US20190175683A1 - Pharmaceutical formulations of c1 esterase inhibitor - Google Patents

Pharmaceutical formulations of c1 esterase inhibitor Download PDF

Info

Publication number
US20190175683A1
US20190175683A1 US16/323,155 US201716323155A US2019175683A1 US 20190175683 A1 US20190175683 A1 US 20190175683A1 US 201716323155 A US201716323155 A US 201716323155A US 2019175683 A1 US2019175683 A1 US 2019175683A1
Authority
US
United States
Prior art keywords
inh
pharmaceutical formulation
formulation according
formulation
formulations
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.)
Abandoned
Application number
US16/323,155
Other languages
English (en)
Inventor
Hubert Metzner
Ernst-Juergen KANZY
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.)
CSL BEHRING GmbH
CSL Behring GmbH Deutschland
Original Assignee
CSL BEHRING GmbH
CSL Behring GmbH Deutschland
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 CSL BEHRING GmbH, CSL Behring GmbH Deutschland filed Critical CSL BEHRING GmbH
Publication of US20190175683A1 publication Critical patent/US20190175683A1/en
Assigned to CSL BEHRING GMBH reassignment CSL BEHRING GMBH ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: KANZY, ERNST-JUERGEN, METZNER, HUBERT
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/005Enzyme inhibitors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/55Protease inhibitors
    • 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
    • A61K47/08Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing oxygen, e.g. ethers, acetals, ketones, quinones, aldehydes, peroxides
    • A61K47/10Alcohols; Phenols; Salts thereof, e.g. glycerol; Polyethylene glycols [PEG]; Poloxamers; PEG/POE alkyl ethers
    • 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
    • A61K47/24Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing atoms other than carbon, hydrogen, oxygen, halogen, nitrogen or sulfur, e.g. cyclomethicone or phospholipids
    • 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
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/14Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles
    • A61K9/19Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles lyophilised, i.e. freeze-dried, solutions or dispersions
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid

Definitions

  • the present invention relates to pharmaceutical formulations comprising the C1 esterase inhibitor (“C1-INH”), exhibiting reduced formation of aggregates of said esterase inhibitor (C1-INH) upon storage.
  • C1 esterase inhibitor (“C1-INH”)
  • C1-INH a plasma glycoprotein with a molecular weight of 104 kDa, belongs to the protein family of serine protease inhibitors (serpins), which regulate the activity of serine proteases by inhibiting their catalytic activity (Bock S C, et al., Biochemistry 1986, 25: 4292-4301).
  • C1-INH inhibits the classical pathway of the complement system by inhibiting the activated serine proteases C1s and C1r.
  • C1-INH is a major inhibitor of the contact activation system due to its ability to inhibit the activated serine proteases factor XIIa (FXIIa), factor XIa (FXIa), and plasma kallikrein (Davis A E, Clin.
  • HAE hereditary angioedema
  • HAE hereditary C1-INH deficiency
  • Type II HAE is associated with normal or elevated antigenic levels of C1-INH of low functional activity.
  • HAE with normal C1-INH also known as type III HAE
  • C1-INH Longhurst H, et al., Lancet 2012, 379: 474-481; Bork K, Allergy Asthma Clin. Immunol. 2010, 6: 15.
  • administration of C1-INH has been shown to prevent edema formation in patients when given prophylactically.
  • C1-INH is currently marketed as. e.g. Berinert® (CSL Behring). Due to its inhibitory effects on the complement and the contact activation systems, C1-INH substitution restores normal homeostatic function and inhibits the excessive formation of vasoactive peptides such as bradykinin, which mediate the formation of angioedema.
  • C1-INH compositions commercially available for the treatment of C1-Inh deficiency up to date are all large volume formulations, i.e., these formulations must be administered by intravenous injection.
  • C1-INH has been shown to prevent edema formation in patients with hereditary angioedema when given prophylactically (Cicardi M et al., Expert Opin. Pharmacother. 2007; 8: 3173-3181)
  • Long-term prophylaxis of HAE aims to prevent or to minimize the number and severity of angioedema attacks.
  • the medications currently available for long-term prophylaxis are in many cases not optimal.
  • Oral antifibrinolytics requiring multiple daily doses are relatively ineffective and frequently associated with significant side effects.
  • Anabolic androgens are convenient to take and usually effective at doses ⁇ 200 mg/day but can be associated with significant risk of serious side effects.
  • Currently available formulations of C1-INH require intravenous access, imposing a burden on the patient, the healthcare provider, or both. Maintenance of intravenous access has required many patients to have ports implanted, which are associated with increased risks of infection and thrombosis. Plasma levels of functional 01-INH fall rapidly following intravenous administration of therapeutic dosages of C1-INH concentrates, reaching near basal levels within 3 days.
  • HMWC high molecular weight components
  • strategies that minimize HMWC formation are highly desired to be developed as early as feasible in product development. This can be achieved by e.g. using an appropriate cell substrate, selecting manufacturing conditions that minimize HMWC formation, employing a purification scheme that removes HMWC to the greatest extent possible, choosing a container system, which minimizes HMWC formation of the protein, and most notably, choosing a formulation that minimizes HMWC formation, degradation and denaturation during storage.
  • formulation components are principally chosen based on their ability to preserve the native conformation of the therapeutic protein by preventing denaturation due to hydrophobic interactions that may lead to HMWC formation, as well as by preventing chemical degradation, including truncation, oxidation, and deamidation (Cleland et al., Crit. Rev. Ther. Drug Carrier Syst. 1993, 10(4): 307-377; Shire et al., J. Pharm. Sci. 2004, 93(6): 1390-1402; Wakankar and Borchardt, J. Pharm. Sci. 2006, 95(11): 2321-2336).
  • HMWC immune responses induced by protein HMWC may depend on the loss or preservation of native epitopes in the HMWC: (a) some antibodies generated by the human subject against HMWC containing native protein may bind to monomeric protein as well as to the HMWC and may inhibit or neutralize product activity; (b) other antibodies to denatured/degraded and hence aggregated protein bind uniquely to the HMWC material, but not to native protein monomers (Guidance for Industry Immunogenicity Assessment for Therapeutic Protein Products, U.S. Department of Health and Human Services, Food and Drug Administration, Center for Drug Evaluation and Research (CDER), Center for Biologics Evaluation and Research (CBER), August 2014).
  • CDER Center for Drug Evaluation and Research
  • CBER Center for Biologics Evaluation and Research
  • WO 2014/145519 discloses C1-INH compositions having about 400 or 500 U/mL C1-INH. It is suggested not to use citrate or citric acid as a buffer substance for subcutaneous administration. The disclosed formulations contain only particular buffer substances in low concentrations with no other excipient added. All disclosed C1-INH formulations have a relatively low overall purity of about 67% monomer content at t 0 . With regard to stability the WO 2014/145519 discloses only data after one week at 40° C. and after two weeks at 25° C., i.e. no long-term stability data are shown and thus long-term stability is unproven.
  • the present invention provides low volume formulations comprising high concentrations of C1-INH and having an increased C1-INH stability.
  • formulations can be administered via subcutaneous injection and can easily be self-administered by the patients themselves.
  • these formulations can also be administered via intravenous injection.
  • the present invention provides one type of formulation, which is suitable, both for using intravenous and subcutaneous treatment, even for self-application by the patients.
  • formulations according to the invention ensure a substantially reduced formation of undesired oligomers and high molecular weight components (HMWC) compared to available C1-INH formulations.
  • HMWC high molecular weight components
  • the present invention further relates to use of such formulations in the acute and/or prophylactic treatment of disorders related to kinin formation.
  • the present invention further relates to kits comprising said C1-INH formulation, methods for preparing such formulations, and uses of such formulations.
  • the present invention relates to a stable pharmaceutical composition
  • a stable pharmaceutical composition comprising (a) C1-INH at a concentration of about 400-2,000 IU/mL and (b) histidine, preferably L-histidine, or a salt/salts thereof at a concentration of about 5-150 mM, preferably 10-80 mM, wherein the formulation does not comprise citrate or di-hydrogen phosphate/hydrogen phosphate.
  • the pharmaceutical formulation of the invention further comprises one or more amino acids, preferably an L-amino acid or a salt thereof at a concentration of 2-150 mM each, preferably about 5-50 mM.
  • the one or more amino acid is selected from arginine, glycin and lysine or any salt(s) thereof.
  • the formulation comprises saccharose (sucrose) at a concentration of about 5-250 mM, preferably 20-200 mM.
  • the formulation further comprises sodium chloride or sodium glutamate at a concentration of about 1-200 mM, preferably 20-150 mM.
  • the pH of the pharmaceutical formulations of this invention is between about 6.0 and 8.0, preferably between about 6.7 and 7.5.
  • the pharmaceutical composition comprises:
  • the pharmaceutical composition may further comprise
  • the C1-INH of this invention is human C1-INH.
  • the human C1-INH is derived from human plasma.
  • the human C1-INH is recombinantly expressed.
  • the pharmaceutical formulation is
  • the pharmaceutical formulation is provided as a lyophilized powder.
  • the formulation can be administered via subcutaneous administration or via intravenous administration whereby optionally said formulation may be self-administered by the patient.
  • Another aspect of the invention refers to pharmaceutical formulations for use
  • kits comprising pharmaceutical formulations of the invention as a lyophilized powder and a respective volume of a suitable liquid for reconstitution.
  • kits comprising the pharmaceutical formulation of the invention and at least one syringe and/or one needle.
  • a syringe prefilled with a liquid pharmaceutical formulation of the invention refers to
  • FIG. 1 HMWC levels by SEC HPLC of three C1-INH formulations over 36 months at 2-8° C.:
  • FIG. 2 HMWC levels by SEC HPLC of three C1-INH formulations over 36 months at 25° C.
  • FIG. 3 HMWC levels by SEC HPLC of three C1-INH formulations over 18 months at 40° C.
  • FIG. 4 Relative C1-INH activity of three C1-INH formulations over 36 months at 2-8° C.
  • FIG. 5 Relative C1-INH activity of three C1-INH formulations over 36 months at 25° C.
  • FIG. 6 Relative C1-INH activity of three C1-INH formulations over 18 months at 40° C.
  • FIG. 7 HMWC levels by SEC HPLC of five C1-INH formulations over 24 months at 2-8° C.:
  • FIG. 8 HMWC levels by SEC HPLC of five C1-INH formulations over 6 months at 40° C.
  • FIG. 9 Relative C1-INH activity of five C1-INH formulations over 24 months at 2-8° C.
  • FIG. 10 Relative C1-INH activity of five C1-INH formulations over 6 months at 40° C.
  • C1 esterase inhibitor or “C1 inhibitor” (“C1-INH”) refers to the proteins or fragments thereof that function as serine protease inhibitors and inhibit proteases associated with the complement system, preferably proteases C1r and C1s as well as MASP-1 and MASP-2, with the kallikrein-kinin system, preferably plasma kallikrein and/or factor XIIa, and with the coagulation system, preferably factor XIa and factor XIIa.
  • the C1-INH can serve as an anti-inflammatory molecule that reduces the selectin-mediated leukocyte adhesion to endothelial cells.
  • C1-INH as used herein can be the native serine protease inhibitor or an active fragment thereof, or it can comprise a recombinant peptide, a synthetic peptide, peptide mimetic, or peptide fragment that provides similar functional properties, such as the inhibition of proteases C1r and C1s, and/or MASP-1 and MASP-2, and/or plasma kallikrein, and/or factor XIIa, and/or factor XIa.
  • the term C1-INH shall also encompass all natural occurring alleles, splice variants and isoforms which have the same or similar functions as the C1-INH.
  • the structure and function of C1-INH see U.S. Pat. Nos. 4,915,945, 5,939,389, 6,248,365, 7,053,176 and WO 2007/073186.
  • U One “unit” (“U”) of C1-INH is equivalent to the C1-INH activity in 1 mL of fresh citrated plasma of healthy donors.
  • the C1-INH may also be determined in “international units” (“IU”). These units are based on the current World Health Organization (WHO) standard for C1-INH concentrates (08/256) which was calibrated in an international collaborative study using normal local human plasma pools. In general, U and IU are equivalent.
  • WHO World Health Organization
  • HAE hereditary angioedema
  • HAE angioedema caused by a low content and low inhibitory activity of C1-INH in the circulation (HAE type I) or by the presence of normal or elevated antigenic levels of C1-INH of low functional activity (HAE type II).
  • HAE as used herein also encompasses HAE with normal C1-INH (also known as HAE type III) which has been described recently in two subcategories: (1) HAE due to mutation in the factor XII gene and, as a result, increased activity of factor XII leading to a high generation of bradykinin, and (2) HAE of unknown genetic cause.
  • edema attacks can occur in various intervals, including a daily, weekly, monthly, or even yearly basis. Furthermore, there are affected patients wherein no edema occurs.
  • angioedema relates to swelling of tissue, for example swelling of skin or mucosa.
  • the swelling can occur, for example, in the face, at hands or feet or on the genitals.
  • swelling can occur in the gastro-intestinal tract or in the respiratory tract.
  • Other organs can also be affected. Swelling persists usually between one and three days. However, remission can already occur after hours or not until weeks.
  • IRI ischemia-reperfusion injury
  • Reperfusion tissue
  • Direct damage to the tissue is caused by the interruption of the blood flow, mainly due to loss of oxygenation to the viable tissue, ultimately leading to infarction if not reversed.
  • the reperfusion of the ischemic tissue may paradoxically cause further “indirect” damage.
  • the direct damage resulting from hypoxia alone is the predominant mechanism.
  • the “indirect” reperfusion mediated damage increasingly contributes to the damage caused.
  • retinopathy as used herein relates to acute or persistent damage of the eye.
  • Retinopathy can be caused by diabetes mellitus (leading to diabetic retinopathy), arterial hypertension (leading to hypertensive retinopathy), prematurity of the newborn (leading to retinopathy of prematurity), exposure to ionizing radiation (radiation retinopathy), direct sunlight exposure (solar retinopathy), sickle cell disease, retinal vascular disease such as retinal vein or artery occlusion, trauma, especially to the head and other diseases or conditions.
  • Many types of retinopathy are proliferative resulting, most often, from neovascularization or the overgrowth of blood vessels.
  • Angiogenesis the sprouting of new vessels is the hallmark precursor that may result in blindness or severe vision loss particularly if the macula becomes affected.
  • retinopathy is caused by genetic diseases.
  • acute treatment or “treatment” as used herein relates to the treatment of a patient displaying acute symptoms.
  • Acute treatment can occur from the appearance of the symptom until the full remission of the symptom.
  • An acute treatment can occur once or several times until the desired therapeutic effect is achieved.
  • prophylactic treatment or “prophylaxis” or “prevention” as used herein relates to the treatment of a patient in order to prevent the occurrence of symptoms. Prophylactic treatment can occur at regular intervals of days, weeks or months. Prophylactic treatment can also occasionally occur.
  • HMWC high molecular weight components
  • HMWC refers to any self-associated, i.e. multimerised or aggregated protein species, in particular of C1-INH, with monomer defined as the smallest functional subunit. HMWC are further classified based on five characteristics: size, reversibility/dissociation, conformation, chemical modification, and morphology (Narhi et al., J. Pharm. Sci. 2012, 101(2): 493-498).
  • HMWC in particular multimers and aggregates
  • the underlying mechanisms by which proteins aggregate may elicit or enhance immune responses include inter alia the following: extensive cross-linking of B-cell receptors, causing efficient B-cell activation (Dintzis et al., J. Immunol. 1989, 143(4): 1239-1244; Bachmann et al., Science 1993, 262(5138): 1448-1451); enhancing antigen uptake, processing, and presentation; and triggering immunostimulatory danger signals (Seong and Matzinger, Nat.
  • finished dosage form (FDF) of a drug has undergone all stages of manufacture, including packaging in its final container and labelling.
  • physiologically acceptable salt refers to salts in formulations that are mainly used in treating medical conditions in humans, in particular to treating or preventing disorders related to kinin formation.
  • a physiological acceptable salt refers to ionic substances which are soluble, i.e. in the liquid, preferably aqueous, state a physiological acceptable salt will be present in form of its dissolved cation(s) and anion(s), and which will not cause serious adverse side events after administration to the human body.
  • the formulations or their finished dosage forms are appropriate for physiological practice together with other excipients.
  • WFI water for injection
  • It is water intended for use in the manufacture of medicines for parenteral administration, the solvent of which is water.
  • it refers to water that is used to dissolve or dilute substances or preparations for parenteral administration.
  • the C1-INH is a plasma-derived or a recombinant C1-INH.
  • said inhibitor is identical to the naturally occurring human protein or a variant thereof.
  • said inhibitor is human C1-INH.
  • said inhibitor is a recombinant analogue of human C1-INH protein.
  • the C1-INH may be modified to improve its bioavailability and/or half-life, to improve its efficacy and/or to reduce its potential side effects.
  • the modification can be introduced during recombinant synthesis or otherwise. Examples for such modifications are glycosylation, PEGylation and HESylation of the C1-INH or an albumin fusion of the described C1-INH.
  • C1-INH comprises a fusion construct between C1-INH and albumin, in particular human albumin.
  • the albumin is a recombinant protein.
  • the C1-INH and albumin proteins are joined directly, or via a linker polypeptide.
  • glycosylation and albumin fusion of proteins see WO 01/79271.
  • the C1-INH can be produced according to methods known to the skilled person.
  • plasma-derived C1-INH can be prepared by collecting blood plasma from several donors. Donors of plasma should be healthy as defined in the art. Preferably, the plasma of several (1000 or more) healthy donors is pooled and optionally further processed.
  • An exemplary process for preparing C1-INH for therapeutic purposes is disclosed in U.S. Pat. No. 4,915,945.
