[go: up one dir, main page]

WO2022005338A1 - Procédé de traitement avec un agent de thérapie génique - Google Patents

Procédé de traitement avec un agent de thérapie génique Download PDF

Info

Publication number
WO2022005338A1
WO2022005338A1 PCT/RU2021/050192 RU2021050192W WO2022005338A1 WO 2022005338 A1 WO2022005338 A1 WO 2022005338A1 RU 2021050192 W RU2021050192 W RU 2021050192W WO 2022005338 A1 WO2022005338 A1 WO 2022005338A1
Authority
WO
WIPO (PCT)
Prior art keywords
antibody
disease
subject
agent
antibodies
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/RU2021/050192
Other languages
English (en)
Russian (ru)
Inventor
Максим Петрович НИКИТИН
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.)
Individual
Original Assignee
Individual
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 Individual filed Critical Individual
Publication of WO2022005338A1 publication Critical patent/WO2022005338A1/fr
Anticipated expiration legal-status Critical
Ceased legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7042Compounds having saccharide radicals and heterocyclic rings
    • A61K31/7052Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression

Definitions

  • This invention relates to methods of treating a subject in need of treatment for a disease caused by loss of function or activity of a protein, or methods of treating a subject in need of treatment of a disease caused by an increase in functional activity or expression of a protein, as well as compositions and substances for carrying out such methods.
  • the invention relates to methods, compositions and substances for improving the delivery of heterologous polynucleotides for therapeutic or research purposes.
  • Gene therapy is one of the most promising areas of modern biomedicine, the widespread use of which in clinical practice promises to revolutionize the entire healthcare system and take a confident step towards preventive and personalized medicine of the future.
  • the issue of efficient and safe delivery of such agents continues to be a key problem of the technology.
  • One of the promising solutions to this problem is to combine the achievements of molecular medicine and nanobiotechnology and create new highly efficient delivery systems by combining gene therapy agents with specially designed nanocarriers based on a number of promising materials.
  • nanoparticles have the ability to combine many different agents at once, such as therapeutic, diagnostic, reporter (fluorescent, magnetic, chromophore), which entails increased functionality unattainable with molecular agents; 2) at the same time, the small size in comparison with the cellular structures of the body makes it possible for nanoparticles to penetrate through the smallest capillaries and then be absorbed by cells, which makes it possible to efficiently deliver drugs and localize high doses of drugs in the immediate vicinity of the cell, or inside the cell itself; 3) the possibility of using biodegradable materials for the manufacture of nanoparticles makes it possible to achieve a controlled release of a drug in the target area within a clearly controlled predetermined time - days, weeks, and even months.
  • nanoparticles and nanoscale objects in nature indicates the fundamental possibility of using these delivery systems not only in the field of drug delivery for the treatment of any complex (difficult to treat) conditions of the body, but also for milder effects on the body, such as their use in nutrition for the transfer and delivery of various vitamins, mineral components, etc. to the body.
  • nanoparticles have already firmly won their position as an indispensable tool for various studies in the field of life sciences, biomedicine, fundamental medicine, theranostics, etc.
  • the development of drug delivery systems has been significantly influenced by nanotechnology. It is expected that new drug delivery systems can make a significant contribution to the pharmaceutical market. Approximately 13% of the current global pharmaceutical market is the sale of products that include a drug delivery system. New drug delivery technologies are an important strategic vehicle for expanding drug markets. Improved drug delivery systems can solve the problems of existing agents related to efficacy, side effects and patient tolerance.
  • nanotechnology allows the delivery of drugs that are insoluble or unstable in the biological environment. For example, due to the high pH in the stomach, some drugs are administered intravenously or by encapsulation in controlled release nanoparticles. Also, nanotechnology can solve the problem of premature loss of efficiency due to rapid elimination from the body and degradation as a result of metabolism.
  • Nanoparticle systems have potential advantages such as prolonging the effective concentrations of drugs in the bloodstream or tissue when injected intravenously, which reduces the frequency of administration; localization of high drug concentrations; reducing toxicity and increasing the stability of drugs.
  • Intravenous drug administration provides rapid redistribution in the circulatory system, followed by delivery to target organs. To avoid peak serum levels that are reached shortly after intravascular injection, administration of drugs carried on stable carriers would allow for gradual release of drugs in the circulatory system following bolus intravenous injection of therapeutic nanoparticles.
  • the active substance is dissolved, captured, adsorbed, attached or encapsulated in the nanoparticle matrix.
  • the nanoparticle matrix can be composed of biodegradable materials such as polymers or proteins.
  • nanoparticles with different properties and release characteristics of encapsulated therapeutic agents can be obtained, in particular, in response to the appearance of specific substances in the immediate vicinity of the particle or intracellular signals (Sahoo SK and Labhasetwar V., Nanotech approaches to drug delivery and imaging, 8, 1112-1120, 2003), (Nikitin, M. P. Shipunova, VO, Deyev, S. M. & Nikitin, PI, Nat. Nanotechnol., 9, 716-722, 2014).
  • Injectable controlled release nanoparticles can provide a pre-programmed duration of therapeutic action, ranging from days to several weeks from a single injection. They can also have significant advantages over conventional drugs, including automatic, patient compliance assurance, and drug targeting to specific tissues or organs.
  • the anticancer agent must release active substances or be activated in a controlled manner.
  • the ability to reach target tissues largely determines the effectiveness of drug use.
  • cytotoxic drugs can infect both malignant and normal cells.
  • a drug delivery system that specifically targets a toxic drug to malignant cells reduces toxicity to healthy cells.
  • Targeted delivery of nanoparticles can be passive or active.
  • Active delivery of the therapeutic agent is most often achieved by conjugating the therapeutic agent or carrier system with a tissue or cell-specific ligand, which can be, for example, a portion of an antibody or a linanda receptor including but not limited to transferrin, mannose, folate, EGFR, etc.
  • This effect is used for passive but relatively specific delivery of drugs encapsulated in nanoparticles or associated with macromolecules such as high molecular weight polymers or proteins (Maeda, H., Adv. Enzyme ReguL, 2001), (Sahoo, S. K. et al. , Bioconjug. Chem., 2002).
  • Nanoparticles can be functionalized to deliver drugs across biological barriers in the target area and to protect drugs from the biological environment elsewhere.
  • the immune response to viral vectors, or artificial agents that have been introduced into the body (subject, patient) throughout his life
  • the complete disappearance of the "effect" of prolonged circulation and rapid elimination of the agent from the bloodstream which significantly reduces the activity of the therapeutic activity of the agent.
  • the closest method is known (W02020016318), in which, in order to universally extend the circulation time of the agent and increase its therapeutic efficacy, in addition to the introduction of the agent, a protease is introduced, which cleaves the immunoglobulins of class G, separating the immunoglobulin Fc fragment from the antigen-binding fragment.
  • This strategy essentially neutralizes the immune response to the components of the agent, which leads to a significant decrease in the elimination of the agent from the bloodstream by the immune system.
  • this method eliminates only the problem of repeated administration of the agent, or the problem of administering the agent to an organism that already had antibodies to the components of the agent. Accordingly, when using an agent that itself has a short circulation time, this method will not be able to influence and increase the circulation time in any way.
  • the required technical result consists in increasing the circulation time of agents that deliver nucleic acids (polynucleotides), increasing the efficiency of delivery of polynucleotides by these agents to targets in the body, as well as reducing the toxicity and increasing the safety of such agents.
  • This invention discloses a universal method of dramatic (significant) increase in the circulation time of nanoparticles in the bloodstream for the delivery of genetic information to the body.
  • MPS mononuclear phagocyte system
  • anti-erythrocyte antibodies have already been approved by the FDA and are widely used in humans for the prevention of Rh syndrome during pregnancy and the treatment of immune thrombocytopenia (ITP).
  • Nanorobots and other new nanoprobes, actuators and nanoautomatics for delivering genes in line with the most daring ideas proposed in the field of materials science, can be introduced into research in the field of in vivo life sciences, and then rapidly improved for clinical use.
  • a method of treating a subject in need of treatment of a disease caused by a loss of function or activity of a protein comprising: (a) administering to said subject an antibody that specifically binds to blood cells; (B) administering to said subject an agent comprising a heterologous polynucleotide that encodes a protein or peptide that provides or complements the function or activity of the protein.
  • a method of treating a disease that is being treated with a gene therapy vector comprising administering to a subject in need thereof a therapeutically effective amount of an antibody that specifically binds to blood cells.
  • a method of treating a subject in need of treatment of a disease caused by an increase in functional activity or protein expression comprising: (a) administering to said subject an antibody that specifically binds to blood cells; (B) administering to said subject an agent comprising a heterologous polynucleotide that is transcribed into a nucleic acid that inhibits, decreases or decreases the expression of an enhancement of the function, activity, or expression of said protein.
  • An alternative method is to influence the body itself, namely the immune system, in particular, the mononuclear phagocytic system (MFS).
  • MFS mononuclear phagocytic system
  • blockade of the reticuloendothelial system mononuclear phagocytic system
  • these methods use the fact of saturation of the phagocytic activity of organisms when these substances are removed from the bloodstream. In this case, the prolongation of circulation in the bloodstream of nanoagents introduced simultaneously with these substances or after them is achieved.
  • the essence of this invention lies in a method for the delivery of gene therapy agents (carrying polynucleotides) by blocking the MFS with a small dose of the administered substance.
  • gene therapy agents carrier polynucleotides
  • antibodies that recognize blood cells are introduced into the body.
  • Such antibodies effectively block MFS due to specific binding to blood cells (or with certain populations of them), and inducing the elimination of these cells from the bloodstream by MFS cells.
  • an increase in efficiency may not necessarily be associated with an increase in circulation time.
  • the present invention helps to reduce the elimination of agents from the bloodstream by macrophages of MFS, these agents can bind just as quickly to their target.
  • the efficiency of delivery will increase due to the fact that the biodistribution of agents changes.
  • the present invention is well suited for repeated administration of the agent.
  • the agent is a virus, including one selected from the group consisting of: adenovirus, associated virus, lentivirus and the like; either a liposomal carrier of the polynucleotide, or a complex of the polynucleotide with various polymers, for example, polyethylene, cationic polymers and lipids, and the like.
  • Disclosed herein are methods for treating a subject in need of treatment for a disease caused by loss of function or activity of a protein, as well as methods for treating a subject in need of treatment for a disease caused by an increase in functional activity or expression of a protein, as well as compositions for performing such methods.
  • a method of treating a subject in need of treatment of a disease caused by loss of function or activity of a protein comprising: (a) administering to said subject an antibody that specifically binds to blood cells; (B) administering to said subject an agent comprising a heterologous polynucleotide that encodes a protein or peptide that provides or complements the function or activity of the protein.
  • a method of treating a subject in need of treatment of a disease caused by an increase in functional activity or protein expression comprising: (a) administering to said subject an antibody that specifically binds to blood cells; (B) administering to said subject an agent comprising a heterologous polynucleotide that is transcribed into a nucleic acid that inhibits, decreases or decreases the expression of an enhancement of the function, activity, or expression of said protein.
  • a method of treating a subject in need of treatment of a disease caused by loss of function or activity of a protein comprising: (a) administering to said subject a vector (or a recombinant viral vector or therapeutic agent) containing a heterologous a polynucleotide that encodes a protein or peptide that provides or complements the function or activity of the protein (here and throughout the endogenous said protein); and (b) administering to the subject an amount of anti-blood cell antibody (an antibody that specifically binds to blood cells) effective to block the mononuclear phagocytic system.
  • a vector or a recombinant viral vector or therapeutic agent
  • a method of treating a subject in need of treatment for a disease caused by loss of function or activity of a protein comprising: (a) administering to said subject a vector (or a recombinant viral vector or therapeutic agent) containing a heterologous polynucleotide that encodes a protein or peptide that provides or complements the function or activity of the protein; and (b) administering to said subject an amount of anti-blood cell antibody (an antibody that specifically binds to blood cells) effective to increase the circulation time of said vector.
  • a method of treating a subject in need of treatment for a disease caused by loss of function or activity of a protein comprising: (a) administering to said subject a vector (or a recombinant viral vector or therapeutic agent) containing a heterologous polynucleotide that encodes a protein or peptide that provides or complements the function or activity of the protein; and (b) administering to said subject an amount of anti-blood cell antibody (an antibody that specifically binds to blood cells) effective to increase the circulation time of said vector by causing blockage of the mononuclear phagocytic system.
  • a vector or a recombinant viral vector or therapeutic agent
  • a heterologous polynucleotide that encodes a protein or peptide that provides or complements the function or activity of the protein
  • an amount of anti-blood cell antibody an antibody that specifically binds to blood cells
  • a method of treating a subject in need of treatment of a disease caused by an increase in functional activity or protein expression comprising: (a) administering to said subject a vector (or a recombinant viral vector or therapeutic agent) containing a heterologous polynucleotide that is transcribed into a nucleic acid that inhibits, reduces or reduces the expression of enhancement of the function, activity or expression of the specified protein; and (b) administering to the subject an amount of anti-blood cell antibody (an antibody that specifically binds to blood cells) effective to block the mononuclear phagocytic system.
  • a vector or a recombinant viral vector or therapeutic agent
  • a method of treating a subject in need of treatment of a disease caused by an increase in functional activity or protein expression comprising: (a) administering to said subject a vector (or a recombinant viral vector or therapeutic agent) containing a heterologous polynucleotide that is transcribed into a nucleic acid that inhibits, reduces or reduces the expression of enhancement of the function, activity or expression of the specified protein; and (b) administering to said subject an amount of anti-blood cell antibody (an antibody that specifically binds to blood cells) effective to increase the circulation time of said vector.
  • a vector or a recombinant viral vector or therapeutic agent
  • a method of treating a subject in need of treatment of a disease caused by an increase in functional activity or protein expression comprising: (a) administering to said subject a vector (or a recombinant viral vector or therapeutic agent) containing a heterologous polynucleotide that is transcribed into a nucleic acid that inhibits, reduces or reduces the expression of enhancement of the function, activity or expression of the specified protein; and (b) administering to said subject an amount of anti-blood cell antibody (an antibody that specifically binds to blood cells) effective to increase the circulation time of said vector by causing blockage of the mononuclear phagocytic system.
  • a vector or a recombinant viral vector or therapeutic agent
  • step (b) is performed no earlier than 1 hour, more preferably 2 hours, more preferably 3 hours, more preferably 6 hours, more preferably 9 hours, more preferably 12 hours, or 24 hours, or 2 days, or 3 days after performing stage (a).
  • step (b) is performed no later than 1 hour, or 2 hours, or 3 hours, or 6 hours, or 9 hours, or 12 hours, or 24 hours, or 2 days, or 3 days after completing step (a).
  • a method further comprising determining a blockage state of the mononuclear phagocytic system by administering a test agent to a subject and analyzing the concentration of the test agent in a biological sample (e.g., blood, or a blood product, e.g., plasma, serum) after a predetermined time (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24 hours and more or after other intervals).
  • a biological sample e.g., blood, or a blood product, e.g., plasma, serum
  • a method further comprising determining a blockage state of the mononuclear phagocytic system by administering a test agent to a subject and analyzing the concentration of the test agent in a biological sample, for example, blood, or a blood product, for example, plasma, serum) after several different time intervals (for example, 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24 hours or through others intervals).
  • a biological sample for example, blood, or a blood product, for example, plasma, serum
  • a method in which the dose of said antibody is selected sufficient to increase the circulation time of said therapeutic agent in the bloodstream by at least 20%, preferably 30%, preferably 50%, preferably 100%, preferably 2.5 times, preferably 4 times, preferably 5 times, preferably 7 times, preferably 10 times, preferably 15 times, preferably 20 times, preferably 30 times (in comparison with the same time characteristic of circulation without administration of antibodies).
  • the method in which the specified antibody, when bound to the specified cells, induces (mediates) phagocytosis of these cells by cells of the mononuclear phagocytic system.
  • the specified antibody is a mixture of several monoclonal antibodies.
  • the specified antibody is an antibody expressed in cell culture.
  • a method in which the dose of said antibody is in the range of 0.1-1 mg / kg of the subject's weight. In addition, a method in which the dose of said antibody is in the range of 1-10 mg / kg of the subject.
  • a method in which the dose of said antibody is in the range of 10-100 mg / kg of the subject's body weight.
  • erythrocytes are further administered to a subject.
  • an erythropoiesis-stimulating substance is additionally administered to the subject.
  • erythropoietin is further administered to a subject.
  • said agent further comprises at least one of the following homopolymers or heteropolymers: linear polyethyleneimine, branched polyethyleneimine, polystyrene, carboxymethyldextran, dextran, polypeptide, polylactide glycolic acid, block copolymer, lipid, transport polymer , non-cationic lipid, cationic lipid.
  • said agent further comprises cytostatic, cytotoxic, or therapeutic compounds.
  • the method in which the said agent further includes a label from the group: fluorescent, magnetic, MPT-contrasting, X-ray contrast, luminescent, radioactive.
  • step (b) is performed concurrently with step (a).
  • step (b) is performed after step (a) is performed.
  • step (b) is performed no earlier than 6 hours after the step (a).
  • step (b) is performed within 3 days after step (a) is performed.
  • step (b) is performed within 24 hours after step (a) is performed.
  • step (b) is performed within 13 hours after step (a) is performed.
  • a method wherein said agent comprises a lentiviral vector, an adenoviral vector, or an adeno-associated virus vector.
  • said agent comprises an adeno-associated virus vector that comprises a VP1, VP2 and / or VP3 capsid protein having 50% or more sequence identity to a VP1, VP2 and / or VP3 capsid protein selected from the group consisting of AAV1, AAV2, AAV3, AAV4, AAV5, AAV6, AAV7, AAV8, AAV9, AAV10, AAV3B, AAV-2i8, RhlO, Rh74; 2 capsid proteins VP1, VP2 and / or VP3,
  • said agent comprises an adeno-associated virus vector that contains a VP1, VP2 and / or VP3 capsid protein having 100% sequence identity to a VP 1, VP2 and / or VP3 capsid protein selected from the group consisting of AAV1 ... , AAV2, AAV3, AAV4, AAV5, AAV6, AAV7, AAV8, AAV9, AAV10, AAV3B, AAV-2i8, RhlO, Rh74, 2 capsid proteins VP1, VP2 and / or VP3.
  • a method further comprising determining a blockage state of a mononuclear phagocytic system by administering a test agent to a subject and analyzing the concentration of the test agent in a biological sample after a predetermined time.
  • the method further comprising determining the blockage state of the mononuclear phagocytic system by comparing the rate of elimination of the test agent before step (a) and the elimination efficiency of the test agent after step (a), but before step (b).
  • a method in which the rate of elimination of a test agent is determined by analyzing the concentration of the test agent in a biological sample after a predetermined time.
  • the method further comprising: before step (a) the introduction of the test agent to the subject and analysis of the concentration of 1 test agent in the biological sample after a predetermined time, after step (a) the introduction of the test agent to the subject and analysis of the concentration 2 of the test agent in the biological sample after time, comparison of concentration 1 and concentration 2.
  • test agent includes a nanoparticle.
  • test agent includes a liposome
  • test agent is fluorescently labeled.
  • the test agent includes a magnetic label.
  • the method in which the specified subject has a disease from the group: a disease of organs (eg, brain, liver, kidney, heart); viral infectious disease (for example, hepatitis B, hepatitis C, HIV, etc.), bacterial or fungal nature; crayfish; type 1 or type 2 diabetes; Alzheimer's disease; Parkinson's disease; epilepsy; a circulatory system disorder (eg, anemia); lung disease (eg, cystic fibrosis); a bleeding disorder (for example, hemophilia A or hemophilia B with or without inhibitors); metabolic disorders (eg, glycogen storage disease); a neurological or neurodegenerative disorder; deficiency of lysosomal acid lipase; deficiency of adenosine deaminase; violation of the accumulation of copper or iron (for example, Wilson's disease, Menkes disease); hereditary angioedema; thalassemia; Huntington
  • the method in which the specified subject has a bleeding disorder of the group: hemophilia A; hemophilia A with inhibitory antibodies; hemophilia B; hemophilia B with inhibitory antibodies; thalassemia; deficiency of any of the blood clotting factors: II, V, VII, VIII, IX, X, XI, XII, von Willebrand factor or combined FV / FVIII deficiency; deficiency of gamma carboxylase or deficiency of vitamin K epoxide reductase C1.
  • a heterologous polynucleotide encodes a protein selected from the group consisting of: insulin, glucagon, parathyroid hormone (PTH), growth hormone (GH), follicle stimulating hormone (FSH), connective tissue growth factor (CTGF), factor vascular endothelial growth (VEGF), nerve growth factor (NGF), brain neurotrophic factor (NTFGM), epidermal growth factor (EGF), growth hormone release factor (VGF), angiopoietins, erythropoietin (EPO), angiostatin, bone morphogenetic protein ( BMI), luteinizing hormone (LH), human chorionic gonadotropin (hCG), granulocyte colony-stimulating factor (GCSF), basic fibroblast growth factor (bFGF), acidic fibroblast growth factor (cGF), transforming growth factor a (TGF), thrombotic growth factor (TFR), insulin growth factors I and II (FRI-1 and FRI-P), TFR,
  • PTH parathyroid
  • a heterologous polynucleotide encodes a protein selected from the group consisting of: interferons a, b and g, stem cell factor, humanized antibodies, IgG, IgM, IgA, IgD and IgE, chimeric immunoglobulins, thrombopoietin (TP) , single-chain antibodies, interleukins (IL-1 to IL-36), monocytic chemoattractant protein, chimeric T-cell receptors, single-chain T-cell receptors, leukemia inhibitory factor, granulocyte-monocyte colony-stimulating factor, tumor ligand Fas, tumor necrosis factors and b, flk-2 / flt3 ligand, T cell receptors, class I and class II MHC molecules, alpha-galactosidase for the treatment of Fabry disease; glucose-6-phosphatase for the treatment of glycogen storage disease type I (GSDI); GSDI
  • heterologous polynucleotide is a small interfering RNA.
  • heterologous polynucleotide is microRNA
  • the method in which the protein encoded by the heterologous polynucleotide contains a gene editing nuclease contains a gene editing nuclease.
  • the gene editing nuclease comprises zinc finger nuclease (ZFN) or an effector transcription activator-like nuclease (TALEN).
  • step (a) and / or step (b) are performed two or more times.
  • a method in which a disease is treated with a gene therapy vector and comprising administering to a subject in need thereof a therapeutically effective amount to the subject of an antibody that specifically binds to blood cells comprising administering to a subject in need thereof a therapeutically effective amount to the subject of an antibody that specifically binds to blood cells.
  • heterologous polynucleotide encodes an inhibitory nucleic acid
  • the specified inhibitory nucleic acid is selected from the group consisting of mRNA, antisense molecule, microRNA, interfering RNA, ribozyme.
  • inhibitory nucleic acid binds to a gene, gene transcript or gene transcript associated with a disease with a polynucleotide repeat.
  • composition for use in a method of treatment comprising said antibodies.
  • composition for use in a method of treatment comprising said antibodies and said agent.
  • composition for use in a method of treatment comprising said antibodies and said agent in one or separate dosage forms.
  • a package which contains: (a) an agent containing a heterologous polynucleotide that encodes a protein or peptide; (b) an antibody that specifically binds to blood cells; and (c) a label with instructions for performing the specified method, in which (a) and (b) are in a separate or the same container.
  • a package which contains: (a) an agent containing a heterologous polynucleotide, which is transcribed into a nucleic acid that inhibits, reduces or reduces the expression of the protein; (b) an antibody that specifically binds to blood cells; and (c) a label with instructions for performing the specified method, in which (a) and (b) are in a separate or the same container.
  • the package (kit, kit), which contains: (a) the specified agent; (b) the specified antibody; and (c) a label with instructions for performing the specified method, in which (a) and (b) are in a separate or the same container.
  • compositions used in the methods described herein for example, compositions containing said antibody, or a composition containing both said antibody and said agent.
  • a method of promoting a product to the market including at least promoting, for the treatment of diseases or conducting scientific research, at least the specified method or the specified composition.
  • a method of promoting a product to the market in which promotion is carried out by an insert in a package with a commercial composition or a substance (or substances) containing at least one of the specified compositions.
  • a business method that includes at least marketing for gene therapy or research or for the delivery of polynucleotides or nucleic acids to targets in the body, or for increasing the circulation time of polynucleotide delivery agents in the body's bloodstream, or for treating or to increase the effectiveness of the treatment of an agent of one or more of said composition or method.
  • lipid includes any chemical compound that is hydrophobic (including cholesterol and its derivatives, or, for example, long-chain saturated or unsaturated aliphatic hydrocarbon chains or those chains substituted with one or more aromatic, cycloaliphatic or heterocyclic groups) and hydrophilic parts (usually phosphate, carboxyl, sulfate, amino, sulfihydryl, nitro and other similar groups) and, for example, allowing chemicals to be incorporated into a micelle or liposome.
  • hydrophobic including cholesterol and its derivatives, or, for example, long-chain saturated or unsaturated aliphatic hydrocarbon chains or those chains substituted with one or more aromatic, cycloaliphatic or heterocyclic groups
  • hydrophilic parts usually phosphate, carboxyl, sulfate, amino, sulfihydryl, nitro and other similar groups
  • non-cationic lipid refers to any of the lipids that exist in either uncharged form, neutral (zwitterionic) or anionic form at physiological pH.
  • lipids include, for example, diacylphosphatidylcholine, diacylphosphatidylethanolamine, ceramide, sphingomyelin, cephalin, cardiolipin, cerebrosides, DOPE, and cholesterol.
  • cationic lipid refers to any of the lipids that carry a positive charge.
  • Such lipids include, but are not limited to, compounds of the type DODAC, DOTMA, DDAB, DOSPER, DOSPA, DOT AP, DC-Choi and DMRIE, as well as commercially available preparations based on them.
  • transport polymer includes polymers of various structures, including linear and branched.
  • pharmaceutically acceptable component such as a salt, carrier, excipient or diluent
  • pharmaceutical agent delivery composition of the present invention is a component that is compatible with the other ingredients of the delivery composition and is suitable for use in animals (including humans) without severe side effects (toxicity, inflammation and allergic reaction). Side effects are considered “severe” when the risk of their occurrence outweighs the benefit of the pharmaceutical agent.
  • transfect or "transgene” is widely used to mean an exogenous compound, such as a polynucleotide sequence, nucleic acid, CRISPR / Cas9 genomic editing system, etc., introduced into a prokaryotic or eukaryotic cell.
  • the nucleic acids are preferably associated with additional molecular components (promoters) that control the expression of the nucleic acid in the target cell population.
  • the expression vector may include inducible or non-inducible promoters upstream of the expressed DNA. Examples of inducible promoters include the Tet promoters, ecdysone, or the steroid-metallothionine.
  • non-inducible promoters examples include long terminal repeat (LTR), simian virus, phosphoglycerate kinase (PGK), b-actin, or cytomegalovirus (CMV) promoters.
  • promoters can allow general (CMV, PGK) or specific expression (eg, alpha-fetoprotein, tyrosinase) of the coding DNA. Delivery systems can contain multiple promoters to enhance transfection.
  • a DNA vector can include an IRES (internal ribosome entry site) between different coding DNA sequences, allowing for translation of more than one polypeptide from the same transcript.
  • IRES internal ribosome entry site
  • composition of a nucleic acid for expression in gene therapy is widely described in the literature and is not the subject of this invention.
  • these terms in the present invention mean a nucleic acid that is intended or has been introduced into a cell or organism.
  • heterologous polynucleotide sequences or heterologous nucleic acids encoding a functional protein or peptide can be used.
  • transgenic and heterologous nucleic acid / polynucleotide sequences are used interchangeably in this invention.
  • in vivo includes injection-based therapy (eg, intratumoral, intramuscular, subcutaneous, intratracheal, intravenous, direct injection into an organ or respiratory tract, injection into organ vessels, injection into the abdominal cavity, intracardiac injection or airway aerosol) into the living organism of animals and humans.
  • injection-based therapy eg, intratumoral, intramuscular, subcutaneous, intratracheal, intravenous, direct injection into an organ or respiratory tract, injection into organ vessels, injection into the abdominal cavity, intracardiac injection or airway aerosol
  • in vivo also includes therapy based on electroporation of a tumor, tissue or organ.
  • composition for delivery of a pharmaceutical agent in accordance with this invention contains any combination of two or more of the listed components: a pharmaceutical agent (including those based on proteins, peptides, nucleic acids, antisense oligonucleotides, ribozymes, DNA and RNA oligonucleotides, a chemotherapeutic anticancer agent, a chemotherapeutic agent disease, diagnostic agent, and any combination of two or more of the above.
  • a pharmaceutical agent including those based on proteins, peptides, nucleic acids, antisense oligonucleotides, ribozymes, DNA and RNA oligonucleotides, a chemotherapeutic anticancer agent, a chemotherapeutic agent disease, diagnostic agent, and any combination of two or more of the above.
  • recombinant vector AAV means that the vector has been subjected to some influence or designed in a way that is not usually found in nature.
  • a specific example of a recombinant AAV vector would be when a nucleic acid that is not normally present in the wild-type AAV genome (heterologous polynucleotide) is inserted into the viral genome.
  • serotype in relation to different viral vectors denotes the composition and structure of its capsid, which are serologically different (ie, there is no cross-reactivity between antibodies to one of the viral strains) from viruses of other serotypes. Differences in cross-reactivity are usually due to differences in the sequences of capsid proteins or antigenic determinants on the surface of the viral vector.
  • patient and “subject” interchangeably refer to an animal, usually a mammal, such as a rodent, cat, dog, and primate. In a particular case, under the subject or patient, according to the present invention, a person can be designated.
  • the pharmaceutical agent suitably contains a nucleic acid such as DNA or RNA.
  • the nucleic acid is preferably associated with control elements for the expression of the nucleic acid in the target cell population.
  • the expression vector may include inducible or non-inducible promoters upstream of the expressed DNA. Examples of inducible promoters include the Tet promoters, ecdysone, or the steroid metal otionine. Examples of constitutive non-inducible promoters include long terminal repeat (LTR), simian virus, phosphoglycerate kinase (PGK), b-actin, or cytomegalovirus (CMV) promoters.
  • LTR long terminal repeat
  • PGK phosphoglycerate kinase
  • CMV cytomegalovirus
  • promoters allow general (CMV, PGK) or specific expression (eg, alpha-fetoprotein, tyrosinase) of the coding DNA.
