WO2021083278A1 - Engineering red blood cells for treating gout and hyperuricemia diseases - Google Patents

Engineering red blood cells for treating gout and hyperuricemia diseases Download PDF

Info

Publication number
WO2021083278A1
WO2021083278A1 PCT/CN2020/124849 CN2020124849W WO2021083278A1 WO 2021083278 A1 WO2021083278 A1 WO 2021083278A1 CN 2020124849 W CN2020124849 W CN 2020124849W WO 2021083278 A1 WO2021083278 A1 WO 2021083278A1
Authority
WO
WIPO (PCT)
Prior art keywords
cells
human
lin
red blood
urat1
Prior art date
Application number
PCT/CN2020/124849
Other languages
French (fr)
Inventor
Xiaofei GAO
Xiaoqian NIE
Yanjie HUANG
Hui Li
Original Assignee
Westlake Therapeutics (Hangzhou) Co. Limited
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 Westlake Therapeutics (Hangzhou) Co. Limited filed Critical Westlake Therapeutics (Hangzhou) Co. Limited
Priority to CN202080076275.7A priority Critical patent/CN114746546A/en
Publication of WO2021083278A1 publication Critical patent/WO2021083278A1/en

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • 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
    • A61K35/18Erythrocytes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/43Enzymes; Proenzymes; Derivatives thereof
    • A61K38/44Oxidoreductases (1)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • 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
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y107/00Oxidoreductases acting on other nitrogenous compounds as donors (1.7)
    • C12Y107/03Oxidoreductases acting on other nitrogenous compounds as donors (1.7) with oxygen as acceptor (1.7.3)
    • C12Y107/03003Factor-independent urate hydroxylase (1.7.3.3), i.e. uricase
    • 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
    • C12N2500/00Specific components of cell culture medium
    • C12N2500/05Inorganic components
    • C12N2500/10Metals; Metal chelators
    • C12N2500/20Transition metals
    • C12N2500/24Iron; Fe chelators; Transferrin
    • C12N2500/25Insulin-transferrin; Insulin-transferrin-selenium
    • 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
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/10Growth factors
    • C12N2501/125Stem cell factor [SCF], c-kit ligand [KL]
    • 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
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/20Cytokines; Chemokines
    • C12N2501/23Interleukins [IL]
    • C12N2501/2303Interleukin-3 (IL-3)
    • 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
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/20Cytokines; Chemokines
    • C12N2501/23Interleukins [IL]
    • C12N2501/2306Interleukin-6 (IL-6)
    • 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
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/20Cytokines; Chemokines
    • C12N2501/26Flt-3 ligand (CD135L, flk-2 ligand)
    • 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
    • C12N2506/00Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells
    • C12N2506/11Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells from blood or immune system 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
    • C12N2510/00Genetically modified 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
    • C12N2740/00Reverse transcribing RNA viruses
    • C12N2740/00011Details
    • C12N2740/10011Retroviridae
    • C12N2740/16011Human Immunodeficiency Virus, HIV
    • C12N2740/16041Use of virus, viral particle or viral elements as a vector
    • C12N2740/16043Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector

Definitions

  • the present disclosure relates to engineering red blood cells, and in particular, to engineering red blood cells carrying URAT1 and/or UOX.
  • the present disclosure also relates to methods for treating hyperuricemia associated diseases.
  • Gout is the most common form of inflammatory arthritis in adults, especially in men, with a prevalence ranging from 1%to 4%globally [1, 2] . Gout occurs when monosodium urate crystal (MSU) deposited in tissues, causing inflammation and intense pain of a gout attack.
  • MSU monosodium urate crystal
  • the biologic precursor to gout is elevated serum uric acid (UA) levels (i.e., hyperuricemia) .
  • UA serum uric acid
  • hyperuricemia is the strongest single risk factor for the development of gout and is universally present in patients with gout, not all individuals with hyperuricemia develop gout. Recent work has emphasized the importance of the innate immune response [2] .
  • urate-lowering agents such as anti-inflammatory drugs (colchicine) , xanthine oxidase inhibitors (allopurinol, febuxostat) or uricosuric agents (probenecid, benzbromarone) induce very slow reduction in UA deposits, not allowing for the rapid resolution of tophi for all patients with gout and are mainly used at the early stage [3] .
  • Urate oxidase (UOX, uricase) is a liver enzyme that metabolizes UA into allantoin, a more water-soluble compound, which is easily excreted by the kidney [2, 3] . All mammals produce UOX, except humans and certain primates. Indeed, during evolution, UOX was inactivated in humans primarily due to missense and frame-shift mutations in the gene encoding this enzyme [2, 4] . Uricase undeniably represents a valuable treatment option for chronic tophaceous gout when conventional urate-lowering agents may not be used.
  • Rasburicase a recombinant UOX from A. flavus, was approved by EMEA in 2001 and the Food and Drug Administration (FDA) in 2002 for tumor lysis syndrome [5, 6] .
  • This agent significantly reduces serum UA levels and acts faster than allopurinol.
  • the recommended dose is 0.2 mg/kg in children and adults [7, 8] .
  • its biological half-life is short---only 21h, so rasburicase is given by infusion once daily for ⁇ 7 days [7] .
  • recent studies have shown that repeated UOX injections could cause anaphylactic reactions with the production of antibodies that neutralize UOX enzyme activity [9] .
  • Pegloticase a recombinant porcine UOX, with a baboon C-terminal sequence, is a modified pegylated recombinant UOX developed to be the first non-immunogenic biologic for treating the hyperuricemia of refractory gout, approved by the FDA for patients with chronic gout [3, 10] .
  • a 6-month study versus placebo showed that pegloticase (infused at 8 mg every 2 weeks) , induced a significant decrease in plasma UA in about 40%of the patients (associated with a tendency for tophi dissolution) [3] . However, the remaining patients had no response, which was correlated with the formation of pegloticase antibodies and infusion reactions [3] . More than 10%of the patients treated with pegloticase had adverse events such as kidney stones, joint pain, anaemia, muscle spasms, dyspnea, headache, nausea, and fever [7] .
  • UOX rasburicase, pegloticase
  • UOX rasburicase, pegloticase
  • PEG PEG-conjugated enzymes
  • PEG may adversely affect the activity of the conjugated enzyme, leading to reduced efficacy in the treatment [12, 13] .
  • the therapeutic enzymes may become inactivated or eliminated in vivo due to short half-life, limited bioavailability, and/or interactions with plasma proteins [14] .
  • red blood cells are the most abundant cells in the blood, occupying a quarter of all human cells, and are widely distributed to the human body through circulation; (2) red blood cells have a long life span (about 120 days for humans and 50 days for mice) . Furthermore, old or damaged red blood cells can be cleared by macrophages in the liver and spleen without causing immune responses; (3) red blood cells are biocompatible, especially when using autologous red blood cells; (4) mature red blood cells have no nuclei, mitochondria, or other organelles.
  • red blood cells protect the drug substance from premature inactivation and/or degradation, as well as the organism from the toxic effects of the drug [15-17] .
  • a major regulator of serum urate is renal excretion of UA.
  • net reabsorption of UA into the blood predominates owing to less excretion of UA than is filtered at the glomerulus.
  • URAT1 a urate–anion exchanger, can transport UA from the urinary lumen, which is an important step for UA reabsorption [2, 18] .
  • an engineered red blood cell comprising:
  • a urate oxidase (UOX) or a functional variant thereof;
  • URAT1 urate transporter 1
  • the URAT1 comprises an amino acid sequence of SEQ ID NO: 2.
  • the UOX comprises an amino acid sequence of SEQ ID NO: 4.
  • the present disclosure provides a method for producing engineered red blood cells, which comprises:
  • Lin-cells lineage negative cells
  • the one or more exogenous nucleic acids comprises a first polynucleotide sequence encoding a urate oxidase (UOX) or a functional variant thereof, a second polynucleotide sequence encoding a urate transporter 1 (URAT1) or a functional variant thereof; or both of the first polynucleotide sequence and the second polynucleotide sequence.
  • UOX urate oxidase
  • URAT1 urate transporter 1
  • the URAT1 comprises an amino acid sequence of SEQ ID NO: 2.
  • the UOX comprises an amino acid sequence of SEQ ID NO: 4.
  • the blood sample is a peripheral blood sample, a cord blood sample or a fetal blood sample.
  • the blood sample is a human peripheral blood sample.
  • the Lin-cells are Lin-CD34-cells.
  • the step 1) includes isolating peripheral blood mononuclear cells (PBMCs) from the peripheral blood sample and isolating Lin-CD34-cells from the PBMCs.
  • PBMCs peripheral blood mononuclear cells
  • the step 1) includes removing lineage positive (Lin+) cells from the peripheral blood sample by using a lineage cell depletion kit.
  • the step 2) including culturing the Lin-cells in a hematopoietic stem cell expansion medium supplemented with a combination of cytokines, and the combination of cytokines comprise fms-like tyrosine kinase 3 ligand (Flt3L) , stem cell factor (SCF) , interleukin 3 (IL-3) , and interleukin 6 (IL-6) .
  • cytokines comprise fms-like tyrosine kinase 3 ligand (Flt3L) , stem cell factor (SCF) , interleukin 3 (IL-3) , and interleukin 6 (IL-6) .
  • the combination of cytokines comprise 50 ng/mL human Flt3L, 50 ng/mL human SCF, 10 ng/mL human IL-3, and 10 ng/mL human IL-6.
  • the hematopoietic stem cell expansion medium is StemSpan TM SFEM serum-free expansion medium.
  • the step 2) including culturing the Lin-cells at 37°C under 5%CO 2 for about 5 days.
  • the step 3) comprising culturing the expanded Lin-cells in a first differentiation medium supplemented with cytokines related to erythroid development.
  • the first differentiation medium is an Iscove's Modified Dulbecco's Medium (IMDM) containing: fetal bovine serum (FBS) or human serum, human plasma, glutamine, BSA, transferrin, insulin, Penicillin-Streptomycin, IL-3, EPO, and SCF.
  • IMDM Iscove's Modified Dulbecco's Medium
  • the first differentiation medium is an Iscove's Modified Dulbecco's Medium (IMDM) containing: 10-15%FBS or human serum, 5-10%human plasma, 1-4 mM glutamine, 1-2%BSA, 300-600 ⁇ g/mL human transferrin, 8-13 ⁇ g/mL human insulin, 2%Penicillin-Streptomycin, 3-5 ng/mL human IL-3, 4-7 U/mL human EPO, and 100 ng/mL human SCF.
  • IMDM Iscove's Modified Dulbecco's Medium
  • step 3 comprising culturing the expanded Lin-CD34-cells at 37°C under 5%CO 2 for about 9 days.
  • the step 4) comprising culturing the erythroid cells in a second differentiation medium, wherein the second differentiation medium is an Iscove's Modified Dulbecco's Medium (IMDM) containing: FBS or human serum, human plasma, glutamine, BSA, transferrin, insulin, Penicillin-Streptomycin, and EPO.
  • IMDM Iscove's Modified Dulbecco's Medium
  • the second differentiation medium is an Iscove's Modified Dulbecco's Medium (IMDM) containing: 15%FBS or human serum, 5-10%human plasma, 1-4 mM glutamine, 1-2%BSA, 300-600 ⁇ g/mL human transferrin, 8-13 ⁇ g/mL human insulin, 2%Penicillin-Streptomycin, and 1-5 U/mL human EPO.
  • IMDM Iscove's Modified Dulbecco's Medium
  • the step 4) comprising culturing erythroid cells at 37°C under 5%CO 2 for about 7 days.
  • the introduction of the one or more exogenous nucleic acids into the expanded Lin-cells is carried out on the 1st day of culturing the expanded Lin-cells in the first differentiation medium.
  • the exogenous nucleic acid is an expression vector.
  • the exogenous nucleic acid is a lentiviral expression vector.
  • the present disclosure provides a pharmaceutical composition comprising the engineered red blood cells and a physiologically acceptable excipient.
  • the present disclosure provides uses of the engineered red blood cells in the preparation of a medicament for treating a disorder associated with elevated serum uric acid levels.
  • the disorder is gout or a hyperuricemia disease.
  • the present disclosure provides a method for the treatment of a disorder associated with elevated serum uric acid levels in a subject, which comprises:
  • the disorder is gout or a hyperuricemia disease
  • the present disclosure provides a method for the treatment of a disorder associated with elevated serum uric acid levels in a subject, which comprises infusing a therapeutically effective amount of the engineered red blood cell into the subject.
  • the method further comprises performing blood typing before the infusion to ensure the subject is compatible with the engineered red blood cell.
  • Fig. 1 shows the RBC engineering strategy for gout and hyperuricemia therapy.
  • Fig. 2 depicts experimental scheme of engineering hematopoietic stem and/or progenitor cells to express URAT1 and UOX, and subsequently differentiating the engineered hematopoietic stem and/or progenitor cells in vitro into mature red blood cells carrying both functional proteins.
  • Fig. 3 depicts representative analysis that can be used to test the functions of engineered RBCs (eRBCs) expressing URAT1 and UOX in vitro, as well as their functions in vivo using established animal models for gout.
  • eRBCs engineered RBCs
  • Fig. 4 shows a workflow of eRBC generation from mouse embryos.
  • Mouse erythroid progenitors were isolated from fetal livers at embryonic 14.5 days. Cells were then infected with MSCV-URAT1/UOX-copGFP before induced into terminally differentiated mouse red cells expressing URAT1 or UTRA1 and UOX.
  • Fig. 5 shows that eRBCs expressing URAT1 and/or UOX have undergone normal erythroid differentiation.
  • FACS analysis of eRBCs generated from Lin-PBMCs of a gout patient at day 8 of the in vitro differentiation.
  • the following are the markers used in the FACS analyses: CD235a, CD117, CD71, Hoechst33342, and GFP (the reporter gene indicating expression of URAT1 and/or UOX) .
  • CD235a, CD117 and CD71 are used to track the progression of erythroid development.
  • Hoechst33342 stains DNA, and therefore, Hoechst 33342 negative cells are enucleated cells.
  • the channels used to detect corresponding signals Y585-PE: PI; B525-FITC: GFP; R660-APC: CD235a; B650-PC5.5: CD71; UV450: Hoechst33342.
  • A eRBC-URAT1 at day 8 of the in vitro erythroid differentiation
  • B eRBC-URAT1+UOX at day 8 of the in vitro erythroid differentiation.
  • eRBC-URAT1 and eRBC-URAT1+UOX generated from Lin-PBMCs of the gout patients reduced the UA concentration in vitro efficiently.
  • A UA concentration of the medium in which eRBCs were cultured. 10 5 eRBCs, eRBC-URAT1 or eRBC-URAT1+UOX were cultured in the medium with 465 ⁇ M UA (mimicking UA concentration of a gout patient) for 24 hours. After the incubation, the UA concentration in the medium was determined by a coupled enzyme reaction (Abcam) .
  • B The consumption rates of UA by eRBC-URAT1 and eRBC-UOX+URAT1 were calculated based on the results from Fig. 6A.
  • FIG. 7 shows tissue distribution of engineered RBCs in NSG mouse by live imaging. DIR-labeled RBCs were injected into NSG mouse via intravenous injection and the subsequent distributions of the labeled red blood cells were examined by in vivo imaging at 1 h, 5 h, 48 h, and 5 days after the injection of red blood cells.
  • Mouse #1 no treatment ; Mouse #2: 2 ⁇ M DIR only in PBS; Mouse #3: 1x10 8 human red blood cells differentiated from PBMCs in vitro and labeled with 2 ⁇ M DIR; Mouse #4: 1x10 8 human red blood cells infected with vector only differentiated from PBMCs in vitro and labeled with 2 ⁇ M DIR; Mouse #5: 1x10 8 human red blood cells transducing with lentiviral vectors encoding phenylalanineammonialyase (PAL) and differentiated from PBMCs in vitro and labeled with 2 ⁇ M DIR; Mouse #6: 5x10 7 human red blood cells transducing with lentiviral vectors encoding glyoxylate and hydroxypyruvate reductase (GRHPR) and differentiated from PBMCs in vitro and labeled with 2 ⁇ M DIR.