  • C1-INH can be collected and concentrated from natural tissue sources using techniques known in the art. Recombinant C1-INH can be prepared by known methods.
  • C1-INH is derived from human plasma. In a further preferred embodiment, C1-INH is prepared by recombinant expression.
  • a commercially available product comprising C1-INH is, e.g., plasma-derived Berinert® (CSL Behring). Berinert® is manufactured according to A. Feussner et al. (Transfusion 2014, 54: 2566-73) and is indicated for treatment of hereditary angioedema and congenital deficiencies.
  • Alternative commercially available products comprising C1-INH are plasma-derived Cetor® (Sanquin), Cinryze® (Shire), and recombinant Ruconest®/Rhucin® (Pharming).
  • the present invention relates to formulations comprising C1-INH. These highly concentrated formulations of the invention are provided in low volume formulations having a long-term stability. The formulations are well-tolerated and suitable for intravenous and in particular subcutaneous administration.
  • the concentration of C1-INH in said formulations is about 400 IU/mL to 2,000 IU/mL, preferably of about 400 IU/mL to 1,200 IU/mL, more preferably of about 400 IU/mL to 1000 IU/mL, more preferably of about 400 IU/mL to 800 IU/mL, more preferably of about 400 IU/mL to 625 IU/mL, and most preferably of about 500 IU/mL or any range in between.
  • Said pharmaceutical formulations of the present invention further comprise histidine, preferably L-histidine, or a salt/salts thereof at a concentration of about 5-150 mM, preferably 10-80 mM. Additionally said formulation neither comprises sodium citrate nor sodium di-hydrogen phosphate/di-sodium hydrogen phosphate as buffer substances.
  • said formulation further comprises one or more amino acids or a salt/salts thereof.
  • the one or more amino acids are selected from arginine, glycin and/or lysine.
  • the amino acids may be L- or D-amino acids, preferably L-amino acids.
  • the concentration of each further amino acid of this invention ranges from about 2 mM to about 150 mM, preferably from about 3 mM to about 130 mM, more preferably from about 5 mM to about 50 mM.
  • the pharmaceutical formulation comprises only one type of further amino acid.
  • the formulations referred to herein do not comprise a buffer compound selected from the following: succinate, tartrate, maleate, acetate, and salts thereof.
  • the formulation comprises saccharose, trehalose, or a combination thereof at a concentration of about 5-250 mM, preferably at a concentration of about 20-200 mM, more preferably at a concentration of about 100-200 mM. In highly preferred embodiments, the concentration is about 140-160 mM.
  • the pharmaceutical formulation further comprises at least one substance selected from the group consisting of sodium chloride, di-sodium EDTA, sodium acetate, sodium glutamate and sodium succinate at a concentration of about 1-200 mM, preferably of about 10-200 mM, more preferably of about 20-150 mM, and even more preferably of about 40-60 mM or about 130-150 mM.
  • the pharmaceutical formulation does not comprise a tissue permability enhancer, such as e.g. hyaluronidase.
  • the pH of the formulation is between about 6.0 and 8.0, between about 6.7 and 7.5, between about 6.8 and 7.4, between about 6.9 and 7.3, between about 7.0 and 7.2. In the most preferred embodiment, the pH of the formulation is 7.0.
  • the pharmaceutical formulation comprises
  • the pharmaceutical formulation comprises
  • the pharmaceutical formulation comprises
  • the pharmaceutical formulation comprises
  • the pharmaceutical formulation comprises:
  • the pharmaceutical formulation comprises:
  • the pharmaceutical formulation comprises:
  • the provided formulation comprises one or more detergents and/or one or more preservatives and/or one or more antioxidants.
  • the pharmaceutical formulation can comprise PS80 (polysorbate 80) and/or PS20 (polysorbate 20).
  • PS80 may be present at a concentration of about 0.5-2 mg/mL.
  • PS20 may be present at a concentration of about 0.5-2 mg/mL.
  • the preservatives and/or antioxidants are selected from the group consisting of benzylalcohol, cresol, phenol, methionine and glutathione.
  • the preservatives and/or antioxidants may be present at a concentration of about 1-5 mM.
  • the provided formulations may comprise pharmaceutical carriers and excipients that are well known in the art (see for example “Pharmaceutical Formulation Development of Peptides and Proteins”, Frokjaer et al., Taylor & Francis 2000 or “Handbook of Pharmaceutical Excipients”, 3 rd edition, Kibbe et al., Pharmaceutical Press 2000).
  • the formulation comprises an absolute amount of C1-INH of about 1,000 IU/FDF, 1,200 IU/FDF, about 1,500 IU/FDF, about 1,800 IU/FDF, about 2,100 IU/FDF, about 2,400 IU/FDF, about 2,700 IU/FDF or about 3,000 IU/FDF or any absolute amount in between.
  • the formulation comprises an absolute amount of C1-INH of at least 1,200 IU per FDF, 1,500 IU per FDF, or at least 1,800 IU per FDF.
  • the formulation comprises an absolute amount of C1-INH of about 1,200 IU-3,000 IU, of about 1,200 IU-2,000 IU, of about 1,200 IU-1,800 IU or of about 1,400 IU-1,600 IU.
  • the liquid pharmaceutical formulation is provided in a volume of about 0.1-10 mL/FDF, about 1-5 mL/FDF or about 3 mL/FDF or any volume in between.
  • the formulation is provided as an aqueous solution in a volume of about 3 mL per FDF, of about 4 mL per FDF or of about 6 mL per FDF.
  • the C1-INH is human C1-INH.
  • the human C1-INH is derived from human plasma, or the human C1-INH is recombinantly expressed.
  • the C1-INH is derived from human plasma.
  • the pharmaceutical formulations of the invention comprises less than 10% of HMWC, preferably less than 8% of HMWC, more preferably less than 5% of HMWC or even more preferably less than 3% of HMWC as determined immediately by SEC-HPLC. Immediately after preparation means within one day after preparation.
  • the formulation is provided as a stable lyophilized powder.
  • the formulation may be lyophilized by a variety of procedures known in the art.
  • the lyophilized powder can be reconstituted with a respective volume of a suitable liquid to obtain the pharmaceutical liquid formulation for injection.
  • suitable liquids are, e.g., water for injection (WFI).
  • the formulation is provided as a stable liquid formulation.
  • the liquid formulation is obtainable by reconstitution of a stable lyophilized powder with a suitable liquid.
  • the components of the pharmaceutical formulation are present in a solution suitable for injection without any further processing, i.e. the formulation is provided as a stable liquid formulation.
  • the components are provided as a stable lyophilized powder and the indicated concentrations are reached upon reconstitution of the lyophilized powder in the respective volume of a suitable liquid.
  • a suitable liquid for reconstitution is e.g. water for injection (WFI).
  • stable formulation refers to pharmaceutical formulations wherein no significant decrease of C1-INH activity is observed after a certain period of time of storage at least at 2-8° C., preferably at about 25° C.
  • no significant decrease of C1-INH activity means at least 70%, 75%, 80%, 85%, 90%, 95% 96%, 97%, 98%, 99% of the C1-INH activity of the original C1-INH activity.
  • C1-INH activity refers to the inhibitory activity of the C1-INH protein in plasma and is indicated in “IU/mL” and can be measured without limitation e.g. by a chromogenic assay (see e.g. example 1).
  • the terms “stable formulation”, “stable lyophilized powder”, “stable lyophilized formulation” or “stable liquid formulation” as used herein refer to formulations wherein no significant increase of HMWC formation is observed after a certain period of time, preferably at least 36 months, of storage at 2-8° C., preferably at about 25° C.
  • the term “no significant increase of HMWC formation” means at most 20%, 15%, 14%, 13%, 12%, 11% 10%, 9%, 8%, 7%, 6%, 5%, 4%, or about 3% protein HMWC at a certain point in time, e.g. after 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 11, 12, 15, 18, 21, 24, or 36 months, i.e.
  • the percentage of HMWC is determined as the percentage of HMWC of the total protein content in the formulation at this certain point of time and this value is at the most 20%, 15%, 14%, 13%, 12%, 11% 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%.
  • the level of protein HMWC of the total protein can be measured without limitation e.g. by SEC HPLC (see e.g. example 1).
  • stable formulation refers to formulations wherein no significant increase of HMWC formation is observed after a certain period of time, preferably at least 18-24 months, of storage at about 40° C.
  • no significant increase of HMWC formation means formation of at most 20%, 15%, 14%, 13%, 12%, 11% 10%, 9%, 8%, 7%, 6%, 5%, or about 4% protein HMWC at a certain point in time, e.g. after 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 11, 12, 15, 18, 21, or 24 months, i.e.
  • the percentage of HMWC is determined as the percentage of HMWC of the total protein content in the formulation at this certain point of time and this value is at the most 20%, 15%, 14%, 13%, 12%, 11% 10%, 9%, 8%, 7%, 6%, 5%, 4%.
  • the level of protein HMWC of the total protein can be measured without limitation e.g. by SEC HPLC (see e.g. example 1).
  • stable formulation refers to formulations wherein no significant increase of fragmentation is observed after a certain period of time, preferably at least 36 months, of storage at 2-8° C., preferably at about 25° C.
  • no significant increase of fragmentation means at most 20%, 15%, 14%, 13%, 12%, 11% 10%, 9%, 8%, 7%, 6%, 5, 4%, or about 3% fragments at a certain point in time, e.g. after 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 11, 12, 15, 18, 21, 24, or 36 months, i.e.
  • the percentage of fragments is determined as the percentage of fragments the total protein content in the formulation at this certain point of time and this value is at the most 20%, 15%, 14%, 13%, 12%, 11% 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%.
  • the level of fragments of the total protein can be measured without limitation e.g. by SEC HPLC.
  • the formulations provided herein upon lyophilization will be stable for a certain period of time, i.e. for at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48 or more months at a temperature of 25° C.
  • the lyophilized formulation will be stable for at least 6 months (25° C.).
  • the lyophilized formulation will be stable for at least 12 months (25° C.).
  • the lyophilized formulation will be stable for at least 24 months (25° C.).
  • the liquid formulation provided herein will be stable for a certain period of time, i.e. for at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36 or more months at a temperature of 2-8° C.
  • the liquid formulation will be stable for at least 6 months (2-8° C.).
  • the liquid formulation will be stable for at least 12 months (2-8° C.).
  • the liquid formulation will be stable for at least 24 months (2-8° C.).
  • the liquid formulation provided herein will be stable for a certain period of time, i.e. for at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24 or more months at a temperature of 25° C.
  • the liquid formulation will be stable for at least 6 months (25° C.).
  • the liquid formulation will be stable for at least 12 months (25° C.).
  • the provided formulations retain at least 70%, preferably at least 80%, more preferably at least 90% and most preferably at least 95% C1-INH activity after 12 months, preferably after 24 months, more preferably after 36 months and most preferably after 48 months of storage at 2-8° C.
  • the provided formulation retains at least 70%, preferably at least 80%, more preferably at least 90% and most preferably at least 95% C1-INH activity after 6 months, preferably after 12 months, more preferably after 24 months and most preferably after 36 months of storage at 25° C.
  • the provided formulation retains at least 30%, preferably at least 40%, more preferably at least 50% and most preferably at least 60% C1-INH activity after 6 month, preferably after 12 months, more preferably after 24 months of storage at 40° C.
  • the provided pharmaceutical formulations can be utilized as stand-by medication, i.e., a patient suffering from hereditary angioedema can always keep such a formulation in close proximity (without the requirement of cooling) in order to have an immediate treatment available upon occurrence of an edema attack.
  • the provided pharmaceutical formulations demonstrate a long-term stability compared to concentrated C1-INH formulations having only a buffer excipient in low concentrations.
  • the provided pharmaceutical formulations are suitable for subcutaneous administration as well as intravenous administration.
  • subcutaneous administration is preferred upon prophylactic treatment of patients suffering from hereditary angioedema and intravenous administration is preferred upon acute treatment of patients suffering from hereditary angioedema.
  • intravenous nor the subcutaneous administration of the provided pharmaceutical formulations causes serious drug-related adverse side events but only minor treatment-emergent adverse effects.
  • the provided formulations can be used for both administrations.
  • the pharmaceutical formulation described herein is suitable for intra-arterial and/or intramuscular administration.
  • the patients can self-administer the provided formulations.
  • Patients can use the provided formulations for prophylactic treatment and, in addition, the same formulation can be used for an acute treatment upon occurrence of an angioedema attack.
  • patients are only supplied with one type of formulation, which is indicative of a high patient compliance and which can be used as required.
  • the provided formulations may achieve a high patient compliance.
  • the provided formulations exhibit a high local tolerance upon subcutaneous and intravenous injection; they are well tolerated, with no serious drug-related adverse events.
  • the subcutaneous administration of the provided formulations is safe and well tolerated with only mild-to-moderate local site reactions.
  • a high local tolerance is achieved upon intra-arterial injection and intramuscular injection.
  • thrombotic events, clot formation, thromboembolic complications do not occur after administration of the described formulations in any of the described doses in a patient. Further, administration of the described formulations does not enhance the thrombogenic risk in a patient.
  • the pharmaceutical formulations of the present invention ensure a good bioavailability of the C1-INH upon subcutaneous as well as upon intravenous administration.
  • kits comprising a lyophilized formulation described herein and the respective amount of a liquid suitable for reconstitution.
  • the suitable liquid is water for injection, preferable deionized sterile water for injection.
  • the kit can additionally comprise a syringe.
  • the syringe is suitable for subcutaneous injection.
  • the syringe is suitable for intravenous injection.
  • the syringe is suitable for subcutaneous injection and intravenous injection.
  • the syringe is suitable for intra-arterial injection and/or intramuscular injection.
  • the kit further can comprise a needle suitable for intravenous injection and/or a needle suitable for subcutaneous injection.
  • the kit can comprise a needle suitable for intra-arterial injection and/or a needle suitable for intramuscular injection.
  • a syringe prefilled with the liquid formulation described herein is provided.
  • the syringe is suitable for subcutaneous injection.
  • the syringe is suitable for intravenous injection.
  • the syringe is suitable for subcutaneous injection and intravenous injection.
  • the syringe is suitable for intra-arterial injection and/or intramuscular injection.
  • a method of reconstituting a lyophilized formulation comprising C1-INH in a suitable solution is provided herein. Furthermore, a method for reconstituting any of the lyophilized formulations described herein is provided.
  • a lyophilized formulation comprising C1-INH for the preparation of a liquid formulation comprising C1-INH is provided.
  • the lyophilized formulation may be any lyophilized formulation described herein.
  • the provided formulations can be used in the treatment of various diseases and conditions.
  • the provided formulations can be used in the treatment and/or prevention of a disorder related to kinin formation, in particular hereditary angioedema (HAE), secondary brain edema, edema of the central nervous system, hypotensive shock, or edema during or after contacting blood with an artificial surface.
  • HAE hereditary angioedema
  • secondary brain edema edema of the central nervous system
  • hypotensive shock edema during or after contacting blood with an artificial surface.
  • a method of treating or preventing a disorder related to kinin formation in particular hereditary angioedema (HAE), secondary brain edema, edema of the central nervous system, hypotensive shock, or edema during or after contacting blood with an artificial surface in a patient, comprising administering a pharmaceutically effective dose of any of the formulations described herein.
  • HAE hereditary angioedema
  • secondary brain edema edema of the central nervous system
  • hypotensive shock or edema during or after contacting blood with an artificial surface in a patient
  • the provided formulations are used in the treatment and/or prophylaxis of hereditary angioedema, in particular HAE type I, HAE type II and/or HAE type III.
  • a method of treating or preventing hereditary angioedema, in particular HAE type I, HAE type II and/or HAE type III in a patient comprising administering a pharmaceutically effective dose of any of the formulations described herein, is provided.
  • the provided formulations can be used in the treatment and/or prophylaxis of an ischemia-reperfusion injury (IRI), in particular wherein the IRI is due to surgical intervention, in particular vascular surgery, cardiac surgery, neurosurgery, trauma surgery, cancer surgery, orthopedic surgery, transplantation, minimally invasive surgery, or insertion of a device for delivery of a pharmacologically active substance or for mechanical removal of complete or partial obstructions.
  • IRI ischemia-reperfusion injury
  • IRI ischemia-reperfusion injury
  • the IRI is due to surgical intervention, in particular vascular surgery, cardiac surgery, neurosurgery, trauma surgery, cancer surgery, orthopedic surgery, transplantation, minimally invasive surgery, or insertion of a device for delivery of a pharmacologically active substance or for mechanical removal of complete or partial obstructions in a patient, comprising administering a pharmaceutically effective dose of any of the formulations described herein.
  • the provided formulations can be used to prevent rejection of a transplanted tissue in a patient.
  • the transplantation can be allotransplantation or xenotransplantation.
  • transplantation can be allotransplantation or xenotransplantation.
  • the provided formulations can be used in the treatment and/or prevention of retinopathy.
  • acute treatment occurs upon treatment of a patient having hereditary angioedema and suffering from an acute angioedema attack.
  • prophylactic treatment of a patient having hereditary angioedema occurs in order to prevent the occurrence of angioedema.
  • Prophylactic treatment of patients suffering from hereditary angioedema can be done regularly and can also occur occasionally, for example before surgical interventions, dental treatments and other symptom-triggering situations such as a situation where a patient realizes an upcoming edema.
  • the provided formulations can be administered via subcutaneous injection. In alternative embodiments, the provided formulations can be administered via intravenous injection. The formulations can be administered continuously by infusion or by bolus injection. In a preferred embodiment, the provided formulations can be administered via subcutaneous injection and via intravenous injection. In a further preferred embodiment, the patient can self-administer the provided formulations.
  • the provided formulation is administered via intravenous injection during acute treatment of a patient. In other embodiments, the provided formulation is administered via subcutaneous injection during prophylactic treatment of a patient.
  • the provided formulations can be administered via intra-arterial injection. In further embodiments, the provided formulations can be administered via intramuscular injection.
  • the formulations described herein may be administered to a patient by any pharmaceutically suitable means of administration.
  • Various delivery systems are known and can be used to administer the composition by any convenient route.
  • the compositions of the invention are administered systemically.
  • the therapeutic proteins of the invention are formulated for parenteral (e.g. intravenous, subcutaneous, intramuscular, intraperitoneal, intracerebral, intrapulmonar, intranasal or transdermal) or enteral (e.g., oral, vaginal or rectal) delivery according to conventional methods.
  • parenteral e.g. intravenous, subcutaneous, intramuscular, intraperitoneal, intracerebral, intrapulmonar, intranasal or transdermal
  • enteral e.g., oral, vaginal or rectal
  • Some formulations encompass slow release systems.
  • a dose of C1-INH of about 1,200 IU, about 1,500 IU, about 1,600 IU, about 1,700 IU, about 1,800 IU, about 1,900 IU, about 2,000 IU, about 2,100 IU, about 2,200 IU, about 2,300 IU, about 2,400 IU, about 2,500 IU, about 2,600 IU, about 2,700 IU, about 2,800 IU, about 2,900 IU, about 3,000 IU, about 3,500 IU, about 4,000 IU, about 4,500 IU, about 5,000 IU, about 5,500 IU or about 6,000 IU or any amount in between is administered to a patient.
  • a dose of about 1,500 IU, about 2,000 IU, about 3,000 IU, about 4,000 IU, about 5,000 IU or about 6,000 IU is administered to a patient.
  • a dose of C1-INH of about 10-100 IU/kg bodyweight, of about 20-90 IU/kg bodyweight, of about 20-80 IU/kg bodyweight, of about 30-70 IU/kg bodyweight, of about 40-60 IU/kg bodyweight or any range in between is administered to a patient.
  • a dose of C1-INH of about 20-80 IU/kg bodyweight, of about 30-80 IU/kg bodyweight, of about 40-80 IU/kg bodyweight, of about 40-60 IU/kg bodyweight, of about 50-60 IU/kg bodyweight or any range in between is administered to a patient.
  • a dose of C1-INH of about 10-60 IU/kg bodyweight, of about 20-40 IU/kg bodyweight, of about 20 IU/kg bodyweight or any range in between is administered to a patient.
  • the target is to reach a mean C1-INH activity level in the prophylactic treatment.
  • a dose of C1-INH is administered, preferably in prophylactic treatment, at intervals of 1, 2, 3, 4, 5, 6 or 7 days.
  • a dose of C1-INH is administered, preferably in prophylactic treatment, at intervals of every 1-2 days, at intervals of every 2-3 days, at intervals of every 3-4 days, at intervals of every 4-5 days, at intervals of every 5-6 days or at intervals of every 6-7 days.
  • a formulation comprising one, two or more FDF(s) of about 1,500 IU of C1-INH is/are administered, preferably subcutaneously, at intervals of every 2-3 days.
  • a formulation comprising one, two or more FDF(s) of about 3,000 IU of C1-INH is/are administered, preferably subcutaneously, at intervals of every 3-4 days.
  • a formulation comprising about 1,500 IU, about 3,000 IU, about 4,000 IU, about 5,000 IU or about 6,000 IU of C1-INH is/are administered, preferably subcutaneously, weekly.
  • a formulation comprising about 1,500 IU, about 3,000 IU, about 4,000 IU, about 5,000 IU or about 6,000 IU of C1-INH is/are administered, preferably subcutaneously, twice-weekly.
  • a formulation comprising a dose of about 20-80 IU per kg bodyweight, preferably about 20-40 IU C1-INH per kg bodyweight is administered, preferably subcutaneously, at intervals of every 2-3 days.
  • a formulation comprising a dose of about 40-60 IU C1-INH per kg bodyweight is administered, preferably subcutaneously, at intervals of every 3-4 days.
  • a formulation comprising a dose of about 40-80 IU C1-INH per kg bodyweight or of about 40-60 IU C1-INH per kg bodyweight is administered, preferably subcutaneously, weekly.
  • a formulation comprising a dose of about 40-80 IU C1-INH per kg bodyweight or of about 40-60 IU C1-INH per kg bodyweight is administered, preferably subcutaneously, twice weekly.
  • one, two or more FDF(s) of about 1,500 IU C1-INH is/are administered to a patient during acute treatment, preferably via intravenous administration.
  • one or more FDF(s) of about 3,000 IU C1-INH is/are administered to a patient during acute treatment, preferably via intravenous administration.
  • one, two or more dose(s) of about 20 IU C1-INH per kg bodyweight is/are administered to a patient during acute treatment, preferably via intravenous administration.
  • one or more dose(s) of about 40 IU C1-INH per kg bodyweight is/are administered to a patient during acute treatment, preferably via intravenous administration.
  • Freeze dried C1-INH concentrate (manufactured based on the Berinert® manufacturing process (according to A. Feussner et al., Transfusion 2014, 54: 2566-73)) was dissolved in WFI. PD10 desalting columns (GE Healthcare) were equilibrated with the target formulation buffer. Then the C1-INH solutions were applied per PD10 column. C1-INH was eluted with the corresponding formulation buffer (see Tables 1 and 2) within the column exclusion volume. C1-INH concentration was determined by chromogenic assay (Berichrom C1-Inhibitor, Siemens) and adjusted to 500 IU/ml using the corresponding formulation buffer.
  • Formulated C1-INH was filtered through 0.2 ⁇ m and then dispensed in aliquots to glass vials, lyophilized and then stoppered with rubber stoppers under aseptic conditions. This was done for each of the different C1-INH formulations disclosed in Table 1 and 2. The respective glass vials were finally transferred to tempered rooms at 2-8° C., 25° C. or 40° C. (Table 1) and 2-8° C. or 40° C. (Table 2).
  • Sample vials of each C1-INH formulation were taken at various time points and tested for C1-INH activity by chromogenic assay and for HMWC formation and fragmentation by SEC HPLC on a TSK-Gel G3000SWXL (Tosoh) using 20 mM NaH 2 PO 4 , 20 mM Na 2 HPO 4 , 100 mM NaCl, pH 7.2 as eluent buffer (see FIGS. 1 to 10 ).

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Engineering & Computer Science (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Oil, Petroleum & Natural Gas (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Immunology (AREA)
  • Molecular Biology (AREA)
  • Biochemistry (AREA)
  • Dermatology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Biophysics (AREA)
  • Inorganic Chemistry (AREA)
  • Diabetes (AREA)
  • Hematology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
US16/323,155 2016-08-05 2017-08-04 Pharmaceutical formulations of c1 esterase inhibitor Abandoned US20190175683A1 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
EP16183030 2016-08-05
EP16183030.2 2016-08-05
PCT/EP2017/069765 WO2018024873A1 (en) 2016-08-05 2017-08-04 Pharmaceutical formulations of c1 esterase inhibitor

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2017/069765 A-371-Of-International WO2018024873A1 (en) 2016-08-05 2017-08-04 Pharmaceutical formulations of c1 esterase inhibitor

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US16/932,919 Continuation US11554156B2 (en) 2016-08-05 2020-07-20 Pharmaceutical formulations of C1 esterase inhibitor

Publications (1)

Publication Number Publication Date
US20190175683A1 true US20190175683A1 (en) 2019-06-13

Family

ID=56615877

Family Applications (2)

Application Number Title Priority Date Filing Date
US16/323,155 Abandoned US20190175683A1 (en) 2016-08-05 2017-08-04 Pharmaceutical formulations of c1 esterase inhibitor
US16/932,919 Active 2037-08-11 US11554156B2 (en) 2016-08-05 2020-07-20 Pharmaceutical formulations of C1 esterase inhibitor

Family Applications After (1)

Application Number Title Priority Date Filing Date
US16/932,919 Active 2037-08-11 US11554156B2 (en) 2016-08-05 2020-07-20 Pharmaceutical formulations of C1 esterase inhibitor

Country Status (5)

Country Link
US (2) US20190175683A1 (de)
EP (1) EP3493832A1 (de)
CN (1) CN109562149A (de)
AU (1) AU2017305856A1 (de)
WO (1) WO2018024873A1 (de)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022109124A1 (en) * 2020-11-20 2022-05-27 Csl Behring Gmbh Method for treating antibody-mediated rejection
US11554156B2 (en) 2016-08-05 2023-01-17 Csl Behring Gmbh Pharmaceutical formulations of C1 esterase inhibitor

Family Cites Families (23)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE3228502A1 (de) 1982-07-30 1984-02-02 Behringwerke Ag, 3550 Marburg Verfahren zur herstellung des c1-inaktivators und seine verwendung
DE4222534A1 (de) 1992-07-09 1994-01-13 Behringwerke Ag Verwendung von Komplement-Inhibitoren zur Herstellung eines Arzneimittels zur Prophylaxe und Therapie von entzündlichen Darm- und Hauterkrankungen sowie Purpura
DE4227762A1 (de) 1992-08-24 1994-03-03 Behringwerke Ag Verwendung eines Kallikrein-Inhibitors zur Herstellung eines Arzneimittels zur Prophylaxe und Therapie bestimmter Krankheiten
WO2001077137A1 (en) 2000-04-12 2001-10-18 Human Genome Sciences, Inc. Albumin fusion proteins
AU6083899A (en) 1999-09-16 2001-04-17 Aventis Behring Gmbh Combination of c1-inh and lung surfactant for the treatment of respiratory disorders
US7067713B2 (en) 2000-01-31 2006-06-27 Pharming Intellectual Property B.V. C1 Inhibitor produced in the milk of transgenic non-human mammals
ES2332402T5 (es) 2000-10-12 2018-05-14 Genentech, Inc. Formulaciones de proteína concentradas de viscosidad reducida
WO2006128497A1 (en) * 2005-06-01 2006-12-07 Novo Nordisk A/S Pharmaceutical formulation of factor xi
SI2380587T1 (en) 2005-12-21 2018-03-30 Pharming Intellectual Property B.V. Use of a C1 inhibitor to prevent ischemic reperfusion injury
DK2109457T3 (en) 2007-02-12 2016-04-11 Csl Behring Gmbh THERAPEUTIC USE OF KAZAL TYPE SERINE PROTEASE INHIBITORS
NZ602685A (en) 2010-03-01 2014-10-31 Cytodyn Inc Concentrated protein formulations and uses thereof
CA2829037C (en) 2011-03-09 2022-05-17 Csl Behring Gmbh Factor xii inhibitors for the administration with medical procedures comprising contact with artificial surfaces
EP2497489A1 (de) 2011-03-09 2012-09-12 CSL Behring GmbH Faktor XII-Hemmer zur Behandlung von schleichender Ischämie und Ischämie in anderen Organen
IL230564B2 (en) 2011-07-22 2023-09-01 Csl Ltd Anticoagulant monoclonal antibodies anti–factor xii/fxiia, a method for their preparation, a pharmaceutical compound containing them and medical uses.
AU2012311483B2 (en) 2011-09-24 2016-03-10 Csl Behring Gmbh Combination therapy using immunoglobulin and C1-inhibitor
EP2623110A1 (de) 2012-01-31 2013-08-07 CSL Behring GmbH Faktor-XII-Hemmer zur Behandlung neurologischer Entzündungserkrankungen
KR102403299B1 (ko) 2013-03-08 2022-06-03 체에스엘 베링 게엠베하 원격 허혈-재관류 손상의 치료 및 예방
DE202014011208U1 (de) 2013-03-15 2018-08-23 Shire Viropharma Incorporated C1-INH Zusammensetzungen für die Vorbeugung und Behandlung von Störungen, die mit C1-Esterasehemmer-Defizienz assoziiert sind
CA2916399C (en) 2013-06-28 2022-08-02 Marc Nolte Combination therapy using a factor xii inhibitor and a c1-inhibitor
TWI694836B (zh) * 2014-05-16 2020-06-01 英商葛蘭素史克智慧財產管理有限公司 抗體調配物
JP6516855B2 (ja) 2015-02-20 2019-05-22 ツェー・エス・エル・ベーリング・ゲー・エム・ベー・ハー C1エステラーゼ阻害剤の医薬製剤
WO2018024873A1 (en) 2016-08-05 2018-02-08 Csl Behring Gmbh Pharmaceutical formulations of c1 esterase inhibitor
JP2019534240A (ja) 2016-08-23 2019-11-28 ツェー・エス・エル・ベーリング・ゲー・エム・ベー・ハー C1エステラーゼ阻害剤欠乏に関連する遺伝性血管浮腫の急性発作を予防する方法

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11554156B2 (en) 2016-08-05 2023-01-17 Csl Behring Gmbh Pharmaceutical formulations of C1 esterase inhibitor
WO2022109124A1 (en) * 2020-11-20 2022-05-27 Csl Behring Gmbh Method for treating antibody-mediated rejection

Also Published As

Publication number Publication date
CN109562149A (zh) 2019-04-02
AU2017305856A1 (en) 2019-03-21
EP3493832A1 (de) 2019-06-12
US20200390845A1 (en) 2020-12-17
US11554156B2 (en) 2023-01-17
WO2018024873A1 (en) 2018-02-08

Similar Documents

Publication Publication Date Title
JP7150804B2 (ja) プラミノーゲンを含む医薬組成物及びその使用
TWI670072B (zh) 凍乾的重組vwf調配物
ES2193159T5 (es) Una formulación de factor de coagulación viii.
JP7175506B2 (ja) 低血圧治療のための単独または併用使用されるアンギオテンシンii
RU2510279C2 (ru) Новые защитные композиции для рекомбинантного фактора viii
US11554156B2 (en) Pharmaceutical formulations of C1 esterase inhibitor
KR20140084208A (ko) 정제된 인자 viii의 재구성 후의 안정성을 향상시키는 방법
IE64080B1 (en) Pharmaceutical for subcutaneous administration containing polypeptides
US20180036394A1 (en) Pharmaceutical formulations of c1 esterase inhibitor
US20220257702A1 (en) Stable albuvirtide compositions
US10441631B2 (en) Therapeutic agent for amniotic fluid embolism
JPH08109140A (ja) 高血圧症予防治療剤
JPH07316072A (ja) 妊娠中毒症治療剤
JPH09132534A (ja) アンチトロンビン−iiiの液状製剤およびその保存 安定化方法
RU2774720C2 (ru) ЛЕЧЕНИЕ ЖЕЛУДОЧНО-КИШЕЧНОГО КРОВОТЕЧЕНИЯ У ПАЦИЕНТОВ С ТЯЖЕЛОЙ ФОРМОЙ БОЛЕЗНИ ФОН ВИЛЛЕБРАНДА ПУТЕМ ВВЕДЕНИЯ РЕКОМБИНАНТНОГО ФфВ
US5888964A (en) Method for increasing placental blood flow
JPH0867636A (ja) 播種性血管内凝固症候群(dic)の予防治療薬
JP3806945B2 (ja) ヒト由来アンチトロンビン−iiiの新規用途
JPH08245419A (ja) ショックに起因する胃粘膜障害予防治療剤

Legal Events

Date Code Title Description
STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

AS Assignment

Owner name: CSL BEHRING GMBH, GERMANY

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:METZNER, HUBERT;KANZY, ERNST-JUERGEN;SIGNING DATES FROM 20170105 TO 20170116;REEL/FRAME:052221/0885

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION