WO2022035197A1 - Formulation pharmaceutique stable - Google Patents

Formulation pharmaceutique stable Download PDF

Info

Publication number
WO2022035197A1
WO2022035197A1 PCT/KR2021/010606 KR2021010606W WO2022035197A1 WO 2022035197 A1 WO2022035197 A1 WO 2022035197A1 KR 2021010606 W KR2021010606 W KR 2021010606W WO 2022035197 A1 WO2022035197 A1 WO 2022035197A1
Authority
WO
WIPO (PCT)
Prior art keywords
seq
binding molecule
pharmaceutical formulation
present
stable pharmaceutical
Prior art date
Application number
PCT/KR2021/010606
Other languages
English (en)
Korean (ko)
Inventor
김광우
김수정
노지원
오준석
이재빈
한원용
Original Assignee
(주)셀트리온
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 (주)셀트리온 filed Critical (주)셀트리온
Publication of WO2022035197A1 publication Critical patent/WO2022035197A1/fr

Links

Images

Classifications

    • 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
    • A61K39/39591Stabilisation, fragmentation
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/08Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from viruses
    • C07K16/10Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from viruses from RNA viruses
    • C07K16/1002Coronaviridae
    • C07K16/1003Severe acute respiratory syndrome coronavirus 2 [SARS‐CoV‐2 or Covid-19]
    • 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
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/42Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum viral
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/02Inorganic compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/16Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing nitrogen, e.g. nitro-, nitroso-, azo-compounds, nitriles, cyanates
    • A61K47/18Amines; Amides; Ureas; Quaternary ammonium compounds; Amino acids; Oligopeptides having up to five amino acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/16Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing nitrogen, e.g. nitro-, nitroso-, azo-compounds, nitriles, cyanates
    • A61K47/18Amines; Amides; Ureas; Quaternary ammonium compounds; Amino acids; Oligopeptides having up to five amino acids
    • A61K47/183Amino acids, e.g. glycine, EDTA or aspartame
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/22Heterocyclic compounds, e.g. ascorbic acid, tocopherol or pyrrolidones
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/26Carbohydrates, e.g. sugar alcohols, amino sugars, nucleic acids, mono-, di- or oligo-saccharides; Derivatives thereof, e.g. polysorbates, sorbitan fatty acid esters or glycyrrhizin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/08Solutions
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/08Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from viruses
    • C07K16/10Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from viruses from RNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding

Definitions

  • the present invention relates to a stable pharmaceutical preparation, and more particularly, to a pharmaceutical preparation capable of stably preserving a molecule binding to a spike protein (S protein) on the surface of SARS-coronavirus-2 .
  • S protein spike protein
  • SARS-coronavirus-2 severe acute respiratory syndrome coronavirus 2, SARS-CoV-2
  • SARS-CoV-2 is a single-stranded RNA coronavirus with a positive sense in genetic sequencing (DNA sequencing).
  • SARS-CoV-2 is contagious to humans and is the cause of coronavirus disease 2019 (COVID-19). The first outbreak of COVID-19 was in Wuhan, Hubei Province, China.
  • SARS-CoV-2 may have mild to severe symptoms such as fever, cough, shortness of breath, and diarrhea. People with complications or diseases and the elderly are more likely to die.
  • coronavirus disease 2019 2019 (COVID-19)
  • the existing treatment is administered to the patient to expect a therapeutic effect.
  • Antiviral agents favipiravir, remdesivir, and galidesivir, which are Ebola treatment or treatment candidates, and hepatitis C treatment ribavirin are being used as COVID-19 treatments.
  • the antimalarial drug Chloroquine has been shown to have a therapeutic effect on COVID-19 and is undergoing public clinical trials.
  • hepatitis C treatment ribavirin may have severe side effects such as anemia, and the antiviral drug interferon is also recommended to be used with caution due to concerns about various side effects.
  • the Korea Centers for Disease Control and Prevention announced that i) that COVID-19 could also spread like influenza for a long time and that it would be included in the surveillance system like influenza, and ii) a coronavirus that spreads among people (4 species) is also prevalent in winter and spring, leaving open the possibility that COVID-19 may also become indigenous (2020.2.17).
  • a formulation capable of more stably preserving a binding molecule targeting the SARS-CoV-2 virus spike protein (S protein) has not yet been developed. Accordingly, as a result of continuous research to develop a formulation having excellent stability, the present inventors have finally completed the present invention.
  • the present inventors have developed a pharmaceutical preparation capable of stably preserving a neutralizing binding molecule that binds to a spike protein (S protein) on the surface of SARS-CoV-2.
  • S protein spike protein
  • the problem to be solved by the present invention is a stable pharmaceutical preparation, preferably comprising a neutralizing binding molecule that binds to the spike protein (S protein) on the surface of SARS-CoV-2 (SARS-CoV-2)
  • S protein spike protein
  • SARS-CoV-2 SARS-CoV-2
  • the present invention provides (A) a neutralizing binding molecule that binds to the spike protein (S protein) on the surface of SARS-coronavirus-2 (SARS-CoV-2), (B) a buffer, (C ) a stabilizer and (D) a surfactant.
  • S protein spike protein
  • SARS-CoV-2 SARS-coronavirus-2
  • Neutralizing binding molecules such as antibodies have larger and more complex structures than conventional organic and inorganic drugs. Therefore, a pharmaceutical preparation containing a neutralizing-binding molecule such as an antibody has a special problem in that the stability of the neutralizing-binding molecule, such as an antibody, must be maintained.
  • the stability of a neutralizing binding molecule may be affected by factors such as ionic strength, pH, temperature, repeated freeze/thaw cycles, and concentration of a neutralizing binding molecule, such as an antibody.
  • Neutralizing binding molecules such as active antibodies, exhibit physical instability including denaturation, aggregation (formation of soluble and insoluble aggregates), precipitation and adsorption, or physical instability, including racemization, beta-elimination, disulfide exchange, hydrolysis, deamidation and oxidation. Loss may occur due to chemical instability.
  • Such physical and chemical instability can potentially result in the formation of neutralizing binding molecule byproducts or derivatives, such as antibodies, with reduced biological activity, increased toxicity, and/or increased immunogenicity of the neutralizing binding molecule, such as an antibody.
  • each neutralizing-binding molecule such as an antibody has its own physicochemical properties, so neutralizing-binding molecules such as other antibodies have been proposed. It is practically impossible to predict the type and amount of excipients to overcome the problem of instability of a neutralizing binding molecule such as a specific antibody in consideration of the conventional formulation used. In addition, it is difficult and time-consuming to derive optimal conditions such as the concentration of neutralizing-binding molecules such as antibodies, pH, and concentration of excipients for maintaining chemically and biologically stable neutralizing-binding molecules such as specific antibodies in pharmaceutical formulations and effort are required.
  • the present invention relates to a high molecular weight of the neutralizing binding molecule in a pharmaceutical formulation comprising a neutralizing binding molecule that binds to a spike protein (S protein) on the surface of SARS-coronavirus-2 and a buffer, a stabilizer and a surfactant together. It was confirmed for the first time that there was an effect of stabilizing its activity by reducing the formation of the number of components and insoluble foreign substances.
  • the present invention relates to a spike protein on the surface of a coronavirus, preferably SARS-coronavirus-2 (SARS-CoV-2), more preferably SARS-coronavirus-2 (SARS-CoV-2). , S protein) to a stable pharmaceutical formulation comprising a neutralizing binding molecule that binds.
  • SARS-CoV-2 SARS-coronavirus-2
  • SARS-CoV-2 SARS-coronavirus-2
  • SARS-CoV-2 SARS-coronavirus-2
  • SARS-CoV-2 SARS-coronavirus-2
  • SARS-CoV-2 SARS-coronavirus-2
  • the binding molecule may bind to the receptor binding domain (RBD) region of the spike protein on the SARS-coronavirus-2 surface.
  • RBD receptor binding domain
  • the SARS-coronavirus-2 (SARS-CoV-2) surface spike protein (S protein, S protein) of the present invention consists of or includes the sequence of SEQ ID NO: 3841. and derivatives and/or variants thereof, but are not limited thereto.
  • the SARS-CoV-2 (SARS-CoV-2) surface spike protein region of the present invention may consist of or include the sequence of SEQ ID NO: 3842. , derivatives and/or variants thereof.
  • the pharmaceutical formulation according to the present invention may be in a liquid form, but is not limited thereto.
  • the neutralizing binding molecule of the present invention exhibits excellent binding ability and/or excellent neutralizing ability to the spike protein of SARS-CoV-2 (SARS-CoV-2). In another embodiment of the present invention, the neutralizing binding molecule of the present invention exhibits excellent binding and/or neutralizing ability to a mutant virus in which the spike protein of SARS-CoV-2 is mutated.
  • the neutralizing binding molecule of the present invention is a binding molecule that binds to the receptor binding domain (RBD) region of the spike protein of SARS-CoV-2 and is screened as a binding molecule with neutralizing ability.
  • the neutralizing binding molecule of the present invention may exhibit excellent neutralizing ability against mutant viruses of other regions of the S protein other than the RBD region, but is not limited thereto.
  • the World Health Organization classifies SARS-Coronavirus-2 into six types based on amino acid changes due to differences in gene sequence. First, it was classified into S and L types, then again into L, V, and G types, and as G was divided into GH and GR, it is classified into a total of six types: S, L, V, G, GH, and GR. At the beginning of the COVID-19 outbreak, types S and V were prevalent in Asia including Wuhan, China, and after that, different types were discovered for each continent. Among them, it has been reported that the GH type has the potential to appear high in transmission power.
  • the neutralizing binding molecule of the present invention is S-type (amino acid at position 614 of the S protein is D), G-type (the amino acid at position 614 of the S protein is G) based on the SARS-CoV-2 virus amino acid mutation. ), may exhibit neutralizing ability in strains such as V type, but is not limited to this strain.
  • An example of the SARS-CoV-2 virus type S is the BetaCoV/Korea/KCDC03/2020 strain, but is not limited thereto.
  • An example of the SARS-CoV-2 virus type G includes, but is not limited to, the hCoV-19/South Korea/KUMC17/2020 strain.
  • the neutralizing binding molecule of the present invention exhibits excellent neutralizing ability even in a mutant virus in which D614G mutation occurs at amino acid position 614 of the spike protein of SARS-CoV-2 (SARS-CoV-2).
  • the neutralizing binding molecule of the present invention is SARS-coronavirus-2 (SARS-CoV-2) surface protein (RBD) mutant proteins A435S, F342L, G476S, K458R, N354D, V367F, V483A, and W436R It exhibits excellent bonding strength.
  • the neutralizing binding molecule of the present invention is a SARS-coronavirus-2 strain isolated to date, for example, UNKNOWN-LR757996 strain (Strain), SARS-CoV-2/Hu of unknown date and place of isolation.
  • the neutralizing binding molecule of the present invention may be a neutralizing binding molecule that binds to a spike protein (S protein) on the surface of SARS-CoV-2.
  • S protein spike protein
  • the neutralizing binding molecule of the present invention may be any one binding molecule selected from the group consisting of binding molecules shown in Table 1, preferably binding molecule No. 89 to No. 194, and No. 248 may be any one binding molecule selected from the group consisting of, more preferably No. 139 binding molecule. In Table 1 below, No. means the number of each binding molecule.
  • the CDRs of the variable region according to the present invention were determined by a conventional method according to the system devised by Kabat et al. (Kabat et al., Sequences of Proteins of Immunological Interest (5th), National Institutes of Health, Bethesda) , MD. (1991)).
  • the CDR numbering used in the present invention uses the Kabat method, but may be determined according to other methods such as the IMGT method, the Chothia method, and the AbM method.
  • a binding molecule comprising a CDR determined by any of the above methods is also included in the present invention.
  • the neutralizing binding molecule of the present invention may be any one binding molecule selected from the group consisting of binding molecules shown in Table 2, preferably binding molecule No. 89 to No. 194, and No. 248 may be any one binding molecule selected from the group consisting of, more preferably No. 139 binding molecule. In Table 2 below, No. means the number of each binding molecule.
  • the neutralizing binding molecule of the present invention may be any one binding molecule selected from the group consisting of binding molecules shown in Table 3, preferably No. 322 or No. 343 binding molecule, more preferably No. 322 binding molecule.
  • No. means the number of each binding molecule.
  • the neutralizing binding molecule of the present invention may be any one binding molecule selected from the group consisting of binding molecules shown in Table 4, preferably No. 322 or No. 343 binding molecule, more preferably No. 322 binding molecule.
  • No. means the number of each binding molecule.
  • the neutralizing binding molecule of the present invention may be an scFv fragment, an scFv-Fc fragment, a Fab fragment, an Fv fragment, a diabody, a chimeric antibody, a humanized antibody, or a human antibody, but is not limited thereto.
  • the neutralizing binding molecule of the present invention may be an scFv-Fc that binds to the SARS-CoV-2 S protein.
  • a neutralizing binding molecule of the present invention comprises a light chain variable region comprising LC CDR1, LC CDR2 and LC CDR3, and a heavy chain variable region comprising HC CDR1, HC CDR2 and HC CDR3,
  • said LC CDR1 comprises the sequence SGX 1 X 2 SNIGX 3 NX 4 X 5 S, wherein X 1 is S, G or R, X 2 is S, N or T, and X 3 is N, D or K , X 4 is Y or F, X 5 is V or I,
  • said LC CDR2 comprises the sequence DNX 6 KRPS, wherein X 6 is N or D;
  • said LC CDR3 comprises the sequence GTWDX 7 X 8 LSX 9 X 10 X 11 , wherein X 7 is S or N, X 8 is S or N, X 9 is A or G, and X 10 is G or V and X 11 is V or R,
  • said HC CDR1 comprises the sequence TSGX 12 GVX 13 , wherein X 12 is M or V, X 13 is G or S,
  • said HC CDR2 comprises the sequence LIDWDDNKYX 14 TTSLKT, wherein X 14 is Y or H;
  • Said HC CDR3 may be a binding molecule comprising the sequence IPGFLRYRNRYYYYGX 15 DV, wherein X 15 is M or V.
  • the neutralizing binding molecule of the present invention comprises a light chain variable region comprising LC CDR1, LC CDR2 and LC CDR3, and a heavy chain variable region comprising HC CDR1, HC CDR2 and HC CDR3, ,
  • LC CDR1 comprises the sequence RASQSISSYLN
  • LC CDR2 comprises the sequence AASSLQS
  • said LC CDR3 comprises the sequence QQSYSTPLT
  • said HC CDR1 comprises the sequence SNYMX 16 , wherein X 16 is T or S;
  • said HC CDR2 comprises the sequence X 17 IYPGGSTX 18 X 19 ADSVX 20 G, wherein X 17 is I or V, X 18 is Y or F, X 19 is Y or F, and X 20 is K or Q ,
  • the HC CDR3 may be a binding molecule comprising the sequence DLPLTGTTLDY or SYDFLTDYTDAFDI.
  • the neutralizing binding molecule of the present invention comprises a light chain variable region comprising a CDR1 region of SEQ ID NO: 829, a CDR2 region of SEQ ID NO: 830, and a CDR3 region of SEQ ID NO: 831; and a heavy chain variable region comprising a CDR1 region of SEQ ID NO: 832, a CDR2 region of SEQ ID NO: 833, and a CDR3 region of SEQ ID NO: 834.
  • the neutralizing binding molecule of the present invention comprises a light chain variable region comprising a CDR1 region of SEQ ID NO: 2507, a CDR2 region of SEQ ID NO: 2508, and a CDR3 region of SEQ ID NO: 2509; and a heavy chain variable region comprising a CDR1 region of SEQ ID NO: 2510, a CDR2 region of SEQ ID NO: 2511, and a CDR3 region of SEQ ID NO: 2512.
  • the neutralizing binding molecule comprises a light chain variable region of the polypeptide sequence of SEQ ID NO: 2017; and 80% to 99%, preferably 85 to 99%, more preferably 90 to 99% identical to the binding molecule comprising the heavy chain variable region of the polypeptide sequence of SEQ ID NO: 2018. It may be a binding molecule comprising a sequence .
  • the neutralizing binding molecule comprises a light chain variable region of the polypeptide sequence of SEQ ID NO: 3523; and 80% to 99%, preferably 85 to 99%, more preferably 90 to 99% identical to the binding molecule comprising the heavy chain variable region of the polypeptide sequence of SEQ ID NO: 3524. .
  • the neutralizing binding molecule comprises a light chain variable region of the polypeptide sequence of SEQ ID NO: 2017; and a heavy chain variable region of the polypeptide sequence of SEQ ID NO: 2018.
  • the neutralizing binding molecule comprises a light chain variable region of the polypeptide sequence of SEQ ID NO: 3523; and a heavy chain variable region of the polypeptide sequence of SEQ ID NO: 3524.
  • the term 'antibody' is used in the broadest sense, specifically, an intact monoclonal antibody, a polyclonal antibody, a multispecific antibody formed from two or more intact antibodies (eg, a bispecific antibody), and antibody fragments exhibiting the desired biological activity.
  • Antibodies are proteins produced by the immune system that are capable of recognizing and binding to specific antigens. In terms of their structure, antibodies usually have a Y-shaped protein consisting of four amino acid chains (two heavy chains and two light chains). Each antibody mainly has two regions: a variable region and a constant region. The variable region located in the distal portion of the arm of Y binds and interacts with the target antigen.
  • variable region comprises a complementarity determining region (CDR) that recognizes and binds a specific binding site on a specific antigen.
  • CDR complementarity determining region
  • the constant region located at the tail of Y is recognized and interacted with by the immune system.
  • Target antigens have multiple binding sites, called epitopes, which are generally recognized by CDRs on multiple antibodies. Each antibody that specifically binds to a different epitope has a different structure. Thus, an antigen may have more than one corresponding antibody.
  • the binding molecule according to the present invention may include a functional variant of the binding molecule.
  • a variant of the invention may compete with a binding molecule of the invention for specific binding to SARS-CoV-2 or its S protein.
  • it is regarded as a functional variant of the binding molecule of the present invention if it has the ability to neutralize SARS-CoV-2.
  • the functional variant includes, but is not limited to, a derivative having a substantially similar primary structural sequence.
  • the functional variant includes in vitro or in vivo modification, modification by chemical and/or biochemical agents.
  • the functional variant is not found in the parental monoclonal antibody of the present invention.
  • Such modifications include, for example, acetylation, acylation, covalent bonding of nucleotides or nucleotide derivatives, covalent bonding of lipids or lipid derivatives, crosslinking, disulfide bond formation, glycosylation, hydroxylation, methylation, oxidation, pegylation, proteolysis. and phosphorylation.
  • the functional variant may optionally comprise an amino acid sequence containing one or more amino acid substitutions, insertions, deletions or combinations thereof compared to the amino acid sequence of the parent antibody.
  • the functional variant may comprise a truncated form of the amino acid sequence at one or both of the amino terminus or the carboxy terminus.
  • a variable region including, but not limited to, a framework structure, a hypervariable region, in particular, a complementarity-determining region (CDR) of a light or heavy chain
  • CDR complementarity-determining region
  • a light or heavy chain region comprises three hypervariable regions, comprising three CDR regions, and a more conserved region, namely a framework region (FR).
  • FR framework region
  • a hypervariable region comprises amino acid residues from a CDR and amino acid residues from a hypervariable loop.
  • Functional variants within the scope of the present invention include about 50%-99%, about 60%-99%, about 80%-99%, about 90%-99%, about 95%-99%, or about 97%-99% amino acid sequence identity.
  • Gap or Bestfit known to those skilled in the art among computer algorithms may be used to optimally align amino acid sequences to be compared and to define similar or identical amino acid residues.
  • the functional variant may be obtained by changing the parent antibody or a part thereof by a known general molecular biological method including PCR method, mutagenesis using oligomeric nucleotides and partial mutagenesis, or by organic synthesis method.
  • the present invention is not limited thereto.
  • the binding molecule may be an immunoconjugate to which one or more tags are additionally bound, for example, an immunoconjugate to which a drug is further attached to the binding molecule.
  • the binding molecule according to the present invention may be used in the form of an antibody-drug conjugate (ADC) to which a drug is bound.
  • ADC antibody-drug conjugate
  • ADCs i.e. immunoconjugates
  • the ADC form can improve the maximum efficacy and minimum toxicity of the drug by increasing the drug-connectivity and drug-releasing properties as well as the selectivity of polyclonal and monoclonal antibodies (mAbs).
  • the concentration of (A) neutralizing binding molecule of the present invention can be freely adjusted within a range that does not substantially adversely affect the stability and viscosity of the stable pharmaceutical preparation according to the present invention.
  • the concentration of the neutralizing binding molecule may be 1 to 240 mg/ml.
  • the concentration of the neutralizing binding molecule may be 1 to 230 mg/ml.
  • the concentration of the neutralizing binding molecule may be 1 to 220 mg/ml.
  • the concentration of the neutralizing binding molecule may be 1 to 210 mg/ml.
  • the concentration of the neutralizing binding molecule may be between 1 and 200 mg/ml.
  • the concentration of the neutralizing binding molecule may be 5 to 240 mg/ml. In another embodiment of the invention, the concentration of the neutralizing binding molecule may be between 5 and 230 mg/ml. In another embodiment of the present invention, the concentration of the neutralizing binding molecule may be 5 to 220 mg/ml. In another embodiment of the invention, the concentration of the neutralizing binding molecule may be between 5 and 210 mg/ml. In another embodiment of the invention, the concentration of the neutralizing binding molecule may be 5 to 200 mg/ml.
  • the concentration of the neutralizing binding molecule may be 10-240 mg/ml. In another embodiment of the invention, the concentration of the neutralizing binding molecule may be between 10 and 230 mg/ml. In another embodiment of the present invention, the concentration of the neutralizing binding molecule may be 10-220 mg/ml. In another embodiment of the invention, the concentration of the neutralizing binding molecule may be between 10 and 210 mg/ml. In another embodiment of the present invention, the concentration of the neutralizing binding molecule may be between 10 and 200 mg/ml.
  • the concentration of the neutralizing binding molecule may be 20 to 240 mg/ml. In another embodiment of the invention, the concentration of the neutralizing binding molecule may be between 20 and 230 mg/ml. In another embodiment of the present invention, the concentration of the neutralizing binding molecule may be between 20 and 220 mg/ml. In another embodiment of the invention, the concentration of the neutralizing binding molecule may be between 20 and 210 mg/ml. In another embodiment of the present invention, the concentration of the neutralizing binding molecule may be between 20 and 200 mg/ml.
  • the concentration of the neutralizing binding molecule may be 30 to 240 mg/ml. In another embodiment of the present invention, the concentration of the neutralizing binding molecule may be between 30 and 230 mg/ml. In another embodiment of the present invention, the concentration of the neutralizing binding molecule may be between 30 and 220 mg/ml. In another embodiment of the present invention, the concentration of the neutralizing binding molecule may be between 30 and 210 mg/ml. In another embodiment of the present invention, the concentration of the neutralizing binding molecule may be between 30 and 200 mg/ml.
  • the concentration of the neutralizing binding molecule may be 40 to 240 mg/ml. In another embodiment of the present invention, the concentration of the neutralizing binding molecule may be between 40 and 230 mg/ml. In another embodiment of the present invention, the concentration of the neutralizing binding molecule may be between 40 and 220 mg/ml. In another embodiment of the present invention, the concentration of the neutralizing binding molecule may be between 40 and 210 mg/ml. In another embodiment of the present invention, the concentration of the neutralizing binding molecule may be between 40 and 200 mg/ml.
  • the concentration of the neutralizing binding molecule may be from 1 to 100 mg/ml. In another embodiment of the present invention, the concentration of the neutralizing binding molecule may be 1 to 90 mg/ml. In another embodiment of the present invention, the concentration of the neutralizing binding molecule may be 1 to 80 mg/ml. In another embodiment of the present invention, the concentration of the neutralizing binding molecule may be between 1 and 70 mg/ml. In another embodiment of the present invention, the concentration of the neutralizing binding molecule may be between 1 and 60 mg/ml.
  • the concentration of the neutralizing binding molecule may be between 5 and 100 mg/ml. According to another embodiment of the present invention, the concentration of the neutralizing binding molecule may be 5 to 90 mg/ml. In another embodiment of the present invention, the concentration of the neutralizing binding molecule may be 5 to 80 mg/ml. In another embodiment of the present invention, the concentration of the neutralizing binding molecule may be 5 to 70 mg/ml. In another embodiment of the present invention, the concentration of the neutralizing binding molecule may be between 5 and 60 mg/ml.
  • the concentration of the neutralizing binding molecule may be between 10 and 100 mg/ml. According to another embodiment of the present invention, the concentration of the neutralizing binding molecule may be 10 to 90 mg/ml. In another embodiment of the present invention, the concentration of the neutralizing binding molecule may be between 10 and 80 mg/ml. In another embodiment of the present invention, the concentration of the neutralizing binding molecule may be between 10 and 70 mg/ml. In another embodiment of the present invention, the concentration of the neutralizing binding molecule may be between 10 and 60 mg/ml.
  • the concentration of the neutralizing binding molecule may be between 20 and 100 mg/ml. According to another embodiment of the present invention, the concentration of the neutralizing binding molecule may be 20 to 90 mg/ml. In another embodiment of the present invention, the concentration of the neutralizing binding molecule may be between 20 and 80 mg/ml. In another embodiment of the present invention, the concentration of the neutralizing binding molecule may be between 20 and 70 mg/ml. In another embodiment of the present invention, the concentration of the neutralizing binding molecule may be between 20 and 60 mg/ml.
  • the concentration of the neutralizing binding molecule may be between 30 and 100 mg/ml. According to another embodiment of the present invention, the concentration of the neutralizing binding molecule may be 30 to 90 mg/ml. In another embodiment of the invention, the concentration of the neutralizing binding molecule may be between 30 and 80 mg/ml. In another embodiment of the present invention, the concentration of the neutralizing binding molecule may be between 30 and 70 mg/ml. In another embodiment of the present invention, the concentration of the neutralizing binding molecule may be between 30 and 60 mg/ml.
  • the concentration of the neutralizing binding molecule may be between 40 and 100 mg/ml. According to another embodiment of the present invention, the concentration of the neutralizing binding molecule may be 40 to 90 mg/ml. In another embodiment of the present invention, the concentration of the neutralizing binding molecule may be 40 to 80 mg/ml. In another embodiment of the present invention, the concentration of the neutralizing binding molecule may be 40 to 70 mg/ml. In another embodiment of the present invention, the concentration of the neutralizing binding molecule may be between 40 and 60 mg/ml.
  • the concentration of the neutralizing binding molecule may be between 50 and 100 mg/ml. According to another embodiment of the present invention, the concentration of the neutralizing binding molecule may be 50 to 90 mg/ml. In another embodiment of the present invention, the concentration of the neutralizing binding molecule may be between 50 and 80 mg/ml. In another embodiment of the present invention, the concentration of the neutralizing binding molecule may be between 50 and 70 mg/ml. In another embodiment of the present invention, the concentration of the neutralizing binding molecule may be between 50 and 60 mg/ml.
  • the buffer according to the present invention is a neutralizing material that minimizes changes in pH due to acid or alkali
  • the buffer is i) histidine, histidine salt or a mixture thereof, ii) acetate, iii) citrate, iv) succinate, v) phosphate, or vi) gluconate, but is not limited thereto.
  • the histidine salt may include histidine chloride, histidine acetate, histidine phosphate, histidine sulfate, and the like.
  • it may be preferable in terms of pH control and stability to include histidine as a buffer.
  • the content of the buffer can be freely adjusted within a range that does not substantially adversely affect the stability and viscosity of the pharmaceutical formulation according to the present invention.
  • the content of the buffer may be 1 to 100 mM.
  • the content of the buffer may be 1 to 90 mM.
  • the content of the buffer may be 1 to 80 mM.
  • the content of the buffer may be 1 to 70 mM.
  • the content of the buffer may be 1 to 60 mM.
  • the content of the buffer may be 1 to 50 mM.
  • the content of the buffer may be 2 to 100 mM. In another embodiment of the present invention, the content of the buffer may be 2 to 90 mM. In another embodiment of the present invention, the content of the buffer may be 2 to 80 mM. In another embodiment of the present invention, the content of the buffer may be 2 to 70 mM. In another embodiment of the present invention, the content of the buffer may be 2 to 60 mM. In another embodiment of the present invention, the content of the buffer may be 2-50 mM.
  • the content of the buffer may be 3 to 100 mM. In another embodiment of the present invention, the content of the buffer may be 3 to 90 mM. In another embodiment of the present invention, the content of the buffer may be 3 to 80 mM. In another embodiment of the present invention, the content of the buffer may be 3 to 70 mM. In another embodiment of the present invention, the content of the buffer may be 3 to 60 mM. In another embodiment of the present invention, the content of the buffer may be 3 to 50 mM.
  • the content of the buffer may be 4 to 100 mM. In another embodiment of the present invention, the content of the buffer may be 4 to 90 mM. In another embodiment of the present invention, the content of the buffer may be 4 to 80 mM. In another embodiment of the present invention, the content of the buffer may be 4 to 70 mM. In another embodiment of the present invention, the content of the buffer may be 4 to 60 mM. In another embodiment of the present invention, the content of the buffer may be 4-50 mM.
  • the content of the buffer may be 5 to 100 mM. In another embodiment of the present invention, the content of the buffer may be 5 to 90 mM. In another embodiment of the present invention, the content of the buffer may be 5 to 80 mM. In another embodiment of the present invention, the content of the buffer may be 5 to 70 mM. In another embodiment of the present invention, the content of the buffer may be 5 to 60 mM. In another embodiment of the present invention, the content of the buffer may be 5 to 50 mM.
  • the content of the buffer may be 1 to 20 mM. In another embodiment of the present invention, the content of the buffer may be 5 to 15 mM. In another embodiment of the present invention, the content of the buffer may be 7 to 13 mM. In another embodiment of the present invention, the content of the buffer may be 10 mM.
  • the stabilizer according to the present invention consists of i) a metal salt, ii) a sugar or a derivative thereof, and iii) an amino acid (provided that it is different from the amino acid contained in the (B) buffer) or a salt thereof. It may be any one or more selected from the group, preferably an amino acid.
  • the stabilizer may be a metal salt, and an anion thereof may be included together.
  • the metal salt may include sodium chloride (NaCl), potassium chloride (KCl), calcium chloride (CaCl), or a mixture of two or more thereof.
  • the stabilizer may be a sugar or a derivative of sugar.
  • the sugar may be a monosaccharide, a disaccharide, an oligosaccharide, a polysaccharide, or a mixture of two or more thereof.
  • monosaccharides include, but are not limited to, glucose, fructose, galactose, and the like.
  • disaccharides include, but are not limited to, sucrose, lactose, maltose, trehalose, and the like.
  • oligosaccharides include, but are not limited to, fructooligosaccharides, galactooligosaccharides, mannan oligosaccharides, and the like.
  • polysaccharides include, but are not limited to, starch, glycogen, cellulose, chitin, pectin, and the like.
  • the sugar derivative may be a sugar alcohol, a sugar acid, or a mixture thereof.
  • sugar alcohols include glycerol, erythritol, threitol, arabitol, xylitol, ribitol, mannitol, sorbitol, galactitol, fusitol, iditol, inositol, bolemitol, isomalt, maltitol, lactitol, maltotriitol , maltotetraitol, polyglycitol, and the like, but is not limited thereto.
  • sugar acids include, but are not limited to, aldonic acid (such as glyceric acid), ulosonic acid (such as neuramic acid), uronic acid (such as glucuronic acid), and aldaric acid (such as tartaric acid).
  • aldonic acid such as glyceric acid
  • ulosonic acid such as neuramic acid
  • uronic acid such as glucuronic acid
  • aldaric acid such as tartaric acid
  • the sugar or its derivative may be sorbitol, mannitol, trehalose, sucrose, or a mixture of two or more thereof, preferably trehalose.
  • the stabilizing agent may be an amino acid or a salt of an amino acid.
  • the amino acid may be glycine, arginine, threonine, methionine, or a mixture of two or more thereof.
  • the salt of the amino acid may be L- arginine monohydrochloride (monohydrochloride), but is not limited thereto.
  • the stabilizer may be preferably L-arginine monohydrochloride.
  • the content of the stabilizer can be freely adjusted within a range that does not substantially adversely affect the stability and viscosity of the pharmaceutical formulation according to the present invention.
  • the content of the stabilizer may be 50 to 300 mM or 1 to 20% (w/v), but is not limited thereto.
  • the content of the stabilizer is 50 to 300 mM, 50 to 290 mM, 50 to 280 mM, 50 to 270 mM, 50 to 260 mM or 50 to 250 mM.
  • the content of the stabilizer is 60 to 300 mM, 60 to 290 mM, 60 to 280 mM, 60 to 270 mM, 60 to 260 mM or from 60 to 250 mM.
  • the content of the stabilizer is 70 to 300 mM, 70 to 290 mM, 70 to 280 mM, 70 to 270 mM, 70 to 260 mM or 70 to 250 mM.
  • the content of the stabilizer is 80 to 300 mM, 80 to 290 mM, 80 to 280 mM, 80 to 270 mM, 80 to 260 mM or 80 to 250 mM.
  • the content of the stabilizer is 90 to 300 mM, 90 to 290 mM, 90 to 280 mM, 90 to 270 mM, 90 to 260 mM or from 90 to 250 mM.
  • the content of the stabilizer is 100 to 300 mM, 100 to 290 mM, 100 to 280 mM, 100 to 270 mM, 100 to 260 mM or from 100 to 250 mM.
  • the content of the stabilizer is 1 to 20% (w/v), based on 100% by volume of the total stable pharmaceutical formulation of the present invention; 1 to 18% (w/v), 1 to 16% (w/v), 1 to 14% (w/v), 1 to 12% (w/v) or 1 to 10% (w/v) days can be 1 to 20% (w/v), based on 100% by volume of the total stable pharmaceutical formulation of the present invention; 1 to 18% (w/v), 1 to 16% (w/v), 1 to 14% (w/v), 1 to 12% (w/v) or 1 to 10% (w/v) days can
  • the content of the stabilizer is 2 to 20% (w/v), based on 100% by volume of the total stable pharmaceutical formulation of the present invention; 2 to 18% (w/v), 2 to 16% (w/v), 2 to 14% (w/v), 2 to 12% (w/v) or 2 to 10% (w/v) days can
  • the content of the stabilizer is 3 to 20% (w/v), based on 100% by volume of the total stable pharmaceutical formulation of the present invention; 3 to 18% (w/v), 3 to 16% (w/v), 3 to 14% (w/v), 3 to 12% (w/v) or 3 to 10% (w/v) days can
  • the content of the stabilizer is 4 to 20% (w/v), based on 100% by volume of the total stable pharmaceutical formulation of the present invention; 4 to 18% (w/v), 4 to 16% (w/v), 4 to 14% (w/v), 4 to 12% (w/v) or 4 to 10% (w/v) days can
  • the content of the stabilizer is 5 to 20% (w/v), based on 100% by volume of the total stable pharmaceutical formulation of the present invention; 5 to 18% (w/v), 5 to 16% (w/v), 5 to 14% (w/v), 5 to 12% (w/v) or 5 to 10% (w/v) days can
  • the stabilizer Within the content range of the stabilizer, high molecular weight or low molecular weight components are maintained low for a long period of time, the content of intact immunoglobulin G component or intact heavy and light chains is maintained high, and long-term stability is excellent and low viscosity. .
  • the surfactant according to the present invention is polyoxyethylene sorbitan fatty acid ester (eg, polysorbate), polyoxyethylene alkyl ether (eg, Brij), alkylphenylpolyoxyethylene ethers (eg Triton-X), polyoxyethylene-polyoxypropylene copolymers (eg Poloxamer, Pluronic), sodium dodecyl sulfate (SDS), poloxamers and mixtures thereof, and the like, but are limited thereto. it is not going to be
  • the surfactant may be polysorbate, poloxamer, or a mixture thereof, preferably polyoxyethylene sorbitan fatty acid ester (polysorbate), poloxamer, or a mixture thereof.
  • the polysorbate may be polysorbate 20, polysorbate 40, polysorbate 60, polysorbate 80, or a mixture of two or more thereof, but is not limited thereto.
  • the poloxamer may be poloxamer 188, but is not limited thereto.
  • the surfactant may most preferably be polysorbate 80.
  • the concentration of the surfactant can be freely adjusted within a range that does not adversely affect the stability and viscosity of the stable pharmaceutical formulation according to the present invention.
  • the concentration of the surfactant according to the present invention is 0.01 to 0.1% (w/v), preferably 0.01 to 0.08% (w/v), more preferably 0.01 to 0.07% (w/v) days can
  • the concentration of the surfactant may be most preferably 0.05% (w/v).
  • the pH of the composition according to the present invention may be 5.0 to 7.0, 5.5 to 7.0, 5.7 to 7.0, 6.0 to 7.0, 6.3 to 7.0 or 6.5 to 7.0.
  • the pH of the composition according to the present invention may be 5.5 to 6.5, 5.7 to 6.5, 6.0 to 6.5, or 6.3 to 6.5.
  • the pH of the composition according to the present invention may be 5.5 to 6.3, 5.7 to 6.3, or 6.0 to 6.3.
  • the pH of the composition according to the present invention may be 5.5 to 6.0, and may be 5.7 to 6.0.
  • the pH of the composition according to the present invention may preferably be 6.0. Within the above pH range, it can exhibit excellent stability and low viscosity for a long period of time.
  • the pH can be adjusted using a buffer, and when the buffer is included in a predetermined amount, the pH of the above range can be exhibited without a separate pH adjusting agent.
  • stable means that the neutralizing binding molecule according to the present invention substantially retains physical stability, chemical stability and/or biological activity during the manufacturing process and/or upon storage/storage.
  • various analytical techniques for measuring the stability of an antibody that are readily available in the art may be used.
  • the physical stability can be evaluated by methods known in the art, which include measuring the sample apparent attenuation of light (absorption or optical density). This light attenuation measurement is related to the turbidity of the formulation.
  • the physical stability can be measured by high molecular weight component content, low molecular weight component content, intact protein amount, the number of insoluble foreign particles, and the like.
  • the chemical stability can be evaluated, for example, by detecting and quantifying a chemically changed form of a neutralizing binding molecule.
  • Such chemical stability includes charge changes (eg, occurring as a result of deamidation or oxidation) that can be assessed, for example, by ion exchange chromatography.
  • Chemical stability can be measured with charge variants (acidic or basic peaks) or the like.
  • the biological activity may be evaluated by a method known in the art, for example, antigen binding affinity may be measured through ELISA.
  • stable pharmaceutical preparation refers to a pharmaceutical preparation satisfying one or more of the following.
  • a pharmaceutical formulation having an absorbance A600 of 0 to 0.0700 or 0 to 0.0400 measured with a spectrophotometer after storage for 4 weeks at a temperature of 5 ⁇ 3° C.;
  • a pharmaceutical formulation having an absorbance A600 of 0 to 0.0700 or 0 to 0.0400 measured with a spectrophotometer after storage for 4 weeks at a temperature of 40 ⁇ 2° C., a relative humidity of 75 ⁇ 5%, and a closed condition;
  • the number of insoluble foreign particles (10.00 ⁇ m ⁇ , ⁇ 400.00 ⁇ m) measured by HIAC after storage for 4 weeks at a temperature of 40 ⁇ 2°C, a relative humidity of 75 ⁇ 5%, and a closed condition is 0 to 200 pharmaceutical formulations ;
  • the number of insoluble foreign particles (25.00 ⁇ m ⁇ , ⁇ 400.00 ⁇ m) measured by HIAC after storage for 4 weeks at a temperature of 40 ⁇ 2°C, a relative humidity of 75 ⁇ 5%, and a closed condition is 0 to 50 pharmaceutical formulations ;
  • the number of insoluble foreign particles (1.00 ⁇ m ⁇ , ⁇ 400.00 ⁇ m) measured by MFI after storage for 4 weeks at a temperature of 40 ⁇ 2°C, a relative humidity of 75 ⁇ 5%, and a closed condition is 0 to 20,000 pharmaceutical formulations ;
  • the number of insoluble foreign particles (10.00 ⁇ m ⁇ , ⁇ 400.00 ⁇ m) measured by MFI after storage for 4 weeks at a temperature of 40 ⁇ 2°C, a relative humidity of 75 ⁇ 5%, and a closed condition is 0 to 300 pharmaceutical formulations ;
  • the number of insoluble foreign particles (25.00 ⁇ m ⁇ , ⁇ 400.00 ⁇ m) measured by MFI after storage for 4 weeks at a temperature of 40 ⁇ 2°C, a relative humidity of 75 ⁇ 5%, and a closed condition is 0 to 30 pharmaceutical formulations ;
  • the stable pharmaceutical formulation of the present invention can be prepared using a known method, and is not limited to a specific manufacturing method. For example, after adjusting the pH by adding a buffer to a solution containing a stabilizer and a surfactant, a neutralizing binding molecule may be added to the mixed solution to prepare a pharmaceutical formulation. In addition, after preparing a solution containing some excipients in the final step of the purification process, the remaining ingredients may be added to prepare a pharmaceutical formulation.
  • the preparation may not include a freeze-drying process during manufacture. If the freeze-drying process is not included, for example, the pharmaceutical formulation of the present invention may be prepared and placed in an airtight container such as a glass vial or pre-filled syringe, which is a primary packaging material, immediately after treatment such as sterilization.
  • an airtight container such as a glass vial or pre-filled syringe, which is a primary packaging material
  • the stable pharmaceutical formulation of the present invention is (A) a neutralizing binding molecule 5 that binds to the spike protein (S protein) on the surface of SARS-CoV-2 (SARS-CoV-2) to 240 mg/ml; (B) 1-50 mM buffer; (C) 50-200 mM stabilizer; and (D) 0.01 to 0.1% (w/v) of a surfactant.
  • S protein spike protein
  • SARS-CoV-2 SARS-CoV-2
  • D 0.01 to 0.1% (w/v) of a surfactant.
  • the present invention provides the above stable pharmaceutical formulation; And it may provide a product comprising a container for accommodating the stable pharmaceutical formulation in a closed state.
  • the stable pharmaceutical formulation is as described above.
  • the container may be formed of a material such as glass, polymer (plastic), or metal, but is not limited thereto.
  • the container may be a bottle, a vial, a cartridge, a syringe (pre-filled syringe), or a tube, but is not limited thereto.
  • the container may be a glass or polymer vial, or a glass or polymer pre-filled syringe.
  • the product may further include instructions for providing a method of use, a method of storage, or both of the stable pharmaceutical formulation.
  • the product may include other tools necessary from a commercial and user point of view, for example, needles, syringes, and the like.
  • binding molecule refers to an intact immunoglobulin, including monoclonal antibodies, such as chimeric, humanized or human monoclonal antibodies, or antigen-binding, which is an immunoglobulin that binds to an antigen. Includes fragments. For example, in binding to the spike protein of SARS-CoV-2, it refers to a variable domain, enzyme, receptor, or protein comprising an immunoglobulin fragment that competes with an intact immunoglobulin. Regardless of structure, the antigen-binding fragment binds to the same antigen recognized by the intact immunoglobulin.
  • the antigen-binding fragment comprises at least two contiguous groups of the amino acid sequence of the antibody, at least 20 contiguous amino acid residues, at least 25 contiguous amino acid residues, at least 30 contiguous amino acid residues, at least 35 contiguous amino acid residues, at least 40 contiguous amino acid residues , at least 50 contiguous amino acid residues, at least 60 contiguous amino acid residues, at least 70 contiguous amino acid residues, at least 80 contiguous amino acid residues, at least 90 contiguous amino acid residues, at least 100 contiguous amino acid residues, at least 125 contiguous amino acid residues, a peptide or polypeptide comprising an amino acid sequence of at least 150 contiguous amino acid residues, at least 175 contiguous amino acid residues, at least 200 contiguous amino acid residues, or at least 250 contiguous amino acid residues.
  • the term "antigen-binding fragment” particularly refers to Fab, F(ab'), F(ab')2, Fv, dAb, Fd, complementarity determining region (CDR) fragments, single-chain antibodies (scFv). , bivalent single-chain antibodies, single-chain phage antibodies, diabodies, triabodies, tetrabodies, polypeptides containing one or more fragments of an immunoglobulin sufficient to bind a particular antigen to the polypeptide. etc.
  • the fragments may be produced synthetically or by enzymatic or chemical digestion of complete immunoglobulins, or may be genetically engineered by recombinant DNA techniques. Methods of production are well known in the art.
  • the stable pharmaceutical formulation according to the present invention has excellent long-term storage stability under temperature conditions such as accelerated conditions and severe conditions, and can maintain excellent stability even under physical stress conditions such as light, freezing/thawing, and shaking.
  • FIG. 10 is a measurement result of the content of the main component at a temperature of 5 ⁇ 3° C. in Examples 1 and 7 to 16.
  • FIG. 10 is a measurement result of the content of the main component at a temperature of 5 ⁇ 3° C. in Examples 1 and 7 to 16.
  • FIG. 11 is a measurement result of the content of the main component at a temperature of 25 ⁇ 2° C. in Examples 1 and 7 to 16.
  • FIG. 11 is a measurement result of the content of the main component at a temperature of 25 ⁇ 2° C. in Examples 1 and 7 to 16.
  • FIG. 12 is a measurement result of the content of the main component at a temperature of 40 ⁇ 2° C. in Examples 1 and 7 to 16.
  • FIG. 12 is a measurement result of the content of the main component at a temperature of 40 ⁇ 2° C. in Examples 1 and 7 to 16.
  • FIG. 23 is a measurement result of intact immunoglobulin G content of Examples 1, 20 to 31, and Comparative Example 1.
  • a buffer solution containing a stabilizer was prepared at a pH that showed the optimal buffering ability, and then SARS-CoV-2 neutralizing binding molecules were added to the solution, and a surfactant was added. was added to reach the target concentration to prepare the corresponding formulation component.
  • the absorbance at 600 nm was measured using a UV-Vis spectrophotometer.
  • the main component content (%) was measured using size exclusion high performance liquid chromatography (Size Exclusion HPLC).
  • the content (pre-peak; %) of the high molecular weight component was measured using size exclusion high performance liquid chromatography (Size Exclusion HPLC).
  • the content (post-peak; %) of low molecular weight components was measured using size exclusion high performance liquid chromatography (Size Exclusion HPLC).
  • the content of intact immunoglobulin G was measured using Labchip GXII, a non-reducing chip-based CE-SDS analysis equipment.
  • the contents of light and heavy chains of the antibody were measured using Labchip GXII, a reduced chip-based CE-SDS analysis equipment.
  • MFI Micro Flow Imaging
  • HIAC 9703 Light-shielding particle counter
  • the oxidation rate (%) of heavy chain Met 263 was measured through peptide mapping by liquid chromatography (LC-MS) through mass spectrometry.
  • SARS-CoV-2 RBD binding affinity (%) was measured by Enzyme-Linked ImmunoSorbent Assay (ELISA).
  • each buffer was prepared to suit each pH, and arginine monohydrochloride, sorbitol and trehalose were added thereto, an antibody was added thereto, and a surfactant was added to the sample of Table 5 were manufactured.
  • the pharmaceutical formulations prepared according to Examples 1 to 3 were stored at a temperature of 5 ⁇ 3° C. and a temperature of 50 ⁇ 2° C., stability after 5 days at a temperature of 5 ⁇ 3° C., and after 3 days and 5 at a temperature of 50 ⁇ 2° C. Stability after one day was measured. For physical stress, freezing and thawing at -40°C were repeated 5 times. The results of the experiment are shown in Tables 6 to 18 and FIGS. 1 to 8 .
  • the antibody used in Experimental Example 1 was No. 1 described in Tables 1 to 2 above. 139 is the binding molecule.
  • the formulation prepared with the following ingredients had excellent stability under temperature conditions such as accelerated conditions and severe conditions, and maintained excellent stability even under physical stress conditions such as freezing/thawing.
  • the main component content and the intact immunoglobulin G content in the formulation containing L-arginine monohydrochloride were more stable under high temperature conditions than the formulation containing sorbitol and trehalose.
  • Example 1 histidine 10 mM 6.0 L-Arginine monohydrochloride 150 mM Polysorbate 800.05% (w/v) 60mg/mL
  • Example 2 histidine 10 mM 6.0 Sorbitol 5% (w/v) Polysorbate 800.05% (w/v) 60mg/mL
  • Example 3 histidine 10 mM 6.0 Trehalose 10% (w/v) Polysorbate 800.05% (w/v) 60mg/mL
  • Example 1 0.0370 0.0055 0.0096 0.0108 0.0068
  • Example 2 0.0121 0.0162 0.0157 0.0412 0.0070
  • Example 3 0.0082 0.0037 0.0090 0.0059 0.0059
  • Example 1 the turbidity of Example 1 was 0.0400 or less under conditions of 5 ⁇ 3° C., 50 ⁇ 2° C. and freeze/thaw stress. In the case of Example 2, the turbidity of Example 1 was higher than that of 50 ⁇ 2° C. conditions.
  • Table 7 shows the measurement results of the main component content (Main peak %) by size exclusion chromatography.
  • Example 1 contained the highest content of main components under the conditions of 5 ⁇ 3 °C and 50 ⁇ 2 °C. It can be seen that the main component content of Examples 1 to 3 is stable to 98.0% or more after 5 days at 50 ⁇ 2° C. condition. The main component content by freezing/thawing stress was similar to Examples 1 to 3, and it was found that it was stable at 98.0% or more (FIG. 1).
  • Table 8 shows the results of size exclusion chromatography high molecular weight component content (pre-peak %) measurement.
  • Example 1 Referring to Table 8, the conditions of 5 ⁇ 3° C. after 5 days and 50 ⁇ 2° C. after 3 days and 5 days of Example 1 showed the lowest content of high molecular weight components. It was found that the high molecular weight component content of Examples 1 to 3 was stable at 50 ⁇ 2° C. after 5 days at 1.00% or less. The high molecular weight content due to freezing/thawing stress was similar to Examples 1 to 3, and it was found that it was stable at 1.00% or less ( FIG. 2 ).
  • Table 9 shows the measurement results of the low molecular weight component content (post-peak %) by size exclusion chromatography.
  • Example 1 0.04 0.04 0.18 0.31 0.05
  • Example 2 0.04 0.04 0.16 0.27 0.04
  • Example 3 0.04 0.05 0.16 0.30 0.03
  • Table 10 below shows the measurement results of intact immunoglobulin G content (non-reducing chip-based CE-SDS).
  • Example 1 the highest intact immunoglobulin G content was shown in Example 1 at 5 ⁇ 3° C. after 5 days and at 50 ⁇ 2° C. after 3 and 5 days. It was found that the content of intact immunoglobulin G of Examples 1 to 3 was stable to 90.0% or more after 5 days at 50 ⁇ 2°C.
  • the high molecular weight content due to freezing/thawing stress was similar to Examples 1 to 3, and it was found that it was stable at 90.0% or more (FIG. 4).
  • Table 11 below shows the measurement results of the antibody light chain and heavy chain content (reduced chip-based CE-SDS).
  • Table 12 below shows the measurement results of the charge variant (main peak %).
  • Example 1 65 65.48 64.67 63.35 65.56
  • Example 2 65.12 65.39 64.21 62.67 65.39
  • Example 3 65.14 64.92 64.03 62.92 65.25
  • Table 13 shows the measurement results of the charge variant (acid peak %).
  • Table 14 shows the measurement results of charge variants (basic peak %).
  • Table 15 shows the measurement results of the number of insoluble foreign particles (10.00 ⁇ m ⁇ , ⁇ 100.00 ⁇ m) measured by MFI.
  • Table 16 shows the measurement results of the number of insoluble foreign particles (25.00 ⁇ m ⁇ , ⁇ 100.00 ⁇ m) measured by MFI.
  • Table 17 below shows the measurement results of the number of insoluble foreign particles (10.00 ⁇ (um)) measured by HIAC.
  • Example 1 is a formulation finally selected according to Experimental Example 1.
  • the prepared pharmaceutical formulation was stored at 5 ⁇ 3°C temperature, 40 ⁇ 2°C temperature and 75 ⁇ 5% relative humidity, and stability after 2 and 4 weeks at 5 ⁇ 3°C temperature and 40 ⁇ 2°C temperature and 75 ⁇ Stability after 2 weeks and 4 weeks at 5% relative humidity was measured. For physical stress, shaking stress was applied at 3000 rpm at room temperature for 4 hours, and the results are shown in Tables 20 to 27 and FIG. 9 .
  • Antibodies used in Experimental Example 2 were No. 1 described in Tables 1 to 2 above. 139 is the binding molecule.
  • Example 4 histidine 10 mM 6.0 L-Arginine monohydrochloride 150 mM Polysorbate 800.05% (w/v) 100mg/mL
  • Example 5 histidine 10 mM 6.0 L-Arginine monohydrochloride 150 mM Polysorbate 800.05% (w/v) 150mg/mL
  • Example 6 histidine 10 mM 6.0 L-Arginine monohydrochloride 150 mM Polysorbate 800.05% (w/v) 200mg/mL
  • Table 21 below shows the measurement results of SARS-CoV-2 RBD binding affinity (ELISA).
  • Table 22 below shows the measurement results of the main component content (Main peak %) by size exclusion chromatography.
  • Table 23 shows the measurement results of high molecular weight components (pre-peak %) by size exclusion chromatography.
  • Example 5 0.50 0.56 1.02 0.63 1.53 0.51
  • Example 6 0.55 0.67 1.19 0.88 1.31 0.65
  • Example 1 0.37 0.40 0.53 0.36 0.65 0.38
  • Table 24 shows the results of measurement of low molecular weight components (post-peak %) by size exclusion chromatography.
  • Table 25 shows the measurement results of the number of insoluble foreign particles (10.00 ⁇ (um)) measured by HIAC.
  • Table 26 shows the measurement results of the number of insoluble foreign particles (25.00 ⁇ (um)) measured by HIAC.
  • Example 26 the number of insoluble foreign particles (25.00 ⁇ (um)) of Examples 4 to 6 was stable to 10 or less under all experimental conditions, and it was found to be at a level similar to the result of Example 1.
  • Table 27 shows the oxidation rate Oxidation (Met 263) measurement results.
  • Example 1 is a formulation finally selected according to Experimental Example 1.
  • the prepared pharmaceutical formulations were stored at 5 ⁇ 3°C temperature, 25 ⁇ 2°C temperature, 60 ⁇ 5% relative humidity, 40 ⁇ 2°C temperature and 75 ⁇ 5% relative humidity, and each temperature condition for 3 weeks and 6 weeks and stability after 9 weeks.
  • shaking stress was applied at 3000 rpm at room temperature for 4 hours, freezing/thawing stress was repeated 5 times at -40 ° C.
  • the results are shown in Tables 29 to 43 and FIGS. 10 to 17 .
  • the antibody used in Experimental Example 3 was No. 1 described in Tables 1 to 2 above. 139 is the binding molecule.
  • Example 7 histidine 5 mM 6.0 L-Arginine monohydrochloride 150 mM Polysorbate 800.05% (w/v) 60mg/mL
  • Example 8 Histidine 15 mM 6.0 L-Arginine monohydrochloride 150 mM Polysorbate 800.05% (w/v) 60mg/mL
  • Example 9 histidine 10 mM 5.7 L-Arginine monohydrochloride 150 mM Polysorbate 800.05% (w/v) 60mg/mL
  • Example 10 histidine 10 mM 6.3 L-Arginine monohydrochloride 150 mM Polysorbate 800.05% (w/v) 60mg/mL
  • Example 11 histidine 10 mM 6.0 L-Arginine monohydrochloride 100 mM Polysorbate 800.05% (w/v) 60mg/mL
  • Example 12 histidine 10 mM 6.0 L-Arginine monohydrochloride 200 mM Polysorbate 800.05%
  • Example 7 0.0114 0.0019 0.0059 0.0039 0.0100 0.0066 0.0095 0.0096 0.0063 0.0104 0.0099 0.0114 0.0121
  • Example 8 0.0121 0.0103 0.0086 0.0076 0.0096 0.0144 0.0075 0.0080 0.0089 0.0148 0.0034 0.0103 0.0231
  • Example 9 0.0071 0.0064 0.0050 0.0082 0.0103 0.0089 0.0064 0.0100 0.0126 0.0090 0.0070 0.0109 0.0171
  • Example 10 0.0097 0.0095 0.0070 0.0072 0.0100 0.0108 0.0085 0.0102 0.0072 0.0111 0.00
  • Table 30 below shows the measurement results of the main component content (Main peak %) by size exclusion chromatography.
  • Example 7 99.47 99.50 99.36 98.96 99.44 99.28 98.53 99.42 99.19 98.24 99.46 99.39 99.12
  • Example 8 99.45 99.53 99.41 98.99 99.44 99.29 98.54 99.45 99.14 98.20 99.48 99.43 99.09
  • Example 9 99.53 99.54 99.46 98.76 99.45 99.36 98.45 99.47 99.15 97.82 99.29 99.46 99.19
  • Example 10 99.26 99.46 99.29 98.87 99.37 99.14 98.43 99.
  • Example 30 As shown in Table 30, it was found that the main component content of Examples 7 to 16 was stable to 99% or more at 5 ⁇ 3° C. temperature conditions, and was stable to 97% or more at 40 ⁇ 2° C. temperature and 75 ⁇ 5% relative humidity conditions. . It was also found that the level was similar to that of Example 1 ( FIGS. 10 , 11 and 12 ).
  • Table 31 shows the measurement results of high molecular weight components (pre-peak %) by size exclusion chromatography.
  • Example 7 0.48 0.45 0.53 0.68 0.51 0.60 0.85 0.52 0.61 0.81 0.47 0.57 0.79
  • Example 8 0.48 0.41 0.49 0.63 0.51 0.59 0.82 0.46 0.65 0.80 0.44 0.53 0.82
  • Example 9 0.41 0.40 0.45 0.66 0.50 0.52 0.78 0.46 0.63 0.89 0.64 0.50 0.68
  • Example 10 0.66 0.49 0.60 0.77 0.59 0.73 0.97 0.55 0.74 0.93 0.52 0.66 0.75
  • Example 11 0.57 0.47 0.55 0.71 0.52 0.63 0.88 0.49 0.64 0.87 0.47 0.59 0.74
  • Example 12 0.49 0.45 0.49 0.67 0.49 0.58 0.85 0.
  • the high molecular weight component content of Examples 7 to 16 is stable to 1.0% or less at 5 ⁇ 3° C. temperature conditions, and is stable to 2.0% or less at 40 ⁇ 2° C. temperature and 75 ⁇ 5% relative humidity conditions. could In addition, it was found that the level was similar to the result of Example 1.
  • Table 32 below shows the measurement results of low molecular weight components (post-peak %) by size exclusion chromatography.
  • Example 7 0.05 0.05 0.12 0.37 0.04 0.12 0.62 0.05 0.20 0.96 0.07 0.04 0.09
  • Example 8 0.07 0.06 0.10 0.37 0.05 0.12 0.64 0.10 0.21 0.99 0.08 0.05 0.09
  • Example 9 0.06 0.06 0.09 0.58 0.05 0.12 0.77 0.06 0.22 1.29 0.07 0.04 0.13
  • Example 10 0.07 0.05 0.11 0.36 0.05 0.13 0.60 0.08 0.21 0.99 0.08 0.04 0.10
  • Example 11 0.06 0.06 0.11 0.39 0.04 0.12 0.62 0.07 0.21 0.95 0.08 0.06 0.11
  • Example 12 0.07 0.05 0.09 0.38 0.04 0.12 0.63 0.
  • Example 32 As shown in Table 32, it can be seen that the low molecular weight component content of Examples 7 to 16 is stable to 0.5% or less at 5 ⁇ 3° C. temperature conditions, and is stable to 1.5% or less at 40 ⁇ 2° C. temperature and 75 ⁇ 5% relative humidity conditions. could In addition, it was found that the level was similar to the result of Example 1.
  • Table 33 below shows the measurement results of the intact immunoglobulin G content (non-reducing chip-based CE-SDS).
  • Example 7 98.15 97.86 98.09 97.80 98.41 98.69 98.67 97.52 97.50 96.59 97.86 98.77 98.62
  • Example 8 98.17 97.90 97.90 97.68 98.43 98.74 98.75 97.61 97.32 96.58 97.74 98.95 98.80
  • Example 9 98.24 97.99 97.46 97.46 98.43 98.77 98.68 97.62 97.52 96.25 97.74 98.91
  • Table 34 below shows the measurement results of the antibody light chain and heavy chain content (reduced chip-based CE-SDS).
  • Table 35 shows the measurement results of the charge variant (main peak %).
  • Example 7 62.29 62.36 62.29 57.88 63.32 62.39 54.34 62.61 62.06 50.73 61.53 63.01 61.81
  • Example 8 62.34 62.61 62.52 58.48 63.37 63.00 54.59 62.91 63.12 49.88 61.51 63.07 61.81
  • Example 9 62.33 62.62 62.03 57.44 62.97 62.12 52.83 62.53 62.34 49.59 61.50 63.07 61.91
  • Example 10 Example 10
  • Table 36 shows the measurement results of the charge variant (acid peak %).
  • Table 37 shows the measurement results of charge variants (basic peak %).
  • Example 7 23.66 23.37 23.01 21.41 23.10 22.23 17.36 23.26 22.08 16.42 24.11 22.86 23.13
  • Example 8 23.53 23.27 22.53 19.99 22.95 21.34 16.30 22.93 21.06 15.83 24.02 22.71 23.09
  • Example 9 23.58 23.41 23.22 21.18 23.12 22.39 17.94 23.36 22.02 16.19 24.01 23.07 23.59
  • Example 10 24.20 23.21 22.41 19.07 22.86 21.06 15.61 23.07 20.69 15.18 23.78 22.59 22.62
  • Example 11 24.20 23.21 22.41 19.07 22.86 21.06 15.61 23.07 20.69 15.18 23.78 22.59 22.62
  • Example 11 24.20 23.21 22.41 19.07 22.86 21.06 15.61 23.07 20.69 15.18 23.78 22.59 22.62
  • Example 11 24.20 23.21 22.41 19.
  • Table 38 shows the oxidation rate oxidation (Met 263) measurement results.
  • Example 7 2.4 2.7 3.4 6.8 4.4
  • Example 8 2.7 2.8 3.4 7.0 4.9
  • Example 9 2.4 2.5 2.7 4.4 4.4
  • Example 10 2.4 2.8 3.9 8.4 4.8
  • Example 11 2.3 3.5 3.3 7.5 5.3
  • Example 12 2.3 2.7 5.4 8.3 5.0
  • Example 13 2.4 2.7 4.7 8.0 5.1
  • Example 14 2.4 2.8 3.8 7.7 4.5
  • Example 15 2.4 2.8 3.7 7.9 4.3
  • Example 16 2.4 2.8 4.0 8.4 5.2
  • Example 1 2.4 2.7 3.5 6.8 4.9
  • Table 39 below shows the measurement results of SARS-CoV-2 RBD binding affinity (ELISA).
  • Example 7 101 99 98 97 106 94 96 108 102 105 107
  • Example 8 100 97 106 95 104 96 101 100 105 102 107
  • Example 9 104 107 103 98 106 105 94 107 112 114
  • Example 10 101 102 98 101 96 96 99 98 107 104 106
  • Example 11 104 100 106 101 101 99 99 103 99 98 102
  • Example 12 97 100 102 101 99 105 99 108 107 113 97
  • Example 14 108 106 106 102 104 86 101 100 98 98 98
  • Table 40 shows the measurement results of the number of insoluble foreign particles (10.00 ⁇ m ⁇ , ⁇ 100.00 ⁇ m) measured by MFI.
  • Table 41 shows the measurement results of the number of insoluble foreign particles (25.00 ⁇ m ⁇ , ⁇ 100.00 ⁇ m) measured by MFI.
  • Table 42 below shows the measurement results of the number of insoluble foreign particles (10.00 ⁇ (um)) measured by HIAC.
  • Table 43 below shows the measurement results of the number of insoluble foreign particles (25.00 ⁇ (um)) measured by HIAC.
  • Example 1 16 mL of the stable liquid pharmaceutical formulation of Example 1 prepared by the method of Experimental Example 1 was stored in an airtight container at 5 ⁇ 3° C./ambient relative humidity. Stability was measured after 1 month, 2 months and 3 months at the above temperature and humidity.
  • Example 1 was found to be stable because the appearance was maintained without any change in appearance after 3 months at 5 ⁇ 3 °C.
  • Example 1 was found to be stable because it was kept constant without a change in the antibody concentration after 3 months at 5 ⁇ 3 °C.
  • Table 46 below shows the measurement results of the antibody light chain and heavy chain content (reduced CE-SDS).
  • Example 1 was found to be stable because it was kept constant without change in the antibody light chain and heavy chain content after 3 months at 5 ⁇ 3 °C.
  • Table 47 below shows the measurement results of intact immunoglobulin G content (non-reduced CE-SDS).
  • Example 1 was found to be stable since it was maintained at 5 ⁇ 3° C. for 3 months without change in the intact immunoglobulin G content.
  • Table 48 below shows the measurement results of SEC-HPLC (main component, high molecular weight component, low molecular weight component content).
  • Example 1 was found to be stable because it was kept constant without changes in the main component, high molecular weight component, and low molecular weight component content measured by SEC-HPLC after 3 months at 5 ⁇ 3 °C.
  • IEC-HPLC (main peak, acidic peak, and basic peak) measurement results are shown in Table 49 below.
  • Example 1 was found to be stable because it was kept constant without changes in the main peak, acidic peak, and basic peak contents measured by IEC-HPLC after 3 months at 5 ⁇ 3°C.
  • Table 50 below shows the results of SARS-CoV-2 RBD binding affinity measurement.
  • Example 1 was found to be stable because the SARS-CoV-2 RBD binding affinity was kept constant without change after 3 months at 5 ⁇ 3°C.
  • each buffer was prepared to suit each pH, and arginine monohydrochloride, sorbitol and trehalose were added thereto, an antibody was added thereto, and a surfactant was added to the sample of Table 51 were manufactured.
  • the prepared pharmaceutical formulation was stored at a temperature of 5 ⁇ 3° C. and 50 ⁇ 2° C., and stability after 5 days at a temperature of 5 ⁇ 3° C. and stability after 3 days and 5 days at a temperature of 50 ⁇ 2° C. were measured.
  • freezing and thawing were repeated 5 times at -40°C, and shaking stress was applied at room temperature at 3000 rpm for 4 hours.
  • the results of the experiment are shown in Tables 52 to 64 and FIGS. 18 to 22 .
  • Antibodies used in Experimental Example 5 were No. 1 described in Tables 3 to 4 above. It is the 322 bond molecule.
  • Table 53 below shows the measurement results of the antibody light chain and heavy chain content (reduced chip-based CE-SDS).
  • Table 54 below shows the measurement results of intact immunoglobulin G content (non-reducing chip-based CE-SDS).
  • Example 17 showed a lower content of intact immunoglobulin G compared to Examples 18 and 19 at 50 ⁇ 2° C. ( FIG. 19 ).
  • Table 55 shows the measurement results of charge variants (main peak %).
  • Table 57 shows the measurement results of charge variants (basic peak %).
  • Table 58 shows the measurement results of the main component content (Main peak %).
  • Example 17 showed a lower main component content than Examples 18 and 19 under the conditions of 50 ⁇ 2 °C.
  • the main component content due to freezing/thawing stress and shaking stress was similar to Examples 17 to 19, and it was found that it was stable at 99.0% or more ( FIG. 20 ).
  • Table 59 shows the measurement results of the high molecular weight component content (pre-peak %).
  • Example 17 was higher than that of Examples 18 and 19 at 50 ⁇ 2° C. ( FIG. 21 ).
  • Table 60 shows the measurement results of the low molecular weight component content (post-peak %).
  • Example 17 the low molecular weight component of Example 17 was higher than that of Examples 18 and 19 under the conditions of 50 ⁇ 2° C. ( FIG. 22 ).
  • Table 61 shows the measurement results of the number of insoluble foreign particles (10.00 ⁇ m ⁇ , ⁇ 100.00 ⁇ m) measured by MFI.
  • Table 62 shows the measurement results of the number of insoluble foreign particles (25.00 ⁇ m ⁇ , ⁇ 100.00 ⁇ m) measured by MFI.
  • Table 63 shows the measurement results of the number of insoluble foreign particles (10.00 ⁇ (um)) measured by HIAC.
  • Table 64 shows the measurement results of the number of insoluble foreign particles (25.00 ⁇ (um)) measured by HIAC.
  • each buffer solution (acetate, citrate, succinate, phosphate) was prepared according to each pH, and arginine monohydrochloride, sodium chloride, sucrose and glycine were added thereto. After addition of the antibody, it was concentrated. Thereafter, surfactants (polysorbates 20 and 80, poloxamer 188) were added (however, Comparative Example 1 was not added) to prepare the samples in Table 65.
  • Example 1 is a formulation finally selected according to Experimental Example 1. The prepared pharmaceutical formulation was stored at 5 ⁇ 3° C. temperature, 40 ⁇ 2° C. temperature, and 75 ⁇ 5% relative humidity, and stability after 2 weeks and 4 weeks at each temperature condition was measured. For physical stress, shaking stress was applied at 3000 rpm for 4 hours at room temperature. The results are shown in Tables 66 to 75 and FIGS. 23 to 25 .
  • the antibody used in Experimental Example 6 was No. 1 described in Tables 1 to 2 above. 139 is the binding molecule.
  • Example 20 Acetate 10 mM 5.5 L-Arginine monohydrochloride 150 mM Polysorbate 800.05% (w/v) 60mg/mL Example 21 Citrate 10 mM 5.5 L-Arginine monohydrochloride 150 mM Polysorbate 800.05% (w/v) 60mg/mL Example 22 Citrate 10 mM 6.0 L-Arginine monohydrochloride 150 mM Polysorbate 800.05% (w/v) 60mg/mL Example 23 Succinate 10 mM 6.5 L-Arginine monohydrochloride 150 mM Polysorbate 800.05% (w/v) 60mg/mL Example 24 Phosphate 10 mM 6.0 L-Arginine monohydrochloride 150 mM Polysorbate 800.05% (w/v) 60mg/mL Example 25 Phosphate 10 mM 6.5 L-Arginine monohydrochloride 150 mM Polysorbate 800.05% (w/v) 60mg/mL Example
  • Example 20 0.0098 0.0124 0.0072 0.0098 0.0112 0.0096
  • Example 21 0.0118 0.0084 0.0091 0.0099 0.0137 0.0135
  • Example 22 0.0134 0.0091 0.0134 0.0110 0.0090 0.0167
  • Example 23 0.0121 0.0086 0.0093 0.0076 0.0086 0.0430
  • Example 24 0.0122 0.0074 0.0083 0.0091 0.0087 0.0113
  • Example 25 0.0059 0.0071 0.0089 0.0088 0.0101 0.0145
  • Example 26 0.0131 0.0104 0.0114 0.0083 0.0146 0.0294
  • Example 27 0.0051 0.0049 0.0027 0.0013 0.0019 0.0023
  • Example 28 0.0020 0.0033 0.0025 0.0088 0.0028 0.0041
  • Example 29 0.0083 0.0090 0.0089 0.0099 0.0085 0.0105
  • Example 30 0.0091
  • Table 67 shows the measurement results of intact immunoglobulin G content (non-reduced CE-SDS).
  • Example 20 98.66 98.23 97.61 97.92 97.22 98.59
  • Example 21 98.65 98.24 97.62 98.00 97.26 98.59
  • Example 22 98.61 98.26 97.49 97.92 97.73 98.63
  • Example 23 98.49 98.22 97.73 98.03 97.69 98.60
  • Example 24 98.64 98.20 97.70 97.92 97.53 98.55
  • Example 25 98.64 98.22 97.62 97.97 96.99 98.50
  • Example 26 98.61 98.21 97.69 98.04 96.92 98.58
  • Example 27 98.61 98.22 97.59 98.00 97.28 98.44
  • Example 21 98.65 98.24 97.62 98.00 97.26 98.59
  • Example 22 98.61
  • Examples 20 to 31 did not differ from Example 1 in the intact immunoglobulin G content after 4 weeks at 5 ⁇ 3° C. and 40 ⁇ 2° C. and 75 ⁇ 5% relative humidity conditions. In addition, it was confirmed that there was no decrease in the effect of shaking stress (FIG. 23).
  • Table 68 shows the measurement results of the antibody light chain and heavy chain content (reduced CE-SDS).
  • Examples 20 to 31 were stable because they were kept constant without change in the light chain and heavy chain contents of the antibody after 4 weeks at 5 ⁇ 3 ° C, 40 ⁇ 2 ° C temperature, and 75 ⁇ 5% relative humidity conditions. knew that it was In addition, it was confirmed that it is stable against shaking stress (FIG. 24).
  • Examples 20 to 31 were stable with a main component content of 99.0% or more after 4 weeks at 5 ⁇ 3°C. Although a decrease in the main component content was seen at 40 ⁇ 2 ° C. temperature and 75 ⁇ 5% relative humidity conditions, it was confirmed that the main component content was stable at 98.0% or more in all examples after 4 weeks, and there was no significant difference from Example 1. could (Fig. 25).
  • Table 70 shows the measurement results of the high molecular weight component content (pre-peak %).
  • Example 20 0.29 0.36 0.60 0.35 0.66 0.31
  • Example 21 0.29 0.37 0.64 0.36 0.67 0.31
  • Example 22 0.31 0.39 0.67 0.38 0.75 0.32
  • Example 23 0.31 0.39 0.71 0.39 0.78 0.36
  • Example 24 0.34 0.43 0.71 0.44 0.82 0.36
  • Example 25 0.39 0.50 0.86 0.51 0.96 0.42
  • Example 26 0.39 0.49 0.91 0.50 1.00 0.40
  • Example 27 0.36 0.40 0.67 0.40 0.94 0.37
  • Example 28 0.34 0.42 0.61 0.43 0.66 0.34
  • Example 29 0.36 0.40 0.66 0.42 0.76 0.35
  • Example 30 0.35 0.39 0.65 0.40 0.71 0.33
  • Example 31 0.31 0.43 0.61 0.40 0.66 0.32
  • Example 1 0.35 0.40 0.66 0.42 0.80 0.34 Comparative Example 1 0.33 0.38 0.66 0.40 0.72 0.34
  • Example 20 to 31 were stable at 1.0% or less of high molecular weight components after 4 weeks and after shaking stress under all temperature conditions. In addition, it was found that there was no significant difference from the results of Example 1.
  • Table 71 shows the measurement results of the content of low molecular weight components (post-peak %).
  • Example 20 0.07 0.06 0.61 0.08 0.98 0.08
  • Example 21 0.06 0.06 0.58 0.08 0.93 0.08
  • Example 22 0.07 0.06 0.35 0.08 0.58 0.06
  • Example 23 0.06 0.06 0.34 0.08 0.54 0.06
  • Example 24 0.06 0.06 0.32 0.07 0.56 0.06
  • Example 25 0.05 0.06 0.31 0.08 0.55 0.04
  • Example 26 0.07 0.06 0.36 0.08 0.61 0.07
  • Example 27 0.05 0.06 0.29 0.07 0.48 0.07
  • Example 28 0.08 0.06 0.26 0.07 0.47 0.04
  • Example 29 0.07 0.06 0.34 0.08 0.58 0.06
  • Example 30 0.08 0.06 0.33 0.08 0.58 0.04
  • Example 31 0.04 0.06 0.32 0.07 0.56 0.04
  • Example 1 0.07 0.06 0.35 0.08 0.61 0.05 Comparative Example 1 0.06 0.06 0.33 0.57 0.06
  • Example 20 to 31 were stable at 1.0% or less of low molecular weight components after 4 weeks and after shaking stress in all temperature conditions. In addition, it was found that there was no significant difference from the results of Example 1.
  • Table 72 shows the measurement results of the number of insoluble foreign particles (10.00 ⁇ m ⁇ , ⁇ 100.00 ⁇ m) measured by MFI.
  • Table 73 shows the measurement results of the number of insoluble foreign particles (25.00 ⁇ m ⁇ , ⁇ 100.00 ⁇ m) measured by MFI.
  • Example 20 2 2 4 2 4 25
  • Example 21 0 0 0 2 2 15
  • Example 22 2 2 6 0 0 28
  • Example 23 0 0 2 4 0 9
  • Example 24 0 0 2 0 4 100
  • Example 25 0 2 0 5 2 0
  • Example 26 2 0 0 0 2 78
  • Example 27 0 0 0 4 0 2
  • Example 28 0 3 2 0 4 21
  • Example 30 0 2 0 4 20 9
  • Example 31 0 0 0 2 0 68
  • Example 20 3 0 23 65 2 2
  • Example 21 3 2 2 2 5 0
  • Example 22 2 0 5 2 3 2
  • Example 23 0 0 28 2 0 2
  • Example 24 0 0 0 2 0 50
  • Example 25 75 2 12 8 2 0
  • Example 26 0 8 12 2 3 18
  • Example 27 7 15 10 20 18 15
  • Example 29 0 0 2 0 5 7
  • Example 30 3 3 3 2 17 365
  • Example 31 0 2 12 2 0 153
  • Example 1 2 0 5 0 8 95 Comparative Example 1 5 0 0 13 2 24603
  • Table 75 shows the measurement results of the number of insoluble foreign particles (25.00 ⁇ (um)) measured by HIAC.
  • Example 20 2 0 3 5 0 2
  • Example 21 0 0 0 0 0 0 0
  • Example 22 2 0 0 0 0 0
  • Example 23 0 0 10 0 0 0
  • Example 24 0 0 0 0 0
  • 32 Example 25
  • Example 26 0 0 10 0 0 0
  • Example 27 0 2 0 2 0 0
  • Example 28 2 0 2 7 0 8
  • Example 29 0 0 0 0 0 0
  • Example 30 0 2 0 0 3 3
  • Example 31 0 0 8 0 0 18
  • Example 1 0 0 2 0 0 0 16195

Abstract

La présente invention concerne une formulation pharmaceutique stable et, plus particulièrement, une formulation pharmaceutique comprenant une molécule de liaison par rapport à la protéine spike (protéine S) de la surface du SARS-coronavirus-2. La formulation pharmaceutique stable selon la présente invention présente une excellente stabilité de stockage à long terme sous des conditions de température de conditions accélérées et de conditions difficiles, et peut maintenir une excellente stabilité même sous des conditions de stress physique telles que la lumière, la congélation/décongélation, et l'agitation.
PCT/KR2021/010606 2020-08-11 2021-08-10 Formulation pharmaceutique stable WO2022035197A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
KR10-2020-0100805 2020-08-11
KR20200100805 2020-08-11

Publications (1)

Publication Number Publication Date
WO2022035197A1 true WO2022035197A1 (fr) 2022-02-17

Family

ID=80248006

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/KR2021/010606 WO2022035197A1 (fr) 2020-08-11 2021-08-10 Formulation pharmaceutique stable

Country Status (2)

Country Link
KR (1) KR20220020228A (fr)
WO (1) WO2022035197A1 (fr)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022261153A1 (fr) * 2021-06-08 2022-12-15 Eli Lilly And Company Formulations pharmaceutiques contenant des anticorps anti-coronavirus 2019
WO2023079086A1 (fr) * 2021-11-05 2023-05-11 Astrazeneca Uk Limited Composition pour le traitement et la prévention de la covid-19

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20060120568A (ko) * 2003-07-22 2006-11-27 크루셀 홀란드 비.브이. 사스-코로나바이러스에 대한 결합분자 및 그것의 용도
KR20200010103A (ko) * 2018-07-19 2020-01-30 (주)셀트리온 안정한 액체 약제학적 제제

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20060120568A (ko) * 2003-07-22 2006-11-27 크루셀 홀란드 비.브이. 사스-코로나바이러스에 대한 결합분자 및 그것의 용도
KR20200010103A (ko) * 2018-07-19 2020-01-30 (주)셀트리온 안정한 액체 약제학적 제제

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
KIM SANG IL, NOH JINSUNG, KIM SUJEONG, CHOI YOUNGGEUN, YOO DUCK KYUN, LEE YONGHEE, LEE HYUNHO, JUNG JONGTAK, KANG CHANG KYUNG, SON: "Stereotypic Neutralizing V H Clonotypes Against SARS-CoV-2 RBD in COVID-19 Patients and the Healthy Population", BIORXIV, 2 July 2020 (2020-07-02), pages 1 - 56, XP055899840, DOI: 10.1101/2020.06.26.174557 *
PARRAY HILAL AHMAD, CHIRANJIVI ADARSH KUMAR, ASTHANA SHAILENDRA, YADAV NAVEEN, SHRIVASTAVA TRIPTI, MANI SHAILENDRA, SHARMA CHANDRE: "Identification of an anti–SARS–CoV-2 receptor-binding domain–directed human monoclonal antibody from a naïve semisynthetic library", JOURNAL OF BIOLOGICAL CHEMISTRY, vol. 295, no. 36, 1 September 2020 (2020-09-01), US , pages 12814 - 12821, XP055899844, ISSN: 0021-9258, DOI: 10.1074/jbc.AC120.014918 *
SHI RUI, SHAN CHAO, DUAN XIAOMIN, CHEN ZHIHAI, LIU PEIPEI, SONG JINWEN, SONG TAO, BI XIAOSHAN, HAN CHAO, WU LIANAO, GAO GE, HU XUE: "A human neutralizing antibody targets the receptor-binding site of SARS-CoV-2", NATURE, vol. 584, no. 7819, 6 August 2020 (2020-08-06), London, pages 120 - 124, XP055891106, ISSN: 0028-0836, DOI: 10.1038/s41586-020-2381-y *

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022261153A1 (fr) * 2021-06-08 2022-12-15 Eli Lilly And Company Formulations pharmaceutiques contenant des anticorps anti-coronavirus 2019
WO2023079086A1 (fr) * 2021-11-05 2023-05-11 Astrazeneca Uk Limited Composition pour le traitement et la prévention de la covid-19

Also Published As

Publication number Publication date
KR20220020228A (ko) 2022-02-18

Similar Documents

Publication Publication Date Title
WO2021194188A1 (fr) Molécule de liaison ayant une activité neutralisante contre le sras-coronavirus-2
WO2022035197A1 (fr) Formulation pharmaceutique stable
WO2017116205A1 (fr) Conjugué persistant d'un triple activateur activant le récepteur du glucagon, du glp-1 et du gip
WO2018004260A1 (fr) Préparation pharmaceutique liquide stable
WO2014158001A1 (fr) Composition comprenant au moins deux molécules de liaison de neutralisation du virus de la grippe a
WO2019066586A1 (fr) Conjugué à action prolongée du dérivé de peptide-2 apparenté au glucagon (glp-2)
WO2018174544A2 (fr) Anticorps se liant spécifiquement à muc1 et son utilisation
WO2019203599A1 (fr) Polysaccharides capsulaires de streptococcus pneumoniae et conjugués immunogènes de ceux-ci
WO2020017901A1 (fr) Préparation pharmaceutique liquide stable
WO2022019671A1 (fr) Molécule de liaison neutralisant le sars-coronavirus 2 du sras qui se lie à l'épitope de la protéine de spicule du sars-coronavirus 2
WO2021060837A1 (fr) Anticorps anti-mers-cov et son utilisation
WO2020101365A1 (fr) Anticorps anti-c-met présentant une stabilité améliorée ou des fragments de liaison à l'antigène de celui-ci
WO2021060791A1 (fr) Nano-perforateur ayant une activité antivirale améliorée
WO2016199964A1 (fr) Anticorps se liant spécifiquement à un peptide isolé dérivé de la vimentine, ou à un fragment de liaison dudit peptide
WO2022211558A1 (fr) Molécule de liaison neutralisant le sars-coronavirus-2 qui se lie à l'épitope de la protéine de spicule du sars-coronavirus-2
WO2016200189A1 (fr) Épitope de protéine g du virus rabique, et molécule de liaison neutralisant le virus rabique s'y liant spécifiquement
WO2020071865A1 (fr) Utilisations thérapeutiques du glucagon et produit combiné comprenant celui-ci
WO2022149837A1 (fr) Anticorps anti-fgfr3 et son utilisation
WO2022015115A1 (fr) Utilisation thérapeutique d'une combinaison contenant un conjugué à action prolongée triplement agoniste ou un triple agoniste
WO2022085905A1 (fr) Anticorps se liant spécifiquement à la protéine de spicule du sars-cov-2 et utilisation associée
WO2021215801A1 (fr) Composition pharmaceutique préventive ou thérapeutique pour l'hyperlipidémie comprenant un triple agoniste agissant sur tous les récepteurs du glucagon, du glp-1 et du gip, ou un conjugué de ceux-ci et procédé préventif ou thérapeutique
WO2020214013A1 (fr) Utilisation thérapeutique, pour l'hyperlipidémie, d'un triple agoniste ayant une activité par rapport à tous les récepteurs du glucagon, glp -1 et gip, ou conjugué de ceux-ci
WO2020138868A1 (fr) Protéine de fusion fc neutralisant un interféron de type 1 et son utilisation
WO2023277620A1 (fr) Utilisation thérapeutique d'une combinaison comprenant un triple activateur présentant une activité sur l'ensemble des récepteurs suivants : au glucagon, glp-1 et gip
WO2022080991A1 (fr) Utilisation d'un triple activateur ayant une activité sur tous les récepteurs du glucagon, du glp-1 et du gip pour le traitement des séquelles d'une maladie infectieuse respiratoire

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: 21856203

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: 21856203

Country of ref document: EP

Kind code of ref document: A1