Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
LIPOSOMES COMPRISING SPHINGOMYELIN
Document Type and Number:
WIPO Patent Application WO/2019/122220
Kind Code:
A1
Abstract:
The invention relates to liposomes. The liposomes comprise sphingomyelin in the lipid bilayer. The liposomes are configured to cross the blood-brain barrier for the treatment of neuro- degenerative diseases and spinal cord injuries. The liposomes are essentially free of ganglioside. The invention also relates to a method of producing liposomes and to the use of liposome as medicament.

Inventors:
HALBHERR STÉFAN (CH)
Application Number:
PCT/EP2018/086352
Publication Date:
June 27, 2019
Filing Date:
December 20, 2018
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
INNOMEDICA HOLDING AG (CH)
International Classes:
A61K9/00; A61K9/127
Domestic Patent References:
WO2007044748A22007-04-19
WO2009150686A12009-12-17
WO2014000857A12014-01-03
WO2008033253A22008-03-20
WO2008033253A22008-03-20
WO2007044748A22007-04-19
WO2009150686A12009-12-17
Foreign References:
US20140004172A12014-01-02
Other References:
SARAIVA C. ET AL., J CONTROLLED RELEASE, 2016
BETZER 0. ET AL., NANOMEDICINE, 2017
N ENGL J MED, vol. 376, no. 18, 4 May 2017 (2017-05-04), pages 1706 - 1708
NATURE, vol. 540, no. 7631, 23 November 2016 (2016-11-23), pages 15 - 16
ALZHEIMERS DEMENT, vol. 12, no. 2, February 2016 (2016-02-01), pages 110 - 120
J. NEUROL. SCI., vol. 324, no. 1-2, 2013, pages 140 - 148
SPINAL CORD, vol. 51, 2013, pages 2 - 9
ACTA ORTOP BRAS, vol. 24, no. 3, May 2016 (2016-05-01), pages 123 - 6
MOL NEUROBIOL, vol. 54, no. 1, January 2017 (2017-01-01), pages 623 - 638
GIM MING ONG ET AL.: "Evaluation of Extrusion Technique for Nanosizing Liposomes", PHARMACEUTICS, vol. 36, no. 8, 2016, pages 5
GIM MING ONG ET AL.: "Evaluation of Extrusion Technique for Nanosizing Liposomes", PHARMACEUTICS, vol. 36, no. 8, 2016
PERRIE ET AL.: "Manufacturing Methods for Liposome Adjuvants, in; Vaccine Adjuvants", METHODS AND PROTOCOLS, METHODS IN MOLECULAR BIOLOGY, vol. 1494, 2017
Attorney, Agent or Firm:
DR. WILMING, Martin et al. (CH)
Download PDF:
Claims:
Claims

1. Liposomes for the treatment of neurodegenerative diseases and spinal cord injuries, comprising sphingomyelin in the lipid bilayer and configured to cross the blood-brain barri er, characterized in that the liposome is essentially free of ganglioside, in particular the lipid bilayer of the lipo some is essentially free of gangliosides .

2. Liposomes according to claim 1, wherein the liposomes addi tionally comprise cholesterol.

3. Liposomes according to one of the preceding claims, wherein the liposomes are essentially free of surface-modifications.

4. Liposomes according to one of the preceding claims, wherein the liposomes have a mean diameter between 10 and 70 nm, preferably between 10 and 50 nm and most preferably 25 to 35 nm.

5. Liposomes according to one of the preceding claims, wherein the neurodegenerative disease is chosen from the group:

tauopathies, in particular Alzheimer's disease; synucleinop- athies, in particular Parkinson's disease; trinucleotide re peat disorder, in particular Chorea Huntington; motor neu rone disease, in particular amyotrophic lateral sclerosis; prion diseases, in particular Creutzfeldt-Jakob Disease;

diseases of the central nervous system, in particular multi ple sclerosis.

6. Liposomes according to one of the preceding claims, wherein at least one active component is comprised and/or encapsu lated in the liposomes.

7. Liposomes according to claim 6, wherein the active component is chosen from the group, consisting of: cholinesterase- inhibitors, in particular donepezil or tacrine; dopamine ag onist, in particular bromocriptin or pramipexol; resvera- trol; nicotinic derivatives, in particular nicotinamide, nicotinic acid, niacin or NAD+ .

8. Liposomes according to one of claims 1 to 5, wherein at

least gangliosides , in particular GM1 gangliosides , are en capsulated in the liposome as an active component.

9. Liposomes according to one of the preceding claims in a lip osomal formulation, wherein the liposomal formulation has a polydispersity index £ 0.15, preferably 0.1 to 0.15.

10. Liposomes according to one of the preceding claims in a lip osomal formulation, wherein the liposomes of the liposomal formulation have a mean relative circularity of 0.95, pref erably between 0.98 and 1.00.

11. Liposomes according to one of the preceding claims in a lip osomal formulation, wherein 90% of the liposomes of the for mulation are unilamellar, preferably 97 to 99% of the lipo somes of the formulation are unilamellar.

12. Method for producing liposomes, preferably liposomes accord ing to one of the claims 1 to 11, comprising the steps of: a) providing lipids and cholesterol in an organic solvent, b) adding an aqueous liquid,

c) sonication to enable liposome formation,

d) optionally: separating the liposomes, characterized in that step c) is carried out such that the liposomes have a mean diameter between 10 and 70 nm, prefer ably 10 to 50 nm and most preferably 25 to 35 nm.

