimethyl sulphoxide or ethanol or methanol or a mixture of them to obtain a viscous knead.
  • Fenretinide salt is added in amounts corresponding to molar ratios varying from 1 :1 to 1 :100 (salt: cyclodextrin).
  • the mixture obtained is stirred to homogeneity and subsequently added to water in amounts ranging from 1 :50 to 1 :100 (w:v, mixture:water).
  • the suspensions obtained are stirred at room temperature for 15-50 min then sonicated at in an ultrasound bath in the dark at 37 °C 1 h. It is then left at 4°C overnight in the dark. Subsequently the suspension is again sonicated 10-30 min at RT, diluted with water and filtered through a 0.2 pm filter. The filtered solution is finally lyophilized.
  • the suspension is sonicated in an ultrasound bath in the dark at 37 °C 1 h. It is then left at 4°C overnight in the dark. Subsequently it is again sonicated 10-30 min at RT, diluted with water and filtered through a 0.2 pm filter. The filtered solution obtained is finally lyophilized.
  • the complexes are analysed by H-NMR (600 MHz; D20) and by spectrophotometric methods to evaluate the mode of interaction between fenretinide and cyclodextrin in the complexes prepared by the different processes. It is well known in the art that complexation may be studied by 1 H-NMR spectroscopy by measuring the difference in the proton chemical shifts between the free drug and the drug inserted into the cyclodextrin cavity and that chemical shift changes are markers of inclusion complexes.
  • the chemical shift changes of the protons from the complexed drug may be either downfield or up field changes depending on the molecular structure of the drug, the cyclodextrin type and the steric conformation of the drug molecule inside the cyclodextrin cavity.
  • the 1 H-NMR spectra of the complexes prepared by the physical mixture process displayed downfield shift changes compared to the fenretinide salt.
  • the complexes obtained by the kneading process and co-precipitation process at contrary, did not show chemical shift changes ( ⁇ 0) in the 1 H-NMR spectra indicating that supramolecuiar rather than inclusion complexes are obtained.
  • the supramolecuiar complexes are drug: cyclodextrin aggregates stabilized by electrostatic interactions without inclusion of the drug in the hydrophobic cavity of the cyclodextrin. They are generally known to be less efficient in raising the solubility of the complexed drug and less predictable in their ability to retain the drug in an aqueous environment and release it in the presence of an absorbing phase.
  • CE capillary electrophoresis
  • HPLC high-liquid phase separation
  • the separations are carried out on a fused-silica capillary of 50 ⁇ internal diameter, with a total length of 23 cm (effective length 19.5 cm)
  • the electrophoretic runs are performed at a constant voltage of 5.0 kV with controlled temperature at 25°C
  • the complexes are dissolved in waterethanol (1 :1 , v:v) mixtures and injected hydrodynamically using a pressure of 2 p.s.i. x s (1 p.s.i.
  • the detection is performed at the wavelength of maximum absorption of fenretinide (340 nm)
  • the running buffer is constituted of an aqueous solution of sodium tetraborate (20 mM; pH 9.2) supplemented with 100 mM of sodium dodecylsulfate.
  • the complex is dissolved in water and isocratically eluted on a C18 column (3 ⁇ , 150 mm x 4.6 mm I.D.) with a mixture of acetonitrile:water:glacial acetic acid (80:18:2, v/v/v) at a flow rate of 1.0 mL/min, at room temperature, according to a described and validated method (J. Chromatogr.
  • the differences between (a1 ) and (a2) are due to the gel filtration through sephadex performed in (a2). Indeed the complex obtained by the process (a1 ) is formed by inclusion complexation + supramolecuiar complexation where inclusion complexation provides 1 :1 m:m (drug: cyclodextrin) ratio while supramolecuiar complexation provides 1 : >1 m:m (drug: cyclodextrin) ratios due to additional cyclodextrin molecules establishing electrostatic interactions with the inclusion 1 : 1 complex. In this way a final 1 :30 m:m (drug: cyclodextrin) ratio complex is obtained by the (a1 ) process.
  • Verification The aqueous solubility of fenretinide from the complexes has been evaluated by UV- Vis-spectroscopy and HPLC methods. Saturated solutions were prepared by placing excess amounts of complexes in water, stirring 12 h in the dark at 25°C and filtering through 0.2 pm. After filtration the saturated solutions were spectrophotometrically analysed at the maximum absorption wavelength of fenretinide (340 nm).
  • the fenretinide-cyclodextrin complex should provide the free drug for absorption into the tumour cells.
  • drug release from the complex should take place in the presence of an absorbing biological phase such as the cell membranes of a tumour cell population.
  • the drug release should be suppressed through circulation in the bloodstream and distribution in the body fluids.
  • the release of the drug from the complex was evaluated by a spectrophotometry analysis of the complex solution in the releasing compartment at the maximum wavelength of fenretinide (340 nm)at appropriate time points.
  • the results, reported in figure 4, show that in the absence of the organic- extracting solvent fenretinide is not released from the complex as no variation in the absorbance is observed over 24 h.. Conversely in the presence of the absorbing phase a progressive decrease in the absorbance is observed indicating a gradual release of the drug from the complex taking place over time.
