WO2022106985A1 - Solid amorphous dispersions of cabozantinib-(s)-malate and processes for the preparation thereof - Google Patents

Solid amorphous dispersions of cabozantinib-(s)-malate and processes for the preparation thereof Download PDF

Info

Publication number
WO2022106985A1
WO2022106985A1 PCT/IB2021/060580 IB2021060580W WO2022106985A1 WO 2022106985 A1 WO2022106985 A1 WO 2022106985A1 IB 2021060580 W IB2021060580 W IB 2021060580W WO 2022106985 A1 WO2022106985 A1 WO 2022106985A1
Authority
WO
WIPO (PCT)
Prior art keywords
cabozantinib
solid dispersion
malate
graft copolymer
solid
Prior art date
Application number
PCT/IB2021/060580
Other languages
French (fr)
Inventor
Daniele Ciceri
Original Assignee
Indena S.P.A.
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 Indena S.P.A. filed Critical Indena S.P.A.
Publication of WO2022106985A1 publication Critical patent/WO2022106985A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/14Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles
    • A61K9/141Intimate drug-carrier mixtures characterised by the carrier, e.g. ordered mixtures, adsorbates, solid solutions, eutectica, co-dried, co-solubilised, co-kneaded, co-milled, co-ground products, co-precipitates, co-evaporates, co-extrudates, co-melts; Drug nanoparticles with adsorbed surface modifiers
    • A61K9/146Intimate drug-carrier mixtures characterised by the carrier, e.g. ordered mixtures, adsorbates, solid solutions, eutectica, co-dried, co-solubilised, co-kneaded, co-milled, co-ground products, co-precipitates, co-evaporates, co-extrudates, co-melts; Drug nanoparticles with adsorbed surface modifiers with organic macromolecular compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines

Abstract

The present invention relates to a solid dispersion of Cabozantinib malate in a graft copolymer and the preparation process thereof.

Description

SOLID AMORPHOUS DISPERSIONS OF CABOZANTINIB-(S)-MALATE AND
PROCESSES FOR THE PREPARATION THEREOF
Field of invention
The present invention relates to a solid dispersion of Cabozantinib malate in a graft copolymer and the preparation process thereof.
Background to the invention
Cabozantinib, having the formula 7V-(4-((6,7-dimethoxyquinolin-4-yl)oxy) phenyl)-N' -(4-fluorophenyl)-cy cl opropran- 1,1 -dicarboxamide (hereinafter called
“Cabozantinib 1” or “Cabozantinib free base 1”)
Figure imgf000002_0001
Formula 1 is an active pharmaceutical ingredient belonging to the class of tyrosine kinase receptor inhibitors (TKRIs), which is active against a broad spectrum of target sites, such as the kinase receptor RET, the mesenchymal epithelial transition factor receptor (MET), vascular endothelial cell growth factor receptor 2 (VEGFR2), and the AXL, FLT3 and c-KIT receptors (Griillich C., Recent Results Cancer Res. 2018; 211 :67-75); said receptors are not only involved in the ordinary cell functions, but also in disease processes, such as oncogenesis, tumour angiogenesis and maintenance of the tumoral microenvironment.
Cabozantinib 1 is approved and marketed for the treatment of medullary thyroid carcinoma (MTC), renal cell carcinoma (RC) and hepatocellular carcinoma (HCC); the formulations Cometriq® and Cabometyx®, the former designed for the treatment of MTC and the second for the treatment of RCC and HCC, contain the malate salt having formula 2 (hereinafter called “Cabozantinib-(S)-malate 2” or “Cabozantinib malate 2”), which is more water-soluble than the free base.
Figure imgf000003_0001
Cabozantinib 1, and the preparation process thereof, were described for the first time in WO 2005/030140; however, because of its low water-solubility, Cabozantinib 1 was considered unsuitable for use in solid dosage forms for oral administration.
According to the Biopharmaceutics Classification System (BCS), Cabozantinib 1 is classified as a class II medicament because of its low solubility and high permeability (Nguyen L et al.. J Clin Pharmacol. 2015; 55(11):1293-302). The low water-solubility of the medicaments belonging to said class represents one of the major critical factors in the development of oral formulations; said administration route is generally preferred because it avoids invasive practices (such as intramuscular or intravenous administration) and, in particular cases, such as cancer patients, is accompanied by increased efficacy of the treatment, thus improving patients’ agreement to and compliance with the treatments.
Initially, the problem of low solubility was overcome by preparing various Cabozantinib 1 salts, including Cabozantinib-(S)-malate 2, disclosed in WO 2010/083414. In particular, said document discloses crystalline forms N-l and N-2 and the amorphous form of Cabozantinib malate 2.
Further known crystalline forms of Cabozantinib malate 2 are as follows: forms Ml, M2, M3 and M4 (WO 2015/177758), II (Chinese patent application no. 108341773) S, M (WO 2018/104954), CSI, CSIII (WO 2020/0507622), C2, C3, C4 and C5 (WO 2020/075196). However, no polymorphic form has proved considerably more soluble than the others.
Solid dispersions are one of the most promising approaches to increasing the oral bioavailability of class II medicaments according to the BCS, and an alternative to the traditional investigations of the various polymorphic crystalline forms of the pharmaceutical active ingredients.
WO 2020/069138 relates to a dosage form comprising an active pharmaceutical ingredient, cyclodextrins and bicarbonate, which would enable the oral bioavailability of the active ingredient to be increased by an amount ranging between 10% and 200%. Cabozantinib malate 2 is cited in a long list of active ingredients, but not indicated as the preferred active ingredient, nor is it exemplified in the experimental section.
However, there is still a need to develop further solid forms of Cabozantinib malate 2 with even greater solubility, a factor which would improve the absorption of the medicament, increase its bioavailability and produce a better therapeutic effect; moreover, increased solubility would enable the amount of medicament administered to be reduced, while ensuring equal efficacy, at the same time reducing the serious side effects such as gastrointestinal perforations and fistulae, haemorrhages, thrombotic events, osteonecrosis of the lower jaw, etc., thereby increasing the safety of the medicament.
Description of figures
Figure 1: X-ray diffractogram of the amorphous solid dispersion of Cabozantinib malate 2 according to Example 1.
Figure 2: IR spectrum of the amorphous solid dispersion of Cabozantinib malate 2 according to Example 1.
Figure 3: TG/DTA profile of the amorphous solid dispersion of Cabozantinib malate 2 according to Example 1. Figure 4: DSC profile of the amorphous solid dispersion of Cabozantinib malate 2 according to Example 1.
Description of the invention
The present invention relates to a solid dispersion wherein Cabozantinib malate 2 is dispersed in a graft copolymer; in particular, the dispersion of Cabozantinib malate 2 in a graft copolymer of polyvinylcaprolactam-polyvinyl acetate-polyethylene glycol (hereinafter called “graft copolymer”) is an amorphous solid dispersion. In the present invention the term “solid dispersion” defines a system of two components, wherein a first component is dispersed in a second component and the resulting system is physically uniform, typically, a. hydrophobic active pharmaceutical ingredient (first component) is dispersed in a hydrophilic polymer material (second component).
The polymer material used in the present invention consists of a graft copolymer obtained by free-radical polymerisation of an N-vinyllactam, vinyl acetate and a polyether as described in US 8,158,686, which is incorporated in full herein for reference. Preferably, the copolymer is obtained by polymerisation of polyvinylcaprolactam, polyvinyl acetate and polyethylene glycol; said copolymer is commercially available from BASF under the Soluplus trademark®.
A first aspect of the invention relates to an amorphous solid dispersion of Cabozantinib malate 2 characterised as follows:
I. the X-ray (X-R.PD) diffraction lattice presents no recognisable peaks;
II. the Fourier transform infrared spectrum (FT-IR) comprises the following absorption frequencies: 2924, 1731, 1631, 1478, 1436, 1320, 1083, 973 and 837 cm'1;
III. the thermogravimetric and differential thermal analysis (TG/DTA) profile is characterised by a weight loss from the temperature of 210°C due to decomposition.
The differential scanning calorimetry (DSC) profile does not exhibit fusion events, only decomposition of the sample at 210°C. A second aspect of the invention relates to a process (alternatively called “method’') for the preparation of the amorphous solid dispersion of Cabozantinib malate 2, which involves the use of an organic solvent.
With special reference to the first aspect of the invention, the IR spectrum referred to in point II exhibits the following absorption frequencies: 2924, 2858, 1731, 1684, 1631, 1507, 1478, 1436, 1369, 1350, 1334, 1320, 1213, 1195, 1083, 973, 944, 837 and 714 cm-1.
With special reference to the second aspect of the invention, the preparation process of an amorphous solid dispersion involves suspending Cabozantinib malate 2 and the graft copolymer in a solvent, heating the suspension until a solution is obtained, and finally, removing the solvent; in the present invention the term “solution” refers to a physically homogeneous mixture wherein one or more ingredients are present in a single phase; the term “suspension” refers to a heterogeneous mixture wherein one or more ingredients are present in two or more phases.
In further detail, the process for preparation of an amorphous solid dispersion of Cabozantinib malate 2 comprises the following steps: a) Cabozantinib malate 2 and the graft copolymer are suspended in an organic solvent to give suspension A; b) suspension A is maintained under reflux and under stirring until a solution B is obtained; c) solution B is filtered to give solution C; d) the solvent of solution C is removed until a solid D is obtained; e) solid D is ground until the amorphous solid dispersion of Cabozantinib 2 is obtained as a powdery solid.
Typically, the ratio of the amount of Cabozantinib malate 2 to graft copolymer in step a) falls into the range 0.1-10; preferably 0.1-2.5. The use of said copolymer has proved particularly advantageous; it not only prevents crystallisation of Cabozantinib malate 2 in the dispersion, but also performs the function of active solubiliser due to the formation of micelles in contact with water.
Typically, the organic solvent used in step a) is selected from acetone, tetrahydrofuran, acetonitrile, methylene chloride, ethyl acetate, methanol and ethanol, preferably tetrahydrofuran.
Typically, suspension A is maintained under stirring for a time ranging between 30 seconds and 60 minutes; preferably for 10 minutes.
Typically, the filtration referred to in step b) is conducted through a 5-15 pm sintered filter (type G4). Typically, a technique selected from distillation, low-pressure distillation, freeze-drying and spray drying is used in step d) to remove the solvent; preferably, low-pressure distillation.
The solubility of the amorphous solid dispersion of Cabozantinib malate 2 was evaluated in water and in simulated biological fluids. In particular, comparative solubility studies were conducted between the amorphous solid dispersion of Cabozantinib malate 2 and the corresponding polymorphic cry stalling form N-2 in water and in various simulated biological fluids, such as fasted-state simulated gastric fluid at pH 1.6 (hereinafter called FaSSGF), fasted-state simulated intestinal fluid at pH 6.5 (hereinafter called FaSSIF), and fed-state simulated intestinal fluid at pH 5 (hereinafter called FeSSIF). As will be seen from the results set out in Table 1 to Example 2 in the Experimental Section, the amorphous dispersion of Cabozantinib malate 2 proved more soluble than the crystalline form, both in water and in simulated biological fluids; in particular, the solubility of the amorphous solid dispersion in FaSSIF is about 100 times greater than that of form N-2, and 28 times greater in FeSSIF, while in FaSSGF and in water, the solubility is 8 and 16 times greater respectively. The solubility data highlight two surprising aspects of the invention; firstly, in view of its ionic and hydrophilic nature, it would be expected that, as taught by Shamma R. et al., the solubility of the copolymer would not change in the gastrointestinal tract (Shamma R. N. et al., Powder Technology 2013, 237, 406-414), but surprisingly, the solubility of the solid dispersion varies significantly between FaSSGF and FaSSIF. A second advantageous aspect that emerges from the solubility data is that the greatest solubility is observed in FaSSIF; this finding is surprisingly useful because, as the administration of both Cometriq® and Cabometyx® is indicated on an empty stomach, the increased solubility under said conditions would enable Cabozantinib malate 2 to be replaced with the solid dispersion, thus significantly reducing the amount of medicament administered.
Experimental section
Materials and methods
Soluplus®, commercially available from BASF, was used as graft copolymer.
X-ray powder diffraction (Fig. 1)
The X-ray diffraction pattern was recorded on a Bruker D2-Phaser diffractometer. The X-ray generator was set to 30 kV and 10 mA, using CuK as radiation source. The sample was prepared in a sample holder and irradiated for an irradiation length of 10 mm. The data were recorded between 2 and 50 29 degrees every 0.02 29 degrees, with a recording time of 3 seconds per 29 degree.
Fourier-transform infrared spectroscopy (FTIR) (Fig. 2)
The infrared spectrum was recorded in attenuated total reflectance (ATR) using the Perkin Elmer Spectrum One Fourier Transform spectrometer, equipped with the Specac ATR Golden Gate accessory. The spectrum results from the acquisition and transformation of 16 scans in the spectral region between 4000-500 cm'1 at a resolution of 4 cm'1.
Thermogravimetry (TG) and differential thermal analysis (DTA) (Fig. 3)
The analysis was conducted with a Seiko TG/DTA7200 simultaneous system instrument and open aluminum crucibles (volume 40 pL). The TG/DT signal was recorded between 30°C and 300°C with a linear heating gradient (10°C/min) under nitrogen flow (200 mL/min). About 10 mg of sample was used for the measurement.
Differential scanning calorimetry (DSC) (Fig. 4)
The analysis was conducted with a Mettler DSC1 System instrument. The heat flow was recorded in a range between 30° and 300°C with linear gradient (10°C/min) and under nitrogen flow (50 mL/min). About 5 mg of sample was used for the measurement, in a sealed and then perforated aluminum crucible (volume 40 pl).
High-performance liquid chromatography (HPLC)
The analysis was conducted with an Agilent 1100 instrument, equipped with a Symmetry shield RP18 column, 150 x 4.6, 5 pm. Table 1 shows the gradient used for the eluent mixture (Eluent A: 10 Mm ammonium formate buffer, pH 6.6; Eluent B: acetonitrile).
Figure imgf000009_0001
Table 1
Examples
Example 1- Preparation of an amorphous solid dispersion of Cabozantinib malate 2
2.5 g of Cabozantinib malate 2 and 10 g of Soluplus® were suspended in 400 mL of tetrahydrofuran. The suspension was heated to reflux for 10 minutes until a solution was obtained, which was then filtered through a sintered filter typically having a pore diameter ranging between 5 and 15 pm. The solvent was removed by distillation under vacuum until a soft mass was obtained. The soft mass was dried under vacuum at 50°C for 24 hours. The resulting solid was ground until a powdery white solid was obtained.
Example 2- Solubility studies in simulated gastrointestinal fluids
The solubility studies on the solid dispersion prepared according to Example 1 were conducted in different biologically relevant media to evaluate its behaviour in the various sections of the gastrointestinal tract. The solubility studies were conducted in water, FaSSIF (pH 6.5), FeSSIF (pH 5) and FaSSGF (pH 1.6).
10-20 mL of water, FaSSIF, FeSSIF or FaSSGF were placed in 40 mL glass vials.
An amount of solid dispersion prepared according to Example 1, in excess of the expected saturated solubility, was added to the vials; the resulting mixtures were maintained under stirring for 2 hours at room temperature. The mixtures were filtered through 0.45 pm PTFE membrane filters. The filtered solutions were analysed with HPLC instrumentation. Table 2 compares the concentrations of Cabozantinib malate 2 and the solid dispersion prepared according to Example 1 in various simulated gastrointestinal fluids.
Figure imgf000010_0001
*50 mg of solid dispersion according to Example 1 contains 10 mg of
Cabozantinib malate 2 Table 2. Concentrations of Cabozantinib malate 2 in water and in various simulated biological fluids expressed in mg/mL.

Claims

1. A solid dispersion of Cabozantinib malate 2 in a graft copolymer.
2. The solid dispersion according to claim 1, wherein the graft copolymer is obtained by free radical polymerisation of an N-vinyl lactam, vinyl acetate and a polyether.
3. The solid dispersion according to claim 2, wherein the graft copolymer is obtained by polymerisation of polyvinyl caprolactam, polyvinyl acetate and polyethylene glycol.
4. The solid dispersion according to claims 1-3 wherein the dispersion is in amorphous form.
5. The solid dispersion according to claims 1-3 with IR spectrum comprising the following absorption frequencies: 2924, 1731, 1631, 1478, 1436, 1320, 1083, 973 837 cm'1.
6. The solid dispersion according to claim 5, wherein the IR spectrum comprises the following absorption frequencies: 2924, 2858, 1731, 1684, 1631, 1507, 1478, 1436, 1369, 1350, 1334, 1320, 1213, 1195, 1083, 973, 944, 837 714 cm'1.
7. Process of preparation of the solid dispersion according to claims 1-6 which involves the use of an organic solvent.
8. The process according to claim 7 comprising the following steps: a. Cabozantinib malate 2 and the graft copolymer are suspended in an organic solvent to give suspension A; b. suspension A is kept under reflux and stirring until a solution B is obtained; c. solution B is filtered to give solution C; d. the solvent of solution C is removed until a solid D is obtained; e. solid D is ground until the amorphous solid dispersion of Cabozantinib 2 is obtained as a powdery solid.
9. The process according to claim 8 wherein in step a), the ratio of Cabozantinib malate 2 to graft copolymer is included in the range 0.1-10.
PCT/IB2021/060580 2020-11-18 2021-11-16 Solid amorphous dispersions of cabozantinib-(s)-malate and processes for the preparation thereof WO2022106985A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
IT102020000027678 2020-11-18
IT102020000027678A IT202000027678A1 (en) 2020-11-18 2020-11-18 CABOZANTINIB-(S)-MALATO AMORPHOUS SOLID DISPERSIONS AND PROCESSES FOR THEIR PREPARATION

Publications (1)

Publication Number Publication Date
WO2022106985A1 true WO2022106985A1 (en) 2022-05-27

Family

ID=74557070

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IB2021/060580 WO2022106985A1 (en) 2020-11-18 2021-11-16 Solid amorphous dispersions of cabozantinib-(s)-malate and processes for the preparation thereof

Country Status (2)

Country Link
IT (1) IT202000027678A1 (en)
WO (1) WO2022106985A1 (en)

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3705118A1 (en) * 2017-10-31 2020-09-09 Samyang Biopharmaceuticals Corporation Oral solid dosage form composition having improved disintegration and preparation method therefor

Family Cites Families (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DK2392564T3 (en) 2003-09-26 2014-01-13 Exelixis Inc c-Met modulators and methods of application
DE102005053066A1 (en) 2005-11-04 2007-05-10 Basf Ag Use of copolymers as solubilizers for sparingly water-soluble compounds
SG173014A1 (en) 2009-01-16 2011-08-29 Exelixis Inc Malate salt of n- (4- { [ 6, 7-bis (methyloxy) quin0lin-4-yl] oxy}phenyl)-n' - (4 -fluorophenyl) cyclopropane-1,1-dicarboxamide, and crystalline forms therof for the treatment of cancer
EP3145913A1 (en) 2014-05-23 2017-03-29 Mylan Laboratories Ltd. Novel polymorphs of cabozantinib (s)-malate and cabozantinib free base
US11261160B2 (en) 2016-12-07 2022-03-01 Msn Laboratories Private Limited, R&D Center Process for the preparation of N-(4-(6,7-dimethoxyquinolin-4-yloxy)phenyl)-N′-(4-fluorophenyl)cyclopropane-1, 1-dicarboxamide, (2S)-hydroxybutanedioate and its polymorphs thereof
CN108341773A (en) 2017-01-21 2018-07-31 南京华威医药科技开发有限公司 The rich crystal form II for Buddhist nun's malate of card
SE1851052A1 (en) 2018-09-05 2020-02-26 Scania Cv Ab Method and control device for controlling vehicle communication during operation of a vehicle
WO2020075196A1 (en) 2018-10-11 2020-04-16 Cipla Limited Polymorphs of n-(4-(6,7-dimethoxyquinolin-4-yloxy) phenyl)-n'-(4-fluorophenyl)cyclopropane-1, 1-dicarboxamide, (s)- malate, methods of production and pharmaceutical uses thereof

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3705118A1 (en) * 2017-10-31 2020-09-09 Samyang Biopharmaceuticals Corporation Oral solid dosage form composition having improved disintegration and preparation method therefor

Also Published As

Publication number Publication date
IT202000027678A1 (en) 2022-05-18

Similar Documents

Publication Publication Date Title
AU2018200931B2 (en) A pharmaceutical composition comprising stable, amorphous hybrid nanoparticles of at least one protein kinase inhibitor and at least one polymeric stabilizing and matrix-forming component
JP2019163337A (en) Crystalline solid form of n-{4-[(6,7-dimethoxyquinolin-4-yl)oxy]phenyl}-n'-(4-fluorophenyl)cyclopropane-1,1-dicarboxamide, production process, and method of use
CN113527203A (en) Novel crystal form of lenvatinib mesylate and preparation method and application thereof
JP2017505796A5 (en)
WO2016165650A1 (en) Co-crystal of olaparib and urea and preparation method therefor
US10208065B2 (en) Crystalline free bases of C-Met inhibitor or crystalline acid salts thereof, and preparation methods and uses thereof
CN111467501B (en) Compositions with improved dissolution properties
CN105294717A (en) EGFR inhibitor salt, crystal form and purpose thereof
IL285017B2 (en) A pharmaceutical composition comprising stable, amorphous hybrid nanopraticles of at least one protein kinase inhibitor and at least one polymeric stabilizing and matrix-forming component
CN109996787B (en) Insoluble compound or solvate thereof, pharmaceutical composition and application thereof
EP2752414A1 (en) Crystalline form of apixaban
WO2022106985A1 (en) Solid amorphous dispersions of cabozantinib-(s)-malate and processes for the preparation thereof
AU2018252643A1 (en) Compound of EOC315 Mod.I crystal form and preparation method therefor
CA3010315A1 (en) Crystalline forms of thienopyrimidine compound
Mali Fast disintegrating tablets of olmesartan medoxomil using solid dispersion technique
US20240016739A1 (en) Novel amorphous active pharmaceutical ingredients
JP2023511675A (en) Salt of arylaminopurine derivative, method for preparation thereof, and use thereof
CN105753869B (en) A kind of eutectic and preparation method thereof of CDK inhibitor and mek inhibitor
CN114685466B (en) Acetinib eutectic salt
CN114685434B (en) Acetinib and glutaric acid eutectic crystal
CN114685433B (en) Apolitinib vanillic acid eutectic salt and preparation thereof
EP2824104A1 (en) Process for the preparation of form III of Vilazodone hydrochloride
AU2016382384A1 (en) Crystalline forms of hydrochloride salts of thienopyrimidine compound
CN114685438A (en) Novel axitinib malate salt
US20040224956A1 (en) Crystal of 6-[4-(4-pyridylamino)phenyl]-4,5-dihydro-3(2h)-pyridazinone hydrochloride trihydrate

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: 21819964

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: 21819964

Country of ref document: EP

Kind code of ref document: A1