13. Method according to claim 12, wherein the organic solvent used in step a) is chosen from the group, consisting of: ethanol, methanol, chloroform and mixtures thereof.

14. Method according to one of claims 12 to 13, wherein the

aqueous liquid used in step b) is chosen from the group, consisting of: water, aqueous buffer solution, aqueous gly cine-solution .

15. Method according to one of claims 12 to 14, wherein the or ganic solvent used in step a) and/or the aqueous liquid used in step b) comprise an active component, preferably chosen from the group: cholinesterase-inhibitors, in particular donepezil or tacrine; dopamine agonist, in particular bromo- criptin and pramipexol; resveratrol; nicotinic derivatives, in particular nicotinamide, nicotinic acid, niacin or NAD+ .

16. Liposomes according to one of claims 1 to 11 for use as a medicament, in particular for use in the treatment of neuro- degenerative diseases and spinal cord injuries.

17. Liposomes according to one of claims 1 to 11 for use in the treatment of neurodegenerative diseases and spinal cord in juries according to claim 16, wherein the treatment compris es the oral or intravenous administration of the liposomes.

18. Liposome according to one of claims 1 to 11 obtainable by a method according to one of claims 12 to 15.

Description:
Liposomes comprising sphingomyelin

The present invention relates to liposomes, a method of produc ing liposomes and liposomes for the use as a medicament.

A liposome is a spherical vesicle having at least one lipid bi layer. Liposomes may also be multivesicular liposomes in which one vesicle contains one or more smaller vesicles. The liposome has an aqueous solution core surrounded by a hydrophobic mem brane in the form of a lipid bilayer.

The use of liposomes for drug delivery has been proposed for a variety of drugs, particularly those which are administered par- enterally. Liposomes have the potential to provide controlled "depot" release of the administered drug over an extended time period, and to reduce side effects of the drug, by limiting the concentration of free drug in the bloodstream. Liposomes can al so alter the tissue distribution and uptake of drugs, in a ther apeutically favorable way, and can increase the convenience of therapy, by allowing less frequent drug administration. For ex ample, liposomes may transport encapsulated active components directly to the disease site, including tumour cells and sites of inflammation. The active component can be directly released from the liposome at the treatment site. Thus, a lower dosage of the active component is required, and side effects are in conse quence limited.

However, depending on the targeted cells, the liposomes need to be modified in order to assure the release of the medicament at the desired treatment site.

The development of drug delivery systems to treat neurodegenera- tive diseases and spinal cord injuries is particularly challeng- ing, as such systems need to reach the brain and/or the spinal cord. However, due to the restrictive nature of the blood-brain barrier, a special layer of tissue constituting a protective barrier between the central nervous system and the systemic blood circulation, the development of such systems remains ra ther challenging.

Different efforts have been made in the past to treat neuro- degenerative diseases with liposomes. WO 2014/000857 describes the use of liposomes comprising phosphatidic acid and/or cardi- olipin as well as apolipoprotein E (ApoE) as the active compo nent in the treatment of Alzheimer's disease. Even though amy loid plaque formation associated with Alzheimer's disease can be reduced at the extra- and intracellular level of the limbic sys tem upon treatment with such liposomes, accumulating evidence from human clinical trials suggests that plaque formation is ra ther a symptom of disease but not the cause. Multiple phase 3 clinical studies have failed to demonstrate that eliminating plaques slows down disease progression in humans. Recent scien tific literature suggests that the particle size of 100 nm de scribed in WO 2014/000857 is too large to efficaciously pass the blood-brain barrier (Saraiva C. et al . 2016 J Controlled Re lease; Betzer 0. et al . 2017 Nanomedicine (London)).

In the example of Alzheimer's disease, further efforts to devel op treatment have been undertaken. One recent example is the de velopment of an antibody-based therapy against Alzheimer intend ing to clear beta-amyloid plaques. However, accumulating evi dences from clinical trials suggest that monoclonal antibodies aiming at amyloid-beta clearance do not provide benefits to Alz heimer patients (N Engl J Med. 2017 May 4 ; 376 ( 18 ) : 1706-1708. and Nature. 2016 Nov 23 ; 540 ( 7631 ) : 15-16. and Alzheimers Dement. 2016 Feb; 12 (2) : 110-120. Therefore, the need to find alternative ap proaches for the treatment of Alzheimer's disease persists.

WO 2008/033253 A2 describes the use of liposome complexes for delivering pharmaceutical agents across the blood-brain barrier for the treatment of neurodegenerative diseases. The liposomes are prepared from phospholipids and are associated with a phar maceutical agent. Further, the liposomes are modified with sial ic acid-containing molecules, such as gangliosides , attached to the liposomes. The sialic acid-containing molecule may serve as a linker between the targeting agent, such as antibody based agents or peptides analogues, and the external surface of the liposome or may be attached to the external surface of the lipo some to prevent scavenging of the liposome by the body' s reticu- lo-endothelial system. In any case, according to WO 2008/033253, sialic acid-containing molecules are required for ensuring transportation of the targeting agent to the brain.

