The present invention relates to a method for producing synthetic extracellular vesicles comprising a lipid bilayer including at least two lipids, optionally one or more extracellular vesicle associated proteins, and optionally one or more nucleic acid molecules. The inventive synthetic extracellular vesicles are formed by emulsification using a mechanic emulsifier in the form of polymer shell stabilized synthetic extracellular vesicles. The inventive method allows producing synthetic extracellular vesicles miming the composition and function of natural extracellular vesicles. Therefore, synthetic extracellular vesicles with specific protein and nucleic acids composition are also disclosed herein, as well as their therapeutic uses.
Extracellular vesicles are membrane-contained small vesicles, secreted by all types of pro- and eukaryotic cells, and play a crucial role in intercellular signaling under both physiological and pathological conditions.
In physiological conditions, extracellular vesicles are important mediators for cell-to-cell and inter-tissue communication, thus playing a role in regulating homeostasis as well as other conditions. In pathological conditions, the information transferred by extracellular vesicles, mainly cancer cell extracellular vesicles, may have detrimental effects. Indeed, extracellular vesicles have been demonstrated to contribute to various pathologies such as tumorigenesis and metastasis, inflammation, and immune system activation.
As a result of the above-mentioned functions, extracellular vesicles may serve as novel tools for various therapeutic and diagnostic applications, such as anti-tumour therapy, pathogen vaccination, immune-modulatory and regenerative therapies and drug delivery. Indeed, extracellular vesicles can be used for the target-specific delivery of nucleic acid molecules, proteins or small molecules into the intracellular environment, where they also act on a genetic level.
The three main categories of extracellular vesicles are apoptotic bodies, shedding microvesicles and exosomes. Microvesicles and exosomes are smaller compared to apoptotic bodies. Additionally they differ from apoptotic bodies in their content, since they rarely contain DNA.
The main function of microvesicles and exosomes is the intercellular transfer of lipids, RNA, and cytosolic proteins, thereby affecting cell metabolism and functions, including, but not limited to migration, cell proliferation and differentiation.
A detailed and precise characterization of the intercellular signalling mechanisms mediated by extracellular vesicles is essential to develop extracellular vesicle-based therapeutic applications. However, the methods of the current art to isolate and purify extracellular vesicles are very complex, long and error prone providing extracellular vesicle preparations with low yield and purity and high variability between different batches, which hamper a correct understanding of extracellular vesicle biology and of their interactions with the environment. Moreover, the exosome preparations oft contain also microvesicles.
Therefore, synthetic and cleaner vesicle formulations are not only highly sought for therapeutic and clinical applications but also to study fundamental aspects of extracellular vesicle biology, signalling, as well as the role of their individual components.
Currently, synthetic exosomes are prepared by two types of methodologies: top-down or bottom-up (Garcia-Manrique P. et al., 2017. Fully Artificial Exosomes: Towards New Theranostic Biomaterials. Trends in Biotechnology). In top-down methodologies, the production of artificial exosomes begins with cultured and eventually engineered cells that are then processed to produce membrane fragments to be used to form the vesicles. Even if these methods enable synthesis of exosomes similarly to their natural counterpart, they still have some drawbacks. Indeed cargo loading is not tightly controlled due to the passive encapsulation of the surrounding medium during membrane fragment self-assembly. Moreover, these exosomes cannot have a defined composition and size, and are usually not homogenous in size. The final purification steps used for exosome isolation are time-consuming and characterized by low purity, yield and reproducibility in term of exosome composition.
The patent applications WO 2019 027847 A1, WO 2019 126068 A1, US 2016 0354313 A1, and US 2016 0354313 A1, and the disclosure of Kooijmans et al. (J. controlled release, 2016, 224, 77-85) refer to extracellular vesicles produced by top-down methodologies.
The international patent application WO 2019 027847 A1 describes the synthesis of bispecific nanoparticle vesicles that are able to redirect immune effector cells towards cancer cells for killing. However, the nanoparticle vesicles are prepared by transducing a population of cells comprising vesicles, such as exosomes, with polynucleotides coding the polypeptides of interest, and thus isolating the transduced vesicles or exosomes. The exosomes released into the culture media are purified using traditional approaches as differential centrifugation, density-gradient- or cushion-based ultracentifugation, precipitation with commercial kits, and affinity and size exclusion chromatography. Thus, the extracellular vesicles or exosomes described in WO 2019 027847 A1 do not possess a membrane bilayer with a defined lipid composition. Moreover, the vesicle preparation can still contain impurities due to the isolation procedure.
The international patent application WO 2019 126068 A1 discloses engineered extracellular vesicles (EVs) produced by using a membrane cloaking platform technology, wherein the cloaking imparts to the EVs enhanced delivery to tissues of interest, such as damaged or dysfunctional tissue. The engineered EV compositions can be used to treat diseases. The EVs are obtained from culture media of not-modified cultured cells using standard methods and then tailored with fluorescent molecules or ligand proteins using the membrane cloaking platform technology. This consists in incubating the exosomes with a lipid anchor molecule, such as DMPE-PEG, bound to a member (e.g. streptavidin) of a coupling moiety and then with a biotinylated antibody or protein of interest.
The US Patent Application US 2016 0354313 A1 discloses a hybridosome, i.e. a hybrid biocompatible carrier, which is synthetized from two different vesicles, one is a naturally secreted vesicle (BDM), one is in vitro produced by using standard methods (EDEM) comprising at least one tunable fusogenic moiety. The hybridome are described as able to deliver bioactive agents into leukocytes or glial cells or into cells during ex-vivo expansion.
The scientific article of Kooijmans et al. discloses the engineering of extracellular vesicles derived from Neuro2A cells or platelets by mixing with nanobody-PEG-micelles, where the nanobodies are specific for a cellular target, such as the epidermal growth factor receptor (EGFR). The disclosed EVs are showed to efficiently target EGFR positive tumor cells, and to be stable in plasma for longer than 60 min post-injection.
The US Patent Application US 2019 202892 A1 discloses extracellular vesicles comprising an immune-modulating component, such as a cytokine or a binding partner of a cytokine (for example IL2, IL7, IL10, IL12, IL15 or others), and optionally a second component such as an activator for a positive co-stimulatory molecule or an activator for a binding partner of a positive co-stimulatory molecule (for example a TNF receptor superfamily member). The extracellular vesicles are obtained by isolation from producer cells using standard methods. In particular, the the extracellular vesicles with an immunomodulating component are obtained by modifying a producer cell with the immunomodulating component, and then obtaining the extracellular vesicles from the conditioned culture media of the modified producer cells.
Bottom-up methodologies to prepare synthetic exosomes involves the preparation of a synthetic bilayer that is then functionalized with selected proteins to mimic desired exosome functions. However, most of these methodologies are characterized by low encapsulation efficiency and high costs, as the methods are adapted from conventional liposome production routes. Moreover, methodologies to synthetize exosomes containing a specific composition of both exosome proteins and nucleic acids such as miRNAs are still missing.
The disclosures US 2017 0128367 A1, and US 2019 343767 A1 refer to synthetic extracellular vesicles or exosomes produced by bottom-up methodologies.
The patent application US 2017 0128367 A1 discloses liposomes comprising a cationic lipid and a lipid covalently conjugated to a PEG derivative, which is bound to a glycosaminoglycan coating the liposome. The PEG serves to stabilize the liposome, whereas the glycosaminoglycan, for example hyaluronic acid, is used to target the cells of interest. The produced liposomes are characterized by a narrow dimension range between 20 and 500 nm.
The patent application US 2019 343767 A1 discloses artificial exosomes comprising rab7, desmoplakin, alpha 2-HS glycoprotein (AHSG), and a cardiolipin or a variant thereof. The exosome can also comprise a cargo molecule as a peptide, a polypeptide, a nucleic acid, a virus, a small molecule, a fluorophore, or a combination thereof. The artificial exosomes are produced by mixing the single components DOPC, cholesterol, and a cardiolipin to form a cardiolipin-containing liposome, and incubating the cardiolipin-containing liposome with rab7, desmoplakin, and AHSG to form an artificial exosome. The artificial exosomes can thereafter optionally be loaded with siRNA molecules.
The disclosure of Weiss et al. (Nature Materials, 2017, 17, 89-96) teaches a high-throughput microfluidic method to generate liposomes to be used as synthetic model cell systems, called protocells, to study interactions of these synthetic cells with physiologically relevant environments such as extracellular matrices, cells or signalling proteins. These mechanically and chemically stable cell-like compartments, called droplet-stabilized GUVs (dsGUVs), can be loaded with biomolecules such as transmembrane and cytoskeleton proteins by microfluidic pico-injection technology. However, this method allows to regulating the diameter of the dsGUVs in the range from 28 μm to 120 μm.
Thus, none of the prior art documents discloses a method to produce fully synthetic extracellular vesicles at high efficiency, high stability, high controlled composition, high purity and reproducibility between different batches.
Therefore, there is still an urgent need for efficient procedures to produce fully synthetic exosomes, or extracellular vesicles with a high defined composition, low variability between different batches, high purity and efficiency.
It is the objective of the present invention to provide synthetic extracellular vesicles assembled with a highly controlled composition and produced surrounded by a stabilizing polymer shell, which can be used for therapeutic applications.
The objective of the present invention is solved by the teaching of the independent claims. Further advantageous features, aspects and details of the invention are evident from the dependent claims, the description, the figures, and the examples of the present application.
The present invention provides a method for high-throughput bottom-up assembly of fully synthetic extracellular vesicles with analogous functionalities to naturally occurring cell-derived extracellular vesicles. The production method is based on charge-mediated assembly of predefined functionalized lipid vesicles and encapsulation of miRNAs within the polymer shell stabilized lipid vesicles. “Charge mediated” assembly refers to the process in which the negative charge of the vesicles and the negative charge on the periphery of the polimer shell stabilized vesicles are complexed by the cations, such as for example Mg2+ cations. Following their release into an aqueous environment, the respective protein-functionalized synthetic extracellular vesicles can interact with target cells thus influencing their functions, such as metabolism, proliferation, or growth.
The synthetic extracellular vesicles obtained by the highly controlled droplet-stabilized assembly provide a robust platform for therapeutic applications and moreover allow getting new insights into fundamental functioning-principles of extracellular vesicles.
In comparison with the prior art methods, a first advantage of the invention is to provide extracellular vesicles with high stability (
The assembly in polymer shell stabilized vesicles allows also encapsulation of nucleic acids at high efficiency (
Moreover, the inventive method allows adjusting the vesicle dimensions by regulating the emulsification speed (Example 2), which is an important factor influencing the activity of the vesicles (
To notice, the emulsification process allows reaching throughput rates much higher than the throughput rates allowed by microfluidic techniques.
Importantly, the use of emulsification to produce synthetic extracellular vesicles has never been suggested in the prior art so far.
The method also allows obtaining extracellular vesicles preparations with high purity (
Moreover, the inventive method allows design and assembly of fully synthetic extracellular vesicles by a polymer shell-stabilized approach that hold a higher therapeutic potential as their laboriously isolated natural analogues (Examples 5-12,
Moreover, the inventive method for bottom-up assembly of extracellular vesicles, allows controlling the quantity of each individual extracellular vesicle components, which is an essential aspect for therapeutic applications, and also to decipher their roles on disease related states, representing an essential advantage in comparison with natural exosomes.
Non-limiting examples of the extracellular vesicle types are from the group of vesicles that include an exosome, a microvesicle, an apoptotic vesicle, and a liposome.
In particular, the present invention is directed to a method for producing synthetic extracellular vesicles comprising:
The present invention is also directed to a method for producing synthetic extracellular vesicles comprising:
According to an aspect of the present invention, the method further comprises after step d) the following steps:
According to a more particular aspect of the present invention, the water phase of step a) comprises at least one lipid coupled to a functional ligand selected from biotin, N-hydroxysuccinimide ester, N-hydroxysulfosuccinimide, nitrilotriacetic acid-nickel, amine, carboxylic acid, maleimide, aromatic maleimid, dithiopyridinyl, pyridyl disulfide, pyridyldithiopropionate, N-benzylguanine, cyanuric chloride, carboxyacyl, cyanur, folate, square, galloyl, glycan, thiol, arginylglycylaspartic acid, a fluorescent dye molecule, a magnetic resonance imaging reagent, a chelator; and
According to a still more particular aspect of the present invention, the water phase of step a) comprises one or more nucleic acid molecules selected from the group comprising miRNA molecules miR-17, miR-18a, miR-19a, miR-19b-1, miR-20a, miR-92a; miR-21, miR-30d-5p, miR-33b, miR-124, miR-125, miR-126, miR-130, miR-132, miR-133b, miR-140-5p, miR-191, miR-222, miR-451, miR-494, miR-575, miR-630, miR-638, miR-1202, miR-1207-5p, miR-1225-5p, miR-1268, miR-6087, miR-92a-3p-e, miR-K12-3, let-7a.
Further miRNA molecules suitable for the method and the synthetic extracellular vesicles disclosed herein are listed in Table 3.
According to a still more particular aspect of the present invention, the extracellular vesicle associated protein, or a fragment thereof, is selected from the group comprising:
Further extracellular vesicle associated proteins suitable for the method and the synthetic extracellular vesicles disclosed herein are listed in Table 4.
According to a still more particular aspect of the present invention, the water phase of step a) comprises at least two lipids selected from the group comprising:
According to a further aspect of the present invention, step d) comprises producing polymer shell stabilized synthetic extracellular vesicles by emulsifying the combined phases at step c) using a mechanic or electronic emulsifier for at least 5 seconds at speed higher than 1,000 rpm.
