US20200230259A1 - Isolation of extracellular vesicles (evs) from red blood cells for gene therapy - Google Patents

Isolation of extracellular vesicles (evs) from red blood cells for gene therapy Download PDF

Info

Publication number
US20200230259A1
US20200230259A1 US16/825,097 US202016825097A US2020230259A1 US 20200230259 A1 US20200230259 A1 US 20200230259A1 US 202016825097 A US202016825097 A US 202016825097A US 2020230259 A1 US2020230259 A1 US 2020230259A1
Authority
US
United States
Prior art keywords
evs
cells
target cells
rna
aso
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US16/825,097
Inventor
Thi Nguyet Minh Le
Jiahai Shi
Muhammad Waqas
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
City University of Hong Kong CityU
Original Assignee
City University of Hong Kong CityU
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by City University of Hong Kong CityU filed Critical City University of Hong Kong CityU
Priority to US16/825,097 priority Critical patent/US20200230259A1/en
Publication of US20200230259A1 publication Critical patent/US20200230259A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/87Introduction of foreign genetic material using processes not otherwise provided for, e.g. co-transformation
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • A61K48/0075Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the delivery route, e.g. oral, subcutaneous
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/14Hydrolases (3)
    • C12N9/16Hydrolases (3) acting on ester bonds (3.1)
    • C12N9/22Ribonucleases RNAses, DNAses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/13Tumour cells, irrespective of tissue of origin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/14Blood; Artificial blood
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • A61K48/0008Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the 'non-active' part of the composition delivered, e.g. wherein such 'non-active' part is not delivered simultaneously with the 'active' part of the composition
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • A61K48/0008Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the 'non-active' part of the composition delivered, e.g. wherein such 'non-active' part is not delivered simultaneously with the 'active' part of the composition
    • A61K48/0025Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the 'non-active' part of the composition delivered, e.g. wherein such 'non-active' part is not delivered simultaneously with the 'active' part of the composition wherein the non-active part clearly interacts with the delivered nucleic acid
    • A61K48/0041Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the 'non-active' part of the composition delivered, e.g. wherein such 'non-active' part is not delivered simultaneously with the 'active' part of the composition wherein the non-active part clearly interacts with the delivered nucleic acid the non-active part being polymeric
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • A61K48/005Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the 'active' part of the composition delivered, i.e. the nucleic acid delivered
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • A61K48/0091Purification or manufacturing processes for gene therapy compositions
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/87Introduction of foreign genetic material using processes not otherwise provided for, e.g. co-transformation
    • C12N15/88Introduction of foreign genetic material using processes not otherwise provided for, e.g. co-transformation using microencapsulation, e.g. using amphiphile liposome vesicle
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/87Introduction of foreign genetic material using processes not otherwise provided for, e.g. co-transformation
    • C12N15/90Stable introduction of foreign DNA into chromosome
    • C12N15/902Stable introduction of foreign DNA into chromosome using homologous recombination
    • C12N15/907Stable introduction of foreign DNA into chromosome using homologous recombination in mammalian cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0634Cells from the blood or the immune system
    • C12N5/0641Erythrocytes
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/483Physical analysis of biological material
    • G01N33/487Physical analysis of biological material of liquid biological material
    • G01N33/49Blood
    • G01N33/491Blood by separating the blood components
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K2035/126Immunoprotecting barriers, e.g. jackets, diffusion chambers
    • A61K2035/128Immunoprotecting barriers, e.g. jackets, diffusion chambers capsules, e.g. microcapsules
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/11Antisense
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/12Type of nucleic acid catalytic nucleic acids, e.g. ribozymes
    • C12N2310/122Hairpin
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/20Type of nucleic acid involving clustered regularly interspaced short palindromic repeats [CRISPRs]