W02007/044748 discloses a pharmaceutical composition of lipo somes containing sphingomyelin to treat disorders involving neu ropathic pain and aberrant muscle contractions associated with bladder hyperactivity disorders. The liposomes are produced by thin film hydration.

W02009/150686 discloses liposomes which are capable of effec tively binding beta amyloid peptide and are useful for the treatment, prevention and diagnosis of Alzheimer's disease. The liposomes are produced by extrusion.

The drawback of such liposomes is a rather laborious and costly industrial scale production. Moreover, the targeting moiety - chemically linked to the liposomal surface - may generate body- foreign molecular structures, which are likely immunogenic and may provoke adverse drug reactions. In contrast, the liposomal membrane of the invention described in this patent application is essentially free of body-foreign molecules, resulting in high biocompatibility .

Another problem lies with the administration of certain active components such as the neuroprotector GM1 ganglioside for the treatment of neurodegenerative diseases. For example, the admin istration of ganglioside GM1 for indications such as Parkinson' s disease has been described to cause difficulties in treatment (J. Neurol. Sci. 2013; 324(1-2): 140-148). Furthermore, the treatment of spinal cord injuries using free GM1 has shown posi tive outcomes in patients (Spinal Cord (2013) 51, 2-9 and Acta Ortop Bras. 2016 May-Jun; 24 (3) : 123-6) . Due to the pharmacokinet ics of GM1, the substance has to be administered subcutaneously or intravenously at high doses. The high dose and route of ad ministration make patients prone to certain types of adverse re actions, such as local pain and swelling at the site of injec tion, erythema, pruritus and hematoma. It is desirable to avoid such side effects and to avoid the use of high amounts of GM1.

Therefore, there is an unmet medical need for an effective drug delivery systems, which can transport active components to the brain and to the spinal cord for the treatment of neurodegenera tive diseases, spinal cord injuries and other neurological dis orders. There is in particular a need to provide delivery sys tems that can overcome the restrictive mechanism imposed by the blood-brain barrier. Ideally, the delivery system can be admin istered non-parenterally, thus avoiding the risks and inconven iences associated with parenteral administration.

It is thus an object of the present invention to address those needs and to provide liposomes suitable as active components and/or as carrier systems in the treatment of neurodegenerative diseases and spinal cord injuries. It is another object of the present invention to provide a method of producing such lipo somes and provide the use of such liposomes as a medicament.

The objects have been solved by liposomes, a method for produc ing liposomes and liposomes for the use as medicament as out lined below.

The invention relates to liposomes, which comprise sphingomyelin (SM) in the lipid bilayer and are essentially free of gangli- osides. In particular, the lipid bilayer of the liposome is es sentially free of gangliosides . The liposomes are configured to cross the blood-brain barrier and are suitable for the treatment of neurodegenerative diseases and spinal cord injuries. The dif ferent properties of the liposome which render it suitable to configure the blood-brain barrier are described below in more detail .

Sphingomyelin belongs to the group of phospholipids and sphin- golipids. It makes up about 10 % of the lipids of the brain. Sphingomyelin tends to be in greatest concentrations in the plasma membrane, and especially in the outer leaflet, of cells.

Liposomes comprising sphingomyelin as described in this inven tion show enhanced stability and enhanced biological properties. These liposomes can act as a medicament. The liposomes may also act as drug carrier system with enhanced pharmacokinetics and therapeutic properties. Surprisingly, it was found that lipo somes essentially free of gangliosides are very efficient in crossing the blood-brain barrier and, after administration, can be found in the brain and spinal cord. Moreover, it has been demonstrated that the crossing efficacy is increased compared to liposomes comprising a significant amount of ganglioside (Figure 4) . Thus, an additional modification with ganglioside is not necessary to cross the blood-brain barrier. The liposomes ac cording to the invention are exceptionally suitable as medica ment and/or drug carriers for active components directed to the treatment of neurodegenerative diseases and spinal cord inju ries .

"Essentially free" in the context of the invention refers to an amount of ganglioside less than 5 %mol, preferably even less than 3 %mol and most preferably less than 1 %mol. It may also be that the liposomes are free of ganglioside.

Sphingomyelin used for the purpose of the present invention can be obtained either by way of synthesis or by way of extraction from natural based components, in particular components of ani mal origin. Preferably, the sphingomyelin used for the purpose of the present invention is Palmitoyl-D-erythro-sphingosine-1- phosphocholine . Palmitoyl-D-erythro-sphingosine-l-phosphocholine corresponds to the body' s own sphingomyelin type phospholipids (dl 8 : 1 /I 6 : 0 ) , resulting in an improved uptake of the liposome into the body, and in particular into the brain and spinal cord. Furthermore, its C16 chain provides a high liposomal stability.

It was further found that the liposomes can be metabolized in clearing organs such as spleen and liver and are thus removed from the body after treatment, avoiding long-term accumulation.

The liposomes may additionally comprise cholesterol (Choi) .

Preferably, the ratio of sphingomyelin and cholesterol in the liposome may vary between 60-40 %mol and 45-55 %mol respective ly. Liposomes comprising sphingomyelin and cholesterol show an enhanced circulation lifetime. They have improved pharmacokinet- ics and therapeutic characteristics. They are biocompatible and biodegradable. Sphingomyelin-cholesterol interaction may lead to cholesterol/sphingolipid-enriched nano- and micro-domains (re ferred to as membrane "rafts") in the plane of plasma and other organelle (e.g. Golgi) membrane. These domains play an important role in regulating synaptic functions and synapse formation, neurotransmitter release and synaptic plasticity (Mol Neurobiol. 2017 Jan; 54 (1) : 623-638) .

