[go: up one dir, main page]

WO2024088213A1 - Combination to induce specific immune tolerance - Google Patents

Combination to induce specific immune tolerance Download PDF

Info

Publication number
WO2024088213A1
WO2024088213A1 PCT/CN2023/125936 CN2023125936W WO2024088213A1 WO 2024088213 A1 WO2024088213 A1 WO 2024088213A1 CN 2023125936 W CN2023125936 W CN 2023125936W WO 2024088213 A1 WO2024088213 A1 WO 2024088213A1
Authority
WO
WIPO (PCT)
Prior art keywords
seq
rbc
agent
sortase
combination
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.)
Ceased
Application number
PCT/CN2023/125936
Other languages
French (fr)
Inventor
Xiaofei GAO
Xiaoqian NIE
Yuehua Liu
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.)
Westlake Therapeutics Shanghai Co Ltd
Original Assignee
Westlake Therapeutics Shanghai Co Ltd
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 Shanghai Co Ltd filed Critical Westlake Therapeutics Shanghai Co Ltd
Priority to CN202380074882.3A priority Critical patent/CN120152732A/en
Publication of WO2024088213A1 publication Critical patent/WO2024088213A1/en
Anticipated expiration legal-status Critical
Ceased legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/06Antigout agents, e.g. antihyperuricemic or uricosuric agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/56Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids
    • A61K31/57Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids substituted in position 17 beta by a chain of two carbon atoms, e.g. pregnane or progesterone
    • A61K31/573Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids substituted in position 17 beta by a chain of two carbon atoms, e.g. pregnane or progesterone substituted in position 21, e.g. cortisone, dexamethasone, prednisone or aldosterone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/66Phosphorus compounds
    • A61K31/675Phosphorus compounds having nitrogen as a ring hetero atom, e.g. pyridoxal phosphate
    • 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
    • A61K38/46Hydrolases (3)
    • A61K38/47Hydrolases (3) acting on glycosyl compounds (3.2), e.g. cellulases, lactases
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/69Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit
    • A61K47/6901Conjugates being cells, cell fragments, viruses, ghosts, red blood cells or viral vectors
    • 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

Definitions

  • the present disclosure relates generally to modified red blood cells (RBCs) , and more particularly to combinations of an agent conjugated to an RBC with an immunosuppressor and uses thereof to induce immune tolerance specific to the agent.
  • RBCs modified red blood cells
  • UOX Urate oxidase
  • uricase Urate oxidase
  • UOX uricase
  • UOX uric acid
  • 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.
  • Uricase 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.
  • FDA Food and Drug Administration
  • This agent significantly reduces serum UA levels and acts faster than allopurinol.
  • the recommended dose is 0.2 mg/kg in children and adults.
  • its biological half-life is short (only 21h) , so rasburicase is given by infusion once daily for ⁇ 7 days.
  • recent studies have shown that repeated UOX injections could cause anaphylactic reactions with the production of antibodies that neutralize UOX enzyme activity [1-6] .
  • UOX rasburicase, pegloticase
  • UOX is potent hypouricemic agents for gout.
  • UOX has significant immunogenicity and it may induce severe allergic reactions.
  • Conjugating therapeutic enzymes to polyethylene glycol (PEG) may reduce immune responses in patients.
  • PEG polyethylene glycol
  • PEG may adversely affect the activity of the conjugated enzyme, leading to reduced efficacy in the treatment.
  • the therapeutic enzymes may become inactivated or eliminated in vivo due to short half-life, limited bioavailability, and/or interactions with plasma proteins.
  • production and purification of the enzymes tend to be time-consuming, and treatments with enzyme replacement therapy are very costly. Therefore, there is a need for new gout therapy that is more efficacious and safer.
  • Red blood cells (RBCs) , the most common cell type in the human body, have been widely investigated as an in vivo drug delivery system for over three decades due to their unique biological properties, including, for example, (i) widespread circulation range throughout the body; (ii) good biocompatibility as a biological material with long in vivo survival time; (iii) large surface to volume ratio; and (iv) no nucleus, mitochondria, and other cellular organelles.
  • RBCs have been reported as an interesting tool for antigen delivery to induce specific immune tolerance (see, e.g., Cremel et al., Int J Pharm 2013 Feb 25; 443 (1-2) : 39-49) .
  • RBCs Aged and damaged RBCs are naturally removed daily in the liver and spleen where phagocytes such as macrophages and monocytes can process and present major histocompatibility complex (MHC) -associated antigens to the T cells.
  • MHC major histocompatibility complex
  • an agent delivered by an RBC in combination with an immunosuppressor induces durable immune tolerance that enables safer and more efficacious treatment of a disease with the agent.
  • a general aspect of the application relates to a combination comprising (a) an agent coupled to a red blood cell (RBC) and (b) an immunosuppressor.
  • the agent can be coupled to the RBC by any means, such as encapsulation within the RBC or surface coupling, and either ex vivo or in vivo.
  • a combination comprising (a) an agent conjugated to a red blood cell (RBC) and (b) an immunosuppressor.
  • RBC red blood cell
  • the agent can be conjugated to the RBC by any means, such as chemical conjugation, binding to a specific receptor such as glycophorin A, sortase-mediated transpeptidation (sortagging) , or passive adsorption.
  • the agent is conjugated to at least one membrane protein of the RBC by a sortase-mediated reaction.
  • the sortase-mediated reaction is a sortase-mediated glycine conjugation and/or a sortase-mediated lysine side chain ⁇ -amino group conjugation.
  • the sortase-mediated glycine conjugation and/or the sortase-mediated lysine side chain ⁇ -amino group conjugation occur at least on glycine (n) and/or lysine ⁇ -amino group at internal sites of the extracellular domain of the at least one endogenous, non-engineered membrane protein, optionally n being 1 or 2.
  • the RBC is a natural RBC, such as a natural human RBC
  • the membrane protein is an endogenous, non-engineered membrane protein.
  • the RBC has not been genetically engineered to express a protein comprising a sortase recognition motif or a nucleophilic acceptor sequence, and preferably the RBC is a natural RBC such as a natural human RBC.
  • the RBC is a genetically engineered RBC
  • the membrane protein of claim 3 is a genetically engineered membrane protein.
  • the RBC has been genetically engineered to express a protein comprising a sortase recognition motif or a nucleophilic acceptor sequence, and preferably the RBC is a genetically engineered human RBC.
  • the sortase is a Sortase A (SrtA) such as a Staphylococcus aureus transpeptidase A variant (mgSrtA) .
  • the mgSrtA comprises an amino acid sequence having at least 60%identity, such as at least 60%, 70%, 80%, 90%, 95%and 100%identity, to the amino acid sequence as set forth in SEQ ID NO: 3.
  • the agent before being linked to the RBC, comprises a sortase recognition motif on its C-terminus.
  • the sortase recognition motif comprises an amino acid sequence selecting from a group consisting of LPXTG (SEQ ID NO: 8) , LPXAG (SEQ ID NO: 9) , LPXSG (SEQ ID NO: 10) , LPXLG (SEQ ID NO: 11) , LPXVG (SEQ ID NO: 12) , LGXTG (SEQ ID NO: 13) , LAXTG (SEQ ID NO: 14) , LSXTG (SEQ ID NO: 15) , NPXTG (SEQ ID NO: 16) , MPXTG (SEQ ID NO: 17) , IPXTG (SEQ ID NO: 18) , SPXTG (SEQ ID NO: 19) , VPXTG (SEQ ID NO: 20) , YPXRG (SEQ ID NO: 21) , LPXTS (SEQ ID NO: 22) and LPXTA (SEQ ID NO:23) , wherein X is any amino acid.
  • the at least one membrane protein on the surface of the RBC comprises a structure of A1-LPXT-P1, in which LPXT is linked to a glycine (n) in P1, and/or a structure of A1-LPXT-P2, in which LPXT is linked to the side chain ⁇ -amino group of lysine in P2, wherein optionally n is 1 or 2,
  • A1 represents the agent
  • P1 and P2 independently represent the extracellular domain of the at least one endogenous, non-engineered membrane protein
  • X represents any amino acids.
  • the sortase has been further modified to enhance its stabilization in circulation and/or reduce its immunogenicity.
  • the sortase has been PEGylated and/or linked to an Fc fragment.
  • the agent comprises a protein, a peptide, an antibody or its functional antibody fragment, an antigen or epitope, a MHC-peptide complex, a drug such as a small molecule drug, an enzyme, a hormone, a cytokine, a growth factor, an antimicrobial agent, a probe, a ligand, a receptor, an immunotolerance-inducing peptide, a targeting moiety or any combination thereof.
  • a drug such as a small molecule drug, an enzyme, a hormone, a cytokine, a growth factor, an antimicrobial agent, a probe, a ligand, a receptor, an immunotolerance-inducing peptide, a targeting moiety or any combination thereof.
  • the agent comprises a urate oxidase (UOX) , such as a human UOX.
  • UOX urate oxidase
  • the agent, before being linked to the RBC comprises the amino acid sequence of SEQ ID NO: 1.
  • the agent, before being linked to the RBC comprises an amino acid sequence that is at least 80%, such as at least, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%or 100%identical to SEQ ID NO: 1.
  • the immunosuppressor is selected from the group consisting of cyclophosphamide (CTX) , methotrexate (MTX) , and dexamethasone (DEX) , preferably CTX,
  • the combination is for use in a method of inducing immune tolerance to the agent in a subject.
  • Also provide is a method of inducing immune tolerance to an agent comprising administering to a subject (a) the agent conjugated to an RBC and (b) an immunosuppressor to thereby induce the immune tolerance to the agent in the subject.
  • the agent conjugated to the RBC and the immunosuppressor are administered to the subject within 7 days, such as within 6, 5, 4, 3 or 2 days.
  • the agent conjugated to the RBC and the immunosuppressor are administered to the subject on the same day.
  • the method further comprises administering to the subject the agent conjugated to the RBC one or more additional times.
  • the agent conjugated to the RBC are administered to the subject one or more additional times after the administration of the immunosuppressor, such as about 7-300 days after the administration of the immunosuppressor, e.g., at least 7, 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, 200, 210, 220, 230, 240, 250, 260, 270, 280, 290, 300 or more days after the administration of the immunosuppressor.
  • the immunosuppressor such as about 7-300 days after the administration of the immunosuppressor, e.g., at least 7, 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, 200, 210, 220, 230, 240, 250, 260, 270, 280, 290, 300 or more days after the administration of the immunosuppressor.
  • the method further comprises administering to the subject the immunosuppressor one or more additional times after the initial administration of the immunosuppressor, such as about 30, 40, 50, 60, 70, 80, 90, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, 200, 210, 220, 230, 240, 250, 260, 270, 280, 290, 300 or more days after the initial administration of the immunosuppressor.
  • additional times such as about 30, 40, 50, 60, 70, 80, 90, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, 200, 210, 220, 230, 240, 250, 260, 270, 280, 290, 300 or more days after the initial administration of the immunosuppressor.
  • the application describes a method of inducing an immune tolerance to a urate oxidase in a subject, the method comprising administering to a subject (a) an effective amount of the urate oxidase conjugated to an RBC and (b) an effective amount of an immunosuppressor selected from the group consisting of cyclophosphamide (CTX) , methotrexate (MTX) , and dexamethasone (DEX) , preferably CTX, to thereby induce the immune tolerance to the urate oxidase in the subject, preferably, the immune tolerance is specific to the urate oxidase in the subject.
  • CTX cyclophosphamide
  • MTX methotrexate
  • DEX dexamethasone
  • the immune tolerance lasts at least 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, 200, 210, 220, 230, 240, 250, 260, 270, 280, 290, 300 or more days after the administration of the immunosuppressor.
  • the RBCs are isolated from autologous blood. In other embodiments, the RBCs are isolated from matching donor blood.
  • the subject is in need of a treatment for a disease.
  • the disease is gout.
  • FIG. 1 shows inhibition of anti-UOX IgG antibody in rats treated with recombinant urate oxidase conjugated to red blood cells (UOX-RBCs) alone or in combination with cyclophosphamide (CTX) , methotrexate (MTX) , or dexamethasone (DEX) at D0 and challenged with UOX-RBCs (2e10 RBCs/kg) alone at D7.
  • UOX-RBCs red blood cells
  • CTX cyclophosphamide
  • MTX methotrexate
  • DEX dexamethasone
  • FIG. 2 shows inhibition of anti-UOX IgG antibodies in rats treated with UOX-RBCs alone, CTX alone, or UOX-RBCs in combination with CTX at day 0 and then repeatedly challenged with UOX-RBCs alone at day 7, day 28, day 56, day 77, day 98, day 119, day 140, day 161, day 182 and day 203.
  • FIG. 3 shows inhibition of anti-UOX IgG antibodies in mice treated with UOX-RBCs alone, CTX alone, or UOX-RBCs in combination with CTX at day 0 and then challenged with UOX-RBCs alone at day 21.
  • FIG. 4 shows inhibition of anti-ovalbumin (OVA) antibodies in mice treated with OVA-RBCs alone or a combination of OVA-RBC and an immunosuppressor (IS; CTX) at day 0 and then challenged with OVA-RBCs alone at days 7 and 21.
  • OVA anti-ovalbumin
  • FIG. 5 shows inhibition of anti-UOX antibodies in mice treated with an immunosuppressor (IS; CTX) only, recombinant UOX proteins and immunosuppressor, or a UOX-RBCs and IS at day 0 and then challenged with UOX proteins at day 7, day 14, and day 21.
  • IS immunosuppressor
  • FIG. 6 shows inhibition of anti-UOX and anti-OVA antibodies in mice treated with UOX-RBCs in combination with immunosuppressor or OVA-RBCs in combination with immunosuppressor or immunosuppressor alone at day 0 and then challenged with UOX-RBC or OVA-RBCs at day 7.
  • FIG. 7 shows inhibition of anti-OVA antibodies in splenectomized or sham-surgery-treated mice administered immunosuppressor (IS; CTX) alone or a combination of IS and OVA-RBCs on day 0 and then challenged with OVA proteins only at days 7, 14 and 21.
  • IS immunosuppressor
  • FIG. 8 shows inhibition of anti-UOX antibodies in mice transfused with splenocytes from mice treated with UOX-RBC in combination with CTX or CTX only, then challenged with UOX-RBCs alone.
  • FIG. 9 shows quantification of FOXP3+CD4+ T cells from mice treated with UOX-RBC in combination with CTX or CTX only and then challenged with UOX-RBCs alone on day 7.
  • FIGs. 10A -10D illustrate that distinct splenic effector Tregs were expanded in the mice treated with UOX-RBCs plus CTX:
  • A Schematic of the experiment schedule;
  • B Frequency of effector Tregs of total splenic Tregs assessed by scRNA-seq;
  • D Typical markers that characterized central Tregs and effector Tregs by scRNA-seq.
  • FIG. 11 shows the pharmacokinetics of UOX-RBCs in Wistar rats administered with rat UOX-RBCs (2e10 RBCs/kg) in combination with CTX (72 mg/kg) at days 0 and 21, or equivalent UOX proteins in combination with CTX (72 mg/kg) administrated daily.
  • FIG. 12 shows pharmacodynamics of UOX-RBCs in Wistar rats administered UOX-RBCs in combination with CTX or UOX-RBCs alone.
  • nucleic acids are written left to right in 5' to 3' orientation; amino acid sequences are written left to right in amino to carboxy orientation, respectively. It is to be understood that this invention is not limited to the particular methodology, protocols, and reagents described, as these may vary, depending upon the context they are used by those of skills in the art.
  • the term “consisting essentially of” in the context of an amino acid sequence is meant the recited amino acid sequence together with additional one, two, three, four or five amino acids at the N-or C-terminus.
  • the terms “patient” , “individual” and “subject” are used in the context of any mammalian recipient of a treatment or combination disclosed herein. Accordingly, the methods and combination disclosed herein may have medical and/or veterinary applications. In a preferred form, the mammal is a human.
  • sequence identity is meant to include the number of exact nucleotide or amino acid matches having regard to an appropriate alignment using a standard algorithm, having regard to the extent that sequences are identical over a window of comparison.
  • a “percentage of sequence identity” is calculated by comparing two optimally aligned sequences over the window of comparison, determining the number of positions at which the identical nucleic acid base (e.g., A, T, C, G) occurs in both sequences to yield the number of matched positions, dividing the number of matched positions by the total number of positions in the window of comparison (i.e., the window size) , and multiplying the result by 100 to yield the percentage of sequence identity.
  • sequence identity may be understood to mean the “match percentage” calculated by the DNASIS computer program (Version 2.5 for windows; available from Hitachi Software engineering Co., Ltd., South San Francisco, California, USA) .
  • Red blood cells (RBCs)
  • the application relates to a combination comprising (a) an agent coupled to a red blood cell (RBC) and (b) an immunosuppressor.
  • RBC red blood cell
  • the agent can be coupled to the RBC by any means, such as encapsulation within the RBC or surface coupling, and either ex vivo or in vivo. Any suitable method can be used to couple an agent to an RBC in view of the present disclosure. See, e.g., Glassman et al., Pharmaceutics. 2020 May; 12 (5) : 440 and the references therein, which are incorporated herein by reference in their entirety.
  • the present disclosure provides a combination comprising an agent conjugated to a red blood cell (RBC) and (b) an immunosuppressor.
  • RBC red blood cell
  • the agent can be conjugated to the RBC by chemical or biological means.
  • the conjugate can be direct or indirect conjugates.
  • Direct conjugates encompass those in which the agent and the RBC are joined by direct covalent chemical linkages.
  • Indirect conjugates encompass those in which the agent and the RBC are joined via an intermediary complex involving a biological molecule.
  • the agent can be conjugated to the RBC using any suitable method, such as by chemical conjugation, binding to a specific receptor such as glycophorin A, sortase-mediated transpeptidation (sortagging) , or passive adsorption. See, e.g., the methods described in US8148321, US9862779, US8940501, US10471099, US20180334661, WO2020/089485, CN106191015B, US9267127, CN109797194A, the content of each of which is incorporated herein by reference in its entirety.
  • the agent is conjugated to at least one endogenous, non-engineered membrane protein of the RBC by a sortase-mediated reaction. In some embodiments, the agent is conjugated to at least one endogenous, non-engineered membrane protein through a sortase-mediated glycine conjugation and/or a sortase-mediated lysine side chain ⁇ -amino conjugation.
  • the sortase-mediated glycine conjugation and/or the sortase-mediated lysine side chain ⁇ -amino group conjugation occur at least on glycine (n) and/or lysine ⁇ -amino group in the extracellular domain (for example at internal sites of the extracellular domain) of the at least one endogenous, non-engineered membrane protein, preferably n being 1 or 2.
  • the sortase-mediated lysine side chain ⁇ -amino group conjugation occurs at ⁇ -amino group of terminal lysine or internal lysine of the extracellular domain.
  • the sortase-mediated glycine conjugation and/or the sortase-mediated lysine side chain ⁇ -amino group conjugation may occur at glycine (n) and/or lysine ⁇ -amino group at terminal (e.g., N-terminal) and/or internal sites of the extracellular domain of at least one endogenous, non-engineered membrane protein, preferably n being 1 or 2.
  • red blood cell refers to a red blood cell (RBC)
  • RBC red blood cell
  • the RBC is a human RBC, such as a human natural RBC.
  • the RBC is a red blood cell that has not been genetically engineered to express a protein comprising a sortase recognition motif or a nucleophilic acceptor sequence. In some embodiments the RBC has not been genetically engineered. Unless otherwise indicated or clearly evident from the context, where the present disclosure refers to sortagging red blood cells it is generally intended to mean red blood cells that have not been genetically engineered for sortagging. In certain embodiments the red blood cells are not genetically engineered.
  • a red blood cell is considered “not genetically engineered for sortagging” if the cell has not been genetically engineered to express a protein comprising a sortase recognition motif or a nucleophilic acceptor sequence in a sortase-catalyzed reaction.
  • the combination comprises a plurality of red blood cells with an agent conjugated thereto.
  • at least a selected percentage of the cells in the combination are modified, i.e., having an agent conjugated thereto by sortase. For example, in some embodiments at least 5%, 10%, 15%, 20%, 25%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 96%, 97%, 98%, 99%, or more of the cells have an agent conjugated thereto.
  • the conjugated agent may be one or more of the agents described herein.
  • two, three, four, five or more different endogenous non-engineered polypeptides expressed by the cell have an agent conjugated thereto via a sortase-mediated reaction.
  • the agents attached to different polypeptides may be the same or the cell may be sortagged with a plurality of different agents.
  • the agent is linked via a sortase recognition motif to the at least one endogenous, non-engineered membrane protein.
  • the sortase recognition motif may be selected from a group consisting of LPXTG, LPXAG, LPXSG, LPXLG, LPXVG, LGXTG, LAXTG, LSXTG, NPXTG, MPXTG, IPXTG, SPXTG, VPXTG, YPXRG, LPXTS and LPXTA, wherein X is any amino acid.
  • the agent linked to the at least one endogenous, non-engineered membrane protein comprises A1-L1-P1, in which L1 is linked to a glycine (n) in P1, and/or a structure of A1-L1-P2, in which L1 is linked to the side chain ⁇ -amino group of lysine in P2, wherein n is preferably 1 or 2;
  • L1 is selected from the group consisting of LPXT, LPXA, LPXS, LPXL, LPXV, LGXT, LAXT, LSXT, NPXT, MPXT, IPXT, SPXT, VPXT, YPXR, LPXT and LPXT;
  • A1 represents the agent;
  • P1 and P2 independently represent the at least one endogenous, non-engineered membrane protein; and
  • X represents any amino acids.
  • the agent conjugated to the at least one endogenous, non-engineered membrane protein comprises A1-LPXT-P1, in which LPXT is linked to a glycine (n) in P1, and/or a structure of A1-LPXT-P2, in which LPXT is linked to the side chain ⁇ -amino group of lysine in P2, wherein n is preferably 1 or 2,
  • A1 represents the agent
  • P1 and P2 independently represent the at least one endogenous, non-engineered membrane protein
  • X represents any amino acids.
  • P1 and P2 may be the same or different.
  • the agent is linked to one or more (e.g., two, three, four, five or more) glycine (n) or lysine side chain ⁇ -amino groups in or within an extracellular domain of the at least one endogenous, non-engineered membrane protein.
  • one or more e.g., two, three, four, five or more
  • glycine (n) or lysine side chain ⁇ -amino groups in or within an extracellular domain of the at least one endogenous, non-engineered membrane protein.
  • genetically engineered red blood cells are modified by using sortase to attach a sortase substrate to a non-genetically engineered endogenous polypeptide of the cell.
  • the red blood cell may, for example, have been genetically engineered to express any of a wide variety of products, e.g., polypeptides or noncoding RNAs, may be genetically engineered to have a deletion of at least a portion of one or more genes, and/or may be genetically engineered to have one or more precise alterations in the sequence of one or more endogenous genes.
  • a non-engineered endogenous polypeptide of such genetically engineered cell is sortagged with any of the various agents described herein.
  • the present disclosure contemplates using autologous red blood cells that are isolated from an individual to whom such isolated red blood cells, after modified in vitro, are to be administered.
  • the present disclosure contemplates using immuno-compatible red blood cells that are of the same blood group as an individual to whom such cells are to be administered (e.g., at least with respect to the ABO blood type system and, in some embodiments, with respect to the D blood group system) or may be of a compatible blood group.
  • non-engineered, “non-genetically modified” and “non-recombinant” as used herein are interchangeable and refer to not being genetically engineered, absence of genetic modification, etc.
  • Non-engineered membrane proteins encompass endogenous proteins.
  • a non-genetically engineered red blood cell does not contain a non-endogenous nucleic acid, e.g., DNA or RNA that originates from a vector, from a different species, or that comprises an artificial sequence, e.g., DNA or RNA that was introduced artificially.
  • a non-engineered cell has not been intentionally contacted with a nucleic acid that is capable of causing a heritable genetic alteration under conditions suitable for uptake of the nucleic acid by the cells.
  • the RBCs suitable for use in the compositions and methods of the application can be autologous or obtained from a matching donor.
  • the RBCs can be isolated from the blood of a subject or a matching donor, optionally genetically engineered, then conjugated to an agent, such as the UOX.
  • Any suitable method for RBC isolation and/or genetic engineering can be used in view of the present disclosure. See, e.g., Glassman et al., Pharmaceutics. 2020 May; 12 (5) : 440 and the references therein, which are incorporated herein by reference in their entirety.
  • Sortases Enzymes identified as “sortases” have been isolated from a variety of Gram-positive bacteria. Sortases, sortase-mediated transacylation reactions, and their use in protein engineering are well known to those of ordinary skills in the art (see, e.g., PCT/US2010/000274 (WO/2010/087994) , and PCT/US2011/033303 (WO/2011/133704) ) .
  • Sortases have been classified into 4 classes, designated A, B, C, and D, based on sequence alignment and phylogenetic analysis of 61 sortases from Gram-positive bacterial genomes (Dramsi S, Trieu-Cuot P, Bierne H, Sorting sortases: a nomenclature proposal for the various sortases of Gram-positive bacteria. Res Microbiol. 156 (3) : 289-97, 2005) . Those skilled in the art can readily assign a sortase to the correct class based on its sequence and/or other characteristics such as those described in Drami, et al., supra.
  • sortase A refers to a class A sortase, usually named SrtA in any particular bacterial species, e.g., SrtA from S. aureus or S. pyogenes.
  • sortase also known as transamidases refers to an enzyme that has transamidase activity. Sortases recognize substrates comprising a sortase recognition motif, e.g., the amino acid sequence LPXTG (SEQ ID NO: 8) . A molecule recognized by a sortase (i.e., comprising a sortase recognition motif) is sometimes termed a “sortase substrate” herein. Sortases tolerate a wide variety of moieties in proximity to the cleavage site, thus allowing for the versatile conjugation of diverse entities so long as the substrate contains a suitably exposed sortase recognition motif and a suitable nucleophile is available.
  • a sortase recognition motif e.g., the amino acid sequence LPXTG (SEQ ID NO: 8) .
  • a molecule recognized by a sortase i.e., comprising a sortase recognition motif
  • Sortases tolerate a wide variety of moieties in proximity to the cle
  • sortase-mediated transacylation reaction “sortase-catalyzed transacylation reaction” , “sortase-mediated reaction” , “sortase-catalyzed reaction” , “sortase reaction” , “sortase-mediated transpeptide reaction” and like terms, are used interchangeably herein to refer to such a reaction.
  • sortase recognition motif “sortase recognition sequence” and “transamidase recognition sequence” with respect to sequences recognized by a transamidase or sortase, are used interchangeably herein.
  • N-terminal glycine e.g., 1, 2, 3, 4, or 5 N-terminal glycines
  • lysine side chain ⁇ -amino group e.g., 1, 2, 3, 4, or 5
  • sortase A is used, such as SrtA from S. aureus.
  • sortases may utilize different sortase recognition sequences and/or different nucleophilic acceptor sequences.
  • the sortase is a sortase A (SrtA) .
  • SrtA recognizes the motif LPXTG (SEQ ID NO: 8) , with common recognition motifs being, e.g., LPKTG, LPATG, LPNTG.
  • LPETG is used.
  • motifs falling outside this consensus may also be recognized.
  • the motif comprises an ‘A’ , ‘S’ , ‘L’ or ‘V’ rather than a ‘T’a t position 4, e.g., LPXAG, LPXSG, LPXLG or LPXVG, e.g., LPNAG or LPESG, LPELG or LPEVG.
  • the motif comprises an ‘A’ rather than a ‘G’ at position 5, e.g., LPXTA, e.g., LPNTA.
  • the motif comprises a ‘G’ or ‘A’ rather than ‘P’ at position 2, e.g., LGXTG or LAXTG, e.g., LGATG or LAETG.
  • the motif comprises an ‘I’ or ‘M’ rather than ‘L’ at position 1, e.g., MPXTG or IPXTG, e.g., MPKTG, IPKTG, IPNTG or IPETG.
  • Diverse recognition motifs of sortase A are described in Pishesha et al. 2018.
  • the sortase recognition sequence is LPXTG, wherein X is a standard or non-standard amino acid.
  • X is selected from D, E, A, N, Q, K, or R.
  • the recognition sequence is selected from LPXTG, LPXAG, LPXSG, LPXLG, LPXVG, LGXTG, LAXTG, LSXTG, NPXTG, MPXTG, IPXTG, SPXTG, VPXTG, YPXRG, LPXTS and LPXTA, wherein X may be any amino acids, such as those selected from D, E, A, N, Q, K, or R in certain embodiments.
  • the present disclosure contemplates using a variant of a naturally occurring sortase.
  • the variant is capable of mediating a glycine (n) conjugation and/or a lysine side chain ⁇ -amino group conjugation, preferably at internal sites of the extracellular domain of the at least one endogenous, non-engineered membrane protein of a red blood cell, preferably n being 1 or 2.
  • Such variants may be produced through processes such as directed evolution, site-specific modification, etc.
  • sortase enzymes e.g., sortase A enzymes
  • NMR or crystal structures of SrtA alone or bound to a sortase recognition sequence see, e.g., Zong Y, et al. J. Biol Chem. 2004, 279, 31383-31389) .
  • the active site and substrate binding pocket of S. aureus SrtA have been identified.
  • One of ordinary skills in the art can generate functional variants by, for example, avoiding deletions or substitutions that would disrupt or substantially alter the active site or substrate binding pocket of a sortase.
  • directed evolution on SrtA can be performed by utilizing the FRET (Fluorescence Resonance Energy Transfer) -based selection assay described in Chen, et al. Sci. Rep. 2016, 6 (1) , 31899.
  • a functional variant of S. aureus SrtA may be those described in CN10619105A and CN109797194A.
  • the S. aureus SrtA variant can be a truncated variant with e.g. 25-60 (e.g., 30, 35, 40, 45, 50, 55, 59 or 60) amino acids being removed from N-terminus (as compared to the wild type S. aureus SrtA) .
  • the present disclosure contemplates a S. aureus SrtA variant (mg SrtA) comprising or consisting essentially of or consisting of an amino acid sequence having at least 60% (e.g., at least 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5%, 99.9%or higher) identity to an amino acid sequence as set forth in SEQ ID NO: 3.
  • the present disclosure provides a nucleic acid encoding the S. aureus SrtA variant, and in some embodiments the nucleic acid is set forth in SEQ ID NO: 4.
  • an agent comprises a protein, a peptide (e.g., an extracellular domain of oligomeric ACE2) , an antibody or its functional antibody fragment, an antigen or epitope, a MHC-peptide complex, a drug such as a small molecule drug (e.g., an antitumor agent such as a chemotherapeutic agent) , an enzyme (e.g., a functional metabolic or therapeutic enzyme) , a hormone, a cytokine, a growth factor, an antimicrobial agent, a probe, a ligand, a receptor, an immunotolerance-inducing peptide, a targeting moiety or any combination thereof.
  • a drug such as a small molecule drug (e.g., an antitumor agent such as a chemotherapeutic agent)
  • an enzyme e.g., a functional metabolic or therapeutic enzyme
  • the agent in addition to a therapeutically active domain such as an enzyme, a drug, a small molecule (such as a small molecule drug (e.g., an antitumor agent such as a chemotherapeutic agent) , a therapeutic protein and a therapeutic antibody as described herein, the agent can further comprise a targeting moiety for targeting the cells and/or agent to a site in the body where the therapeutic activity is desired.
  • the targeting moiety binds to a target present at such a site. Any targeting moiety can be used, e.g., an antibody.
  • the site can be any organ or tissue, e.g., respiratory tract (e.g., lung) , bone, kidney, liver, pancreas, skin, cardiovascular system (e.g., heart) , smooth or skeletal muscle, gastrointestinal tract, eye, blood vessel surfaces, etc.
  • respiratory tract e.g., lung
  • bone e.g., kidney
  • liver e.g., pancreas
  • cardiovascular system e.g., heart
  • smooth or skeletal muscle e.g., smooth or skeletal muscle
  • gastrointestinal tract e.g., eye
  • blood vessel surfaces e.g., etc.
  • a protein is an enzyme such as a functional metabolic or therapeutic enzyme, e.g., an enzyme that plays a role in metabolism or other physiological processes in a mammal.
  • a protein is an enzyme that plays a role in carbohydrate metabolism, amino acid metabolism, organic acid metabolism, porphyrin metabolism, purine and/or pyrimidine metabolism.
  • Deficiencies of enzymes or other proteins can lead to a variety of diseases, e.g., diseases associated with defects in carbohydrate metabolism, amino acid metabolism, organic acid metabolism, purine or pyrimidine metabolism, and blood clotting, among others. Metabolic diseases are characterized by the lack of functional enzymes or excessive intake of metabolites.
  • the metabolites deposition in the circulation and tissues causes tissue damage.
  • the present disclosure contemplates modifying membrane proteins of RBCs with functional metabolic enzymes.
  • the enzymes targeted RBCs will uptake metabolites in plasma of patients.
  • the agent comprises urate oxidase (UOX) .
  • immunosuppressor and “immunosuppressant” are used interchangeably and refer to an immunosuppressive drug.
  • Immunosuppressive drugs can be classified into five groups: glucocorticoids, cytostatics, antibodies, drugs acting on immunophilins and other drugs.
  • the immunosuppressor is a glucocorticoid such as prednisone, dexamethasone (DEX) , or hydrocortisone.
  • the immunosuppressor is a cytostatic, such as cyclophosphamide (CTX) or methotrexate (MTX) .
  • CTX cyclophosphamide
  • MTX methotrexate
  • the immunosuppressor is selected from the group consisting of dexamethasone (DEX) , cyclophosphamide (CTX) and methotrexate (MTX) .
  • a first pharmaceutical composition comprising an agent conjugated to a red blood cell (RBC) for use with a second pharmaceutical composition comprising an immunosuppressor.
  • RBC red blood cell
  • a first composition comprises a plurality of red blood cells.
  • at least a selected percentage of the cells in a first composition are modified, i.e., having an agent conjugated thereto by sortase. For example, in some embodiments at least 5%, 10%, 15%, 20%, 25%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 96%, 97%, 98%, 99%, or more of the cells have an agent conjugated thereto.
  • two or more red blood cells or red blood cell populations conjugated with different agents are included.
  • a first composition comprises sortagged blood red cells, wherein the cells are sortagged with any agent of interest.
  • a first composition comprises an effective amount of cells, e.g., up to about 10 14 cells, e.g., about 10, 10 2 , 10 3 , 10 4 , 10 5 , 5 ⁇ 10 5 , 10 6 , 5 ⁇ 10 6 , 10 7 , 5 ⁇ 10 7 , 10 8 , 5 ⁇ 10 8 , 10 9 , 5 ⁇ 10 9 , 10 10 , 5 ⁇ 10 10 , 10 11 , 5 ⁇ 10 11 , 10 12 , 5 ⁇ 10 12 , 10 13 , 5 ⁇ 10 13 , or 10 14 cells.
  • the number of cells may range between any two of the afore-mentioned numbers.
  • an effective amount refers to an amount sufficient to achieve a biological response or effect of interest, e.g., reducing one or more symptoms or manifestations of a disease or condition or modulating an immune response.
  • a first composition administered to a subject comprises up to about 10 14 cells, e.g., about 10 3 , 10 4 , 10 5 , 10 6 , 10 7 , 10 8 , 10 9 , 10 10 , 10 11 , 10 12 , 10 13 or 10 14 cells, or any intervening number or range.
  • physiologically acceptable carrier is meant a solid or liquid filler, diluent or encapsulating substance that may be safely used in systemic administration. Depending upon the particular route of administration, a variety of carriers, diluent and excipients well known in the art may be used.
  • These can be selected from a group including sugars, starches, cellulose and its derivatives, malt, gelatine, talc, calcium sulfate, vegetable oils, synthetic oils, polyols, alginic acid, phosphate buffered solutions, emulsifiers, isotonic saline and salts such as mineral acid salts including hydrochlorides, bromides and sulfates, organic acids such as acetates, propionates and malonates, water and pyrogen-free water.
  • sugars starches, cellulose and its derivatives, malt, gelatine, talc, calcium sulfate, vegetable oils, synthetic oils, polyols, alginic acid, phosphate buffered solutions, emulsifiers, isotonic saline and salts such as mineral acid salts including hydrochlorides, bromides and sulfates, organic acids such as acetates, propionates and malonates, water and pyrogen-free
  • a combination or composition of the application is for use in a method of inducing immune tolerance to an agent.
  • immune tolerance or “immunotolerance” refers to a state of unresponsiveness of the immune system to an agent that would otherwise have the capacity to elicit an immune response in a subject.
  • a combination or composition of the application is for use in a method of reducing the immunogenicity of an agent in a subject.
  • immunogenicity refers to the ability of a foreign substance, such as an antigen, to provoke an immune response.
  • Also provided is a method of inducing immune tolerance to an agent comprising administering to the subject a combination comprising administering to a subject (a) an effective amount of the agent conjugated to an RBC and (b) an effective amount of an immunosuppressor to thereby induce the immune tolerance to the agent in the subject.
  • the induced immune tolerance is specific to the agent.
  • the agent conjugated to the RBC and the immunosuppressor can be administered to the subject in any order, e.g., the agent conjugated to the RBC can be administered before, after or at the same time with the immunosuppressor.
  • the agent conjugated to the RBC and the immunosuppressor are administered to the subject within 7 days, such as within 6, 5, 4, 3 or 2 days.
  • the agent conjugated to the RBC and the immunosuppressor are administered to the subject on the same day.
  • the subject is in need of a treatment for a disease.
  • treating refers to a therapeutic intervention that at least partly ameliorates, eliminates or reduces a symptom or pathological sign of a pathogen-associated disease, disorder or condition after it has begun to develop. Treatment need not be absolute to be beneficial to the subject. The beneficial effect can be determined using any methods or standards known to the ordinarily skilled artisan.
  • the method as described herein further comprises administering the agent conjugated red blood cells to a subject, e.g., directly into the circulatory system, e.g., intravenously, by injection or infusion, one or more times.
  • a method of delivering an agent to a subject in need thereof comprising administering the combination as described herein to the subject.
  • delivery refers to transportation of a molecule or agent to a desired cell or tissue site. Delivery can be to the cell surface, cell membrane, cell endosome, within the cell membrane, nucleus or within the nucleus, or any other desired area of the cell.
  • a subject receives a single dose of the agent conjugated red blood cells, or receives multiple doses of the agent conjugated red blood cells, e.g., between 2 and 5, 10, 20, or more doses, over a course of treatment.
  • a dose or total cell number may be expressed as cells/kg.
  • a dose may be about 10 3 , 10 4 , 10 5 , 10 6 , 10 7 , 10 8 cells/kg.
  • a course of treatment lasts for about 1 week to 12 months or more e.g., 1, 2, 3 or 4 weeks or 2, 3, 4, 5 or 6 months.
  • a subject is treated about every 2-4 weeks.
  • the number of cells, doses, and/or dosing interval may be selected based on various factors such as the weight, and/or blood volume of the subject, the condition being treated, response of the subject, etc.
  • the exact number of cells required may vary from subject to subject, depending on factors such as the species, age, weight, sex, and general condition of the subject, the severity of the disease or disorder, the particular cell (s) , the identity and activity of agent (s) conjugated to the cells, mode of administration, concurrent therapies, and the like.
  • the UOX- (GS) 3 -LPETGG used in this Example contains the amino acid sequence of SEQ ID NO: 1 shown below:
  • the nucleotide sequence encoding the UOX- (GS) 3 -LPETGG used in this Example contains the polynucleotide sequence of SEQ ID NO: 2 shown below:
  • UOX Aspergillus flavus uricase
  • GenScript GenScript
  • Subclones were generated by standard PCR procedure and inserted into the pET-30a vector with coding sequence for a C-terminal (GS) 3 (SEQ ID NO: 5) linker followed by a sortase recognition sequence (LPETGG) (SEQ ID NO: 6) . All constructs were verified by sequencing and then transformed in E. coli BL21 (DE3) for protein expression.
  • a single transformed colony was inoculated into 10 ml Luria-Bertani (LB) medium supplemented with ampicillin (100 ⁇ g/ml, Beyotime Biotechnology, Suzhou, China) , grew overnight at 37°C with 220 rpm shaking. Next day, this 10 ml culture was transferred to 1 L fresh LB medium and grew at 37°C with 220 rpm shaking until OD600 reached 0.6. The temperature of the culture was then lowered to 20°C and 1 mM IPTG (Sigma, St. Louis, MO) was added for induction.
  • LB Luria-Bertani
  • a cell pellet was collected by centrifugation, resuspended in low salt lysis buffer (50 mM Tris 8.8, 50 mM NaCl) and then lysed with sonication.
  • a supernatant containing the recombinant protein UOX- (GS) 3 -LPETGG was collected by centrifugation at 10,000 rpm for 1 h, and loaded on a Q Sepharose FF column (Cytiva, Marlborough, USA) pre-equilibrated with buffer A (20 mM Tris 8.8) .
  • the column was washed with the buffer A until the absorbance at 280 nm and conductivity became stable and then eluted with a linear gradient of 0- 1 M NaCl in 20 mM Tris 8.8. Fractions corresponding to the elution peak were analyzed by SDS-PAGE and the purest fractions were pooled. The pooled elution was diluted by a buffer (20mM Tris8.0) , then loaded on a Diamond MixA column (Bestchrom, Shanghai, China) and eluted with a linear gradient of 0-1 M NaCl in 20 mM Tris 8.0. Fractions corresponding to the elution peak were analyzed by SDS-PAGE and the purest fractions were pooled.
  • the elution sample was loaded on a UniHR Phenyl-80L column (NanMicr) , and washed with 60%gradient buffer B (20 mM Tris 7.5) , then eluted with 100%buffer B (20mM Tris7.5) . Concentration of the elution was detected with Amicon Ultra-15 Centrifugal Filter Unit (Millipore, Darmstadt, Germany) .
  • Red blood cells were separated from peripheral blood of C57BL/6J mice and rats (jh-lab animal) by density gradient centrifugation, respectively. The separated red blood cells were washed with PBS for 3 times. Then the RBCs were pretreated with 2.5 mM Tris (2-carboxyethyl) phosphine hydrochloride (TCEP) (Sigma) for 1 hr at room temperature. The pretreated RBCs were washed with PBS for 3 times.
  • TCEP Tris (2-carboxyethyl) phosphine hydrochloride
  • G peptide-mal (6-maleimidohexanoic acid) -K (GGG) -GGG-K (GGG) -GGG-K (GGG) -NH2 (SEQ ID NO: 7) , also called G peptide-mal was synthesized with more than 99%purity (Beijing Scilight Biotechnology Led. Co. ) .
  • G peptide-mal was dissolved in phosphate buffer to a final concentration of 10 mg/mL. Then, 1 ⁇ 10 9 RBCs were contacted with 0.17mg/mL G peptide-mal for 30 mins at 30°C to obtain RBCs modified with G peptide-mal, named as G peptide-mal-RBCs.
  • the obtained G peptide-mal-RBCs were then washed with PBS for 3 times.
  • the G peptide-mal-RBCs were either used immediately or stored at 4°C for further use.
  • G peptide-mal-RBCs (1 ⁇ 10 9 /mL) were conjugated with UOX- (GS) 3 -LPETGG via a sortase (mg SrtA) catalyzed conjugation reaction.
  • concentration of mg SrtA was 10 ⁇ M and the UOX- (GS) 3-LPETGG substrate was in the range of 25 ⁇ M-100 ⁇ M.
  • UOX- (GS) 3-LPET-G peptide-mal-RBCs also described as UOX-RBCs below
  • the mg SrtA used in the conjugation reaction of this Example contains the amino acid sequence of SEQ ID NO: 3 below:
  • the nucleotide sequence encoding the used mg SrtA contains the nucleotide sequence of SEQ ID NO: 4 below:
  • OVA-RBCs were prepared by SMCC (succinimidyl-trans-4- (N-maleimidylmethyl) cyclohexane-1-carboxylate) labeling.
  • SMCC succinimidyl-trans-4- (N-maleimidylmethyl) cyclohexane-1-carboxylate
  • NHS N-hydroxysuccinimide
  • maleimide groups that allow covalent conjugation of amine-and sulfhydryl-containing molecules.
  • NHS esters react with primary amines to form amide bonds
  • maleimides react with sulfhydryl groups to form stable thioether bonds.
  • Red blood cells were separated from peripheral blood of C57BL/6J mice and rats (jh-lab animal) by density gradient centrifugation, respectively.
  • the separated red blood cells were washed with PBS for 3 times. Then the RBCs were pretreated with 2.5 mM TCEP for 1 hr at room temperature to expose sulfhydryl and then washed with PBS for 3 times. Amine containing OVA protein (InvivoGen) was react with SMCC NHS ester for 1h at RT and ultrafiltration at 4200g, 20 min for 3 times to remove remnant SMCC and obtain OVA-SMCC. Finally, sulfhydryl exposed RBCs were conjugated with OVA-SMCC, the final products, OVA-RBCs, were stored at 2–8 °C.
  • UOX-RBCs The capacity of UOX-RBCs to induce immune tolerance with different immunosuppressors was evaluated in rats.
  • the Wistar rats (Vital River) were randomly divided into 4 groups each contained 3 rats.
  • the 4 groups were respectively treated with, (1) UOX-RBCs (2e10 RBCs/kg) only, (2) UOX-RBCs (2e10 RBCs/kg) in combination with cyclophosphamide (CTX, 72 mg/kg, BAXTER) , (3) UOX-RBCs (2e10 RBCs/kg) in combination with methotrexate (MTX, 50 mg/kg, Zhengqing company) , (4) UOX-RBCs (2e10 RBCs/kg) in combination with dexamethasone (DEX, 3 mg/kg, cspcbaike) .
  • DEX dexamethasone
  • the Wistar rats were administrated intravenously with UOX-RBCs in combination with CTX, or MTX, or DEX at day 0 (D0) (tolerance induction) . Then, these rats were challenged with UOX-RBCs (2e10 RBCs/kg) alone at day 7 (D7) .
  • the anti UOX immunogenicity was evaluated using plasma collected at D0, D7, D14 and D21.
  • Anti-UOX IgG antibodies were measured by indirect ELISA using the coating of the antigen (UOX proteins) at 0.5 ⁇ g/mL.
  • HRP-conjugated goat anti-rat IgG (Abcam) or HRP-conjugated goat anti-mouse IgG (Abcam) was used to detect IgG antibodies, respectively, in rats samples.
  • TMB Solarbio
  • the optical density (OD) of the wells was read immediately after adding an H2SO4 stop solution (Solarbio) at a wavelength 450 nm. Data analysis was performed using GraphPad Prism 8.
  • a four-parameter logistic curve-fit graph was prepared with the dilution on the x axis (log scale) and the OD value on the y axis (linear scale) , and the half maximum value (EC50) for each sample was determined.
  • Fig. 1 The results (Fig. 1) showed that rats treated with UOX-RBCs in combination with MTX or DEX had an attenuated immune response to UOX, although the mean titers increased significantly at D14 (DEX) and D21 (MTX) .
  • the rats treated with UOX-RBCs in combination with CTX showed no detectable antibody titers against UOX up to D21. Therefore, the capacity of an immunosuppressor to induce immune tolerance in combination with UOX-RBCs is CTX>MTX>DEX.
  • Example 4 A single dose of engineered RBCs in combination with CTX provided durable inhibition of specific immune responses in rodents.
  • mice were treated with OVA-RBCs (obtained in example 2) (2e10 RBCs/kg) in combination with CTX (240 mg/kg) and repeated challenged them with OVA-RBC (2e10 RBCs/kg) alone as described in Fig. 4. These animals were randomly divided into 2 groups each contained 3 mice.
  • Anti-OVA IgG antibodies were measured by indirect ELISA using the coating of the antigen (OVA proteins) at 0.5 ⁇ g/mL.
  • HRP-conjugated goat anti-mouse IgG (Abcam) was used to detect IgG antibodies, respectively, in rat samples.
  • mice were challenged with an unrelated antigen after the tolerization period. These animals were randomly divided into 5 groups each contained 2 mice as described in Fig. 6. Mice tolerized to OVA-RBC (2e10 RBCs/kg) in combination with CTX (240 mg/kg) and then challenged with UOX-RBCs (2e10 RBCs/kg) and OVA-RBCs (2e10 RBCs/kg) , respectively, showed a normal immune response to UOX but no response to OVA (Fig. 6) . Importantly, animals that received CTX only during the tolerization period showed robust immune responses to both UOX and OVA.
  • UOX protein also named UOX- (GS) 3-LPETGG, SEQ ID NO: 1
  • CTX 240 mg/kg
  • an antigen conjugated to RBC such as UOX-RBCs
  • an immunosuppressor such as CTX
  • CTX immunosuppressor
  • unconjugated agent e.g., UOX protein
  • RBCs were accumulated primarily in the spleen and liver. Specifically, antigenic peptides, attached to erythrocyte membranes, are captured in the spleen along with erythrocytes [12] .
  • CTX erythrocytes
  • splenocytes were harvested as source of donor cells. Mice then received 10 million unfractionated splenocytes from the mice treated with UOX-RBC in combination with CTX or CTX only. One day after, these mice were challenged with UOX-RBCs alone. A substantial reduction in anti UOX immunogenicity was observed in the mice that received splenocytes from tolerance induced mice, comparing with the mice received splenocytes from CTX injected mice (Fig. 8) . This suggests a role of the spleen as a site where tolerance induction is initiated.
  • FOXP3 (FOXP3-PE, biolegend) intracellular staining was performed in PBS +2%FBS.
  • Flow cytometry measurements were performed using a Cytoflex (Beckman Coulter) and data were analyzed using version 9.8.2 of FlowJo software.
  • expansion of Tregs in the spleen was assessed in the mice treated with UOX-RBCs in combination with CTX until D21 (Fig. 9) .
  • mice were treated with CTX with or without UOX-RBCs on day 0 and challenged with UOX-RBCs only on day 7, and the splenocytes were isolated for single cell transcriptome analysis on day 21 (FIG. 10A) .
  • the frequency of effector Tregs of total splenic Tregs were assessed by scRNA-seq and the results of which are shown in FIG. 10B.
  • FIG. 10D shows the typical markers that characterized central Tregs and effector Tregs by scRNA-seq.
  • UOX-RBCs The pharmacokinetics of UOX-RBCs were evaluated by detecting percentage rat UOX-RBCs in vivo through flow cytometry. Briefly, Wistar rats (Vital River) were administered i.v. with rat UOX-RBCs (2e10 RBCs/kg) in combination with CTX (72 mg/kg) , equivalent UOX proteins in combination with CTX (72 mg/kg) were administrated daily. Each group contained 4 rats.
  • Example 8 UOX-RBCs in combination with CTX allowed for repeated dosing and enabled sustained control of uric acid levels.
  • UOX-RBCs The pharmacodynamics of UOX-RBCs in combination with CTX were evaluated. Briefly, Wistar rats (Vital River) were administrated i.v. with rat UOX-RBCs (2e10 RBCs/kg) in combination with CTX (72 mg/kg) , equivalent UOX-RBCs were used as control. Each group contained 4 rats. The plasma was collected at D (-1) , D7, D14, D21, D28, D35, D42 and D49 for uric acid evaluation according to the manufacturing protocol (abcam) . The results (Fig. 12) showed that Wistars rats treated with UOX-RBC in combination with CTX inhibited the ADA response, enabling the sustained maintenance of low serum uric acid levels.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Medicinal Chemistry (AREA)
  • Epidemiology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Organic Chemistry (AREA)
  • Zoology (AREA)
  • Hematology (AREA)
  • Immunology (AREA)
  • Virology (AREA)
  • Cell Biology (AREA)
  • Genetics & Genomics (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Wood Science & Technology (AREA)
  • Biomedical Technology (AREA)
  • Biotechnology (AREA)
  • General Engineering & Computer Science (AREA)
  • Developmental Biology & Embryology (AREA)
  • Biochemistry (AREA)
  • Pain & Pain Management (AREA)
  • Rheumatology (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

A combination comprises an agent conjugated to a red blood cell (RBC) and an immunosuppressor. The uses of the combination are inducing immune tolerance specific to the agent in a subject.

Description

COMBINATION TO INDUCE SPECIFIC IMMUNE TOLERANCE
CROSS-REFERENCE TO RELATED APPLICATIONS
This application claims the benefit of International Application No. PCT/CN2022/127009 filed October 24, 2022, the content of which is incorporated by reference herein in its entirety.
TECHNICAL FIELD
The present disclosure relates generally to modified red blood cells (RBCs) , and more particularly to combinations of an agent conjugated to an RBC with an immunosuppressor and uses thereof to induce immune tolerance specific to the agent.
REFERENCE TO SEQUENCE LISTING SUBMITTED ELECTRONICALLY
The contents of the electronic sequence listing (070900-2WO2_Sequence Listing. xml; Size: 22, 615 bytes; and Date of Creation: October 17, 2023) is herein incorporated by reference in its entirety.
BACKGROUND
Urate oxidase (UOX, uricase) is a liver enzyme that metabolizes uric acid (UA) into allantoin, a more water-soluble compound, which is easily excreted by the kidney. 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. Uricase 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. This agent significantly reduces serum UA levels and acts faster than allopurinol. The recommended dose is 0.2 mg/kg in children and adults. However, its biological half-life is short (only 21h) , so rasburicase is given by infusion once daily for ≤ 7 days. In addition, recent studies  have shown that repeated UOX injections could cause anaphylactic reactions with the production of antibodies that neutralize UOX enzyme activity [1-6] .
Available recombinant UOX (rasburicase, pegloticase) drugs are potent hypouricemic agents for gout. However, there are several limitations for the current therapy. For example, UOX has significant immunogenicity and it may induce severe allergic reactions. Conjugating therapeutic enzymes to polyethylene glycol (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. In addition, the therapeutic enzymes may become inactivated or eliminated in vivo due to short half-life, limited bioavailability, and/or interactions with plasma proteins. Furthermore, production and purification of the enzymes tend to be time-consuming, and treatments with enzyme replacement therapy are very costly. Therefore, there is a need for new gout therapy that is more efficacious and safer.
Red blood cells (RBCs) , the most common cell type in the human body, have been widely investigated as an in vivo drug delivery system for over three decades due to their unique biological properties, including, for example, (i) widespread circulation range throughout the body; (ii) good biocompatibility as a biological material with long in vivo survival time; (iii) large surface to volume ratio; and (iv) no nucleus, mitochondria, and other cellular organelles. RBCs have been reported as an interesting tool for antigen delivery to induce specific immune tolerance (see, e.g., Cremel et al., Int J Pharm 2013 Feb 25; 443 (1-2) : 39-49) . Aged and damaged RBCs are naturally removed daily in the liver and spleen where phagocytes such as macrophages and monocytes can process and present major histocompatibility complex (MHC) -associated antigens to the T cells. Previous studies have shown the capacity of RBCs to induce specific immune tolerance to multiple sclerosis, type 1 diabetes and celiac disease, which are all T-cell driven autoimmune diseases with well-defined antigens [7-8] . However, the capacity of RBC alone to induce immune tolerance is limited.
BRIEF SUMMARY
It is now discovered that an agent delivered by an RBC in combination with an immunosuppressor induces durable immune tolerance that enables safer and more efficacious treatment of a disease with the agent.
A general aspect of the application relates to a combination comprising (a) an agent coupled to a red blood cell (RBC) and (b) an immunosuppressor. The agent can be coupled to the RBC by any means, such as encapsulation within the RBC or surface coupling, and either ex vivo or in vivo.
In one general aspect, provided is a combination comprising (a) an agent conjugated to a red blood cell (RBC) and (b) an immunosuppressor. The agent can be conjugated to the RBC by any means, such as chemical conjugation, binding to a specific receptor such as glycophorin A, sortase-mediated transpeptidation (sortagging) , or passive adsorption.
In certain embodiments, the agent is conjugated to at least one membrane protein of the RBC by a sortase-mediated reaction. In certain embodiments, the sortase-mediated reaction is a sortase-mediated glycine conjugation and/or a sortase-mediated lysine side chain ε-amino group conjugation.
In certain embodiments, the sortase-mediated glycine conjugation and/or the sortase-mediated lysine side chain ε-amino group conjugation occur at least on glycine (n) and/or lysine ε-amino group at internal sites of the extracellular domain of the at least one endogenous, non-engineered membrane protein, optionally n being 1 or 2.
In certain embodiments, the RBC is a natural RBC, such as a natural human RBC, and the membrane protein is an endogenous, non-engineered membrane protein. In particular, the RBC has not been genetically engineered to express a protein comprising a sortase recognition motif or a nucleophilic acceptor sequence, and preferably the RBC is a natural RBC such as a natural human RBC.
In certain embodiments, the RBC is a genetically engineered RBC, and the membrane protein of claim 3 is a genetically engineered membrane protein. In particular, the RBC has been genetically engineered to express a protein comprising a sortase recognition motif or a nucleophilic acceptor sequence, and preferably the RBC is a genetically engineered human RBC.
In certain embodiments, the sortase is a Sortase A (SrtA) such as a Staphylococcus aureus transpeptidase A variant (mgSrtA) . In certain embodiments, the mgSrtA comprises an amino acid sequence having at least 60%identity, such as at least 60%, 70%, 80%, 90%, 95%and 100%identity, to the amino acid sequence as set forth in SEQ ID NO: 3.
In certain embodiments, the agent, before being linked to the RBC, comprises a sortase recognition motif on its C-terminus.
In certain embodiments, the sortase recognition motif comprises an amino acid sequence selecting from a group consisting of LPXTG (SEQ ID NO: 8) , LPXAG (SEQ ID NO: 9) , LPXSG (SEQ ID NO: 10) , LPXLG (SEQ ID NO: 11) , LPXVG (SEQ ID NO: 12) , LGXTG (SEQ ID NO: 13) , LAXTG (SEQ ID NO: 14) , LSXTG (SEQ ID NO: 15) , NPXTG (SEQ ID NO: 16) , MPXTG (SEQ ID NO: 17) , IPXTG (SEQ ID NO: 18) , SPXTG (SEQ ID NO: 19) , VPXTG (SEQ ID NO: 20) , YPXRG (SEQ ID NO: 21) , LPXTS (SEQ ID NO: 22) and LPXTA (SEQ ID NO:23) , wherein X is any amino acid.
In certain embodiments, the at least one membrane protein on the surface of the RBC comprises a structure of A1-LPXT-P1, in which LPXT is linked to a glycine (n) in P1, and/or a structure of A1-LPXT-P2, in which LPXT is linked to the side chain ε-amino group of lysine in P2, wherein optionally n is 1 or 2, A1 represents the agent, P1 and P2 independently represent the extracellular domain of the at least one endogenous, non-engineered membrane protein, and X represents any amino acids.
In certain embodiments, the sortase has been further modified to enhance its stabilization in circulation and/or reduce its immunogenicity.
In certain embodiments, the sortase has been PEGylated and/or linked to an Fc fragment.
In certain embodiments, the agent comprises a protein, a peptide, an antibody or its functional antibody fragment, an antigen or epitope, a MHC-peptide complex, a drug such as a small molecule drug, an enzyme, a hormone, a cytokine, a growth factor, an antimicrobial agent, a probe, a ligand, a receptor, an immunotolerance-inducing peptide, a targeting moiety or any combination thereof.
In certain embodiments, the agent comprises a urate oxidase (UOX) , such as a human UOX. In certain embodiments, the agent, before being linked to the RBC, comprises the amino acid sequence of SEQ ID NO: 1. In certain embodiments, the agent, before being linked to the RBC, comprises an amino acid sequence that is at least 80%, such as at least, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%or 100%identical to SEQ ID NO: 1. In certain embodiments, the immunosuppressor is selected from the group consisting of cyclophosphamide (CTX) , methotrexate (MTX) , and dexamethasone (DEX) , preferably CTX,
In certain embodiments, the combination is for use in a method of inducing immune tolerance to the agent in a subject.
Also provide is a method of inducing immune tolerance to an agent, the method comprising administering to a subject (a) the agent conjugated to an RBC and (b) an immunosuppressor to thereby induce the immune tolerance to the agent in the subject. In certain embodiments, the agent conjugated to the RBC and the immunosuppressor are administered to the subject within 7 days, such as within 6, 5, 4, 3 or 2 days. Preferably the agent conjugated to the RBC and the immunosuppressor are administered to the subject on the same day. In certain embodiments, the method further comprises administering to the subject the agent conjugated to the RBC one or more additional times. Preferably, the agent conjugated to the RBC are administered to the subject one or more additional times after the administration of the immunosuppressor, such as about 7-300 days after the administration of the immunosuppressor, e.g., at least 7, 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, 200, 210, 220, 230, 240, 250, 260, 270, 280, 290, 300 or more days after the administration of the immunosuppressor. In certain embodiments, the method further comprises administering to the subject the immunosuppressor one or more additional times after the initial administration of the immunosuppressor, such as about 30, 40, 50, 60, 70, 80, 90, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, 200, 210, 220, 230, 240, 250, 260, 270, 280, 290, 300 or more days after the initial administration of the immunosuppressor.
In certain embodiment, the application describes a method of inducing an immune tolerance to a urate oxidase in a subject, the method comprising administering to a subject (a) an effective amount of the urate oxidase conjugated to an RBC and (b) an effective amount of an immunosuppressor selected from the group consisting of cyclophosphamide (CTX) , methotrexate (MTX) , and dexamethasone (DEX) , preferably CTX, to thereby induce the immune tolerance to the urate oxidase in the subject, preferably, the immune tolerance is specific to the urate oxidase in the subject. In certain embodiments, the immune tolerance lasts at least 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, 200, 210, 220, 230, 240, 250, 260, 270, 280, 290, 300 or more days after the administration of the immunosuppressor.
In certain embodiments, the RBCs are isolated from autologous blood. In other embodiments, the RBCs are isolated from matching donor blood.
In certain embodiments, the subject is in need of a treatment for a disease. In preferred embodiments, the disease is gout.
BRIEF DESCRIPTION OF THE DRAWINGS
The patent or application file contains at least one drawing executed in color. Copies of this patent or patent application publication with color drawings will be provided by the Office upon request and payment of the necessary fees.
In the drawings, embodiments of the present disclosure are illustrated by way of example. It is to be expressly understood that the description and drawings are only for the purpose of illustration and as an aid to understanding, and are not intended as a definition of the limits of the invention.
FIG. 1 shows inhibition of anti-UOX IgG antibody in rats treated with recombinant urate oxidase conjugated to red blood cells (UOX-RBCs) alone or in combination with cyclophosphamide (CTX) , methotrexate (MTX) , or dexamethasone (DEX) at D0 and challenged with UOX-RBCs (2e10 RBCs/kg) alone at D7.
FIG. 2 shows inhibition of anti-UOX IgG antibodies in rats treated with UOX-RBCs alone, CTX alone, or UOX-RBCs in combination with CTX at day 0 and then repeatedly challenged with UOX-RBCs alone at day 7, day 28, day 56, day 77, day 98, day 119, day 140, day 161, day 182 and day 203.
FIG. 3 shows inhibition of anti-UOX IgG antibodies in mice treated with UOX-RBCs alone, CTX alone, or UOX-RBCs in combination with CTX at day 0 and then challenged with UOX-RBCs alone at day 21.
FIG. 4 shows inhibition of anti-ovalbumin (OVA) antibodies in mice treated with OVA-RBCs alone or a combination of OVA-RBC and an immunosuppressor (IS; CTX) at day 0 and then challenged with OVA-RBCs alone at days 7 and 21.
FIG. 5 shows inhibition of anti-UOX antibodies in mice treated with an immunosuppressor (IS; CTX) only, recombinant UOX proteins and immunosuppressor, or a UOX-RBCs and IS at day 0 and then challenged with UOX proteins at day 7, day 14, and day 21.
FIG. 6 shows inhibition of anti-UOX and anti-OVA antibodies in mice treated with UOX-RBCs in combination with immunosuppressor or OVA-RBCs in combination with immunosuppressor or immunosuppressor alone at day 0 and then challenged with UOX-RBC or OVA-RBCs at day 7.
FIG. 7 shows inhibition of anti-OVA antibodies in splenectomized or sham-surgery-treated mice administered immunosuppressor (IS; CTX) alone or a combination of IS and OVA-RBCs on day 0 and then challenged with OVA proteins only at days 7, 14 and 21.
FIG. 8 shows inhibition of anti-UOX antibodies in mice transfused with splenocytes from mice treated with UOX-RBC in combination with CTX or CTX only, then challenged with UOX-RBCs alone.
FIG. 9 shows quantification of FOXP3+CD4+ T cells from mice treated with UOX-RBC in combination with CTX or CTX only and then challenged with UOX-RBCs alone on day 7.
FIGs. 10A -10D illustrate that distinct splenic effector Tregs were expanded in the mice treated with UOX-RBCs plus CTX: (A) Schematic of the experiment schedule; (B) Frequency of effector Tregs of total splenic Tregs assessed by scRNA-seq; (C) Frequency of effector Treg of total CD4 T cells (n=3 per group) assessed by flow cytometry measurements; and (D) Typical markers that characterized central Tregs and effector Tregs by scRNA-seq.
FIG. 11 shows the pharmacokinetics of UOX-RBCs in Wistar rats administered with rat UOX-RBCs (2e10 RBCs/kg) in combination with CTX (72 mg/kg) at days 0 and 21, or equivalent UOX proteins in combination with CTX (72 mg/kg) administrated daily.
FIG. 12 shows pharmacodynamics of UOX-RBCs in Wistar rats administered UOX-RBCs in combination with CTX or UOX-RBCs alone.
DETAILED DESCRIPTION
For the purposes of promoting an understanding of the principles of the present disclosure, reference will now be made to the embodiments illustrated in the drawings, and specific language will be used to describe the same. It will nevertheless be understood that no limitation of the scope of this disclosure is thereby intended.
In the present disclosure, unless otherwise specified, the scientific and technical terms used herein have the meanings as generally understood by a person skilled in the art. Although any methods and materials similar or equivalent to those described herein find use in the practice of the present invention, the preferred methods and materials are described herein. Accordingly, the terms defined herein are more fully described by reference to the Specification as a whole.
As used herein, the singular terms "a, " "an, " and "the" include the plural reference unless the context clearly indicates otherwise. Unless otherwise indicated, nucleic acids are written left to right in 5' to 3' orientation; amino acid sequences are written left to right in amino to carboxy orientation, respectively. It is to be understood that this invention is not limited to the particular methodology, protocols, and reagents described, as these may vary, depending upon the context they are used by those of skills in the art.
As used herein, the term “consisting essentially of” in the context of an amino acid sequence is meant the recited amino acid sequence together with additional one, two, three, four or five amino acids at the N-or C-terminus.
Unless the context requires otherwise, the terms “comprise” , “comprises” and “comprising” , or similar terms are intended to mean a non-exclusive inclusion, such that a recited list of elements or features does not include those stated or listed elements solely, but may include other elements or features that are not listed or stated.
As used herein, the terms “patient” , “individual” and “subject” are used in the context of any mammalian recipient of a treatment or combination disclosed herein. Accordingly, the methods and combination disclosed herein may have medical and/or veterinary applications. In a preferred form, the mammal is a human.
As used herein, the term “sequence identity” is meant to include the number of exact nucleotide or amino acid matches having regard to an appropriate alignment using a standard algorithm, having regard to the extent that sequences are identical over a window of comparison. Thus, a “percentage of sequence identity” is calculated by comparing two optimally aligned sequences over the window of comparison, determining the number of positions at which the identical nucleic acid base (e.g., A, T, C, G) occurs in both sequences to yield the number of matched positions, dividing the number of matched positions by the total number of positions in the window of comparison (i.e., the window size) , and multiplying the result by 100 to yield the percentage of sequence identity. For example, “sequence identity” may be understood to mean the “match percentage” calculated by the DNASIS computer program (Version 2.5 for windows; available from Hitachi Software engineering Co., Ltd., South San Francisco, California, USA) .
Recent studies have discovered mutant sortases with different specificities in motif recognition [9] . For instance, Ge et al. showed that an evolved SrtA variant (mg SrtA) is capable of recognizing the N-terminus of G1-modified peptide, which cannot be achieved by wt  SrtA [10] . In addition, membrane proteins with a single glycine at the N-terminus are much more abundant than those with 3×glycines. Ge et al. made an N-terminal sequence analysis of human membrane proteome with a predicted N-terminal glycine (s) . The list of 182 proteins that contain N-terminal glycine residues after enzymatic removal of the signal peptide or the initiator methionine residue according to the previous study [11] . Among them, 176 proteins (96.70%) contain a single glycine residue at the N-terminus, 4 proteins (2.20%) contain a GG residue at the N-terminus, while only 2 proteins (1.10%) contain a G (n≥3) residue at the N-terminus. None of the 182 proteins is known to be expressed on the surface of mature human red blood cells.
Red blood cells (RBCs)
The application relates to a combination comprising (a) an agent coupled to a red blood cell (RBC) and (b) an immunosuppressor. The agent can be coupled to the RBC by any means, such as encapsulation within the RBC or surface coupling, and either ex vivo or in vivo. Any suitable method can be used to couple an agent to an RBC in view of the present disclosure. See, e.g., Glassman et al., Pharmaceutics. 2020 May; 12 (5) : 440 and the references therein, which are incorporated herein by reference in their entirety.
In one general aspect, the present disclosure provides a combination comprising an agent conjugated to a red blood cell (RBC) and (b) an immunosuppressor. The agent can be conjugated to the RBC by chemical or biological means. The conjugate can be direct or indirect conjugates. Direct conjugates encompass those in which the agent and the RBC are joined by direct covalent chemical linkages. Indirect conjugates encompass those in which the agent and the RBC are joined via an intermediary complex involving a biological molecule.
The agent can be conjugated to the RBC using any suitable method, such as by chemical conjugation, binding to a specific receptor such as glycophorin A, sortase-mediated transpeptidation (sortagging) , or passive adsorption. See, e.g., the methods described in US8148321, US9862779, US8940501, US10471099, US20180334661, WO2020/089485, CN106191015B, US9267127, CN109797194A, the content of each of which is incorporated herein by reference in its entirety.
In certain embodiments, the agent is conjugated to at least one endogenous, non-engineered membrane protein of the RBC by a sortase-mediated reaction. In some embodiments, the agent is conjugated to at least one endogenous, non-engineered membrane protein through a sortase-mediated glycine conjugation and/or a sortase-mediated lysine side chain ε-amino  conjugation. In some embodiments, the sortase-mediated glycine conjugation and/or the sortase-mediated lysine side chain ε-amino group conjugation occur at least on glycine (n) and/or lysine ε-amino group in the extracellular domain (for example at internal sites of the extracellular domain) of the at least one endogenous, non-engineered membrane protein, preferably n being 1 or 2. In some embodiments, without being limited to any theory, the sortase-mediated glycine conjugation may occur at exposed glycine (n=1 or 2) of previously unreported membrane proteins due to tissue-specific mRNA splicing and protein translation during erythropoiesis. In some embodiments, the exposed glycine (n=1 or 2) may be N-terminal exposed glycine (n=1 or 2) . In some embodiments, the sortase-mediated lysine side chain ε-amino group conjugation occurs at ε-amino group of terminal lysine or internal lysine of the extracellular domain. In some embodiments, the sortase-mediated glycine conjugation and/or the sortase-mediated lysine side chain ε-amino group conjugation may occur at glycine (n) and/or lysine ε-amino group at terminal (e.g., N-terminal) and/or internal sites of the extracellular domain of at least one endogenous, non-engineered membrane protein, preferably n being 1 or 2.
Unless otherwise indicated or clearly evident from the context, where the present disclosure refers to a red blood cell (RBC) , it is generally intended to mean a mature red blood cell. In certain embodiments, the RBC is a human RBC, such as a human natural RBC.
In some embodiments, the RBC is a red blood cell that has not been genetically engineered to express a protein comprising a sortase recognition motif or a nucleophilic acceptor sequence. In some embodiments the RBC has not been genetically engineered. Unless otherwise indicated or clearly evident from the context, where the present disclosure refers to sortagging red blood cells it is generally intended to mean red blood cells that have not been genetically engineered for sortagging. In certain embodiments the red blood cells are not genetically engineered.
A red blood cell is considered “not genetically engineered for sortagging” if the cell has not been genetically engineered to express a protein comprising a sortase recognition motif or a nucleophilic acceptor sequence in a sortase-catalyzed reaction.
In some embodiments, the combination comprises a plurality of red blood cells with an agent conjugated thereto. In some embodiments, at least a selected percentage of the cells in the combination are modified, i.e., having an agent conjugated thereto by sortase. For example, in some embodiments at least 5%, 10%, 15%, 20%, 25%, 30%, 40%, 50%, 60%, 70%, 80%,  90%, 95%, 96%, 97%, 98%, 99%, or more of the cells have an agent conjugated thereto. In some embodiments, the conjugated agent may be one or more of the agents described herein.
In some embodiments, two, three, four, five or more different endogenous non-engineered polypeptides expressed by the cell have an agent conjugated thereto via a sortase-mediated reaction. The agents attached to different polypeptides may be the same or the cell may be sortagged with a plurality of different agents.
Red blood cell conjugates are described in International Publication No. WO 2021/185359, the contents of which are incorporated herein in their entirety. In some embodiments, the agent is linked via a sortase recognition motif to the at least one endogenous, non-engineered membrane protein. In some embodiments, the sortase recognition motif may be selected from a group consisting of LPXTG, LPXAG, LPXSG, LPXLG, LPXVG, LGXTG, LAXTG, LSXTG, NPXTG, MPXTG, IPXTG, SPXTG, VPXTG, YPXRG, LPXTS and LPXTA, wherein X is any amino acid. It can be understood that after the agent linked to the membrane protein, the last (e.g., 5th from the direction of N-terminal to C-terminal) residue of the sortase recognition motif is replaced by the amino acid on which the linkage occurs, as described elsewhere herein. For example, the agent linked to the at least one endogenous, non-engineered membrane protein comprises A1-L1-P1, in which L1 is linked to a glycine (n) in P1, and/or a structure of A1-L1-P2, in which L1 is linked to the side chain ε-amino group of lysine in P2, wherein n is preferably 1 or 2; L1 is selected from the group consisting of LPXT, LPXA, LPXS, LPXL, LPXV, LGXT, LAXT, LSXT, NPXT, MPXT, IPXT, SPXT, VPXT, YPXR, LPXT and LPXT; A1 represents the agent; P1 and P2 independently represent the at least one endogenous, non-engineered membrane protein; and X represents any amino acids. In some embodiments, the agent conjugated to the at least one endogenous, non-engineered membrane protein comprises A1-LPXT-P1, in which LPXT is linked to a glycine (n) in P1, and/or a structure of A1-LPXT-P2, in which LPXT is linked to the side chain ε-amino group of lysine in P2, wherein n is preferably 1 or 2, A1 represents the agent, P1 and P2 independently represent the at least one endogenous, non-engineered membrane protein, and X represents any amino acids. In some embodiments, P1 and P2 may be the same or different. In some embodiments, the agent is linked to one or more (e.g., two, three, four, five or more) glycine (n) or lysine side chain ε-amino groups in or within an extracellular domain of the at least one endogenous, non-engineered membrane protein.
In some embodiments, genetically engineered red blood cells are modified by using sortase to attach a sortase substrate to a non-genetically engineered endogenous polypeptide of the cell. The red blood cell may, for example, have been genetically engineered to express any of a wide variety of products, e.g., polypeptides or noncoding RNAs, may be genetically engineered to have a deletion of at least a portion of one or more genes, and/or may be genetically engineered to have one or more precise alterations in the sequence of one or more endogenous genes. In certain embodiments, a non-engineered endogenous polypeptide of such genetically engineered cell is sortagged with any of the various agents described herein.
In some embodiments, the present disclosure contemplates using autologous red blood cells that are isolated from an individual to whom such isolated red blood cells, after modified in vitro, are to be administered. In some embodiments, the present disclosure contemplates using immuno-compatible red blood cells that are of the same blood group as an individual to whom such cells are to be administered (e.g., at least with respect to the ABO blood type system and, in some embodiments, with respect to the D blood group system) or may be of a compatible blood group.
The terms “non-engineered, “non-genetically modified” and “non-recombinant” as used herein are interchangeable and refer to not being genetically engineered, absence of genetic modification, etc. Non-engineered membrane proteins encompass endogenous proteins. In certain embodiments, a non-genetically engineered red blood cell does not contain a non-endogenous nucleic acid, e.g., DNA or RNA that originates from a vector, from a different species, or that comprises an artificial sequence, e.g., DNA or RNA that was introduced artificially. In certain embodiments, a non-engineered cell has not been intentionally contacted with a nucleic acid that is capable of causing a heritable genetic alteration under conditions suitable for uptake of the nucleic acid by the cells.
The RBCs suitable for use in the compositions and methods of the application can be autologous or obtained from a matching donor. The RBCs can be isolated from the blood of a subject or a matching donor, optionally genetically engineered, then conjugated to an agent, such as the UOX. Any suitable method for RBC isolation and/or genetic engineering can be used in view of the present disclosure. See, e.g., Glassman et al., Pharmaceutics. 2020 May; 12 (5) : 440 and the references therein, which are incorporated herein by reference in their entirety.
Sortase
Enzymes identified as “sortases” have been isolated from a variety of Gram-positive bacteria. Sortases, sortase-mediated transacylation reactions, and their use in protein engineering are well known to those of ordinary skills in the art (see, e.g., PCT/US2010/000274 (WO/2010/087994) , and PCT/US2011/033303 (WO/2011/133704) ) . Sortases have been classified into 4 classes, designated A, B, C, and D, based on sequence alignment and phylogenetic analysis of 61 sortases from Gram-positive bacterial genomes (Dramsi S, Trieu-Cuot P, Bierne H, Sorting sortases: a nomenclature proposal for the various sortases of Gram-positive bacteria. Res Microbiol. 156 (3) : 289-97, 2005) . Those skilled in the art can readily assign a sortase to the correct class based on its sequence and/or other characteristics such as those described in Drami, et al., supra. The term “sortase A” as used herein refers to a class A sortase, usually named SrtA in any particular bacterial species, e.g., SrtA from S. aureus or S. pyogenes.
The term “sortase” also known as transamidases refers to an enzyme that has transamidase activity. Sortases recognize substrates comprising a sortase recognition motif, e.g., the amino acid sequence LPXTG (SEQ ID NO: 8) . A molecule recognized by a sortase (i.e., comprising a sortase recognition motif) is sometimes termed a “sortase substrate” herein. Sortases tolerate a wide variety of moieties in proximity to the cleavage site, thus allowing for the versatile conjugation of diverse entities so long as the substrate contains a suitably exposed sortase recognition motif and a suitable nucleophile is available. The terms “sortase-mediated transacylation reaction” , “sortase-catalyzed transacylation reaction” , “sortase-mediated reaction” , “sortase-catalyzed reaction” , “sortase reaction” , “sortase-mediated transpeptide reaction” and like terms, are used interchangeably herein to refer to such a reaction. The terms “sortase recognition motif” , “sortase recognition sequence” and “transamidase recognition sequence” with respect to sequences recognized by a transamidase or sortase, are used interchangeably herein. The term “nucleophilic acceptor sequence” refers to an amino acid sequence capable of serving as a nucleophile in a sortase-catalyzed reaction, e.g., a sequence comprising an N-terminal glycine (e.g., 1, 2, 3, 4, or 5 N-terminal glycines) or in some embodiments comprising internal glycines (n=1 or 2) or lysine side chain ε-amino group.
The present disclosure encompasses embodiments relating to any of the sortase classes known in the art (e.g., a sortase A, B, C or D from any bacterial species or strain) . In some embodiments, sortase A is used, such as SrtA from S. aureus. In some embodiments it is  contemplated to use two or more sortases. In some embodiments the sortases may utilize different sortase recognition sequences and/or different nucleophilic acceptor sequences.
In some embodiments, the sortase is a sortase A (SrtA) . SrtA recognizes the motif LPXTG (SEQ ID NO: 8) , with common recognition motifs being, e.g., LPKTG, LPATG, LPNTG. In some embodiments LPETG is used. However, motifs falling outside this consensus may also be recognized. For example, in some embodiments the motif comprises an ‘A’ , ‘S’ , ‘L’ or ‘V’ rather than a ‘T’a t position 4, e.g., LPXAG, LPXSG, LPXLG or LPXVG, e.g., LPNAG or LPESG, LPELG or LPEVG. In some embodiments the motif comprises an ‘A’ rather than a ‘G’ at position 5, e.g., LPXTA, e.g., LPNTA. In some embodiments the motif comprises a ‘G’ or ‘A’ rather than ‘P’ at position 2, e.g., LGXTG or LAXTG, e.g., LGATG or LAETG. In some embodiments the motif comprises an ‘I’ or ‘M’ rather than ‘L’ at position 1, e.g., MPXTG or IPXTG, e.g., MPKTG, IPKTG, IPNTG or IPETG. Diverse recognition motifs of sortase A are described in Pishesha et al. 2018.
In some embodiments, the sortase recognition sequence is LPXTG, wherein X is a standard or non-standard amino acid. In some embodiments, X is selected from D, E, A, N, Q, K, or R. In some embodiments, the recognition sequence is selected from LPXTG, LPXAG, LPXSG, LPXLG, LPXVG, LGXTG, LAXTG, LSXTG, NPXTG, MPXTG, IPXTG, SPXTG, VPXTG, YPXRG, LPXTS and LPXTA, wherein X may be any amino acids, such as those selected from D, E, A, N, Q, K, or R in certain embodiments.
In some embodiments, the present disclosure contemplates using a variant of a naturally occurring sortase. In some embodiments, the variant is capable of mediating a glycine (n) conjugation and/or a lysine side chain ε-amino group conjugation, preferably at internal sites of the extracellular domain of the at least one endogenous, non-engineered membrane protein of a red blood cell, preferably n being 1 or 2. Such variants may be produced through processes such as directed evolution, site-specific modification, etc. Considerable structural information regarding sortase enzymes, e.g., sortase A enzymes, is available, including NMR or crystal structures of SrtA alone or bound to a sortase recognition sequence (see, e.g., Zong Y, et al. J. Biol Chem. 2004, 279, 31383-31389) . The active site and substrate binding pocket of S. aureus SrtA have been identified. One of ordinary skills in the art can generate functional variants by, for example, avoiding deletions or substitutions that would disrupt or substantially alter the active site or substrate binding pocket of a sortase. In some embodiments, directed evolution on  SrtA can be performed by utilizing the FRET (Fluorescence Resonance Energy Transfer) -based selection assay described in Chen, et al. Sci. Rep. 2016, 6 (1) , 31899. In some embodiments, a functional variant of S. aureus SrtA may be those described in CN10619105A and CN109797194A. In some embodiments, the S. aureus SrtA variant can be a truncated variant with e.g. 25-60 (e.g., 30, 35, 40, 45, 50, 55, 59 or 60) amino acids being removed from N-terminus (as compared to the wild type S. aureus SrtA) .
In some embodiments, the present disclosure contemplates a S. aureus SrtA variant (mg SrtA) comprising or consisting essentially of or consisting of an amino acid sequence having at least 60% (e.g., at least 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5%, 99.9%or higher) identity to an amino acid sequence as set forth in SEQ ID NO: 3. In some embodiments, the present disclosure provides a nucleic acid encoding the S. aureus SrtA variant, and in some embodiments the nucleic acid is set forth in SEQ ID NO: 4.
Agents
Depending on the intended applications of the modified red blood cells, a wide variety of agents such as a binding agent, a therapeutic agent or a detection agent can be contemplated in the present disclosure. In some embodiments, an agent comprises a protein, a peptide (e.g., an extracellular domain of oligomeric ACE2) , an antibody or its functional antibody fragment, an antigen or epitope, a MHC-peptide complex, a drug such as a small molecule drug (e.g., an antitumor agent such as a chemotherapeutic agent) , an enzyme (e.g., a functional metabolic or therapeutic enzyme) , a hormone, a cytokine, a growth factor, an antimicrobial agent, a probe, a ligand, a receptor, an immunotolerance-inducing peptide, a targeting moiety or any combination thereof.
In some embodiments, in addition to a therapeutically active domain such as an enzyme, a drug, a small molecule (such as a small molecule drug (e.g., an antitumor agent such as a chemotherapeutic agent) , a therapeutic protein and a therapeutic antibody as described herein, the agent can further comprise a targeting moiety for targeting the cells and/or agent to a site in the body where the therapeutic activity is desired. The targeting moiety binds to a target present at such a site. Any targeting moiety can be used, e.g., an antibody. The site can be any organ or tissue, e.g., respiratory tract (e.g., lung) , bone, kidney, liver, pancreas, skin, cardiovascular system (e.g., heart) , smooth or skeletal muscle, gastrointestinal tract, eye, blood vessel surfaces, etc.
In some embodiments, a protein is an enzyme such as a functional metabolic or therapeutic enzyme, e.g., an enzyme that plays a role in metabolism or other physiological processes in a mammal. In some embodiments a protein is an enzyme that plays a role in carbohydrate metabolism, amino acid metabolism, organic acid metabolism, porphyrin metabolism, purine and/or pyrimidine metabolism. Deficiencies of enzymes or other proteins can lead to a variety of diseases, e.g., diseases associated with defects in carbohydrate metabolism, amino acid metabolism, organic acid metabolism, purine or pyrimidine metabolism, and blood clotting, among others. Metabolic diseases are characterized by the lack of functional enzymes or excessive intake of metabolites. Thus, the metabolites deposition in the circulation and tissues causes tissue damage. Due to the wide distribution in human body of RBCs, the present disclosure contemplates modifying membrane proteins of RBCs with functional metabolic enzymes. The enzymes targeted RBCs will uptake metabolites in plasma of patients.
In certain embodiments, the agent comprises urate oxidase (UOX) .
Immunosuppressors
As used herein, “immunosuppressor” and “immunosuppressant” are used interchangeably and refer to an immunosuppressive drug. Immunosuppressive drugs can be classified into five groups: glucocorticoids, cytostatics, antibodies, drugs acting on immunophilins and other drugs. In certain embodiments, the immunosuppressor is a glucocorticoid such as prednisone, dexamethasone (DEX) , or hydrocortisone. In certain embodiments, the immunosuppressor is a cytostatic, such as cyclophosphamide (CTX) or methotrexate (MTX) . In certain embodiments, the immunosuppressor is selected from the group consisting of dexamethasone (DEX) , cyclophosphamide (CTX) and methotrexate (MTX) .
Composition
Also provided is a first pharmaceutical composition comprising an agent conjugated to a red blood cell (RBC) for use with a second pharmaceutical composition comprising an immunosuppressor.
In some embodiments, a first composition comprises a plurality of red blood cells. In some embodiments, at least a selected percentage of the cells in a first composition are modified, i.e., having an agent conjugated thereto by sortase. For example, in some embodiments at least 5%, 10%, 15%, 20%, 25%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 96%, 97%, 98%, 99%,  or more of the cells have an agent conjugated thereto. In some embodiments, two or more red blood cells or red blood cell populations conjugated with different agents are included.
In some embodiments, a first composition comprises sortagged blood red cells, wherein the cells are sortagged with any agent of interest. In some embodiments, a first composition comprises an effective amount of cells, e.g., up to about 1014 cells, e.g., about 10, 102 , 103 , 104 , 105 , 5×105 , 106 , 5×106 , 107 , 5×107 , 108 , 5×108 , 109 , 5×109 , 1010, 5×1010, 1011, 5×1011, 1012, 5×1012, 1013, 5×1013, or 1014 cells. In some embodiments the number of cells may range between any two of the afore-mentioned numbers.
As used herein, the term “an effective amount” refers to an amount sufficient to achieve a biological response or effect of interest, e.g., reducing one or more symptoms or manifestations of a disease or condition or modulating an immune response. In some embodiments a first composition administered to a subject comprises up to about 1014 cells, e.g., about 103, 104, 105, 106, 107, 108, 109, 1010, 1011, 1012, 1013 or 1014 cells, or any intervening number or range.
As used herein, the term “aphysiologically acceptable carrier” is meant a solid or liquid filler, diluent or encapsulating substance that may be safely used in systemic administration. Depending upon the particular route of administration, a variety of carriers, diluent and excipients well known in the art may be used. These can be selected from a group including sugars, starches, cellulose and its derivatives, malt, gelatine, talc, calcium sulfate, vegetable oils, synthetic oils, polyols, alginic acid, phosphate buffered solutions, emulsifiers, isotonic saline and salts such as mineral acid salts including hydrochlorides, bromides and sulfates, organic acids such as acetates, propionates and malonates, water and pyrogen-free water.
Use
In certain embodiments, a combination or composition of the application is for use in a method of inducing immune tolerance to an agent. As used herein, “immune tolerance” or “immunotolerance” refers to a state of unresponsiveness of the immune system to an agent that would otherwise have the capacity to elicit an immune response in a subject.
In certain embodiments, a combination or composition of the application is for use in a method of reducing the immunogenicity of an agent in a subject. As used herein,  “immunogenicity” refers to the ability of a foreign substance, such as an antigen, to provoke an immune response.
Also provided is a method of inducing immune tolerance to an agent, the method comprising administering to the subject a combination comprising administering to a subject (a) an effective amount of the agent conjugated to an RBC and (b) an effective amount of an immunosuppressor to thereby induce the immune tolerance to the agent in the subject. Preferably, the induced immune tolerance is specific to the agent.
The agent conjugated to the RBC and the immunosuppressor can be administered to the subject in any order, e.g., the agent conjugated to the RBC can be administered before, after or at the same time with the immunosuppressor. In certain embodiment, the agent conjugated to the RBC and the immunosuppressor are administered to the subject within 7 days, such as within 6, 5, 4, 3 or 2 days. Preferably, the agent conjugated to the RBC and the immunosuppressor are administered to the subject on the same day.
In certain embodiments, the subject is in need of a treatment for a disease.
As used herein, “treating” , “treat” or “treatment” refers to a therapeutic intervention that at least partly ameliorates, eliminates or reduces a symptom or pathological sign of a pathogen-associated disease, disorder or condition after it has begun to develop. Treatment need not be absolute to be beneficial to the subject. The beneficial effect can be determined using any methods or standards known to the ordinarily skilled artisan.
In some embodiments, the method as described herein further comprises administering the agent conjugated red blood cells to a subject, e.g., directly into the circulatory system, e.g., intravenously, by injection or infusion, one or more times.
In another aspect, provided is a method of delivering an agent to a subject in need thereof, comprising administering the combination as described herein to the subject. The term "delivery" or “delivering” refers to transportation of a molecule or agent to a desired cell or tissue site. Delivery can be to the cell surface, cell membrane, cell endosome, within the cell membrane, nucleus or within the nucleus, or any other desired area of the cell.
In some embodiments, a subject receives a single dose of the agent conjugated red blood cells, or receives multiple doses of the agent conjugated red blood cells, e.g., between 2 and 5, 10, 20, or more doses, over a course of treatment. In some embodiments a dose or total cell number may be expressed as cells/kg. For example, a dose may be about 103, 104, 105, 106,  107, 108 cells/kg. In some embodiments a course of treatment lasts for about 1 week to 12 months or more e.g., 1, 2, 3 or 4 weeks or 2, 3, 4, 5 or 6 months. In some embodiments a subject is treated about every 2-4 weeks. One of ordinary skills in the art will appreciate that the number of cells, doses, and/or dosing interval may be selected based on various factors such as the weight, and/or blood volume of the subject, the condition being treated, response of the subject, etc. The exact number of cells required may vary from subject to subject, depending on factors such as the species, age, weight, sex, and general condition of the subject, the severity of the disease or disorder, the particular cell (s) , the identity and activity of agent (s) conjugated to the cells, mode of administration, concurrent therapies, and the like.
It will be appreciated by those skilled in the art that other variations of the embodiments described herein may also be practiced without departing from the scope of the invention. Other modifications are therefore possible.
Although the disclosure has been described and illustrated in exemplary forms with a certain degree of particularity, it is noted that the description and illustrations have been made by way of example only. Numerous changes in the details of construction and combination and arrangement of parts and steps may be made. Accordingly, such changes are intended to be included in the invention, the scope of which is defined by the claims.
EXAMPLES
Examples are provided below further illustrating different features of the present invention and methodology for practicing the invention. The provided examples do not limit the claimed invention.
Example 1. Preparation of UOX-RBCs
Purification of UOX- (GS) 3-LPETGG protein
The UOX- (GS) 3-LPETGG used in this Example contains the amino acid sequence of SEQ ID NO: 1 shown below:
The nucleotide sequence encoding the UOX- (GS) 3-LPETGG used in this Example contains the polynucleotide sequence of SEQ ID NO: 2 shown below:
The coding sequence of UOX (Aspergillus flavus uricase) was codon optimized for expression in E. coli and synthesized by GenScript. Subclones were generated by standard PCR procedure and inserted into the pET-30a vector with coding sequence for a C-terminal (GS) 3 (SEQ ID NO: 5) linker followed by a sortase recognition sequence (LPETGG) (SEQ ID NO: 6) . All constructs were verified by sequencing and then transformed in E. coli BL21 (DE3) for protein expression.
A single transformed colony was inoculated into 10 ml Luria-Bertani (LB) medium supplemented with ampicillin (100 μg/ml, Beyotime Biotechnology, Suzhou, China) , grew overnight at 37℃ with 220 rpm shaking. Next day, this 10 ml culture was transferred to 1 L fresh LB medium and grew at 37℃ with 220 rpm shaking until OD600 reached 0.6. The temperature of the culture was then lowered to 20℃ and 1 mM IPTG (Sigma, St. Louis, MO) was added for induction.
After induction, a cell pellet was collected by centrifugation, resuspended in low salt lysis buffer (50 mM Tris 8.8, 50 mM NaCl) and then lysed with sonication. A supernatant containing the recombinant protein UOX- (GS) 3-LPETGG was collected by centrifugation at 10,000 rpm for 1 h, and loaded on a Q Sepharose FF column (Cytiva, Marlborough, USA) pre-equilibrated with buffer A (20 mM Tris 8.8) . The column was washed with the buffer A until the absorbance at 280 nm and conductivity became stable and then eluted with a linear gradient of 0- 1 M NaCl in 20 mM Tris 8.8. Fractions corresponding to the elution peak were analyzed by SDS-PAGE and the purest fractions were pooled. The pooled elution was diluted by a buffer (20mM Tris8.0) , then loaded on a Diamond MixA column (Bestchrom, Shanghai, China) and eluted with a linear gradient of 0-1 M NaCl in 20 mM Tris 8.0. Fractions corresponding to the elution peak were analyzed by SDS-PAGE and the purest fractions were pooled. After adding equal volume buffer (40mM Tris pH7.5, 2M (NH42SO4) , the elution sample was loaded on a UniHR Phenyl-80L column (NanMicr) , and washed with 60%gradient buffer B (20 mM Tris 7.5) , then eluted with 100%buffer B (20mM Tris7.5) . Concentration of the elution was detected with Amicon Ultra-15 Centrifugal Filter Unit (Millipore, Darmstadt, Germany) . Concentrated elution was loaded on a EzLoad 16/60 Chromdex 200 pg (Bestchrom) pre-equilibrated with phosphate buffered saline (PBS) , and then the target protein peak was collected.
Preparation of RBCs labeled with UOX- (GS) 3-LPETGG
Red blood cells were separated from peripheral blood of C57BL/6J mice and rats (jh-lab animal) by density gradient centrifugation, respectively. The separated red blood cells were washed with PBS for 3 times. Then the RBCs were pretreated with 2.5 mM Tris (2-carboxyethyl) phosphine hydrochloride (TCEP) (Sigma) for 1 hr at room temperature. The pretreated RBCs were washed with PBS for 3 times. (6-maleimidohexanoic acid) -K (GGG) -GGG-K (GGG) -GGG-K (GGG) -NH2 (SEQ ID NO: 7) , also called G peptide-mal was synthesized with more than 99%purity (Beijing Scilight Biotechnology Led. Co. ) . G peptide-mal was dissolved in phosphate buffer to a final concentration of 10 mg/mL. Then, 1×109 RBCs were contacted with 0.17mg/mL G peptide-mal for 30 mins at 30℃ to obtain RBCs modified with G peptide-mal, named as G peptide-mal-RBCs. The obtained G peptide-mal-RBCs were then washed with PBS for 3 times. The G peptide-mal-RBCs were either used immediately or stored at 4℃ for further use.
G peptide-mal-RBCs (1×109/mL) were conjugated with UOX- (GS) 3-LPETGG via a sortase (mg SrtA) catalyzed conjugation reaction. In the conjugation reaction, concentration of mg SrtA was 10 μM and the UOX- (GS) 3-LPETGG substrate was in the range of 25 μM-100 μM. After the conjugation, the final products, UOX- (GS) 3-LPET-G peptide-mal-RBCs (also described as UOX-RBCs below) , were stored at 2–8 ℃.
The mg SrtA used in the conjugation reaction of this Example contains the amino acid sequence of SEQ ID NO: 3 below:
The nucleotide sequence encoding the used mg SrtA contains the nucleotide sequence of SEQ ID NO: 4 below:
Example 2. Preparation of OVA-RBCs
OVA-RBCs were prepared by SMCC (succinimidyl-trans-4- (N-maleimidylmethyl) cyclohexane-1-carboxylate) labeling. Briefly, SMCC (Sigma) is a hetero-bifunctional crosslinker that contain N-hydroxysuccinimide (NHS) ester and maleimide groups that allow covalent conjugation of amine-and sulfhydryl-containing molecules. NHS esters react with primary amines to form amide bonds, while maleimides react with sulfhydryl groups to form stable thioether bonds. Red blood cells were separated from peripheral blood of C57BL/6J mice and rats (jh-lab animal) by density gradient centrifugation, respectively. The separated red blood cells were washed with PBS for 3 times. Then the RBCs were pretreated with 2.5 mM TCEP for 1 hr at room temperature to expose sulfhydryl and then washed with PBS for 3 times. Amine containing OVA protein (InvivoGen) was react with SMCC NHS ester for 1h at RT and ultrafiltration at 4200g, 20 min for 3 times to remove remnant SMCC and obtain OVA-SMCC. Finally, sulfhydryl exposed RBCs were conjugated with OVA-SMCC, the final products, OVA-RBCs, were stored at 2–8 ℃.
Example 3. Immunosuppressors screening
The capacity of UOX-RBCs to induce immune tolerance with different immunosuppressors was evaluated in rats. The Wistar rats (Vital River) were randomly divided  into 4 groups each contained 3 rats. The 4 groups were respectively treated with, (1) UOX-RBCs (2e10 RBCs/kg) only, (2) UOX-RBCs (2e10 RBCs/kg) in combination with cyclophosphamide (CTX, 72 mg/kg, BAXTER) , (3) UOX-RBCs (2e10 RBCs/kg) in combination with methotrexate (MTX, 50 mg/kg, Zhengqing company) , (4) UOX-RBCs (2e10 RBCs/kg) in combination with dexamethasone (DEX, 3 mg/kg, cspcbaike) .
In particular, the Wistar rats were administrated intravenously with UOX-RBCs in combination with CTX, or MTX, or DEX at day 0 (D0) (tolerance induction) . Then, these rats were challenged with UOX-RBCs (2e10 RBCs/kg) alone at day 7 (D7) . The anti UOX immunogenicity was evaluated using plasma collected at D0, D7, D14 and D21.
Anti-UOX IgG antibodies were measured by indirect ELISA using the coating of the antigen (UOX proteins) at 0.5 μg/mL. HRP-conjugated goat anti-rat IgG (Abcam) or HRP-conjugated goat anti-mouse IgG (Abcam) was used to detect IgG antibodies, respectively, in rats samples. After incubation for 1 h at 37℃ in the dark, TMB (Solarbio) was added. The optical density (OD) of the wells was read immediately after adding an H2SO4 stop solution (Solarbio) at a wavelength 450 nm. Data analysis was performed using GraphPad Prism 8. A four-parameter logistic curve-fit graph was prepared with the dilution on the x axis (log scale) and the OD value on the y axis (linear scale) , and the half maximum value (EC50) for each sample was determined.
The results (Fig. 1) showed that rats treated with UOX-RBCs in combination with MTX or DEX had an attenuated immune response to UOX, although the mean titers increased significantly at D14 (DEX) and D21 (MTX) . The rats treated with UOX-RBCs in combination with CTX showed no detectable antibody titers against UOX up to D21. Therefore, the capacity of an immunosuppressor to induce immune tolerance in combination with UOX-RBCs is CTX>MTX>DEX.
Example 4. A single dose of engineered RBCs in combination with CTX provided durable inhibition of specific immune responses in rodents.
To assess whether UOX-RBCs in combination with CTX could induce immunological tolerance and not just transient immunosuppression, rats (vital River) were treated with UOX-RBCs in combination with CTX at D0 and then repeatedly challenged with UOX-RBCs alone at D7, D28, D56, D77, D98, D119, D140, D161, D182 and D203 as described in Fig. 2. These animals were randomly divided into 3 groups each contained 4 rats and were  treated with: (1) UOX-RBCs only (2e10 RBCs/kg) , (2) UOX-RBCs (2e10 RBCs/kg) in combination with CTX (72 mg/kg) , (3) CTX only (72 mg/kg) . The anti UOX immunogenicity at D0, D7, D14, D28, D49, D70, D91, D112, D133, D154, D175, D196 and D224 was analyzed. The preparation of UOX-RBCs and analysis of anti UOX immunogenicity were described in Example 3.
The results in Fig. 2 showed that UOX-RBCs co-treated with one-time injection of CTX successfully prevented the formation of anti-UOX antibodies during the UOX-RBCs-challenging period, even at Day 224 after ten times of repeated UOX-RBCs challenging in rats. In contrast, animals that received a delayed-immunization with UOX-RBCs (e.g., animals that were pre-treated with CTX on D0, challenged with UOX-RBCs alone on D7 and after) and animals that were treated with UOX-RBCs alone on D0, D7 and after showed a robust immune response during the UOX-RBCs-challenging period. These results demonstrated that UOX-RBCs plus CTX combination therapy induced sustained efficient immune tolerance rather than a transient immune suppression.
The capacity of UOX-RBCs in combination with CTX to induce immune tolerance in C57/B6 mice (jh-labanimal) was also evaluated. These animals were randomly divided into 4 groups each contained 3 mice as described in Fig. 3. UOX-RBCs (2e10 RBCs/kg) in combination with CTX (240 mg/kg) was also efficacious in inhibiting anti-UOX antibody responses in C57/B6 mice (Fig. 3) .
To assess the effect of different antigen conjugated to RBC on tolerance induction, mice were treated with OVA-RBCs (obtained in example 2) (2e10 RBCs/kg) in combination with CTX (240 mg/kg) and repeated challenged them with OVA-RBC (2e10 RBCs/kg) alone as described in Fig. 4. These animals were randomly divided into 2 groups each contained 3 mice. Anti-OVA IgG antibodies were measured by indirect ELISA using the coating of the antigen (OVA proteins) at 0.5 μg/mL. HRP-conjugated goat anti-mouse IgG (Abcam) was used to detect IgG antibodies, respectively, in rat samples. After incubation for 1 h at 37℃ in the dark, TMB (Solarbio) was added. The optical density (OD) of the wells was read immediately after adding an H2SO4 stop solution (Solarbio) at a wavelength 450 nm. Data analysis was performed using GraphPad Prism 8. A four-parameter logistic curve-fit graph was prepared with the dilution on the x axis (log scale) and the OD value on the y axis (linear scale) , and the half maximum value  (EC50) for each sample was determined. The results showed that OVA-RBC in combination with CTX was also effective in inhibiting the antibody response to OVA.
Example 5. Specificity of tolerance induced by UOX-RBCs in combination with CTX
To further evaluate whether UOX-RBCs in combination with CTX induces antigen-specific immunological tolerance rather than chronic immune suppression, mice were challenged with an unrelated antigen after the tolerization period. These animals were randomly divided into 5 groups each contained 2 mice as described in Fig. 6. Mice tolerized to OVA-RBC (2e10 RBCs/kg) in combination with CTX (240 mg/kg) and then challenged with UOX-RBCs (2e10 RBCs/kg) and OVA-RBCs (2e10 RBCs/kg) , respectively, showed a normal immune response to UOX but no response to OVA (Fig. 6) . Importantly, animals that received CTX only during the tolerization period showed robust immune responses to both UOX and OVA.
To investigate the effect of RBCs, the efficacy of UOX-RBCs in combination with CTX was compared with that of the unconjugated UOX protein in combination with CTX in C57/B6 mice (jh-labanimal) . These animals were randomly divided into 3 groups each contained 3 mice as described in Fig. 5. UOX-RBCs (2e10 RBCs/kg) in combination with CTX (240 mg/kg) was efficacious in inhibiting anti-UOX antibody responses in C57/B6 mice. However, equivalent amount of UOX protein (also named UOX- (GS) 3-LPETGG, SEQ ID NO: 1) that were not conjugated to RBCs in combination with CTX (240 mg/kg) failed to induce durable immune tolerance, confirming that the antigen conjugated to RBC was critical to induce durable immune tolerance.
Together these data show that an antigen conjugated to RBC, such as UOX-RBCs, in combination with an immunosuppressor, such as CTX, but not the immunosuppressor (e.g., CTX) alone or unconjugated agent (e.g., UOX protein) in combination with CTX, was capable of inducing durable and specific immunological tolerance to the antigen.
Example 6. Spleen was an important site where tolerance induction is initiated.
RBCs were accumulated primarily in the spleen and liver. Specifically, antigenic peptides, attached to erythrocyte membranes, are captured in the spleen along with erythrocytes [12] . To explore the possible mechanisms of antigen conjugated to RBC in combination with CTX, the tolerance induction in both splenectomized or sham-surgery-treated mice was evaluated. These C57/B6 mice were divided into 4 groups each contained 3 mice. The results (Fig. 7) showed that OVA-RBCs (2e10 RBCs/kg) in combination with CTX (240 mg/kg)  did not induce robust immune tolerance in splenectomized mice, indicating that the spleen is the major organ for induction of tolerance.
One week after tolerance induction, splenocytes were harvested as source of donor cells. Mice then received 10 million unfractionated splenocytes from the mice treated with UOX-RBC in combination with CTX or CTX only. One day after, these mice were challenged with UOX-RBCs alone. A substantial reduction in anti UOX immunogenicity was observed in the mice that received splenocytes from tolerance induced mice, comparing with the mice received splenocytes from CTX injected mice (Fig. 8) . This suggests a role of the spleen as a site where tolerance induction is initiated.
Regularly T cells (FOXP3+CD4+, Treg) being central for induction of robust lone-term tolerance, frequencies of FOXP3+CD4+ T cells were assessed on D10, D14 and D21 by flow cytometry. For staining of splenocytes, cells were exposed for 5 min at RT to RBC lysis buffer (solarbio) to lyse RBC. The following staining steps were performed on ice. Cells were washed with PBS, strained for 15 min for surface staining (CD8-BV711, CD4-PC5.5, biolegend) and finally fixed for 15 min using FIX &PERM Cell Fixation &Permeabilization Kit (thermofisher) . FOXP3 (FOXP3-PE, biolegend) intracellular staining was performed in PBS +2%FBS. Flow cytometry measurements were performed using a Cytoflex (Beckman Coulter) and data were analyzed using version 9.8.2 of FlowJo software. Interestingly, expansion of Tregs in the spleen was assessed in the mice treated with UOX-RBCs in combination with CTX until D21 (Fig. 9) .
In addition, it was shown that distinct splenic effector Tregs were expanded in the mice treated with UOX-RBCs plus CTX. In particular, the mice (n=1 per group, jh-labanimal) were treated with CTX with or without UOX-RBCs on day 0 and challenged with UOX-RBCs only on day 7, and the splenocytes were isolated for single cell transcriptome analysis on day 21 (FIG. 10A) . The frequency of effector Tregs of total splenic Tregs were assessed by scRNA-seq and the results of which are shown in FIG. 10B. The frequency of effector Treg of total CD4 T cells (n=3 per group) assessed by flow cytometry measurements is shown in FIG. 10C. FIG. 10D shows the typical markers that characterized central Tregs and effector Tregs by scRNA-seq.
Example7. UOX-RBCs in combination with CTX has significantly prolonged half-life of UOX proteins in rodents
The pharmacokinetics of UOX-RBCs were evaluated by detecting percentage rat UOX-RBCs in vivo through flow cytometry. Briefly, Wistar rats (Vital River) were administered i.v. with rat UOX-RBCs (2e10 RBCs/kg) in combination with CTX (72 mg/kg) , equivalent UOX proteins in combination with CTX (72 mg/kg) were administrated daily. Each group contained 4 rats.
Whole blood was collected into blood collection tubes with K2-EDTA by tail-nick bleed at 0.5 hr (D0) , D1, D3, D7, D14 and D21 following the UOX-RBCs administration. Samples were analyzed on a Beckman Coulter CytoFLEX LX, and the percentage and mean fluorescent intensity of UOX-RBCs was analyzed by using FlowJoTM software. Results showed that UOX-RBCs in combination with CTX had a significant prolonged half-life compared to UOX proteins and allowing for repeated dosing (Fig. 11) .
Example 8. UOX-RBCs in combination with CTX allowed for repeated dosing and enabled sustained control of uric acid levels.
The pharmacodynamics of UOX-RBCs in combination with CTX were evaluated. Briefly, Wistar rats (Vital River) were administrated i.v. with rat UOX-RBCs (2e10 RBCs/kg) in combination with CTX (72 mg/kg) , equivalent UOX-RBCs were used as control. Each group contained 4 rats. The plasma was collected at D (-1) , D7, D14, D21, D28, D35, D42 and D49 for uric acid evaluation according to the manufacturing protocol (abcam) . The results (Fig. 12) showed that Wistars rats treated with UOX-RBC in combination with CTX inhibited the ADA response, enabling the sustained maintenance of low serum uric acid levels.
References
[1] Garay, R.P., et al., Therapeutic perspectives on uricases for gout. Joint Bone Spine, 2012. 79 (3) : p. 237-42.
[2] 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.
[3] 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.
[4] 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.
[5] Sestigiani E., et al., Efficacy and (pharmaco) kinetics of one single dose of rasburicase in patients with chronic kidney disease. Nephron Clin Pract. 2008; 108 (4) : c265-71
[6] 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.
[7] Raposo CJ, Cserny JD, Serena G, Chow JN, Cho P, Liu H, Kotler D, Sharei A, Bernstein H, John S. Engineered RBCs Encapsulating Antigen Induce Multi-Modal Antigen-Specific Tolerance and Protect Against Type 1 Diabetes. Front Immunol. 2022, 13: 869669.
[8] Pishesha N, Bilate AM, Wibowo MC, Huang NJ, Li Z, Deshycka R, Bousbaine D, Li H, Patterson HC, Dougan SK, Maruyama T, Lodish HF, Ploegh HL. Engineered erythrocytes covalently linked to antigenic peptides can protect against autoimmune disease. Proc Natl Acad Sci U S A. 2017, 114 (12) : 3157-3162.
[9] Antos JM, Truttmann MC, Ploegh HL, Recent Advances in Sortase-Catalyzed Ligation Methodology. Curr Opin Struct Biol. 2016, 38: 111–118.
[10] Y. Ge, L. Chen, S. Liu, J. Zhao, H. Zhang, and P.R. Chen, Enzyme-Mediated Intercellular Proximity Labeling for Detecting Cell-Cell Interactions. J. Am. Chem. Soc., vol. 141, no. 5, pp. 1833–1837, 201.
[11] Swee, L.K.; Lourido, S.; Bell, G.W.; Ingram, J.R.; Ploegh, H. L. One-step Enzymatic Modification of the Cell Surface Redirects Cellular Cytotoxicity and Parasite Tropism. ACS Chem. Biol. 2015, 10: 460-465.
[12] Ukidve, A., et al., Erythrocyte-driven immunization via biomimicry of their natural antigen-presenting function. Proc Natl Acad Sci U S A. 2020, 117 (30) 17727-17736.

Claims (40)

  1. A combination comprising (a) an agent coupled to, more particularly conjugated to, a red blood cell (RBC) and (b) an immunosuppressor.
  2. The combination of claim 1, wherein the agent is conjugated to the RBC by chemical conjugation, binding to a specific receptor such as glycophorin A, sortase-mediated transpeptidation (sortagging) , or passive adsorption.
  3. The combination of claim 2, wherein the agent is conjugated to at least one membrane protein of the RBC by the sortase-mediated transpeptidation, preferably by a sortase-mediated glycine conjugation and/or a sortase-mediated lysine side chain ε-amino group conjugation.
  4. The combination of any one of claims 1-3, wherein the RBC is a natural RBC, such as a natural human RBC, and the membrane protein of claim 3 is an endogenous, non-engineered membrane protein.
  5. The combination of any one of claims 1-3, wherein the RBC is a genetically engineered RBC, and the membrane protein of claim 3 is a genetically engineered membrane protein.
  6. The combination of any one of claims 3-5, wherein the sortase is a Sortase A (SrtA) .
  7. The combination of claim 6, wherein the sortase A is a Staphylococcus aureus transpeptidase A variant (mgSrtA) .
  8. The combination of claim 7, wherein the mgSrtA comprises an amino acid sequence having at least 60%identity, such as at least 60%, 70%, 80%, 90%, 95%and 100%identity, to the amino acid sequence of SEQ ID NO: 3.
  9. The combination of any of claims 1-8, wherein the agent, before being linked to the RBC, comprises a sortase recognition motif on its C-terminus.
  10. The combination of claim 9, wherein the sortase recognition motif comprises an amino acid sequence selected from the group consisting of LPXTG (SEQ ID NO: 8) , LPXAG (SEQ ID NO: 9) , LPXSG (SEQ ID NO: 10) , LPXLG (SEQ ID NO: 11) , LPXVG (SEQ ID NO: 12) , LGXTG (SEQ ID NO: 13) , LAXTG (SEQ ID NO: 14) , LSXTG (SEQ ID NO: 15) , NPXTG (SEQ ID NO: 16) , MPXTG (SEQ ID NO: 17) , IPXTG (SEQ ID NO: 18) , SPXTG (SEQ ID NO: 19) , VPXTG (SEQ ID NO: 20) , YPXRG (SEQ ID NO: 21) , LPXTS  (SEQ ID NO: 22) and LPXTA (SEQ ID NO: 23) , wherein X is any amino acid.
  11. The combination of any one of claims 1-10, wherein the agent comprises a protein, a peptide, an antibody or its functional antibody fragment, an antigen or epitope, a MHC-peptide complex, a drug such as a small molecule drug, an enzyme, a hormone, a cytokine, a growth factor, an antimicrobial agent, a probe, a ligand, a receptor, an immunotolerance-inducing peptide, a targeting moiety or any combination thereof, in particular, the agent comprises urate oxidase (UOX) , preferably, the agent, before being linked to the RBC, comprises the amino acid sequence of SEQ ID NO: 1.
  12. The combination of any one of claims 1-11, wherein the immunosuppressor is selected from the group consisting of cyclophosphamide (CTX) , methotrexate (MTX) , and dexamethasone (DEX) .
  13. The combination of claim 12, wherein the immunosuppressor is cyclophosphamide (CTX) .
  14. A combination comprising (a) an agent comprising a urate oxidase (UOX) conjugated to an RBC and (b) an immunosuppressor selected from the group consisting of cyclophosphamide (CTX) , methotrexate (MTX) , and dexamethasone (DEX) , preferably CTX.
  15. The combination of any one of claims 1-14 for use in a method of inducing immune tolerance to the agent in a subject in need thereof.
  16. The combination of claim 15, wherein the subject is in need of a treatment for a disease, such as gout.
  17. A method of inducing an immune tolerance to an agent in a subject, the method comprising administering to a subject (a) an effective amount of the agent coupled to, more particularly conjugated to, an RBC and (b) an effective amount of an immunosuppressor to thereby induce the immune tolerance to the agent in the subject.
  18. The method of claim 17, wherein the agent conjugated to the RBC and the immunosuppressor are administered to the subject within 7 days, such as within 6, 5, 4, 3 or 2 days, preferably the agent conjugated to the RBC and the immunosuppressor are administered to the subject on the same day.
  19. The method of claim 17 or 18, further comprising administering to the subject the agent conjugated to the RBC one or more additional times, preferably after the administration of the immunosuppressor.
  20. The method of any one of claims 17-19, wherein the agent is conjugated to the RBC by chemical conjugation, binding to a specific receptor such as glycophorin A, sortase-mediated transpeptidation (sortagging) , or passive adsorption.
  21. The method of claim 20, wherein the agent is conjugated to at least one membrane protein of the RBC by the sortase-mediated transpeptidation, preferably by a sortase-mediated glycine conjugation and/or a sortase-mediated lysine side chain ε-amino group conjugation.
  22. The method of any one of claims 17-21, wherein the RBC is a natural RBC, such as a natural human RBC, and the membrane protein of claim 21 is an endogenous, non-engineered membrane protein.
  23. The method of any one of claims 17-21, wherein the RBC is a genetically engineered RBC, and the membrane protein of claim 21 is a genetically engineered membrane protein.
  24. The method of any one of claims 21-23, wherein the sortase is a Sortase A (SrtA) .
  25. The method of claim 24, wherein the sortase A is a Staphylococcus aureus transpeptidase A variant (mgSrtA) .
  26. The method of claim 25, wherein the mgSrtA comprises an amino acid sequence having at least 60%identity, such as at least 60%, 70%, 80%, 90%, 95%and 100%identity, to the amino acid sequence of SEQ ID NO: 3.
  27. The method of any of claims 17-26, wherein the agent, before being linked to the RBC, comprises a sortase recognition motif on its C-terminus.
  28. The method of claim 27, wherein the sortase recognition motif comprises an amino acid sequence selected from the group consisting of LPXTG (SEQ ID NO: 8) , LPXAG (SEQ ID NO: 9) , LPXSG (SEQ ID NO: 10) , LPXLG (SEQ ID NO: 11) , LPXVG (SEQ ID NO: 12) , LGXTG (SEQ ID NO: 13) , LAXTG (SEQ ID NO: 14) , LSXTG (SEQ ID NO: 15) , NPXTG (SEQ ID NO: 16) , MPXTG (SEQ ID NO: 17) , IPXTG (SEQ ID NO: 18) , SPXTG (SEQ ID NO: 19) , VPXTG (SEQ ID NO: 20) , YPXRG (SEQ ID NO: 21) , LPXTS (SEQ ID NO: 22) and LPXTA (SEQ ID NO: 23) , wherein X is any amino acid.
  29. The method of any one of claims 17-28, wherein the agent comprises urate oxidase (UOX) , preferably, the agent, before being linked to the RBC, comprises the amino acid sequence of SEQ ID NO: 1.
  30. The method of any one of claims 17-29, wherein the immunosuppressor is selected from the group consisting of cyclophosphamide (CTX) , methotrexate (MTX) , and dexamethasone (DEX) .
  31. The method of claim 30, wherein the immunosuppressor is cyclophosphamide (CTX) .
  32. A method of inducing an immune tolerance specific to a urate oxidase in a subject, the method comprising administering to a subject (a) an effective amount of an agent comprising the urate oxidase conjugated to an RBC and (b) an effective amount of an immunosuppressor selected from the group consisting of cyclophosphamide (CTX) , methotrexate (MTX) , and dexamethasone (DEX) , preferably CTX, to thereby induce the immune tolerance specific to the urate oxidase in the subject.
  33. The method of claim 32, wherein the urate oxidase conjugated to the RBC and the immunosuppressor are administered to the subject within 7 days, such as within 6, 5, 4, 3 or 2 days, preferably the urate oxidase conjugated to the RBC and the immunosuppressor are administered to the subject on the same day.
  34. The method of claim 32 or 33, further comprising administering to the subject the urate oxidase conjugated to the RBC one or more additional times, preferably after the administration of the immunosuppressor.
  35. The method of any one of claims 17-34, wherein the subject is in need of a treatment for a disease.
  36. The method of claim 35, wherein the disease is gout.
  37. The method of any one of claims 17-36, wherein the RBC is autologous.
  38. The method of any one of claims 17-36, wherein the RBC is from a matching donor.
  39. The effective amount of the agent conjugated to the RBC for use in the method of any one of claims 17-38.
  40. The effective amount of the immunosuppressor for use in the method of any one of claims 17-38.
PCT/CN2023/125936 2022-10-24 2023-10-23 Combination to induce specific immune tolerance Ceased WO2024088213A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202380074882.3A CN120152732A (en) 2022-10-24 2023-10-23 Combinations that induce specific immune tolerance

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN2022127009 2022-10-24
CNPCT/CN2022/127009 2022-10-24

Publications (1)

Publication Number Publication Date
WO2024088213A1 true WO2024088213A1 (en) 2024-05-02

Family

ID=90830102

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2023/125936 Ceased WO2024088213A1 (en) 2022-10-24 2023-10-23 Combination to induce specific immune tolerance

Country Status (2)

Country Link
CN (1) CN120152732A (en)
WO (1) WO2024088213A1 (en)

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014183071A2 (en) * 2013-05-10 2014-11-13 Whitehead Institute For Biomedical Research In vitro production of red blood cells with sortaggable proteins
CN109797194A (en) * 2019-01-24 2019-05-24 北京大学 It marks cell membrane surface and studies the enzyme and method of cell-ECM interaction
CN110612122A (en) * 2017-03-11 2019-12-24 西莱克塔生物科技公司 Methods and compositions related to combination therapy with anti-inflammatory agents and synthetic nanocarriers comprising immunosuppressants
WO2020160325A1 (en) * 2019-01-30 2020-08-06 Horizon Pharma Rheumatology Llc Reducing immunogenicity to pegloticase
WO2022035828A1 (en) * 2020-08-10 2022-02-17 Horizon Therapeutics Ireland Dac Methods of treating gout
WO2022089605A1 (en) * 2020-10-30 2022-05-05 Westlake Therapeutics (Hangzhou) Co. Limited Modified red blood cells and uses thereof for delivering agents

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014183071A2 (en) * 2013-05-10 2014-11-13 Whitehead Institute For Biomedical Research In vitro production of red blood cells with sortaggable proteins
CN110612122A (en) * 2017-03-11 2019-12-24 西莱克塔生物科技公司 Methods and compositions related to combination therapy with anti-inflammatory agents and synthetic nanocarriers comprising immunosuppressants
CN109797194A (en) * 2019-01-24 2019-05-24 北京大学 It marks cell membrane surface and studies the enzyme and method of cell-ECM interaction
WO2020160325A1 (en) * 2019-01-30 2020-08-06 Horizon Pharma Rheumatology Llc Reducing immunogenicity to pegloticase
WO2022035828A1 (en) * 2020-08-10 2022-02-17 Horizon Therapeutics Ireland Dac Methods of treating gout
WO2022089605A1 (en) * 2020-10-30 2022-05-05 Westlake Therapeutics (Hangzhou) Co. Limited Modified red blood cells and uses thereof for delivering agents

Also Published As

Publication number Publication date
CN120152732A (en) 2025-06-13

Similar Documents

Publication Publication Date Title
JP6763832B2 (en) In the prevention and / or treatment of infectious diseases, autoimmune diseases, immune responses to allogeneic factors, allergic diseases, tumors, transplant rejection reactions, and immune responses to viral vectors used for gene therapy or gene vaccination. Immunogenic peptide for use
JP5913120B2 (en) Human lipocalin 2 (Lcn2, hNGAL) mutein having affinity for a given target
JPH09509944A (en) Products and methods for T cell regulation
JP2016028604A (en) Tear lipocalin mutein that binds to IL-4Rα
WO2022089605A1 (en) Modified red blood cells and uses thereof for delivering agents
CN115335064B (en) Modified erythrocytes and their use for delivering a medicament
WO2023284742A1 (en) Cells modified by conjugated n-terminal glycine and uses thereof
WO2024088213A1 (en) Combination to induce specific immune tolerance
WO2022166913A1 (en) Modified red blood cells and uses thereof for treating hyperuricemia and gout
CN115916805A (en) Immunogenic peptides with extended oxidoreductase motifs
JP5594691B2 (en) Modified protein
WO2023103963A1 (en) Modified cells and uses thereof for delivering agents
WO2024067295A1 (en) Modified red blood cell and use thereof for delivering medicament
HK40084691A (en) Modified red blood cells and uses thereof for delivering agents
WO2021007436A1 (en) Tricyclic peptide as protein binders and modulators and uses thereof
HK40038219A (en) Immunogenic peptides for use in the prevention and/or treatment of infectious diseases, autoimmune diseases, immune responses to allofactors, allergic diseases, tumors, graft rejection and immune responses against viral vectors used for gene therapy or gene vaccination
HK40038219B (en) Immunogenic peptides for use in the prevention and/or treatment of infectious diseases, autoimmune diseases, immune responses to allofactors, allergic diseases, tumors, graft rejection and immune responses against viral vectors used for gene therapy or gene vaccination
HK1185886A (en) Immunogenic peptides for use in the prevention and/or treatment of infectious diseases, autoimmune diseases, immune responses to allofactors, allergic diseases, tumors, graft rejection and immune responses against viral vectors used for gene therapy or gene vaccination
HK1185886B (en) Immunogenic peptides for use in the prevention and/or treatment of infectious diseases, autoimmune diseases, immune responses to allofactors, allergic diseases, tumors, graft rejection and immune responses against viral vectors used for gene therapy or gene vaccination

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: 23881791

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 202380074882.3

Country of ref document: CN

NENP Non-entry into the national phase

Ref country code: DE

WWP Wipo information: published in national office

Ref document number: 202380074882.3

Country of ref document: CN

122 Ep: pct application non-entry in european phase

Ref document number: 23881791

Country of ref document: EP

Kind code of ref document: A1