Definitions

  • the present invention relates to the field of molecular biology and genome editing, more specifically the transfer of genetic materials to recipient cells by extracellular vesicles (EVs).
  • EVs extracellular vesicles
  • RNA therapeutics including antisense oligo nucleotides (ASOs), small-interfering RNA (siRNAs), synthetic mRNAs and genome editing RNA-protein complexes are emerging modalities for therapies targeting the human genomes at high specificity and great flexibility.
  • ASOs and siRNAs have been widely used as the tools for gene knockdown in biomedical research. Their ability to silence any gene of interest offers a great potential for targeting disease-prevalent genes.
  • Various chemical modifications or conjugations can be used to keep ASOs and siRNAs stable and enhance their binding specificity.
  • Common methods for RNA transfection including nucleofection, lipofection and electroporation are only suitable for ex vivo delivery. Viral transduction and nanoparticles are often used for in vivo delivery of RNAs and DNAs however, these methods are usually ineffective, immunogenic and toxic.
  • CRISPR clustered regularly interspaced short palindromic repeats
  • a CRISPR system involves two main components: a Cas9 enzyme and a guide (gRNA).
  • the gRNA contains a targeting sequence for DNA binding and a scaffold sequence for Cas9 binding.
  • Cas9 nuclease is often used to “knockout” target genes hence it can be applied for deletion or suppression of oncogenes that are essential for cancer initiation or progression.
  • CRISPR system offers a great flexibility in targeting any gene of interest hence, potential CRISPR based therapies can be designed based on the genetic mutation in individual patients.
  • An advantage of the CRISPR system is its ability to completely ablate the expression of disease genes which can only be suppressed partially by RNA interference methods with ASOs or siRNAs.
  • multiple gRNAs can be employed to suppress or activate multiple genes simultaneously, hence increasing the treatment efficacy and reducing resistance potentially caused by new mutations in the target genes.
  • the applications of CRISPR technology have evolved very quickly from bench to bedside targeting different diseases. Clinical trials of CRISPR-mediated modification of T cells for cancer therapies have started in China and in the USA. Many other CRISPR-based therapies are under development. However, most of these therapies rely on ex vivo modification of the target cells or systemic delivery of the CRISPR system using virus or nanoparticles that can target very few cell types such as hepatocytes.
  • AML Acute myeloid leukemia
  • PB peripheral blood
  • BM bone marrow
  • RNA interference and genome editing methods are emerging as new approaches to target these abnormalities.
  • delivery of RNAs to AML cells for gene therapies has proven challenging, especially for in vivo treatments.
  • Common gene therapy delivery vehicles such as adeno-associated virus (AAV) and lipid nanoparticles (LNPs) are mostly ineffective or toxic in AML models.
  • AAV adeno-associated virus
  • LNPs lipid nanoparticles
  • RNA delivery to target cells comprising the steps of: a) purification of extracellular vesicles (EVs) from red blood cells (RBCs); b) electroporation of the EVs with RNAs to form RNA-loaded EVs; and c) applying the RNA-loaded EVs to the target cells.
  • EVs extracellular vesicles
  • RBCs red blood cells
  • the advantage of using EVs (including microvesicles and exosomes) from RBCs is that the RBCs are the most abundant blood cells hence a large amount of EVs can be obtained and purified from RBC units that are available at any blood bank.
  • the RBCs are derived from a human. They are also nontoxic, unlike synthetic transfection reagents.
  • RBC EVs do not contain oncogenic DNA/RNA or growth factors that are usually abundant in EVs from cancer cells or stem cells, hence RBC EVs do not post any transformation risks to recipient cells.
  • the RBCs are derived from a mammal preferably a human and treated with ionophore in particular calcium ionophore.
  • the EVs are purified using ultracentrifugation with a sucrose cushion.
  • sucrose cushion refers to a sucrose gradient which establishes itself during a centrifugation.
  • the sucrose gradient is prepared by using a solution of about 40% to about 70%, about 50% to about 60%, or about 60% of sucrose.
  • the electroporated EVs comprises antisense oligonucleotides (ASO), mRNAs and plasmids.
  • ASO antisense oligonucleotides
  • mRNAs mRNAs
  • plasmids plasmids.
  • the ASO comprises or consists of SEQ ID NO: 1.
  • the target cells comprise cancer cells, or are cancer cells.
  • the target cells comprise leukemia cells in particular acute myeloid leukemia (AML) cells, breast cancer cells, or a combination of AML cells and breast cancer cells.
  • AML acute myeloid leukemia
  • the EVs are electroporated with ASO antagonizing miR-125b for knockdown of miR-125b in target cells as described above.
  • the ASO antagonizing miR-125b comprises or consists of SEQ ID NO: 1.
  • the growth of the target cells is suppressed.
  • the EVs are electroporated with a small chemical such as dextran.
  • the method comprises administering to the target cells the RNA-loaded EVs which modulate an apoptosis-related gene expression, thereby inducing apoptosis in the target cells.
  • a method for delivery of an antisense oligonucleotide (ASO) to target cells to suppress gene expression comprising the steps of: a) purification of extracellular vesicles (EVs) from red blood cells (RBCs); b) electroporation of the EVs with RNAs to form RNA-loaded EVs; and c) applying the RNA-loaded EVs to the target cells.
  • ASO antisense oligonucleotide
  • the RBCs are derived from a mammal preferably a human, and treated with ionophore in particular calcium ionophore.
  • the RNA is an ASO antagonizing miR-125b to inhibit the oncogenic miR-125b in the target cells.
  • the ASO antagonizing miR-125b comprises or consists of SEQ ID NO: 1.
  • the target cells comprise cancer cells or are cancer cells.
  • the target cells comprise leukemia cells in particular AML, cells, breast cancer cells, or a combination of AML cells and breast cancer cells.
  • a method of RNA delivery to target cells for a CRISPR genome editing system comprising the steps of: a) purification of extracellular vesicles (EVs) from red blood cells (RBCs), wherein the RBCs are preferably derived from a human and treated with ionophore in particular calcium ionophore; b) electroporation of the EVs with RNAs which may be Cas9 mRNAs and/or gRNAs to form RNA-loaded EVs; and c) applying the RNA-loaded EVs to the target cells.
  • CRISPR is a method that enables robust and precise modifications of genomic DNA for a wide range of applications in research and medicine. The system can be designed to target genomic DNA directly.
  • the EVs are electroporated with Cas9 mRNA and gRNA.
  • Cas9 mRNA comprises or consists of SEQ ID NO: 2.
  • the gRNA is eGFP gRNA comprising or consisting of SEQ ID NO: 3.
  • the EVs are electroporated with Cas9 and gRNA plasmids.
  • the target cells comprise cancer cells or are cancer cells.
  • the target cells comprise leukemia cells or are leukemia cells.
  • the target cells comprise leukemia cells in particular AML cells, breast cancer cells, or a combination of AML cells and breast cancer cells.
  • a method of treating cancer by delivery of RNA to target cells comprising the steps of: a) purification of extracellular vesicles (EVs) from red blood cells (RBCs) which are preferably derived from a mammal in particular a human and treated with ionophore in particular calcium ionophore; b) electroporation of the EVs with RNAs to form RNA-loaded EVs; and c) applying the RNA-loaded EVs to the target cells thereby inhibiting the growth of the target cells, wherein the target cells comprise cancer cells.
  • EVs extracellular vesicles
  • RBCs red blood cells
  • the target cells comprise leukemia cells, breast cancer cells, or a combination of leukemia cells and breast cancer cells. In another embodiment, the target cells comprise acute myeloid leukemia cells.
  • the step c) comprises a step of administering the RNA-loaded EVs to a subject having the target cells via a local or systemic administration.
  • Local administration refers to the delivery of the RNA-loaded EVs directly to the site of action, and includes, but not limiting to, intratumoral administration.
  • Systemic administration refers to the delivery of the RNA-loaded EVs via circulatory system, and includes, but not limiting to, intravenous injection.
  • the growth of the target cells is suppressed after the step c).
  • FIG. 1 a is a schematic diagram showing the process of collecting the Evs from human red blood cells (RBCs).
  • FIG. 1 b is a plot showing the concentration and the size distribution of RBC EVs.
  • FIG. 1 c shows the expression of ALIX, TSG101, and hemoglobin A in cell lysates and EVs, via Western blot analysis.
  • FIG. 2 a is a schematic presentation of EV electroporation.
  • FIG. 2 b show the results obtained from FACS analysis of AF647 fluorescence and forward scatter (FSC) of the beads that were incubated with electroporated EVs (E-EVs) or unelectroporated EVs (UE-EVs).
  • FSC fluorescence and forward scatter
  • FIG. 3 a is a schematic presentation of the EV uptake assay.
  • FIG. 3 b shows the expression of HBA relative to GAPDH, via Western blot analysis.
  • FIG. 3 c shows the uptake of RBC EVs by leukemia MOLM13 cells, via FACS analysis.
  • FIG. 4 a is a schematic presentation of Dextran delivery.
  • FIG. 4 b shows RBC EVs deliver dextran to leukemia MOLM13 cells, via FACS analysis.
  • FIG. 5 a is a schematic presentation of ASO delivery.
  • FIG. 5 b shows the results obtained from FACS analysis, where the MOLM13 cells were untreated or incubated with FAM ASO or with electroporated EVs (E-EVs) or with unelectroporated EVs (UE-EVs).
  • FIG. 5 c is a plot showing the results of FIG. 5 b .
  • FIG. 5 d is a diagram showing the average percentage of FAM-positive cells after treatments. In particular, the results reveal that RBC EVs deliver antisense oligonucleotides (ASO) to leukemia MOLM13 cells.
  • ASO antisense oligonucleotides
  • FIG. 6 a shows the results obtained from FACS analysis of AF647 fluorescence in MOLM13 cells that were untreated, incubated with Dextran AF647 (Dex-647) alone, with Dex-647 and unelectroporated RBC EVs (UE-EVs), with Dex-647 loaded LipofectaminTM 3000 (Lipo3000), with Dex-647 loaded INTERFERin or with Dex-647 electroporated RBC EVs (E-EVs) for 24 hours.
  • UE-EVs Dex-647 loaded LipofectaminTM 3000 (Lipo3000)
  • E-EVs Dex-647 loaded INTERFERin
  • 6 b shows the results obtained from FACS analysis of FAM fluorescence in MOLM13 cells that were untreated, incubated with FAM-ASO alone, with FAM-ASO and unelectroporated RBC EVs (UE-EVs), with FAM-ASO loaded Lipo3000, with FAM-ASO loaded INTERFERin or with FAM-ASO electroporated RBC EVs (E-EVs) for 24 hours.
  • UE-EVs FAM-ASO loaded Lipo3000
  • E-EVs FAM-ASO electroporated RBC EVs
  • FIG. 7 a shows the percentage of cell death/viability of MOLM13 cells after treatments with Dextran AF647 (Dex-647) alone, with Dex-647 and unelectroporated RBC EVs (UE-EVs), with Dex-647 loaded LipofectaminTM 3000 (Lipo3000), with Dex-647 loaded INTERFERin or with Dex-647 electroporated RBC EVs (E-EVs) for 24 hours.
  • Dextran AF647 Dex-647
  • UE-EVs Dex-647 and unelectroporated RBC EVs
  • Dex-647 loaded LipofectaminTM 3000 Lipo3000
  • Dex-647 loaded INTERFERin or with Dex-647 electroporated RBC EVs (E-EVs) for 24 hours.
  • FIG. 7 a shows the percentage of cell death/viability of MOLM13 cells after treatments with Dextran AF647 (Dex-647) alone, with Dex-647 and une
  • 7 b shows the percentage of cell death/viability of MOLM13 cells after treatments with FAM-ASO alone, with FAM-ASO and unelectroporated RBC EVs (UE-EVs), with FAM-ASO loaded Lipo3000, with FAM-ASO loaded INTERFERin or with FAM-ASO electroporated RBC EVs (E-EVs) for 24 hours
  • FIG. 8 a is a schematic presentation of miR-125b ASO delivery.
  • FIG. 8 b shows the expression of miR-125b in MOLM13 cells after treatment with unelectroporated RBC EVs (UE-EVs), with negative control (NC)-ASO electroporated RBC EVs (E-EVs) or anti-miR-125b ASO (125b-ASO) electroporated RBC EVs for 72 hours.
  • UE-EVs unelectroporated RBC EVs
  • NC negative control
  • E-EVs negative control
  • 125b-ASO anti-miR-125b ASO
  • FIG. 8 c shows the expression of miR-125a in MOLM13 cells after treatment with unelectroporated RBC EVs (UE-EVs), with negative control (NC)-ASO electroporated RBC EVs (E-EVs) or anti-miR-125b ASO (125b-ASO) electroporated RBC EVs for 72 hours.
  • FIG. 8 d shows the expression of BAK1 relative to GAPDH in MOLM13 cells treated the same as in FIGS. 8 b and 8 c .
  • FIG. 8 e shows the number of MOLM13 cells untreated, or treated with UE-EVs or with ASO electroporated EVs as indicated.
  • FIG. 9 a is a schematic presentation of miR-125b ASO delivery.
  • FIG. 9 b shows the expression of miR-125b in CA1a cells after treatment with unelectroporated RBC EVs (UE-EVs), with negative control (NC)-ASO electroporated RBC EVs (E-EVs) or anti-miR-125b ASO (125b-ASO) electroporated RBC EVs for 72 hours.
  • UE-EVs unelectroporated RBC EVs
  • NC negative control
  • E-EVs negative control
  • 125b-ASO anti-miR-125b ASO
  • FIG. 9 c shows the expression of miR-125a in CA1a cells after treatment with unelectroporated RBC EVs (UE-EVs), with negative control (NC)-ASO electroporated RBC EVs (E-EVs) or anti-miR-125b ASO (125b-ASO) electroporated RBC EVs for 72 hours.
  • FIG. 9 d shows the results of crystal violet staining of CA1a cells after treatments as indicated above.
  • FIG. 10 a is a schematic presentation of Cas9 mRNA delivery.
  • FIG. 10 b shows the levels of Cas9 mRNA in MOLM13 cells after treatment with unelectroporated EVs or with EVs that were electroporated with 5, 10 or 20 ⁇ g Cas9 mRNA, determined by qRT-PCR after 24 hours of treatment.
  • FIG. 10 c show representative images of MOLM13 cells after treatments as indicated above.
  • FIG. 10 d shows the average percentage of MOLM13 cells stained positive for HA-Cas9 protein as shown in FIG. 10 c.
  • FIG. 11 a is a schematic presentation of the RNA delivery.
  • FIG. 11 b shows the results obtained from FACS analysis of GFP in NOMO1-GFP cells after treatment with unelectroporated EVs or EVs electroporated with Cas9 and gRNA.
  • FIG. 12 a is a schematic presentation of plasmid delivery.
  • FIG. 12 b shows the results obtained from FACS analysis of GFP in 293T-eGFP cells untreated, or incubated with unelectroporated EVs (UE-EVs) or with plasmid electroporated EVs (E-EVs) as indicated.
  • FIG. 12 c shows the plot prepared from the results of FIG. 12 b.
  • FIG. 13 a is a schematic presentation of an in vivo EV uptake assay.
  • FIG. 13 b shows the fluorescent images of nude mice bearing untreated tumors on the right and tumors injected with PKH26-labeled EVs on the left.
  • FIG. 13 c shows the ex vivo fluorescent images of the tumors at 72 hours post-treatment.
  • FIG. 13 d shows the total radiance efficiency (photons/second) of fluorescent signals in the tumors 24-72 hours after the injection of PKH26-labeled EVs.
  • the present invention relates to the field of molecular biology and genome editing. More specifically the transfer of genetic materials to recipient cells by extracellular vesicles (EVs) and the method of purification or isolation of exosomes from Red Blood Cells.
  • EVs extracellular vesicles
  • EVs Extracellular vesicles
  • EVs secreted by many cell types contain RNAs that function to alter the phenotypes of other cells.
  • EVs contain not only RNAs but also proteins that stabilize RNAs and facilitate the functions of RNAs in the target cells.
  • EV-mediated delivery of RNAs is an attractive platform because the natural biocompatibility of EVs is the solution to overcome most in vivo delivery hurdles.
  • EVs are generally nontoxic and non-immunogenic. They are taken up readily by many cell types but they do possess some antiphagocytic markers such as CD47 that help them to evade the phagocytosis by macrophages of the reticuloendothelial system.
  • EVs are able to extravasate well through the interendothelial junctions and even cross the blood-brain barrier hence, they are greatly versatile drug carriers.
  • delivery by EVs is not hampered by the multidrug resistance mechanism caused by overexpression of P-glycoproteins that tumor cells often exhibit to eliminate many chemical compounds.
  • EVs from cancer cell lines and stem cells which are very costly due to the large-scale cell culture that requires various supplements.
  • EVs from cancer and stem cells may contain oncogenic proteins or growth factors that promote cancer growth.
  • EVs from plasma and blood cells are safer for cancer therapies.
  • RBCs EVs do not contain oncogenic DNA/RNA or growth factors that are usually abundant in EVs from cancer cells or stem cells, hence, RBC EVs do not post any transformation risks to recipient cells.
  • RBCs EVs are also nontoxic unlike synthetic transfection reagents.
  • an RNA is selected to inhibit expression of a target gene by binding to a miRNA or editing the target genomic DNA. Further, there is provided a novel method for the purification of EVs from red blood cells (RBCs) and incorporation of RNAs in EVs for gene therapies against cancer, including acute myeloid leukemia and breast cancer.
  • RBCs red blood cells
  • RBCs Blood samples were obtained by Red Cross from healthy donors in Hong Kong with informed consents.
  • RBCs were separated from plasma and white blood cells by centrifugation and treated with 10 mM calcium ionophore (Sigma) overnight.
  • the purification of EVs were optimized with multiple centrifugation steps including the removal of protein contamination using a 60% sucrose cushion (ultracentrifugation at 100,000 ⁇ g) that yields a homogenous population of EVs with an average diameter of ⁇ 140 nm.
  • These EVs are enriched in EV markers, ALIX and TSG101, as shown by Western blot analysis. They also contain hemoglobin A which is a major protein from RBCs.
  • FIG. 1 a Culture supernatants were collected from ionophore-treated human red blood cells and subjected to multiple steps of centrifugation to remove dead cells and debris. EVs were purified by ultracentrifugation with 60% sucrose cushion and washed with phosphate buffer saline (PBS) by ultracentrifugation (100,000 ⁇ g).
  • FIG. 1 b Concentrations and the size distribution of RBC EVs were measured by a Nanosight nanoparticle analyzer.
  • FIG. 1 c Western blot analysis of ALIX, TSG101 (EV markers) and Hemoglobin A (RBC marker) relative to GAPDH in the cell lysate and EVs purified from RBCs.
  • an electroporation protocol was optimized for the RBC EVs using Dextran conjugated with Alexa Fluor® 647 (AF647, Thermo Fisher Scientific) tested at different voltages using a Gene Pulser Xcell electroporator (BioRad). Electroporated EVs were added to latex beads and analyzed for AF647 using flow cytometry. It was found that 250 V was the optimal voltage, which resulted in 93.6% AF647 positive EV-bound beads.
  • FIG. 2 a Schematic presentation of EV electroporation: 50 ⁇ g RBC EVs were mixed with 4 ⁇ g Alexa Fluor® 647 (AF647) labeled Dextran and electroporated at different voltages from 50 to 250 V. EVs were incubated with latex beads overnight and analyzed by fluorescent activated cell sorting (FACS).
  • FIG. 2 b FACS analysis of AF647 fluorescence and forward scatter (FSC) of the beads that were incubated with electroporated EVs (E-EVs) or unelectroporated EVs (UE-EVs). The percentage of AF647 positive beads are indicated above the gates.
  • FSC fluorescent activated cell sorting
  • the RBC-derived EVs was labelled with a fluorescent membrane dye called Bodipy® TR (Thermo Fisher). Labeled EVs were washed extensively using the sucrose cushion, mock electroporated and added to the AML MOLM13 cells. After 24 hours of incubation with EVs, Western blot analysis of MOML13 cells showed a clear uptake of Hemoglobin A (HBA) protein which was absent in the untreated cells. Importantly, treatment with RBC EVs did not affect the viability of AML cells as shown by FACS analysis.
  • HBA Hemoglobin A
  • MOLM13 cells became 100% Bodipy positive after the incubation with Bodipy-labeled EVs, indicating that all the cells took up the fluorescent RBC EVs. Electroporation increased the uptake of HBA but not Bodipy by MOLM13 cells.
  • FIG. 3 a Schematic presentation of the EV uptake assay: 50 ⁇ g RBC EVs were labeled with Bodipy TR (a red fluorescent dye), washed twice, mock electroporated at 250 V, and incubated with MOLM13 cells for 24 hours.
  • FIG. 3 b Western blot analysis of hemoglobin A (HBA) relative to GAPDH and
  • FIG. 3 c FACS analysis of live cells, gated based on size scatter (SSC) and forward scatter (FSC), and BODIPY fluorescence in MOLM13 cells that were untreated or incubated with electroporated EVs (E-EVs) or unelectroporated EVs (UE-EVs).
  • SSC size scatter
  • FSC forward scatter
  • FIG. 4 a Schematic presentation of Dextran delivery: 50-100 ⁇ g RBC EVs were mixed with 4 ⁇ g Dextran AF647 and electroporated at 250 V. Electroporated EVs were incubated with MOLM13 cells for 24 hours.
  • FIG. 4 b FACS analysis of Dextran AF647 fluorescence in MOLM13 cells that were untreated or incubated with 50-100 ⁇ g Dextran AF647 electroporated EVs (E-EVs) or 100 ⁇ g unelectroporated (UE-EVs).
