WO2024067295A1 - Globules rouges modifiés et leur utilisation pour administrer un médicament - Google Patents

Globules rouges modifiés et leur utilisation pour administrer un médicament Download PDF

Info

Publication number
WO2024067295A1
WO2024067295A1 PCT/CN2023/120066 CN2023120066W WO2024067295A1 WO 2024067295 A1 WO2024067295 A1 WO 2024067295A1 CN 2023120066 W CN2023120066 W CN 2023120066W WO 2024067295 A1 WO2024067295 A1 WO 2024067295A1
Authority
WO
WIPO (PCT)
Prior art keywords
ggg
sortase
red blood
linker
small peptide
Prior art date
Application number
PCT/CN2023/120066
Other languages
English (en)
Chinese (zh)
Inventor
高晓飞
聂小千
刘璇
黄彦杰
Original Assignee
西湖生物医药科技(杭州)有限公司
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 西湖生物医药科技(杭州)有限公司 filed Critical 西湖生物医药科技(杭州)有限公司
Publication of WO2024067295A1 publication Critical patent/WO2024067295A1/fr

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues

Definitions

  • the present invention relates generally to modified red blood cells (RBCs), and more particularly to covalently modified RBCs and their use for the delivery of drugs and probes.
  • RBCs modified red blood cells
  • Red blood cells are the most common cell type in the human body and have been extensively studied as an ideal in vivo drug delivery system for more than 30 years due to their unique biological properties, including: (i) a wide range of in vivo circulation and a long survival time in vivo; (ii) high biosafety, low immunogenicity, and good biocompatibility as biomaterials; (iii) a large surface area to volume ratio; and (iv) the absence of nuclei, mitochondria, and other organelles.
  • red blood cells which can be mainly divided into: genetically modified red blood cell carriers, non-genetically modified red blood cell carriers and red blood cell membrane modification, such as installing proteins by direct encapsulation, non-covalent linkage of exogenous peptides, or by fusing proteins with RBC surface protein-specific antibodies.
  • modified red blood cells have limitations in in vivo applications. For example, encapsulation can damage the cell membrane, thereby affecting the in vivo survival rate of engineered cells.
  • the non-covalent connection between polymer particles and red blood cells is easily dissociated, and the payload will be quickly degraded in vivo.
  • Bacterial sortases are transpeptidases that can modify proteins in a covalent and site-specific manner.
  • Wild-type sortase A (wtSrtA) from Staphylococcus aureus recognizes the LPXTG motif and cuts between threonine and glycine to form a covalent acyl-enzyme intermediate between the enzyme and the substrate protein.
  • This intermediate is disintegrated by nucleophilic attack of oligoglycine of a peptide or protein, wherein the peptide or protein typically has three consecutive oligoglycine residues (3 ⁇ glycine, G 3 ) at the N-terminus.
  • the present invention provides a modified red blood cell (RBC).
  • RBC red blood cell
  • the present invention provides a modified erythrocyte, wherein the erythrocyte is treated with a reducing agent so that the disulfide bonds in the extracellular domain of at least one endogenous membrane protein of the erythrocyte (e.g., at an internal site of the extracellular domain) are reduced to have free thiol groups.
  • the present invention provides a modified erythrocyte, wherein a linker comprising a maleimido alkyl chain ( C 2-8 ) is covalently bound to a free thiol group on the surface of the erythrocyte membrane.
  • the linker is covalently bound to a free thiol group on the surface of the erythrocyte membrane through 6-maleimidocaproic acid or 4-maleimidobutyric acid contained therein, thereby obtaining an erythrocyte carrying the linker.
  • the linker comprises a small peptide containing oligoglycine (also referred to herein as a "G-containing small peptide").
  • the G-containing small peptide contained in the linker is a linear small peptide or a branched small peptide.
  • the branched small peptide comprises 2 or more branching units, wherein one or more active agents are each coupled to the corresponding branching unit.
  • the branching units have the same structure.
  • the branching unit is composed of an amino acid sequence K (GGG), wherein the glycine (G) in the brackets is conjugated with the side chain ⁇ -amino group of the adjacent lysine (K) to form a branch chain, and the lysine forms a peptide bond with other amino acids through its ⁇ -amino group to form the main chain of the "G-containing small peptide".
  • an extension chain can be added between K and G in the branch unit K (GGG), such as COCH 2 CH 2 -PEG 6 -NH.
  • the G-containing small peptide has a structure selected from the following: GGGSK (SEQ ID NO: 11), K(GGG)-GGG-K(GGG) (SEQ ID NO: 12), K(GGG)-GGG-K(GGG)-GGG-K(GGG) (SEQ ID NO: 13), K(GGG)-GGG-K(GGG)-GGG-K(GGG)-GGG-K(GGG) (SEQ ID NO: 14), or K(GGG)-GGG-K(GGG)-GGG-K(GGG)-GGG-K(GGG)-GGG-K(GGG) (SEQ ID NO: 15), or K[(COCH 2 CH 2 -PEG 6 -NH)-GGG]-GGG-K[(COCH 2 CH 2 -PEG 6 -NH)-GGG]-GGG-K[(COCH 2 CH 2 -PEG 6 -NH)-GGG]-NH 2 (SEQ ID NO: 11),
  • an extension chain can be added between K and G in the branching unit K (GGG), such as COCH 2 CH 2 -PEG 6 -NH.
  • GGG branching unit K
  • the above oligoglycine reacts with an active agent containing a sortase recognition motif under the mediation of a sortase to be conjugated together.
  • a plurality of identical or different active agents are conjugated to a linker comprising a plurality of branching units through the above reaction.
  • the linker comprises a G-containing small peptide and a maleimido alkyl chain (C 2-8 ), and is connected to the membrane protein of the erythrocyte through its maleimido alkyl chain (C 2-8 ), and is connected to the active agent containing the sortase recognition motif through a sortase-mediated reaction by means of its G-containing small peptide.
  • the linker consists of a G-containing small peptide and a maleimido alkyl chain (C 2-8 ).
  • the linker consists of a G-containing small peptide and 6-maleimido hexanoic acid.
  • multiple identical or different active agents are conjugated to erythrocytes at the same time through the linker.
  • at least two active agents are conjugated to erythrocytes at the same time.
  • the above-mentioned active agents are the same active agents.
  • the linker has a structure as shown in Table 1 or Table 3.
  • red blood cells may be conjugated to multiple such linker molecules on their membrane surface.
  • the erythrocytes have not been genetically engineered to express a protein comprising a sortase recognition motif or a nucleophilic receptor sequence, and preferably the erythrocytes are natural erythrocytes, such as natural human erythrocytes.
  • the present invention provides erythrocytes conjugated with an active agent, wherein the active agent is conjugated to the erythrocytes carrying the linker described above.
  • the active agent is modified to comprise a recognition motif for a sortase and is attached to a linker on an erythrocyte via a sortase-mediated reaction, and preferably via sortase-mediated glycine conjugation and/or sortase-mediated lysine side chain epsilon-amino conjugation.
  • the sortase is sortase A (SrtA), such as Staphylococcus aureus transpeptidase A, such as Staphylococcus aureus transpeptidase A variant (mgSrtA).
  • SrtA sortase A
  • mgSrtA comprises, consists essentially of, or consists of an amino acid sequence having at least 60% identity to the amino acid sequence shown in SEQ ID NO:3.
  • the sortase recognition motif comprises or consists essentially of or consists of an amino acid sequence selected from the 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 sortase recognition motif provided herein is LPETG.
  • the sortase recognition motif can be modified to improve its efficiency of recognition, preferably, LPETG is modified to improve its affinity with the sortase, for example, G is added to the C-terminal of the recognition sequence, for example, the modified sequence is LPETGG.
  • the active agent comprises a binding agent, a therapeutic agent or a detection agent, including, for example, a protein, an antibody or a functional antibody fragment thereof, an antigen such as a tumor antigen, an MHC-peptide complex, a drug such as a small molecule drug (e.g., an anti-tumor agent, such as a chemotherapeutic agent), an enzyme (e.g., a functional metabolic enzyme such as UOX, or a therapeutic enzyme), a hormone, a cytokine, a growth factor, an antimicrobial agent, a probe, a ligand, a receptor, an immune tolerance inducing peptide, a targeting moiety, a prodrug, or any combination thereof.
  • a drug such as a small molecule drug (e.g., an anti-tumor agent, such as a chemotherapeutic agent), an enzyme (e.g., a functional metabolic enzyme such as UOX, or a therapeutic enzyme), a hormone, a cytokine
  • the red blood cells after the transamidation reaction of sortase, the red blood cells have a conjugated structure of "linker-LPXT-active agent" on their membrane surface.
  • the active agent can be linked to the sortase recognition motif via a flexible peptide segment.
  • the active agent is a peptide molecule, and preferably the flexible peptide segment fuses the sortase recognition motif to the C-terminus of the active agent peptide molecule.
  • (GS) n is, for example, (GS) 2, (GS) 3, (GS) 4 .
  • the red blood cells conjugated with the active agent have the following structures: UOX-LPET-G1-RBC, UOX-LPET-G2-RBC, UOX-LPET-G3-RBC, UOX-LPET-G4-RBC, UOX-LPET-G5-RBC, UOX-LPET-G6-RBC, anti-PD1 mAb-LPET-G3-RBC, anti-PD1 mAb-LPET-G1-RBC, or anti-PD1 mAb-1-LPET-GAASK-RBC.
  • the present application provides a linker molecule, which is composed of a G-containing small peptide and a maleimido alkyl chain (C 2-8 ).
  • the maleimido alkyl chain (C 2-8 ) is 6-maleimidocaproic acid, 4-maleimidobutyric acid.
  • the G-containing small peptide is a linear or branched small peptide.
  • the branched small peptide comprises 2 or more branching units, wherein one or more active agents are each coupled to the corresponding branching unit.
  • the branching units have the same structure.
  • the branching unit consists of the amino acid sequence K (GGG), wherein the glycine in the brackets is conjugated to the side chain ⁇ -amino group of lysine to form a branch, and lysine forms a peptide bond with other amino acids through its ⁇ -amino group to form the main chain of the G-containing small peptide.
  • the G-containing small peptide has a structure selected from the following: GGGSK, K(GGG)-GGG-K(GGG), K(GGG)-GGG-K(GGG)-GGG-K(GGG), K(GGG)-GGG-K(GGG)-GGG-K(GGG)-GGG-K(GGG) or K(GGG)-GGG-K(GGG)-GGG-K(GGG)-GGG-K(GGG)-GGG-K(GGG)-GGG-K(GGG), wherein the glycine in the brackets is conjugated to the ⁇ -amino group of the lysine side chain to form a branch.
  • the small peptide is conjugated together by reacting the oligoglycine in the branch chain with an active agent containing a sortase recognition motif under the mediation of sortase.
  • an active agent containing a sortase recognition motif under the mediation of sortase.
  • multiple identical or different active agents are conjugated to a linker comprising multiple branching units through the above reaction.
  • the linker is connected to the membrane protein of the erythrocyte through its maleimido alkyl chain (C2-8), and is connected to the active agent containing the sortase recognition motif through a sortase-mediated reaction by means of its G-containing small peptide.
  • multiple identical or different active agents are conjugated to the erythrocyte at the same time through the linker.
  • at least two active agents are conjugated to the erythrocyte at the same time.
  • the above-mentioned active agents are the same active agent.
  • the linker has a structure as shown in Table 1.
  • linker described in this aspect in modifying erythrocytes, wherein the erythrocytes and an active agent are coupled together via the linker.
  • the present application provides a method for preparing the red blood cells described in the first aspect, comprising:
  • step 3 In the presence of sortase, contacting the erythrocytes obtained in step 1) with the active agent obtained in step 2) under conditions suitable for the reaction of the sortase, so that the sortase conjugates the active agent to the endogenous membrane protein of the erythrocyte via a linker.
  • the erythrocytes are treated with a reducing agent so that disulfide bonds in the extracellular domain (eg, at an internal site of the extracellular domain) of at least one endogenous membrane protein of the erythrocyte are reduced to have free sulfhydryl groups.
  • the linker molecule is linked to the free sulfhydryl group on the extracellular domain of the endogenous membrane protein of erythrocytes through the 6-maleimidocaproic acid contained in the linker molecule.
  • the active agent is linked to the G-containing small peptide in the linker molecule through the sortase recognition motif LPXTG, and after the transamidation reaction of the sortase, an active agent-LPXT-linker structure is formed.
  • multiple active agents are conjugated to the linker via a linker molecule having a branching unit. In a more specific embodiment, multiple active agents are conjugated to the red blood cells via a linker molecule having a branching unit.
  • the present invention provides a modified erythrocyte obtained by the method of the third aspect, wherein the membrane surface of the erythrocyte is conjugated with an active agent via a linker.
  • the present invention provides a composition comprising the red blood cells described in the first aspect.
  • the composition is a pharmaceutical composition, optionally comprising a pharmaceutically acceptable carrier that is compatible with red blood cells.
  • the present invention provides a method for diagnosing, treating or preventing a disease in a subject in need thereof, comprising administering to the subject red blood cells or the composition as described in the present application.
  • the disease is selected from a tumor or cancer, a metabolic disease, a bacterial infection, a viral infection, an autoimmune disease, and an inflammatory disease.
  • the present invention provides a method of delivering an active agent to a subject in need thereof, comprising administering to the subject a red blood cell or a composition as described in the present disclosure.
  • the present invention provides a method for increasing the plasma half-life of an active agent, comprising:
  • step 1) conjugating the active agent obtained in step 1) with the erythrocytes obtained in step 2) in the presence of a sortase under suitable conditions, wherein the conditions are suitable for the sortase to conjugate the sortase substrate to at least one endogenous non-engineered membrane protein of the erythrocyte through a sortase-mediated reaction, preferably through sortase-mediated glycine conjugation and/or sortase-mediated lysine side chain ⁇ -amino conjugation.
  • the method further comprises administering the active agent conjugated to the red blood cells to the subject, eg, directly into the circulatory system, eg, intravenously.
  • the present invention provides the use of red blood cells or compositions as described herein in the preparation of a medicament for treating or preventing a disease, or in the preparation of a diagnostic agent for diagnosing a disorder, condition or disease, or in the preparation of a medicament for delivering an active agent.
  • the disease is selected from a tumor or cancer, a metabolic disease, a bacterial infection, a viral infection, an autoimmune disease, and an inflammatory disease.
  • the medicament is a vaccine.
  • the present invention provides red blood cells or compositions of the present disclosure for use in diagnosing, treating or preventing a disease in a subject in need thereof.
  • the disease is selected from a tumor or cancer, a metabolic disease, a bacterial infection, a viral infection, an autoimmune disease, and an inflammatory disease.
  • FIG1 shows the effect of using red blood cells containing different linkers on the plasma urate concentration in mice.
  • FIG. 2 shows the effect of different linkers on the drug loading capacity of red blood cells
  • Figure 3 Flow cytometry was used to detect the effect of G1 linker or G3 linker on the coupling efficiency on the surface of natural red blood cells.
  • Control group unlabeled red blood cells
  • experimental group red blood cells labeled with eGFP-LPET-G1, red blood cells labeled with eGFP-LPET-G3.
  • the histogram shows the eGPF signal on the surface of red blood cells after incubation with the corresponding molecules.
  • nucleic acids are written from left to right in a 5' to 3' orientation; amino acid sequences are written from left to right in an amino to carboxyl orientation. It should be understood that the present invention is not limited to the particular methodology, protocols and reagents described, as they may vary depending on the specific circumstances used by those skilled in the art.
  • patient refers to any mammal to which the treatment or composition disclosed herein can be applied.
  • methods and compositions disclosed herein can have medical and/or veterinary applications.
  • the mammal is a human.
  • sequence identity refers to the number of completely matching nucleotides or amino acids after appropriate alignment using a standard algorithm, with respect to the degree of identity of the sequences over a comparison window.
  • percentage of sequence identity is calculated by comparing two optimally aligned sequences over a comparison window, determining the number of positions at which the same nucleic acid base (e.g., A, T, C, G) appears in the two sequences to produce the number of matching positions, dividing the number of matching positions by The total number of positions in the comparison window (i.e., the window size) is then multiplied by 100 to generate the percent sequence identity.
  • sequence identity may be understood to mean the "percent match” calculated by the DNASIS computer program (version 2.5 for Windows; available from Hitachi Software Engineering, Inc., South San Francisco, California, USA).
  • mutation refers to replacing at least one amino acid residue in the parent amino acid sequence with a different amino acid residue.
  • the one or more replacement residues can be "naturally occurring amino acid residues" or "non-naturally occurring amino acid residues". Examples of non-naturally occurring amino acid residues include norleucine, ornithine, norvaline, homoserine, aib and other amino acid residue analogs.
  • position refers to the position of an amino acid residue in the amino acid sequence of a polypeptide. In any case, the positions are numbered sequentially, with the first amino acid residue being numbered 1.
  • mammals include, but are not limited to, domesticated animals (e.g., cows, sheep, cats, dogs, and horses), primates (e.g., humans and non-human primates such as monkeys), rabbits, and rodents (e.g., mice and rats).
  • domesticated animals e.g., cows, sheep, cats, dogs, and horses
  • primates e.g., humans and non-human primates such as monkeys
  • rabbits e.g., mice and rats
  • rodents e.g., mice and rats.
  • the individual is a human.
  • cancer and “cancerous” refer to the physiological condition in mammals that is typically characterized by unregulated cell growth.
  • examples of cancer include, but are not limited to, solid tumors such as lung cancer, lymphoma, breast cancer, liver cancer, bladder cancer, skin cancer, melanoma, colon cancer, rectal cancer, ovarian cancer, cervical cancer, prostate cancer, pancreatic adenocarcinoma, esophageal cancer, head and neck squamous cell carcinoma, thyroid cancer, glioblastoma, glioma, and blood tumors such as leukemia and lymphoma.
  • solid tumors such as lung cancer, lymphoma, breast cancer, liver cancer, bladder cancer, skin cancer, melanoma, colon cancer, rectal cancer, ovarian cancer, cervical cancer, prostate cancer, pancreatic adenocarcinoma, esophageal cancer, head and neck squamous cell carcinoma, thyroid cancer, glioblastoma, glioma, and blood tumors such
  • drug loading refers to the amount of drug loaded per unit weight or per unit volume or per single red blood cell.
  • drug loading of red blood cells is usually measured in ⁇ g/mL as a dosage unit.
  • treatment refers to clinical intervention intended to alter the natural course of a disease in the individual being treated. Desired therapeutic effects include, but are not limited to, preventing the appearance or recurrence of the disease, alleviating symptoms, reducing any direct or indirect pathological consequences of the disease, preventing metastasis, reducing the rate of disease progression, ameliorating or relieving the disease state, and alleviating or improving prognosis.
  • prevention includes inhibition of the occurrence or development of a disease or disorder or symptoms of a particular disease or disorder.
  • subjects with a family history of cancer are candidates for preventive regimens.
  • prevention refers to the administration of a drug before the signs or symptoms of cancer occur, particularly in a subject at risk for cancer.
  • the term "effective amount” refers to such an amount or dosage of the modified red blood cells or compositions of the present invention, which produces the desired effect in a patient in need of treatment or prevention after being administered to the patient in a single or multiple doses.
  • the effective amount can be easily determined by the attending physician who is a person skilled in the art by considering a variety of factors such as the species of the mammal; body weight, age and general health; the specific disease involved; the extent or severity of the disease; the response of the individual patient; the specific antibody administered; the mode of administration; the bioavailability characteristics of the administered formulation; the selected dosing regimen; and the use of any concomitant therapy.
  • a "therapeutically effective amount” refers to an amount that effectively achieves the desired therapeutic outcome at the desired dosage and for the desired period of time.
  • the therapeutically effective amount of the modified erythrocytes or compositions of the present invention can vary according to a variety of factors such as disease state, age, sex, and weight of the individual. Relative to untreated subjects, a "therapeutically effective amount” preferably suppresses measurable parameters (e.g., uric acid content, tumor growth rate, tumor volume, etc.) by at least about 20%, more preferably at least about 40%, even more preferably at least about 50%, 60% or 70%, and still more preferably at least about 80% or 90%.
  • measurable parameters e.g., uric acid content, tumor growth rate, tumor volume, etc.
  • prophylactically effective amount refers to an amount effective to achieve the desired preventive result at the required dosage and for the required period of time. Typically, since a prophylactic dose is used in a subject before or at an earlier stage of the disease, the prophylactically effective amount will be less than the therapeutically effective amount.
  • composition refers to a composition that is in a form that permits the biological activity of the active ingredient contained therein to be effective, and that contains no additional ingredients that are unacceptably toxic to a subject to which the composition would be administered.
  • Red blood cells (RBC)
  • red blood cells are the largest blood cells in the circulatory system. Unlike other blood cells, red blood cells lack a nucleus and are flexible. They can change their shape to adapt to human blood vessels and are mainly responsible for the oxygen supply function in the body. Key protein markers on the surface of red blood cells allow them to circulate in the body for a long time without being cleared by macrophages, thus having a long half-life. This property makes them an excellent candidate for drug carriers. Mature red blood cells without nuclei do not contain any genetic material, so they have good safety compared to other gene and cell therapies.
  • the present invention provides red blood cells conjugated with an active agent, wherein the active agent is connected to the extracellular domain of at least one endogenous membrane protein of RBC through a linker.
  • the red blood cells are modified so that their membrane proteins are covalently linked to the linker.
  • the linker is covalently linked to the free sulfhydryl or amino group of the membrane protein through the maleimide portion contained therein.
  • the red blood cells are modified with a reducing agent so that the disulfide bonds in some endogenous membrane proteins on the surface of the red blood cells are reduced to free sulfhydryls in preparation for connection with the maleimide portion of the linker.
  • the endogenous membrane protein of the modified red blood cells is connected to a linker comprising a maleimide portion and a G-containing small peptide.
  • the red blood cells carrying the linker are contacted with an active agent containing a sortase recognition motif and a conjugation reaction occurs, thereby obtaining red blood cells carrying the active agent.
  • the RBC is a human RBC, such as a human naive RBC.
  • the RBC is not genetically engineered.
  • the invention provides red blood cells having an active agent conjugated thereto by a sortase-mediated reaction.
  • the conjugated active agent can be one or more active agents described herein.
  • the active agent can be conjugated to the red blood cell via a linker listed in Table 1.
  • the active agent after connection comprises the first 4 amino acid residues of the sortase recognition motif, which is, for example, selected from LPXT, LPXA, LPXS, LPXL, LPXV, LGXT, LAXT, LSXT, NPXT, MPXT, IPXT, SPXT, VPXT, YPXR, LPXT and LPXT; X represents any amino acid.
  • the present invention contemplates the use of autologous red blood cells isolated from an individual, which are modified in vitro and then administered to the individual.
  • the present invention contemplates the use of immunocompatible red blood cells that have the same blood type (e.g., at least with respect to the ABO blood group system, and in some embodiments, with respect to the D blood group system) or may be a compatible blood type as the individual to whom the cells are to be administered.
  • sortase also known as transamidase, refers to an enzyme having transamidase activity.
  • Transamidase enzymes generally catalyze the formation of a peptide bond (amide bond) between an acyl donor and a nucleophilic acyl acceptor.
  • Sortase enzymes recognize substrates comprising a sortase recognition motif, such as the amino acid sequence LPXTG. Sortase enzymes cleave the recognition motif between residues threonine and glycine. Molecules recognized by sortase enzymes (i.e., comprising a sortase recognition motif) are sometimes referred to herein as "sortase substrates".
  • sortase enzymes will be apparent to those skilled in the art, including but not limited to sortase A, sortase B, sortase C, and sortase D.
  • the amino acid sequences of sortase enzymes and the nucleotide sequences encoding them are known to those skilled in the art.
  • the sortase enzyme is Staphylococcus aureus sortase A.
  • sortase A recognizes a substrate containing the LPXTG amino acid sequence motif and cleaves the amide bond between Thr and Gly with the help of the active site Cys to produce a sortase A-substrate thioester intermediate; then, the thioester acyl-enzyme intermediate is decomposed by nucleophilic attack of the amino group of a second substrate containing oligoglycine to produce a covalently linked conjugate molecule and regenerate sortase A.
  • a soluble truncated sortase A lacking the transmembrane region can be used, such as, for S. aureus, a truncated SrtA comprising amino acid residues 60 to 206.
  • the sortase A-mediated reaction results in the attachment of molecules containing a sortase recognition sequence (sortase motif) to molecules containing a sortase receptor sequence (e.g., one or more N-terminal glycine residues).
  • SrtA recognizes the motif LPXTG, wherein common recognition motifs include, for example, LPKTG, LPATG, LPNTG.
  • LPETG is used.
  • motifs falling outside the consensus sequence can also be recognized.
  • the 4th position of the motif comprises "A”, “S”, “L” or “V” instead of "T”, such as LPXAG, LPXSG, LPXLG or LPXVG, such as LPNAG or LPESG, LPELG or LPEVG.
  • the 5th position of the motif comprises "A” instead of "G", such as LPXTA, such as LPNTA.
  • the 2nd position of the motif comprises "G” or “A” instead of “P”, such as LGXTG or LAXTG, such as LGATG or LAETG.
  • the 1st position of the motif comprises "I” or “M” instead of "L”, such as MPXTG or IPXTG, such as MPKTG, IPKTG, IPNTG or IPETG.
  • 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 can be any amino acid, such as an amino acid selected from D, E, A, N, Q, K or R in certain embodiments.
  • the sortase recognition motif provided herein is LPETG.
  • the sortase recognition motif can be modified to improve its efficiency of recognition, preferably, LPETG is modified to improve its affinity with the sortase, such as adding G at the C-terminus of the recognition sequence, such as the modified sequence is LPETGG.
  • the present invention contemplates the use of naturally occurring variants of sortases.
  • a large amount of structural information is available for sortases such as sortase A, including NMR or crystal structures of SrtA alone or in combination with 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 Staphylococcus aureus SrtA have been determined.
  • One of ordinary skill in the art can generate functional variants by, for example, deletions or substitutions that do not destroy or significantly change the active site or substrate binding pocket of the sortase.
  • directed evolution of SrtA can be performed by utilizing a FRET (fluorescence resonance energy transfer)-based selection assay described by Chen et al. Sci. Rep. 2016, 6 (1), 31899.
  • the functional variants of Staphylococcus aureus SrtA can be those described in CN10619105A and CN109797194A.
  • the S. aureus SrtA variant can be a truncated variant, for example, with 25-60 (eg, 30, 35, 40, 45, 50, 55, 59, or 60) amino acids removed from the N-terminus (compared to wild-type S. aureus SrtA).
  • the functional variant of S. aureus SrtA useful in the present invention can be a S. aureus SrtA variant comprising one or more mutations in D124G, Y187L, E189R and F200L at amino acid positions D124, Y187, E189 and F200, and optionally further comprising one or more mutations in P94S/R, D160N, D165A, K190E and K196T.
  • the above-mentioned mutant amino acid positions are numbered according to the numbering of wild-type S. aureus SrtA.
  • the sortase is a Staphylococcus aureus transpeptidase A variant (mgSrtA) comprising, consisting essentially of, or consisting of the amino acid sequence:
  • the sequence has 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 more) identity with the amino acid sequence set forth in SEQ ID NO:3.
  • a sortase A variant having higher transamidase activity than naturally occurring sortase A can be used.
  • the activity of the sortase A variant is at least about 10, 15, 20, 40, 60, 80, 100, 120, 140, 160, 180, or 200 times that of wild-type S. aureus sortase.
  • such a sortase variant is used in the compositions or methods of the invention.
  • the sortase variant comprises any one or more of the following substitutions relative to wild-type S.
  • aureus SrtA P94S/R, E105K, E108A, E108Q, D124G, D160N, D165A, Y187L, E189R, K190E, K196T, and F200L mutations.
  • the SrtA variant can have 25-60 (eg, 30, 35, 40, 45, 50, 55, 59, or 60) amino acids removed from the N-terminus.
  • the sortase variants may also contain 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14 or 15 conservative amino acid mutations. Conservative amino acid mutations that do not significantly affect protein activity are well known in the art.
  • Covalent conjugates comprising two entities that are not covalently bound in vivo can be obtained in vitro by using sortases, in particular sortase A.
  • linker refers to a bifunctional or multifunctional molecule that can connect (conjugate) two molecules or entities together, usually with two reactive functions.
  • the linkers used in conjugates can be roughly divided into non-cleavable or cleavable. They can also be divided into straight linkers and branched linkers according to whether the linker is branched. Branched linkers can contain branching units, each of which can be coupled to at least one drug or molecule.
  • the linker mentioned in the present application comprises two parts, namely, a "maleimido alkyl chain (C 2-8 ) part" and a "G-containing small peptide” part.
  • the "G-containing small peptide” part is a branched small peptide comprising 2 or more branch units.
  • the branch unit can be conjugated to the same or different molecules.
  • the terms “branch unit” and “branching unit” can be used interchangeably.
  • the linker can be conjugated to multiple active molecules by virtue of its branching unit.
  • multiple linkers can be conjugated to the membrane surface of erythrocytes.
  • the branching unit is an amino acid sequence K (GGG), wherein a glycine residue adjacent to lysine is conjugated to the side chain ⁇ -amino group of lysine, so that oligoglycine forms a branch chain, and lysine forms a peptide bond with other amino acids through its ⁇ -amino group to form the main chain of a G-containing small peptide.
  • GGG amino acid sequence K
  • the linker is covalently bound to the free thiol groups on the surface of the erythrocyte membrane through its "maleimido alkyl chain (C 2-8 ) part", thereby being conjugated to the erythrocyte membrane.
  • the "maleimido alkyl chain (C 2-8 ) part” is 6-maleimido hexanoic acid or 4-maleimido butyric acid.
  • urate oxidase refers to an enzyme that oxidizes slightly soluble uric acid into more soluble allantoin. Urate oxidase exists in many species, but higher animals such as humans and apes lack biologically active urate oxidase because the urate oxidase gene has mutated in animals, so uric acid exists as the end product of purine metabolism in humans and some other primates.
  • Gout is the most common inflammatory arthritis in adults, especially in adult men, with a global prevalence of 1% to 4%. Gout occurs when monosodium urate crystals (MSU) are deposited in tissues, causing inflammation and severe pain of gout attacks.
  • MSU monosodium urate crystals
  • the biological precursor of gout is elevated serum uric acid (UA) levels (i.e., hyperuricemia).
  • UA serum uric acid
  • uricase is undoubtedly a valuable treatment option for chronic tophaceous gout.
  • UOX rasburicase, pegloticase
  • current therapies have several limitations. First, UOX is significantly immunogenic and may cause severe allergic reactions. Second, these therapeutic enzymes may be inactivated or cleared in vivo due to their short half-life, limited bioavailability, and/or interactions with plasma proteins.
  • the recognition motif LPETG of the sortase was coupled to the UOX protein from Aspergillus flavus using a flexible peptide (GS) 3.
  • GS flexible peptide
  • the recognition motif LPETG can be modified.
  • the affinity is increased by adding G to the C-terminus of the recognition motif. Therefore, in this embodiment, a fusion protein of UOX and LPETGG was constructed, and the obtained fusion protein was named UOX-LPETGG, and its amino acid sequence is shown in SEQ ID NO: 1:
  • the nucleotide sequence encoding the fusion protein UOX-LPETGG is shown in SEQ ID NO: 2:
  • the coding sequence of UOX-LPETG was synthesized by GenScript, and the sequence was divided into three parts: the nucleic acid sequence encoding the UOX protein, the nucleic acid sequence encoding (GS) 3 , and the nucleic acid sequence encoding the C-terminal LPETGG of the fusion protein. After the accuracy of the sequence was confirmed by sequencing, the complete coding nucleic acid was constructed into a suitable expression vector and then transformed into Escherichia coli BL21 (DE3, Tiangen) for protein expression.
  • the transformed single colony was inoculated into 10 ml Luria-Bertani (LB) medium containing ampicillin (100 ⁇ g/ml, Bio-Tech), and cultured at 37°C and 220 rpm with shaking. The next day, 10 ml of the culture was transferred to 1 L fresh LB medium and cultured at 37°C and 220 rpm with shaking until OD600 reached 0.6. The culture temperature was lowered to 20°C, and 1 mM IPTG (sigma) was added for induction.
  • LB Luria-Bertani
  • the cell pellet was collected by centrifugation, resuspended in low salt lysis buffer (50mM Tris 8.8, 50mM NaCl), and then sonicated.
  • the supernatant containing UOX-LPETGG protein was collected by centrifugation at 10,000rpm for 1 hour and loaded onto a Q Sepharose FF column (Cytiva, Marlborough, USA) pre-equilibrated with QA buffer (20mM Tris 8.8). The column was washed with QA buffer until the absorbance was 280nm and the conductivity was stable, and then eluted with a linear gradient solution of 20mM Tris pH8.8 containing 0-1M NaCl.
  • the components corresponding to the elution peak were analyzed by SDS-PAGE, and the purest components were pooled and merged.
  • the combined eluate was diluted with buffer (20mM Tris8.0) and then loaded onto a Diamond MixA column (Borgron (Shanghai) Biotechnology Co., Ltd.) and eluted with a linear gradient solution of 20mM Tris pH 8.0 containing 0-1M NaCl.
  • the components corresponding to the elution peak were analyzed by SDS-PAGE, and the purest components were pooled.
  • the eluted sample was loaded onto a UniHR Phenyl-80L column (Suzhou Na Micro Technology Co., Ltd.), washed with 60% gradient buffer B (20mM Tris7.5), and then eluted with 100% buffer B (20mM Tris7.5).
  • the elution concentration was detected using an Amicon Ultra-15 centrifugal filter device (Millipore).
  • the concentrated eluate was loaded onto an EzLoad 16/60 Chromdex 200pg (Borgron (Shanghai) Biotechnology Co., Ltd.) pre-equilibrated with PBS, and then the target protein peak was collected.
  • the active agent can be conjugated to the membrane surface of erythrocytes using a general straight linker, but the drug loading capacity of erythrocytes obtained in this way sometimes cannot meet clinical needs. Therefore, this example studies the effect of different linkers on the drug loading capacity of erythrocytes.
  • a linker containing a linear and a linear G-containing peptide is prepared.
  • the linker contains only one oligoglycine (e.g., GGG) that can react with an active agent containing a sortase recognition sequence
  • the G-containing peptide is called a G1 peptide
  • the linker contains two oligoglycines that can react with an active agent containing a sortase recognition sequence
  • the G-containing peptide is called a G2 peptide
  • the linker contains three oligoglycines that can react with an active agent containing a sortase recognition sequence the G-containing peptide is called a G3 peptide
  • G4 peptide and G5 peptide are obtained respectively.
  • the linker containing the corresponding peptide can also be called G1, G2, G3, G4, G5.
  • the linker obtained is a branch linker.
  • Zhongke Yaguang was commissioned to prepare and synthesize the various linkers described in Table 1, which contain G1, G2, G3, G4, and G5 small peptides respectively.
  • Red blood cells were separated from the peripheral blood of Wistar rats (purchased from Beijing Weitong Lihua Experimental Animal Technology Co., Ltd.) by density gradient centrifugation.
  • the separated red blood cells were rinsed with PBS three times, and then pretreated with 5mM tri(2-carboxyethyl)phosphine (TCEP, Sigma) at 30°C for 1hr.
  • TCEP tri(2-carboxyethyl)phosphine
  • the pretreated red blood cells were washed with PBS three times, and the linkers containing G1-G5 small peptides described in Table 1 were reacted with the above-treated red blood cells.
  • each linker was dissolved with PBS solution and mixed with the red blood cells pretreated with TCEP, and the final reaction concentration of the linker was 0.625mM. At 30°C, the reaction was carried out for 15min to obtain red blood cells carrying different linkers. The red blood cells carrying different linkers were washed with PBS three times to obtain linker-modified red blood cells, which were named Gn-RBC.
  • amino acid sequence of mg SrtA is shown in SEQ ID NO:3:
  • the nucleotide sequence of mg SrtA is shown in SEQ ID NO:4:
  • the conjugation effect of UOX protein with erythrocyte membrane was detected by sandwich ELISA.
  • the wells of PVC microtiter plates were coated with anti-UOX antibody-1 (purchased from Hangzhou Huaan Biotechnology Co., Ltd.) at a concentration of 0.5 ⁇ g/mL in ELISA coating buffer (pH 9.6, purchased from Beijing Solebow Technology Co., Ltd.) overnight at 4°C; the coating solution was removed and the wells were rinsed twice with 200 ⁇ L PBS; 200 ⁇ L blocking buffer (5% skim milk/PBS) was added to each well, and the remaining protein binding sites in the coated wells were blocked at 37°C for 1 hour; and washed twice with 200 ⁇ L PBS.
  • ELISA coating buffer pH 9.6, purchased from Beijing Solebow Technology Co., Ltd.
  • UOX-LPET-Gn-RBC was lysed with RIPA buffer (R&D) at 4°C for 10 minutes, and then 100 ⁇ L lysis solution was added to each well of the plate.
  • Each plate included a positive control (duplicate) and a blank control, incubated at 37°C for 1 hour. The solution was removed and washed twice with 200 ⁇ L PBS.
  • TMB solution purchased from Beijing Solebold Technology Co., Ltd.
  • stop solution purchased from Beijing Solebold Technology Co., Ltd.
  • the results are shown in Table 2. As the number of G-containing small peptides contained in the linker increases, the drug loading of the engineered erythrocytes obtained after reacting with erythrocytes increases significantly.
  • the UOX content in the erythrocytes carrying UOX conjugated by the G2 linker is 2.5 times that of the erythrocytes carrying UOX conjugated by the G1 linker, and the UOX content in the erythrocytes carrying UOX conjugated by the G3, G4 or G5 linkers is more than 3 times that of the erythrocytes carrying UOX conjugated by the G1 linker.
  • G2, G3, G4 or G5 linkers to modify erythrocytes makes the drug loading efficiency of the modified erythrocytes higher than that of the G1 linker.
  • the urate level in the plasma of mice treated with UOX-LPET-G3-RBC, UOX-LPET-G4-RBC, and UOX-LPET-G5-RBC was significantly lower than the urate level in the plasma of mice treated with UOX-LPET-G1-RBC, indicating that the red blood cells modified with G3, G4 or G5 linkers all make the in vivo efficacy (ability to reduce plasma urate) of the modified red blood cells higher than the efficacy of red blood cells modified with G1 linkers.
  • Red blood cells were isolated from the peripheral blood of C57/B6 mice (Shanghai Jihui Experimental Animal Breeding Co., Ltd.) by density gradient centrifugation. The isolated red blood cells were washed three times with PBS and then pre-warmed with 2.5 mM TCEP (Sigma) at 30°C. Treat for 1 hour. The pretreated red blood cells were washed 3 times with PBS, and the treated red blood cells were reacted with the connectors containing G1 and G3 described in Table 1 and the connector containing G6 in Table 3. Specifically, each connector (G1, G3, G6) was dissolved with a PBS solution and mixed with the red blood cells pretreated with TCEP, and the final reaction concentration of the connector was 0.625mM.
  • red blood cells carrying different connectors were washed three times with PBS to obtain connector-modified red blood cells, and the modified red blood cells were named Gn-RBC based on the name of the connector, where n is an integer of 1-5.
  • the conjugation effect of UOX protein and erythrocyte membrane was detected by sandwich ELISA. Specifically, in ELISA coating buffer (pH 9.6, purchased from Beijing Solebow Technology Co., Ltd.), anti-UOX antibody-1 (purchased from Beijing Solebow Technology Co., Ltd.) at a concentration of 0.5 ⁇ g/mL was used.
  • ELISA coating buffer pH 9.6, purchased from Beijing Solebow Technology Co., Ltd.
  • anti-UOX antibody-1 purchased from Beijing Solebow Technology Co., Ltd.
  • the wells of PVC microtiter plates were coated with RIPA buffer (R&D, Hangzhou Huaan Biotechnology Co., Ltd.) at 4°C overnight; the coating solution was removed and the wells were rinsed twice with 200 ⁇ L PBS; free protein binding sites in the wells were blocked with 200 ⁇ L blocking buffer (5% skim milk/PBS) at 37°C for 1 hour; and washed twice with 200 ⁇ L PBS.
  • UOX-LPET-Gn-RBCs were lysed with RIPA buffer (R&D) at 4°C for 10 min, and then 100 ⁇ L of lysis solution was added to each well of the plate. Each plate included a positive control (duplicate) and a blank control and incubated at 37°C for 1 hour.
  • the solution was removed and washed twice with 200 ⁇ L PBS.
  • 100 ⁇ L of diluted detection anti-UOX antibody-2 solution (1 ⁇ g/mL, conjugated with HRP, purchased from Hangzhou Huaan Biotechnology Co., Ltd.) was added to each well and incubated at 37°C for 1 hour. Washed 4 times with 200 ⁇ L PBS.
  • TMB solution purchased from Beijing Solebow Technology Co., Ltd.
  • an equal volume of stop solution purchasedd from Beijing Solebow Technology Co., Ltd. was added, and the optical density at 450 nm was detected.
  • SrtA SEQ ID NO: 3
  • eGFP-LPETG cDNA was cloned into the pET vector and transformed into Escherichia coli BL21 (DE3) cells for protein expression.
  • the transformed cells were cultured at 37°C until OD600 reached 0.6, and then 500 ⁇ M IPTG (sigma) was added. After culture at 37°C for 4 hours, the cells were collected by centrifugation and lysed with pre-cooled lysis buffer (20 mM Tris-HCl, pH 7.8, 500 mM NaCl). The lysate was sonicated on ice (5 seconds on, 5 seconds off, 60 cycles, 25% power, Branson Sonifier 550 ultrasonic cell disruptor).
  • amino acid sequence of eGFP-LEPTG is shown in SEQ ID NO:5:
  • the nucleotide sequence of eGFP-LEPTG is shown in SEQ ID NO:6:
  • Red blood cells were isolated from the peripheral blood of C57/B6 mice (Shanghai Jihui Experimental Animal Breeding Co., Ltd.) by density gradient centrifugation, respectively, using the same method as in Example 2.
  • the pretreated red blood cells were washed 3 times with PBS and modified with the G1 and G3 connectors described in Table 1, respectively.
  • each connector was dissolved with a PBS solution and mixed with the red blood cells pretreated with TCEP, and the final reaction concentration of the connector was 0.625 mM.
  • the reaction was carried out for 15 minutes to obtain red blood cells carrying different connectors.
  • the red blood cells carrying different connectors were washed three times with PBS to obtain connector-modified red blood cells, and the modified red blood cells were named Gn-RBC based on the names of the connectors.
  • eGFP-LPETG SEQ ID NO: 5
  • sortase mg SrtA
  • concentration of mg SrtA 10 ⁇ M
  • eGFP-LEPTG substrate 25 ⁇ M.
  • the final product after conjugation was named eGFP-LPET-Gn-RBC and stored at 2-8°C.
  • eGFP eGFP-LPETG coupling to Gn-RBC was characterized by the eGFP (FITC) fluorescence signal on the cell membrane.
  • the heavy chain amino acid sequence of anti-PD1 mAb-LPETGG is shown in SEQ ID NO:7:
  • the heavy chain nucleotide sequence of anti-PD1 mAb-LPETGG is shown in SEQ ID NO:8:
  • amino acid sequence of the light chain of the anti-PD1 antibody is shown in SEQ ID NO:9:
  • the nucleotide sequence of the light chain of the anti-PD1 antibody is shown in SEQ ID NO: 10:
  • the nucleotide sequences encoding the above heavy chains or light chains were inserted into the expression vector pcDNA3.1, respectively.
  • Each successfully constructed vector was transfected into CHO-S cells using the ExpiCHO TM expression system (ThermoFisher) according to the manufacturer's instructions.
  • the transfected cells were cultured in ExpiCHO TM expression medium to express the corresponding heavy chain or light chain, thereby assembling the corresponding anti-PD1 antibody. Since LPETGG is linked to the C-terminus of the antibody heavy chain, it is called anti-PD1Ab-LPETGG.
  • the culture supernatant with anti-PD1 mAb-LPETGG protein was then harvested and purified using protein A affinity chromatography (Cytiva), Q Sepharose FF column (Cytiva), and Bestdex G-25 (Borgron (Shanghai) Biotechnology Co., Ltd.) according to the manufacturer's instructions, and the purified target protein was concentrated and stored at -80°C.
  • Red blood cells were separated from the peripheral blood of C57/B6 mice (Shanghai Jihui Experimental Animal Breeding Co., Ltd.) by density gradient centrifugation. The separated red blood cells were washed 3 times with PBS. The red blood cells were then pretreated with 2.5mM TCEP (sigma) at 30°C for 1 hour. The pretreated red blood cells were washed 3 times with PBS, and the treated red blood cells were reacted with the connectors containing G1 and G3 described in Table 1, respectively. Specifically, each connector was dissolved with a PBS solution and mixed with the red blood cells pretreated with TCEP, and the final reaction concentration of the connector was 0.625mM. At 30°C, react for 15 minutes to obtain red blood cells carrying different connectors. The red blood cells carrying different connectors were washed three times with PBS to obtain connector-modified red blood cells, and the modified red blood cells were named Gn-RBC based on the name of the connector, where n is an integer of 1-5.
  • the amount of anti-PD1 mAb conjugated to RBC was measured by sandwich ELISA. Briefly, the wells of PVC microtiter plates were coated with a concentration of 0.5 ⁇ g/mL of captured human PD-1 His tag (ACRO) in ELISA coating buffer (pH 9.6, purchased from Beijing Solebao Technology Co., Ltd.) at 4°C overnight; the coating solution was removed and the plate was washed twice with 200 ⁇ L PBS; 200 ⁇ L blocking buffer (5% skim milk/PBS) was added to each well, and the remaining protein binding sites in the coated wells were blocked at 37°C for 1 h; the plate was washed twice with 200 ⁇ L PBS; anti-PD1 mAb-LPET-Gn-RBC was lysed with RIPA buffer (R&D) at 4°C for 10 minutes.
  • RIPA buffer R&D

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Chemical & Material Sciences (AREA)
  • Biomedical Technology (AREA)
  • Organic Chemistry (AREA)
  • Microbiology (AREA)
  • Wood Science & Technology (AREA)
  • Genetics & Genomics (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Health & Medical Sciences (AREA)
  • Biotechnology (AREA)
  • Zoology (AREA)
  • Mycology (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Cell Biology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Epidemiology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Medicinal Chemistry (AREA)
  • Biochemistry (AREA)
  • General Engineering & Computer Science (AREA)
  • Immunology (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

La présente invention concerne un globule rouge modifié. Un agent actif est conjugué à une partie extracellulaire d'une protéine membranaire d'un globule rouge au moyen d'un lieur, le lieur comprenant un petit peptide contenant G et une chaîne maléimidoalkyle (C2-8). La chaîne maléimidoalkyle (C2-8) est conjuguée à la protéine membranaire du globule rouge, et le petit peptide contenant G est conjugué à un agent actif contenant un motif de reconnaissance d'une enzyme de tri au moyen d'une réaction médiée par une enzyme de tri. La présente invention concerne également un RBC modifié de manière covalente et son utilisation pour administrer un médicament et une sonde.
PCT/CN2023/120066 2022-09-28 2023-09-20 Globules rouges modifiés et leur utilisation pour administrer un médicament WO2024067295A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CNPCT/CN2022/122203 2022-09-28
CN2022122203 2022-09-28

Publications (1)

Publication Number Publication Date
WO2024067295A1 true WO2024067295A1 (fr) 2024-04-04

Family

ID=90476216

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2023/120066 WO2024067295A1 (fr) 2022-09-28 2023-09-20 Globules rouges modifiés et leur utilisation pour administrer un médicament

Country Status (1)

Country Link
WO (1) WO2024067295A1 (fr)

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20160122707A1 (en) * 2013-05-10 2016-05-05 Whitehead Institute For Biomedical Research Protein modification of living cells using sortase
WO2022089605A1 (fr) * 2020-10-30 2022-05-05 Westlake Therapeutics (Hangzhou) Co. Limited Globules rouges modifiés et utilisations correspondantes pour l'administration d'agents
WO2022164392A1 (fr) * 2021-01-29 2022-08-04 National University Of Singapore Globules rouges à surface modifiée et leurs procédés de génération
WO2022166913A1 (fr) * 2021-02-04 2022-08-11 Westlake Therapeutics (Hangzhou) Co. Limited Globules rouges modifiés et utilisations de ces derniers pour traiter l'hyperuricémie et la goutte
WO2023284742A1 (fr) * 2021-07-13 2023-01-19 Westlake Therapeutics (Hangzhou) Co. Limited Cellules modifiées par une glycine n-terminale conjuguée et leurs utilisations
WO2023134573A1 (fr) * 2022-01-12 2023-07-20 Westlake Therapeutics (Shanghai) Co., Limited Cellules modifiées et leurs utilisations

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20160122707A1 (en) * 2013-05-10 2016-05-05 Whitehead Institute For Biomedical Research Protein modification of living cells using sortase
WO2022089605A1 (fr) * 2020-10-30 2022-05-05 Westlake Therapeutics (Hangzhou) Co. Limited Globules rouges modifiés et utilisations correspondantes pour l'administration d'agents
WO2022164392A1 (fr) * 2021-01-29 2022-08-04 National University Of Singapore Globules rouges à surface modifiée et leurs procédés de génération
WO2022166913A1 (fr) * 2021-02-04 2022-08-11 Westlake Therapeutics (Hangzhou) Co. Limited Globules rouges modifiés et utilisations de ces derniers pour traiter l'hyperuricémie et la goutte
WO2023284742A1 (fr) * 2021-07-13 2023-01-19 Westlake Therapeutics (Hangzhou) Co. Limited Cellules modifiées par une glycine n-terminale conjuguée et leurs utilisations
WO2023134573A1 (fr) * 2022-01-12 2023-07-20 Westlake Therapeutics (Shanghai) Co., Limited Cellules modifiées et leurs utilisations

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
SHI, JIAHAI ET AL.: "Engineered Red Blood Cells as Carriers for Systemic Delivery of a Wide Array of Functional Probes", PNAS, vol. 111, no. 28, 15 July 2014 (2014-07-15), XP055189994, DOI: 10.1073/pnas.1409861111 *

Similar Documents

Publication Publication Date Title
US10232014B2 (en) Tear lipocalin muteins binding IL-4 R alpha
TWI710570B (zh) 用於治療代謝異常之組成物及方法
US10206980B2 (en) IL-15 heterodimeric protein and uses thereof
JP2021006024A (ja) 所定の標的に対して親和性を有するヒトリポカリン2(Lcn2、hNGAL)の突然変異タンパク質
CN104145015A (zh) 靶向的溶酶体酶化合物
JP2001504334A (ja) プロテアーゼ活性化可能なPseudomonas体外毒素A様プロタンパク質
CN104136606A (zh) 靶向的艾杜糖-2-硫酸酯酶化合物
JP5966000B2 (ja) 腫瘍ターゲティング腫瘍壊死因子関連アポトーシス誘導リガンド変異体及びその応用
SG173332A1 (en) Muteins of tear lipocalin and methods for obtaining the same
JP2017506886A (ja) P97−ids融合タンパク質
JP2015521463A (ja) 標的化イズロニダーゼ化合物
TW200938632A (en) Recombinant TAT-HOXB4H protein for use as a stimulant of hematopoiesis in vivo and pharmaceutical composition containing the same
CN105452457A (zh) 用于治疗高胱氨酸尿症的胱硫醚β-合酶
JP2021511030A (ja) C3b結合ポリペプチド
JP3469247B2 (ja) 組換えリボヌクレアーゼタンパク質
US20170267731A1 (en) Disintegrin variants and pharmaceutical uses thereof
JPH10513347A (ja) 組換え型ヒトα−フェトプロテインおよびその利用
JP2019520392A (ja) ヒト酵素媒介性シスチン枯渇
WO2023284742A1 (fr) Cellules modifiées par une glycine n-terminale conjuguée et leurs utilisations
CN104955946A (zh) 靶向的艾杜糖醛酸-2-硫酸酯酶化合物
TW201838647A (zh) 對胰島素受體有降低親和性之胰島素類似物之接合物及其用途
CZ186996A3 (en) Method of selective provocation of methionine insufficiency for malignant cells of mammals
WO2020228791A1 (fr) Cellules immunitaires à prolifération de protéine mutante il-2
US20230406927A1 (en) Masked single-domain antibodies and methods thereof
WO2024067295A1 (fr) Globules rouges modifiés et leur utilisation pour administrer un médicament

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: 23870527

Country of ref document: EP

Kind code of ref document: A1