  • Virus or plasmid based delivery systems can contain multiple promoters to enhance transfection.
  • the vector DNA can include an IRES (internal ribosome entry site) between different coding DNA sequences, allowing for translation of more than one polypeptide from the same transcript.
  • the plasmid or virus can also express a pharmaceutical agent.
  • the composition of a nucleic acid for expression in gene therapy is widely described in the literature.
  • the cell to be treated can include any cell of animal, plant, or bacterial origin that is sensitive to intracellular delivery of a pharmaceutical agent using a delivery composition, either in vivo or in vitro.
  • suitable cellular targets include, but are not limited to, epithelial cells, endothelial cells, keratinocytes, fibroblasts, muscle cells, hepatocytes; blood cells such as T-lymphocytes, B-lymphocytes, monocytes, macrophages, neutrophils, eosinophils, megakaryocytes, granulocytes; various stem cells or progenitor cells, in particular hematopoietic stem cells or progenitor cells, for example, derived from bone marrow, umbilical cord blood, peripheral blood, fetal liver and the like.
  • the cell is selected from the group consisting of lung cells, liver cells, endothelial cells, muscle cells, skin cells.
  • polypeptides are used interchangeably in this invention and may be used interchangeably.
  • Polypeptides can be either full-length native sequences or their separate functional fragments, modified forms or sequence variants, if they retain certain functionality of the original full-length protein.
  • such peptides, proteins and polypeptides encoded by polynucleotide sequences may optionally be identical to an endogenous protein that is defective or insufficient or insufficient in expression in the treated mammal.
  • Effective amount or “sufficient amount” means that amount of a drug that provides (with single or multiple administration, alone or in combination with one or more other compositions, treatments, protocols or agents) the expected or desired effect of any duration in the subject. , to any measurable degree.
  • a detectable or measurable improvement includes a subjective or objective decrease, decrease, inhibition, suppression, limitation or control of the occurrence, frequency, the severity, progression, or duration of the disease or complication caused by the disease.
  • said antibodies that specifically bind to blood cells may be antibodies that recognize antigenic determinants, receptors, cell surface markers of various cellular subsets.
  • said antibodies can recognize and bind with high or low affinity and / or avidity to leukocytes, erythrocytes, platelets, lymphocytes, T cells, B cells, etc.
  • the antibody recognizes and binds said cell receptors using its antigen-binding site.
  • the mentioned cellular receptor can be present only on a given subpopulation of blood cells, and also on various other cells of the body.
  • antibodies are used against the CD47 receptor, which is actively expressed on the membrane of erythrocytes, as well as many other cells, and is responsible for recognition and identification of “self” cells by macrophages and other phagocytic cells (friend or foe signaling).
  • an antibody full-length or only an antigen-recognition fragment, for example, Fab or Fab2 fragment
  • SIRPa this receptor or its ligand
  • the specificity of an antibody to a specific receptor, or a specific cellular subpopulation, or a cell in general, is determined by the usual traditional considerations and approaches of cell and molecular biology.
  • the specificity of antibodies is measured using cygometric, microscopic, or enzyme-linked immunosorbent assays to calculate the amount of antibodies bound to a cell.
  • antibodies are considered to be specifically binding, which bind to a given subpopulation of more than 20 molecules per cell, preferably 30 molecules per cell, preferably 50 molecules per cell, preferably 100 molecules per cell, preferably 200 molecules per cell, preferably 300 molecules per cell preferably 400 molecules per cell, preferably 500 molecules per cell, preferably 600 molecules per cell, preferably 700 molecules per cell, preferably 1000 molecules per cell.
  • antibodies are selected that have the desired properties of the effector functions of the Fc fragment.
  • antibodies are used that have a potent activity in inducing Fc receptor dependent phagocytosis.
  • an antibody is used that has an affinity for one or more types of Fc receptor, for example, FcyRI (CD64), FcyRIIA (CD32), FcyRIIB (CD32), FcyRIIIA (CD16a), FcyRIIIB (CD 16b), FcaRI (or CD89), FceRI, FceRII
  • the methods use antibodies with a receptor that provides minimal or no complement activation when complexed with said cells.
  • antibodies are used that cause minimal or no aggregation of blood cells, for example, agglutination of red blood cells.
  • either antibodies of the same isotype or a mixture of antibodies of different isotypes against one or different epitopes of said cells are used.
  • autologous antibodies and / or allogeneic antibodies isolated from the blood of donors eg, anti-Rh antibodies
  • xenogeneic or a mixture of xenogeneic antibodies against blood cells and allogeneic antibodies that recognize said xenogeneic antibodies are used.
  • anti-erythrocyte antibodies in particular, anti-RhD
  • ITP immune thrombocytopenia
  • RhD Rh- diseases in humans.
  • ITP immune thrombocytopenia
  • anti-RhD antibodies are used.
  • said antibodies against blood cells are isolated from material from patients with AICA, and in this way a more efficient repertoire of antibodies can be obtained.
  • the selection of suitable clones of antibodies should be based on safety considerations.
  • Test parameters can include parameters, concerning the health of red blood cells (lysis and deposition in tissues), as well as the pro-inflammatory response (level of cytokines in the blood, edema, thrombosis, renal dysfunction and pulmonary function).
  • MPS cytoblockade may further intensify these efforts due to the ability of the method to significantly expand the capabilities of nanoparticle medicine for the treatment of the many diseases that require targeted delivery of drugs and genetic constructs.
  • MPS-cytoblockade is not a mutually exclusive alternative to stele-coatings, but represents a new strategy that can be synergistically combined with other approaches to prolong the circulation of nanoparticles.
  • Rozrolimupab a mixture of 25 anti-red blood cell monoclonal antibodies developed for the treatment of immune thrombocytopenia.
  • said method includes the additional step of introducing blood or its various components into the body.
  • blood or its various components can be cells, or subpopulations of cells, against which the specified antibody has been targeted.
  • it can be whole blood, including donated blood.
  • This step is useful for minimizing or eliminating the side effects of this delivery method (therapy), for example, leveling and normalizing the hematocrit when using antibodies that specifically bind to erythrocytes (and cause blockage of MFS due to increased rhytrophagocytosis).
  • an additional step can be used to introduce various substances used to therapeutically increase the level of these cell subpopulations.
  • it can be erythropoietin, which causes the active production of red blood cells in the body.
  • said antibodies are monoclonal antibodies, polyclonal antibodies, a mixture of various monoclonal and or polyclonal antibodies, full-length antibodies, antibody fragments, peptides having the functionality described herein for said antibodies.
  • the specified antibody can be isolated from blood, or obtained in cell culture, obtained in bacterial culture, obtained in eukaryotic culture, obtained using immunization technologies, hybridomas, phage display, and the like.
  • the antibody can be a humanized antibody, or an antibody with an altered sequence to reduce the immunogenicity of the antibody in the body, or otherwise modify the antibody, such as altering the effector function of the antibody to obtain the desired characteristics (see above).
  • said blood, blood, or substance components may be formulated in compositions used in said methods. Moreover, they can be in the same dosage form as the agent and / or antibody, or in separate dosage forms.
  • the agent is a viral delivery system.
  • Viral systems include nucleic acid delivery systems (NK), in which various mammalian viruses and constructs based on them are used as a transport system.
  • NK nucleic acid delivery systems
  • Gammaretroviruses, lentiviruses, adeno- and adeno-associated viruses, herpes simplex viruses, rhinoviruses, etc. can be used as such transport constructs (vectors) (Herzog et al., 2010).
  • the main directions of improving methods based on this group of NK delivery systems are regulation of their specificity and efficiency of penetration into various types of cells, increasing stability and safety, primarily to eliminate the risk of cancer and other virus-associated diseases.
  • the surface glycoprotein is modified by replacing it with glycoprotein G from the vesicular stomatitis virus or its chemical binding to proteins that specifically recognize certain types of cells (Vannucci et al., 2013).
  • adenoviruses whose genomic DNA does not integrate into the host's DNA, but coexists in the nucleus in the form of episomes (Choi & Yun, 2013), non-pathogenic adeno-associated viruses ( Mingozzi & High, 2011) or inactivated herpes simplex virus (Manservigi et al., 2010).
  • NK delivery vehicles capable of penetrating the main physiological barriers, including the blood-brain barrier, and therefore promising for the therapy of diseases of the central nervous system are described (Duque et al., 2009; Foust et al.
  • an increase in the efficiency of transfection using viral vectors is achieved by a combination of viral and non-viral delivery systems, for example, for the treatment of cystic fibrosis (Marshall et al., 1994), small cell lung cancer (Waelti & Gliick, 1998), delivery of DNA to neurons of the brain (Wu et al., 1996) and others.
  • the above methods of viral DNA delivery use efficient mechanisms of penetration into cells, which have been refined in the process of evolution. Nevertheless, a number of limitations inherent in viral systems (the difficulty of industrial scaling, limitations on the size of the transfect, potential immunotoxicity) necessitate the search for new delivery systems that are free of these disadvantages.
  • a big problem when using viral vectors is the response of the adaptive immune system of the body, which consists in the production of antigen-specific immunoglobulin (eg, IgG) antibodies, leading to inhibition or clearance of the delivery system.
  • immunoglobulin eg, IgG
  • animals and humans can naturally be exposed to wild-type viruses, gene therapy with such viruses can be hindered by the presence of pre-existing antibodies (Calcedo & Wilson, 2013) and may lead to the exclusion of certain subjects from trials or gene therapy.
  • the emergence of antibodies against carrier viruses after primary transfection can dramatically impair the efficiency of re-transfection using the same or cross-reactive vectors (Boutin et al., 2010).
  • the agent is a non-viral delivery system.
  • the agent may include nanoparticles that mimic high or low density lipoproteins, which are made on the basis of lipoprotein proteins Apo-A1, ApoB-100, Apo-I, Apo E (Amin, AR, & Amin, N.K. , 2018), (McMahon, K. M. et al., Cancer Treat. Res., 2015).
  • Lipoproteins are carriers of hydrophobic substances in the body, and thus are ideal for the transport of hydrophobic drugs as well as hydrophilic molecules such as nucleic acids modified by hydrophobic molecules. Like albumin, lipoproteins are taken up by cells through endocytosis.
  • Endocytosis modulating receptors SR-B1 are overexpressed on the surface of cancer cells, such as breast cancer. Also, modification of nanoparticles with folic acid inhibits binding to SR-B 1 and induces binding to FR-a of ovarian cancer overexpressed on cells (Corbin, I. R., Methods in molecular biology, 2013.).
  • minicircular DNA vectors are used (Gill et al., 2009; Mayrhofer et al., 2009).
  • Such structures in contrast to DNA plasmids of bacterial origin, have significantly fewer immunogenic regions and can effectively avoid destruction by the intracellular immunity system.
  • a DNA vector is formed by optimizing the components of the expression cassette (Lu et al., 2013) or by attaching to a special carrier (scaffold) that allows the vector episomal construct to replicate along with the host genomic DNA during cell division (Argyros et al., 2011), or in the presence of helper molecules that facilitate the transmembrane transport of the vector construct - for example, CPP peptides, Cell-Penetrating Peptides (Ramakrishna et al., 2014), streptococcal SLO toxin (Walev et al., 2001), or calcium phosphate molecules (Ebina et al., 2013).
  • helper molecules that facilitate the transmembrane transport of the vector construct - for example, CPP peptides, Cell-Penetrating Peptides (Ramakrishna et al., 2014), streptococcal SLO toxin (Walev et al., 2001), or calcium
  • transfection methods based on cationic lipid (liposomes) or polymer constructs are used (Jin et al., 2014).
  • cationic liposomes are the most commonly used non-viral transfection system in vitro (P. L. Feigner et al., 1987; Remy et al., 1994) and in vivo (Nabel et al., 1993; Zhu et al., 1993).
  • the relatively low stability of liposomes to the action of physiological factors leads to a relatively low efficiency of transfection (Marshall, M., 1995) and determines the need to search for new methods to increase the efficiency liposomal transfection.
  • physiological factors for example, blood components
  • the components for the synthesis of liposomes of a new generation are used as described in the literature (J. H. Feigner et al., 1994; Y.
  • polymeric systems for delivery of nucleic acids are used.
  • they are created by the method of condensing DNA molecules on polycations such as polyethyleneimine, polylysine, lipopolyamine, dendrimers based on polyamidoamines, which act as an effective buffer system that suppresses the destructive action of lysosomes and facilitates the subsequent intracellular release of the transfect.
  • the transfect delivery is carried out using various particulate carriers with a high specific surface area, which may include nanoparticles of noble metals (primarily gold), condensation products of nucleic acids to form, for example, "nanoclub” (Sun et al. , 2014, 2015), mesoporous microspheres of silicon dioxide, carbon nanotubes.
  • nanoparticles of noble metals primarily gold
  • condensation products of nucleic acids to form for example, "nanoclub” (Sun et al. , 2014, 2015), mesoporous microspheres of silicon dioxide, carbon nanotubes.
  • the nucleic acids of the present invention can be obtained by any currently known method, including, but not limited to, chemical, biological, genetic, or enzymatic synthesis methods, and arbitrary combinations thereof.
  • the heterologous polynucleotide encodes a protein selected from the group consisting of: interferons a, b, and g, stem cell factor, humanized antibodies, IgG, IgM, IgA, IgD and IgE, chimeric immunoglobulins, thrombopoietin (TPO), single chain antibodies , interleukins (from IL-1 to IL-36), monocyte chemoattractant protein, chimeric T-cell receptors, single-chain T-cell receptors, leukemia-inhibiting factor, granulocygar-monocyte colony-stimulating factor, ligand Fas, tumor necrosis factors a and b, flk-2 / flt3 ligand, T cell receptors, MHC class I and class P molecules, alpha-galactosidase for the treatment of Fabry disease; glucose-6-phosphatase for the treatment of storage disease glycogen type I (GSDI);
  • the heterologous polynucleotide encodes an inhibitory nucleic acid.
  • the inhibitory nucleic acid is selected from the group consisting of: small RNA, small DNA, small interfering RNA, microRNA, ribozyme, insensitive molecule, RNA interference.
  • the inhibitory nucleic acid binds to a gene, gene transcript, or gene transcript associated with a polynucleotide repeat disease selected from the group consisting of an androgen receptor on the X chromosome in spinbulbar muscle atrophy; a gene associated with dentorubral atrophy; human voltage-gated calcium channel; TATA binding protein; Ataxin 8 opposite chain; beta isoforms of the regulatory subunit B of serine / threonine protein phosphatase 2A in spinocerebellar ataxia; gene Huntington; apolipoprotein B and proprotein convertase, subtilisin / kexin type 9, HIV-TAR, the gene for the transactivator response element of the human immunodeficiency virus in HIV infection; C-C chemokine receptor in HIV infection; HIV-Tat, the gene for the transcriptional transcription of the human immunodeficiency virus in HIV infection; unstable X-oligophrenia 1 with unstable X-syndrome and X-
  • the administration of said antibody causes blocking (saturation) of the phagocytic ability of the MFS and, as a consequence, decreases the rate of elimination of agents from the bloodstream (from the plasma compartment, i.e. absorbed by the cell), which is expressed in an increase in the circulation of nanoscale (supramolecular) agents.
  • the fact of blocking is confirmed directly during therapy between the administration of the antibody and the agent, by means of the administration of a test agent, according to the registration of the pharmacokinetics of which (according to any standard quantitative characteristic, for example, according to concentration after a fixed period of time, for example, 10, 20 , 30, 45, 60 minutes and any other interval) assess the achievement of the state of blocking the MFS.
  • this step may not be necessary to minimize the amount of drugs administered to the patient.
  • One aspect of the present invention is a method for delivering a pharmaceutical (therapeutic) agent into a cell.
  • the delivery of a pharmaceutical agent can be carried out in vitro into a cell, ex vivo, and in vivo into a cell and / or into a tissue of an organism.
  • the target cell for the therapy of this invention can be any cell of animal, plant, or bacterial origin that is responsive to delivery of a pharmaceutical agent using an in vivo or in vitro delivery composition.
  • suitable cellular targets include, but are not limited to, lung, liver, endothelial and muscle cells, skin cells, epithelial and endothelial cells, keratinocytes, fibroblasts, muscle cells, hepatocytes; blood cells such as T-lymphocytes, B-lymphocytes, monocytes, macrophages, neutrophils, eosinophils, megakaryocytes, granulocytes; various stem cells or progenitor cells, in particular hematopoietic stem cells or progenitor cells, including those derived from bone marrow, umbilical cord blood, peripheral blood, fetal liver, etc.
  • diseases and disorders can be treated with the methods of the present invention, including, but not limited to: diseases of organs (eg, brain, liver, kidney, heart); viral infectious disease (for example, hepatitis B, hepatitis C, HIV, etc.), bacterial or fungal nature; crayfish; type 1 or type 2 diabetes; Alzheimer's disease; Parkinson's disease; epilepsy; a circulatory system disorder (eg, anemia); lung disease (eg, cystic fibrosis); a bleeding disorder (for example, hemophilia A or hemophilia B with inhibitors or without them); metabolic disorders (eg, glycogen storage disease); a neurological or neurodegenerative disorder; deficiency of lysosomal acid lipase; deficiency of adenosine deaminase; violation of the accumulation of copper or iron (for example, Wilson's disease, Menkes disease); hereditary angioedema; thalassemia; Huntington's disease; amyotrophic lateral lateral
  • Various diseases and disorders can be treated with the methods of the present invention, including, but not limited to, bleeding disorders: hemophilia A; hemophilia A with inhibitory antibodies; hemophilia B; hemophilia B with inhibitory antibodies; thalassemia; deficiency of any of the blood clotting factors: II, V, VII, VIII, IX, X, XI, XII, von Willebrand factor or combined FV / FVIII deficiency; deficiency of gamma carboxylase or deficiency of vitamin K epoxide reductase C1.
  • Various diseases and disorders can be treated with the methods of the present invention, including, but not limited to: anemia; bleeding associated with stroke, tissue damage, trauma, thrombosis, thrombocytopenia, coagulopathy, disseminated intravascular coagulation (DIC); excessive anticoagulation associated with heparin, low molecular weight heparin, warfarin, low molecular weight antithrombotic agents (i.e., FXa inhibitors), pentasaccharide, platelet disorders such as Bernard Soulier's syndrome, Glanzmann's thrombasthenia or platelet pool deficiency.
  • anemia bleeding associated with stroke, tissue damage, trauma, thrombosis, thrombocytopenia, coagulopathy, disseminated intravascular coagulation (DIC); excessive anticoagulation associated with heparin, low molecular weight heparin, warfarin, low molecular weight antithrombotic agents (i.e., FXa inhibitors), pentasaccharide, plate
  • the disease is a neurodegenerative disease.
  • the neurodegenerative disease or CNS disease is Parkinson's disease, Alzheimer's disease, spinal muscular atrophy, Huntington's disease, primary lateral sclerosis, amyotrophic lateral sclerosis, hereditary spastic hemiplegia, Kennedy's disease.
  • the heterologous polynucleotide encodes a protein selected from the group consisting of: insulin, glucagon, parathyroid hormone (PTH), growth hormone (GH), follicle stimulating hormone (FSH), connective tissue growth factor (CTGF), vascular endothelial growth factor (VEGF), nerve growth factor (NGF), brain neurotrophic factor (NTFGM), epidermal growth factor (EGF), growth hormone release factor (HRGF), angiopoietins, erythropoietin (EPO), angiostatin, bone morphogenetic protein (BMP), luteinizing hormone (LH), human chorionic gonadotropin (hCG), granulocyte colony-stimulating factor (G-CSF), basic fibroblast growth factor (bFGF), acid fibroblast growth factor (cGF), transforming growth factor a (TGFa), platelet-derived growth factor (TGF) , insulin growth factors I and II (FRI-1 and FRI-P), TGF,
  • PTH
  • Various diseases and disorders can be treated with the methods of the present invention, including, but not limited to: proliferative diseases (cancer, tumors); diabetes types 1 and 2; organ diseases (eg heart, kidney, liver, brain); cardiovascular diseases (restenosis, ischemia, dyslipidemia, homozygous familial hypercholesterolemia, etc.); viral (for example, hepatitis B and C, HIV, etc.), bacterial or fungal infections; neurological or neurodegenerative disorders (eg, Parkinson's disease, Alzheimer's disease, Huntington's disease, ALS, hereditary spastic hemiplegia, primary lateral sclerosis, spinal muscle atrophy, Kennedy's disease); epilepsy; eye diseases (retinitis pigmentosa, Leber's congenital amaurosis, Leber's hereditary visual neuropathy and Stargardt's disease); metabolic disorders (for example, glycogen storage disease); lung disease (eg cystic fibrosis); disorders of the transport of metals (for example, Wilson's
  • the invention provides a method of treating a disease in a patient in need of such treatment, the method comprising administering to the patient an effective amount of any of the compositions described herein (including embodiments).
  • the composition in this case should be considered both the specified agent and the specified antibody - in separate dosage forms or a single one.
  • composition for delivery of a pharmaceutical agent in accordance with this invention may contain any combination of two or more of the listed components: a pharmaceutical agent (including those based on proteins, peptides, nucleic acids and oligonucleotides, antisense oligonucleotides, ribozymes, etc.), chemotherapeutic anticancer agent, chemotherapy drug, and diagnostic component.
  • a pharmaceutical agent including those based on proteins, peptides, nucleic acids and oligonucleotides, antisense oligonucleotides, ribozymes, etc.
  • chemotherapeutic anticancer agent including those based on proteins, peptides, nucleic acids and oligonucleotides, antisense oligonucleotides, ribozymes, etc.
  • chemotherapy drug including those based on proteins, peptides, nucleic acids and oligonucleotides, antisense oligonucleotides, ribozymes, etc
  • the dose of the administered drug according to this invention may vary depending on the type of disease, the severity of its initial stage, progression, severity, frequency, duration and likelihood of complications, the desired result of treatment, previous or simultaneous treatment, general health, age, sex, immunological status subject and other factors, the assessment of which should be carried out by qualified specialists. Depending on their decision based on the analysis of factors such as adverse side effects, complications or other risk factors, the frequency and duration of the dose may be changed. The person skilled in the art will understand the factors that can influence the dosage and timing required to achieve the expected effect of a therapeutic drug.
  • the effective dose of the drug can vary depending on several factors, including, but not limited to: the method of administration; the level of expression of the heterologous polynucleotide required to achieve a therapeutic effect; the nature of the disease; the severity of the host's immune response to the vector used, to the heterologous polynucleotide or its expression product (protein or peptide), etc.
  • a person skilled in the art can independently determine the range of the drug to be administered based on his own knowledge of a particular disease or disorder.
  • compositions of the invention may be administered alone or may be coadministered to a patient. This includes the simultaneous or sequential administration of the compositions individually or in combination (more than one composition).
  • the drugs can also be combined, when desired, with other active substances (for example, to reduce metabolic degradation).
  • the compositions described herein can be used in combination with each other, with other active agents known to be useful in treating a disease, or with additional agents that may be ineffective on their own, but may contribute to the effectiveness of the agent or antibody.
  • compositions of the present invention can also be administered by injection, that is, intravenously, intramuscularly, intradermally, subcutaneously, rectally, or intraperitoneally.
  • the compositions described herein can be administered by inhalation, for example, intra-nasally.
  • the compositions of the present invention can be administered transdermally as well as eye drops. It is also contemplated that multiple routes of administration (eg, intramuscular, oral, transdermal) can be used to administer the compositions described herein (including embodiments).
  • the present invention also relates to pharmaceutical compositions comprising a pharmaceutically acceptable excipient and one or more compositions of the invention.
  • the compositions of this invention can be administered by any means known in the medical field, as well as by novel means.
  • the compositions may include administration to a subject intravenously, intradermally, intraarterially, intraperitoneally, intracranially, intraarticularly, intraprostatically, intratracheally, intranasally, intravitreal, intravaginally, intrarectally, locally, intramuscularly, intramuscularly, intrathecally, intrauterinely, intramuscularly, subconjunctively.
  • Administration can be local, for example, at the site of the disease (eg, tumor in the case of cancer) or systemic.
  • pharmaceutically acceptable carriers can be solid or liquid.
  • Solid form preparations include powders, tablets, pills, capsules, cachets, suppositories, and dispersible granules.
  • a solid carrier can be one or more substances that can also act as diluents, flavors, binders, preservatives, disintegrating agents, or encapsulating material.
  • the carrier is a finely divided, dispersed solid in admixture with a finely divided active component (eg, the compositions provided herein).
  • a finely divided active component eg, the compositions provided herein.
  • the active composition is mixed with a carrier having the necessary binding properties in suitable proportions and compacted in the desired shape and size. Powders and tablets can contain from about 5% to about 70% of the active compositions.
  • injectable, sterile solutions preferably oily or aqueous solutions, as well as dispersions, suspensions, emulsions or implants, including rectal suppositories
  • carriers for parenteral administration include aqueous solutions of dextrose, saline, pure water, buffers, ethanol, glycerin, propylene glycol, peanut oil, sesame oil, polyoxyethylene polymer polymers, and the like. It is convenient to use ampoules for dosing.
  • the compositions can also be incorporated into liposomes or administered via transdermal pumps or patches.
  • the pharmaceutical preparation is preferably in unit dosage form.
  • the preparation is subdivided into unit doses containing appropriate amounts of the active ingredient (agent and / or antibody).
  • a unit dosage form can be a packaged preparation, a sachet containing discrete quantities of preparation, such as packaged tablets, capsules and powders in vials or ampoules.
  • the dosage form can be in the form of a frozen dispersion.
  • compositions described herein may further include components to provide sustained release and / or comfort.
  • Such components include high molecular weight, anionic mucomimetic polymers, gelling polysaccharides, and micronized drug carrier substrates. These components can serve a variety of functions as they can also act as a dispersion medium to aid in the formation of nanoclusters.
  • Nanocluster dispersions can be loaded into devices (entities) known to those skilled in the art of drug delivery to further provide controlled (e.g., sustained) release, including liposomes, microspheres, capsules, osmotic pumps, coating of polymer shells, matrices, and implantable devices. In another embodiment, the nanocluster dispersions can be dried and then loaded into these devices.
  • compositions described herein are suitable for parenteral administration, such as intravenous, administration, or administration into a body cavity or organ lumen.
  • parenteral administration such as intravenous, administration, or administration into a body cavity or organ lumen.
  • acceptable carriers and solvents that can be used are water and Ringer's solution, isotonic sodium chloride.
  • sterile fixed oils can be conventionally used as a solvent or suspending medium. Any soft fixed oil can be used for this purpose, including synthetic mono- or diglycerides.
  • fatty acids such as oleic acid can also be used for the preparation of injectables. These solutions are sterile and usually do not contain unwanted substances. These formulations can be sterilized by conventional, well-known sterilization techniques (eg, filtration).
  • the intravenous preparation can be a sterile injectable preparation such as a sterile injectable aqueous dispersion.
  • composition comprising said agent and / or said antibody described herein
  • a therapeutically effective amount can be initially determined from cell culture experiments.
  • concentrations will be those concentrations of active compound (s) that are capable of achieving the effects described herein, as measured using the methods described herein or known in the art, both in terms of transfection efficiency and in terms of erythrophagocytosis or phagocytosis by macrophages of cells. with which these antibodies bind.
  • therapeutically effective amounts for use in humans can also be determined in animal models.
  • a human dose can be calculated to achieve a concentration that has been found to be effective in animals.
  • Dosage in humans can be adjusted by monitoring the effectiveness of the compounds and adjusting the dosage up or down as described above. Adjusting the dose for maximum efficacy in humans based on the methods described above and other methods is within the capabilities of the skilled artisan.
  • testing of the body's hemolytic response to the introduction of antibodies can be performed in vitro before starting treatment.
  • a strong decrease in the level of hematocrit or the level of other cell populations of the blood can be compensated by the transfusion of donated blood or its components.
  • the dosage of said agent and antibody may vary depending on the requirements of the patient and the compound used.
  • the dose administered to the patient should be sufficient to produce a beneficial therapeutic response over time in the patient.
  • the size of the dose will also be determined by the presence, nature and extent of any adverse side effects. Monitoring of hemolysis and hematocrit during and after treatment is important. Determining the correct dosage for a particular situation is within the skill of the art. Typically, treatment begins with lower doses that are less than the optimal dose of the compound. Thereafter, the dosage is increased in small increments until the optimal effect for the specific conditions is achieved.
  • nucleic acid delivery systems there is no or rudimentary data on the immune response to the test formulation of the agent, for example, upon repeated administration, except for those cases when the actual goal was immuno-modulation (He, C. et al., Nat Commun. 2016), (Prasad, S. et al., J Autoimmun, 2018). It is known that in a healthy liver, Kupffer cells determine immunological tolerance to antigens, and the induction of immunosuppressive Treg cells with inhibition of helper CD4 cells (Heymann, F. et al., 2015).
  • liver cells are damaged, a large number of monocytes from other organs enter the liver (Taske, F., J Hepatol, 2017), the immune response is activated, and immunosuppression induced by Kupper cells is inhibited (Heymann, F. et al., 2015 ).
  • the methods of treatment according to the present invention may include combination therapy, which involves the additional use of any compound, agent, drug, as well as a therapeutic effect, regimen or protocol, having the desired therapeutic, beneficial, additive, synergistic or complementary activity or effect.
  • combination compositions and methods of treatment may include second active agents such as biologics (proteins), agents (eg, immunosuppressants), and drugs.
  • second active agents such as biologics (proteins), agents (eg, immunosuppressants), and drugs.
  • Such substances, agents, drugs and methods of treatment can be administered or carried out before, simultaneously with or after any other method of treatment in accordance with the present invention, for example, a therapeutic method for treating a subject with lysosomes.
  • said antibodies induce a transient (transient) increase in IL-10 and MCP-1 chemokine, as well as TNF-a (e.g., anti-erythrocyte antibodies - see Robak, T. et al. Blood 120, 3670- 3676 (2012)).
  • TNF-a e.g., anti-erythrocyte antibodies - see Robak, T. et al. Blood 120, 3670- 3676 (2012).
  • the probability is predicted (estimated, the probability is studied) of an increase in these cytokines based on the analysis of their genome, in particular, for example, by the Fc receptor genotype (for example, the F cy 111 A l 58VV genotype gives a strong increase in the level of cytokines, a F cy 111 A- 158 FF - weak).
  • a decrease in the expression of a protein or nucleic acid encoded by a delivered polynucleotide in macrophages and other actively phagocytic cells of the body is achieved, but an increase in expression in target cells (i.e., in certain cell populations that are the target of therapy).
  • a therapeutic agent (with the required properties of a polynucleotide), a dose (antibodies and an agent), a dosage regimen, specificity of antibodies for the patient the expression of the therapeutic construct either in the body as a whole or in the cells of the phagocytic system, but at the same time an increase in the expression of the therapeutic construct in the targeted population of cells was provided, which seems to be an additional great advantage of this method of gene therapy.
  • the introduction of these antibodies before or simultaneously with the introduction can significantly reduce the side effects from the introduction of a therapeutic agent, up to a decrease in deaths.
  • interleukin-10 many "protective" mechanisms of action of interleukin-10 are known (see Viejo-Borbolla A, Schildberg FA, Burgert HG. Mechanisms of Extracellular Immunomodulation Mediated by Infectious Agents. J Immunol Res. 2017; 2017: 5107527. Doi: 10.1155 / 2017 / 5107527 - the entire content of this document is included in this description by reference), among which it is important to note: increasing the level of interleukin-10, especially local, reduces excessive inflammation and possible tissue damage from such excessive inflammation; in addition, interleukin-10 is a prosurvival factor for activated NK cells, inhibiting activation-induced cell death.
  • this makes it possible to increase the dose of the therapeutic agent, while remaining in a safe range, and thereby significantly increase the efficiency of delivery of the genetic construct (polynucleotide) to target cells and other drugs and substances carried by the agents.
  • Fig. 1 Comparison of the kinetics of the removal of magnetic particles from the bloodstream of mice as described in Example 7: experimental group (solid line) - mice, which 12 hours before the introduction of the agent were injected with 100 ⁇ l with 25 ⁇ g of antibodies against IgG2a 34-3C erythrocytes; control group (dashed line) - mice, which 12 hours before the introduction of the agent were injected with 100 ⁇ l of saline. A significant prolongation of the circulation time of particles is seen in the case of administration to mice with cytoblockade.
  • Fig. 2 Measurement of induction of cytoblockade using gold test agents and strip strips (lateral flow) as described in Example 8. Left - test strip measurement before the administration of antibodies, on the right - after administration of antibodies and induction of cytoblockade.
  • Fig.Z Demonstration of the increase in the efficiency of lung transfection with polyplexes after administration of antibodies 34-3C as described in Example 11.
  • Fig. 4. Demonstration of an increase in absolute (maximum) transfection efficiency with polyplexes after administration of 34-3C antibodies as described in Example 12.
  • Balb / c or C57BL / 6J mice are injected intraperitoneally, intravenously or retroorbital with 6-100 ⁇ g of IgG2a 34-3C antibodies against erythrocytes, or 12-100 ⁇ g of TER-119 antibodies, or 1-5 mg of polyclonal rabbit antibodies against mouse red blood cells , or antibodies against CD47 (for example, clone MIAP 410).
  • the cytoblock begins to fail almost immediately, the peak is reached depending on the selected antibody - after 3-12 hours. After 5-7 days, the effect of cytoblockade stops.
  • Example 2 Measurement of induction of cytoblockade using fluorescent test agents (quantitative verification of the state of MFS blockage induced by antibodies against erythrocytes TER-119).
  • Balb / c mice were retroorbitally injected with 300 ⁇ g of test agents - 110 nm Spherotech Carboxyl Fluorescent Yellow fluorescent particles. After 10 minutes, a blood sample with heparin is taken, 10 ⁇ l of plasma is isolated, diluted with phosphate-saline solution to 100 ⁇ l, and the fluorescent signal of the sample is measured with excitation at 488 + -5 nm, emission at 520 + -10 nm.
  • cytoblockade is induced in as described in Example 1, using a retroorbital injection of 100 ⁇ l with 25 ⁇ g of TER-119 in physical. solution. After 5 hours, repeat the procedure for introducing the test agent and measuring its plasma concentration after 10 minutes of circulation. The measured fluorescent signal after the introduction of antibodies is 4.5 times higher than before the introduction, therefore, cytoblockade is induced.
  • Example 3 Confirmation of the procedure for measuring the induction of cytoblockade using fluorescent test agents - control.
  • Balb / c mice were retroorbitally injected with 300 ⁇ g of test agents - 110 nm Spherotech Carboxyl Fluorescent Yellow fluorescent particles. After 10 minutes, a blood sample with heparin is taken, 10 ⁇ l of plasma is isolated, diluted with phosphate-saline solution to 100 ⁇ l, and the fluorescent signal of the sample is measured with excitation at 488 + -5 nm, emission at 520 + -10 nm.
  • Example 4 Measurement of induction of cytoblockade using fluorescent test agents (quantitative verification of the state of blockage of MFS induced by antibodies against erythrocytes 34-3 SU
  • Balb / c mice are retroorbitally injected with 300 ⁇ g of test agents - fluorescent particles 0.2 ⁇ m Spherotech Carboxyl Fluorescent Nile Red. After 10 minutes, a blood sample with heparin is taken, 10 ⁇ l of plasma is isolated, diluted with phosphate-saline solution to 100 ⁇ l, and the fluorescent signal of the sample is measured with excitation at 520 + -10 nm, emission at 600 + -20 nm.
  • cytoblockade is induced in as described in Example 1, using a retroorbital injection of 100 ⁇ l with 25 ⁇ g 34-3C in physical. solution. After 5 hours, repeat the procedure for introducing the test agent and measuring its plasma concentration after 10 minutes of circulation. The measured fluorescent signal after the introduction of antibodies is 8.3 times higher than before the introduction, therefore, the cytoblockade is induced.
  • Example 5 Measurement of induction of cytoblockade using fluorescent test agents (quantitative verification of the state of MFS blockage induced by antibodies against CD47 MIAP 410).
  • Balb / c mice are retroorbitally injected with 300 ⁇ g of test agents - 0.05 ⁇ m fluorescent particles Spherotech Carboxyl Fluorescent Pink. After 20 minutes, a blood sample with heparin is taken, 10 ⁇ l of plasma is isolated, diluted with phosphate-saline solution to 100 ⁇ l, and the fluorescent signal of the sample is measured with excitation at 560 + -10 nm, emission at 590 + -10 nm.
  • cytoblockade was induced as described in Example 1 using a retroorbital injection of 100 ⁇ l with 50 ⁇ g of anti-CD47 clone MIAP 410 in saline. After 5 hours, repeat the procedure for introducing the test agent and measuring its plasma concentration after 20 minutes of circulation. The measured fluorescent signal after the introduction of antibodies is 5.4 times higher than before the introduction, therefore, cytoblockade is induced.
  • Example 6 Measurement of the induction of cytoblockade using magnetic test agents
  • mice were retroorbally injected with 300 ⁇ g test agents - 240 nm Estapor magnetic particles. After 20 minutes, a heparinized blood sample (50 ⁇ l) was taken and the particle content was measured using the MPQ method (as described in Nikitin et al. Nat Nanotechnol. 2014).
  • cytoblockade was induced as described in Example 1 using a retroorbital injection of 100 ⁇ l with 25 ⁇ g 34-3C in saline. After 5 hours, repeat the procedure for introducing the test agent and measuring its plasma concentration after 20 minutes of circulation.
  • the measured magnetic signal after the introduction of antibodies is 133 times higher than before the injection (since before the injection, the signal is actually equal to noise, i.e. the particles are not actually detected, and after the introduction of antibodies a significant signal from the particles is recorded), therefore, cytoblockade is induced.
  • Example 7 The effect of cytoblockade on the rate of elimination of particles from the bloodstream.
  • Example 8 Measurement of induction of cytoblockade using gold test agents and strip strips (lateral flow).
  • Test agents - 50 nm gold nanoparticles conjugated to bovine serum albumin conjugated with chloramphenicol are injected retroorbital to Balb / c mice test agents - 50 nm gold nanoparticles. After 10 minutes, a blood sample with heparin is taken, 10 ⁇ l of plasma is isolated, diluted with a solution of 0.1% Tween 20, 1% BSA and passed through a test strip (lateral flow strip). As Fig. 2 shows, a strip on a test line with applied antibodies against chloramphenicol (clone CAPB10) are practically invisible visually (there are practically no particles).
  • cytoblockade is induced in as described in Example 1, using a retroorbital injection of 100 ⁇ l with 25 ⁇ g 34-3C in physical. solution. After 5 hours, repeat the procedure for introducing the test agent and measuring its plasma concentration after 10 minutes of circulation. As Fig. 2 shows, a strip of gold particles on the test line with applied antibodies against chloramphenicol (clone CAPB10) is clearly and clearly visible (particles are well detected), therefore, cytoblockade is induced.
  • Example 9 Synthesis of a polyplex for delivery of a heterologous polynucleotide based on polyethyleneimine.
  • Example 10 Reduction of the toxic effect of a polyplex for delivery of a heterologous polynucleotide after administration of antibodies 34-3C.
  • mice are injected with 25 ⁇ g of 34-3 C antibodies (as described in Example 1), and then, 12 hours later, a dose of polyplexes from Example 9 is injected equal to the LD50 for intact mice (which did not receive the antibody) - 80 ⁇ g by DNA. As a result, none of the 8 mice died. Those. antibody 34-3C significantly reduced the toxic effect of polyplexes.
  • Example 11 Increase in the efficiency of lung transfection with polyplexes after administration of antibodies 34-3C.
  • mice from Example 10 including surviving mice that were injected with the agent without the introduction of antibodies
  • the organs are extracted and homogenized, after which the luciferase substrate is added according to the manufacturer's recommendations (kit Promega Luciferase Assay System) and the luminescent signal reflecting the expression of the delivered gene.
  • the luciferase substrate is added according to the manufacturer's recommendations (kit Promega Luciferase Assay System) and the luminescent signal reflecting the expression of the delivered gene.
  • control mice which were not injected with 34-3C antibodies
  • a half-dose was injected - 40 ⁇ g for DNA, which nevertheless turned out to be lethal (1 mouse out of 4 died).
  • a comparison of the luminescence distribution is shown in FIG. 2. Significantly higher efficiency of transfection in mice with cytoblockade is seen.
  • Example 12 Increase in absolute (maximum) efficiency of transfection with polyplexes after administration of antibodies 34-3C.
  • mice from Example 10 which were injected with antibodies, are euthanized, the organs are extracted and homogenized, after which the luciferase substrate is added according to the manufacturer's recommendations (kit Promega Luciferase Assay System) and a luminescent signal is read, reflecting the expression of the delivered gene.
  • the luciferase substrate is added according to the manufacturer's recommendations (kit Promega Luciferase Assay System) and a luminescent signal is read, reflecting the expression of the delivered gene.
  • control mice which were not injected with 34-3C antibodies
  • a half-dose was injected - 50 ⁇ g for DNA, which nevertheless turned out to be lethal (1 mouse out of 4 died).
  • a comparison of the luminescence distribution is shown in FIG. 4. It can be seen that transfection even with only one lethal outcome in the control group is already significantly lower than in the group with injected antibodies.
  • Example 13 Increase in the efficiency of heart transfection with the AAV9 virus after administration of antibodies 34-3C.
  • mice are injected with 25 ⁇ g of 34-3C antibodies (as described in Example 1) or an equal volume of saline (in the control group). Then, 12 hours later, both groups are injected with 2.5 * 10 L 12 viral particles of the AAV9 virus (encoding the firefly luciferase gene). After 3 weeks, euthanasia is performed, the heart is extracted and homogenized. Then the luciferase substrate is added to the homogenate according to the manufacturer's recommendations (kit Promega Luciferase Assay System) and the luminescent signal is read, reflecting the expression of the delivered gene. The luminescent signal in the group that received the 34-3C antibody is, on average, 2.5 times higher than in the samples of the control group.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Engineering & Computer Science (AREA)
  • Epidemiology (AREA)
  • Microbiology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Genetics & Genomics (AREA)
  • Mycology (AREA)
  • Organic Chemistry (AREA)
  • Immunology (AREA)
  • Biotechnology (AREA)
  • Wood Science & Technology (AREA)
  • Zoology (AREA)
  • General Engineering & Computer Science (AREA)
  • Molecular Biology (AREA)
  • Biomedical Technology (AREA)
  • Biophysics (AREA)
  • Physics & Mathematics (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Plant Pathology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Biochemistry (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)

Abstract

L'invention se rapporte au domaine de la bio-médecine, de la nano-médecine, et concerne plus précisément des procédés de thérapie génique, d'administration d'acides nucléiques dans l'organisme, ainsi que des procédés et des compositions de traitement d'un sujet. L'invention concerne essentiellement un procédé (et une composition pour sa mise en oeuvre) de traitement d'un sujet chez qui on doit traiter une maladie induite par des pertes de fonction ou d'activité protéique, lequel consiste à: administrer audit sujet un anticorps se liant spécifiquement aux cellules sanguines; administrer audit sujet un agent contenant un polynucléotide hétérologue qui code une protéine, qui assure ou complète la fonction ou l'activité de la protéine, ou administrer audit sujet un agent contenant un polynucléotide hétérologue qui se transcrit en un acide nucléique qui inhibe ou diminue l'expression de l'amplification de la fonction, de l'activité ou de l'expression de ladite protéine. Le résultat technique de la présente invention consiste en une augmentation du temps de circulation des agents assurant l'administration d'acides nucléiques (polynucléotides), une augmentation de l'efficacité d'administration des polynucléotides par lesdits agents vers des cibles de l'organisme, ainsi qu'une diminution de la toxicité et une augmentation de la sécurité de ces agents.
PCT/RU2021/050192 2020-07-03 2021-07-01 Procédé de traitement avec un agent de thérapie génique Ceased WO2022005338A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
RU2020122141 2020-07-03
RU2020122141A RU2020122141A (ru) 2020-07-03 2020-07-03 Способ и композиция на основе генотерапевтического агента и антитела, связывающегося с клетками крови

Publications (1)

Publication Number Publication Date
WO2022005338A1 true WO2022005338A1 (fr) 2022-01-06

Family

ID=79317810

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/RU2021/050192 Ceased WO2022005338A1 (fr) 2020-07-03 2021-07-01 Procédé de traitement avec un agent de thérapie génique

Country Status (2)

Country Link
RU (1) RU2020122141A (fr)
WO (1) WO2022005338A1 (fr)

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2019018439A1 (fr) * 2017-07-17 2019-01-24 Spark Therapeutics, Inc. Méthodes d'aphérèse et utilisations associées
WO2019051135A1 (fr) * 2017-09-06 2019-03-14 Fred Hutchinson Cancer Research Center Méthodes pour améliorer une thérapie cellulaire adoptive
WO2020016318A1 (fr) * 2018-07-17 2020-01-23 INSERM (Institut National de la Santé et de la Recherche Médicale) Compositions et procédés pour augmenter ou améliorer la transduction de vecteurs de thérapie génique et pour éliminer ou réduire des immunoglobulines

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2019018439A1 (fr) * 2017-07-17 2019-01-24 Spark Therapeutics, Inc. Méthodes d'aphérèse et utilisations associées
WO2019051135A1 (fr) * 2017-09-06 2019-03-14 Fred Hutchinson Cancer Research Center Méthodes pour améliorer une thérapie cellulaire adoptive
WO2020016318A1 (fr) * 2018-07-17 2020-01-23 INSERM (Institut National de la Santé et de la Recherche Médicale) Compositions et procédés pour augmenter ou améliorer la transduction de vecteurs de thérapie génique et pour éliminer ou réduire des immunoglobulines

Also Published As

Publication number Publication date
RU2020122141A (ru) 2022-01-04
RU2020122141A3 (fr) 2022-01-04

Similar Documents

Publication Publication Date Title
Rohner et al. Unlocking the promise of mRNA therapeutics
Terstappen et al. Strategies for delivering therapeutics across the blood–brain barrier
Stanimirovic et al. Emerging technologies for delivery of biotherapeutics and gene therapy across the blood–brain barrier
Guan et al. Nanotechnologies in delivery of mRNA therapeutics using nonviral vector-based delivery systems
Templeton et al. New directions in liposome gene delivery
JP2023179414A (ja) アフェレーシスの方法及び使用
Steffens et al. Directing the way—receptor and chemical targeting strategies for nucleic acid delivery
AU2022262771A9 (en) Novel compositions with brain-specific targeting motifs and compositions containing same
US20230416757A1 (en) Sod1 dual expression vectors and uses thereof
Schuh et al. Nanotechnology applied to treatment of mucopolysaccharidoses
EP2656837A1 (fr) Nanoparticules lipidiques pour le traitement de maladies oculaires
US20240043494A1 (en) Vesicle Targeting Proteins And Uses Of Same
JP2024501284A (ja) 非ウイルス遺伝子治療を強化する方法
US20250032550A1 (en) A novel way for brain-specific delivery of molecules for the treatment of brain diseases
Chen et al. Unleashing the potential of mRNA: Overcoming delivery challenges with nanoparticles
WO2022005338A1 (fr) Procédé de traitement avec un agent de thérapie génique
US20220339294A1 (en) Nanoparticles for selective tissue or cellular uptake
Erel-Akbaba et al. Rabies virus-mimicking liposomes for targeted gene therapy in Alzheimer’s disease
WO2025015749A1 (fr) Complexe support d'administration de médicament ciblant le système nerveux central, procédé de préparation d'un complexe support d'administration de médicament, et son utilisation
Guan et al. How Advanced are Nanocarriers for Effective Subretinal Injection?
WO2022159722A1 (fr) Utilisation de nouvelles cassettes de site de liaison de miarn pour le déciblage de cellules présentatrices d'antigène de l'expression de transgènes par des vecteurs de thérapie génique raav
WO2022005339A1 (fr) Composition pour thérapie et diagnostic à base d'anticorps
Aundhia et al. Impact and Significance of Viral Vectors for siRNA Delivery in the Treatment of Alzheimer’s Disease
Wairkar et al. Targeted delivery of biopharmaceuticals for neurodegenerative disorders
WO2020172263A1 (fr) Apport de macromolécules au système nerveux central par l'intermédiaire de la circulation sanguine

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: 21833164

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 21833164

Country of ref document: EP

Kind code of ref document: A1