A preferred embodiment of the present invention is directed to a synthetic extracellular vesicle having a hydrodynamic radius between 70 nm and 5000 nm, comprising:
A further preferred embodiment of the present invention is directed to a synthetic extracellular vesicle having a hydrodynamic radius between 70 nm and 5000 nm, comprising:
A particularly preferred embodiment of the present invention is directed to a synthetic extracellular vesicle having a hydrodynamic radius between 70 nm and 5000 nm, with the composition described above and specifically comprising:
Another particularly preferred embodiment of the present invention is directed to a synthetic extracellular vesicle having a hydrodynamic radius between 70 nm and 5000 nm, with the composition described above and specifically comprising:
Another particularly preferred embodiment of the present invention is directed to a synthetic extracellular vesicle having a hydrodynamic radius between 70 nm and 5000 nm, with the composition described above and specifically comprising:
Another particularly preferred embodiment of the present invention is directed to a synthetic extracellular vesicle having a hydrodynamic radius between 70 nm and 5000 nm, with the composition described above and specifically comprising:
Another particularly preferred embodiment of the present invention is directed to a synthetic extracellular vesicle having a hydrodynamic radius between 70 nm and 5000 nm, with the composition described above and specifically comprising:
A further particularly preferred embodiment of the present invention is directed to a synthetic extracellular vesicle having a hydrodynamic radius between 70 nm and 5000 nm, with the composition described above and specifically comprising:
A more particularly preferred embodiment of the present invention is directed to a synthetic extracellular vesicle as described above for use in the treatment of a disorder selected from the group comprising inflammation, cancer, rheumatic disorder, severe graft versus host disease, osteoarthritis, cardiovascular disorder, epithelial diseases, neurodegenerative disorders, autoimmune disorders, bone and cartilage disorders, osteoporosis, renal osteodystrophy, Paget's disease of bone, osteopetrosis, rickets, neurological disorders, intoxication, neuroendocrinology disorders, endocrinology disorders, genetic disorders, infectious diseases, dental disorders, cosmetic procedures, coagulation disorders, dermatoses, diabetes, age-associated disorders.
Unless specifically noted, the embodiments describing “cell-derived vesicles” or “extracellular vesicles” shall include “exosomes”, “liposomes”, “microvesicles” and “apoptotic vesicles” alone or in combination. When the term “exosome” is used as an example, it is understood that liposomes and microvesicles can be substituted therein.
As used herein, the term “extracellular vesicle” refers to a cell-derived vesicle comprising a membrane that encloses an internal space. Extracellular vesicles comprise all membrane-bound vesicles that have a smaller diameter (here determined as hydrodynamic radius) than the cell from which they are derived. Generally extracellular vesicles range in diameter (hydrodynamic radius) from 20 nm to 1000 nm, and can comprise various macromolecular cargo either within the internal space, displayed on the external surface of the extracellular vesicle, and/or spanning the membrane. The cargo can comprise nucleic acids, proteins, carbohydrates, lipids, small molecules, and/or combinations thereof. Two types of extracellular vesicles are exosomes and microvesicles.
As used herein the term “exosome” refers to a cell-derived small (between 20-300 nm in diameter or hydrodynamic radius, more preferably 20-1000 nm in diameter or hydrodynamic radius) vesicle comprising a membrane that encloses an internal space, and which is generated from the cell by direct plasma membrane budding or by fusion of the late endosome with the plasma membrane. The exosome is a species of extracellular vesicle. The exosome comprises lipid or fatty acid and proteins and optionally comprises a), a polynucleotide (e.g., a nucleic acid, RNA, or DNA), a sugar (e.g., a simple sugar, polysaccharide, or glycan), a functional agent (e.g., a therapeutic agent) or other molecules. The exosome can be derived from a producer cell using a technique known in the prior art, and isolated from the producer cell based on its size, density, biochemical parameters, or a combination thereof.
Microvesicles, on the other hand, are released from a cell upon direct budding from the plasma membrane (PM). Microvesicles are typically larger than exosomes and range from approximately 100 nm to 1 μm.
Although these two vesicle-types, microvesicles, and exosomes, are separate classes of vesicles, due to the fact that they overlap in size, and since the commonly used non-specific protocols for exosome isolation and purification rely solely on the vesicle size differences, it is a fact that in many of the reports published, the exosome samples used are impure, since they probably also include microvesicles and large protein aggregates. Because of this, it has been proposed that the term “extracellular vesicles” (EVs) be used as a general term for all small vesicles/particles, including both vesicle types, and also apoptotic bodies or vesicles.
As used herein the term “synthetic exosome” refers to a synthetic exosome that is not secreted, released, or otherwise produced by a cell in vitro or in vivo. As used herein the term “synthetic exosome” refers to a synthetic exosome generated synthetically from a starting lipid mixture, into which one or more polypeptides and/or nucleic acids may be incorporated. Similarly, the term “synthetic extracellular vesicle” refers to a synthetic extracellular vesicle that is not secreted, released, or otherwise produced by a cell in vitro or in vivo. As used herein the term “synthetic extracellular vesicle” refers to a synthetic extracellular vesicle generated synthetically from a starting lipid mixture, into which one or more polypeptides and/or nucleic acids may be incorporated.
“Liposomes” are microscopic vesicles consisting of concentric lipid bilayers. Structurally, liposomes range in size and shape from long tubes to spheres, with dimensions from a few hundred Angstroms to fractions of a millimeter. Vesicle-forming lipids are selected to achieve a specified degree of fluidity or rigidity of the final complex providing the lipid composition of the outer layer.
“Apoptotic bodies” or “apoptotic vesicles” are released during cell death (apoptosis) and are heterogeneously shaped vesicles with sizes between 50-5000 nm. They are formed from the plasma membrane, and they contain DNA, RNA, histones, and signalling molecules. They usually have high amounts of phosphatidylserine in their membranes, since the outer membrane of apoptotic cells is enriched in PS.
“Membrane” as used herein comprises a lipid bilayer that separates an interior space from an exterior space and comprises one or more biological compounds, typically lipids, and optionally polypeptides and/or carbohydrates such as glycan and/or nucleic acids, and/or other macromolecules. In some embodiments, the membrane comprises lipids and fatty acids. In some embodiments, the membrane comprises phospholipids, glycolipids, fatty acids, sphingolipids, phosphoglycerides, sterols, cholesterols, and phosphatidylserines. The extracellular vesicle comprises a membrane as defined herein.
In some embodiments, the extracellular vesicle or exosome further comprises one or more macromolecule in their lumen.
The term “macromolecule” as used herein is selected from the group comprising an extracellular vesicle associated protein, a carbohydrate, a nucleic acid, a polypeptide, a cell receptor, an imaging probe.
As used herein, the term “homogeneous” in reference to a population of extracellular vesicles refers to population of vesicles that have the same or a similar amount of one or more proteins, or one or more nucleic acid molecules, or one or more macromolecule. A homogenous population is one wherein about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, about 98%, or 100% of the vesicles share the one or more proteins, or one or more nucleic acid molecules, or one or more macromolecule.
As used herein, the term “heterogeneous” in reference to a population of engineered vesicles refers to population of vesicles that have differing identity or differing amount of one or more proteins, or one or more nucleic acid molecules, or one or more macromolecule.
Moreover, as used herein, the term “homogeneous” in reference to a population of extracellular vesicles also refers to population of vesicles that have the same or a similar size. A homogenous population in size is one wherein the coefficient of variation calculated as [(standard deviation/average size)*100] is lower than 15%, preferably lower than 13%, preferably lower than 10%, preferably lower than 8%, preferably lower than 7%, most preferably lower than 5%.
The extracellular vesicle or exosome can interact with the target cell via membrane fusion and deliver nucleic acid molecules or intracellular proteins or functional proteins to the surface or cytoplasm of a target cell. In some embodiments, membrane fusion occurs between the extracellular vesicle or exosome and the plasma membrane of a target cell. In other embodiments, membrane fusion occurs between the extracellular vesicle or exosome and an endosomal membrane of a target cell.
As used herein, the term “modulate”, “modulating”, “modify”, and/or “modulator” generally refers to the ability to alter, by increase or decrease, e.g., directly or indirectly promoting, stimulating, up-regulating or interfering with/inhibiting/down-regulating a specific concentration, level, expression, function or behaviour, such as, e.g., to act as an antagonist or agonist. In some instances, a modulator can increase and/or decrease a certain concentration, level, activity or function relative to a control, or relative to the average level of activity that would generally be expected or relative to a control level of activity.
In some embodiments, the extracellular vesicle has a hydrodynamic radius between 20-5000 nm, such as between about 20-150 nm, 20-500 nm, 20-1000 nm, 20-2000, nm, 20-3000 nm, 20-4000 nm, 20-5000 nm, 30-150 nm, 30-500 nm, 30-1000 nm, 30-2000, nm, 30-3000 nm, 30-4000 nm, 30-5000 nm, 40-150 nm, 40-500 nm, 40-1000 nm, 40-2000, nm, 40-3000 nm, 40-4000 nm, 40-5000 nm, 70-2000, nm, 70-3000 nm, 70-4000 nm, 70-5000 nm, 50-150 nm, 50-500 nm, 50-1000 nm, 50-2000, nm, 50-3000 nm, 50-4000 nm, 50-5000 nm, 100-150 nm, 100-500 nm, 100-1000 nm, 100-2000, nm, 100-3000 nm, 100-4000 nm, 100-5000 nm, 500-1000 nm, 500-2000, nm, 500-3000 nm, 500-4000 nm, 500-5000 nm.
In other embodiments, the extracellular vesicle has a hydrodynamic radius between about 20-1000 nm, such as between about 20-100 nm, 20-200 nm, 20-300 nm, 20-400 nm, 20-500 nm, 20-600 nm, 20-700 nm, 20-800 nm, 20-900 nm, 30-100 nm, 30-200 nm, 30-300 nm, 30-400 nm, 30-500 nm, 30-600 nm, 30-700 nm, 30-800 nm, 30-900 nm, 40-100 nm, 40-200 nm, 40-300 nm, 40-400 nm, 40-500 nm, 40-600 nm, 40-700 nm, 40-800 nm, 40-900 nm, 50-150 nm, 50-500 nm, 50-750 nm, 100-200 nm, 100-500 nm, or 500-1000 nm.
In another embodiment, a population of the extracellular vesicles described herein comprise a population wherein 90% of the extracellular vesicles have a hydrodynamic radius 20-5000 nm. In another embodiment, a population of the extracellular vesicles described herein comprise a population wherein 95% of the extracellular vesicles have a hydrodynamic radius 20-5000 nm. In another embodiment, a population of the extracellular vesicles described herein comprise a population wherein 99% of the extracellular vesicles have a hydrodynamic radius 20-5000 nm. In another embodiment, a population of the extracellular vesicles described herein comprise a population wherein 90% of the extracellular vesicles have a hydrodynamic radius 20-1000 nm. In another embodiment, a population of the extracellular vesicles described herein comprise a population wherein 95% of the extracellular vesicles have a hydrodynamic radius 20-1000 nm. In another embodiment, a population of the extracellular vesicles described herein comprise a population wherein 99% of the extracellular vesicles have a hydrodynamic radius 20-1000 nm. In another embodiment, a population of the extracellular vesicles described herein comprise a population wherein 90% of the extracellular vesicles have a hydrodynamic radius 20-500 nm. In another embodiment, a population of the extracellular vesicles described herein comprise a population wherein 95% of the extracellular vesicles have a hydrodynamic radius 20-500 nm. In another embodiment, a population of the extracellular vesicles described herein comprise a population wherein 99% of the extracellular vesicles have a hydrodynamic radius 20-500 nm.
In certain embodiments, the extracellular vesicle is an exosome. In certain embodiments, the extracellular vesicle is a microvesicle.
In some embodiments, the exosome has a hydrodynamic radius between about 20-5000 nm, such as between about 20-150 nm, 20-500 nm, 20-1000 nm, 20-2000, nm, 20-3000 nm, 20-4000 nm, 20-5000 nm, 30-150 nm, 30-500 nm, 30-1000 nm, 30-2000, nm, 30-3000 nm, 30-4000 nm, 30-5000 nm, 40-150 nm, 40-500 nm, 40-1000 nm, 40-2000, nm, 40-3000 nm, 40-4000 nm, 40-5000 nm, 50-150 nm, 50-500 nm, 50-1000 nm, 50-2000, nm, 50-3000 nm, 50-4000 nm, 50-5000 nm, 70-2000, nm, 70-3000 nm, 70-4000 nm, 70-5000 nm, 100-150 nm, 100-500 nm, 100-1000 nm, 100-2000, nm, 100-3000 nm, 100-4000 nm, 100-5000 nm, 500-1000 nm, 500-2000, nm, 500-3000 nm, 500-4000 nm, 500-5000 nm.
In other embodiments, the exosome has a hydrodynamic radius between about 20-1000 nm, such as between about 20-100 nm, 20-200 nm, 20-300 nm, 20-400 nm, 20-500 nm, 20-600 nm, 20-700 nm, 20-800 nm, 20-900 nm, 30-100 nm, 30-200 nm, 30-300 nm, 30-400 nm, 30-500 nm, 30-600 nm, 30-700 nm, 30-800 nm, 30-900 nm, 40-100 nm, 40-200 nm, 40-300 nm, 40-400 nm, 40-500 nm, 40-600 nm, 40-700 nm, 40-800 nm, 40-900 nm, 50-150 nm, 50-500 nm, 50-750 nm, 100-200 nm, 100-500 nm, or 500-1000 nm.
In another embodiment, a population of the exosomes described herein comprise a population wherein 90% of the exosomes have a hydrodynamic radius 20-5000 nm. In another embodiment, a population of the exosomes described herein comprise a population wherein 95% of the exosomes have a hydrodynamic radius 20-5000 nm. In another embodiment, a population of the exosomes described herein comprise a population wherein 99% of the exosomes have a hydrodynamic radius 20-5000 nm. In another embodiment, a population of the exosomes described herein comprise a population wherein 90% of the exosomes have a hydrodynamic radius 20-1000 nm. In another embodiment, a population of the exosomes described herein comprise a population wherein 95% of the exosomes have a hydrodynamic radius 20-1000 nm. In another embodiment, a population of the exosomes described herein comprise a population wherein 99% of the exosomes have a hydrodynamic radius 20-1000 nm. In another embodiment, a population of the exosomes described herein comprise a population wherein 90% of the exosomes have a hydrodynamic radius 20-500 nm. In another embodiment, a population of the exosomes described herein comprise a population wherein 95% of the exosomes have a hydrodynamic radius 20-500 nm. In another embodiment, a population of the exosomes described herein comprise a population wherein 99% of the exosomes have a hydrodynamic radius 20-500 nm.
The present invention is directed to a method for producing synthetic extracellular vesicles comprising:
The present invention is further directed to a method for producing synthetic extracellular vesicles comprising:
In certain embodiments, the extracellular vesicle is an exosome. In certain embodiments, the extracellular vesicle is a microvesicle.
Therefore, a more particular embodiment of the invention is directed to a method for producing a synthetic exosome comprising:
Another still more particular embodiment of the invention is also directed to a method for producing a synthetic exosome comprising:
In accordance with optional step d′), the polymer shell is removed from the polymer shell-stabilized synthetic extracellular vesicles. Since the polymer shell is not necessary any more after assembling the vesicles with all the required components, it is actually preferred to perform the step d′) so as to obtain the synthetic extracellular vesicles into an aqueous phase.
The inventors have found that the synthetic extracellular vesicles can be efficiently released from the polymer shell by adding a deemulsifier surfactant to the polymer shell stabilized synthetic extracellular vesicles formed after emulsification. The deemulsifier surfactant destabilizes the structure of the surrounding polymer shell and thus, allows releasing the synthetic extracellular vesicles from the polymer shell into an aqueous buffer, also named “release buffer”.
The deemulsifier surfactant is preferably selected from the group comprising 1H, 1H,2H,2H-perfluoro-1-octanol; 1H, 1H-perfluoro-1-pentanol; 1H, 1H-perfluor-1-octanol; 1H, 1H, 8H-perfluoro-1-octanol.
The deemulsifier surfactant is preferentially added at a ratio ranging from 1:1 to 10:1 with the intraluminal buffer (also named production buffer).
Thereafter, the synthetic extracellular vesicles are usually centrifuged after release from the polymer shell to allow purification from vesicles of unwanted dimensions and other impurities.
The centrifugation can be performed for a time comprised between 5-60 min and at acceleration comprised between 800 g-100,000 g depending on the dimension of the synthetic extracellular vesicles of interest.
Moreover, for synthetic extracellular vesicles with hydrodynamic radius comprised between 100-1000 nm, the centrifugation is preferentially performed at acceleration comprised between 30,000-60,000 g and a time comprised between 10-60 min. For synthetic extracellular vesicles with hydrodynamic radius comprised between 1000-3000 nm, the centrifugation is preferentially performed at acceleration comprised between 10,000-30,000 g and a time comprised between 10-60 min. For synthetic extracellular vesicles with hydrodynamic radius comprised between 3000-5000 nm, the centrifugation is preferentially performed at acceleration comprised between 5,000-20,000 g and a time comprised between 10-60 min.
The synthetic extracellular vesicles synthetized following the inventive method, released into an aqueous medium and then purified by centrifugation (
Thus, present invention is directed to method for producing synthetic extracellular vesicles comprising:
A particular embodiment of the invention is directed to a method for producing synthetic extracellular vesicles comprising:
A more particular embodiment of the invention is directed to a method for producing synthetic extracellular vesicles comprising:
A more particular embodiment of the invention is directed to a method for producing synthetic extracellular vesicles comprising:
A more particular embodiment of the invention is directed to a method for producing synthetic extracellular vesicles comprising:
A more particular embodiment of the invention is directed to a method for producing synthetic extracellular vesicles comprising:
In the inventive methods, the synthetic extracellular vesicles can be decorated with the proteins of interest after release into an aqueous solution, as described above, wherein the proteins are preferably known to be associated with extracellular vesicles.
The proteins can be coupled to the synthetic extracellular vesicles by applying bio-orthogonal surface chemistry such as N-hydroxysuccinimide ester and/or NTA-poly-histidine tag coupling, or they can be added to the water phase of step a) or can be integrated into or on the polymer shell stabilized synthetic extracellular vesicles by microfluidic technology such as pico-injection.
These procedures allow obtaining synthetic extracellular vesicles comprising proteins at a well-defined ratio protein:lipid, and with very low degree of variation in protein composition between different batches.
Preferred protein:lipid ratios as used herein are between 1:20 to 1:100, 1:40 to 1:100, 1:50 to 1:100, 1:20 to 1:200, 1:40 to 1:200, 1:50 to 1:200, 1:75 to 1:200.
When assessing the exosome protein content, the inventor found that K562-derived exosomes isolated from conditioned media and those provided by a commercial distributer, differed greatly in their protein content, underscoring the degree of variation between different vesicle preparation methods (
Important to mention, by applying bio-orthogonal surface chemistry such as N-hydroxysuccinimide ester and/or NTA-poly-histidine tag coupling, or by using microfluidic technologies, the protein to lipid ratio can be precisely adjusted. This ratio is also very homogenous among the vesicle population.
Thus, the inventive synthetic extracellular vesicles show an outstanding improvement in comparison to the non-adjustable extracellular vesicles obtained by prior art methods, such as differential centrifugation of cell culture medium, or membrane fragmentation of engineered cells.
The wording “extracellular vesicle associated protein” refers to proteins that are enriched in exosomes and extracellular vesicles in comparison to cells. Therefore “extracellular vesicle associated proteins” can also be used as marker of exosomes or other extracellular vesicles. Thus the term “extracellular vesicle associated proteins” has the same meaning as “extracellular vesicle protein marker” or “extracellular vesicle marker”.
Therefore, in specific embodiments, the extracellular vesicles or exosomes comprise one or more proteins on their surface or in their lumen, wherein said proteins are selected from a group of proteins that was recently identified to be enriched on the surface or inside extracellular vesicles, and were thus defined as “extracellular vesicle associated proteins” (Thery et al., 2018, Minimal information for studies of extracellular vesicles 2018; Exocarta Top100 proteins). A list of extracellular vesicle associated proteins suitable for the method and the extracellular vesicles disclosed herein are listed in Table 4.
As used herein the term “fragment” or “active fragment” of a protein refers to a fragment of that protein that retains the ability to be specifically coupled to the extracellular vesicle or exosome. The term “fragment” or “active fragment” of a protein also refers to a fragment of that protein that retains the ability to exert its function in the target cell.
For example, in the case of membrane proteins, a protein fragment refers to a cytosolic domain, a transmembrane domain or an extracellular domain of said protein.
For example, in the case of enzymes, a protein fragment refers to a catalytic domain of that enzyme.
For example, in the case of antibodies, a protein fragment refers to a fragment of the antibody that retains its capacity to bind specifically to the antigen. The antibody or antigen-binding fragment can be derived from natural sources, or partly or wholly synthetically produced. In some embodiments, the antibody is a monoclonal antibody. In some of these embodiments, the monoclonal antibody is an IgG antibody. In certain embodiments, the monoclonal antibody is an IgG1, IgG2, IgG3, or IgG4. In some other embodiments, the antibody is a polyclonal antibody. In certain embodiments, the antibody fragment, also named antigen-binding fragment, is selected from antigen-binding fragment (Fab), Fab′, and F(ab′)2, F(ab)2, a viable fragment (Fv), and Fd fragments. In certain embodiments, the antigen-binding fragment is a Single-chain variable fragment (scFv) or (ScFv)2 fragment. In certain other embodiments, the antibody or antigen-binding fragment is a single-domain antibody. In some embodiments, the antibody or antigen binding fragment is a bispecific or multispecific antibody.
For example, in the case of protein antigens, a protein fragment refers to a fragment of the antigen that retains its capacity to induce an immune response in a human or animal, and/or to be specifically recognized by an antibody.
Preferably, a suitable protein fragment of TSG101 (protein ID Q99816) comprises the amino acids 1-145, a suitable protein fragment of CD9 (protein ID P21926) comprises the amino acids 112-195, a suitable protein fragment of CD81 (protein ID P35762), comprises the amino acids 113-201, a suitable protein protein fragment of CD63 (protein ID P08962) comprises the amino acids 103-203, a suitable protein fragment of RANK (protein ID O35305) comprises the amino acids 31-214, a suitable protein fragment of FasL (protein ID NM_000639.1) comprises the amino acids 134-281, a suitable protein fragment of ICAM-1 (protein ID P05362) comprises the amino acids 1-480.
As used herein the term protein or a fragment thereof also include “variant” of a protein or of a fragment thereof, and refers to a protein or fragment that shares a certain amino acid sequence identity with the reference protein or fragment upon alignment by a method known in the art. A variant of a protein or of a fragment thereof can include a substitution, insertion, deletion, frameshift or rearrangement in another protein. In some embodiments variants share at least 70%, 80%, 85%, 90%, 95% or 99% sequence identity with the reference protein or with the fragment thereof.
Recitation of any protein provided herein encompasses a functional variant of the protein. The term “functional variant” of a protein refers to a variant of the protein that retains the ability to be specifically targeted to exosomes.
The percentage of “sequence identity” is determined by comparing two optimally aligned protein or polypeptide sequences over a “comparison window” on the full length of the reference sequence. A “comparison window” as used herein, refers to the optimal alignment between the reference and variant sequence after that the two sequences are optimally aligned, wherein the variant nucleic acid or polypeptide sequence in the comparison window may comprise additions or deletions (i.e., gaps) of 20 percent or less, usually 5 to 15 percent, or 10 to 12 percent, as compared to the reference sequences (which does not comprise additions or deletions) for optimal alignment. Identity percentage is calculated by determining the number of positions at which the identical amino acid residues occurs in both sequences to yield the number of matched positions, dividing the number of matched positions by the total number of positions in the reference sequence (i.e., the full length in amino acid or nucleotide) and multiplying the results by 100 to yield the percentage of sequence identity. Two protein or polypeptide sequences are said to be “identical” if the sequence of nucleotides or amino acids in the two sequences is the same when optimally aligned as described above.
The percentage of “sequence identity” can be determined on the comparison window defined above with the help of blastp with the “BLAST 2 Sequences” tool available at the NCBI website. (Tatusova A. et al., FEMS Microbiol Lett. 1999, 174:247-250).
Alternatively, a variant sequence may also be any amino acid sequence resulting from allowed substitutions at any number of positions of the parent sequence according to the formula below:
According to the present invention, the extracellular vesicle associated protein is preferentially selected from the group comprising:
Therefore, one embodiment of the present invention is directed to a method for producing synthetic extracellular vesicles comprising:
A further embodiment of the present invention is directed to a method for producing synthetic extracellular vesicles comprising:
Therefore, one embodiment of the present invention is directed to a method for producing synthetic extracellular vesicles comprising:
A further embodiment of the present invention is directed to a method for producing synthetic extracellular vesicles comprising:
Further extracellular vesicle associated proteins suitable for the method and the synthetic extracellular vesicles disclosed herein are listed in Table 4.
For bio-orthogonal surface chemistry, suitable functional ligands are selected from the group comprising biotin, N-hydroxysuccinimide ester, N-hydroxysulfosuccinimide, nitrilotriacetic acid-nickel, amine, carboxylic acid, maleimide, aromatic maleimid, dithiopyridinyl, pyridyl disulfide, pyridyldithiopropionate, N-benzylguanine, cyanuric chloride, carboxyacyl, cyanur, folate, square, galloyl, glycan, thiol, arginylglycylaspartic acid, a fluorescent dye molecule, a magnetic resonance imaging reagent, a chelator.
Table 2 lists the possible functional ligands that can be attached to the lipids, their function and the interacting moieties.
These functional ligands react with particular moieties at high affinity, as for example the ligand biotin reacts with the moiety streptavidin or avidin. Thus, proteins and other macromolecules of interest can be coupled to the surface of the released extracellular vesicles by using the interaction at high affinity between a functional ligand and the respective reacting moiety.
Thus, a particular embodiment of the invention is directed to a method for producing synthetic extracellular vesicles comprising:
A more particular embodiment of the invention is directed to a method for producing synthetic extracellular vesicles comprising:
Thus, a particular embodiment of the invention is directed to a method for producing synthetic extracellular vesicles comprising:
A more particular embodiment of the invention is directed to a method for producing synthetic extracellular vesicles comprising:
The phrase “nucleic acid molecule” refers to a single or double-stranded polymer of deoxyribonucleotide or ribonucleotide bases. It includes chromosomal DNA and self-replicating plasmids, vectors, DNA, cDNA, mRNA, siRNA, antisense nucleotide sequence, shRNA, piRNA, snRNA, lncRNA, PNA, left handed DNA, Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR) guide RNA.
Abbreviations used in this application include the following: “mRNA” refers to messenger RNA, “miRNA” refers to microRNA, “siRNA” refers to small interfering RNA, “antisense nucleotide sequence” refers to a single stranded sequence that is complementary to a nucleotide sequence of interest, “shRNA” refers to small or short hairpin RNA, “lncRNA” refers to long non-coding RNA, and “dsDNA” refers to double stranded DNA.
As used herein, the term “microRNAs” or “miRNAs” refers to post-transcriptional regulators that typically bind to complementary sequences in the three prime untranslated regions (3′ UTRs) of target messenger RNA transcripts (mRNAs), usually resulting in gene silencing. Typically, miRNAs are short, non-coding ribonucleic acid (RNA) molecules, for example, 21 or 22 nucleotides long. The terms “microRNA” and “miRNA” are used interchangeably.
The content of miRNA molecules is preferably comprised between 75 pg/1012 vesicles-1000 pg/1012 vesicles, between 75 pg/1012 vesicles-5000 pg/1012 vesicles, 75 pg/1012 vesicles-10,000 pg/1012 vesicles, between 75 pg/1012 vesicles-20,000 pg/1012 vesicle, between 75 pg/1012 vesicles-50,000 pg/1012 vesicles, between 75 pg/1012 vesicles-100,000 pg/1012 vesicles, between 75 pg/1012 vesicles-150,000 pg/1012 vesicles, between 75 pg/1012 vesicles-200,000 pg/1012 vesicles, between 75 pg/1012 vesicles-300,000 pg/1012 vesicles, between 500 pg/1012 vesicles-1000 pg/1012 vesicles, between 500 pg/1012 vesicles-5000 pg/1012 vesicles, 500 pg/1012 vesicles-10,000 pg/1012 vesicles, between 500 pg/1012 vesicles-20,000 pg/1012 vesicle, 500 pg/1012 vesicles-50,000 pg/1012 vesicles, between 500 pg/1012 vesicles-100,000 pg/1012 vesicles, between 500 pg/1012 vesicles-150,000 pg/1012 vesicles, between 500 pg/1012 vesicles-200,000 pg/1012 vesicles, between 500 pg/1012 vesicles-300,000 pg/1012 vesicles, between 5000 pg/1012 vesicles-10,000 pg/1012 vesicles, between 5000 pg/1012 vesicles-20,000 pg/1012 vesicle, 5000 pg/1012 vesicles-50,000 pg/1012 vesicles, between 5000 pg/1012 vesicles-100,000 pg/1012 vesicles, between 5000 pg/1012 vesicles-150,000 pg/1012 vesicles, between 5000 pg/1012 vesicles-200,000 pg/1012 vesicles, between 5000 pg/1012 vesicles-300,000 pg/1012 vesicles.
In one aspect, the therapeutic agent is a short interfering RNA, also known as siRNA. Methods to prepare and screen interfering RNA and select for the ability to block polynucleotide expression are known in the art and non-limiting examples of which are shown below. These interfering RNA are provided by this invention alone or in combination with a suitable vector or within a host cell. Compositions containing the RNAi are further provided. RNAi is useful to knock-out or knock-down select functions in a cell or tissue as known in the art.
siRNA sequences can be designed by obtaining the target mRNA sequence and determining an appropriate siRNA complementary sequence. siRNAs of the invention are designed to interact with a target sequence, meaning they complement a target sequence sufficiently to hybridize to that sequence. An siRNA can be 100% identical to the target sequence. However, homology of the siRNA sequence to the target sequence can be less than 100% as long as the siRNA can hybridize to the target sequence. Thus, for example, the siRNA molecule can be at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the target sequence or the complement of the target sequence. Therefore, siRNA molecules with insertions, deletions or single point mutations relative to a target may also be used. The generation of several different siRNA sequences per target mRNA is recommended to allow screening for the optimal target sequence. A homology search, such as a BLAST search, should be performed to ensure that the siRNA sequence does not contain homology to any known mammalian gene.
As a general guide, siRNAs that include one or more of the following conditions are particularly useful in gene silencing in mammalian cells:GC ratio of between 45-55%, no runs of more than 9 G/C residues, G/C at the 5′ end of the sense strand; NU at the 5′ end of the antisense strand; and at least 5 NU residues in the first 7 bases of the 5′ terminal of the antisense strand.
siRNA are, in general, from about 10 to about 30 nucleotides in length. For example, the siRNA can be 10-30 nucleotides long, 12-28 nucleotides long, 15-25 nucleotides long, 19-23 nucleotides long, or 21-23 nucleotides long. When an siRNA contains two strands of different lengths, the longer of the strands designates the length of the siRNA. In this situation, the unpaired nucleotides of the longer strand would form an overhang.
The term siRNA includes short hairpin RNAs (shRNAs). shRNAs comprise a single strand of RNA that forms a stem-loop structure, where the stem consists of the complementary sense and antisense strands that comprise a double-stranded siRNA, and the loop is a linker of varying size. The stem structure of shRNAs generally is from about 10 to about 30 nucleotides long. For example, the stem can be 10-30 nucleotides long, 12-28 nucleotides long, 15-25 nucleotides long, 19-23 nucleotides long, or 21-23 nucleotides long.
Tools to assist siRNA design are readily available to the public. For example, a computer-based siRNA design tool is available on the internet at www.dharmacon.com.
In some embodiments, the extracellular vesicle or exosome delivers their nucleic acid or intracellular protein or functional protein to a cell target. The delivery can occur in vitro or in a subject.
Preferentially, the extracellular vesicles disclosed herein include miRNA molecules selected from the group comprising miR-17, miR-18a, miR-19a, miR-19b-1, miR-20a, miR-92a; miR-21, miR-30d-5p, miR-33b, miR-124, miR-125, miR-126, miR-130, miR-132, miR-133b, miR-140-5p, miR-191, miR-222, miR-451, miR-494, miR-575, miR-630, miR-638, miR-1202, miR-1207-5p, miR-1225-5p, miR-1268, miR-6087, miR-92a-3p-e, miR-K12-3, let-7a.
Furthermore, the extracellular vesicles disclosed herein can include one or more miRNA molecules as listed in Table 3.
Thus, one embodiment of the present invention is directed to a method for producing synthetic extracellular vesicles comprising:
Another embodiment of the present invention is directed to a method for producing synthetic extracellular vesicles comprising:
A further embodiment of the present invention is directed to a method for producing synthetic extracellular vesicles comprising:
A more particular embodiment of the invention is directed to a method for producing synthetic extracellular vesicles comprising:
Lipids are the major scaffolding components of extracellular vesicles such as exosomes, and pivotal for their signaling capabilities. Therefore, synthetic extracellular vesicles were assembled with lipid compositions resembling those found in natural synthetic extracellular vesicles (
The lipid composition of the final synthetic extracellular vesicles can be easily fine-tuned on the basis of the composition of the initial lipid solution, as no lipid ratio change was observed during the emulsification and release procedures.
This technology also allows to finely regulating the charge of the synthetic extracellular vesicles by adjusting the ratio of cationic, neutral and anionic lipids.
The molar percentage (mol %) of a lipid is measured as the moles of a lipid of interest on the total lipid moles of the vesicle.
In some embodiments, the molar percentage (mol %) of a cationic lipid typically comprises from 0% to 10%, from 10% to 20%, from 10% to 30%, from 10% to 40%, %, from 10% to 50%, from 10% to 60%, from 20% to 30%, from 20% to 40%, from 20% to 50%, from 20% to 60% of the total lipid present in vesicle.
In some embodiments, the molar percentage (mol %) of an anionic lipid typically comprises from 0% to 10%, from 10% to 20%, from 10% to 30%, from 10% to 40%, %, from 10% to 50%, from 10% to 60%, from 20% to 30%, from 20% to 40%, from 20% to 50%, from 20% to 60% of the total lipid present in vesicle.
In some embodiments, the molar percentage (mol %) of neutral lipid typically comprises from 49% to 99%, from 49% to 89%, from 49% to 79%, from 49% to 69%, %, from 59% to 99%, from 59% to 89%, from 59% to 79%, from 59% to 69% of the total lipid present in vesicle.
The present invention is not particularly limited concerning the chemical nature of the at least one lipid contained in the water phase of step a) and thus in the inner space of the polymer shell stabilized synthetic extracellular vesicle, as long as it is able to form a lipid bilayer. Good results are in particular achieved with phospholipids and in particular with a lipid being selected from the group comprising phosphocholine, phosphocholine derivatives, phosphoethanolamine, phosphoethanolamine derivatives, phosphatidylcholine, phosphatidylcholine derivatives, phosphatidylglycerol, phosphatidylglycerol derivatives and arbitrary combinations of two or more of the aforementioned lipids.
At least one of the lipids is an amphiphilic lipid, defined as having a hydrophilic and a hydrophobic portion, typically a hydrophilic head and a hydrophobic tail. The hydrophobic portion typically orients into a hydrophobic phase, e.g., within the bilayer, while the hydrophilic portion typically orients toward the aqueous phase, e.g., outside the bilayer, and possibly between adjacent apposed bilayer surfaces. The hydrophilic portion may comprise polar or charged groups such as carbohydrates, phosphate, carboxylic, sulfato, amino, sulfhydryl, nitro, hydroxy and other like groups. The hydrophobic portion may comprise apolar groups that include without limitation long chain saturated and unsaturated aliphatic hydrocarbon groups and groups substituted by one or more aromatic, cyclo-aliphatic or heterocyclic groups. Examples of amphipathic lipids include, but are not limited to, phospholipids, aminolipids and sphingolipids.
Typically, the lipids are phospholipids. Phospholipids include without limitation phosphatidylcholine, phosphatidylethanolamine, phosphatidylglycerol, phosphatidylinositol, phosphatidylserine, and their derivatives. It is to be understood that other lipid membrane components, such as cholesterol, sphingomyelin, cardiolipin, etc. may be used.
Lipids can be “uncharged lipids” or “charged lipids.” “Uncharged lipids” refer to lipids that do not carry any charged or ionizable groups such as phosphate groups or choline groups. Examples of uncharged lipids include, but are not limited to, diacyl glycerols and prostaglandins.
“Charged lipids” include neutrally charged, i.e. zwitterionic lipids, cationic lipids and anionic lipids. Generally, lipids bearing a net positive or negative charge exhibit poor solubility in oil phases.
Neutral lipids exist in an uncharged or neutral zwitterionic form at a selected pH.
“Zwitterionic lipids” carry both positively-charged groups and ionizable groups such as amino groups and choline groups that bear a net positive charge, and negatively-charged groups and ionizable groups, such as phosphates, sulfates and carboxylates. Examples of zwitterionic lipids include, but are not limited to, phosphorylcholine and phosphorylethanolamine
“Anionic lipids” are lipids negatively charged at physiological pH. “Cationic lipids” are lipids positively charged at physiological pH.
Further suitable lipids are pH sensitive lipids. A “pH-sensitive” lipid refers to a lipid whose ability to form and/or maintain formation of a lipid bilayer depends at least in part on the pH of the surrounding environment. Synthetic extracellular vesicles containing such lipids are destabilized under acidic conditions of the endocytotic pathway. Therefore, the encapsulated content is delivered into the intracellular bio-environment through destabilization or its fusion with the endosomal membrane.
Specific examples of the lipids suitable to synthetize the synthetic extracellular vesicles according to the method disclosed herein are listed in Table 1.
Preferably, the lipids are biodegradable in order to allow release of the internal proteins or nucleic acid molecules in vivo and/or in vitro. Biodegradable lipids include but are not limited to 1,2-dioleoyl-sn-glycero-3-phosphocholine (dioleoyl-phosphocholine, DOPC), anionic 1,2-di-(9Z-octadecenoyl)-sn-glycero-3-phospho-(1′-rac-glycerol) (dioleoyl-phosphoglycerol, DOPG), and 1,2-distearoyl-sn-glycero-3-phosphoethanolamine (distearoyl-phosphoethanolamine, DSPE).
According to an embodiment of the present invention, the at least one lipid comprised in the water phase of step a) is a lipid coupled with a functional ligand and/or with polyethylenglycol. Specific examples of the suitable functional ligands, the reacting moieties, and of the functionalized lipids containing are listed in Table 2.
Functionalized and non-functionalized lipids are available from a number of commercial sources including Avanti Polar Lipids (Alabaster, Ala.).
Sulfhydryls, also called thiols, exist in proteins in the side-chain of cysteine (Cys, C) amino acids. Sulfhydryl-reactive chemical groups include haloacetyls, maleimides, aziridines, acryloyls, arylating agents, vinylsulfones, pyridyl disulfides, TNB-thiols and disulfide reducing agents.
Different lipids which are offered for thioether conjugation contain maleimide, aromatic maleimides such as N-[4-(p-maleimidophenyl)-butyryl] (MPB) or 4-(N-maleimidomethyl)cyclohexane-1-carboxylate (MCC) group. The maleimide function group of MCC which contains an aliphatic cyclohexane ring is more stable toward hydrolysis in aqueous reaction environments rather than the aromatic phenyl group of MPB
Carbohydrates are selected from the group comprising β-galactose, α-mannose-, β-mannose-, and α-fucose. It has been shown that said carbohydrates can be conjugated to cholesterols to be incorporated into liposomes, and in vitro results showed that the sugar-conjugated liposomes are efficiently recognized by cells that overexpress carbohydrate-binding receptors on their surface (Rajabi and Mousa, 2016, Current Pharmaceutical Biotechnology, 17, 8).
SNAP-tag is a self-labeling protein tag commercially available in various expression vectors. SNAP-tag is a 182 residues polypeptide (19.4 kDa) that can be fused to any protein of interest and further specifically and covalently tagged with a suitable ligand, such as a fluorescent dye.
A functional ligand for coupling to lipids for carry out the present invention is preferably selected from the group comprising biotin, N-hydroxysuccinimide ester, nitrilotriacetic acid-nickel, amine, carboxylic acid, maleimides, dithiopyridinyl, pyridyl disulfide, pyridyldithiopropionate, N-benzylguanine, carboxyacyl, cyanur, folate, square, galloyl, glycan, thiol, arginylglycylaspartic acid, a fluorescent dye molecule, a magnetic resonance imaging reagent, a chelator.
Therefore, one embodiment of the present invention is directed to a method for producing synthetic extracellular vesicles comprising:
A further embodiment of the present invention is directed to a method for producing synthetic extracellular vesicles comprising:
A particular embodiment of the present invention is directed to a method for producing synthetic extracellular vesicles comprising:
A further particular embodiment of the present invention is directed to a method for producing synthetic extracellular vesicles comprising:
In order to allow a good dispersion of the polymer shell stabilized vesicles in the oil phase and in order to allow a good dispersion of the lipid containing aqueous phase within the polymer shell of the vesicle, it is preferred that the polymer shell is made of an amphiphilic copolymer with a hydrophobic end arranged at the outer side and a hydrophilic end arranged at the inner side of the polymer shell.
This may be achieved by forming the polymer shell of the extracellular vesicle, from a diblock copolymer, or a triblock copolymer, to form a water-in-oil droplet.
Good results are particularly obtained, if the polymer shell of the droplet is made of a diblock copolymer consisting of an hydrophobic block arranged at the outer side and a hydrophilic block arranged at the inner side of the polymer shell, or a triblock copolymer consisting of two hydrophobic polymer blocks and a hydrophilic polymer block, and wherein the oil phase comprises a fluorosurfactant triblock, so that the one or two hydrophobic polymer blocks are arranged at the outer side and the hydrophilic polymer block is arranged at the inner side of the polymer shell.
The hydrophobic block may be, but is not restricted to members, e.g. selected from the group consisting of perfluorinated polymers, such as perfluorinated polyethers, polystyrene or poly(olefin oxides), such as poly(propylene oxide), whereas the hydrophilic block may be selected e.g. from polyether glycols, polyetheramine, polyacrylate acid, polymethylacrylate acid or poly[poly(ethylene glycol) methyl ether methacrylate].
Likewise, good results are obtained, if the polymer shell of the droplet is made of a triblock copolymer consisting of two hydrophobic perfluorinated polymer end blocks and therebetween a hydrophilic polyether glycol block, wherein the triblock copolymer is folded so that the hydrophobic perfluorinated polymer blocks are arranged at the outer side and that the hydrophilic polyether glycol block is arranged at the inner side of the polymer shell. Examples for the hydrophobic blocks and the hydrophilic blocks are the same as those mentioned above.
Preferably, the perfluorinated polymer block is a perfluorinated polyether block (PFPE) and more preferably a perfluorinated polyether block having a weight average molecular weight of 1,000 to 10,000 g/mol. Likewise preferably, the polyether glycol (PEG) and polyetheramine (JEFFAMINE) blocks have preferably a weight average molecular weight of 100 to 50,000 g/mol. More specifically, suitable examples for the respective copolymers are PFPE-carboxylic acid (Krytox, MW 2500 or 7000 g/mol) and suitable examples for the respective diblock copolymers are PFPE (7000 g/mol)-PEG (1400 g/mol), PFPE (7000 g/mol)-PEG (600 g/mol), PFPE (2500 g/mol)-PEG (600 g/mol), PFPE (4000 g/mol)-PEG (600 g/mol), PFPE (4000 g/mol)-PEG (1400 g/mol), PFPE (2000 g/mol)-PEG (600 g/mol), PFPE (7000 g/mol)-JEFFAMINE (600 g/mol), PFPE (7000 g/mol)-JEFFAMINE (900 g/mol), PFPE (2500 g/mol)-JEFFAMINE (600 g/mol), PFPE (2500 g/mol)-JEFFAMINE (900 g/mol), PFPE (4000 g/mol)-JEFFAMINE (900 g/mol), PFPE (2500 g/mol)-JEFFAMINE (600 g/mol), PFPE (2000 g/mol)-JEFFAMINE (600 g/mol), PFPE (2000 g/mol)-JEFFAMINE (900 g/mol) and suitable examples for the respective triblock copolymers are PFPE (7000 g/mol)-PEG (1400 g/mol)-PFPE (7000 g/mol), PFPE (7000 g/mol)-PEG (600 g/mol)-PFPE (7000 g/mol), PFPE (4000 g/mol)-PEG (1400 g/mol)-PFPE (4000 g/mol) PFPE (2500 g/mol)-PEG (600 g/mol)-PFPE (2500 g/mol), PFPE (2000 g/mol)-PEG (600 g/mol)-PFPE (2000 g/mol), PFPE (7000 g/mol)-JEFFAMINE (900 g/mol)-PFPE (7000 g/mol) PFPE (7000 g/mol)-JEFFAMINE (600 g/mol)-PFPE (7000 g/mol), PFPE (4000 g/mol)-JEFFAMINE (900 g/mol)-PFPE (4000 g/mol), PFPE (4000 g/mol)-JEFFAMINE (600 g/mol)-PFPE (4000 g/mol), PFPE (2500 g/mol)-JEFFAMINE (900 g/mol)-PFPE (2500 g/mol), PFPE (2500 g/mol)-JEFFAMINE (600 g/mol)-PFPE (2500 g/mol), PFPE (2000 g/mol)-JEFFAMINE (900 g/mol)-PFPE (2000 g/mol) and PFPE (2000 g/mol)-JEFFAMINE (600 g/mol)-PFPE (2000 g/mol). The molecular weight is determined by gel permeation chromatography using a polystyrene standard.
Therefore, the present invention is directed to a method for producing synthetic extracellular vesicles comprising:
Moreover, the present invention is directed to a method for producing synthetic extracellular vesicles comprising:
In one embodiment, the present invention is directed to a method for producing synthetic extracellular vesicles comprising:
In one embodiment, the present invention is directed to a method for producing synthetic extracellular vesicles comprising:
One embodiment of the present invention is directed to a method for producing synthetic extracellular vesicles comprising:
Moreover, the present invention is directed to a method for producing synthetic extracellular vesicles comprising:
The emulsification procedure is usually performed with a mechanically or electronic emulsifier, for at least 5 seconds at speed higher than 1,000 rpm. This procedure holds the considerable advantage to regulate the vesicle dimension by changing the time and shear stress of emulsification. As shown in Example 2, synthetic extracellular vesicles radii between 292 nm±12 nm, (coefficient of variation (CV)=4.1%; n=3) were obtained by emulsification for 30 sec at 30,000 rpm and radii of 627 nm±15 nm, (CV=2.4%; n=3) were obtained by emulsification for 30 sec at 14,000 rpm.
Notably, this procedure allowed obtaining extracellular vesicles very homogenous in size, as the coefficient of variation of the synthetized extracellular vesicles varied between 2.4%, for vesicles of size 292 nm±12 nm, and 4.1% for vesicles of size 627 nm±15 nm, which are variation levels much lower than observed in the natural exosome samples. Indeed the variation value of commercial K562 exosomes was CV=42.5% for dimensions 468 nm±199 nm, (n=3), and that of exosomes isolated from conditioned K562 cell culture medium was CV=13.3%, for dimensions 240 nm±32 nm (n=3).
Another embodiment of the present invention is directed to a method for producing synthetic extracellular vesicles comprising:
Another particular embodiment of the present invention is directed to a method for producing synthetic extracellular vesicles comprising:
A further embodiment of the present invention is directed to a method for producing synthetic extracellular vesicles comprising:
A further particular embodiment of the present invention is directed to a method for producing synthetic extracellular vesicles comprising:
A particular embodiment of the present invention is directed to a synthetic extracellular vesicle having a diameter between 70 nm and 5000 nm, comprising:
A more particular embodiment of the present invention is directed to a synthetic extracellular vesicle having a hydrodynamic radius between 70 nm and 5000 nm, comprising:
A still more particular embodiment of the present invention is directed to a synthetic extracellular vesicle having a hydrodynamic radius between 70 nm and 5000 nm, comprising:
As mentioned above, in certain embodiments, the synthetic extracellular vesicle is an exosome. In certain embodiments, the synthetic extracellular vesicle is a microvesicle.
A particularly preferred embodiment of the present invention is directed to a synthetic extracellular vesicle between 70 nm and 5000 nm with the composition described above and specifically comprising:
Another particularly preferred embodiment of the present invention is directed to a synthetic extracellular vesicle between 70 nm and 5000 nm with the composition described above and specifically comprising:
Another particularly preferred embodiment of the present invention is directed to a synthetic extracellular vesicle between 70 nm and 5000 nm with the composition described above and specifically comprising:
Another particularly preferred embodiment of the present invention is directed to a synthetic extracellular vesicle between 70 nm and 5000 nm with the composition described above and specifically comprising:
Another particularly preferred embodiment of the present invention is directed to a synthetic extracellular vesicle between 70 nm and 5000 nm with the composition described above and specifically comprising:
Another particularly preferred embodiment of the present invention is directed to a synthetic extracellular vesicle between 70 nm and 5000 nm specifically comprising:
A further particularly preferred embodiment of the present invention is directed to a synthetic extracellular vesicle between 70 nm and 5000 nm specifically comprising:
A further more particularly preferred embodiment of the present invention is directed to a synthetic extracellular vesicle between 70 nm and 5000 nm specifically comprising:
The examples of the present invention show that the synthetic extracellular vesicles are able to deliver their protein and nucleic acid contents into target cells, thus affecting their gene expression, protein expression, signalling pathways and metabolism.
Thus, the inventive synthetic extracellular pathways can be used for therapy of a wide range of disorders by acting at cellular levels.
For example, it has been here shown that the synthetic extracellular vesicles resembling those of fibrocyte origin can stimulate epithelial cell proliferation, migration, and collagen deposition, ultimately leading to wound healing.
Therefore, one embodiment of the present invention is directed to a synthetic extracellular vesicle having a hydrodynamic radius between 70 nm and 5000 nm, comprising:
A particular embodiment of the present invention is directed to a synthetic extracellular vesicle having a hydrodynamic radius between 70 nm and 5000 nm, comprising:
A more particular embodiment of the present invention is directed to a synthetic extracellular vesicle having a hydrodynamic radius between 70 nm and 5000 nm, comprising:
In certain embodiments, the extracellular vesicle is an exosome. In certain embodiments, the extracellular vesicle is a microvesicle.
One preferred embodiment of the present invention is directed to a synthetic extracellular vesicle between 70 nm and 5000 nm specifically comprising:
A preferred embodiment of the present invention is directed to a synthetic extracellular vesicle between 70 nm and 5000 nm specifically comprising:
A particularly preferred embodiment of the present invention is directed to a synthetic extracellular vesicle between 70 nm and 5000 nm specifically comprising:
A particularly preferred embodiment of the present invention is directed to a synthetic extracellular vesicle between 70 nm and 5000 nm specifically comprising:
Another particularly preferred embodiment of the present invention is directed to a synthetic extracellular vesicle between 70 nm and 5000 nm specifically comprising:
Another particularly preferred embodiment of the present invention is directed to a synthetic extracellular vesicle between 70 nm and 5000 nm specifically comprising:
Another particularly preferred embodiment of the present invention is directed to a synthetic extracellular vesicle between 70 nm and 5000 nm specifically comprising:
Another particularly preferred embodiment of the present invention is directed to a synthetic extracellular vesicle between 70 nm and 5000 nm specifically comprising:
Another particularly preferred embodiment of the present invention is directed to a synthetic extracellular vesicle between 70 nm and 5000 nm specifically comprising:
Another more particularly preferred embodiment of the present invention is directed to a synthetic extracellular vesicle between 70 nm and 5000 nm specifically comprising:
Another more particularly preferred embodiment of the present invention is directed to a synthetic extracellular vesicle between 70 nm and 5000 nm specifically comprising:
Also described herein is a method for treating or ameliorating a disorder comprising administering to a patient suffering from said disorder a therapeutically effective amount of a synthetic extracellular vesicle as disclosed herein, wherein the disorder is selected from the group comprising inflammation, cancer, rheumatic disorder, severe graft versus host disease, osteoarthritis, cardiovascular disorder, epithelial diseases, neurodegenerative disorders, autoimmune disorders, bone and cartilage disorders, osteoporosis, renal osteodystrophy, Paget's disease of bone, osteopetrosis, rickets, neurological disorders, intoxication, neuroendocrinology disorders, endocrinology disorders, genetic disorders, infectious diseases, dental disorders, cosmetic procedures, coagulation disorders, dermatoses, diabetes, age-associated disorders.
Also described herein is a method for treating or ameliorating a disorder comprising administering to a patient suffering from said disorder a therapeutically effective amount of a synthetic extracellular vesicle having a hydrodynamic radius between 70 nm and 5000 nm, comprising:
Also described herein is a method for treating or ameliorating a disorder comprising administering to a patient suffering from said disorder a therapeutically effective amount of a synthetic extracellular vesicle as disclosed herein, wherein the disorder is selected from the group comprising inflammation, cancer, rheumatic disorder, severe graft versus host disease, osteoarthritis, cardiovascular disorder, epithelial diseases, neurodegenerative disorders, autoimmune disorders, bone and cartilage disorders, osteoporosis, renal osteodystrophy, Paget's disease of bone, osteopetrosis, rickets, neurological disorders, intoxication, neuroendocrinology disorders, endocrinology disorders, genetic disorders, infectious diseases, dental disorders, cosmetic procedures, coagulation disorders, dermatoses, diabetes, age-associated disorders.
Also described herein is a method for treating or ameliorating a disorder comprising administering to a patient suffering from said disorder a therapeutically effective amount of a synthetic extracellular vesicle having a hydrodynamic radius between 70 nm and 5000 nm, comprising:
Also described herein is a method for treating or ameliorating a disorder comprising administering to a patient suffering from said disorder a therapeutically effective amount of a synthetic extracellular vesicle as disclosed herein, wherein the disorder is selected from the group comprising inflammation, cancer, rheumatic disorder, severe graft versus host disease, osteoarthritis, cardiovascular disorder, epithelial diseases, neurodegenerative disorders, autoimmune disorders, bone and cartilage disorders, osteoporosis, renal osteodystrophy, Paget's disease of bone, osteopetrosis, rickets, neurological disorders, intoxication, neuroendocrinology disorders, endocrinology disorders, genetic disorders, infectious diseases, dental disorders, cosmetic procedures, coagulation disorders, dermatoses, diabetes, age-associated disorders.
Also described herein is a method for treating or ameliorating a disorder comprising administering to a patient suffering from said disorder a therapeutically effective amount of a synthetic extracellular vesicle having a hydrodynamic radius between 70 nm and 5000 nm, comprising:
Also described herein is a method for treating or ameliorating a disorder comprising administering to a patient suffering from said disorder a therapeutically effective amount of a synthetic extracellular vesicle between 70 nm and 5000 nm with the composition described above and specifically comprising:
Also described herein is a method for treating or ameliorating a disorder comprising administering to a patient suffering from said disorder a therapeutically effective amount of a synthetic extracellular vesicle between 70 nm and 5000 nm with the composition described above and specifically comprising:
Also described herein is a method for treating or ameliorating a disorder comprising administering to a patient suffering from said disorder a therapeutically effective amount of a synthetic extracellular vesicle between 70 nm and 5000 nm with the composition described above, and specifically comprising:
Also described herein is a method for treating or ameliorating a disorder comprising administering to a patient suffering from said disorder a therapeutically effective amount of a synthetic extracellular vesicle between 70 nm and 5000 nm with the composition described above, and specifically comprising:
Also described herein is a method for treating or ameliorating a disorder comprising administering to a patient suffering from said disorder a therapeutically effective amount of a synthetic extracellular vesicle between 70 nm and 5000 nm with the composition described above, and specifically comprising:
Further described herein is a method for treating or ameliorating a disorder comprising administering to a patient suffering from said disorder a therapeutically effective amount of a synthetic extracellular vesicle between 70 nm and 5000 nm with the composition described above, and specifically comprising:
“Disorder” is any condition that would benefit from treatment with a substance/molecule or method described herein.
“Cell proliferative disorder” and “proliferative disorder” refer to disorders that are associated with some degree of abnormal cell proliferation, such as cancer.
“Cancer” and “cancerous” refer to, or describe a physiological condition in mammals that is typically characterized by a cell proliferative disorder. Cancer generally can include, but is not limited to, carcinoma, lymphoma (e.g., Hodgkin's and non-Hodgkin's lymphoma), blastoma, sarcoma, and leukemia. More specific examples of cancer can include, squamous cell cancer, small-cell lung cancer, non-small cell lung cancer, adenocarcinoma of the lung, squamous carcinoma of the lung, cancer of the peritoneum, hepatocellular cancer, gastrointestinal cancer, pancreatic cancer, glioma, cervical cancer, ovarian cancer, liver cancer, bladder cancer, hepatoma, breast cancer, colon cancer, colorectal cancer, endometrial or uterine carcinoma, salivary gland carcinoma, kidney cancer, liver cancer, prostate cancer, vulval cancer, thyroid cancer, hepatic carcinoma, leukemia and other lymphoproliferative disorders, and various types of head and neck cancer.
“Tumour” refers to all neoplastic cell growth and proliferation, whether malignant or benign, and all pre-cancerous and cancerous cells and tissues. The terms “cancer”, “cancerous”, “cell proliferative disorder”, “proliferative disorder”, and “tumour” are not mutually exclusive as referred to herein.
“Cardiovascular disorders” include but are not limited to disorders of the heart and the vascular system like congestive heart failure, myocardial infarction, ischemic diseases of the heart, all kinds of atrial and ventricular arrhythmias, hypertensive vascular diseases, peripheral vascular diseases, and atherosclerosis.
“Metastasis” refers to the spread of cancer and/or tumour from its primary site to other places in the body of an individual.
The term “neurodegenerative disease” or “neurological disorder” or “neuroinflammatory disorder” refers to any disease, disorder, or condition affecting the central or peripheral nervous system. Preferred examples of neurodegenerative diseases and neuroinflammatory disorders are selected from the group comprising or consisting of: Alzheimer's disease, Parkinson's disease, Creutzfeldt Jakob disease (CJD), new variant of Creutzfeldt Jakobs disease (nvCJD), Hallervorden Spatz disease, Huntington's disease, multisystem atrophy, dementia, frontotemporal dementia, motor neuron disorders of multiple spontaneous or genetic background, amyotrophic lateral sclerosis (ALS), spinal muscular atrophy, spinocerebellar atrophies (SCAs), schizophrenia, affective disorders, major depression, meningoencephalitis, bacterial meningoencephalitis, viral meningoencephalitis, CNS autoimmune disorders, multiple sclerosis (MS), acute ischemic/hypoxic lesions, stroke, CNS and spinal cord trauma, head and spinal trauma, brain traumatic injuries, arteriosclerosis, atherosclerosis, microangiopathic dementia, Binswanger' disease (Leukoaraiosis), retinal degeneration, cochlear degeneration, macular degeneration, cochlear deafness, AIDS-related dementia, retinitis pigmentosa, fragile X-associated tremor/ataxia syndrome (FXTAS), progressive supranuclear palsy (PSP), striatonigral degeneration (SND), olivopontocerebellear degeneration (OPCD), Shy Drager syndrome (SDS), age dependant memory deficits, neurodevelopmental disorders associated with dementia, Down's Syndrome, synucleinopathies, superoxide dismutase mutations, trinucleotide repeat disorders as Huntington's Disease, trauma, hypoxia, vascular diseases, vascular inflammations, CNS-ageing. Also age dependant decrease of stem cell renewal may be addressed.
“Aging-associated disorders and diseases” are most often seen with increasing frequency with increasing senescence. Examples of aging-associated diseases are atherosclerosis and cardiovascular disease, cancer, arthritis, cataracts, osteoporosis, type 2 diabetes, hypertension and Alzheimer's disease. The incidence of all of these diseases increases exponentially with age.
“Rheumatic diseases” are characterized by inflammation that affects the connecting or supporting structures of the body; most commonly the joints, but also sometimes the tendons, ligaments, bones, and muscles. Some rheumatic diseases even affect the organs. These diseases can ultimately cause loss of function in those body parts. Preferred examples of rheumatic diseases and are selected from the group comprising or consisting of: osteoarthritis, rheumatoid arthritis, fibromyalgia, systemic lupus erythematosus, gout, juvenile idiopathic arthritis, arthritis, scleroderma.
“Epithelial diseases” include acne, atopic eczema, atopic dermatitis, contact dermatitis, impetigo, psoriasis, sunburn, sweating disorders, yeast infections of the mucous membranes.
“Endocrinology disorders” include diabetes, adrenal insufficiency, cushing's disease, gigantism, hyperthyroidism, hypothyroidism, hypopituitarism, polycystic ovary syndrome.
Neuroendocrine disorders are disorders that affect the interaction between the nervous system and the endocrine system. Examples of neuroendocrine disorders include diabetes insipidus, Kallman syndrome, neuroendocrine cancer, and neuroendocrine tumors (NETs), which are neoplasms that arise from cells of the endocrine and nervous systems.
“Bone and cartilage disorders” include diseases or injuries that affect human bones and cartilage.
“Osteoarthritis” is one of the leading causes of disability in adults worldwide. It is a degenerative disease of the joints secondary to many predisposing factors, most notably age, joint injury, altered mechanical stress, and obesity. All these processes cause a local chronic inflammatory response resulting in the progressive joint failure characteristic of osteoarthritis.
“Osteoporosis” is the result of cumulative bone loss during aging. Nevertheless, a wide variety of diseases, medications, and lifestyles can cause or contribute to the development of osteoporosis. In addition, the immune system participates in the regulation of bone homeostasis through production of cytokines and inflammatory mediators with subsequent activation of cartilage-degrading proteinases.
“Paget's disease” is a chronic skeletal disorder, caused by enhanced bone resorption followed by abnormal bone formation, in which a potential cross talk between the bone and the immune system takes place.
Other bone related disorders include renal osteodystrophy, osteopetrosis, rickets.
“Cartilage disorders” include osteoarthritis, costochondritis enchondromatosis, herniation, achondroplasia, relapsing polychondritis, chondroma, chondrosarcoma.
“Treatment”, “treat” or “treating” refer to clinical intervention in an attempt to alter the natural course of a disorder in the individual being treated, and can be performed either for prophylaxis or during the course of clinical pathology. Desired results of treatment can include, but are not limited to, preventing occurrence or recurrence of the disorder, alleviation of symptoms, diminishment of any direct or indirect pathological consequences of the disorder, preventing metastasis, decreasing the rate of progression, amelioration or palliation of a disease state, and remission or improved prognosis. For example, treatment can include administration of a therapeutically effective amount of a pharmaceutical formulation comprising a synthetic extracellular vesicle disclosed herein to a subject to delay development or slow progression of a disorder, wherein the disorder is selected from the group comprising inflammation, cancer, rheumatic disorder, severe graft versus host disease, osteoarthritis, cardiovascular disorder, epithelial diseases, neurodegenerative disorders, autoimmune disorders, bone and cartilage disorders, osteoporosis, renal osteodystrophy, Paget's disease of bone, osteopetrosis, rickets, neurological disorders, intoxication, neuroendocrinology disorders, endocrinology disorders, genetic disorders, infectious diseases, dental disorders, cosmetic procedures, coagulation disorders, dermatoses, diabetes, age-associated disorders.
“Pharmaceutical formulation” refers to a preparation in a form that allows the biological activity of the active ingredient (s) to be effective, and which contain no additional components which are toxic to the subjects to which the formulation is administered.
“Pharmaceutically acceptable carrier” refers to an ingredient in a pharmaceutical formulation, other than an active ingredient, which is nontoxic to the subject to whom it is administered. A pharmaceutically acceptable carrier includes, but is not limited to, a buffer, excipient, stabilizer, or preservative.
“Therapeutically effective amount” refers to the amount of an active ingredient or agent (e.g., a pharmaceutical formulation) to achieve a desired therapeutic or prophylactic result, e.g., to treat or prevent a disease or disorder in a subject. In the case of a cancer, the therapeutically effective amount of the therapeutic agent is an amount that reduces the number of cancer cells; reduces the primary tumour size; inhibits (i.e. slows to some extent and preferably stop) cancer cell infiltration into peripheral organs; inhibits (i.e. slows to some extent and preferably stop) tumour metastasis; inhibits, to some extent, tumour growth; and/or relieves to some extent one or more of the symptoms associated with the cancer. To the extent the drug may prevent growth and/or kill existing cancer cells, it may be cytostatic and/or cytotoxic. For cancer therapy, efficacy in vivo can, for example, be measured by assessing the duration of survival, time to disease progression (TTP), the response rates (RR), duration of response, and/or quality of life.
“Individual” or “subject” refers to a mammal, including but not limited to, domesticated animals (e.g. cows, sheep, cats, dogs, and horses), primates (e.g. humans and non-human primates such as monkeys), rabbits, and rodents (e.g. mice and rats).
A “therapeutic agent” or “therapeutic molecule” includes a compound or molecule that, when present in an effective amount, produces a desired therapeutic effect, pharmacologic and/or physiologic effect on a subject in need thereof. It includes any compound, e.g. a small molecule drug, or a biologic (e.g., a polypeptide drug or a nucleic acid drug) that when administered to a subject has a measurable or conveyable effect on the subject, e.g., it alleviates or decreases a symptom of a disease, disorder or condition.
As used herein, the term “antibody” encompasses an immunoglobulin whether natural or partly or wholly synthetically produced, and fragments thereof. The term also covers any protein having a binding domain that is homologous to an immunoglobulin binding domain. “Antibody” further includes a polypeptide comprising a framework region from an immunoglobulin gene or fragments thereof that specifically binds and recognizes an antigen. Use of the term antibody is meant to include whole antibodies, polyclonal, monoclonal and recombinant antibodies, fragments thereof, and further includes single-chain antibodies, humanized antibodies, murine antibodies, chimeric, mouse-human, mouse-primate, primate-human monoclonal antibodies, anti-idiotype antibodies, antibody fragments, such as, e.g., scFv, (scFv)2, Fab, Fab′, and F(ab′)2, F(abl)2, Fv, dAb, and Fd fragments, diabodies, and antibody-related polypeptides. Antibody includes bispecific antibodies and multispecific antibodies so long as they exhibit the desired biological activity or function.
18:1 DOPG 1,2-dioleoyl-sn-glycero-3-phospho-(1′-rac-glycerol), 18:1 DOPC 1,2-dioleoyl-sn-glycero-3-phosphocholesteroline, 18:1 DOPE 1,2-dioleoyl-sn-glycero-3-phosphoethanolamine, LissRhod PE 1,2-dioleoyl-sn-glycero-3-phosphoethanolamine-N-(lissamine rhodamine B sulfonyl), 18:1 DGS-NTA(Ni) 1,2-dioleoyl-sn-glycero-3-[(N-(5-amino-1-carboxypentyl)iminodiacetic acid)succinyl] (nickel salt), 18:1 1,2-dioleoyl-sn-glycero-3-phospho-(1′-myo-inositol) (ammonium salt), 18:1 1,2-di-(9Z-octadecenoyl)-sn-glycero-3-phospho-L-serine (sodium salt), 18:1 1,2-di-(9Z-octadecenoyl)-sn-glycero-3-phosphate (sodium salt), cholesterol, 18:1 1-2-di-(9Z-octadecenoyl)-sn-glycerol, 18:0 N-stearoyl-D-erythro-sphingosylphosphorylcholine and extrude set with 50 nm pore size polycarbonate filter membranes were purchased from Avanti Polar Lipids, USA. All lipids were stored in chloroform at −20° C. and used without further purification. Hoechst 33342, CellTracker Green CMFDA dye, wheat germ agglutinin (WGA)-AlexaFluor conjugates (obtained from Thermo Fisher scientific, Invitrogen), Dulbecco's Modified Eagle Medium (DMEM) high Glucose, heat inactivated as well as exosome depleted fetal bovine serum, recombinant N-terminal His-tagged human CD9 (amino acids 103-203), penicillin-streptomycin (10,000 U/mL), L-Glutamine (200 mM), Alexa Fluor 488 NHS Ester, trypsin-EDTA (0.05%) with phenol red and phosphate buffered saline were purchased from Thermo Fischer Scientific, Germany. 1H,1H,2H,2H-Perfluoro-1-octanol (PFO) de-emulsifier and human male plasma serum were purchased from Sigma Aldrich, Germany. Bovine albumin fraction V (BSA) was purchased from Carl Roth, Germany. HaCaT cells were obtained from CLS cell line service, Germany. A431, K562, MC 3T3, and BJ cell lines as well as Iscove's Modified Dulbecco's Medium were obtained from ATCC, USA. Atto488 conjugated 1,2-dipalmitoyl-sn-glycero-3-phosphoethanolamine was purchased from Atto-Tec GmbH, Germany. Caspase 8 (Cleaved Asp391) monoclonal antibody (clone S.147.8) was purchased from Thermo Fischer Scientific, Germany. Purified Mouse Anti-ERK1/2 (pT202/pY204) was purchased from BD biosciences (Cat. No. 612358). Recombinant N-terminal His-tagged human CD9 (protein ID P21926 amino acids 112-195) was purchased from Novus Biologicals, Germany. Recombinant N-terminal His-tagged human TSG101 (protein ID Q99816, amino acids 1-145) was purchased from Fitzgerald, USA. Recombinant N-terminal His-tagged human CD81 (protein ID P35762, amino acids 113-201) was purchased from MyBioSource, USA. Recombinant N-terminal His-tagged human CD63 (protein ID P08962, amino acids Ala 103-Val 203) was purchased from Thermo Fischer Scientific, Germany. Recombinant His-tagged RANK (protein ID O35305, amino acids 31-214) was obtained from Abcam, Germany. Recombinant his-tagged FasL amino acids N-Met-His8 (Pro134-Leu281) (protein ID NM_000639.1) was obtained from BioLegend, USA. His tagged ICAM-1 (protein ID P05362) recombinant human protein (Met 1-Glu 480), was obtained from Thermo Fischer Scientific, Germany. miRIDIAN micro RNA mimics (hsa-miR-21-5p, hsa-miR124-3p, hsa-miR-125b-5p, hsa-miR-126-5p, hsa-miR-130a-3p, hsa-miR-132-3p) were purchased from Horizon Dharmacon, USA. K562 exosomes (HBM-K562) were obtained from Hansa BioMed Lonza, Switzerland. 4-well cell exclusion inserts were purchased from Ibidi, Germany. Pre-wounded full thickness human organotypic skin cultures, respective culture media and histological sample preparation services were purchased from MatTek Cooperation, USA. FC-40 oil was purchased from Iolitec, Germany. ELISA kit for quantification of human pro-collagen I alpha was obtained from Abcam, UK.
Exosome Isolation from K562 Cell Cultures
K562 extracellular vesicles were isolated from conditioned cell culture medium by differential centrifugation. For this, K562 cells were cultured in 50 ml of Iscove's modified Dulbecco's Medium for 48 hours in suspension with 10% exosome free serum at 37° C. and 5% CO2 atmosphere. The final cell concentration was 5×105 cells/ml. After incubation, the cell suspension was centrifuged at 300 g at 4° C. for 10 minutes to remove the cells. The supernatant was filtered through a 0.22 μm filter and centrifuged at 125,000 g at 4° C. for 75 min with a Beckmann Coulter Optima XE-100 ultracentrifuge in a JA-20 fixed angle rotor (k-factor 770). The pellet was washed with 50 ml ice-cold PBS and centrifuged again under the same conditions. The exosome pellet was resuspended in 1 ml PBS. The total protein concentration of this exosome suspension was assessed by measuring the absorbance at 280 nm with a Nanodrop ND-1000 spectrophotometer.
Confocal microscopy was performed with a laser scanning microscope LSM 800 (Carl Zeiss AG). Images were acquired with a 20× (Objective Plan-Apochromat 20×/0.8 M27, Carl Zeiss AG) and a 63× immersion oil objective (Plan-Apochromat 63×/1.40 Oil DIC, Carl Zeiss AG). Images were analyzed with ImageJ (NIH) and adjustments of image brightness and contrast or background corrections were performed always on the whole image and special care was taken not to obscure or eliminate any information from the original image. For bright field imaging a Leica DMi8 inverted fluorescent microscope equipped with a sCMOS camera and 10×HC PL Fluotar (NA 0.32, PH1) objective was used.
For analysis of extracellular vesicle uptake into HaCaT cells, rhodamine B labeled extracellular vesicles were incubated with HaCaT cells in Nunc LabTek 8-well chambers. Immediately after addition of the extracellular vesicles to the cells, 5 μg/ml of AlexaFluor (obtained from Thermo Fischer Scientific, Invitrogen) conjugated wheat germ agglutinin (WGA) was added to the medium. Cells were incubated for 24 hours and subsequently imaged by confocal laser scanning microscopy. WGA binds to specific sugar residues on the outer cell membrane and is endocytosed along with them during membrane turn-over and endocytotic processes, staining intracellular endosomal vesicles.
Alexa488 labelled CD9 was produced by incubating NHS functionalized Alexa488 with recombinant CD9 in a twofold molar excess for 2 hours at 37° C. in PBS. Subsequently, free NHS was quenched by adding a 10-fold molar excess of glycine.
Staining of HaCaT cells with CellTracker Green was performed by incubating 20 μM of cell Tracker Green CMFDA dye for 60 min. To remove excess dye and non-uptaken synthetic extracellular vesicles, cells were rinsed twice with PBS.
For analysis of synthetic extracellular vesicles conjugated with recombinant extracellular domain of FasL (
The protein to lipid ratio on vesicle membranes (
Analysis of the hydrodynamic radius of vesicles was performed with a Malvern Zetasizer Nano ZS system. Samples were diluted to a final lipid concentration of 15 μM in PBS filtered with through a 0.22 μm filter. The temperature equilibration time was set to 300 s at 25° C. Three individual measurements for each sample were performed at a scattering angle of 173° based on the built-in automatic run-number selection. The material refractive index was set to 1.4233 and solvent properties were set to η=0.8882, n=1.33 and ε=79.0.
Size polydispersion index (PDI) was assessed from hydrodynamic radius measurements based on dynamic light scattering. A Malvern Zetasizer Nano ZS was used to perform dynamic light scattering (DLS) measurements and PDI was derived from the automatically calculated size distribution analysis.
HaCaT, BJ and A431 cells were cultured in Dulbecco's Modified Eagle Medium supplemented with 4.5 g/I glucose, 1% L-glutamine, 1% penicillin/streptomycin and 10% fetal bovine serum. Cells were routinely cultured at 37° C. and 5% CO2 atmosphere and passaged at approx. 80% confluency using 0.05% trypsin/EDTA treatment. K562 cells were cultured in suspension in Iscove's modified Dulbecco's Medium supplemented with 10% exosomes free fetal bovine serum. K562 cells were splitted every other day by transferring 3 ml of cell suspension to 10 ml of fresh cell culture medium.
To evaluate serum stability (
For proliferation analysis, a previously reported Hoechst 33342 intensity analysis was applied. To this end, HaCaT and A431 cells were seeded at a density of 15.000 cells/well in a flat-bottom transparent 96-well plate in 200 μl culture medium. Cells were seeded together with corresponding extracellular vesicles and incubated for 48 hours. Subsequently, cells were washed twice with 100 μl PBS and incubated for 10 min with icecold culture medium supplemented with 10 μM Hoechst 33342. After removal of the culture medium and 2×washing with PBS, Hoechst 33342 intensity was measured at four individual positions in each well using an Infinite M200 TECAN plate reader controlled by TECAN iControl software with an in-built gain optimization and excitation/emission setting adjusted to 380/460 nm. Measurements were performed in triplicates.
For in vitro 2D wound healing assays, 4-well silicone cell exclusion cell culture inserts with a gap width of 500 μm were used in 12-well plastic plates. Cells were seeded at a cell density of 40,000 cells/well and allowed to adhere overnight in 110 μl culture medium (2 ml of culture medium were added to the well outside of the inserts). Extracellular vesicles were incubated (at final lipid concentration of 10 μM) with the cell monolayer for 24 hours. Subsequently, the inserts were carefully removed using sterile tweezers and the wound was allowed to close for 16 hours. For quantification, culture medium was removed and cell layers were fixed with ice-cold 4% paraformaldehyde for a minimum of 20 min. The wound sides were then imaged by phase contract microscopy and the cell free area was quantified manually with ImageJ software.
For analysis of human organotypic full thickness skin models, pre-wounded human epidermal keratinocytes (obtained from neonatal-foreskin normal tissue of a single donor) and fibroblasts 3D cultures were obtained from a commercial distributer (MatTek corporation). Skins were cultured at an air-liquid interface following manufacturer's suggestions. For wound closure analysis, tissues were allowed to equilibrate for 16 hours after arrival at 37° C. in a 5% CO2 atmosphere. Subsequently, 2 μl of the extracellular vesicle solution (or respective buffer controls) were pipetted onto the wound side and the wound was allowed to heal for 48 hours at 37° C. in a 5% CO2 atmosphere. Tissues were then fixed with 10% formalin solution overnight at 4° C. Wound size was quantified from histological H/E slices. For each wound, six individual slices and three individual wounds were analyzed.
For gel-electrophoretic analysis of protein content of K562 exosomes and synthetic extracellular vesicles, a NuPAGE bold Bis-Tris 4-12% gradient gel was used with MES running buffer. Electrophoresis was performed at 200V for 35 min under denaturating conditions with a total of 3 μg (for natural exosomes) or 500 ng (for synthetic extracellular vesicles) of protein loaded onto each lane. Protein staining was performed with Coomassie R250. Line intensity profiles of the respective lanes were measured by ImageJ software.
For quantification of in vitro collagen deposition, BJ dermal fibroblast were seeded in 96-well flat bottom transparent cell culture plates at a density of 20,000 cells/well. 24 hours after seeding, cells were washed twice with PBS and 200 μl of fresh cell culture medium was added together with synthetic extracellular vesicles (to a final lipids concentration of 10 μM) to the cells. Cells were incubated for 24 hours with synthetic extracellular vesicles. Subsequently human pro-collagen I alpha in the medium was quantified by enzyme-linked immunosorbent assay (Abcam ELISA Kit) following the manufacturer's instructions.
The following examples are included to demonstrate preferred embodiments of the invention. It should be appreciated by those of skill in the art that the techniques disclosed in the examples which follow represent techniques discovered by the inventor to function well in the practice of the invention, and thus can be considered to constitute preferred modes for its practice. However, those of skill in the art should, in light of the present disclosure, appreciate that many changes can be made in the specific embodiments which are disclosed and still obtain a like or similar result without departing from the scope of the invention.
Further modifications and alternative embodiments of various aspects of the invention will be apparent to those skilled in the art in view of this description. Accordingly, this description is to be construed as illustrative only and is for the purpose of teaching those skilled in the art the general manner of carrying out the invention. It is to be understood that the forms of the invention shown and described herein are to be taken as examples of embodiments. Elements and materials may be substituted for those illustrated and described herein, parts and processes may be reversed, and certain features of the invention may be utilized independently, all as would be apparent to one skilled in the art after having the benefit of this description of the invention. Changes may be made in the elements described herein without departing from the scope of the invention as described in the following claims.
Synthetic extracellular vesicles were produced by shear stress emulsification (
The so obtained solution, representing the water phase of step a), was then diluted to a final concentration of 3 mM with PBS, or eventually PBS containing the desired miRIDIAN miRNA mimic components at concentration 40-145 nM.
In this example, polymer shell-stabilized extracellular vesicles were produced from small unilamellar vesicles containing 41 mol % cholesterol, 16 mol % SM, 15 mol % DOPC, 11 mol % DOPS, 6 mol % DOPE, 5 mol % DOPG, 2 mol % PA, 1 mol % DAG, 1 mol % PI, 1 mol % LissRhod PE, 1 mol % DGS-NTA(Ni2+) in PBS containing miRIDIAN RNA (40-145 nM). This lipid composition resembles that of natural extracellular vesicles. However, the technology allows for the integration of an almost unrestricted number of possible lipid types into synthetic exosome membrane.
This water phase was then combined with an oil phase at a ratio 1:2. The oil phase consisted of FC-40 oil containing the fluorosurfactant triblock PEG2500-PFPE600-PEG2500 at a final concentration of 1.25 mM.
The combined water phase and oil phase was then emulsified using an Ultra Turrax IKA T10 basic emulsifier for, exemplarily, 60 sec at approx. 26,300 rpm. The resulting polymer shell stabilized synthetic extracellular vesicles were incubated for at least 2 hours at 4° C. in the dark.
Release of the polymer shell stabilized extracellular vesicles into an aqueous release buffer was performed by removing excess oil phase and adding the release buffer (PBS) and 1H,1H,2H,2H-perfluoro-1-octanol (PFO) to the mixture in a 1:1:1 ratio of aqueous release buffer:PFO. aqueous intraluminal buffer. After 30 min of equilibration, the layer containing the extracellular vesicles was transferred into a 2 ml microtube and PBS was added to a final volume of 2 ml.
This solution was centrifuged at >10,000 g for 15 min.
The supernatant was discarded and the extracellular vesicle pellet was resuspended in PBS. The extracellular vesicles were also released into PBS containing 0.1% BSA to block unspecific protein-lipid interactions
In order to conjugate the released extracellular vesicles with CD peptides, the total amount of NTA(Ni2+) functionalized lipids was calculated according to the lipid ratio. Thus, the His-tagged CD-peptides (CD9, TSG101, or CD63, or CD81) were added to the extracellular vesicle solution in a 1:2 excess and allowed to conjugate for 1 hour at 37° C. protected from light. After this phase, 500 nM Hoechst33342 was eventually added to the extracellular vesicles to visualize nucleic acids by confocal microscopy analysis.
The extracellular vesicle solution was subsequently centrifuged at >10,000 rpm for 15 min. The supernatant containing unbound peptides was disposed and the extracellular vesicle pellet resuspended in PBS.
In order to compare the lipid ratio of the formed extracellular vesicles, and that of the small unilamellar vesicles, the total lipid concentration of the extracellular vesicles was determined by quantifying the fluorescence from the integrated rhodamine B or Atto488 conjugated lipids, which was referenced to a small unilamellar vesicle standard dilution curve.
The results of quantitative electrospray-ionization tandem mass spectrometry revealed that fully synthetic extracellular vesicles had the lipid composition cholesterol:SM:DOPC:DOPS:DOPE:DOPG:PA:DAG:PI 43:16:15:11:6:5:2:1:1, resembling not only the lipid composition of natural extracellular vesicles, but also that of the original small unilamellar vesicles (
The fully synthetic extracellular vesicles produced as described in Example 1 were compared to natural extracellular vesicles in term of purity, protein composition, dimension, and variability between different batches.
Interestingly, the fully synthetic extracellular vesicles produced according to the invention contained considerably less contaminating aggregates and non-vesicular particles compared to exosomes isolated by differential centrifugation from conditioned K562 erythroleukemia cell media or commercially available exosomes from the same cell line (see
Moreover, when assessing the protein content of the respective vesicles by denaturating polyacrylamide gel-electrophoresis, it was found that exosomes isolated from conditioned K562 media and K562 commercial exosomes, differed greatly in their protein content, underscoring the degree of variation between different vesicle preparation methods (
Additionally the biophysical similarity of fully synthetic extracellular vesicles to natural extracellular vesicles was evaluated by dynamic light scattering. The results showed that the size of polymer shell stabilized extracellular vesicles and therefore the hydrodynamic radius of fully synthetic extracellular vesicles can be fine-tuned by adjusting the shear stress used during emulsification, producing fully synthetic extracellular vesicles radii between 292 nm±12 nm, (CV=4.1%; n=3) by emulsification for 30 sec at 30,000 rpm, and radii between 627 nm±15 nm, (CV=2.4%; n=3) by emulsification for 30 sec at 14,000 rpm. The zeta potential of synthetic extracellular vesicles containing the above-mentioned lipid formulation was −12.3 mV (±0.7 mV, n=3). Thus, size and zeta potential of the fully synthetic extracellular vesicles are comparable to those of natural extracellular vesicles reported in literature (Vogel, R. et al. High-Resolution Single Particle Zeta Potential Characterization of Biological Nanoparticles using Tunable Resistive Pulse Sensing, Scientific reports 7, 2017).
Moreover, the hydrodynamic radius values of the synthetic extracellular vesicles were very similar to those of commercial K562 exosomes (468 nm±199 nm, CV=42.5%, and −11.8 mV±0.9 mV, n=3) and of exosomes isolated from conditioned K562 cell culture medium (240 nm±32 nm, CV=13.3%, and −11.3 mV±0.5 mV, n=3).
With respect to particle size distribution characterization, a parameter used to define the size range of the lipidic carrier systems is called the “polydispersity index” (PDI). PDI is basically a representation of the distribution of size populations within a given sample. The term “polydispersity” (or “dispersity” as recommended by IUPAC) is used to describe the degree of non-uniformity of a size distribution of particles (Danaei et al., Pharmaceutics 2018, 10, 57). Also known as the heterogeneity index, PDI is a number calculated from a two-parameter fit to the correlation data (the cumulants analysis). This index is dimensionless. The numerical value of PDI ranges from 0.0 (for a perfectly uniform sample with respect to the particle size) to 1.0 (for a highly polydisperse sample with multiple particle size populations). In drug delivery applications using lipid-based carriers, such as liposome and nanoliposome formulations, a PDI of 0.3 and below is considered to be acceptable and indicates a homogenous population of phospholipid vesicles.
Dynamic scattering analysis showed that the polydispersion index of an exemplary sample of fully synthetic extracellular vesicles is 0.098, demonstrating that the vesicles are homogenous in size.
Vesicle stability was evaluated by measuring the uptake rate by target cells of two synthetic extracellular vesicle samples preserved for two different time intervals (
These results confirm that fully synthetic extracellular vesicles can be assembled from individually adjustable synthetic lipid precursors to precisely match the lipid composition and therefore biophysical characteristic (membrane charge, dimensions) of natural extracellular vesicles. Importantly, the fully synthetic extracellular vesicles prepared according to the inventive method showed the technical advantage to be much more reproducible, pure, stable and homogenous in size (coefficient of variation and PDI) in comparison with the exosomes obtained according to prior art methods.
Although lipids play an important role in extracellular vesicle communication, for therapeutic applications the main physiological functions of extracellular vesicles is commonly attributed to their micro RNA (miRNA) cargo and to the peripheral membrane proteins and receptor ligands on their surface.
The inventors have here produced synthetic exosomes miming fibrocyte-derived exosomes, in order to show the potential of the inventive method to produce synthetic exosomes for therapeutic application.
Natural fibrocyte-derived exosomes contain miRNAs including miR-21, miR-124, miR-125, miR-126, miR-130 and miR-132 and protein components including CD9, CD63 and CD81, and have been shown to promote wound-healing. Therefore, synthetic exosomes where prepared using synthetic miRIDIAN mimics of the miRNA described in the natural exosomes at a concentration typically found in natural exosomes (750 pg/1012 vesicles), by mixing the miRNA solution with the initial water phase containing the lipids (
Confocal microscopy analysis of the prepared synthetic exosomes showed luminal distribution of the miRNAs and peripheral distribution of Alexa-488 labeled CD9, overlapping with the lipid fluorescence of Liss Rhodamin PE integrated into the lipid bilayer (
The interaction of fully synthetic extracellular vesicles synthetized as explained in Example 3 with target cells was analysed by incubating the keratinocytes HaCaT cells with said vesicles for 24 hours. These experiments showed that fluorescently labeled synthetic extracellular vesicles are internalized by HaCaT cells via an endosomal pathway (
Importantly, these results suggest that the content of the fully synthetic extracellular vesicles is delivered into the target cells, so that they could be used for intracellular cargo delivery and for therapy of different disorders.
It has been shown that human fibrocyte-derived exosomes can have pro-proliferative effect, accelerates the collective migratory behavior of dermal keratinocytes and enhances collagen deposition of dermal fibroblasts, ultimately promoting wound closure in a diabetic mice model. Therefore, the wound healing effect of the synthetic extracellular vesicles synthetized as in Example 3 was tested studying their effect on cell proliferation, migration and collagen deposition, which is crucial for wound closure and healing.
The pro-proliferative effect on spontaneously immortalized keratinocytes Hacat cells, was assessed by quantifying keratinocyte number via nuclear staining after 48 h of incubation with the fully synthetic extracellular vesicles.
In order to decipher the contribution of the individual biomolecular components of the fully synthetic extracellular vesicles, fully synthetic extracellular vesicles lacking miRNAs, but decorated with the single tetraspanins or combinations of them were produced and analysed. Interestingly, co-presentation of CD9 and CD63 or CD9 and CD81 leaded to a synergistic effect on proliferation (
The contribution of the single miRNAs was further evaluated by producing CD9, CD63 and CD81 biofunctionalized synthetic extracellular vesicles loaded with the individual human miRNA mimics (
Thus, these results show that fully synthetic extracellular vesicles can act as appropriate carriers to convey miRNA-based signaling information and thereby acting on post-transcriptional gene regulation by which they mirror a central mechanism of extracellular vesicle signaling.
The effect of extracellular vesicles on epithelial cell migration was assessed performing in vitro cell exclusion wound healing assays of collectively migrating keratinocyte monolayers treated for 24 h with the fully synthetic extracellular vesicles (
Building on these observation, the ratios of the different miRNAs and tetraspanins could be varied, in order to assess the influence of individual signaling pathways.
The effect of fully synthetic extracellular vesicles on pro-collagen-la deposition of BJ dermal fibroblasts was assessed by enzyme-linked immunosorbent assay (ELISA) after a 24 hours treatment with the synthetic extracellular vesicles (
Taken all these finding together, the in vitro assembled vesicles, like their natural equivalents, comprise the ability to boost three of the very fundamental processes critical for wound healing: proliferation, migration and collagen deposition.
In order to assess the ability of fully synthetic extracellular vesicles to promote epithelial regeneration of wounded skin, wounded organotypic full-thickness human skin models (
It is known that proliferation, migration and collagen deposition play a critical role in development and progression of epithelial carcinomas. Therefore, the effect of fully synthetic extracellular vesicles miming the fibrocyte derived vesicle was additionally tested on A431 human vulvar squamous carcinoma cells.
The results revealed that A431 carcinoma cells responded to fully synthetic extracellular vesicle treatment by accelerated in vitro collective migration, a central requirement for tumor invasion and metastasis, but displayed an altered sensitivity for the respective fully synthetic extracellular vesicle surface proteins (
The inventive method was applied to produce synthetic extracellular vesicles containing a nicotinamide phosphoribosyltransferase intracellular protein, as previous studies showed that these vesicles play a role in treatment of age-related disorders, and increase of lifespan.
Therefore, the inventors have here produced synthetic extracellular vesicles according to the inventive method and specifically comprising the functional protein nicotinamide phosphoribosyltransferase, and cytosolic proteins such as Apoptosis-Linked Gene 2-Interacting Protein X (ALIX), tumour susceptibility gene 101 protein (TSG101). These synthetic extracellular vesicles did not comprise transferrin and albumin.
The inventors aimed to synthetize synthetic extracellular vesicles with immunoregulatory properties that could be used to treat immune disorders, autoimmune disorders, inflammatory disorders, such as rheumatoid arthritis, and cancer, e.g. by cancer immunotherapy.
This type of synthetic extracellular vesicles can comprise transmembrane proteins such as MHCII, CD80, CD86, CD11c, MHCI, integrin α-chains, integrin β-chains, ICAM-1, and CD71; functional proteins such as cytokines, interleukins, IL4, growth factors, milk fat globule-EGF factor 8 protein (MFGE8), Fas, Fas Ligand (FasL), RANK, RANK Ligand (RANKL), indolamin-2,3-dioxygenase, cytotoxic T-lymphocyte-associated protein 4-immunoglobulin fusion protein (CTLA4-Ig), tumor necrosis factor-related apoptosis-inducing ligand (Apo2L, TRAIL).
Fas (also named CD95, Apo-1) is a membrane receptor from the TNF family expressed on surface of almost all cells in the human body. Upon activation by Fas ligand (FasL), Fas induces apoptosis by activation of caspase signalling. Fas mediated apoptosis plays a major role in immunobiology, and especially by death induction of infected and cancerous cells mediated by cytotoxic T-cells and by natural killer cells (NK-cells). Moreover, Fas induced apoptosis is crucial for regulation of T-cell physiology in inflammatory diseases. Major producers of FasL are T-cells and NK-cells which release FasL primarily from intracellular reservoirs in a form bound to extracellular vesicles resembling exosomes.
Synthetic extracellular vesicles presenting FasL could therefore be useful not only to investigate further Fas-signalling, but also to provide new therapy option in immune disorders, inflammatory disorders, neurodegenerative disorders, or cancer.
In order to produce synthetic extracellular vesicles presenting FasL, synthetic extracellular vesicles were produced by the inventive method based on mechanical emulsification at 14,000 rpm for 30 sec with the following composition: 20 mol % DOPG, 78 mol % DOPC, 1 mol % DGS-NTA(Ni2+) and 1 mol % LissRhod PE. Obtained synthetic extracellular vesicles had a mean hydrodynamic radius of 606 nm, as measured by dynamic light scattering (DLS). Recombinant his-tagged FasL amino acids N-Met-His8 (BioLegend, USA) was coupled on the surface of these synthetic extracellular vesicles by applying bio-orthogonal surface chemistry NTA-poly-histidine tag coupling. The FasL present on synthetic extracellular vesicles is referred to as vesicular FasL to differentiate from soluble FasL (sFasL). Cryo-electron microscopy analysis demonstrated correct coupling of FasL on vesicle surface (
FasL-synthetic vesicles were then tested for their pro-apoptotic activity on T-cells, which is important for therapy of inflammatory and autoimmune diseases. To this aim, Jurkat T-cells were incubated for 24 hours with FasL-synthetic vesicles and apoptotis was quantified by measuring propidium iodide (PI) staining intensity on a plate reader. Results showed that vesicle bound recombinant FasL is able to induce apoptosis in the T-cells, but not soluble FasL (
Thereafter, FasL-synthetic vesicles were tested for their ability to activate caspase-8, which has been shown also for natural FasL-vesicles. To this aim, human dermal fibroblast BJ cells were incubated with FasL-synthetic vesicles for 2 hours and then stained with antibodies recognizing activated caspase-8, i.e. cleaved at Asp391 (
We further evaluated the pro-apoptotic concentration range of FasL presenting synthetic vesicles on Jurkat T-cells and on K562 granulocytic cells, which are frequently used to assess FasL mediated apoptosis on NK cells. We again found that the addition of soluble FasL (sFasL) did not display any cytotoxic effect at all tested concentrations (
Thereafter, the inventive method was used to assemble pro-apoptotic synthetic vesicles exposing not only FasL but also the intercellular adhesion molecule ICAM-1, which is presented on natural occurring extracellular vesicles with immunological activity. Synthetic extracellular vesicles presenting FasL and ICAM-1, were produced as described above by mechanical emulsification at 14,000 rpm for 30 sec. Obtained synthetic extracellular vesicles had a mean hydrodynamic radius of 606 nm. The pro-apoptotic potential of these vesicles was tested on Jurkat T-cells using different FasL to ICAM-1 ratios, and measuring cell death by propidium iodide (PI) staining It was found that the killing efficiency of the synthetic vesicles depends on the FasL to ICAM-1 ratio (
Moreover, in order to analyse the effect of vesicle membrane density of FasL on the pro-apoptotic potential, vesicles harbouring 5 mol % DGS-NTA(Ni2+) were compared to vesicles with 1 mol % DGS-NTA(Ni2+). Jurkat cells were incubated with FasL-extracellular vesicles (vFasL) with 1 mol % DGS-NTA(Ni2+) at 1×108 vesicles/ml or vFasL) with 5 mol % DGS-NTA(Ni2+) at 2×107 vesicles/ml, in order to have the same final FasL concentration in the two samples. Results showed that vesicles with higher FasL density (5 mol % DGS-NTA(Ni2+)) displayed reduced killing-efficiency.
The molar percentage of DGS-NTA(Ni2+) is a measure of the density of FasL on the vesicles as confirmed by labelling the vesicles with gold-nanoparticle conjugated antibodies and subsequent transmission electron microscopy imaging (
The inventive method was applied to produce synthetic extracellular vesicles resembling those derived from mesenchymal stem cells. Natural extracellular vesicles derived from mesenchymal stem cells have been shown to have a wide range of potential therapeutic effects, such as alleviation of severe graft versus host disease, osteoarthritis and promotion of cartilage extracellular matrix homeostasis.
The synthetic extracellular vesicles resembling those of mesenchymal stem cell origin were prepared with transmembrane proteins such as CD29, CD44, CD90, CD73, Sca-1, tetraspanin proteins CD9, CD63, and CD81, functional proteins such as Wnta and Wntb, nucleic acid molecules such as miR-140-5p, miR-92a-3p-e, nucleic acid molecules such as miRNAs miR-33b, miR-451, miR-575, miR-630, miR-638, miR-1202, miR-1207-5p, miR-1225-5p, miR-1268, miR-K12-3.
Emerging evidence suggests that stem cell derived exosomes and their microRNA cargo mediate cell therapy derived neurorestorative effects in patients after stroke. In particular, these exosomes can play a role in angiogenesis, neurogenesis, vascular remodeling, white matter remodeling, and also modulate inflammatory and immune responses at the local and systemic level.
In order to reproduce exosomes with such properties, the inventive method was applied to produce synthetic extracellular vesicle specifically comprising: transmembrane proteins such as CD29, CD44, CD90, CD73, CD44, Sca-1, tetraspanin proteins CD9, CD63, and CD81; nucleic acid molecules such as miR-17, miR-18a, miR-19a, miR-19b-1, miR-20a, miR-92a, let-7a, miR-21, miR124, miR126, miR-133b, miR-191, miR-222, miR-494, miR-6087, miR-30d-5p; miR-33b, miR-451, miR-575, miR-630, miR-638, miR-1202, miR-1207-5p, miR-1225-5p, miR-1268, miR-K12-3.
Osteoclast to osteoblast signalling is crucial for bone homeostasis in order to assure correct bone formation and resorption. Receptor activator of nuclear factor-κB (RANK) is pivotal for this interaction. It has been shown, that maturing osteoclasts secrete vesicles presenting RANK on their surface, which then binds to RANKL on osteoblast surface, thus boosting mineralization of osteoblasts by activation of “reverse signalling”. After RANK ligand (RANKL)-RANK binding, downstream MAP-kinase signalling is activated. The RANK-exposing vesicles have been shown to be crucial for physiological signalling and represent potential therapeutic targets to treat bone diseases such as osteoporosis.
Therefore, synthetic extracellular vesicles exposing RANK on their surface were produced applying the inventive method based on emulsification using a mechanic emulsifier, and tested for their activating effect on pre-osteoblast cells.
Synthetic extracellular vesicles were produced the following lipid composition: 20 mol % DOPG, 78 mol % DOPC, 1 mol % DGS-NTA(Ni2+) and 1 mol % LissRhod PE. Recombinant His-tagged RANK (amino acids 31-214) (protein ID O35305) was coupled on the surface of these synthetic extracellular vesicles by applying bio-orthogonal surface chemistry NTA-poly-histidine tag coupling, at a protein lipid (LissRhodPE) ratio of 1:100. Two samples of such synthetic extracellular vesicles were produced having hydrodynamic radii of 292 nm (emulsification at 30,000 rpm, 30 sec) and 615 nm (emulsification at 14,000 rpm, 30 sec), respectively, in order to optimize the vesicle dimension for maximal functionality. Synthetic extracellular vesicles were incubated with MC 3T3 cells, a model of pre-osteoblast cells, for 24 hours in 96 well plates at concentration 10 μM (vesicle lipid moles/medium volume). Cell samples were subsequently stained for p(202/204) ERK with an anti-p(202/204) ERK1/2-Alexa488 conjugated antibody and staining intensity was quantified using an Infinite M200 TECAN plate reader controlled by TECAN iControl software with an in-built gain optimization and excitation/emission setting adjusted to 488/512 nm. Here, phosphorylation of ERK at amino acids 202/204 was quantified as a measure of vesicular RANK potency in specific pre-osteoblast cell activation. As a control, soluble RANK was added to control cells at the same concentration as used to produce vesicular RANK (soluble or vesicle RANK/medium volume=100 ng/ml).
The results revealed that RANK exposed on synthetic extracellular vesicles can induce RANK signalling more strongly compared to soluble RANK (
cerevisiae)
cerevisiae)
Number | Date | Country | Kind |
---|---|---|---|
20155012.6 | Jan 2020 | EP | regional |
Filing Document | Filing Date | Country | Kind |
---|---|---|---|
PCT/EP2021/052145 | 1/29/2021 | WO |