  • E-EVs E-EVs
  • UE-EVs unelectroporated
  • RNA oligonucleotide green fluorescent labeled scrambled RNA oligonucleotide (Shanghai GenePharma), about 7 kDa, that is often used as a negative control antisense oligonucleotide (ASO).
  • FAM green fluorescent labeled scrambled RNA oligonucleotide
  • MOLM13 cells MOLM13 cells
  • FIG. 5 a Schematic presentation of ASO delivery: 75 ⁇ g RBC EVs were electroporated with 400 pmole FAM fluorescent labeled scrambled ASO ( ⁇ 7 kDa) and incubated with MOLM13 cells for 24 hours.
  • FIGS. 5 b -5 d FACS analysis of FAM fluorescence in MOLM13 cells that were untreated or incubated with FAM ASO or with electroporated EVs (E-EVs) or with unelectroporated EVs (UE-EVs). The average percentage+SEM of FAM-positive cells were calculated from 3 independent experiments as shown in FIG. 5 d.
  • Dextran AF647 and FAM ASO were then compared with that of two commercialized lipofection reagents, LipofectamineTM 3000 (Thermo Fisher Scientific) and INTERFERinTM (Polyplus transfection) that are commonly used for transfection of nucleic acids in mammalian cells. Consistent with previous experiments, RBC EVs delivered Dextran AF647 and FAM ASO to ⁇ 75% MOLM13 cells. LipofectamineTM 3000 archived only 3% and 55% delivery of Dextran AF647 and FAM ASO whereas INTERFERin archived only 2.7% and 38.7% delivery of Dextran AF647 and FAM ASO respectively in MOML13 cells.
  • LipofectamineTM 3000 and INTERFERin were not a surprise since blood cells including AML cells are referred to as “difficult-to-transfect” cell types by the manufacturers. Hence, the 75% delivery efficiency archived by RBC EVs was a great improvement.
  • FIG. 6 a FACS analysis of AF647 fluorescence in MOLM13 cells that were untreated, incubated with 4 ⁇ g Dextran AF647 (Dex-647) alone, with Dex-647 and unelectroporated RBC EVs (UE-EVs), with Dex-647 loaded LipofectaminTM 3000 (Lipo3000), with Dex-647 loaded INTERFERin or with Dex-647 electroporated RBC EVs (E-EVs) for 24 hours.
  • UE-EVs Dex-647 loaded LipofectaminTM 3000 (Lipo3000)
  • E-EVs Dex-647 loaded INTERFERin
  • 6 b FACS analysis of FAM fluorescence in MOLM13 cells that were untreated, incubated with 2 ⁇ mole FAM-ASO alone, with FAM-ASO and unelectroporated RBC EVs (UE-EVs), with FAM-ASO loaded Lipo3000, with FAM-ASO loaded INTERFERin or with FAM-ASO electroporated RBC EVs (E-EVs) for 24 hours.
  • UE-EVs FAM-ASO and unelectroporated RBC EVs
  • E-EVs FAM-ASO electroporated RBC EVs
  • RBC EVs exhibit no toxicity to the cells in contrast to about 20-30% increase in cell death caused by the transfection using LipofectamineTM 3000 and INTERFERin.
  • FIG. 7 a MOLM13 cells that were untreated, incubated with 4 ⁇ g Dextran AF647 (Dex-647) alone, with Dex-647 and unelectroporated RBC EVs (UE-EVs), with Dex-647 loaded LipofectaminTM 3000 (Lipo3000), with Dex-647 loaded INTERFERin or with Dex-647 electroporated RBC EVs (E-EVs) for 24 hours;
  • PI Propidium iodide
  • miR-125b is upregulated in different types of cancer including AML and other leukemia. It has been shown that miR-125b suppresses apoptosis by regulating multiple genes in the p53 network. miR-125b also promotes proliferation of hematopoietic stem cells and leukemia cells in both humans and mouse models.
  • An anti-miR-125b ASO (Shanghai Gene Pharma) comprising a sequence of SEQ ID NO: 1 was loaded into RBC EVs using electroporation and treated MOLM13 cells with these EVs.
  • miR-125b the level of miR-125b was suppressed by 80-95% in a dose-dependent manner.
  • miR-125a the homologue of miR-125b, was also suppressed by 50-80% due to the sequence similarity to miR-125b.
  • Inhibition of miR-125 led to a significant increase in BAK1, a target of miR-125a/b which regulates apoptosis.
  • Treatment with miR-125b ASO loaded EVs also dampened the growth of MOLM13 cells significantly after 3-4 days of incubation. Hence, the inhibition of miR-125b using ASO in RBC EVs may represent a new approach for AML treatment.
  • miR-125b preferably comprises or consists of SEQ ID NO: 4 and miR-125a preferably comprises or consists of SEQ ID NO: 5.
  • miR-125b consists of SEQ ID NO: 4 and miR-125a consists of SEQ ID NO: 5.
  • FIG. 8 a Schematic presentation of miR-125b ASO delivery: 25-100 ⁇ g RBC EVs were electroporated with 2 ⁇ mole anti-miR-125b ASO and incubated with MOLM13 cells.
  • Anti-miR-125b ASO in this embodiment consists of SEQ ID NO: 1.
  • FIGS. 8 a Schematic presentation of miR-125b ASO delivery: 25-100 ⁇ g RBC EVs were electroporated with 2 ⁇ mole anti-miR-125b ASO and incubated with MOLM13 cells.
  • Anti-miR-125b ASO in this embodiment consists of SEQ ID NO: 1.
  • FIG. 8 b - c Expression of miR-125b and miR-125a relative to U6b snRNA in MOLM13 cells that were untreated, incubated with 100 ug unelectroporated RBC EVs (UE-EVs), with negative control (NC)-ASO electroporated RBC EVs (E-EVs) or anti-miR-125b ASO (125b-ASO) electroporated RBC EVs for 72 hours, as determined by Taqman qRT-PCR, presented as average and SEM.
  • FIG. 8 d Expression of BAK1 relative to GAPDH in MOLM13 cells treated the same as in FIG. 8 b .
  • FIG. 8 e Number of MOLM13 cells untreated, or treated with UE-EVs or with ASO electroporated EVs as indicated.
  • One-way Anova test ** P ⁇ 0.01; *** P ⁇ 0.001.
  • RBC EVs were tested for the delivery of miR-125b ASO to breast cancer MCF10aCA1a (CA1a) cells.
  • the inventors observed 80-90% knockdown of miR-125a and miR-125b in CA1a cells treated with miR-125b ASO loaded EVs. As a consequence, the knockdown of miR-125s suppressed the proliferation of CA1a cells.
  • FIG. 9 a Schematic presentation of miR-125b ASO delivery: 25-50 ⁇ g RBC EVs were electroporated with 2 ⁇ mole anti-miR-125b ASO and incubated with CA1a cells.
  • FIG. 9 b - c Expression of miR-125b and miR-125a relative to U6b snRNA in CA1a cells that were untreated, incubated with unelectroporated RBC EVs (UE-EVs), with negative control (NC)-ASO electroporated RBC EVs (E-EVs) or anti-miR-125b ASO (125b-ASO) electroporated RBC EVs for 72 hours, as determined by Taqman qRT-PCR, presented as average and SEM.
  • UE-EVs unelectroporated RBC EVs
  • NC negative control
  • E-EVs negative control
  • E-EVs anti-miR-125b ASO
  • 125b-ASO anti-mi
  • FIG. 10 a Schematic presentation of Cas9 mRNA delivery: RBC EVs were electroporated with Cas9 mRNA and incubated with MOLM13 cells for 24 or 48 hours.
  • FIG. 10 a Schematic presentation of Cas9 mRNA delivery: RBC EVs were electroporated with Cas9 mRNA and incubated with MOLM13 cells for 24 or 48 hours.
  • FIG. 10 b The levels of Cas9 mRNA relative to GAPDH mRNA in MOLM13 cells that were untreated, incubated with unelectroporated EVs or with EVs that were electrop
  • FIG. 10 c Representative images of MOLM13 cells that were untreated, or incubated for 48 hours with unelectroporated EVs or with EVs that were electroporated with 10 ⁇ g Cas9 mRNAs. The cells were stained for HA-Cas9 protein (using green dye, not seen in black and white images) and nuclear DNA (Hoechst, blue dye, not seen in black and white images).
  • FIG. 10 d Average percentage of MOLM13 cells stained positive for HA-Cas9 protein as shown in (c).
  • Cas9 mRNA preferably comprises or consists of SEQ ID NO: 2 and eGFP gRNA preferably comprises or consists of SEQ ID NO: 3.
  • Cas9 mRNA consists of SEQ ID NO: 2 and eGFP gRNA consists of SEQ ID NO: 3.
  • FIG. 11 a Schematic presentation of the RNA delivery: RBC EVs were electroporated with Cas9 mRNA and anti-GFP gRNA and incubated with NOMO1-GFP cells for 7 days.
  • FIG. 11 b FACS analysis of GFP in NOMO1-GFP cells that were untreated, incubated with unelectroporated EVs or EVs electroporated with Cas9 and gRNA. The percentages of GFP-negative cells are shown above the gate.
  • RBC EVs were electroporated with two plasmids, one expressing SpCas9 and one expressing gRNA against eGFP. Electroporated EVs were incubated with human embryonic kidney HEK-293T cells that homogenously express eGFP. After 96 hours, it was found that 13.8% GFP-negative cells resulted from the EV treatment, compared to 3.52% GFP-negative in the untreated population. Treatment with electroporated EVs showed a distinct peak of GFP-negative cells that suggests a homologous knockout of eGFP by the delivery of Cas9 and gRNA plasmids. Therefore, RBC EVs are able to deliver not only RNA but also plasmid DNA for genome editing. Moreover, the delivery is applicable to HEK-293T solid cancer cells.
  • FIG. 12 a Schematic presentation of plasmid delivery: RBC EVs were electroporated with Cas9 plasmid and eGFP gRNA plasmid and incubated with eGFP expressing 293T cells for 96 hours.
  • FIGS. 12 b -12 c FACS analysis of GFP in 293T-eGFP cells untreated, or incubated with unelectroporated EVs (UE-EVs) or with plasmid electroporated EVs (E-EVs) as indicated.
  • the GFP negative cells are indicated by the percentages in FIG. 12 b and the arrow in FIG. 12 c.
  • FIG. 13 a Schematic presentation of an in vivo EV uptake assay.
  • FIG. 13 b Fluorescent images of nude mice bearing untreated tumors on the top and tumors injected with PKH26-labeled (red dye, not seen in black and white images) EVs on the bottom.
  • FIG. 13 c Ex vivo fluorescent images of the tumors at 72 hours post-treatment.
  • FIG. 13 d Total radiance efficiency (photons/second) of fluorescent signals in the tumors 24-72 hours after the injection of PKH26-labeled EVs.
  • CA1a cells were implanted in the mice, in the flank at 2 sides ( FIG.
  • FIG. 13 a shows ex vivo fluorescent images of the tumors at 72 hours post-treatment and proves that PKH26-labeled EVs were taken up by tumor cells.
  • the total radiance efficiency (photons/second) of fluorescent signals in the tumors decreased gradually 24 to 72 hours after the injection of PKH26-labeled EVs as shown in FIG. 13 d.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Genetics & Genomics (AREA)
  • Chemical & Material Sciences (AREA)
  • Biomedical Technology (AREA)
  • Biotechnology (AREA)
  • General Health & Medical Sciences (AREA)
  • Molecular Biology (AREA)
  • Zoology (AREA)
  • Organic Chemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Wood Science & Technology (AREA)
  • Medicinal Chemistry (AREA)
  • General Engineering & Computer Science (AREA)
  • Biochemistry (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Physics & Mathematics (AREA)
  • Microbiology (AREA)
  • Hematology (AREA)
  • Cell Biology (AREA)
  • Biophysics (AREA)
  • Immunology (AREA)
  • Plant Pathology (AREA)
  • Virology (AREA)
  • Developmental Biology & Embryology (AREA)
  • Food Science & Technology (AREA)
  • Analytical Chemistry (AREA)
  • General Physics & Mathematics (AREA)
  • Urology & Nephrology (AREA)
  • Pathology (AREA)
  • Ecology (AREA)
  • Mycology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)