  • PAL phenylalanineammonialyase
  • GSHPR hydroxypyruvate
  • FIG. 8 shows an example of images of tissue samples after the injection of red blood cells.
  • DIR-stained red blood cells were intravenously injected into each mouse. Seven days after the injection, the mice were sacrificed, and the tissue samples were imaged.
  • Mouse #5 1x10 8 human red blood cells transducing with lentiviral vectors encoding phenylalanineammonialyase (PAL) and differentiated from PBMCs in vitro and labeled with 2 ⁇ M DIR;
  • Fig. 9 shows that engineered mouse eRBC-URAT1+UOX reduced UA concentration in a hyperuricemia and gout mouse model.
  • (A) Hyperuricemia was induced by intraperitoneal injection of MSU crystal suspension at the dose of 250 mg/kg, and the serum UA concentration was analyzed by a coupled enzyme reaction;
  • B Hyperuricemia was induced 3 days after 2x10 8 eRBC-URAT1 or eRBC-URAT1+UOX cells or PBS (Blank) were intravenously injected into the mice. UA concentration in the serum was measured to assess the therapeutic effects of the eRBCs.
  • an element means one element or more than one element.
  • Lineage negative cells or “Lin-cells” as used herein refers to cells that are essentially free of lineage markers.
  • Lineage markers are characteristic of cell lineages. Exemplary lineage markers are CD1c, CD3, CD11c, CD14, CD15, CD16, CD20, CD41, CD56, CD203c, CD235a, and/or BDCA2.
  • lineage negative cells are essentially not stained by the lineage antibodies.
  • Lineage negative cells comprise stem and progenitor cells. Accordingly, lineage negative cells also show stem and progenitor cell activity. Lin-cells or a blood cell population enriched for lineage negative cells can be purified by enriching a blood cell population that is essentially free of lineage markers.
  • the lineage negative cells can be purified by depleting cells that are positive for at least one lineage marker selected from the group consisting of CD1c, CD3, CD11c, CD14, CD15, CD16, CD20, CD41, CD56, CD203c, CD235a, and/or BDCA2.
  • a lineage cell depletion kit can be used to perform the purification.
  • lineage positive (Lin+) cells are a mix of all cells expressing mature cell lineage markers. Examples of lineage positive cells include T cells, B cells, NK cells, dendritic cells, monocytes, granulocytes, erythroid cells, and their committed precursors.
  • the term “functional variant” means any variant comprising substitution, deletion or addition of one or a few to plural amino acids to UOX or URAT1, provided that the variant substantially retains the same function as UOX or URAT1 possesses.
  • culture refers to maintaining cells in a medium with or without cell expansion or differentiation for any period of time.
  • differentiation refers to a process by which a less specialized cell such as a stem cell develops or matures to possess a more distinct form and function with a concomitant loss of potential.
  • Cells that are less specialized can be differentiated into cells that are more specialized by culturing the cells under particular conditions or in specific media as known in the art.
  • pharmaceutically acceptable excipient refers to a pharmaceutically acceptable material, composition or vehicle, such as a liquid or solid filler, diluent, carrier, manufacturing aid (e.g., lubricant, talc magnesium) , solvent or encapsulating material, involved in carrying or transporting a therapeutic compound for administration to a subject.
  • a pharmaceutically acceptable material such as a liquid or solid filler, diluent, carrier, manufacturing aid (e.g., lubricant, talc magnesium) , solvent or encapsulating material, involved in carrying or transporting a therapeutic compound for administration to a subject.
  • manufacturing aid e.g., lubricant, talc magnesium
  • solvent or encapsulating material involved in carrying or transporting a therapeutic compound for administration to a subject.
  • FLT3L fms-like tyrosine kinase 3 ligand
  • FMS-like tyrosine kinase 3 ligand Flt-3 ligand
  • FLT3LG is a protein which promotes HSCs differentiation to various hematopoietic lineages.
  • FLT3LG is structurally homologous to stem cell factor (SCF) and colony stimulating factor 1 (CSF-1) . FLT3LG increases the number of cells by activating hematopoietic progenitor cells.
  • SCF stem cell factor
  • CSF-1 colony stimulating factor 1
  • SCF Stem cell factor
  • Kit ligand also known as stem cell factor (SCF)
  • SCF belongs to the SCF family of type I transmembrane glycoproteins.
  • KITLG is a ligand for the receptor-type protein tyrosine kinase KIT.
  • SCF plays an important role in regulating cell survival and proliferation, hematopoiesis, stem cell maintenance, cell development, migration and function.
  • Interleukin 3 is a glycoprotein belonging to the hematopoietic growth factor family, which exhibits multi-lineage activity in preclinical in vitro and in vivo studies. Hematopoietic progenitor cells proliferate and differentiate into mature red blood cells, mast cells, megakaryocytes and granulocytes with the help of IL-3 protein.
  • Interleukin-6 is a multifunctional cytokine that regulates immune response, hematopoietic function, acute phase response and inflammatory response. Synergistically promotes hematopoietic cell proliferation with IL-3.
  • Erythropoietin is a major erythropoietin that interacts with various other growth factors (IL-3, IL-6, glucocorticoids, and SCF) that develop erythroid lineages from pluripotent progenitor cells.
  • Explosive forming unit-erythrocyte (BFU-E) cells begin to express erythropoietin receptors and are sensitive to erythropoietin. It is an important erythroid hematopoietic cytokine.
  • Holo human transferrin is a major ferritin in plasma, which forms a complex with iron ions for hemoglobin production in red blood cells.
  • CD235a also known as blood glycoprotein A, is a single transmembrane glycoprotein dominantly expressed on the surface of both mature red blood cells and erythroid precursor cells, is a lineage specific marker for red blood cells.
  • CD117 is mast cell/stem cell growth factor receptor, expressed on the surface of hematopoietic stem cells and other cells.
  • CD71 also known as Transferrin receptor 1
  • Transferrin receptor 1 is a transmembrane glycoprotein composed of two disulfide-bonded monomers linked by two disulfide bonds. Each monomer binds to a full transferrin molecule to produce an iron-transferrin-transferrin receptor complex that enters the cell by endocytosis.
  • PI Propidium Iodide binds to double-stranded DNA. PI cannot cross intact plasma membrane and therefore will only be present in DNA of cells where the plasma membrane has been compromised/permeabilized.
  • Hoechst33342 is a fluorescent dye used for DNA staining of living cells.
  • copGFP is a green fluorescent protein, which is a commonly used fluorescent reporter.
  • the lentivirus expressing URAT1 and UOX genes was produced by using MSCV-URAT1-copGFP (or MSCV-UOX-copGFP) , pSPAX2, VSVG at the ratio of 2: 1: 1.
  • MSCV-URAT1-copGFP or MSCV-UOX-copGFP
  • pSPAX2 VSVG
  • VSVG a 6-well plate as an example
  • 2 ⁇ g MSCV-URAT1-copGFP (or MSCV-UOX-copGFP) , 1 ⁇ g pSPAX2 and 1 ⁇ g VSVG were transfected by using calcium phosphate into HEK 293T cells for generating virus. 12 hours post transfection, calcium phosphate was replaced by fresh DMEM medium containing 10%FBS. The supernatant was collected at 48 hours and 72 hours post transfection and were filtered through a 0.45 ⁇ m filters.
  • the filtered supernatants are subjected to ultracentrifugation and concentrated at a speed of 70000 RCF for 2 hours at a temperature of 4°C. After centrifugation, the supernatant was removed and pellets containing viral particles were resuspended using differentiation stage 1 medium (described herein blow) .
  • the virus titer was quantified by using p24 ELISA kit, and stored at -80°C.
  • Example 2 Human red blood cells engineered to express URAT1 and UOX
  • Mature red blood cells are differentiated from early erythroid progenitor cells derived from hematopoietic stem cells.
  • hematopoietic stem cells due to the difficulty in obtaining hematopoietic stem cells from human bone marrows, we have developed an in vitro human erythroid differentiation culture system by using Lin-CD34+ HSCs or Lin-CD34-peripheral blood mononuclear cells (PBMCs) to generate mature human red cells.
  • PBMCs Lin-CD34-peripheral blood mononuclear cells
  • peripheral blood from gout patients or healthy subjects with matched blood types or umbilical cord blood was diluted 1: 1 with phosphate buffer, and PBMCs or perenrichment HSCs were isolated by centrifugation at 1200 x g for 15 minutes, using a lymphocyte separation solution (LymphoprepTM, STEMCELL Technologies) .
  • Lin-CD34-PBMCs from peripheral blood were isolated by using a Human Lineage Cell Depletion Set-DM kit (BD Biosciences) ; Lin-CD34+ HSCs from cord blood samples were isolated by using EasySep TM Human Cord Blood CD34 Positive Selection Kit II (Stemcell Technologies) .
  • Lin-CD34-PBMCs or Lin-CD34+ HSCs cells were cultured in hematopoietic stem cell expansion medium (StemSpanTM SFEM, STEMCELL Technologies) supplemented with penicillin-streptomycin (Gibco) and a combination of cytokines containing 50 ng/ml recombinant human fms-like tyrosine kinase 3 ligand (Flt3L) , 50 ng/ml recombinant human stem cell factor (SCF) , 10 ng/ml recombinant human interleukin 3 (IL-3) , 10 ng/ml recombinant human interleukin 6 (IL-6) or StemSpanTM CC100 (STEMCELL Technologies) . Cells were cultured at 37°C under 5%CO 2 .
  • StemSpanTM CC100 StemSpanTM CC100
  • the cultured cells were switched to the differentiation stage 1 medium consisting of IMDM (Iscove's Modified Dulbecco's Medium, Sigma-Aldrich) , 10-15%fetal bovine serum (FBS, Gibco) or human serum, 5-10%human plasma, 1-4 mM glutamine, 1-2%BSA (Albumin from bovine serum) , 300-600 ⁇ g/mL human transferrin (holo human transferrin, Sigma -Aldrich) , 8-13 ⁇ g/mL recombinant human insulin (Sigma-Aldrich) , 2%Penicillin-Streptomycin (Gibco) , 3-5 ng/mL recombinant human interleukin III (rhIL) -3, Peprotech) , 4-7 U/mL recombinant human erythropoietin (rhEpo, Amgen) , 100 ng/mL recombinant human stem cell factor
  • IMDM Iscove
  • the differentiation stage 2 medium was consisting of IMDM (Iscove's Modified Dulbecco's Medium, Sigma-Aldrich) , 15%fetal bovine serum (FBS, Gibco) or 15%autologous serum, 5-10%human plasma (Plasma) , 1-4 mM glutamine, 1-2%BSA (Albumin from bovine serum) , 300-600 ⁇ g/mL human transferrin (holo human transferrin, Sigma-Aldrich) , 8-13 ⁇ g/mL recombinant human insulin (Sigma-Aldrich) , 2%Penicillin-Streptomycin (Gibco) , 1-5 U/mL recombinant human erythropoietin (rhEpo, Amgen) . Cells were cultured another 7 days, during which the medium was changed with fresh differentiation stage 2 medium every 2 days.
  • IMDM Iscove's Modified Dulbecco's Medium, Sigma-Aldrich
  • URAT1 forward primer 5’-TCCAGACGGTGTACGAGATG-3’ (SEQ ID NO: 5)
  • URAT1 reverse primer 5’-GGATGTCCACGACACCAATGA-3’ (SEQ ID NO: 6)
  • UOX forward primer 5’-CGGTGTCCAGACGGTGTACG-3’ (SEQ ID NO: 7)
  • URAT1 reverse primer 5’–GGTCCAGCGGTGGCAGACAAT-3’ (SEQ ID NO: 8)
  • eRBCs were incubated with serum from gout patients.
  • the content of UA in the patient serum was measured at different time points to determine the consumption rates of UA by the engineered red blood cells in vitro. The results were showed in Fig. 6.
  • DIR labelled eRBCs were injected into the immune-deficient NSG mice through intravenous injection, and tissue distribution and survival of the labelled eRBCs cells in the mouse was analysed by live imaging, flow cytometry, immunohistochemistry, etc. Some results were showed in Fig. 7 and Fig. 8.
  • Example 6 Analysis of the function of eRBC-URAT1 or URAT1+UOX to consume the UA in vivo
  • ChromPure Rat IgG (Jackson ImmunoResearch, #012-000-003) at 50 ⁇ L/mouse were added and incubated with cells at 4°C for 5min.
  • Biotinylated anti-mouse TER119 antibodies (BD Pharmingen, #553672) at 1 ⁇ L/1*10 6 cells were added to cells and incubated at 4°C for 15 min.
  • Ms Lineage Panel (Fisher Scientific (Thermo Fisher Scientific) #BDB559971 ) were added to cells at the concentration of 2 ⁇ L /1*10 6 cells and incubated at 4C for 15 min.
  • Streptavidin Particles Plus -DM magnetic beads
  • streptavidin coated magnetic beads were removed by using the magnetic holder.
  • Cells were seeded into a 24-well plate at the concentration of 2*10 5 /mL at 37°C. Cells were infected with lentivirus as described previously.
  • the model of hyperuricemia was induced by intraperitoneal injection of 250mg/kg of MSU crystal suspension as described [20] .
  • Blood were collected from tail vein at 0, 2, 5, 8, 24h, analysing their serum UA concentration.
  • mice The function of mouse eRBC-URAT1+UOX in hyperuricemia mice
  • mice 2x10 8 functional mouse eRBCs were intravenously injected into mice. 3 days after injection, these mice were induced hyperuricemia by intraperitoneal injection of 250mg/kg of MSU crystal suspension, analysing their serum UA concentration at 0, 2, 5, 8, 24h, 48h.

Abstract

Provided are engineered red blood cells comprising a urate oxidase (UOX) or a functional variant thereof; a urate transporter 1 (URAT1) or a functional variant thereof; or a UOX or a functional variant thereof and a URAT1 or a functional variant thereof. Provided are the uses of the engineered red blood cells in treating hyperuricemia associated diseases.

Description

Engineering Red Blood Cells for Treating Gout and Hyperuricemia Diseases FIELD OF THE INVENTION
The present disclosure relates to engineering red blood cells, and in particular, to engineering red blood cells carrying URAT1 and/or UOX. The present disclosure also relates to methods for treating hyperuricemia associated diseases.
BACKGROUND
Gout is the most common form of inflammatory arthritis in adults, especially in men, with a prevalence ranging from 1%to 4%globally [1, 2] . Gout occurs when monosodium urate crystal (MSU) deposited in tissues, causing inflammation and intense pain of a gout attack. The biologic precursor to gout is elevated serum uric acid (UA) levels (i.e., hyperuricemia) . Although hyperuricemia is the strongest single risk factor for the development of gout and is universally present in patients with gout, not all individuals with hyperuricemia develop gout. Recent work has emphasized the importance of the innate immune response [2] .
Conventional urate-lowering agents such as anti-inflammatory drugs (colchicine) , xanthine oxidase inhibitors (allopurinol, febuxostat) or uricosuric agents (probenecid, benzbromarone) induce very slow reduction in UA deposits, not allowing for the rapid resolution of tophi for all patients with gout and are mainly used at the early stage [3] .
Urate oxidase (UOX, uricase) is a liver enzyme that metabolizes UA into allantoin, a more water-soluble compound, which is easily excreted by the kidney [2, 3] . All mammals produce UOX, except humans and certain primates. Indeed, during evolution, UOX was inactivated in humans primarily due to missense and frame-shift mutations in the gene encoding this enzyme [2, 4] . Uricase undeniably represents a valuable treatment option for chronic tophaceous gout when conventional urate-lowering agents may not be used.
Rasburicase, a recombinant UOX from A. flavus, was approved by EMEA in 2001 
Figure PCTCN2020124849-appb-000001
and the Food and Drug Administration (FDA) in 2002
Figure PCTCN2020124849-appb-000002
for tumor lysis syndrome [5, 6] . This agent significantly reduces serum UA levels and acts faster than allopurinol. The recommended dose is 0.2 mg/kg in children and adults [7, 8] . However, its biological half-life is short---only 21h, so rasburicase is given by infusion once daily for ≤ 7 days [7] . In addition, recent studies have shown that repeated UOX injections could cause anaphylactic reactions with the production of antibodies that neutralize UOX enzyme activity [9] .
Pegloticase, a recombinant porcine UOX, with a baboon C-terminal sequence, is a modified pegylated recombinant UOX developed to be the first non-immunogenic biologic for  treating the hyperuricemia of refractory gout, approved by the FDA for patients with chronic gout [3, 10] . A 6-month study versus placebo showed that pegloticase (infused at 8 mg every 2 weeks) , induced a significant decrease in plasma UA in about 40%of the patients (associated with a tendency for tophi dissolution) [3] . However, the remaining patients had no response, which was correlated with the formation of pegloticase antibodies and infusion reactions [3] . More than 10%of the patients treated with pegloticase had adverse events such as kidney stones, joint pain, anaemia, muscle spasms, dyspnea, headache, nausea, and fever [7] .
Available recombinant UOX (rasburicase, pegloticase) drugs are potent hypouricemic agents for gout. However, there are several limitations for the current therapy. First, UOX has significant immunogenicity and it may induce severe allergic reactions [11] . Conjugating therapeutic enzymes to PEG may reduce immune responses in patients. However, studies showed that many patients treated with PEG-conjugated enzymes developed anti-PEG antibodies. Moreover, PEG may adversely affect the activity of the conjugated enzyme, leading to reduced efficacy in the treatment [12, 13] . Second, the therapeutic enzymes may become inactivated or eliminated in vivo due to short half-life, limited bioavailability, and/or interactions with plasma proteins [14] . Third, production and purification of the enzymes tend to be time-consuming, and thus treatments with enzyme replacement therapy are very costly. The cost of treatment (calculated on an annual basis) is estimated to be about € 7200 for rasburicase and € 41, 240 for pegloticase [3] . Therefore, we need new gout therapy that is more efficacious and safer.
Engineered RBCs (eRBCs) are attractive carriers for the introduction of novel therapeutics, and have the following benefits: (1) red blood cells are the most abundant cells in the blood, occupying a quarter of all human cells, and are widely distributed to the human body through circulation; (2) red blood cells have a long life span (about 120 days for humans and 50 days for mice) . Furthermore, old or damaged red blood cells can be cleared by macrophages in the liver and spleen without causing immune responses; (3) red blood cells are biocompatible, especially when using autologous red blood cells; (4) mature red blood cells have no nuclei, mitochondria, or other organelles. Thus, any modification made to the DNA of RBC precursors is eliminated upon their enucleation and cannot lead to abnormal growth or tumorigenicity after their transfusion into a recipient; and (5) red blood cells protect the drug substance from premature inactivation and/or degradation, as well as the organism from the toxic effects of the drug [15-17] .
A major regulator of serum urate is renal excretion of UA. In humans, net reabsorption of UA into the blood predominates owing to less excretion of UA than is filtered at the  glomerulus. URAT1, a urate–anion exchanger, can transport UA from the urinary lumen, which is an important step for UA reabsorption [2, 18] .
A significant reduction in immunogenicity, the prolonging half-life of UOX and rapid resolution of tophi are the key objectives in the development of new gout therapy.
SUMMARY
In one aspect, the present disclosure provides an engineered red blood cell (eRBC) comprising:
a urate oxidase (UOX) or a functional variant thereof;.
a urate transporter 1 (URAT1) or a functional variant thereof; or
a UOX or a functional variant thereof and a URAT1 or a functional variant thereof.
In some embodiments, the URAT1 comprises an amino acid sequence of SEQ ID NO: 2.
In some embodiments, the UOX comprises an amino acid sequence of SEQ ID NO: 4.
In another aspect, the present disclosure provides a method for producing engineered red blood cells, which comprises:
1) collecting lineage negative cells (Lin-cells) from a blood sample or Lin-CD34+ cells from a bone marrow sample,
2) expanding the Lin-or Lin-CD34+ cells;
3) culturing the expanded Lin-or Lin-CD34+ cells to induce them to differentiate into erythroid cells; and, concurrently with the differentiation, introducing one or more exogenous nucleic acids into the expanded Lin-cells or Lin-CD34+ cells;
4) culturing the erythroid cells to induce enucleation,
wherein the one or more exogenous nucleic acids comprises a first polynucleotide sequence encoding a urate oxidase (UOX) or a functional variant thereof, a second polynucleotide sequence encoding a urate transporter 1 (URAT1) or a functional variant thereof; or both of the first polynucleotide sequence and the second polynucleotide sequence.
In some embodiments, the URAT1 comprises an amino acid sequence of SEQ ID NO: 2.
In some embodiments, the UOX comprises an amino acid sequence of SEQ ID NO: 4.
In some embodiments, the blood sample is a peripheral blood sample, a cord blood sample or a fetal blood sample.
In some embodiments, the blood sample is a human peripheral blood sample.
In some embodiments, the Lin-cells are Lin-CD34-cells.
In some embodiments, the step 1) includes isolating peripheral blood mononuclear cells (PBMCs) from the peripheral blood sample and isolating Lin-CD34-cells from the PBMCs.
In some embodiments, the step 1) includes removing lineage positive (Lin+) cells from the peripheral blood sample by using a lineage cell depletion kit.
In some embodiments, the step 2) including culturing the Lin-cells in a hematopoietic stem cell expansion medium supplemented with a combination of cytokines, and the combination of cytokines comprise fms-like tyrosine kinase 3 ligand (Flt3L) , stem cell factor (SCF) , interleukin 3 (IL-3) , and interleukin 6 (IL-6) .
In some embodiments, the combination of cytokines comprise 50 ng/mL human Flt3L, 50 ng/mL human SCF, 10 ng/mL human IL-3, and 10 ng/mL human IL-6.
In some embodiments, the hematopoietic stem cell expansion medium is StemSpan TM SFEM serum-free expansion medium.
In some embodiments, the step 2) including culturing the Lin-cells at 37℃ under 5%CO 2 for about 5 days.
In some embodiments, the step 3) comprising culturing the expanded Lin-cells in a first differentiation medium supplemented with cytokines related to erythroid development.
In some embodiments, the first differentiation medium is an Iscove's Modified Dulbecco's Medium (IMDM) containing: fetal bovine serum (FBS) or human serum, human plasma, glutamine, BSA, transferrin, insulin, Penicillin-Streptomycin, IL-3, EPO, and SCF.
In some embodiments, the first differentiation medium is an Iscove's Modified Dulbecco's Medium (IMDM) containing: 10-15%FBS or human serum, 5-10%human plasma, 1-4 mM glutamine, 1-2%BSA, 300-600 μg/mL human transferrin, 8-13 μg/mL human insulin, 2%Penicillin-Streptomycin, 3-5 ng/mL human IL-3, 4-7 U/mL human EPO, and 100 ng/mL human SCF.
In some embodiments, wherein the step 3) comprising culturing the expanded Lin-CD34-cells at 37℃ under 5%CO 2 for about 9 days.
In some embodiments, the step 4) comprising culturing the erythroid cells in a second differentiation medium, wherein the second differentiation medium is an Iscove's Modified Dulbecco's Medium (IMDM) containing: FBS or human serum, human plasma, glutamine, BSA, transferrin, insulin, Penicillin-Streptomycin, and EPO.
In some embodiments, the second differentiation medium is an Iscove's Modified Dulbecco's Medium (IMDM) containing: 15%FBS or human serum, 5-10%human plasma, 1-4 mM glutamine, 1-2%BSA, 300-600 μg/mL human transferrin, 8-13 μg/mL human insulin, 2%Penicillin-Streptomycin, and 1-5 U/mL human EPO.
In some embodiments, the step 4) comprising culturing erythroid cells at 37℃ under 5%CO 2 for about 7 days.
In some embodiments, in the step 3) the introduction of the one or more exogenous nucleic acids into the expanded Lin-cells is carried out on the 1st day of culturing the expanded Lin-cells in the first differentiation medium.
In some embodiments, the exogenous nucleic acid is an expression vector.
In some embodiments, the exogenous nucleic acid is a lentiviral expression vector.
In another aspect, the present disclosure provides a pharmaceutical composition comprising the engineered red blood cells and a physiologically acceptable excipient.
In another aspect, the present disclosure provides uses of the engineered red blood cells in the preparation of a medicament for treating a disorder associated with elevated serum uric acid levels.
In some embodiments, the disorder is gout or a hyperuricemia disease.
In another aspect, the present disclosure provides a method for the treatment of a disorder associated with elevated serum uric acid levels in a subject, which comprises:
a) taking a blood sample or a bone marrow sample from the subject,
b) producing engineered red blood cells by using the method described above; and
c) infusing a therapeutically effective amount of the engineered red blood cells into the subject.
In some embodiments, the disorder is gout or a hyperuricemia disease
In another aspect, the present disclosure provides a method for the treatment of a disorder associated with elevated serum uric acid levels in a subject, which comprises infusing a therapeutically effective amount of the engineered red blood cell into the subject.
In some embodiments, the method further comprises performing blood typing before the infusion to ensure the subject is compatible with the engineered red blood cell.
BRIEF DESCRIPTION OF THE DRAWINGS
Fig. 1 shows the RBC engineering strategy for gout and hyperuricemia therapy.
Fig. 2 depicts experimental scheme of engineering hematopoietic stem and/or progenitor cells to express URAT1 and UOX, and subsequently differentiating the engineered hematopoietic stem and/or progenitor cells in vitro into mature red blood cells carrying both functional proteins.
Fig. 3 depicts representative analysis that can be used to test the functions of engineered RBCs (eRBCs) expressing URAT1 and UOX in vitro, as well as their functions in vivo using established animal models for gout.
Fig. 4 shows a workflow of eRBC generation from mouse embryos. Mouse erythroid progenitors were isolated from fetal livers at embryonic 14.5 days. Cells were then infected  with MSCV-URAT1/UOX-copGFP before induced into terminally differentiated mouse red cells expressing URAT1 or UTRA1 and UOX.
Fig. 5 shows that eRBCs expressing URAT1 and/or UOX have undergone normal erythroid differentiation. Here shows an example of FACS analysis of eRBCs generated from Lin-PBMCs of a gout patient at day 8 of the in vitro differentiation. The following are the markers used in the FACS analyses: CD235a, CD117, CD71, Hoechst33342, and GFP (the reporter gene indicating expression of URAT1 and/or UOX) . CD235a, CD117 and CD71 are used to track the progression of erythroid development. Hoechst33342 stains DNA, and therefore, Hoechst 33342 negative cells are enucleated cells. The channels used to detect corresponding signals: Y585-PE: PI; B525-FITC: GFP; R660-APC: CD235a; B650-PC5.5: CD71; UV450: Hoechst33342. (A) . eRBC-URAT1 at day 8 of the in vitro erythroid differentiation; (B) . eRBC-URAT1+UOX at day 8 of the in vitro erythroid differentiation.
Fig. 6 Both eRBC-URAT1 and eRBC-URAT1+UOX generated from Lin-PBMCs of the gout patients reduced the UA concentration in vitro efficiently. (A) . UA concentration of the medium in which eRBCs were cultured. 10 5 eRBCs, eRBC-URAT1 or eRBC-URAT1+UOX were cultured in the medium with 465 μM UA (mimicking UA concentration of a gout patient) for 24 hours. After the incubation, the UA concentration in the medium was determined by a coupled enzyme reaction (Abcam) . (B) . The consumption rates of UA by eRBC-URAT1 and eRBC-UOX+URAT1 were calculated based on the results from Fig. 6A.
FIG. 7 shows tissue distribution of engineered RBCs in NSG mouse by live imaging. DIR-labeled RBCs were injected into NSG mouse via intravenous injection and the subsequent distributions of the labeled red blood cells were examined by in vivo imaging at 1 h, 5 h, 48 h, and 5 days after the injection of red blood cells. Mouse #1: no treatment ; Mouse #2: 2 μM DIR only in PBS; Mouse #3: 1x10 8 human red blood cells differentiated from PBMCs in vitro and labeled with 2 μM DIR; Mouse #4: 1x10 8 human red blood cells infected with vector only differentiated from PBMCs in vitro and labeled with 2 μM DIR; Mouse #5: 1x10 8 human red blood cells transducing with lentiviral vectors encoding phenylalanineammonialyase (PAL) and differentiated from PBMCs in vitro and labeled with 2 μM DIR; Mouse #6: 5x10 7 human red blood cells transducing with lentiviral vectors encoding glyoxylate and hydroxypyruvate reductase (GRHPR) and differentiated from PBMCs in vitro and labeled with 2 μM DIR.
FIG. 8 shows an example of images of tissue samples after the injection of red blood cells. DIR-stained red blood cells were intravenously injected into each mouse. Seven days after the injection, the mice were sacrificed, and the tissue samples were imaged. Mouse #1: no treatment ; Mouse #2: 2 μM DIR only in PBS; Mouse #4: 1x10 8 human red blood cells infected  with vector only differentiated from PBMCs in vitro and labeled with 2 μM DIR; Mouse #5: 1x10 8 human red blood cells transducing with lentiviral vectors encoding phenylalanineammonialyase (PAL) and differentiated from PBMCs in vitro and labeled with 2 μM DIR; Mouse #6: 5x10 7 human red blood cells transducing with lentiviral vectors encoding glyoxylate and hydroxypyruvate reductase (GRHPR) and differentiated from PBMCs in vitro and labeled with 2 μM DIR.
Fig. 9 shows that engineered mouse eRBC-URAT1+UOX reduced UA concentration in a hyperuricemia and gout mouse model. A representative functional assay of mouse eRBC-URAT1 and eRBC-UOX+URAT1 in vivo. (A) . Hyperuricemia was induced by intraperitoneal injection of MSU crystal suspension at the dose of 250 mg/kg, and the serum UA concentration was analyzed by a coupled enzyme reaction; (B) . Hyperuricemia was induced 3 days after 2x10 8 eRBC-URAT1 or eRBC-URAT1+UOX cells or PBS (Blank) were intravenously injected into the mice. UA concentration in the serum was measured to assess the therapeutic effects of the eRBCs.
DETAILED DESCRIPTION
Unless defined otherwise, technical and scientific terms used herein have the same meaning as commonly understood by a person of ordinary skill in the art. Any methods, devices and materials similar or equivalent to those described herein can be used in the practice of this invention. The following definitions are provided to facilitate understanding of certain terms used frequently herein and are not meant to limit the scope of the present disclosure.
The articles “a” and “an” are used herein to refer to one or to more than one (i.e., to at least one) of the grammatical object of the article. By way of example, “an element" means one element or more than one element.
The term “lineage negative cells” or “Lin-cells” as used herein refers to cells that are essentially free of lineage markers. Lineage markers are characteristic of cell lineages. Exemplary lineage markers are CD1c, CD3, CD11c, CD14, CD15, CD16, CD20, CD41, CD56, CD203c, CD235a, and/or BDCA2. In fact, lineage negative cells are essentially not stained by the lineage antibodies. Lineage negative cells comprise stem and progenitor cells. Accordingly, lineage negative cells also show stem and progenitor cell activity. Lin-cells or a blood cell population enriched for lineage negative cells can be purified by enriching a blood cell population that is essentially free of lineage markers. For example, the lineage negative cells can be purified by depleting cells that are positive for at least one lineage marker selected from the group consisting of CD1c, CD3, CD11c, CD14, CD15, CD16, CD20, CD41, CD56, CD203c, CD235a, and/or BDCA2. In some cases, a lineage cell depletion kit can be  used to perform the purification. To the contrary, lineage positive (Lin+) cells are a mix of all cells expressing mature cell lineage markers. Examples of lineage positive cells include T cells, B cells, NK cells, dendritic cells, monocytes, granulocytes, erythroid cells, and their committed precursors.
The term “functional variant” means any variant comprising substitution, deletion or addition of one or a few to plural amino acids to UOX or URAT1, provided that the variant substantially retains the same function as UOX or URAT1 possesses.
The term “culturing” as used herein refers to maintaining cells in a medium with or without cell expansion or differentiation for any period of time.
The term “differentiation” as used herein refers to a process by which a less specialized cell such as a stem cell develops or matures to possess a more distinct form and function with a concomitant loss of potential. Cells that are less specialized can be differentiated into cells that are more specialized by culturing the cells under particular conditions or in specific media as known in the art.
The term “pharmaceutically acceptable excipient” as used herein refers to a pharmaceutically acceptable material, composition or vehicle, such as a liquid or solid filler, diluent, carrier, manufacturing aid (e.g., lubricant, talc magnesium) , solvent or encapsulating material, involved in carrying or transporting a therapeutic compound for administration to a subject. Each excipient should be “acceptable” in the sense of being compatible with the other ingredients of the formulation and not injurious to the subject.
fms-like tyrosine kinase 3 ligand (Flt3L) or FMS-like tyrosine kinase 3 ligand (Flt-3 ligand) , also known as FL, Flt3L and FLT3LG, is a protein which promotes HSCs differentiation to various hematopoietic lineages. FLT3LG is structurally homologous to stem cell factor (SCF) and colony stimulating factor 1 (CSF-1) . FLT3LG increases the number of cells by activating hematopoietic progenitor cells.
Stem cell factor (SCF) or Kit ligand (KITLG) , also known as stem cell factor (SCF) , belongs to the SCF family of type I transmembrane glycoproteins. KITLG is a ligand for the receptor-type protein tyrosine kinase KIT. SCF plays an important role in regulating cell survival and proliferation, hematopoiesis, stem cell maintenance, cell development, migration and function.
Interleukin 3 (IL-3) is a glycoprotein belonging to the hematopoietic growth factor family, which exhibits multi-lineage activity in preclinical in vitro and in vivo studies. Hematopoietic progenitor cells proliferate and differentiate into mature red blood cells, mast cells, megakaryocytes and granulocytes with the help of IL-3 protein.
Interleukin-6 (IL-6) is a multifunctional cytokine that regulates immune response, hematopoietic function, acute phase response and inflammatory response. Synergistically promotes hematopoietic cell proliferation with IL-3.
Erythropoietin (Epo) is a major erythropoietin that interacts with various other growth factors (IL-3, IL-6, glucocorticoids, and SCF) that develop erythroid lineages from pluripotent progenitor cells. Explosive forming unit-erythrocyte (BFU-E) cells begin to express erythropoietin receptors and are sensitive to erythropoietin. It is an important erythroid hematopoietic cytokine.
Holo human transferrin is a major ferritin in plasma, which forms a complex with iron ions for hemoglobin production in red blood cells.
CD235a, also known as blood glycoprotein A, is a single transmembrane glycoprotein dominantly expressed on the surface of both mature red blood cells and erythroid precursor cells, is a lineage specific marker for red blood cells.
CD117 is mast cell/stem cell growth factor receptor, expressed on the surface of hematopoietic stem cells and other cells.
CD71, also known as Transferrin receptor 1, is a transmembrane glycoprotein composed of two disulfide-bonded monomers linked by two disulfide bonds. Each monomer binds to a full transferrin molecule to produce an iron-transferrin-transferrin receptor complex that enters the cell by endocytosis.
Propidium Iodide (PI) binds to double-stranded DNA. PI cannot cross intact plasma membrane and therefore will only be present in DNA of cells where the plasma membrane has been compromised/permeabilized.
Hoechst33342 is a fluorescent dye used for DNA staining of living cells.
copGFP is a green fluorescent protein, which is a commonly used fluorescent reporter.
We have established an in vitro RBC regeneration culture technology, which can increase proliferation of erythroid progenitors and enable efficient and stable production of enucleated RBCs. Normal human RBCs can be produced in culture from hematopoietic stem and progenitor cells, but the source of hematopoietic stem cells limits the clinical application, such as bone marrow (BM) aspirates, mobilized peripheral blood, and umbilical cord blood (UCB) . We generate a series of novel methods using lineage negative CD34 negative peripheral blood mononuclear cells (Lin-CD34-PBMCs) or lineage negative CD34 positive hematopoietic stem cells (Lin-CD34+ HSCs) to induce erythroid cell proliferation and differentiation, which lays a strong foundation for the eRBC therapy.
We use RBCs as new carriers to deliver UOX to the patient, via designing eRBCs to express both URAT1 and UOX, which can increase UA consumption rate and further improve  the consumption efficiency of UA (Fig. 1 and Fig. 2) . A series of experiments have been carried out to confirm the in vitro and in vivo functions of eRBCs carrying URAT1 or carrying both URAT1 and UOX (Fig. 3) .
EXAMPLES
Example 1. Lentivirus preparation
Design of MSCV-URAT1-copGFP and MSCV-UOX-copGFP plasmids: the cDNA sequences of URAT1 (Genbank, AY358183.1) and UOX (Genbank, X61765.1) are listed as followings. The synthesized DNA of the genes were cloned in the lentiviral vector MSCV.
The DNA sequence of URAT1: (SEQ ID NO: 1)
Figure PCTCN2020124849-appb-000003
The amino acid sequence of URAT1: (SEQ ID NO: 2)
Figure PCTCN2020124849-appb-000004
The DNA sequence of UOX: (SEQ ID NO: 3)
Figure PCTCN2020124849-appb-000005
The amino acid sequence of UOX: (SEQ ID NO: 4)
Figure PCTCN2020124849-appb-000006
The lentivirus expressing URAT1 and UOX genes was produced by using MSCV-URAT1-copGFP (or MSCV-UOX-copGFP) , pSPAX2, VSVG at the ratio of 2: 1: 1. Using 6-well plate as an example, 2 μg MSCV-URAT1-copGFP (or MSCV-UOX-copGFP) , 1 μg pSPAX2 and 1 μg VSVG were transfected by using calcium phosphate into HEK 293T cells for generating virus. 12 hours post transfection, calcium phosphate was replaced by fresh DMEM medium containing 10%FBS. The supernatant was collected at 48 hours and 72 hours post transfection and were filtered through a 0.45 μm filters.
The filtered supernatants are subjected to ultracentrifugation and concentrated at a speed of 70000 RCF for 2 hours at a temperature of 4℃. After centrifugation, the supernatant was removed and pellets containing viral particles were resuspended using differentiation stage 1 medium (described herein blow) . The virus titer was quantified by using p24 ELISA kit, and stored at -80℃.
Example 2. Human red blood cells engineered to express URAT1 and UOX
Mature red blood cells are differentiated from early erythroid progenitor cells derived from hematopoietic stem cells. However, due to the difficulty in obtaining hematopoietic stem cells from human bone marrows, we have developed an in vitro human erythroid differentiation culture system by using Lin-CD34+ HSCs or Lin-CD34-peripheral blood  mononuclear cells (PBMCs) to generate mature human red cells. The detailed steps are as follows.
1) The peripheral blood from gout patients or healthy subjects with matched blood types or umbilical cord blood was diluted 1: 1 with phosphate buffer, and PBMCs or perenrichment HSCs were isolated by centrifugation at 1200 x g for 15 minutes, using a lymphocyte separation solution (LymphoprepTM, STEMCELL Technologies) .
2) Lin-CD34-PBMCs from peripheral blood were isolated by using a Human Lineage Cell Depletion Set-DM kit (BD Biosciences) ; Lin-CD34+ HSCs from cord blood samples were isolated by using EasySep TM Human Cord Blood CD34 Positive Selection Kit II (Stemcell Technologies) .
3) The isolated Lin-CD34-PBMCs or Lin-CD34+ HSCs cells were cultured in hematopoietic stem cell expansion medium (StemSpanTM SFEM, STEMCELL Technologies) supplemented with penicillin-streptomycin (Gibco) and a combination of cytokines containing 50 ng/ml recombinant human fms-like tyrosine kinase 3 ligand (Flt3L) , 50 ng/ml recombinant human stem cell factor (SCF) , 10 ng/ml recombinant human interleukin 3 (IL-3) , 10 ng/ml recombinant human interleukin 6 (IL-6) or StemSpanTM CC100 (STEMCELL Technologies) . Cells were cultured at 37℃ under 5%CO 2 .
4) At day 5 during the culture, the cultured cells were switched to the differentiation stage 1 medium consisting of IMDM (Iscove's Modified Dulbecco's Medium, Sigma-Aldrich) , 10-15%fetal bovine serum (FBS, Gibco) or human serum, 5-10%human plasma, 1-4 mM glutamine, 1-2%BSA (Albumin from bovine serum) , 300-600 μg/mL human transferrin (holo human transferrin, Sigma -Aldrich) , 8-13 μg/mL recombinant human insulin (Sigma-Aldrich) , 2%Penicillin-Streptomycin (Gibco) , 3-5 ng/mL recombinant human interleukin III (rhIL) -3, Peprotech) , 4-7 U/mL recombinant human erythropoietin (rhEpo, Amgen) , 100 ng/mL recombinant human stem cell factor (rhSCF, Peprotech) . Cells were cultured_at 37℃ under 5%CO 2.
5) At day 6 during the culture, cells were infected with lentivirus expressing URAT1, or URAT1 and UOX. Cultured cells were resuspended with the differentiation stage 1 medium at a density of 1 x 10 6 /mL and infected with the virus at the concentration of 5×10 7 TU/ml-5×10 8 TU/ml with polybrene (Merck, 10ng/mL) . Centrifugal infection was performed using a horizontal rotor centrifuge for 90 min at a speed of 500 x g and 32℃.
6) 12 hours post infection, medium was replaced with fresh differentiation stage 2 medium at 37 ℃under 5%CO 2. The differentiation stage 2 medium was consisting of IMDM (Iscove's Modified Dulbecco's Medium, Sigma-Aldrich) , 15%fetal bovine serum (FBS, Gibco) or 15%autologous serum, 5-10%human plasma (Plasma) , 1-4 mM glutamine, 1-2%BSA  (Albumin from bovine serum) , 300-600 μg/mL human transferrin (holo human transferrin, Sigma-Aldrich) , 8-13 μg/mL recombinant human insulin (Sigma-Aldrich) , 2%Penicillin-Streptomycin (Gibco) , 1-5 U/mL recombinant human erythropoietin (rhEpo, Amgen) . Cells were cultured another 7 days, during which the medium was changed with fresh differentiation stage 2 medium every 2 days. The expression of CD235a, CD117, CD71, Hoechst 33342 on cells during the final differentiation in vitro were determined by flow cytometry.
During the culturing, we performed FACS analyses for CD235a, CD117, CD71, Hoechst33342, and GFP (the reporter gene indicating expression of URAT1 and/or UOX) . A typical result was showed in Fig 5.
Example 3. Analysis of URAT1 and UOX expression on engineered RBCs.
Total RNA was isolated from 10 6 eRBC-URAT1+UOX and eRBC-control, using RNeasy mini kit (QIAGEN) according to the manufacturer’s protocol. cDNA was synthesized using a PrimeScript RT Reagent Kit with gDNA Eraser (Takara) with oligo (dT) primers. qRT-PCR was performed using Terra TM qPCR Direct TB Green (Takara) . The following are the primer sequences used for qRT-PCR:
URAT1 forward primer: 5’-TCCAGACGGTGTACGAGATG-3’ (SEQ ID NO: 5)
URAT1 reverse primer: 5’-GGATGTCCACGACACCAATGA-3’ (SEQ ID NO: 6)
UOX forward primer: 5’-CGGTGTCCAGACGGTGTACG-3’ (SEQ ID NO: 7)
URAT1 reverse primer: 5’–GGTCCAGCGGTGGCAGACAAT-3’ (SEQ ID NO: 8)
18s forward primer: 5’-GCTTAATTTGACTCAACACGGGA-3’ (SEQ ID NO: 9)
18s reverse primer: 5’-AGCTATCAATCTGTCAATCCTGTC-3’ (SEQ ID NO: 10)
Example 4. Analysis of function of eRBC-URAT1 or URAT1+UOX in vitro
eRBCs were incubated with serum from gout patients. The content of UA in the patient serum was measured at different time points to determine the consumption rates of UA by the engineered red blood cells in vitro. The results were showed in Fig. 6.
Example 5. Tissue distribution and the lifespan of human eRBCs in NSG mice
DIR labelled eRBCs were injected into the immune-deficient NSG mice through intravenous injection, and tissue distribution and survival of the labelled eRBCs cells in the mouse was analysed by live imaging, flow cytometry, immunohistochemistry, etc. Some results were showed in Fig. 7 and Fig. 8.
Example 6. Analysis of the function of eRBC-URAT1 or URAT1+UOX to consume the UA  in vivo
Engineering mouse red blood cells to express URAT1 and UOX (Fig. 4)
1. Isolating mouse fetal liver cells (FLC) from embryos at E13.5-14.5 days as described previously [19] .
3. Single-cell suspension solution was collected by pipetting up and down, which were subsequently filtered by 25μm strainers (BD Falcon 35-2235) .
4. Cells were centrifuged at 1500 RPM for 5 min, and re-suspend by the red cell lysis buffer (Ammonium Chloride Solution from Stemcell) for 10 mins at 4℃.
5. Cells were centrifuged again at 1500 RPM for 5 min. The lysis buffer was removed and cells were resuspended with 10 mL PBS-2%FBS.
6. ChromPure Rat IgG (Jackson ImmunoResearch, #012-000-003) at 50 μL/mouse were added and incubated with cells at 4℃ for 5min.
7. Biotinylated anti-mouse TER119 antibodies (BD Pharmingen, #553672) at 1 μL/1*10 6 cells were added to cells and incubated at 4℃ for 15 min.
8. Ms Lineage Panel (Fisher Scientific (Thermo Fisher Scientific) #BDB559971 ) were added to cells at the concentration of 2 μL /1*10 6 cells and incubated at 4C for 15 min.
9. Cells were washed with 10 times volume of PBS and were centrifuged at 1500 RPM for 5 min at 4℃.
10. Streptavidin Particles Plus -DM (magnetic beads) (BD Pharmigen , #557812) (5μL/1*10 6 cells) were added to cells and incubated at 4C for 30min. Afterwards, streptavidin coated magnetic beads were removed by using the magnetic holder.
11. The remaining cells, which were lineage negative mouse cells, were transferred to new tubes.
14. Cells were seeded into a 24-well plate at the concentration of 2*10 5/mL at 37℃. Cells were infected with lentivirus as described previously.
15. After the infection, cells were cultured into the differentiated medium for 44-48 hours for the maturation.
Mouse model of hyperuricemia
The model of hyperuricemia was induced by intraperitoneal injection of 250mg/kg of MSU crystal suspension as described [20] . Blood were collected from tail vein at 0, 2, 5, 8, 24h, analysing their serum UA concentration.
The function of mouse eRBC-URAT1+UOX in hyperuricemia mice
2x10 8 functional mouse eRBCs were intravenously injected into mice. 3 days after injection, these mice were induced hyperuricemia by intraperitoneal injection of 250mg/kg of MSU crystal suspension, analysing their serum UA concentration at 0, 2, 5, 8, 24h, 48h.
Here, we assessed the therapeutic functions of eRBC-URAT1+UOX in our mouse model of hyperuricemia. We injected 2×10 8 eRBCs-URAT1+UOX intravenously 3 days before MSU injection. The eRBCs-URAT1+UOX significantly alleviated the elevated serum UA in the mouse model (Fig. 9) .
References:
[1] . Smith, E., et al., The global burden of gout: estimates from the Global Burden of Disease 2010 study. Ann Rheum Dis, 2014. 73 (8) : p. 1470-6.
[2] . Merriman, T.R. and N. Dalbeth, The genetic basis of hyperuricaemia and gout. Joint Bone Spine, 2011. 78 (1) : p. 35-40.
[3] . Garay, R.P., et al., Therapeutic perspectives on uricases for gout. Joint Bone Spine, 2012. 79 (3) : p. 237-42.
[4] . Wu, X.W., et al., Two independent mutational events in the loss of urate oxidase during hominoid evolution. J Mol Evol, 1992. 34 (1) : p. 78-84.
[5] . Cortes, J., et al., Control of plasma uric acid in adults at risk for tumor Lysis syndrome: efficacy and safety of rasburicase alone and rasburicase followed by allopurinol compared with allopurinol alone--results of a multicenter phase III study. J Clin Oncol, 2010. 28 (27) : p. 4207-13.
[6] . Coiffier, B., et al., Efficacy and safety of rasburicase (recombinant urate oxidase) for the prevention and treatment of hyperuricemia during induction chemotherapy of aggressive non-Hodgkin's lymphoma: results of the GRAAL1 (Groupe d'Etude des Lymphomes de l'Adulte Trial on Rasburicase Activity in Adult Lymphoma) study. J Clin Oncol, 2003. 21 (23) : p. 4402-6.
[7] . George, R.J. and J.S. Sundy, Pegloticase for treating refractory chronic gout. Drugs Today (Barc) , 2012. 48 (7) : p. 441-9.
[8] . Galardy, P.J., et al., Rasburicase in the prevention of laboratory/clinical tumour lysis syndrome in children with advanced mature B-NHL: a Children's Oncology Group Report. Br J Haematol, 2013. 163 (3) : p. 365-72.
[9] . Richette, P. and T. Bardin, Successful treatment with rasburicase of a tophaceous gout in a patient allergic to allopurinol. Nat Clin Pract Rheumatol, 2006. 2 (6) : p. 338-42; quiz 343.
[10] . Sherman, M.R., M.G. Saifer and F. Perez-Ruiz, PEG-uricase in the management of  treatment-resistant gout and hyperuricemia. Adv Drug Deliv Rev, 2008. 60 (1) : p. 59-68.
[11] . Richette, P. and T. Bardin, Successful treatment with rasburicase of a tophaceous gout in a patient allergic to allopurinol. Nat Clin Pract Rheumatol, 2006. 2 (6) : p. 338-42; quiz 343.
[12] . Ekladious, I., Y.L. Colson and M.W. Grinstaff, Polymer-drug conjugate therapeutics: advances, insights and prospects. Nat Rev Drug Discov, 2019. 18 (4) : p. 273-294.
[13] . Wraith, J.E., Limitations of enzyme replacement therapy: Current and future. Journal of Inherited Metabolic Disease, 2006. 29 (2-3) : p. 442-447.
[14] . Solomon, M. and S. Muro, Lysosomal enzyme replacement therapies: Historical development, clinical outcomes, and future perspectives. Adv Drug Deliv Rev, 2017. 118: p. 109-134.
[15] . Shi, J., et al., Engineered red blood cells as carriers for systemic delivery of a wide array of functional probes. Proc Natl Acad Sci U S A, 2014. 111 (28) : p. 10131-6.
[16] . Pishesha, N., et al., Engineered erythrocytes covalently linked to antigenic peptides can protect against autoimmune disease. Proc Natl Acad Sci U S A, 2017. 114 (12) : p. 3157-3162.
[17] . Huang, N.J., et al., Genetically engineered red cells expressing single domain camelid antibodies confer long-term protection against botulinum neurotoxin. Nat Commun, 2017. 8 (1) : p. 423.
[18] . Nakanishi, T., et al., Functional cooperation of URAT1 (SLC22A12) and URATv1 (SLC2A9) in renal reabsorption of urate. Nephrol Dial Transplant, 2013. 28 (3) : p. 603-11.
[19] . Flygare, J., et al., HIF1alpha synergizes with glucocorticoids to promote BFU-E progenitor self-renewal. Blood, 2011. 117 (12) : p. 3435-44.
[20] . Chen, G.L., W. Wei and S.Y. Xu, Effect and mechanism of total saponin of Dioscorea on animal experimental hyperuricemia. Am J Chin Med, 2006. 34 (1) : p. 77-85.

Claims (32)

  1. An engineered red blood cell (eRBC) comprising:
    a urate oxidase (UOX) or a functional variant thereof;.
    a urate transporter 1 (URAT1) or a functional variant thereof; or
    a UOX or a functional variant thereof and a URAT1 or a functional variant thereof.
  2. The engineered red blood cell of claim 1, wherein the URAT1 comprises an amino acid sequence of SEQ ID NO: 2.
  3. The engineered red blood cell of claim 1 or claim 2, wherein the UOX comprises an amino acid sequence of SEQ ID NO: 4.
  4. A method for producing engineered red blood cells comprising:
    1) collecting lineage negative cells (Lin-cells) from a blood sample or Lin-CD34+ cells from a bone marrow sample,
    2) expanding the Lin-cells or Lin-CD34+ cells;
    3) culturing the expanded Lin-cells or Lin-CD34+ cells to induce them to differentiate into erythroid cells; and, concurrently with the differentiation, introducing one or more exogenous nucleic acids into the expanded Lin-cells or Lin-CD34+ cells; and
    4) culturing the erythroid cells to induce enucleation,
    wherein the one or more exogenous nucleic acids comprises a first polynucleotide sequence encoding a urate oxidase (UOX) or a functional variant thereof, a second polynucleotide sequence encoding a urate transporter 1 (URAT1) or a functional variant thereof; or both of the first polynucleotide sequence and the second polynucleotide sequence.
  5. The method of claim 4, wherein the URAT1 comprises an amino acid sequence of SEQ ID NO: 2.
  6. The method of claim 4 or claim 5, wherein the UOX comprises an amino acid sequence of SEQ ID NO: 4.
  7. The method of any one of claims 4-6, wherein the blood sample is a peripheral blood sample, a cord blood sample or a fetal blood sample.
  8. The method of any one of claims 4-7, wherein the blood sample is a human peripheral blood sample.
  9. The method of any one of claims 4-8, wherein the Lin-cells are Lin-CD34-cells.
  10. The method of any one of claims 4-9, wherein the step 1) includes isolating peripheral blood mononuclear cells (PBMCs) from the peripheral blood sample and isolating Lin-CD34-cells from the PBMCs.
  11. The method of any one of claims 4-10, wherein the step 1) comprising removing lineage positive (Lin+) cells from the peripheral blood sample by using a lineage cell depletion kit.
  12. The method of any one of claims 4-11, wherein the step 2) including culturing the Lin-cells in a hematopoietic stem cell expansion medium supplemented with a combination of cytokines, and wherein combination of cytokines comprise fms-like tyrosine kinase 3 ligand (Flt3L) , stem cell factor (SCF) , interleukin 3 (IL-3) , and interleukin 6 (IL-6) .
  13. The method of any one of claims 4-12, wherein the combination of cytokines comprise 50 ng/mL human Flt3L, 50 ng/mL human SCF, 10 ng/mL human IL-3, and 10 ng/mL human IL-6.
  14. The method of any one of claims 4-13, wherein the hematopoietic stem cell expansion medium is StemSpan TM SFEM serum-free expansion medium.
  15. The method of any one of claims 4-14, wherein the step 2) including culturing the Lin-cells at 37℃ under 5%CO 2 for about 5 days.
  16. The method of any one of claims 4-15, wherein the step 3) comprising culturing the expanded Lin-cells in a first differentiation medium supplemented with cytokines related to erythroid development.
  17. The method of any one of claims 4-16, wherein the first differentiation medium is an Iscove's Modified Dulbecco's Medium (IMDM) containing: fetal bovine serum (FBS) or human serum, human plasma, glutamine, BSA, transferrin, insulin, Penicillin-Streptomycin, IL-3, EPO, and SCF.
  18. The method of any one of claims 4-17, wherein the first differentiation medium is an Iscove's Modified Dulbecco's Medium (IMDM) containing: 10-15%FBS or human serum, 5-10%human plasma, 1-4 mM glutamine, 1-2%BSA, 300-600 μg/mL human transferrin, 8-13 μg/mL human insulin, 2%Penicillin-Streptomycin, 3-5 ng/mL human IL-3, 4-7 U/mL human EPO, and 100 ng/mL human SCF.
  19. The method of any one of claims 4-18, wherein the step 3) comprising culturing the expanded Lin-CD34-cells at 37℃ under 5%CO 2 for about 9 days.
  20. The method of any one of claims 4-19, wherein the step 4) comprising culturing the erythroid cells in a second differentiation medium, wherein the second differentiation medium is an Iscove's Modified Dulbecco's Medium (IMDM) containing: FBS or human serum, human plasma, glutamine, BSA, transferrin, insulin, Penicillin-Streptomycin, and EPO.
  21. The method of any one of claims 4-20, wherein the second differentiation medium is an Iscove's Modified Dulbecco's Medium (IMDM) containing: 15%FBS or human serum,  5-10%human plasma, 1-4 mM glutamine, 1-2%BSA, 300-600 μg/mL human transferrin, 8-13 μg/mL human insulin, 2%Penicillin-Streptomycin, and 1-5 U/mL human EPO.
  22. The method of any one of claims 4-21, wherein the step 4) comprising culturing erythroid cells at 37℃ under 5%CO 2 for about 7 days.
  23. The method of any one of claims 4-22, wherein, in the step 3) , the introduction of the one or more exogenous nucleic acids into the expanded Lin-cells is carried out on the 1st day of culturing the expanded Lin-cells in the first differentiation medium.
  24. The method of any one of claims 4-23, wherein the exogenous nucleic acid is an expression vector.
  25. The method of any one of claims 4-24, wherein the exogenous nucleic acid is a lentiviral expression vector.
  26. A pharmaceutical composition comprising an engineered red blood cell of any one of claims 1-3 and a physiologically acceptable excipient.
  27. Use of engineered red blood cells of any one of claims 1-3 in the preparation of a medicament for treating a disorder associated with elevated serum uric acid levels.
  28. The use of claim 27, wherein the disorder is gout or a hyperuricemia disease.
  29. A method for the treatment of a disorder associated with elevated serum uric acid levels in a subject comprising:
    a) taking a blood sample or a bone marrow sample from the subject,
    b) producing engineered red blood cells by using a method of any one of the claims 4-24; and
    c) infusing a therapeutically effective amount of the engineered red blood cells into the subject.
  30. The method of claim 29, wherein the disorder is gout or a hyperuricemia disease
  31. A method for the treatment of a disorder associated with elevated serum uric acid levels in a subject comprising infusing a therapeutically effective amount of the engineered red blood cell of any one of claims 1-3 into the subject.
  32. The method of claim 31 further comprising performing blood typing before the infusion to ensure the subject is compatible with the engineered red blood cell.
PCT/CN2020/124849 2019-10-29 2020-10-29 Engineering red blood cells for treating gout and hyperuricemia diseases WO2021083278A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202080076275.7A CN114746546A (en) 2019-10-29 2020-10-29 Engineered erythrocytes for the treatment of gout and hyperuricemia

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN2019113940 2019-10-29
CNPCT/CN2019/113940 2019-10-29

Publications (1)

Publication Number Publication Date
WO2021083278A1 true WO2021083278A1 (en) 2021-05-06

Family

ID=75715850

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2020/124849 WO2021083278A1 (en) 2019-10-29 2020-10-29 Engineering red blood cells for treating gout and hyperuricemia diseases

Country Status (2)

Country Link
CN (1) CN114746546A (en)
WO (1) WO2021083278A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022166913A1 (en) * 2021-02-04 2022-08-11 Westlake Therapeutics (Hangzhou) Co. Limited Modified red blood cells and uses thereof for treating hyperuricemia and gout

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2024017192A1 (en) * 2022-07-18 2024-01-25 西湖生物医药科技(上海)有限公司 Drug delivery system comprising blood cells

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20070274977A1 (en) * 2005-04-11 2007-11-29 Jacob Hartman Methods for Lowering Elevated Uric Acid Levels Using Intravenous Injections of PEG-Uricase
WO2010031859A1 (en) * 2008-09-18 2010-03-25 Centre National De La Recherche Scientifique (Cnrs) Use of a compound capable of reducing the uric acid level for the prevention and/or the treatment of lung inflammation and fibrosis
WO2010133298A1 (en) * 2009-05-19 2010-11-25 Eth Zurich Control of uric acid homeostasis
US20180258406A1 (en) * 2015-05-15 2018-09-13 Medimmune, Llc. Improved Uricase Sequences and Methods of Treatment
CN109010806A (en) * 2018-07-26 2018-12-18 南开大学 A kind of degradable internal uric acid simultaneously realizes the collaboration complex enzyme and its preparation method and application that by-product is removed
WO2019183292A1 (en) * 2018-03-20 2019-09-26 Rubius Therapeutics, Inc. Therapeutic cell systems and methods for treating hyperuricemia and gout

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN110129273B (en) * 2019-05-10 2020-09-08 西湖生物医药科技(杭州)有限公司 Genetically engineered erythrocyte carrying anti-PD-1 single-chain antibody and preparation method thereof

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20070274977A1 (en) * 2005-04-11 2007-11-29 Jacob Hartman Methods for Lowering Elevated Uric Acid Levels Using Intravenous Injections of PEG-Uricase
WO2010031859A1 (en) * 2008-09-18 2010-03-25 Centre National De La Recherche Scientifique (Cnrs) Use of a compound capable of reducing the uric acid level for the prevention and/or the treatment of lung inflammation and fibrosis
WO2010133298A1 (en) * 2009-05-19 2010-11-25 Eth Zurich Control of uric acid homeostasis
US20180258406A1 (en) * 2015-05-15 2018-09-13 Medimmune, Llc. Improved Uricase Sequences and Methods of Treatment
WO2019183292A1 (en) * 2018-03-20 2019-09-26 Rubius Therapeutics, Inc. Therapeutic cell systems and methods for treating hyperuricemia and gout
CN109010806A (en) * 2018-07-26 2018-12-18 南开大学 A kind of degradable internal uric acid simultaneously realizes the collaboration complex enzyme and its preparation method and application that by-product is removed

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
PUI C-H, RELLING MV, LASCOMBES F, HARRISON PL, STRUXIANO A, MONDESIR J-M, RIBEIRO RC, SANDLUND JT, RIVERA GK, EVANS WE, MAHMOUD HH: "Urate oxidase in prevention and treatment of hyperuricemia associated with lymphoid malignancies", LEUKEMIA, vol. 11, no. 11, 1 November 1997 (1997-11-01), pages 1813 - 1816, XP055807953, ISSN: 0887-6924, DOI: 10.1038/sj.leu.2400850 *

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022166913A1 (en) * 2021-02-04 2022-08-11 Westlake Therapeutics (Hangzhou) Co. Limited Modified red blood cells and uses thereof for treating hyperuricemia and gout

Also Published As

Publication number Publication date
CN114746546A (en) 2022-07-12

Similar Documents

Publication Publication Date Title
CN107429232B (en) Immunoregulatory enhanced cells and methods of use and production thereof
US20200338131A1 (en) Method
JP6971986B2 (en) Mesenchymal stem cells to enhance the antitumor activity of immunotherapy
WO2021083278A1 (en) Engineering red blood cells for treating gout and hyperuricemia diseases
AU2014338555A1 (en) Method
JP2021532776A (en) Methods for genetic modification of hematopoietic cells
EP4010462A1 (en) Cellular compositions comprising viral vectors and methods of treatment
US20210154237A1 (en) Compositions and methods for stem cell transplant
Valtieri et al. Efficient transfer of selectable and membrane reporter genes in hematopoietic progenitor and stem cells purified from human peripheral blood
KR20230145381A (en) Modified Stem Cell Compositions and Methods of Use
AU2020459825A1 (en) Therapeutic agent for dystrophic epidermolysis bullosa
KR20060035640A (en) Method for transplanting lymphohematopoietic cells into mammal
US20030059413A1 (en) Retroviral vectors encoding interferon alpha and uses thereof
WO2021073613A1 (en) Engineered red blood cells presenting specific cancer neoantigen with artificial mhc
WO2020219353A1 (en) Red blood cells expressing von willebrand factor protease and methods of use thereof
WO2023086595A1 (en) Biomarkers of megakaryocyte-derived extracellular vesicles
CN115997009A (en) Methods and compositions for in vivo transduction of hematopoietic stem and progenitor cells
CN110656091A (en) Application of recombinant mesenchymal stem cells in preparation of medicine for treating myocardial infarction
CN114853902A (en) Chimeric antigen receptor, expression gene thereof, CAR-modified NK cell and application
WO2007148609A1 (en) Ex vivo/in vivo proliferation of hematopoietic stem cell or hematopoietic progenitor cell by timp-3

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 20882051

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 20882051

Country of ref document: EP

Kind code of ref document: A1