The liposomes may essentially be free of surface modifications. By "essentially free" in the context of the modification, it is meant that the modification constitute less than 5 %mol of the liposome, preferably even less than 3 %mol and most preferably less than 1 %mol. The liposome may also be completely free of surface modifications. The surface modification referred to are folic acid, peptides, antibodies, sugars, polyethylene glycol, monoclonal antibodies, fractions of monoclonal antibodies or surface proteins.

Side effects, caused by such modification, can thus be avoided. With this present innovation, a smaller liposomal diameter can be reached allowing a facilitated crossing of the blood-brain barrier. Moreover, the risks of an immune reaction may be lower when the body's own lipids are used. Liposomes without surface modification provide in this case a higher biological compliance avoiding amongst others an enhanced clearance rate. According to the current state of the art, the liposomal surface modification and active targeting is technically very challenging, which may also lead to inefficient biodistributions and lower cost benefit ratio. Further relevant aspects of the present invention may not only be the reduced costs but also the amount of manufacturing steps leading to a facilitated large-scale production. GM1 is known as neuroprotector. Further, GM1 may interact with a number of proteins that form precipitates in diseases of the central nervous system (CNS) including alpha-synuclein (Parkin son's disease), amyloid-beta (Alzheimer's disease), and hunting- tin (Huntington's disease) . GM1 and its derivatives are known to penetrate the blood-brain barrier and the neuronal plasma mem brane. Administration of LIGA20, a derivative of GM1 has also been demonstrated to reduce Parkinson' s symptoms in a rodent model of Parkinson's disease.

Thus, GM1 and derivatives may be inserted in the aqueous com partment of the liposome as an active component in the treatment of neurodegenerative diseases and spinal cord injuries. If in corporated as active component into the aqueous phase of the liposome, GM1 may be present in an amount between 5 and 15 %mol, preferably 9 to 11 %mol and most preferably 10%mol.

The surface charge of the liposome is an important consideration in the preparation of liposome formulations and a first analyti cal indication on the insertion of ganglioside GM1. If the gan- glioside GM1 is inserted into the liposomal lipid bilayer, the liposome shows a more negative Zeta-potential than the base ves icle lipid bilayer constituted of sphingomyelin and cholesterol. The Zeta-potential can be analysed using a DLS-device and lies in the range of -10 to -60 mV. Liposomes with SM/Chol show a Zeta-potential of -10 mV, SM/Chol/GMl liposomes show a Zeta- potential of -49 mV. GM1 is negatively charged at pH ³ 5, thus liposomes carrying GM1 become negatively charged.

By measuring the Zeta-potential, it can be determined whether the liposome is essentially free of gangliosides .

Preferably, the liposomes have a mean diameter between 10 and 70 nm, preferably between 10 and 50 nm and most preferably 25 to 35 nm. The mean diameter is determined by cryo transmission elec tron microscopy (cryoTEM) with a standard deviation of approx.

10 nm.

Liposomes of a mean diameter not exceeding 50 nm are more likely to pass the blood-brain barrier. In addition, they are opsonized less rapidly and at a lower extent than their larger counter parts and are cleared less rapidly by the reticuloendothelial system.

It is preferred that formulations based on such liposomes have a polydispersity index of £ 0.15, more preferably a polydispersity index from 0.10 to 0.15, and are therefore essentially monodis- perse. The polydispersity index is determined by dynamic light scattering (DLS) . A polydispersity index £ 0.15 is superior over the polydispersity indices of liposomal formulations known in the art. Liposomal formulations known in the art, available by extrusion, homogenization, and sonication procedures, typically show polydispersity indices of 0.2 to 0.4 (Gim Ming Ong et al . , Evaluation of Extrusion Technique for Nanosizing Liposomes, Pharmaceutics 2016 (8) 36, p. 5). Essentially monodisperse lipo somal formulations are beneficial for reproducibility purposes, industrial scale production and compliant with marketing author ization requirements.

The circularity and the lamellarity of the liposomes in a formu lation are determined by cryo transmission electron microscopy (cryoTEM) . Preferably, the liposomes have a relative circularity of 0.95 and most preferably of 0.98 to 1.00. A circularity of 1.00 represents an absolute circle according to the standard physic rules. Preferably, the liposomes are unilamellar and hold one inner compartment. The liposomes of a liposomal formulation according to the invention are preferably to 90% unilamellar and most preferably 97% to 99% unilamellar.

A homogeneous circularity and unilamellarity of the liposomal dispersion provides a controlled and industrially scalable manu facturing process.

In a preferred embodiment of the invention, the mean diameter of a formulation based on liposomes according to the invention af ter 6 months, preferably after 12 months, from manufacturing is between 10 and 70 nm, preferably between 10 and 50 nm and most preferably 25 to 35 nm. It is particularly preferred that the mean diameter of the liposomes in a formulation after 6 months, preferably after 12 months from manufacturing is essentially the same as the mean diameter of the liposomes in the formulation immediately after manufacturing (Figure 7).

In a preferred embodiment of the invention, the polydispersity index of a formulation based on liposomes according to the in vention after 6 months, preferably after 12 months from manufac turing is £ 0.15, preferably 0.1 to 0.15. It is particularly preferred that the polydispersity index of the liposomes after 6 months, preferably after 12 months, from manufacturing is essen tially the same as the polydispersity index of the liposomes im mediately after manufacturing (Figure 8).

The liposomes according to the invention are thus particularly stable. The controllability and longevity of the size of lipo somes is beneficial for manufacturing, storage, shelf life and patient safety proposes.

The neurodegenerative disease treatable with the liposomes may be chosen from the group: tauopathies, in particular Alzheimer's disease; synucleinopathies , in particular Parkinson's disease; trinucleotide repeat disorder, in particular Chorea Huntington; motor neurone disease, in particular amyotrophic lateral sclero sis; prion diseases, in particular Creutzfeldt-Jakob Disease; diseases of the central nervous system, in particular multiple sclerosis .

Spinal cord injuries as used in the context of the invention re fer to all types of spinal cord injuries, complete and incom plete ones.

Spinal cord injuries can be addressed with the liposome of this invention, preferably with the addition of ganglioside, for ex ample GM1, in the inner aqueous compartment of the liposome.

GM1, as a neuroprotector, has been shown to restrain the second ary damages (co lateral biochemical damages triggered upon cell death) upon primary damages (mechanical damages) . Furthermore, it has been observed to partially restore the sensory part of the concerned area(s) (Acta Ortop Bras. 2016 May-Jun; 24 (3) : 123- 6) .

Preferably, at least one active component is comprised and/or encapsulated in the liposomes. It may be also possible to com prise or encapsulate more than one active component. For exam ple, it is possible to comprise or encapsulate active components that show a synergistic effect upon release. At least one active component can also be comprised in the liposomal bilayer and an other at least one active component can be encapsulated in the same liposome. It is further possible, that the liposomes are in the form of multivesicular liposome and wherein different active components form part of the same or different smaller vesicles in the multivesicular liposome.

By "comprised in the liposome" it is meant that the active com ponent forms part of the lipid bilayer or is incorporated in the lipid bilayer, respectively. By "encapsulated in the liposome" it is meant, that the active component is enclosed in the inner aqueous compartment of the vesicle.

The term "active component" may include pharmacologically active drugs as well as pro-drugs. Pro-drugs are medications or com pounds that, after administration, are metabolized into pharma cologically active drugs.

The active component can be selected from the group consisting of small or large organic or inorganic molecules, nucleic acids, nucleic acids analogues and derivatives, peptides, peptidomimet- ics, protein, antibodies and antigen binding fragments thereof, monosaccharides, disaccharides, trisaccharides, oligosaccha rides, lipids, glycosaminoglycans , an extract made from biologi cal material, and any combination thereof.

The liposome itself can also be an active component, loaded and unloaded .

Those kinds of liposomes offer a broad range of applications.

The advantage of liposomes comprising or encapsulating active components can be found in an enhanced therapeutic effect. The liposomes may transfer the active components to the site of ac tion. Since the liposomal membrane is structurally similar to biological membranes, the liposomes may merge with the cellular membranes. Upon merging, the liposomal contents may be emptied into the cell where the active component can act. The use of liposomes as drug carrier system may reduce the side effects as sociated with the administration of the respective active compo nent and related to high systematic absorption of the active component. The active component can be accumulated at the de sired target. The components of the liposome bilayer may be me tabolised in the liver and/or spleen. Preferably, the active component is chosen from the group, con sisting of: cholinesterase-inhibitors, in particular donepezil or tacrine; dopamine agonist, in particular bromocriptin or pramipexol; resveratrol; nicotinic derivatives, in particular nicotinamide, nicotinic acid, niacin or NAD+ .

Sphingomyelin and/or cholesterol can be chosen as active compo nents too.

In a preferred embodiment of the invention, at least gangli- osides, in particular GM1 gangliosides , are encapsulated in the liposome as an active component.

A further aspect of the invention is a method for producing lip osomes, preferably liposomes as previously described. The method comprises the steps of: a) providing lipids and cholesterol in an organic solvent, b) adding an aqueous liquid,

c) sonication to enable liposome formation,

d) optionally: separating the liposomes,

Step c) is carried out such that the liposomes have a mean diam eter between 10 and 70 nm, preferably 10 to 50 nm and most pref erably 25 to 35 nm, measured by cryo transmission electron mi croscopy (cryoTEM) .

Preferably, the lipids and cholesterol in the organic solvent provided in step a) are not subjected to thin film hydration. By "thin-film hydration" a conventional method for the preparation of liposomes , involving the step of making a thin lipid film in a round-bottom flask by the removal of organic solvent, is meant . Using this method, heterogeneous liposomes are formed up- on the addition and agitation of a dispersion medium. Finally, after extrusion through polycarbonate membranes, homogeneous small liposomes are obtained.

In a preferred embodiment of the invention, the liposomes are not subject to a surface modification step, such that the lipo somes are essentially free of surface modifications. By "surface modification step" is meant incorporation of folic acid, pep tides, antibodies, sugars, polyethylene glycol, monoclonal anti bodies, fractions of monoclonal antibodies or surface proteins into the lipid bilayer of the liposome or chemical coupling of such compounds to the liposomal surface.

More preferably, the lipids and cholesterol are not subject to extrusion, i.e. the process does not comprise an extrusion step. By "extrusion" is meant a conventional technique for the prepa ration of liposomes, where a liposomal formulation is passed through a membrane of defined pore size. Extrusion processes have been discussed in the art as being the method of choice for liposome production (Gim Ming Ong et al . , Evaluation of Extru sion Technique for Nanosizing Liposomes, Pharmaceutics 2016 (8) 36; Perrie et al . , Manufacturing Methods for Liposome Adjuvants, in; Vaccine Adjuvants: Methods and Protocols, Methods in Molecu lar Biology, vol. 1494, 2017) .

It has been found that liposomes produced by sonication accord ing to this invention are smaller, less polydisperse, more sta ble and less prone to degradation than liposomes obtainable by conventional techniques.

Preferably, the aqueous solution in step b) is an aqueous buffer solution. Upon adding the aqueous liquid, the solved lipids and cholesterol precipitate. The final ratio of organic solvent in step a) and the aqueous liquid in step b) may be 1:9, meaning that the organic solvent is 10 % of the total liquid mixture.

Too high solvent concentration in the end product can lead to liposomal instability and/or degradation.

The sonication is preferably performed with an amplitude of at least 60 mih and for at least 1 hour. The sonication can be per formed up to 24 hours.

The separation step can be achieved by centrifugation; filtra tion; field flow fractionation (FFF) ; dialysis; chromatographic methods, preferably gel-permeations-chromatography .

The liposomes are separated from remaining substances of the liquid mixture, such as organic solvent, salts and/or deter gents. Preferably, step d) is performed by buffer exchange.

Preferably, steps c) and d) do not require extrusion or any oth er separation method for the generation of a homogenous liposo mal distribution. It is preferred that the liposomes are kept in the original mixture.

The liposome distribution is preferably at least 90% unilamellar and most preferably between 97% and 99% unilamellar. Preferably, the liposomes hold a circularity of 0.95 and most preferably be tween 0.98 and 1.00. Circularity and the lamellarity have been determined based on images recorded with a cryoTEM JEOL JEM- 2100F. In liposomal formulations according to the invention, the ratio of spherical liposomes to broken particles and/or aggre gates in weight-% is higher than 9:1, measured by cryo- transmission electron microscopy.

The method has the advantage, that small homogeneous liposome can be obtained in one sonication step avoiding thin-film hydra- tion and extrusion and other elaborated and costly steps. Lipo somes with a mean diameter of less than 50 nm have a higher ten dency to be stable and to cross the blood-brain barrier. In oth er words, the passing of the blood-brain barrier is facilitated by the small diameter of the liposomes.

At least a part of the lipids used in step a) may be chosen from the group: phospholipids, natural phosphatidylcholine and in particular sphingomyelin; glycolipids, in particular gangli- oside; and a combination thereof. It is also possible to use further components such as cholesterol which greatly contribute to the liposomal stability.

These lipids have the advantage of being stable and resistant. Further, they are biocompatible.

Preferably, the organic solvent used in step a) is chosen from the group, consisting of: ethanol, methanol, chloroform and mix tures thereof. Most preferably, organic solvents with high de gree of purity are used, e.g. ethanol or methanol absolute

>99.99%. Even more preferably, no thin-film hydration is needed.

The used lipids show a good solubility in these organic sol vents. By using organic solvents with a high degree of purity contamination of the liposomes with impurities is avoided.

The aqueous liquid used in step b) may be chosen from the group, consisting of: water, aqueous buffer solution, aqueous glycine- solution. Preferably, aqueous buffer solutions with a physiolog ical salt concentration, e.g. PBS (lOmM phosphate, pH 7.2-7.4, 0.9 % NaCl) can be used. It is also possible to use the follow ing aqueous buffer solutions: 150mM ammonium sulphate, 150nm calcium acetate, 150mM magnesium acetate, 150mM manganese ace tate, 150mM iron chloride, or 150mM copper sulphate. The aqueous liquid enhances the liposome formation. By using physiological salt concentration, the interior of the liposome resembles the physiological conditions in the body.

Preferably, the organic solvent used in step a) and/or the aque ous liquid used in step b) comprise an active component. The ac tive component is preferably chosen from the groups previously described. The active component is incorporated to the aqueous phase or the solvent, depending on their chemical properties.

It is also possible, that the organic solvent comprises a first active component and the aqueous liquid comprises a second ac tive component. These components may be chosen such that they show a synergistic effect.

A further advantage of having an organic and an aqueous solvent present in the preparation method of the liposomes can be found in a broader access towards active components. Components with a higher solubility in the organic solvent than the aqueous liquid may be equally used and vice versa.

Preferably, the active component should fulfil the following criteria :

show an amphiphilic solubility in water, meaning having a logD value between -2 and +2,

comprise at least one weak acid- or base group.

Under certain conditions, it may also be possible to use active components with a logD value > +2. Substances having a logD be tween -2 and +2 can be encapsulated by remote loading. Molecules with a logD beyond this range may be loaded by membrane encapsu lation.

The use of an additionally active component enhances the thera peutic effect. Due to their similarity with cell membranes, the liposomes may merge with the cell membrane and may specifically act as an active component at the target site. The liposomes may also release the encapsulated or comprised active components in to the cell after merging of the liposome with the cell mem brane .

The liposomes as previously described may be used as a medica ment, in particular for use in the treatment of neurodegenera- tive diseases and spinal cord injuries.

The neurodegenerative disease may be chosen from the group as previously described. The spinal cord injuries refer to all types of spinal cord injuries.

Preferably, the liposomes as previously described, in the treat ment of neurodegenerative diseases and spinal cord injuries as previously described, are administered orally or intravenously.

If administered orally, the liposome composition can be in form of a solid or a drinkable solution. It may be in the form of dragees, tablets, granulate, capsules, powder, an emulsion, sus pension or syrup. The liposomes as previously described have the advantage of having a stability resisting the conditions associ ated with passing the gastrointestinal passage. By oral admin istration, side effects associated with a subcutaneously or in travenously delivery can be avoided.

Further, if administered orally, the liposomal composition can include further ingredients. The addition of flavours would pro vide a more pleasant taste, enteric coatings e.g. on the tablets would provide an additional protection against the acid. Basic ingredients such as hydrogen carbonate may provide a stomach- friendly administration. Also vitamins or minerals could be in cluded . The oral administration has the advantage of being easier appli cable than intravenously. A patient would be able to take the medicament in accordance with the prescription and without the need of trained personal.

An intravenous administration can be of advantage if uptake of the liposomes through the gastrointestinal track is less fa voured, for example due to the patient's health condition.

For intravenous injection, the liposomes may be present in solved or suspended form. The amount of liquid may be in the range of 0.1 - 20 ml and is dose dependent. The injectable solu tion can comprise further ingredients, such as stabilising agents. It can also comprise physiological compatible ingredi ents such as salt, in particular sodium chloride or alcohol, preferably ethanol.

A further aspect of the invention is liposomes as previously de scribed obtainable by a method as previously described.

The invention will be further outlined in the following:

Figure 1: In vivo biodistribution of liposomes comprising sphingomyelin labelled with ICG according to the invention .

Figure 2: In vivo biodistribution of liposomes comprising sphingomyelin labelled with DiR according to the invention . Figure 3 : In vivo biodistribution of liposomes comprising sphingomyelin and GM1 labelled with DiR as compar ative example to figure 2.

Figure 4 : Graphic representation of the biodistribution

analysis in the brain, spinal cord, liver and spleen from the in vivo biodistribution images of figure 2 and 3.

Figure 5 : Characterization of the liposomes without surface modification by cryoTEM: (A) Visualization low magnification, (B) Visualization high magnifica tion, (C) Qualitative assessment, (D) Quantitative diameter distribution.

Figure 6: Quantitative characterisation of the liposomes without surface modification by cryoTEM. (A) cir cularity distribution, (B) lamellarity diagram.

Figure 7 : Characterization of size stability of the lipo somes over time, measured by dynamic light scat tering DSC.

Figure 8 : Characterization of polydispersity stability of the liposomes over time, measured by dynamic light scattering DSC.

Figures 9/10: In-vivo fluorescence of different liposomal formu lations in spinal cord and brain.

Figure 11 : In-vivo fluorescence of liposomal formulations with different lipid compositions in the brain Figure 1 shows the in vivo biodistribution of sphingomyelin lip osomes labelled with Indocyaninegreen (ICG) . Mice were treated intravenously with liposomes carrying near-infrared dye and bio distribution was analysed 24 hours post-injection. Analysis was performed with a GE Healthcare explore Optix. Signals of the ICG were found in brain (A) and the spinal cord (B) . Total liposome lipid injection was 45 mg/kg carrying 1:200 weight-to-weight ICG. Further signals could be found in the clearance organs liv er (C) and spleen (D) , indicating that after treatment the lipo somes can be removed from the body.

Figure 2 shows the in vivo biodistribution of sphingomyelin lip osomes labelled with DiR. Different mice A, B, C were treated intravenously with liposomes carrying near-infrared dye and bio distribution was analysed 24 and 48 hours post-injection in a ventral view and a dorsal view. Analysis was performed with an optical imaging system, IVIS Spectrum of Perkin Elmer. Signals of the DiR were found in the brain (circle) and spinal cord (rectangle) . Total liposome lipid injection was 15 mg/kg carry ing 50 mg/ml DiR. Further signals could be found in the clear ance organs liver (plain arrow) and spleen (doted arrow) , indi cating that after treatment the liposomes can be removed from the body. The fluorescence scale is termed in the following unit: total Radiant efficiency [p/s ] / [yW/cm 2 ] .

Figure 3 shows a comparative example of the in vivo biodistribu tion of a liposome with sphingomyelin and GM1, labelled with DiR. Mice were treated intravenously with liposomes carrying near-infrared dye and biodistribution was analysed 24 and 48 hours post-injection. Analysis was performed with an optical im aging system, IVIS Spectrum of Perkin Elmer. Signals of the DiR were found in brain (circle) and the spinal cord (rectangle) . Total liposome lipid injection was 25 mg/kg carrying 50yg/ml DiR. Further signals could be found in the clearance organs liv er (plain arrow) and spleen (doted arrow) , indicating that after treatment the liposomes can be removed from the body. The fluo rescence scale is termed in the following unit: total Radiant efficiency [p/s ] / [yW/cm 2 ] . Even though the liposomes are found in the same organs as the liposomes presented in figure 2, the biodistribution is less distinct compared to the essentially GMl-free liposomes in figure 2.

Figure 4 shows a graphic representation of the biodistribution analysis in the brain, spinal cord, liver and spleen from the in vivo biodistribution images of Figure 2 and Figure 3. Figure 4A shows the normalised fluorescence of the biodistribution of the liposome without ganglioside in four different tissues: brain, spinal cord, liver and spleen. The biodistribution is displayed for two different time points: 24 and 48 hours. The bars repre sent the standard deviation to the mean. Figure 4B shows the normalised fluorescence of the biodistribution of the liposome with ganglioside.

It was surprisingly found, that the in vivo biodistribution of the liposome essentially lacking ganglioside (Fig. 4A) is higher than the in vivo biodistribution of the liposome comprising gan glioside (Fig. 4B) .

Figure 5 shows the characterization of the liposomes without surface modification, liposomes were visualized using Cryo

Transmission Electron Microscope JEOL JEM-2100F and a TVIPS Tern- Cam camera (JEOL Ltd., Japan) . Figure 5A shows an image of the liposomes at low magnification (20000x) . Figure 5B shows the liposomes at high magnification (80000x) . Figure 5C shows a qualitative assessment done by ocular/visual observation of the liposomal distribution. Figure 5D shows the size distribution of the liposomes of this invention. In order to quantify the mean diameter of the liposomes N (liposomes) = 5128 were analysed (Vi- ronova Analyzer Software, Vironova, Sweden) . The mean diameter of the liposomes is 30.46 nm with a standard deviation of 10.10 nm.

Figure 6 comprises two further tests for a quantitative charac terisation of the liposomes without surface modification by cry- oTEM (Vironova Analyzer Software, Vironova, Sweden) . Figure 6A shows the circularity distribution of 5128 liposomes. Figure 6B shows the lamellarity grade of the liposomal distribution. 98% of 5128 liposomes have been characterised as unilamellar.

Figures 7 and 8 show the size and polydispersity stability of liposomes according to the invention over time, measured by dy namic light scattering. The liposomal formulations were obtained according to the method described above, by using sphingomyelin and cholesterol in a 1:1 molar ratio. The liposomes were com pletely free of gangliosides , surface modifications, and did not comprise or encapsulate an active component. The liposomal for mulations were stored in PBS at a pH-value of 6.8 and a tempera ture of 4°C. Size and polydispersity were determined by DLS standard methods. It shall be noted that the values measured by dynamic light scattering are slightly higher than the values ob tainable by cryoTEM due to the impact of the hydrodynamic radius of liposomes on DLS measurements. A diameter of 60 nm as indi cated in Figure 7 corresponds to a mean diameter in the range of 10 and 50 nm when measured by Cryo Transmission Electron Micros copy .

The dotted curve shows the results of a small scale production batch of liposomal formulation as described above, while the dashed curve shows the results of an upscale production, i.e. a batch size of 2 litres. As can be seen from Figures 7 and 8, both the size and polydispersity of the liposomal formulations from Q3 2017 to Q3 2018, i.e. during storage time of one year, remained essentially unchanged.

Figures 9 and 10 show relative in-vivo fluorescence of different liposomal formulations in the spinal cord and brain of mice. In both charts, the liposomes of groups 1 to 4 were obtained ac cording to the method described above, by using only sphingomye lin and cholesterol in a 1:1 molar ratio. Gr . 5 is a control group of free DiR in PBS. In Gr.l, synthetic sphingomyelin was used and GM1 was comprised in the liposomes. In Gr . 2, synthetic sphingomyelin was used and the liposome was completely free from surface modifications, in particular free from GM1. In Gr . 3, sphingomyelin of animal origin was used and GM1 was comprised in the liposomes. In Gr. 4 sphingomyelin of animal origin was used and the liposome was completely free from surface modifications, in particular free from GM1. For all four test groups, DiR was added as a labelling agent. The measurements were performed by NIR imaging technique.

Figures 9 and 10 show the accumulation of the four different kinds of liposomes in the spinal cord and brain respectively in 0. lh, 4h, 24h and 48 h post-injection. It can be seen that the presence of GM1 does not significantly affect the ability of the liposomes to target the central nervous system. The same holds true for the use of synthetic sphingomyelin compared to the use of sphingomyelin of animal origin.

Figure 11 shows the relative in-vivo fluorescence of liposomal formulations with different lipid compositions in the brain of mice. The liposomes of groups 1 to 3 were obtained according to the method described above by using lipids and cholesterol in a 1:1 molar ratio. In group 1, phosphatidylcholine and sphingomye- lin in combination were used as lipids. In group 2, phosphati dylcholine alone was used as a lipid. In group 3, sphingomyelin alone was used as a lipid. In all three test groups, DiR was added to the formulations as a labelling agent. The measurements were performed by NIR imaging technique.

Figure 11 shows the accumulation of the three different kinds of liposomes in the brain of mice after 24h and 48 h post

injection. It can be seen that the composition consisting of sphingomyelin and cholesterol alone results in superior longevi ty of circulation and CNS bioavailability of the liposome com pared to the other variants (Grps 1 and 2) .