Abstract

A method of RNA delivery using extracellular vesicles (EVs) derived from red blood cells (RBCs). The method comprises the purification and electroporation of the EVs and applying the RNA-loaded EVs to target cells. The method further comprises the treatment of cancer using the RNA-loaded EVs.

Description

    SEQUENCE LISTING
  • The Sequence Listing file entitled “sequencelisting” having a size of 6,395 bytes and a creation date of Aug. 16, 2017, that was filed with the patent application is incorporated herein by reference in its entirety.
  • FIELD OF INVENTION
  • The present invention relates to the field of molecular biology and genome editing, more specifically the transfer of genetic materials to recipient cells by extracellular vesicles (EVs).
  • BACKGROUND
  • RNA therapeutics including antisense oligo nucleotides (ASOs), small-interfering RNA (siRNAs), synthetic mRNAs and genome editing RNA-protein complexes are emerging modalities for therapies targeting the human genomes at high specificity and great flexibility. ASOs and siRNAs have been widely used as the tools for gene knockdown in biomedical research. Their ability to silence any gene of interest offers a great potential for targeting disease-prevalent genes. Various chemical modifications or conjugations can be used to keep ASOs and siRNAs stable and enhance their binding specificity. Common methods for RNA transfection including nucleofection, lipofection and electroporation are only suitable for ex vivo delivery. Viral transduction and nanoparticles are often used for in vivo delivery of RNAs and DNAs however, these methods are usually ineffective, immunogenic and toxic.
  • One of the most recent breakthroughs in Science is a new technology for genome editing, the clustered regularly interspaced short palindromic repeats (CRISPR) method that enables robust and precise modifications of genomic DNA for a wide range of applications in research and medicine. CRISPR is an ideal tool for correction of genetic abnormalities in cancer as the system can be designed to target genomic DNA directly. A CRISPR system involves two main components: a Cas9 enzyme and a guide (gRNA). The gRNA contains a targeting sequence for DNA binding and a scaffold sequence for Cas9 binding. Cas9 nuclease is often used to “knockout” target genes hence it can be applied for deletion or suppression of oncogenes that are essential for cancer initiation or progression. Similar to ASOs and siRNAs, the CRISPR system offers a great flexibility in targeting any gene of interest hence, potential CRISPR based therapies can be designed based on the genetic mutation in individual patients. An advantage of the CRISPR system is its ability to completely ablate the expression of disease genes which can only be suppressed partially by RNA interference methods with ASOs or siRNAs. Furthermore, multiple gRNAs can be employed to suppress or activate multiple genes simultaneously, hence increasing the treatment efficacy and reducing resistance potentially caused by new mutations in the target genes. The applications of CRISPR technology have evolved very quickly from bench to bedside targeting different diseases. Clinical trials of CRISPR-mediated modification of T cells for cancer therapies have started in China and in the USA. Many other CRISPR-based therapies are under development. However, most of these therapies rely on ex vivo modification of the target cells or systemic delivery of the CRISPR system using virus or nanoparticles that can target very few cell types such as hepatocytes.
  • Acute myeloid leukemia (AML) is the most aggressive type of blood cancer that affects nearly 352,000 people per year with the 5-year prevalence of 1.5%. AML is characterized by the increase of myeloblasts in the peripheral blood (PB) and the bone marrow (BM). 30-40% AML patients (mostly under 60 years old) response well to chemotherapy and hematopoietic stem cell transplantation. However, the response rate is much lower in older patients as they cannot tolerate the toxicity of chemotherapy. Moreover, almost all the patients relapse after a certain time due to drug resistance. Hence, new treatment strategies are desirable to increase the response rate, reduce toxicity and combat drug resistance. Recent advances in genomics have provided better understanding of the genetic and epigenetic abnormalities in AML and suggest new specific therapeutic targets. RNA interference and genome editing methods are emerging as new approaches to target these abnormalities. However, delivery of RNAs to AML cells for gene therapies has proven challenging, especially for in vivo treatments. Common gene therapy delivery vehicles such as adeno-associated virus (AAV) and lipid nanoparticles (LNPs) are mostly ineffective or toxic in AML models.
  • Therefore, there is a desire to improve the delivery efficiency and reduce toxicity of gene therapies for cancer.
  • SUMMARY OF INVENTION
  • In one aspect of the invention, there is provided a method for RNA delivery to target cells comprising the steps of: a) purification of extracellular vesicles (EVs) from red blood cells (RBCs); b) electroporation of the EVs with RNAs to form RNA-loaded EVs; and c) applying the RNA-loaded EVs to the target cells.
  • The advantage of using EVs (including microvesicles and exosomes) from RBCs is that the RBCs are the most abundant blood cells hence a large amount of EVs can be obtained and purified from RBC units that are available at any blood bank. Preferably, the RBCs are derived from a human. They are also nontoxic, unlike synthetic transfection reagents. RBC EVs do not contain oncogenic DNA/RNA or growth factors that are usually abundant in EVs from cancer cells or stem cells, hence RBC EVs do not post any transformation risks to recipient cells.
  • In one embodiment, the RBCs are derived from a mammal preferably a human and treated with ionophore in particular calcium ionophore. The EVs are purified using ultracentrifugation with a sucrose cushion. The term “sucrose cushion” refers to a sucrose gradient which establishes itself during a centrifugation. In an embodiment, the sucrose gradient is prepared by using a solution of about 40% to about 70%, about 50% to about 60%, or about 60% of sucrose.
  • In another embodiment, the electroporated EVs comprises antisense oligonucleotides (ASO), mRNAs and plasmids. Preferably, the ASO comprises or consists of SEQ ID NO: 1.
  • In a further embodiment, the target cells comprise cancer cells, or are cancer cells. In another embodiment, the target cells comprise leukemia cells in particular acute myeloid leukemia (AML) cells, breast cancer cells, or a combination of AML cells and breast cancer cells.
  • In another embodiment, the EVs are electroporated with ASO antagonizing miR-125b for knockdown of miR-125b in target cells as described above. Preferably, the ASO antagonizing miR-125b comprises or consists of SEQ ID NO: 1.
  • In another embodiment, the growth of the target cells is suppressed. In a further embodiment, the EVs are electroporated with a small chemical such as dextran.
  • In another embodiment, the method comprises administering to the target cells the RNA-loaded EVs which modulate an apoptosis-related gene expression, thereby inducing apoptosis in the target cells.
  • In a second aspect of the invention, there is provided a method for delivery of an antisense oligonucleotide (ASO) to target cells to suppress gene expression, comprising the steps of: a) purification of extracellular vesicles (EVs) from red blood cells (RBCs); b) electroporation of the EVs with RNAs to form RNA-loaded EVs; and c) applying the RNA-loaded EVs to the target cells.
  • In an embodiment, as described above, the RBCs are derived from a mammal preferably a human, and treated with ionophore in particular calcium ionophore.
  • In one embodiment, the RNA is an ASO antagonizing miR-125b to inhibit the oncogenic miR-125b in the target cells. Preferably, the ASO antagonizing miR-125b comprises or consists of SEQ ID NO: 1.
  • In another embodiment, the target cells comprise cancer cells or are cancer cells. In another embodiment, the target cells comprise leukemia cells in particular AML, cells, breast cancer cells, or a combination of AML cells and breast cancer cells.
  • In a third aspect of the invention, there is provided a method of RNA delivery to target cells for a CRISPR genome editing system comprising the steps of: a) purification of extracellular vesicles (EVs) from red blood cells (RBCs), wherein the RBCs are preferably derived from a human and treated with ionophore in particular calcium ionophore; b) electroporation of the EVs with RNAs which may be Cas9 mRNAs and/or gRNAs to form RNA-loaded EVs; and c) applying the RNA-loaded EVs to the target cells. CRISPR is a method that enables robust and precise modifications of genomic DNA for a wide range of applications in research and medicine. The system can be designed to target genomic DNA directly.
  • In one embodiment, the EVs are electroporated with Cas9 mRNA and gRNA. Preferably, Cas9 mRNA comprises or consists of SEQ ID NO: 2. Further, the gRNA is eGFP gRNA comprising or consisting of SEQ ID NO: 3.
  • In another embodiment, the EVs are electroporated with Cas9 and gRNA plasmids. In another embodiment, the target cells comprise cancer cells or are cancer cells.
  • In a further embodiment, the target cells comprise leukemia cells or are leukemia cells. In a particular embodiment, the target cells comprise leukemia cells in particular AML cells, breast cancer cells, or a combination of AML cells and breast cancer cells.
  • In a fourth aspect of the invention, there is provided a method of treating cancer by delivery of RNA to target cells comprising the steps of: a) purification of extracellular vesicles (EVs) from red blood cells (RBCs) which are preferably derived from a mammal in particular a human and treated with ionophore in particular calcium ionophore; b) electroporation of the EVs with RNAs to form RNA-loaded EVs; and c) applying the RNA-loaded EVs to the target cells thereby inhibiting the growth of the target cells, wherein the target cells comprise cancer cells.
  • In one embodiment, the target cells comprise leukemia cells, breast cancer cells, or a combination of leukemia cells and breast cancer cells. In another embodiment, the target cells comprise acute myeloid leukemia cells.
  • In another embodiment, the step c) comprises a step of administering the RNA-loaded EVs to a subject having the target cells via a local or systemic administration. Local administration refers to the delivery of the RNA-loaded EVs directly to the site of action, and includes, but not limiting to, intratumoral administration. Systemic administration refers to the delivery of the RNA-loaded EVs via circulatory system, and includes, but not limiting to, intravenous injection.
  • In a further embodiment, the growth of the target cells is suppressed after the step c).
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1a is a schematic diagram showing the process of collecting the Evs from human red blood cells (RBCs). FIG. 1b is a plot showing the concentration and the size distribution of RBC EVs. FIG. 1c shows the expression of ALIX, TSG101, and hemoglobin A in cell lysates and EVs, via Western blot analysis.
  • FIG. 2a is a schematic presentation of EV electroporation. FIG. 2b show the results obtained from FACS analysis of AF647 fluorescence and forward scatter (FSC) of the beads that were incubated with electroporated EVs (E-EVs) or unelectroporated EVs (UE-EVs).
  • FIG. 3a is a schematic presentation of the EV uptake assay. FIG. 3b shows the expression of HBA relative to GAPDH, via Western blot analysis. FIG. 3c shows the uptake of RBC EVs by leukemia MOLM13 cells, via FACS analysis.
  • FIG. 4a is a schematic presentation of Dextran delivery. FIG. 4b shows RBC EVs deliver dextran to leukemia MOLM13 cells, via FACS analysis.
  • FIG. 5a is a schematic presentation of ASO delivery. FIG. 5b shows the results obtained from FACS analysis, where the MOLM13 cells were untreated or incubated with FAM ASO or with electroporated EVs (E-EVs) or with unelectroporated EVs (UE-EVs). FIG. 5c is a plot showing the results of FIG. 5b . FIG. 5d is a diagram showing the average percentage of FAM-positive cells after treatments. In particular, the results reveal that RBC EVs deliver antisense oligonucleotides (ASO) to leukemia MOLM13 cells.