  • a measure of the drug release may be obtained by the difference between the initial absorbance (Ao) and the absorbance at each time point (At) compared to the initial absorbance (Ao-At)/Ao, In this case a 70% release was obtained at 24 h and 58% after 12h.
  • cancer stem cells display a high sensitivity to the fenretinide:HPBCD complex with IC50 values ranging from 0.04 to 7 ⁇ , in contrast to commercial ceil lines tested, which showed an average IC50 around 25 ⁇ (see Table 5 below).
  • the fenretinide:HPBCD complex represents a candidate for anticancer therapies directed against the CSC pool, which may be used both as a complement to conventional treatments based on chemotherapy (which are usually unable to eradicate CSC) and as monotherapy in cases where chemotherapy is ineffective.
  • the fenretinide:HPBCD complex obtained by the process of the invention steps a-e has been evaluated for its in vitro activity both on commercial cancer cell lines (Calu3, H1299, A549, H1975, H1781 , H460, H292, Calul ) and on CSC lines obtained from solid tumours (non-small cell lung cancer, breast cancer, colorectal cancer, glioblastoma, melanoma, sarcoma and ovarian cancer).
  • CSC were isolated from surgical specimens through mechanical and enzymatic dissociation followed by selection in serum-free medium and subsequently maintained in culture as multicellular spheroids (Figure 7A) in serum-free medium containing 20 ng/ml epidermal growth factor (EGF) and 10 ng/ml basic fibroblast growth factor (bFGF) in a humidified 95% air/5% C0 2 atmosphere as described in (Francescangeli F. et al., Proliferation state and Polo-like kinase- 1 dependence of tumorigenic colon cancer cells, Stem Cells 2012 30(9); 1819-30).
  • CSC lines were routinely characterized for common tumour genetic mutations and validated for their ability to reproduce the tumour of origin when injected in immune-compromised mice.
  • fenretinide:HPBCD complex To test the activity of the fenretinide:HPBCD complex, we first compared pure fenretinide dissolved in EtOH to fenretinide:HPBCD dissolved in water. For viability assays, cells were seeded in 96-well plates (3500cells/well) as triplicates. Multicellular CSC spheroids were dissociated with TRIPLE Express (Invitrogen) as a single cells suspension 24 hours before plating. Cells maintained in medium alone were used as controls. We used a concentration of 10 ⁇ for commercial cell lines and of 1 ⁇ for CSC lines and cell death was quantitatively evaluated with the Cell Titer GLO assay (Promega) after 72 hours of incubation.
  • TRIPLE Express Invitrogen
  • Figure 7 shows the effect of fenretinide: HPBCD on CSC lines derived from solid tumours (colorectal cancer, glioblastoma, sarcoma, breast cancer, melanoma, ovarian cancer).
  • Figure 8 shows the effect of fenretinide:HPBCD on CSC lines derived from different histotypes of non-small cell lung cancer (NSCLC).
  • NSCLC non-small cell lung cancer
  • the fenretinide:HPBCD complex obtained by the process of the invention steps a-e has been evaluated for its in vivo activity by treating immune- compromised mice intravenously with the compound as described below.
  • mice Female 6-8 weeks old NOD.Cg-Prkdc sc ' ll2rg im,Wi '/SzJ (NSG) mice were subcutaneously injected with 2,5 x 10 5 lung CSC re-suspended in 100 ⁇ of a 1 :1 solution of atrigel - serum-free medium containing 20 ng/ml EGF and 10 ng/ml bFGF.
  • Pharmacological treatment with fenretinide:HPBCD or the excipient (cyclodextrins) was started when tumour xenografts reached an approximate volume of 100 mm 3 and consisted in tail vein injections of 2 mg/Kg fenretinide:HPBCD twice a week for three consecutive weeks.
  • Tumour volume was evaluated by using an external digital caliper, and mice weight was evaluated at the indicated times as an indicator of potential toxic effects. Additionally, the potential toxicity of fenretinide:HPBCD was evaluated by evaluating blood parameters at the end of the experiment, as shown in Figure 1 1 B. Treatment with the fenretinide:HPBCD complex resulted in significant inhibition of the growth of CSC- derived xenografts ( Figure 10). Importantly, mice treated with fenretinide:HPBCD did not experience any weight loss nor they showed any sign of drug-related toxicity such as body weight loss or modifications of blood parameters ( Figure 1 1 ), indicating that fenretinide treatment does not elicit toxic side effects.
  • Supramolecular aggregates prepared by the process of the invention (fenretinide salt-CM complex in the form of liposomes):
  • Fenretinide potassium salt has been mixed with phosphatidylcholine in the presence of tributyrine and ethanol as dispersion promoters in molar ratios 1 : 3 : 1.5 : 15 respectively.
  • the mixture has been homogeneously suspended in phosphate buffer pH 7.0 at a concentration of 4.0 mg/mL fenretinide.
  • After stirring 30 min at 25 °C in the dark the suspension has been analyzed by optical microscopy and dynamic light scattering to evaluate the morphology and dimensions of the supramolecular aggregates.
  • mice Female 6-8 weeks old NOD. Cg-Prkdcscid ll2rgtm1Wjl/SzJ (NSG) mice were subcutaneously injected with 5 x 10 5 colon cancer stem cells (CSC, SA54) resuspended in 100 ⁇ of a 1 :1 solution of Matrigel - serum-free medium containing 20 ng/ml EOF and 10 ng/ml bFGF.
  • CSC colon cancer stem cells

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Public Health (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Epidemiology (AREA)
  • Medicinal Preparation (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

La présente invention concerne de nouveaux complexes solubles dans l'eau de molécules porteuses de fenrétinide, leur utilisation médicale, des compositions pharmaceutiques les comprenant, des procédés de préparation desdits complexes et des utilisations de ces complexes.
PCT/IB2015/056862 2014-09-10 2015-09-08 Complexes de fenrétinide WO2016038534A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
ITRM20140511 2014-09-10
ITRM2014A000511 2014-09-10

Publications (1)

Publication Number Publication Date
WO2016038534A1 true WO2016038534A1 (fr) 2016-03-17

Family

ID=51799248

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IB2015/056862 WO2016038534A1 (fr) 2014-09-10 2015-09-08 Complexes de fenrétinide

Country Status (1)

Country Link
WO (1) WO2016038534A1 (fr)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN110124049A (zh) * 2019-04-29 2019-08-16 大连医科大学 聚乙二醇化芬维a胺前体药物及其用途
IT201800006278A1 (it) * 2018-06-13 2019-12-13 Bionanofenretinide nuova formulazione antitumorale

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009016663A1 (fr) * 2007-07-31 2009-02-05 Istituto Giannina Gaslini Conjugués de dextrine amphiphiles et leur utilisation dans des formulations pharmaceutiques en tant qu'agents complexants pour des médicaments hydrophobes pour améliorer la solubilité aqueuse et par conséquent l'efficacité thérapeutique de médicaments complexés

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009016663A1 (fr) * 2007-07-31 2009-02-05 Istituto Giannina Gaslini Conjugués de dextrine amphiphiles et leur utilisation dans des formulations pharmaceutiques en tant qu'agents complexants pour des médicaments hydrophobes pour améliorer la solubilité aqueuse et par conséquent l'efficacité thérapeutique de médicaments complexés

Non-Patent Citations (29)

* Cited by examiner, † Cited by third party
Title
BREWSTER ME; LOFTSSON T, ADV DRUG DELIV REV., vol. 59, 2007, pages 645 - 66
BREWSTER ME; LOFTSSON T: "Cyclodextrins as pharmaceutical solubilizers", ADV DRUG DELIV REV., vol. 59, 2007, pages 645 - 66
CAROSIO R ET AL., J PHARM PHARMACOL., vol. 64, no. 2, February 2012 (2012-02-01), pages 228 - 36
CAROSIO R; PISTOIA V; ORIENTI I; FORMELLI F; CAVADINI E; MANGRAVITI S; MONTALDO PG; OGNIO E; EMIONITE L; ZUCCARI G: "Enhanced anti-neuroblastoma activity of a fenretinide complexed form after intravenous administration", J PHARM PHARMACOL., vol. 64, no. 2, February 2012 (2012-02-01), pages 228 - 36
DURANTE ET AL., ONCOTARGET, vol. 5, no. 13, 15 July 2014 (2014-07-15), pages 4811 - 20
DURANTE S; ORIENTI I; TETI G; SALVATORE V; FOCAROLI S; TESEI A; PIGNATTA S; FALCONI M: "Anti-tumor activity of fenretinide complexed with human serum albumin in lung cancer xenograft mouse model", ONCOTARGET, vol. 5, no. 13, 15 July 2014 (2014-07-15), pages 4811 - 20
FRANCESCANGELI F. ET AL.: "Proliferation state and Polo-like kinase-I dependence of tumorigenic colon cancer cells", STEM CELLS, vol. 30, no. 9, 2012, pages 1819 - 30
FRANCISCO B. T.: "Review: Cyclodextrin Inclusion Complexes Probed by NMR Techniques, Magnetic Resonance Spectroscopy", 2012, article "Pessine, Adriana Calderini and Guilherme L. Alexandrino"
J. CHROMATOGR. B, vol. 808, 2004, pages 125 - 130
LOFTSSON T.; MASSON M.; BREWSTER M.E., J. PHARM. SCI., vol. 93, 2004, pages 1091 - 1099
LOFTSSON T; MÁSSON M; BREWSTER ME: "Self-association of cyclodextrins and cyclodextrin complexes", J PHARM SCI., vol. 93, no. 5, May 2004 (2004-05-01), pages 1091 - 9
MAURER BJ ET AL., CLIN CANCER RES, vol. 13, 2007, pages 3079 - 86
MAURER BJ; KALOUS 0; YESAIR DW; WU X; JANEBA J; MALDONADO V; KHANKALDYYAN V ET AL.: "Improved oral delivery of N-(4-hydroxyphenyl) retinamide with a novel LYM-X-SORB organized lipid complex", CLIN CANCER RES, vol. 13, 2007, pages 3079 - 86
NORIKO TAKAHASHI ET AL: "Effects on M5076-Hepatic Metastasis of Retinoic Acid and N-(4-Hydroxyphenyl) Retinamide, Fenretinide Entrapped in SG-Liposomes", BIOLOGICAL & PHARMACEUTICAL BULLETIN, vol. 26, no. 7, 1 July 2003 (2003-07-01), pages 1060 - 1063, XP055000740, ISSN: 0918-6158, DOI: 10.1248/bpb.26.1060 *
ORIENTI ET AL., BIOMACROMOLECULES, vol. 7, no. 11, November 2006 (2006-11-01), pages 3157 - 63
ORIENTI I ET AL., BIOMACROMOLECULES, vol. 8, no. 10, October 2007 (2007-10-01), pages 3258 - 62
ORIENTI I ET AL., NANOMEDICINE, vol. 8, 2012, pages 880 - 90
ORIENTI I; ZUCCARI G; BERGAMANTE V; CAROSIO R; GOTTI R; CILLI M; MONTALDO PG: "Fenretinide-polyvinylalcohol conjugates: new systems allowing fenretinide intravenous administration", BIOMACROMOLECULES, vol. 8, no. 10, October 2007 (2007-10-01), pages 3258 - 62
ORIENTI I; ZUCCARI G; BERGAMANTE V; MILEO E; LUCARINI M; CAROSIO R; MONTALDO PG: "Amphiphilic poly(vinyl alcohol) derivatives as complexing agents for fenretinide", BIOMACROMOLECULES, vol. 7, no. 11, November 2006 (2006-11-01), pages 3157 - 63
ORIENTI I; ZUCCARI G; CAROSIO R; MONTALDO PG: "Improvement of aqueous solubility of fenretinide and other hydrophobic anti-tumor drugs by complexation with amphiphilic dextrins", DRUG DELIV., vol. 16, no. 7, October 2009 (2009-10-01), pages 389 - 98
ORIENTI I; ZUCCARI G; FALCONI M; TETI G; ILLINGWORTH NA; VEAL GJ: "Novel micelles based on amphiphilic branched PEG as carriers for fenretinide", NANOMEDICINE, vol. 8, 2012, pages 880 - 90
RAFFAGHELLO L; PAGNAN G; PASTORINO F; COSIMO E; BRIGNOLE C; MARIMPIETRI D; BOGENMANN E; PONZONI M; MONTALDO PG: "Immunoliposomal fenretinide: a novel antitumoral drug for human neuroblastoma", CANCER LETT., vol. 197, no. 1-2, 18 July 2003 (2003-07-18), pages 151 - 5
RAINER KUEFER ET AL: "Antagonistic Effects of Sodium Butyrate and N-(4-Hydroxyphenyl)-retinamide on Prostate Cancer", NEOPLASIA, vol. 9, no. 3, 1 March 2007 (2007-03-01), pages 246 - 253, XP055187960, ISSN: 1476-5586, DOI: 10.1593/neo.06766 *
RAJENDIRAN N; MOHANDOSS T; VENKATESH G: "Investigation of inclusion complexes of sulfamerazine with ?- and ?-cyclodextrins: an experimental and theoreticalstudy", SPECTROCHIM ACTA A MOL BIOMOL SPECTROSC., vol. 124, 21 January 2014 (2014-01-21), pages 441 - 50
RAJEWSKI RA; STELLA VJ: "Pharmaceutical applications of cyclodextrins. 2", IN VIVO DRUG DELIVERY. J PHARM SCI., vol. 85, 1996, pages 1142 - 69
TAKAHASHI N; TAMAGAWA K; SHIMIZU K; FUKUI T; MAITANI Y: "Effects on M5076-hepatic metastasis of retinoic acid and N-(4-hydroxyphenyl) retinamide, fenretinide entrapped in SG-liposomes", BIOL PHARM BULL., vol. 26, no. 7, July 2003 (2003-07-01), pages 1060 - 3
VENTURA CA ET AL., MOL BIOMOL SPECTROSC., 2014
VENTURA CA; GIANNONE I; MUSUMECI T; PIGNATELLO R; RAGNI L; LANDOLFI C; MILANESE C; PAOLINO D; PUGLISI G: "Physico-chemical characterization of disoxaril-dimethyl-beta-cyclodextrin inclusion complex and in vitro permeation studies", EUR J MED CHEM., vol. 41, no. 2, 4 January 2006 (2006-01-04), pages 233 - 40
ZHU, G. ET AL.: "Polymeric retinoid prodrug PG-4HPR ennhances the radation response of lung cancer", ONCOLOGY REPORTS, vol. 18, no. 3, 1 September 2007 (2007-09-01), pages 645 - 651, XP002750835 *

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
IT201800006278A1 (it) * 2018-06-13 2019-12-13 Bionanofenretinide nuova formulazione antitumorale
CN110124049A (zh) * 2019-04-29 2019-08-16 大连医科大学 聚乙二醇化芬维a胺前体药物及其用途
CN110124049B (zh) * 2019-04-29 2023-09-22 大连医科大学 聚乙二醇化芬维a胺前体药物及其用途

Similar Documents

Publication Publication Date Title
Arpicco et al. Hyaluronic acid-coated liposomes for active targeting of gemcitabine
JP5770336B2 (ja) リポソーム組成物の製造方法
Zou et al. Peptide-modified vemurafenib-loaded liposomes for targeted inhibition of melanoma via the skin
Cheng et al. GE11-modified liposomes for non-small cell lung cancer targeting: preparation, ex vitro and in vivo evaluation
ES2762224T3 (es) Liposomas que comprenden lípidos conjugados con polímero y usos relacionados
CN102369008B (zh) 脂质体组合物
Liu et al. Asialoglycoprotein receptor-targeted liposomes loaded with a norcantharimide derivative for hepatocyte-selective targeting
Yin et al. A pH-sensitive hyaluronic acid prodrug modified with lactoferrin for glioma dual-targeted treatment
EA011612B1 (ru) Состав с иринотеканом
EP3372232A1 (fr) Agent thérapeutique antitumoral contenant une composition liposomale à base de gemcitabine et kit associé
WO2019133914A1 (fr) Procédé de traitement de tumeurs solides contenant des caractéristiques d'hypoxie et/ou de stroma
Wang et al. Tyrosine modified irinotecan-loaded liposomes capable of simultaneously targeting LAT1 and ATB0,+ for efficient tumor therapy
Han et al. Free paclitaxel-loaded E-selectin binding peptide modified micelle self-assembled from hyaluronic acid-paclitaxel conjugate inhibit breast cancer metastasis in a murine model
Liu et al. Inhibition of growth and metastasis of breast cancer by targeted delivery of 17-hydroxy-jolkinolide B via hyaluronic acid-coated liposomes
Gu et al. A6 peptide-tagged, ultra-small and reduction-sensitive polymersomal vincristine sulfate as a smart and specific treatment for CD44+ acute myeloid leukemia
Huang et al. NIR-II light evokes DNA cross-linking for chemotherapy and immunogenic cell death
Zhou et al. A facile and universal method to achieve liposomal remote loading of non-ionizable drugs with outstanding safety profiles and therapeutic effect
Hu et al. Reduction-triggered polycyclodextrin supramolecular nanocage induces immunogenic cell death for improved chemotherapy
Zhou et al. A linear polyethylenimine (LPEI) drug conjugate with reversible charge to overcome multidrug resistance in cancer cells
Baek et al. Tailoring renal-clearable zwitterionic cyclodextrin for colorectal cancer-selective drug delivery
Lai et al. Effect of Kupffer cells depletion on ABC phenomenon induced by Kupffer cells-targeted liposomes
WO2018172942A1 (fr) Nanoparticules de quercétine
Jang et al. π-Hyaluronan nanocarriers for CD44-targeted and pH-boosted aromatic drug delivery
US20180296529A1 (en) Liposome composition co-encapsulating doxorubicin and a prodrug of mitomycin c
WO2016038534A1 (fr) Complexes de fenrétinide

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: 15767327

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 15767327

Country of ref document: EP

Kind code of ref document: A1