  • FIG. 6a shows the results obtained from FACS analysis of AF647 fluorescence in MOLM13 cells that were untreated, incubated with Dextran AF647 (Dex-647) alone, with Dex-647 and unelectroporated RBC EVs (UE-EVs), with Dex-647 loaded Lipofectamin™ 3000 (Lipo3000), with Dex-647 loaded INTERFERin or with Dex-647 electroporated RBC EVs (E-EVs) for 24 hours. FIG. 6b shows the results obtained from FACS analysis of FAM fluorescence in MOLM13 cells that were untreated, incubated with FAM-ASO alone, with FAM-ASO and unelectroporated RBC EVs (UE-EVs), with FAM-ASO loaded Lipo3000, with FAM-ASO loaded INTERFERin or with FAM-ASO electroporated RBC EVs (E-EVs) for 24 hours.
  • FIG. 7a shows the percentage of cell death/viability of MOLM13 cells after treatments with Dextran AF647 (Dex-647) alone, with Dex-647 and unelectroporated RBC EVs (UE-EVs), with Dex-647 loaded Lipofectamin™ 3000 (Lipo3000), with Dex-647 loaded INTERFERin or with Dex-647 electroporated RBC EVs (E-EVs) for 24 hours. FIG. 7b shows the percentage of cell death/viability of MOLM13 cells after treatments with FAM-ASO alone, with FAM-ASO and unelectroporated RBC EVs (UE-EVs), with FAM-ASO loaded Lipo3000, with FAM-ASO loaded INTERFERin or with FAM-ASO electroporated RBC EVs (E-EVs) for 24 hours
  • FIG. 8a is a schematic presentation of miR-125b ASO delivery. FIG. 8b shows the expression of miR-125b in MOLM13 cells after treatment with unelectroporated RBC EVs (UE-EVs), with negative control (NC)-ASO electroporated RBC EVs (E-EVs) or anti-miR-125b ASO (125b-ASO) electroporated RBC EVs for 72 hours. FIG. 8c shows the expression of miR-125a in MOLM13 cells after treatment with unelectroporated RBC EVs (UE-EVs), with negative control (NC)-ASO electroporated RBC EVs (E-EVs) or anti-miR-125b ASO (125b-ASO) electroporated RBC EVs for 72 hours. FIG. 8d shows the expression of BAK1 relative to GAPDH in MOLM13 cells treated the same as in FIGS. 8b and 8c . FIG. 8e shows the number of MOLM13 cells untreated, or treated with UE-EVs or with ASO electroporated EVs as indicated.
  • FIG. 9a is a schematic presentation of miR-125b ASO delivery. FIG. 9b shows the expression of miR-125b in CA1a cells after treatment with unelectroporated RBC EVs (UE-EVs), with negative control (NC)-ASO electroporated RBC EVs (E-EVs) or anti-miR-125b ASO (125b-ASO) electroporated RBC EVs for 72 hours. FIG. 9c shows the expression of miR-125a in CA1a cells after treatment with unelectroporated RBC EVs (UE-EVs), with negative control (NC)-ASO electroporated RBC EVs (E-EVs) or anti-miR-125b ASO (125b-ASO) electroporated RBC EVs for 72 hours. FIG. 9d shows the results of crystal violet staining of CA1a cells after treatments as indicated above.
  • FIG. 10a is a schematic presentation of Cas9 mRNA delivery. FIG. 10b shows the levels of Cas9 mRNA in MOLM13 cells after treatment with unelectroporated EVs or with EVs that were electroporated with 5, 10 or 20 μg Cas9 mRNA, determined by qRT-PCR after 24 hours of treatment. FIG. 10c show representative images of MOLM13 cells after treatments as indicated above. FIG. 10d shows the average percentage of MOLM13 cells stained positive for HA-Cas9 protein as shown in FIG. 10 c.
  • FIG. 11a is a schematic presentation of the RNA delivery. FIG. 11b shows the results obtained from FACS analysis of GFP in NOMO1-GFP cells after treatment with unelectroporated EVs or EVs electroporated with Cas9 and gRNA.
  • FIG. 12a is a schematic presentation of plasmid delivery. FIG. 12b shows the results obtained from FACS analysis of GFP in 293T-eGFP cells untreated, or incubated with unelectroporated EVs (UE-EVs) or with plasmid electroporated EVs (E-EVs) as indicated. FIG. 12c shows the plot prepared from the results of FIG. 12 b.
  • FIG. 13a is a schematic presentation of an in vivo EV uptake assay. FIG. 13b shows the fluorescent images of nude mice bearing untreated tumors on the right and tumors injected with PKH26-labeled EVs on the left. FIG. 13c shows the ex vivo fluorescent images of the tumors at 72 hours post-treatment. FIG. 13d shows the total radiance efficiency (photons/second) of fluorescent signals in the tumors 24-72 hours after the injection of PKH26-labeled EVs.
  • DETAILED DESCRIPTION OF THE INVENTION
  • The present invention relates to the field of molecular biology and genome editing. More specifically the transfer of genetic materials to recipient cells by extracellular vesicles (EVs) and the method of purification or isolation of exosomes from Red Blood Cells.
  • Cells release into the extracellular environment, diverse types of membrane vesicles of endosomal and plasma membrane origin, called exosomes and microvesicles, respectively. These extracellular vesicles (EVs) represent an important mode of intercellular communication by serving as vehicles for transfer between cells of membrane and cytosolic proteins, lipids, and RNA.
  • EVs secreted by many cell types contain RNAs that function to alter the phenotypes of other cells. EVs contain not only RNAs but also proteins that stabilize RNAs and facilitate the functions of RNAs in the target cells.
  • EV-mediated delivery of RNAs is an attractive platform because the natural biocompatibility of EVs is the solution to overcome most in vivo delivery hurdles. EVs are generally nontoxic and non-immunogenic. They are taken up readily by many cell types but they do possess some antiphagocytic markers such as CD47 that help them to evade the phagocytosis by macrophages of the reticuloendothelial system. Moreover, EVs are able to extravasate well through the interendothelial junctions and even cross the blood-brain barrier hence, they are greatly versatile drug carriers. Of clinical value, delivery by EVs is not hampered by the multidrug resistance mechanism caused by overexpression of P-glycoproteins that tumor cells often exhibit to eliminate many chemical compounds.
  • For therapeutic delivery, many research groups have attempted to produce EVs from cancer cell lines and stem cells which are very costly due to the large-scale cell culture that requires various supplements. Moreover, EVs from cancer and stem cells may contain oncogenic proteins or growth factors that promote cancer growth. EVs from plasma and blood cells are safer for cancer therapies. RBCs EVs do not contain oncogenic DNA/RNA or growth factors that are usually abundant in EVs from cancer cells or stem cells, hence, RBC EVs do not post any transformation risks to recipient cells. RBCs EVs are also nontoxic unlike synthetic transfection reagents. A recent article by Wahlgren et al describes a protocol for isolation of plasma exosomes, small EVs that are derived from multivesicular bodies, and electroporation of these exosomes with siRNAs. They demonstrated that siRNA-loaded exosomes are taken up by monocytes and lymphocytes leading to significant knockdown of the target genes. This method is probably applicable to cancer therapies however, plasma exosomes are normally very heterogeneous because they are derived from different cell types in the circulation and the yield of exosomes from plasma is low. RBCs on the other hand are homogenous as RBCs from each individual are the same.
  • In the present invention, an RNA is selected to inhibit expression of a target gene by binding to a miRNA or editing the target genomic DNA. Further, there is provided a novel method for the purification of EVs from red blood cells (RBCs) and incorporation of RNAs in EVs for gene therapies against cancer, including acute myeloid leukemia and breast cancer.
  • EXAMPLES
  • The present invention is described by reference to the following Examples, which are offered by way of illustration and are not intended to limit the invention in any manner.
  • Example 1 Materials and Methods
  • Blood samples were obtained by Red Cross from healthy donors in Hong Kong with informed consents. RBCs were separated from plasma and white blood cells by centrifugation and treated with 10 mM calcium ionophore (Sigma) overnight. The purification of EVs were optimized with multiple centrifugation steps including the removal of protein contamination using a 60% sucrose cushion (ultracentrifugation at 100,000×g) that yields a homogenous population of EVs with an average diameter of ˜140 nm. Each unit of RBCs, isolated from ˜300 ml blood, yield 7.1 mg EVs on average. These EVs are enriched in EV markers, ALIX and TSG101, as shown by Western blot analysis. They also contain hemoglobin A which is a major protein from RBCs.
  • FIG. 1a : Culture supernatants were collected from ionophore-treated human red blood cells and subjected to multiple steps of centrifugation to remove dead cells and debris. EVs were purified by ultracentrifugation with 60% sucrose cushion and washed with phosphate buffer saline (PBS) by ultracentrifugation (100,000×g). FIG. 1b : Concentrations and the size distribution of RBC EVs were measured by a Nanosight nanoparticle analyzer. FIG. 1c : Western blot analysis of ALIX, TSG101 (EV markers) and Hemoglobin A (RBC marker) relative to GAPDH in the cell lysate and EVs purified from RBCs.
  • Subsequently, an electroporation protocol was optimized for the RBC EVs using Dextran conjugated with Alexa Fluor® 647 (AF647, Thermo Fisher Scientific) tested at different voltages using a Gene Pulser Xcell electroporator (BioRad). Electroporated EVs were added to latex beads and analyzed for AF647 using flow cytometry. It was found that 250 V was the optimal voltage, which resulted in 93.6% AF647 positive EV-bound beads.
  • FIG. 2a : Schematic presentation of EV electroporation: 50 μg RBC EVs were mixed with 4 μg Alexa Fluor® 647 (AF647) labeled Dextran and electroporated at different voltages from 50 to 250 V. EVs were incubated with latex beads overnight and analyzed by fluorescent activated cell sorting (FACS). FIG. 2b : FACS analysis of AF647 fluorescence and forward scatter (FSC) of the beads that were incubated with electroporated EVs (E-EVs) or unelectroporated EVs (UE-EVs). The percentage of AF647 positive beads are indicated above the gates.
  • To measure the uptake of EVs by AML cells, the RBC-derived EVs was labelled with a fluorescent membrane dye called Bodipy® TR (Thermo Fisher). Labeled EVs were washed extensively using the sucrose cushion, mock electroporated and added to the AML MOLM13 cells. After 24 hours of incubation with EVs, Western blot analysis of MOML13 cells showed a clear uptake of Hemoglobin A (HBA) protein which was absent in the untreated cells. Importantly, treatment with RBC EVs did not affect the viability of AML cells as shown by FACS analysis. MOLM13 cells became 100% Bodipy positive after the incubation with Bodipy-labeled EVs, indicating that all the cells took up the fluorescent RBC EVs. Electroporation increased the uptake of HBA but not Bodipy by MOLM13 cells.
  • FIG. 3a : Schematic presentation of the EV uptake assay: 50 μg RBC EVs were labeled with Bodipy TR (a red fluorescent dye), washed twice, mock electroporated at 250 V, and incubated with MOLM13 cells for 24 hours. FIG. 3b : Western blot analysis of hemoglobin A (HBA) relative to GAPDH and; FIG. 3c : FACS analysis of live cells, gated based on size scatter (SSC) and forward scatter (FSC), and BODIPY fluorescence in MOLM13 cells that were untreated or incubated with electroporated EVs (E-EVs) or unelectroporated EVs (UE-EVs).
  • Different amounts of EVs was further electroporated with Dextran AF647 and it was found that the best delivery with 75 μg EVs resulted in 68.6% cells positive for AF647. Therefore, 75 μg EVs was used for subsequent experiments.
  • FIG. 4a : Schematic presentation of Dextran delivery: 50-100 μg RBC EVs were mixed with 4 μg Dextran AF647 and electroporated at 250 V. Electroporated EVs were incubated with MOLM13 cells for 24 hours. FIG. 4b : FACS analysis of Dextran AF647 fluorescence in MOLM13 cells that were untreated or incubated with 50-100 μg Dextran AF647 electroporated EVs (E-EVs) or 100 μg unelectroporated (UE-EVs).
  • Testing the delivery of RNA was started with an FAM (green fluorescent) labeled scrambled RNA oligonucleotide (Shanghai GenePharma), about 7 kDa, that is often used as a negative control antisense oligonucleotide (ASO). RBC EVs were electroporated with the FAM ASO and incubated with MOLM13 cells. After 24 hours, it was observed that ˜70% uptake of FAM ASO by MOLM13 cells. Similar uptake was observed in NOMO-1 cells, another AML cell line (data not shown).
  • FIG. 5a : Schematic presentation of ASO delivery: 75 μg RBC EVs were electroporated with 400 pmole FAM fluorescent labeled scrambled ASO (˜7 kDa) and incubated with MOLM13 cells for 24 hours. FIGS. 5b-5d : FACS analysis of FAM fluorescence in MOLM13 cells that were untreated or incubated with FAM ASO or with electroporated EVs (E-EVs) or with unelectroporated EVs (UE-EVs). The average percentage+SEM of FAM-positive cells were calculated from 3 independent experiments as shown in FIG. 5 d.
  • The delivery of Dextran AF647 and FAM ASO by RBC EVs was then compared with that of two commercialized lipofection reagents, Lipofectamine™ 3000 (Thermo Fisher Scientific) and INTERFERin™ (Polyplus transfection) that are commonly used for transfection of nucleic acids in mammalian cells. Consistent with previous experiments, RBC EVs delivered Dextran AF647 and FAM ASO to ˜75% MOLM13 cells. Lipofectamine™ 3000 archived only 3% and 55% delivery of Dextran AF647 and FAM ASO whereas INTERFERin archived only 2.7% and 38.7% delivery of Dextran AF647 and FAM ASO respectively in MOML13 cells. The poor delivery observed with Lipofectamine™ 3000 and INTERFERin was not a surprise since blood cells including AML cells are referred to as “difficult-to-transfect” cell types by the manufacturers. Hence, the 75% delivery efficiency archived by RBC EVs was a great improvement.
  • FIG. 6a : FACS analysis of AF647 fluorescence in MOLM13 cells that were untreated, incubated with 4 μg Dextran AF647 (Dex-647) alone, with Dex-647 and unelectroporated RBC EVs (UE-EVs), with Dex-647 loaded Lipofectamin™ 3000 (Lipo3000), with Dex-647 loaded INTERFERin or with Dex-647 electroporated RBC EVs (E-EVs) for 24 hours. FIG. 6b : FACS analysis of FAM fluorescence in MOLM13 cells that were untreated, incubated with 2 μmole FAM-ASO alone, with FAM-ASO and unelectroporated RBC EVs (UE-EVs), with FAM-ASO loaded Lipo3000, with FAM-ASO loaded INTERFERin or with FAM-ASO electroporated RBC EVs (E-EVs) for 24 hours.
  • Moreover, RBC EVs exhibit no toxicity to the cells in contrast to about 20-30% increase in cell death caused by the transfection using Lipofectamine™ 3000 and INTERFERin.
  • The percentage of cell death was determined based on Propidium iodide (PI) staining and FACS analysis as shown in FIG. 7a : MOLM13 cells that were untreated, incubated with 4 μg Dextran AF647 (Dex-647) alone, with Dex-647 and unelectroporated RBC EVs (UE-EVs), with Dex-647 loaded Lipofectamin™ 3000 (Lipo3000), with Dex-647 loaded INTERFERin or with Dex-647 electroporated RBC EVs (E-EVs) for 24 hours; FIG. 7b : MOLM13 cells that were untreated, incubated with 2 μmole FAM-ASO alone, with FAM-ASO and unelectroporated RBC EVs (UE-EVs), with FAM-ASO loaded Lipo3000, with FAM-ASO loaded INTERFERin or with FAM-ASO electroporated RBC EVs (E-EVs) for 24 hours. The average cell death and SEM were calculated from three independent experiments. One-way Anova test: ** P<0.05; ** P<0.01.
  • The therapeutic potential of RBC EVs to deliver an ASO that antagonizes the oncogenic miR-125b in AML cells was further tested. miR-125b is upregulated in different types of cancer including AML and other leukemia. It has been shown that miR-125b suppresses apoptosis by regulating multiple genes in the p53 network. miR-125b also promotes proliferation of hematopoietic stem cells and leukemia cells in both humans and mouse models. An anti-miR-125b ASO (Shanghai Gene Pharma) comprising a sequence of SEQ ID NO: 1 was loaded into RBC EVs using electroporation and treated MOLM13 cells with these EVs. After 72 hours, it was found that the level of miR-125b was suppressed by 80-95% in a dose-dependent manner. miR-125a, the homologue of miR-125b, was also suppressed by 50-80% due to the sequence similarity to miR-125b. Inhibition of miR-125 led to a significant increase in BAK1, a target of miR-125a/b which regulates apoptosis. Treatment with miR-125b ASO loaded EVs also dampened the growth of MOLM13 cells significantly after 3-4 days of incubation. Hence, the inhibition of miR-125b using ASO in RBC EVs may represent a new approach for AML treatment.
  • As described here, miR-125b preferably comprises or consists of SEQ ID NO: 4 and miR-125a preferably comprises or consists of SEQ ID NO: 5. In particular, miR-125b consists of SEQ ID NO: 4 and miR-125a consists of SEQ ID NO: 5.
  • FIG. 8a : Schematic presentation of miR-125b ASO delivery: 25-100 μg RBC EVs were electroporated with 2 μmole anti-miR-125b ASO and incubated with MOLM13 cells. Anti-miR-125b ASO in this embodiment consists of SEQ ID NO: 1. FIGS. 8b-c : Expression of miR-125b and miR-125a relative to U6b snRNA in MOLM13 cells that were untreated, incubated with 100 ug unelectroporated RBC EVs (UE-EVs), with negative control (NC)-ASO electroporated RBC EVs (E-EVs) or anti-miR-125b ASO (125b-ASO) electroporated RBC EVs for 72 hours, as determined by Taqman qRT-PCR, presented as average and SEM. FIG. 8d : Expression of BAK1 relative to GAPDH in MOLM13 cells treated the same as in FIG. 8b . FIG. 8e : Number of MOLM13 cells untreated, or treated with UE-EVs or with ASO electroporated EVs as indicated. One-way Anova test: ** P<0.01; *** P<0.001.
  • Similarly, RBC EVs were tested for the delivery of miR-125b ASO to breast cancer MCF10aCA1a (CA1a) cells. The inventors observed 80-90% knockdown of miR-125a and miR-125b in CA1a cells treated with miR-125b ASO loaded EVs. As a consequence, the knockdown of miR-125s suppressed the proliferation of CA1a cells.
  • FIG. 9a : Schematic presentation of miR-125b ASO delivery: 25-50 μg RBC EVs were electroporated with 2 μmole anti-miR-125b ASO and incubated with CA1a cells. FIG. 9b-c : Expression of miR-125b and miR-125a relative to U6b snRNA in CA1a cells that were untreated, incubated with unelectroporated RBC EVs (UE-EVs), with negative control (NC)-ASO electroporated RBC EVs (E-EVs) or anti-miR-125b ASO (125b-ASO) electroporated RBC EVs for 72 hours, as determined by Taqman qRT-PCR, presented as average and SEM. FIG. 9d : Crystal violet staining of CA1a cells untreated, or treated with UE-EVs or with ASO electroporated EVs as indicated. Bar graph represent the average number of cells counted in crystal-violet stained wells (n=3). One-way Anova test: ** P<0.01.
  • To test the feasibility of CRISPR delivery using RBC EVs, synthetic SpCas9 mRNA (Trilink) was electroporated into RBC EVs using the protocol that was optimized for Dextran and ASO. As the result, a large amount of Cas9 mRNAs was detected in MOLM13 cells after a 24-hour incubation with the electroporated EVs, using qRT-PCR. Furthermore, using immunostaining of the HA-tag, Cas9 protein was found in the nuclei (overlapped with a nuclear stain) of ˜50% MOLM13 cells at 48-hour post-treatment. This suggests that RBC EVs can be used to deliver the CRISPR Cas9 system.
  • FIG. 10a : Schematic presentation of Cas9 mRNA delivery: RBC EVs were electroporated with Cas9 mRNA and incubated with MOLM13 cells for 24 or 48 hours. FIG. 10b : The levels of Cas9 mRNA relative to GAPDH mRNA in MOLM13 cells that were untreated, incubated with unelectroporated EVs or with EVs that were electroporated with 5, 10 or 20 μg Cas9 mRNA, determined by qRT-PCR after 24 hours of treatment. Values are presented as mean±SEM (n=3). FIG. 10c : Representative images of MOLM13 cells that were untreated, or incubated for 48 hours with unelectroporated EVs or with EVs that were electroporated with 10 μg Cas9 mRNAs. The cells were stained for HA-Cas9 protein (using green dye, not seen in black and white images) and nuclear DNA (Hoechst, blue dye, not seen in black and white images). FIG. 10d : Average percentage of MOLM13 cells stained positive for HA-Cas9 protein as shown in (c).
  • Subsequently, the inventors delivered Cas9 mRNA together with an anti-eGFP gRNA in RBC EVs to AML cells, NOMO1, that are labeled with eGFP. After one week, the inventors observed a complete knockout of eGFP in 32.9% NOMO1 cells. Hence, the RNAs delivered by RBC EVs were able to execute a CRISPR knockout of eGFP. As described herein, Cas9 mRNA preferably comprises or consists of SEQ ID NO: 2 and eGFP gRNA preferably comprises or consists of SEQ ID NO: 3. In particular, Cas9 mRNA consists of SEQ ID NO: 2 and eGFP gRNA consists of SEQ ID NO: 3.
  • FIG. 11a : Schematic presentation of the RNA delivery: RBC EVs were electroporated with Cas9 mRNA and anti-GFP gRNA and incubated with NOMO1-GFP cells for 7 days. FIG. 11b : FACS analysis of GFP in NOMO1-GFP cells that were untreated, incubated with unelectroporated EVs or EVs electroporated with Cas9 and gRNA. The percentages of GFP-negative cells are shown above the gate.
  • In addition, the delivery of plasmids by RBC EVs was also tested. RBC EVs were electroporated with two plasmids, one expressing SpCas9 and one expressing gRNA against eGFP. Electroporated EVs were incubated with human embryonic kidney HEK-293T cells that homogenously express eGFP. After 96 hours, it was found that 13.8% GFP-negative cells resulted from the EV treatment, compared to 3.52% GFP-negative in the untreated population. Treatment with electroporated EVs showed a distinct peak of GFP-negative cells that suggests a homologous knockout of eGFP by the delivery of Cas9 and gRNA plasmids. Therefore, RBC EVs are able to deliver not only RNA but also plasmid DNA for genome editing. Moreover, the delivery is applicable to HEK-293T solid cancer cells.
  • FIG. 12a Schematic presentation of plasmid delivery: RBC EVs were electroporated with Cas9 plasmid and eGFP gRNA plasmid and incubated with eGFP expressing 293T cells for 96 hours. FIGS. 12b-12c FACS analysis of GFP in 293T-eGFP cells untreated, or incubated with unelectroporated EVs (UE-EVs) or with plasmid electroporated EVs (E-EVs) as indicated. The GFP negative cells are indicated by the percentages in FIG. 12b and the arrow in FIG. 12 c.
  • FIG. 13a : Schematic presentation of an in vivo EV uptake assay. FIG. 13b : Fluorescent images of nude mice bearing untreated tumors on the top and tumors injected with PKH26-labeled (red dye, not seen in black and white images) EVs on the bottom. FIG. 13c : Ex vivo fluorescent images of the tumors at 72 hours post-treatment. FIG. 13d : Total radiance efficiency (photons/second) of fluorescent signals in the tumors 24-72 hours after the injection of PKH26-labeled EVs. To determine whether the RBC EVs are taken up by tumor cells in vivo, CA1a cells were implanted in the mice, in the flank at 2 sides (FIG. 13a ). The tumor size is about 7 mm. 100 μg of PKH26 labelled EVs were then injected intratumorally. Fluorescent live imaging was done every day for 3 days (72 hours). Images of nude mice bearing untreated tumors and tumors injected with PKH26-labeled (red dye note seen in black and white images) EVs were taken (FIG. 13b ). With reference to FIG. 13c , it shows ex vivo fluorescent images of the tumors at 72 hours post-treatment and proves that PKH26-labeled EVs were taken up by tumor cells. The total radiance efficiency (photons/second) of fluorescent signals in the tumors decreased gradually 24 to 72 hours after the injection of PKH26-labeled EVs as shown in FIG. 13 d.
  • It will be appreciated by persons skilled in the art that numerous variations and/or modifications may be made to the present invention as shown in the specific embodiments without departing from the spirit or scope of the invention as broadly described. The present embodiments are, therefore, to be considered in all respects as illustrative and not restrictive.
  • It will also be appreciated by persons skilled in the art that the present invention may also include further additional modifications made to the method which does not affect the overall functioning of the method.
  • Any reference to prior art contained herein is not to be taken as an admission that the information is common general knowledge, unless otherwise indicated. It is to be understood that, if any prior art information is referred to herein, such reference does not constitute an admission that the information forms a part of the common general knowledge in the art, any other country.

Claims (24)

1. A method for RNA delivery to target cells comprising:
a) purification of extracellular vesicles (EVs) from red blood cells (RBCs);
b) electroporation of the EVs with RNAs to form RNA-loaded EVs; and
c) applying the RNA-loaded EVs to the target cells.
2. The method of claim 1, wherein the RBCs are derived from a human and treated with calcium ionophore.
3. The method of claim 2, wherein the EVs are purified from treated RBCs using ultracentrifugation with a sucrose cushion.
4. The method of claim 1, wherein the RNAs comprise antisense oligonucleotides (ASO) and mRNAs.
5. The method of claim 1, wherein the target cells comprise cancer cells.
6. The method of claim 1, wherein the target cells comprise acute myeloid leukemia (AML) cells, breast cancer cells, or a combination of AML cells and breast cancer cells.
7. The method of claim 1, wherein the EVs are electroporated with ASO antagonizing miR-125b.
8. The method of claim 1, wherein the growth of the target cells is suppressed.
9. The method of claim 1, wherein the EVs are electroporated with dextran.
10. The method of claim 1, comprising administering to the target cells the RNA-loaded EVs which modulate an apoptosis-related gene expression, thereby inducing apoptosis in the target cells.
11. A method for delivery of an antisense oligonucleotide (ASO) to target cells to suppress gene expression, wherein the method comprises:
a) purification of extracellular vesicles (EVs) from red blood cells (RBCs);
b) electroporation of the EVs with RNAs to form RNA-loaded EVs; and
c) applying the RNA-loaded EVs to the target cells.
12. The method of claim 11, wherein the RBCs are derived from a human and treated with calcium ionophore.
13. The method of claim 11, wherein the RNA is an ASO antagonizing miR-125b to inhibit the oncogenic miR-125b in the target cells.
14. The method of claim 11, wherein the target cells are acute myeloid leukemia (AML) cells, breast cancer cells, or a combination of AML cells and breast cancer cells.
15. A method of RNA delivery to target cells for a CRISPR genome editing system comprising:
a) purification of extracellular vesicles (EVs) from red blood cells (RBCs);
b) electroporation of the EVs with RNAs to form RNA-loaded EVs; and
c) applying the RNA-loaded EVs to the target cells.
16. The method of claim 15, wherein the EVs are electroporated with Cas9 mRNA and gRNA.
17. The method of claim 15, wherein the EVs are electroporated with Cas9 and gRNA plasmids.
18. The method of claim 15, wherein the target cells are cancer cells.
19. The method of claim 15, wherein the target cells are leukemia cells.
20. A method of treating cancer by delivery of RNA to target cells comprising:
a) purification of extracellular vesicles (EVs) from red blood cells (RBCs);
b) electroporation of the EVs with RNAs to form RNA-loaded EVs; and
c) applying the RNA-loaded EVs to the target cells thereby inhibiting the growth of the target cells, wherein the target cells comprise cancer cells.
21. The method of claim 20, wherein the target cells comprises leukemia cells, breast cancer cells, or a combination of leukemia cells and breast cancer cells.
22. The method of claim 20, wherein the target cells comprise acute myeloid leukemia cells.
23. The method of claim 20, wherein the step c) comprises a step of administering the RNA-loaded EVs to a subject having the target cells via a local or systemic administration.
24. The method of claim 20, wherein the growth of the target cells is suppressed after the step c).
US16/825,097 2017-08-16 2020-03-20 Isolation of extracellular vesicles (evs) from red blood cells for gene therapy Abandoned US20200230259A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US16/825,097 US20200230259A1 (en) 2017-08-16 2020-03-20 Isolation of extracellular vesicles (evs) from red blood cells for gene therapy

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US15/678,363 US10709797B2 (en) 2017-08-16 2017-08-16 Isolation of extracellular vesicles (EVs) from red blood cells for gene therapy
US16/825,097 US20200230259A1 (en) 2017-08-16 2020-03-20 Isolation of extracellular vesicles (evs) from red blood cells for gene therapy

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US15/678,363 Continuation US10709797B2 (en) 2017-08-16 2017-08-16 Isolation of extracellular vesicles (EVs) from red blood cells for gene therapy

Publications (1)

Publication Number Publication Date
US20200230259A1 true US20200230259A1 (en) 2020-07-23

Family

ID=65360994

Family Applications (2)

Application Number Title Priority Date Filing Date
US15/678,363 Active 2037-10-28 US10709797B2 (en) 2017-08-16 2017-08-16 Isolation of extracellular vesicles (EVs) from red blood cells for gene therapy
US16/825,097 Abandoned US20200230259A1 (en) 2017-08-16 2020-03-20 Isolation of extracellular vesicles (evs) from red blood cells for gene therapy

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US15/678,363 Active 2037-10-28 US10709797B2 (en) 2017-08-16 2017-08-16 Isolation of extracellular vesicles (EVs) from red blood cells for gene therapy

Country Status (2)

Country Link
US (2) US10709797B2 (en)
CN (1) CN109402176A (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023077149A1 (en) * 2021-11-01 2023-05-04 Sigma-Aldrich Co. Llc Electroporation enhancers for crispr-cas systems
US11970718B2 (en) 2020-01-13 2024-04-30 Carmine Therapeutics Pte. Ltd. Nucleic acid loaded extracellular vesicles

Families Citing this family (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2021030769A1 (en) * 2019-08-14 2021-02-18 Codiak Biosciences, Inc. Extracellular vesicles with nras antisense oligonucleotides
US20230121065A1 (en) * 2020-03-26 2023-04-20 National University Of Singapore Method of delivering nucleic acid to immune cells using rbcev
US20230226218A1 (en) 2020-05-11 2023-07-20 Erytech Pharma Red Cell Extracellular Vesicles (RCEVs) Containing Cargoes and Methods of Use and Production Thereof
CN117940117A (en) * 2021-06-30 2024-04-26 株式会社普里莫里斯治疗 Pharmaceutical composition containing cell-derived natural or artificial nanovesicles loaded with antisense oligonucleotide-based drugs
WO2023169594A1 (en) * 2022-03-08 2023-09-14 中山大学 Application of blood-derived sample in preparation of vesicles
WO2023185697A2 (en) 2022-03-29 2023-10-05 Accuredit Therapeutics (Suzhou) Co., Ltd. Compositions and methods for treatment of transthyretin amyloidosis
CN114591905B (en) * 2022-04-01 2022-09-02 北京大学口腔医学院 Method for preparing apoptotic vesicles from human erythrocytes and application of apoptotic vesicles
WO2024061296A2 (en) 2022-09-22 2024-03-28 Accuredit Therapeutics (Suzhou) Co., Ltd. Compositions and methods for treatment of hypercholesterolemia and/or cardiovascular disease
CN115887679B (en) * 2022-11-21 2024-03-22 国家纳米科学中心 Gene-chemotherapy nano drug co-delivery system, preparation method and application thereof

Family Cites Families (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9085778B2 (en) 2006-05-03 2015-07-21 VL27, Inc. Exosome transfer of nucleic acids to cells
US8329161B2 (en) 2008-05-01 2012-12-11 National Health Research Institutes Red blood cell-derived vesicles as a nanoparticle drug delivery system
EP3079726B1 (en) * 2013-12-12 2018-12-05 The Broad Institute, Inc. Delivery, use and therapeutic applications of the crispr-cas systems and compositions for targeting disorders and diseases using particle delivery components
US20160331686A1 (en) * 2015-05-12 2016-11-17 Clsn Laboratories, Inc. Compositions and Methods for Yeast Extracellular Vesicles as Delivery Systems
WO2016187717A1 (en) 2015-05-26 2016-12-01 Exerkine Corporation Exosomes useful for genome editing
WO2017054085A1 (en) 2015-10-01 2017-04-06 University Of Ottawa Exosome packaging of nucleic acids
RU2608509C1 (en) 2016-02-11 2017-01-18 Елена Сергеевна Кастарнова Method of obtaining blood exosomes
JP2019528674A (en) 2016-09-30 2019-10-17 セレックス ライフ サイエンシズ,インコーポレーテッド Compositions containing exosomes loaded with proteins and methods for their preparation and delivery

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
AMA Style Insider (Sept 2012) downloaded from https://amastyleinsider.com/2012/09/14/around-about-approximately/ on March 24, 2023 *
e.insights (June 2014) downloaded from https://www.editage.com/insights/scientific-writing-difference-in-meaning-of-about-around-and-approximately on March 24, 2023 *
Zhang et al. (Erythrocyte, 2019 doi: 10.5772/intechopen.81758, pages 1-16) *

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11970718B2 (en) 2020-01-13 2024-04-30 Carmine Therapeutics Pte. Ltd. Nucleic acid loaded extracellular vesicles
WO2023077149A1 (en) * 2021-11-01 2023-05-04 Sigma-Aldrich Co. Llc Electroporation enhancers for crispr-cas systems

Also Published As

Publication number Publication date
US20190054192A1 (en) 2019-02-21
CN109402176A (en) 2019-03-01
US10709797B2 (en) 2020-07-14

Similar Documents

Publication Publication Date Title
US20200230259A1 (en) Isolation of extracellular vesicles (evs) from red blood cells for gene therapy
Usman et al. Efficient RNA drug delivery using red blood cell extracellular vesicles
US10959952B2 (en) Use of exosomes for the treatment of disease
RU2747722C2 (en) RNA-guided destruction of human JC virus and other poliomaviruses
JP2019512458A (en) Eradication of human JC virus and other polyoma viruses induced by RNA
WO2020060479A1 (en) Cargo loaded extracellular vesicles
US11970718B2 (en) Nucleic acid loaded extracellular vesicles
WO2018237369A2 (en) Lipid nanoparticle (lnp)-mediated delivery of a crispr-expressing plasmid dna for treating chronic hepatitis b virus infection
JP2024059816A (en) Methods and compositions for treating cancer using exosome-associated gene editing
WO2019191444A1 (en) Use of exosomes for targeted delivery of therapeutic agents
US20210369858A1 (en) Use of exosomes for targeted delivery of therapeutic agents
JP2021522164A (en) Therapeutic regulation of tumor suppressors using exosomes
JP2021521126A (en) Therapeutic targeting method for oncogenes using exosomes
US20240123034A1 (en) Mrnas encoding granulocyte-macrophage colony stimulating factor for treating parkinson&#39;s disease
JP6857730B2 (en) H-1 PV expressing RNAi effectors targeting CDK9
CN115887679A (en) Gene-chemotherapy nano-drug co-delivery system, preparation method and application thereof
IL302316A (en) Targeting of src-3 in immune cells as an immunomodulatory therapeutic for the treatment of cancer
CN117159495A (en) Lipid nanoparticle and application thereof

Legal Events

Date Code Title Description
STPP Information on status: patent application and granting procedure in general

Free format text: APPLICATION DISPATCHED FROM PREEXAM, NOT YET DOCKETED

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: FINAL REJECTION MAILED

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION