EP1414498A2 - Stabilized formulations of interferons with sulfoalkyl ether cyclodextrins - Google Patents

Stabilized formulations of interferons with sulfoalkyl ether cyclodextrins

Info

Publication number
EP1414498A2
EP1414498A2 EP02748632A EP02748632A EP1414498A2 EP 1414498 A2 EP1414498 A2 EP 1414498A2 EP 02748632 A EP02748632 A EP 02748632A EP 02748632 A EP02748632 A EP 02748632A EP 1414498 A2 EP1414498 A2 EP 1414498A2
Authority
EP
European Patent Office
Prior art keywords
interferon
polypeptide
composition according
interferon gamma
amino acid
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.)
Withdrawn
Application number
EP02748632A
Other languages
German (de)
English (en)
French (fr)
Inventor
Jorn Drustrup
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.)
Maxygen ApS
Maxygen Holdings Ltd
Original Assignee
Maxygen ApS
Maxygen Holdings Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Maxygen ApS, Maxygen Holdings Ltd filed Critical Maxygen ApS
Publication of EP1414498A2 publication Critical patent/EP1414498A2/en
Withdrawn legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/555Interferons [IFN]
    • C07K14/565IFN-beta
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • 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
    • 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
    • A61P31/16Antivirals for RNA viruses for influenza or rhinoviruses
    • 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
    • A61P31/20Antivirals for DNA viruses
    • 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
    • A61P31/20Antivirals for DNA viruses
    • A61P31/22Antivirals for DNA viruses for herpes viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00

Definitions

  • the present invention relates to interferon compositions, such as pharmaceutical interferon compositions and methods of their preparation.
  • interferon compositions such as pharmaceutical interferon compositions and methods of their preparation.
  • stabilized compositions comprising an interferon polypeptide and a sulfoalkyl ether cyclodextrin derivative.
  • Interferons are important cytokines characterized by antiviral, antiproliferative, and immunomodulatory activities. These activities form a basis for the clinical benefits that have been observed in a number of diseases, including hepatitis, various cancers and multiple sclerosis.
  • the interferons are divided into the type I and type II classes. Type I interferons includes interferons ⁇ , beta, ⁇ and ⁇ , whereas interferon ⁇ is the only known member of the distinct type II class. Interferons are reviewed by Aggarwall and Gutterman, in Human Cytokines, Vol I, Blackwell Science, Inc. 1996.
  • Interferon beta and variants and conjugates thereof are described in WO 01/15736, and in PCT/DK02/00128, the contents of which are incorporated herein by reference.
  • Interferon gamma is a cytokine produced by T-lymphocytes and natural killer cells and exists as ahomodimer of two noncovalently bound polypeptide subunits.
  • the mature form of each monomer comprises 143 amino acid residues (shown in SEQ ID NO: 2) and the precursor form thereof, including the signal sequence, comprises 166 amino acid residues (shown in SEQ ID NO: 3).
  • Each subunit has two potential N-glycosylation sites (Aggarwal et al., Human
  • Interferon gamma variants and conjugates thereof are described in WO 01/36001, the content of which is incorporated herein by reference.
  • Experimental 3D structures of wild-type human interferon gamma determined by X-ray crystallography have been reported by Ealick et al. Science 252:698-702 (1991) who reported the C-alpha trace of an interferon gamma homodimer.
  • Walter et al. Nature 376:230-235 (1995) disclosed the structure of an interferon gamma homodimer in complex with two molecules of a soluble form of the interferon gamma receptor. The coordinates ofthis structure, however, have never been made publicly available. Thiel et al.
  • Structure 8:927-936 (2000) showed the structure of an interferon gamma homodimer in complex with two molecules of a soluble form of the interferon gamma receptor having a third molecule of the receptor in the structure not making interactions with the interferon gamma homodimer.
  • Recombiant human interferon gamma is usually applicable via parenteral, preferably via subcutaneous, injection.
  • One problem recognized in connection with formulation of interferons into pharmaceutical products is aggregation of the interferon polypeptide. This problem has been attempted solved by use of various stabilizers e.g. as described in WO 98/28007, WO 99/15193 and WO 01/24814.
  • Commercially available interferon beta products have been stabilized by human serum albumin.
  • compositions comprise a sulfoalkyl ether cyclodextrin derivative as a stabilizing agent.
  • the invention relates to a stabilized pharmaceutical composition
  • a stabilized pharmaceutical composition comprising an interferon polypeptide and a sulfoalkyl ether cyclodextrin derivative.
  • interferon polypeptide is intended to indicate a polypeptide exhibiting interferon activity, e.g. as defined by Aggarwel and Gutterman, op cit.
  • the interferon polypeptide is typically selected from the group consisting of interferon alpha, interferon beta, interferon omega, interferon tau, interferon epsilon and interferon gamma.
  • the invention relates to a primary product container comprising an interferon polypeptide and a sulfoalkyl ether cyclodextrin derivative.
  • the invention relates to a method for increasing stability of an interferon polypeptide formulated into a pharmaceutical composition, said method comprising incorporating into said composition a sulfoalkyl ether cyclodextrin derivative and optionally a buffering agent.
  • the invention relates to a method of subjecting a mammal to interferon therapy, which method comprises administrering a therapeutically effective amount of a composition comprising an interferon polypeptide and a sulfoalkyl ether cyclodextrin derivative.
  • the invention relates to a pharmaceutical composition
  • a pharmaceutical composition comprising an interferon polypeptide and a sulfoalkyl ether cyclodextrin derivative.
  • the invention relates to use of a composition comprising an interferon polypeptide and a sulfoalkyl ether cyclodextrin derivative for the manufacture of a medicament for treatment of a disease or disorder.
  • this invention provides compositions and methods for treating most types of viral infections, cancers or tumors or tumour angiogenesis, Crohn's disease, ulcerative colitis, Guillain-Barre syndrome, glioma, idiopathic pulmonary fibrosis, abnormal cell growth, or for immunomodulation in any suitable animal, preferably mammal, and in particular human.
  • the composition of the invention may be used in the treatment of osteosarcoma, basal cell carcinoma, ovarian carcinoma, cervical dysplasia, cervical carcinoma, laryngeal papillomatosis, mycosis fungoides, glioma, acute myeloid leukemia, multiple myeloma, Hodgkin s disease, melanoma, breast carcinoma, non-small cell lung cancer, malignant melanoma (adjuvant, late stage, as well as prophylactic), carcinoid tumour, B-cell lymphoma, T-cell lymphoma, follicular lymphoma, Kaposi's sarcoma, chronic myelogenous leukaemia, renal cell carcinoma, recurrent superfiecial bladder cancer, colorectal carcinoma, hairy cell leukaemia, and viral infections such as papilloma virus, viral hepatitis, herpes genitalis, herpes zoster, herpetic keratitis, herpe
  • polypeptide or composition of the invention may be used for the treatment of multiple sclerosis (MS), such as any of the generally recognized four types of MS (benign, relapsing remitting MS (RRMS), primary progressive MS (PPMS) and secondary progressive MS (SPMS)) and for monosymptomatic MS), cancer or tumours, hepatitis, e.g. hepatitis B and hepatitis C, or a herpes infection (the latter treatment optionally being combined with a treatment with IL-10).
  • MS multiple sclerosis
  • RRMS relapsing remitting MS
  • PPMS primary progressive MS
  • SPMS secondary progressive MS
  • cancer or tumours cancer or tumours
  • hepatitis e.g. hepatitis B and hepatitis C
  • herpes infection the latter treatment optionally being combined with a treatment with IL-10.
  • the composition of the invention may be used for treatment of any of the medical indications 5 described in WO 01/36001 , in particular interstitial pulmonary diseases, most particularly idiopathic pulmonary fibrosis.
  • Interferon gamma has been suggested for treatment of interstitial lung diseases (IPF) for which purpose interferon gamma can be used in combination with prednisolone.
  • IPF interstitial lung diseases
  • granulomatous diseases In addition to IPF, granulomatous diseases, certain mycobacterial infections, kidney cancer, osteopetrosis, scleroderma , hepatitis B, hepatitis C, septic shock, and l o rheumatoid arthritis may be treated with interferon gamma.
  • the invention relates to a kit comprising a composition according to the invention.
  • the term "stabilized” is intended to mean that the composition has increased storage stability as compared to a composition which does not comprise the sulfoalkyl ether cyclodextrin derivative.
  • the increased storage stability is observed in a liquid formulation stored as such or stored in a frozen state and thawed prior to use, in a dried form, 0 e.g. lyophilized, spray-dried or air-dried form, for later reconstitution into a liquid form prior to use, in a solid form, e.g. intented for pulmonary or nasal delivery, and/or in any other form, e.g. made for a special drug delivery system (such as microspheres or the like).
  • the increased storage stability is typically measured in terms of increased bioactivity as compared to the reference composition when subjected to the same storage conditions.
  • the increased storage 5 stability is intended to comprise physical and/or chemical stability.
  • bioactivity is intended to mean an in vitro and/or in vivo bioactivity as dete ⁇ nined by any suitable assay.
  • bioactivity can be measured in terms of antiviral activity, antiproliferative activity, immunomodulatory activity, receptor binding/activation activity, etc. according to methods known in the art relevant for the interferon polypeptide of
  • interferon polypeptides which for other reasons loose activity during storage, e.g. as a consequence of chemical or other physical degradation during storage, maybe stabilized according to the present invention.
  • aggregate formation is intended to mean a physical interaction between the interferon polypeptides that results in formation of oligomers. Aggregate formation is undesirable since in most cases this leads to reduced or even lost bioactivity and/or increased immunogenicity.
  • the composition of the invention is a liquid composition aggregates may remain soluble or be in the form of large visible aggregates that precipitate out of solution.
  • aggregates may have been formed during preparation thereof resulting in an inferior formulation. Such aggregate formation may be measured by visual inspection, or be measured in any suitable spectrophotometric device.
  • the present invention is generally applicable to all types of interferon including interferons isolated from natural sources (e.g. human leukocytes or fibroblasts), recombinantly produced naturally occurring or variant interferons, as well as chemically synthesized interferons.
  • the interferon polypeptide may be, but is not limited to, a type I interferon or a type II interferon, e.g. selected from the group consisting of interferon alpha, interferon beta, interferon omega, interferon tau, interferon epsilon and interferon gamma.
  • the interferon polypeptide may have the amino acid sequence of that found in nature (wildtype interferon) or may be a variant of such wildtype interferon.
  • the interferon polypeptide may be a variant of a wildtype interferon comprising one or more amino acid modifications, i.e. deletions, insertions or substitutions, and exhibiting interferon activity.
  • modifications can be made in a site-specific manner (e.g. by use of site-directed mutagenesis) or in a random or semi-random manner, e.g. by use of random or localized random mutagenesis, e.g. as defined in WO 01/04287 or by use of directed evolution technology, e.g. as described by Stemmer, Bio/Technology 13:549-553 (1995), US 5,605,793, US 5,830,721, US 5,811,238, etc.
  • the variant comprises at most 15 amino acid modifications, e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14 or 15 modifications.
  • the interferon polypeptide is a human interferon (having the amino acid sequence of a naturally occurring human interferon) or a variant thereof.
  • conjugated polypeptide is intended to indicate a heterogeneous (in the sense of composite or chimeric) molecule formed by the covalent attachment of one or more interferon polypeptide(s) to one or more non- polypeptide moieties.
  • covalent attachment means that the interferon polypeptide and the non-polypeptide moiety are either directly covalently joined to one another, or else are indirectly covalently joined to one another through an intervening moiety or moieties, such as a bridge, spacer, or linkage moiety or moieties.
  • a conjugated interferon polypeptide is soluble at relevant concentrations and conditions, i.e. soluble in physiological fluids such as blood.
  • conjugated interferon polypeptides include glycosylated and/or PEGylated interferon polypeptides.
  • non-conjugated polypeptide may be used about the polypeptide part of the conjugated interferon polypeptide.
  • interferon polypeptides may, if desired, comprise other modifications. These may, for example, include addition of one or more extra residues at the N-terminus, e.g. addition of a Met residue at the N-te ⁇ ninus and/as truncation of one or more C-terminal residues as well as "conservative acid substitutions", i.e. substitutions performed within groups of amino acids with similar characteristics, e.g. small amino acids, acidic amino acids, polar amino acids, basic amino acids, hydrophobic amino acids and aromatic amino acids. Examples of conservative substitutions may, in particular, be selected from the groups listed in the table below.
  • random mutagenesis refers to a mutagenic process that is random with respect to the site of mutation within the subject nucleic acid, and that is random with respect to the mutations introduced, e.g., chemical mutagenesis, uv or ⁇ irradiation, passage through repair deficient cells, etc.
  • localized mutagenesis is used to indicate that the mutagenic process occurs preferentially in a predetermined portion or subsequence of the subject nucleic acid.
  • site directed mutagenesis refers to an alteration at a predetermined nucleotide position or positions, normally with the aim of altering one or more amino acid residues of the encoded amino acid sequence.
  • the site-directed mutagenesis is normally designed on the basis of an analysis of a primary or tertiary (e.g. model) structure of the polypeptide to be modified.
  • attachment group is intended to indicate an amino acid residue group capable of coupling to the relevant non-polypeptide moiety such as a polymer molecule or a sugar moiety. Useful attachment groups and their matching non-polypeptide moieties are apparent from the table below.
  • attachment group is used in an unconventional way to indicate the amino acid residues constituting an N-glycosylation site (with the sequence N-X'-S/T/C-X", wherein X' is any amino acid residue except proline, X" any amino acid residue that may or may not be identical to X' and that preferably is different from proline, N is asparagine and S/T/C is either serine, threonine or cysteine, preferably serine or threonine, and most preferably threonine).
  • amino acid residue comprising an attachment group for the non-polypeptide moiety as used in connection with alterations of the amino acid sequence of the parent polypeptide is to be understood as one, two or all of the amino acid residues constituting an N- glycosylation site is/are to be altered in such a manner that either a functional N- glycosylation site is introduced into the amino acid sequence or removed from said sequence.
  • removal and/or introduction of amino acid residues comprising an attachment group for a non-polypeptide moiety is primarily of interest when the interferon polypeptide is in the form of a conjugate having one or more attached non- polypeptide moieties.
  • By removing and/or introducing amino acid residues comprising an attachment group for a non-polypeptide moiety it is possible to optimize the number and distribution of non-polypeptide moieties conjugated to the interferon polypeptide (e.g.
  • the interferon polypeptide is boosted or otherwise altered in the content of the specific amino acid residues to which the relevant non- polypeptide moiety binds, whereby a more efficient, specific and/or extensive conjugation is achieved.
  • attachment groups By removal of one or more attachment groups it is possible to avoid conjugation to the non-polypeptide moiety in parts of the polypeptide in which such conjugation is disadvantageous, e.g. to an amino acid residue located at or near a functional site of the polypeptide (since conjugation at such a site may result in inactivation or reduced interferon activity of the resulting conjugate due to impaired receptor recognition). Further, it may be advantageous to remove an attachment group located closely to another attachment group in order to avoid heterogeneous conjugation to such groups.
  • amino acid residue comprising an attachment group for a non-polypeptide moiety is selected on the basis of the nature of the non-polypeptide moiety and, in most instances, on the basis of the conjugation method to be used.
  • the non-polypeptide moiety is a polymer molecule, such as a polyethylene glycol or polyalkylene oxide derived molecule
  • amino acid residues capable of functioning as an attachment group may be selected from the group consisting of lysine, cysteine, aspartic acid, glutamic acid and arginine.
  • the attachment group is an in vivo glycosylation site, preferably an N-glycosylation site.
  • the amino acid residue to be introduced or removed is normally located in a surface exposed position of the interferon polypeptide, preferably in a position that is occupied by an amino acid residue which has more than 25% of its side chain exposed to the solvent, preferably more than 50% of its side chain exposed to the solvent.
  • Such positions can be identified on the basis of an analysis of a 3D structure of the interferon polypeptide or by any other suitable method, e.g. as described in WO 01/15736 wherein interferon beta is used as an example.
  • Substitutions that lead to introduction of an additional N-glycosylation site at positions exposed at the surface of the interferon beta polypeptide and occupied by amino acid residues having more than 25% of the side chain exposed to the surface include:
  • Substitutions that lead to introduction of an additional N-glycosylation site at positions exposed at the surface of the interferon beta polypeptide having more than 50% of the side chain exposed to the surface include:
  • Substitutions that lead to introduction of an N-glycosylation site by only one amino acid substitution include: L6S/T, RI IN, D39S/T, Q72N, D73N, S75N, L88S T, Y92S/T, L98S/T, D110N, LI 16N, E137N, R159N and L160S/T, the substitutions being indicated relative to the amino acid sequence of wildtype human interferon beta shown in SEQ ID NO 1.
  • a substitution is preferred that is selected from the group consisting of L6S/T, RI IN, D39S/T, Q72N, D73N, S75N, L88S/T, Y92S/T, L98S/T, Dl ION and LI 16N, more preferably from the group consisting of L6S/T, D39S/T, D73N, S75N, L88S/T, D110N, L116N and E137N; and most preferably selected from the group consisting of L6S/T, D39S/T, D73N, S75N, L88S/T, D110N and L116N.
  • the introduced amino acid residue comprising an attachment group for a non- polypeptide moiety creates a new N-glycosylation site or a new PEGylation site.
  • the interferon polypeptide comprises a glycosylation site
  • the utilization of such site may be optimised. This can be achieved by modification of an amino acid residue located close to said glycosylation site, the modification being of a type resulting in an increasing glycosylation.
  • the in vivo glycosylation site is an N-glycosylation site, but it can also be an O-glycosylation site.
  • the term "increased glycosylation” is intended to indicate increased levels of attached carbohydrate molecules, normally obtained as a consequence of increased (or better) utilization of glycosylation site(s).
  • the increased glycosylation may be determined by any suitable method known in the art for analyzing attached carbohydrate structures.
  • the term "increased degree of in vivo N-glycosylation” or “increased degree of N-glycosylation” is intended to indicate increased levels of attached carbohydrate molecules, normally obtained as a consequence of increased (or better) utilization of N-glycosylation site(s).
  • interferon gamma it is well-known (Hooker et al., 1998, J. Interferon and Cytokine Res.
  • interferon gamma activity is intended to indicate that the interferon gamma polypeptide has one or more of the functions of native human interferon gamma or recombinant human interferon gamma, including the capability to bind to an interferon gamma receptor and cause transduction of the signal transduced upon human interferon gamma-binding of its receptor as determined in vitro or in vivo (i.e. in vitro or in vivo bioactivity).
  • the interferon gamma receptor has been described by Aguet et al. (Cell 55:273-280, 1988) and Calderon et al. (Proc. Natl. Acad. Sci. USA 85:4837-4841, 1988).
  • a suitable assay for testing interferon gamma activity is the assay entitled "Primary Assay" disclosed herein.
  • interferon gamma polypeptide is a polypeptide exhibiting interferon gamma activity, and is used herein about the interferon gamma polypeptide in monomer or dimeric form, as appropriate. For instance, when specific substitutions are indicated these are normally indicated relative to the interferon gamma polypeptide monomer. It will be understood that the term “interferon gamma polypeptide” also encompasses C- terminally truncated and variant forms of the wild-type interferon gamma polypeptide. Specific examples of such variants include variants with modifications such as S99T, E38N+S40T as well as C-terminally truncated forms thereof. More examples of suitable modifications are given below.
  • the variant forms of the interferon gamma polypeptide differs in 1-15 amino acid residues (such as in 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14 or 15 amino acid residues), e.g. in 1-10 amino acid residues, in 1-5 amino acid residues or in 1-3 amino acid residues compared to human interferon gamma (or where a truncated form thereof is desired: compared to the corresponding truncated human interferon gamma).
  • interferon gamma polypeptide also encompasses interferon gamma polypeptides (having the human wild-type sequence or a variant thereof), wherein 1-15 the interferon gamma polypeptide is C-terminally truncated with 1-15 amino acid residues.
  • interferon gamma polypeptide which has been C-terminally truncated with 3 amino acid residues.
  • human interferon gamma is intended to mean the mature form of wild-type human interferon gamma having the amino sequence shown in SEQ ID NO: 2.
  • recombinant human interferon gamma is intended to cover the mature form of wild-type human interferon gamma having the amino acid sequence shown in SEQ ID NO: 2 which has been produced by recombinant means.
  • Actimmune® refers to the 140 amino acid form of interferon gamma (disclosed in SEQ ID NO: 4) achieved by fermentation of a genetically engineered E.coli bacterium. Further information of Actimmune® is available on www.actimmune.com.
  • An amino acid residue "located close to" a glycosylation site is usually located in position -4, -3, -2, -1, +1, +2, +3 or +4 relative to the amino acid residue of the glycosylation site to which the carbohydrate is attached. Such positions are selected from position -1, +1, or +3, in particular in position +1 or +3.
  • the amino acid residue located close to an N-glycosylation site may be located in position ⁇ -, -3, -2, -1 relative to the N-residue, at position X' or X" (in which case the amino acid residue to be introduced is preferably different from proline), or at position +1 relative to the X" residue.
  • the amino acid modification is normally a substitution, the substitution being made with any other amino acid residue that gives rise to an increased glycosylation of the interferon polypeptide as compared to that of the unmodified polypeptide.
  • Such other amino acid residue may be determined by trial and error type of experiments (i.e. by substitution of the amino acid residue of the relevant position to any other amino acid residue, and determination of the resulting glycosylation of the resulting variant) .
  • the amino acid residue in said position must be either Ser, Thr or Cys.
  • the modification of the amino acid residue in position +2 relative to the in vivo N-glycosylation site is a substitution where the amino acid residue in question is replaced with a Thr residue. If, on the other hand, said amino acid residue is already a Thr residue it is normally not preferred or necessary to perform any substitutions in that position.
  • X' should not be Pro and preferably not Trp, Asp, Glu and Leu.
  • the amino acid residue to be introduced is preferably selected form the group consisting of Phe, Asn, Gin, Tyr, Val, Ala, Met, He, Lys, Gly, Arg, Thr, His, Cys and Ser, more preferably Ala, Met, He, Lys, Gly, Arg, Thr, His, Cys and Ser, in particular Ala or Ser.
  • the amino acid residue to be introduced is preferably selected from the group consisting of His, Asp, Ala, Met, Asn, Thr, Arg, Ser and Cys, more preferably Thr, Arg, Ser and Cys.
  • Such modifications are particular relevant if the X' residue is a Ser residue.
  • a free cysteine of the interferon polypeptide may be removed, e.g. by substitution with another amino acid residue such as a neutral amino acid residue such as Gly, Val, Ala, Leu, He, Tyr, Phe, His, Trp, Ser, Thr or Met, preferably Ser or Thr, e.g. as described in US 4,959,314 or EP 192 811.
  • the interferon polypeptide can be derivatized by a non-polypeptide moiety, e.g. a polymer molecule such as polyethylene glycol or a sugar moiety (when the interferon polypeptide is glycosylated).
  • non-polypeptide moiety is intended to indicate a molecule that is capable of conjugating to an attachment group of the polypeptide of the invention.
  • Preferred examples of such molecule include polymer molecules, sugar moieties, lipophilic compounds, or organic derivatizing agents.
  • polymer molecule is defined as a molecule formed by covalent linkage of two or more monomers, wherein none of the monomers is an amino acid residue, except where the polymer is human albumin or another abundant plasma protein.
  • polymer may be used interchangeably with the term “polymer molecule”.
  • the term is intended to cover carbohydrate molecules attached by in vitro glycosylation, i.e. a synthetic glycosylation performed in vitro normally involving covalently linking a carbohydrate molecule to an attachment group of the polypeptide, optionally using a cross-linking agent.
  • Carbohydrate molecules attached by in vivo glycosylation, such as N- or O-glycosylation (as further described below)) are referred to herein as "a sugar moiety”.
  • non-polypeptide moieties such as polymer molecule(s) or sugar moieties in the conjugate
  • every reference to "a non-polypeptide moiety" contained in a conjugate or otherwise used in the present invention shall be a reference to one or more non-polypeptide moieties, such as polymer molecule(s) or sugar moieties, in the conjugate.
  • the interferon polypeptide comprises an introduced amino acid residue comprising an attachment group for a non-polypeptide moiety
  • the non-polypeptide moiety is preferably attached to such amino acid residue.
  • the invention in a first aspect relates to a stabilized composition comprising an interferon polypeptide and a sulfoalkyl ether cyclodextrin derivative. In a second aspect the invention relates to a stabilized pharmaceutical composition comprising an interferon polypeptide and a sulfoalkyl ether cyclodextrin derivative.
  • the sulfoalkyl ether cyclodextrin derivative is present in a concentration from 1 mg/ml to 150 mg/ml, such as from 5 mg/ml to 100 mg/ml.
  • the interferon polypeptide comprises at least one introduced and/or at least one removed amino acid residue comprising an attachment group for a non-polypeptide moiety. In a further embodiment the interferon polypeptide comprises at least one introduced and at least one removed amino acid residue comprising an attachment group for a non- polypeptide moiety. In a further embodiment the interferon polypeptide comprises at least one introduced amino acid residue comprising an attachment group for a non-polypeptide moiety. In a further embodiment the interferon polypeptide comprises at least one removed amino acid residue comprising an attachment group for a non-polypeptide moiety.
  • non-polypeptide moiety comprises a polymer molecule, such as polyethylene glycol, or a sugar moiety.
  • polymer molecule comprises a polyethylene glycol.
  • non-polypeptide moiety comprises a sugar moiety, in particular the sugar moiety obtained from expression of the interferon polypeptide in a mammalian cell, preferably a CHO cell.
  • the interferon polypeptide comprises at least one introduced glycosylation site, and at least one sugar moiety attached to an introduced glycosylation site. In a further embodiment the interferon polypeptide comprises at least one introduced glycosylation site, and at least one sugar moiety attached to an introduced glycosylation site, and a polymer molecule, such as polyethylene glycol. In a particular preferred embodiment the interferon polypeptide comprises 3 sugar moieties each sugar moiety being attached to an N-glycosylation site. In a preferred embodiment, the interferon polypeptide is glycosylated and/or PEGylated.
  • the interferon polypeptide is glycosylated and PEGylated. In a further embodiment, the interferon polypeptide is glycosylated. In a further embodiment, the interferon polypeptide is PEGylated. When the interferon polypeptide is glycosylated it is preferably N- glycosylated. When the interferon polypeptide is glycosylated it usually comprises 1-5 sugar moieties, such as 1-3 sugar moieties. In a further embodiment, the interferon polypeptide is N- glycosylated, and comprises 1-5 sugar moieties, such as 1-3 sugar moieties.
  • the interferon polypeptide When the interferon polypeptide is PEGylated it usually comprises 1-5 polyethylene glycol (PEG) molecules. In a further embodiment the interferon polypeptide comprises 1-5 PEG molecules, such as 1, 2 or 3 PEG molecules. In a further embodiment each PEG molecule has a molecular weight of about 5 kDa (kilo Dalton) to 100 kDa. In a further embodiment each PEG molecule has a molecular weight of about 10 kDa to 40 kDa. In a further embodiment each PEG molecule has a molecular weight of about 12 kDa. In a further embodiment each PEG molecule has a molecular weight of about 20 kDa.
  • PEG polyethylene glycol
  • the interferon polypeptide comprises 1-5 PEG molecules, such as 1, 2 or 3 PEG molecules.
  • each PEG molecule has a molecular weight of about 5 kDa (kilo Dalton) to 100 kDa. In
  • the interferon polypeptide comprises 1-3 PEG molecules each having a molecular weight of about 12 kDa, or 1 PEG molecule having a molecular weight of about 20 kDa.
  • Suitable PEG molecules are available from Shearwater Polymers, Inc. and Enzon, Inc. and may be selected from SS-PEG, NPC-PEG, aldehyd-PEG, mPEG-SPA, mPEG-SCM, mPEG- BTC, SC-PEG, tresylatedmPEG (US 5,880,255), or oxycarbonyl-oxy-N-dicarboxyimide-PEG (US 5,122,614).
  • the interferon polypeptide is an interferon beta polypeptide, e.g. wildtype human interferon beta or a variant thereof, optionally conjugated to a non-polypeptide moiety.
  • the interferon beta polypeptide can be a variant of wildtype human interferon beta, wherein the cysteine residue in position 17 has been deleted or substituted with another amino acid residue, e.g. a neutral amino acid residue as mentioned above.
  • the interferon beta polypeptide comprises the C17S mutation, the substitution being indicated relative to the amino acid sequence of wildtype human interferon beta shown in SEQ ID NO 1.
  • interferon beta polypeptide is any of those described in WO 01/15736. In another preferred embodiment the interferon beta polypeptide is any of those described in PCT/DK02/00128.
  • Preferred variants include a variant with at least one introduced glycosylation site, e.g. in position S2, Q49, Q51 , or F 111 ; and a variant with at least one removed PEGylation site, e.g. in position K19, K33, K45 or K123.
  • the interferon beta polypeptide is a conjugate comprising at least one first non-polypeptide moiety covalently attached to an interferon ⁇ polypeptide, the amino acid sequence of which differs from that of wild-type human interferon ⁇ in at least one introduced and at least one removed amino acid residue comprising an attachment group for said first non-polypeptide moiety.
  • the interferon beta polypeptide is a conjugate comprising at least one first non-polypeptide moiety conjugated to at least one lysine residue of an interferon ⁇ polypeptide, the amino acid sequence of which differs from that of wild-type human interferon ⁇ in at least one introduced and/or at least one removed lysine residue.
  • the interferon beta polypeptide is a conjugate comprising at least one first non-polypeptide moiety conjugated to at least one cysteine residue of an interferon ⁇ polypeptide, the amino acid sequence of which differs from at least one introduce cysteine residue into a position that in wild-type human interferon ⁇ is occupied by a surface exposed amino acid residue.
  • the interferon beta polypeptide is a conjugate comprising at least one first non-polypeptide moiety having an acid group as an attachment group, which moiety is conjugated to at least one aspartic acid or glutamic acid residue of an interferon ⁇ polypeptide, the amino acid sequence of which differs from that of wild-type human interferon ⁇ in at least one introduced and/or at least one removed aspartic acid or glutamic acid residue.
  • the first non-polypeptide moiety comprises a polymer molecule, such as polyethylene glycol, or a sugar moiety.
  • the interferon beta polypeptide is a conjugate comprising at least one polymer molecule and at least one sugar moiety covalently attached to an interferon ⁇ polypeptide, the amino acid sequence of which differs from that of wild-type human interferon ⁇ in a) at least one introduced and/or at least one removed amino acid residue comprising an attachment group for the polymer molecule, and b) at least one introduced and/or at least one removed amino acid residue comprising an attachment group for the sugar moiety, provided that when the attachment group for the polymer molecule is a cysteine residue, and the sugar moiety is an N-linked sugar moiety, a cysteine residue is not inserted in such a manner that an N-glycosylation site is destroyed.
  • the interferon beta polypeptide is a conjugate comprising an interferon ⁇ polypeptide, the amino acid sequence of which differs from that of wild-type human interferon ⁇ in at least one introduced glycosylation site, the conjugate further comprising at least one un-PEGylated sugar moiety attached to an introduced glycosylation site.
  • the interferon beta polypeptide is a conjugate comprising an interferon ⁇ polypeptide, the amino acid sequence of which differs from that of wild-type human interferon ⁇ in at least one introduced glycosylation site, the conjugate further comprising at least one sugar moiety attached to an introduced glycosylation site.
  • the invention relates to a stabilized composition
  • a stabilized composition comprising: a) a conjugate comprising an interferon ⁇ polypeptide, the amino acid sequence of which differs from that of wild-type human interferon ⁇ in at least one introduced glycosylation site, the conjugate further comprising at least one sugar moiety attached to an introduced glycosylation site, and b) a sulfoalkyl ether cyclodextrin derivative.
  • the interferon beta polypeptide is a conjugate comprising an interferon ⁇ polypeptide, the amino acid sequence of which differs from that of wild-type human interferon ⁇ in that a glycosylation site has been introduced or removed by way of introduction or removal of amino acid residue(s) constituting a part of a glycosylation site in a position that in wildtype human interferon ⁇ is occupied by a surface exposed amino acid residue.
  • the interferon beta polypeptide is a conjugate comprising an interferon ⁇ polypeptide, the amino acid sequence of which differs from that of wild-type human interferon ⁇ in that a glycosylation site has been introduced by way of introduction of amino acid residue(s) constituting a part of a glycosylation site in a position that in wildtype human interferon ⁇ is occupied by a surface exposed amino acid residue.
  • interferon beta polypeptide is a conjugate comprising a sugar moiety covalently attached to an interferon ⁇ polypeptide, the amino acid sequence of which differs from that of wild-type human interferon ⁇ in at least one removed glycosylation site.
  • the interferon beta polypeptide is a glycosylated variant of a parent interferon ⁇ polypeptide comprising at least one in vivo glycosylation site, wherein an amino acid residue of said parent polypeptide located close to said glycosylation site has been modified to obtain a variant polypeptide having increased glycosylation as compared to the glycosylation of the parent interferon ⁇ polypeptide.
  • the interferon beta polypeptide comprises amino acid substitutions selected from the group consisting of K19R+K45R+K123R; K19Q+K45R+K123R; K19R+K45Q+K123R;
  • Specific interferon beta variants of interest for the present invention comprises amino acid substitutions selected from the group consisting of
  • each of the specified variants is considered an embodiment of the interferon beta variant.
  • one embodiment is C17S+Q49N+Q51T+D110F+F11 IN+Rl 13T; and another embodiment is C17S+K19R+K33R+K45R+Q49N+Q51T+D110F+F11 IN+Rl 13T.
  • Particularly preferred embodiments of the above specified variants are such wherein the indicated substitutions are the only substitutions relative to the wildtype human interferon beta shown in SEQ ID NO 1.
  • variants may be in the form of conjugates, which further comprise one or more non-polypeptide moieties.
  • the variant is glycosylated and/or PEGylated.
  • the interferon beta polypeptide is glycosylated it is preferably N-glycosylated.
  • the interferon beta polypeptide is glycosylated it usually comprises 1-5 sugar moieties, such as 1-3 sugar moieties.
  • the interferon beta polypeptide is N-glycosylated, and comprises 1-5 sugar moieties, such as 1-3 sugar moieties.
  • the interferon beta polypeptide comprises at least one introduced glycosylation site it is preferred that the polypeptide further comprises at least one sugar moiety attached to the introduced glycosylation site.
  • the interferon beta polypeptide comprises 2-5 sugar moieties, such as 2-3 sugar moieties.
  • the interferon beta polypeptide is N-glycosylated, and comprises 2-5 sugar moieties, such as 2-3 sugar moieties.
  • the interferon beta polypeptide comprises 3 sugar moieties each sugar moiety being attached to an N-glycosylation site.
  • the interferon beta polypeptide When the interferon beta polypeptide is PEGylated it usually comprises 1-5 polyethylene glycol (PEG) molecules. In a further embodiment the interferon beta polypeptide comprises 1-5 PEG molecules, such as 1, 2 or 3 PEG molecules. In a further embodiment each PEG molecule has a molecular weight of about 5 kDa (kilo Dalton) to 100 kDa. In a further embodiment each PEG molecule has a molecular weight of about 10 kDa to 40 kDa. In a further embodiment each PEG molecule has a molecular weight of about 12 kDa. In a further embodiment each PEG molecule has a molecular weight of about 20 kDa.
  • PEG polyethylene glycol
  • the interferon beta polypeptide comprises 1-5 PEG molecules, such as 1, 2 or 3 PEG molecules.
  • each PEG molecule has a molecular weight of about 5 kDa (kilo Dalton) to 100 k
  • the interferon beta polypeptide comprises 1-3 PEG molecules each having a molecular weight of about 12 kDa, or 1 PEG molecule having a molecular weight of about 20 kDa. More preferably the interferon beta polypeptide comprises at least one sugar moiety attached to an introduced glycosylation site and 1 PEG molecule having a molecular weight of about 20 kDa. Most preferably the interferon beta polypeptide comprises 3 sugar moieties attached to 3 N- glycosylation sites and 1 PEG molecule having a molecular weight of about 20 kDa.
  • the interferon polypeptide may be produced according to methods known in the art.
  • the interferon polypeptide is produced recombinantly by expression from a glycosylating host cell (as described in detail in WO 01/15736, PCT/DK02/00128, and in WO 01/36001).
  • the expression host cell may be selected from fungal (filamentous fungal or yeast), insect, mammalian animal cells, from transgenic plant cells or from transgenic animals.
  • the glycosylation may be achieved in the human body when using a nucleotide sequence encoding the polypeptide part of a conjugate of the invention or a polypeptide of the invention in gene therapy.
  • the host cell is a mammalian cell, such as a CHO cell, BHK or HEK cell, e.g. HEK293, or an insect cell, such as an SF9 cell, or a yeast cell, e.g. Saccharomyces cerevisiae, Pichia pastoris or any other suitable glycosylating host.
  • suitable mammalian host cells include Chinese hamster ovary (CHO) cell lines, (e.g. CHO-K1; ATCC CCL-61), Green Monkey cell lines (COS) (e.g. COS 1 (ATCC CRL-1650), COS 7 (ATCC CRL- 1651)); mouse cells (e.g.
  • sugar moieties attached to the polypeptide by in vivo glycosylation are further modified by use of glycosyltransferases, e.g. using the glycoAdvanceTM technology marketed by Neose, Horsham, PA, USA. Thereby, it is possible to, e.g., increase the sialyation of the glycosylated polypeptide following expression and in vivo glycosylation by CHO cells.
  • the interferon polypeptide is purified according to methods known in the art to obtain an interferon preparation of sufficient purity to be useful as a drug.
  • purification methods involve ultrafiltration, diafiltration, cation exchange chromatography (eg. S-Sepharose from Pharmacia), Hydrofobic Interaction Chromatography, hydroxyapatite chromatography, and/or separation on Sephacryl column.
  • interferon polypeptide is an interferon gamma polypeptide, e.g. wildtype human interferon gamma or a variant thereof, optionally conjugated to a non- polypeptide moiety, such as any of the variants or conjugates described in WO 01/36001.
  • interferon gamma polypeptide is selected from an interferon gamma variant, such as any one of those disclosed in the section "Interferon gamma variants" below.
  • glycosylation of the naturally occurring N-glycosylation site located in position 97 of human interferon gamma may be increased, i.e. an increased fraction of fully, or substantially fully, glycosylated interferon gamma polypeptides may be obtained, by substituting the serine residue located in position 99 of human interferon gamma with a threonine residue.
  • substitution S99T about 90% of the interferon gamma polypeptides present in the harvested medium utilized both N-glycosylation site, whereas only about 60% of the recombinantly produced human interferon gamma polypeptides present in the harvested medium was fully glycosylated.
  • the interferon gamma polypeptide comprises the substitution S99T.
  • in vivo glycosylation sites which may have been introduced into the sequence (see the section entitled "Interferon gamma variants where the non-polypeptide moiety is a sugar moiety") may be optimised.
  • the in vivo glycosylation site is an N-glycosylation site, but also an O- glycosylation site is contemplated as relevant.
  • This optimisation may be achieved by performing a modification, preferably a substitution, in a position, which is located close to a glycosylation site, in particular close to an in vivo N-glycosylation site.
  • N-glycosylation site at position 97 may be further optimised by performing a modification, such as a substitution, in a position selected from the group consisting of E93, K94, L95, T96, Y98, V100 and T101 (i.e. at position -4, -3, -2, -1, +1, +3 or +4 relative to N97).
  • a modification such as a substitution
  • substitutions performed in position 98 include Y98F, Y98N, Y98Q, Y98V, Y98A, Y98M, Y98I, Y98K, Y98G, Y98R, Y98T, Y98H, Y98C and Y98S, preferably Y98A, Y98M, Y98I, Y98K, Y98G, Y98R, Y98T, Y98H, Y98C and Y98S, in particular Y98S.
  • substitutions performed in position 100 include V100H, V100D, V100A, V100M, V100N, V100T, V100R, V100S, or V100C, in particular VI 00T, V100R, V100S or VIOOC.
  • this site may be further optimised by performing a modification, such as a substitution, in a position selected from the group consisting of D21, V22, A23, D24, G26, L28 and F29 (i.e. at position -A, -3, -2, -1, +1, +3 or +4 relative to N25).
  • substitutions performed in position 26 include G26F, G26N, G26Y, G26Q, G26V, G26A, G26M, G26I, G26K, G26R, G26T, G26H, G26C and G26S, preferably G26A, G26M, G26I, G26K, G26R, G26T, G26H, G26C and G26S, more preferably G26A and G26S, in particular G26A.
  • substitutions performed in position 28 include G28H, G28D, G28A, G28M, G28N, G28T, G28R, G28S, or G28S, in particular G28A, G28T, G28R, G28S or G28C.
  • any of the modifications mentioned in connection with optimisation of glycosylation at position 97 may be combined with any of the mentioned in connection with optimisation of glycosylation at position 25.
  • Interferon gamma variants comprising attachment groups for non-polypeptide moieties
  • Another class of interesting modifications that may be introduced include modifications, which serve to increase the AUC when administered subcutaneously and/or the serum half-life when administered intravenously.
  • the interferon gamma polypeptide comprises at least one introduced and/or at least one removed amino acid residue comprising an attachment group for a non-polypeptide.
  • the total number of amino acid residues to be modified in accordance with this embodiment typically does not exceed 15.
  • the amino acid sequence comprises 1-10, such as 1-5 e.g. 1-3 modifications compared to SEQ ID NO: 2 (or a C-terminally truncated forms thereof).
  • the modification(s) is/are a substitution(s).
  • the polypeptide may comprise other modifications, e.g. substitutions, that are not related to introduction and/or removal of amino acid residues comprising an attachment group for the non-polypeptide moiety.
  • modifications include conservative amino acid substitutions and/or introduction of Cys-Tyr-Cys or Met at the N-terminus.
  • the exact number of attachment groups available for conjugation and present in the interferon gamma polypeptide in dimeric form is dependent on the effect desired to be achieved by the conjugation.
  • the effect to be obtained is, e.g., dependent on the nature and degree of conjugation (e.g. the identity of the non-polypeptide moiety, the number of non-polypeptide moieties desirable or possible to conjugate to the polypeptide, where they should be conjugated or where conjugation should be avoided, etc.).
  • amino acid residue comprising an attachment group for a non-polypeptide moiety is selected on the basis of the nature of the non-polypeptide moiety part of choice and, in most instances, on the basis of the conjugation method to be used.
  • the non-polypeptide moiety is a polymer molecule, such as a polyethylene glycol- or polyalkylene oxide- derived molecule
  • amino acid residues capable of functioning as an attachment group may be selected from the group consisting of cysteine, lysine, aspartic acid, glutamic acid and arginine.
  • cysteine is preferred.
  • the attachment group is, e.g. an in vivo glycosylation site, preferably an N-glycosylation site.
  • the position of the polypeptide to be modified is conveniently selected as follows: The position is preferably located at the surface of the interferon gamma polypeptide, and more preferably occupied by an amino acid residue that has more than 25% of its side chain exposed to the solvent, preferably more than 50% of its side chain exposed to the solvent, as determined on the basis of a 3D structure or model of interferon gamma in its dimeric form, the structure or model optionally further comprising one or two interferon gamma receptor molecules.
  • the position is preferably located at the surface of the interferon gamma polypeptide, and more preferably occupied by an amino acid residue that has more than 25% of its side chain exposed to the solvent, preferably more than 50% of its side chain exposed to the solvent, as determined on the basis of a 3D structure or model of interferon gamma in its dimeric form, the structure or model optionally further comprising one or two interferon gamma receptor molecules.
  • loop regions of interferon gamma may be modified by modification of one or more amino acid residues located in the loop regions of interferon gamma since most amino acid residues within these loop regions are exposed to the surface and located sufficiently far away from functional sites so that non-polypeptide moieties, such as polymer molecules, in particular PEG molecules, and/or N-glycosylation sites, may be introduced without impairing the function of the polypeptide.
  • non-polypeptide moieties such as polymer molecules, in particular PEG molecules, and/or N-glycosylation sites
  • amino acid residues constituting said loop regions are residues N16-K37 (the "A-B loop"), F60-S65 (the “B-C loop”), N83-S84 (the "C-D loop”) and Y98-L103 (the "D-E loop”).
  • attachment groups located at the receptor-binding site of interferon gamma has preferably been removed, preferably by substitution of the amino acid residue comprising such group.
  • the amino acid residues constituting the interferon gamma receptor-binding site are Ql, D2, Y4, V5, E9, K12, G18, H19, S20, D21, V22, A23, D24, N25, G26, T27, L30, K34, K37, K108, Hi ll, E112, 1114, Q115, A118, E119 (see also Example B herein).
  • the distance between amino acid residues located at the surface of the interferon gamma polypeptide is calculated on the basis of a 3D structure of the interferon gamma dimeric polypeptide. More specifically, the distance between the CB's of the amino acid residues comprising such attachment groups, or the distance between the functional group (NZ for lysine, CG for aspartic acid, CD for glutamic acid, SG for cysteine) of one and the CB of another amino acid residue comprising an attachment group are determined. In case of glycine, CA is used instead of CB.
  • interferon gamma polypeptide part any of said distances is preferably more than 8 A, in particular more than lOA in order to avoid or reduce heterogeneous conjugation.
  • interferon gamma exists as a dimeric polypeptide.
  • the polypeptide is normally in homodimeric form (e.g. prepared by association of two interferon gamma polypeptide polypeptides prepared as described herein).
  • the interferon gamma polypeptide may be provided in single chain form, wherein two interferon gamma polypeptide monomers are linked via a peptide bond or a peptide linker.
  • interferon gamma polypeptide in single chain form has the advantage that the two constituent interferon gamma polypeptides may be different which can be advantageous, e.g., to enable asymmetric mutagenesis of the polypeptides.
  • PEGylation sites can be removed from the receptor- binding site from one of the monomers, but retained in the other.
  • PEGylation one monomer has an intact receptor-binding site, whereas the other may be fully PEGylated (and thus provide significantly increased molecular weight).
  • the interferon gamma polypeptide comprises at least one introduced and/or at least one removed glycosylation site, i.e. the non-polypeptide moiety is a sugar moiety.
  • the glycosylation site is an in vivo glycosylation site, i.e. the non-polypeptide moiety is a sugar moiety, e.g. an O-linked or N-linked sugar moiety, preferably an N-linked sugar moiety.
  • the interferon gamma polypeptide comprises at least one introduced glycosylation site, in particular an introduced in vivo N-glycosylation site.
  • the introduced glycosylation site is introduced by a substitution.
  • an in vivo N-glycosylation site may be introduced into a position of the interferon gamma polypeptide comprising an amino acid residue exposed to the surface.
  • said surface-exposed amino acid residue has at least 25% of the side chain exposed to the surface, in particular at least 50% of its side chain exposed to the surface. Details regarding determination of such positions can be found in Example A herein.
  • the N-glycosylation site is introduced in such a way that the N-residue of said site is located in said position.
  • an O-glycosylation site is introduced so that the S or T residue making up such site is located in said position.
  • the in vivo glycosylation site in particular the N residue of the N-glycosylation site or the S or T residue of the O-glycosylation site, is located within the 118 N-terminal amino acid residues of the interferon gamma polypeptide, more preferably within the 97 N-terminal amino acid residues. Still more preferably, the in vivo glycosylation site is introduced into a position wherein only one mutation is required to create the site (i.e. where any other amino acid residues required for creating a functional glycosylation site is already present in the polypeptide).
  • substitutions that lead to introduction of an additional N- glycosylation site at positions exposed at the surface of the interferon gamma polypeptide and occupied by amino acid residues having at least 25% of the side chain exposed to the surface include: Q1N + P3S/T, P3N+V5S/T, K6N+A8S/T, E9N+L11S/T, K12S/T, K13N+F15S/T, Y14N+N16S/T, G18S/T, G18N, G18N+S20T, H19N+D21S/T, D21N+A23S/T, G26N+L28S/T, G31N+L33S/T, K34N+W36S/T, K37S/T, K37N+E39S/T, E38N, E38N+S40T, E39N+D41S/T, S40N+R42S/T, K55N
  • Substitutions that lead to introduction of an additional N-glycosylation site at positions exposed at the surface of the interferon gamma polypeptide having at least 50% of the side chain exposed to the surface include:
  • S/T indicates a substitution to a serine or threonine residue, preferably a
  • Substitutions where only one amino acid substitution is required to introduce an N-glycosylation site include K12S/T, G18S/T, G18N, K37S/T, E38N, M45N, I49N, K61S/T, D63N, Q67N, V70N, K80S/T, F82N, N85S/T, K87S/T, K94N, Q106S/T, E119N, A124N, K130N and R140N, in particular K12S/T, G18N, G18S/T, K37S/T, E38N, K61S/T, D63N, Q67N, K80S/T, N85S/T, K94N, Q106S/T, A124N and K130N (positions with more than 25% of its site chain exposed to the surface in a structure without receptor molecule), or more preferably G18N, E38N, D63N, Q67N, K94N, A124N and K130N (positions with more than
  • the mutations Q1N+P3S/T, E9N+L11S/T, G18N, G18N+S20T, H19N+D21S/T, D21N+A23S/T, G26N+L28S/T, K34N+W36S/T, K37N+E39S/T, E119N and El 19N+S121T should normally not be performed, unless a reduced receptor affinity is desired.
  • Particular preferred interferon gamma polypeptides include at least one substitution selected from the group consisting of K12S, K12T, G18S, G18T, E38N, E38N+S40T, K61S, K61T, N85S, N85T, K94N, Q106S and Q106T, more preferably selected from the group consisting of K12T, G18T, E38N+S40T, K61T, N85T, K94N and Q106T, even more preferably selected from the group consisting of K12T, G18T, E38N+S40T, K61T and 85T, in particular E38N+S40T. It will be understood that the above-identified substitution are preferably combined with the S99T mutation.
  • interferon gamma polypeptides include substitutions selected from the group consisting of K12S+S99T, K12T+S99T, G18S+S99T, G18T+S99T, E38N+S99T, E38N+S40T+S99T, K61S+S99T, K61T+S99T, N85S+S99T, N85T+S99T, K94N+S99T, S99T+Q106S and S99T+Q106T, more preferably selected from the group consisting of K12T+S99T, G18T+S99T, E38N+S40T+S99T, K61T+S99T, N85T+S99T, K94N+S99T and S99T+Q106T, even more preferably selected from the group consisting of K12T+S99T, G18T+S99T, E38N+S40T+S99T, K61T+S99T and N85T+S
  • the interferon gamma polypeptide comprises at least one introduced cysteine residue.
  • the cysteine residue is introduced by substitution.
  • a cysteine residue may be introduced into a position of the interferon gamma polypeptide comprising an amino acid residue exposed to the surface.
  • said surface-exposed amino acid residue has at least 25% of the side chain exposed to the surface, in particular at least 50% of its side chain exposed to the surface.
  • substitutions that lead to introduction of a cysteine residue at positions exposed at the surface of the interferon gamma polypeptide and occupied by amino acid residue having at least 25% of the side chain exposed to the surface include: QIC, D2C, P3C, K6C, E9C, N10C, K13C,
  • Substitutions that lead to introduction of a cysteine residue at positions exposed at the surface of the interferon gamma polypeptide and occupied by amino acid residue having at least 50% of the side chain exposed to the surface include: P3C, K6C, N10C, K13C, N16C, D21C, N25C, G26C, G31C, K34C,
  • said cysteine residue is introduced by a substitution selected from the group consisting of N10C, N16C, E38C, N59C, N83C, K94C, N104C and
  • any of the above-mentioned modified interferon gamma polypeptides further comprises the substitution S99T.
  • substitution S99T the following substitutions are particularly preferred Nl 0C+S99T, Nl 6C+S99T,
  • the introduced cysteine residue(s) may preferably be conjugated to a non-polypeptide moiety, such as PEG or more preferably mPEG.
  • a non-polypeptide moiety such as PEG or more preferably mPEG.
  • the conjugation of the interferon gamma variant and the activated polymer molecules is conducted by use of any conventional method, e.g. as described in the following references
  • activated PEG polymers particularly preferred for coupling to cysteine residues include the following Unear PEGs: vinylsulfone-PEG (VS-PEG), preferably vinylsulfone-mPEG (VS-mPEG); maleimide-PEG (MAL-PEG), preferably maleimide-mPEG (MAL-mPEG) and orthopyridyl-disulfide-PEG (OPSS-PEG), preferably orthopyridyl-disulfide-mPEG (OPSS-mPEG).
  • VS-PEG vinylsulfone-PEG
  • MAL-PEG maleimide-mPEG
  • OPSS-PEG orthopyridyl-disulfide-mPEG
  • PEG or mPEG polymers will have a size of about 5 kDa, about 10 kD, about 12 kDa or about 20 kDa.
  • the interferon gamma variant is usually treated with a reducing agent, such as dithiothreitol (DDT) prior to PEGylation.
  • DDT dithiothreitol
  • the reducing agent is subsequently removed by any conventional method, such as by desalting. Conjugation of PEG to a cysteine residue typically takes place in a suitable buffer at pH 6-9 at temperatures varying from 4°C to 25°C for periods up to 16 hours.
  • the interferon gamma polypeptide 0 comprises at least one introduced N-glycosyltion site and at least one introduced cysteine residue.
  • the cysteine residue and/or the N-glycosylation site are introduced by substitution.
  • Such polypeptides may be prepared by selecting the residues described in the two preceding sections describing suitable positions for introducing N- glycosylation sites and cysteine residues, respectively.
  • the interferon gamma polypeptide comprises substitutions selected from the group consisting of K12T+N16C, K12T+E38C, K12T+N59C, K12T+N83C, K12T+K94C, K12T+N104C, K12T+A124C, G18T+N10C, G18T+E38C, G18T+N59C, G18T+N83C, G18T+K94C, G18T+N104C, G18T+A124C, E38N+S40T+N10C, E38N+S40T+N16C, E38N+S40T+N59C, E38N+S40T+N83C, E38N+S40T+K94C, 0 E38N+S40T+N104C, E38N+S40T+A124C, K61T+N10C, K61T+N16C, K61T+E38C, K61T+N
  • any of the above-mentioned modified interferon gamma polypeptides further comprises the substitution S99T, i.e. the interferon gamma polypeptide comprises substitutions selected from the group consisting of K12T+N16C+S99T, K12T+E38C+S99T, K12T+N59C+S99T, K12T+N83C+S99T, K12T+K94C+S99T, K12T+N104C+S99T, K12T+A124C+S99T, G18T+N10C+S99T, G18T+E38C+S99T, G18T+N59C+S99T, G18T+N83C+S99T, G18T+K94C+S99T, G18T+N104C+S99T, G18T+A124C+S99T, E38N+S40T+N10C+S99T, E38N+S40T+N16C+
  • the introduced cysteine residue(s) may preferably be conjugated to a non-polypeptide moiety, such as PEG or more preferably mPEG.
  • a non-polypeptide moiety such as PEG or more preferably mPEG.
  • the conjugation between the cysteine-containing polypeptide variant and the polymer molecule may be achieved in any suitable manner, e.g. as described in the section entitled "Interferon gamma variants where the non-polypeptide moiety is a molecule, which has cysteine as an attachment group".
  • Interferon samma variants with a reduced receptor affinity One way to increase the serum half-life of an interferon gamma polypeptide would be to decrease the receptor-mediated internalisation and thereby decrease the receptor-mediated clearance.
  • the receptor mediated internalisation is dependent upon the affinity of the interferon gamma dimer for the interferon gamma receptor complex and, accordingly, an interferon gamma polypeptide with a decreased affinity to the interferon gamma receptor complex is expected to be internalised, and hence cleared, to a lesser extent.
  • the affinity of the interferon gamma dimer to its receptor complex may be decreased by performing one or more modifications, in particular substitutions, in the recpetor binding region of the interferon gamma polypeptide.
  • the amino acid residues which constitute the receptor binding region is defined in Example B herein.
  • One class of substitutions that may be performed is conservative amino acid substitutions.
  • the modification performed gives rise to the introduction of anN- glycosylation site.
  • the interferon gamma polypeptide comprises at least one modification in the receptor binding site (as defined herein). More particularly, the interferon gamma polypeptide comprises at least one modification, preferably a substitution, which creates an in vivo N-glycosylation site, in said receptor binding region.
  • substitutions may be selected from the group consisting of Q1N+P3S/T, D2N+Y4S/T, Y4N+K6S/T, V5N+E7S/T, E9N+L11S/T, K12N+Y14S/T, G18N, G18N+S20T, H19N+D21S/T, S20N+V22S/T, D21N+A23S/T, V22N+D24S/T, D24N+G26S/T, G26N+L28S/T, L30N+I32S/T, K34N+W36S/T, K37N+E39S/T, K108N+I110S/T, H111N+L113S/T, E112N+I114S/T, I114N+V116S/T, Ql 15N+M117S/T, Al 18N+L120S/T, El 19N and El 19N+S121T, preferably from
  • Such variants are contemplated to exhibit a reduced receptor affinity as compared to human interferon gamma or Actimmune®.
  • the receptor affinity may be measured by any suitable assay and will be known to the person skilled in the art.
  • One example of a suitable assay for determining the receptor binding affinity is the BIAcore® assay described in Michiels et al. Int. J. Biochem. Cell Biol. 30:505-516 (1998).
  • interferon gamma polypeptides having reduced receptor affinity will exhibit a reduced interferon gamma activity, e.g. when tested in the "Primary Assay" described herein.
  • the interferon gamma polypeptide may exhibit 1- 95% of the interferon gamma activity of Actimunne® or human interferon gamma, e.g. 1-75%, such as 1-50%, e.g. 1-20% or 1-10% of the interferon gamma activity of Actimunne® or human interferon gamma in its glycosylated form.
  • any of the above-mentioned modifications giving rise to a reduced receptor binding affinity may be combined with any of the other modifications disclosed herein, in particular the modifications mentioned in the sections entitled “Interferon gamma variants with optimised N-glycosylation sites", “Interferon gamma variants comprising attachment groups for non-polypeptide moieties", “Inteferon gamma variants where the non-polypeptide moiety is a sugar moiety", “Interferon gamma variants where the non-polypeptide moiety is a molecule, which has cysteine as an attachment group” and "Interferon gamma variants where a first non-polypeptide moiety is a sugar moiety and a second non-polypeptide moiety is a molecule, which has cysteine as an attachment group", such as the modifications selected from the group consisting of E38N, S40T, S99T and combinations thereof, in particular E38N+S40T+S99T.
  • the interferon gamma polypeptide comprises at least one introduced glycosylation site, and at least one sugar moiety attached to an introduced glycosylation site. In a further embodiment the interferon gamma polypeptide comprises at least one introduced glycosylation site, and at least one sugar moiety attached to an introduced glycosylation site, and a polymer molecule, such as polyethylene glycol. In a particular preferred embodiment the interferon gamma polypeptide comprises 3 sugar moieties each sugar 5 moiety being attached to an N-glycosylation site.
  • Determination of C-terminal truncation of purified samples of interferon gamma polypeptides can be carried out in a number of ways. l o One way of elucidating C-terminal truncations of interferon gamma polypeptides relies on accurate mass determinations by mass spectrometry. Unfortunately, the glycosylation of interferon gamma is heterogeneous thus making it extremely difficult to dete ⁇ nine an accurate mass directly on the glycoprotein. Therefore, different levels of enzymatic deglycosylation are typically used in combination with mass spectrometry.
  • the entire glycan part of the interferon gamma polypeptide is cleaved of using the endo-glycosidase PNGase F followed by accurate mass determination using either ESI mass spectrometry or MALDI-TOF mass spectrometry. Comparing the experimental masses to the known amino acid sequence of interferon gamma makes it possible to determine the sites of C-terminal truncation.
  • sialic acid of the glycan part of the interferon gamma polypeptide is cleaved off instead of the entire glycan. In some cases this is sufficient to reduce the heterogeneity of the sample to a level where the sites of C- terminal truncations can be deduced following accurate mass determination using either ESI mass spectrometry or MALDI-TOF mass spectrometry.
  • interferon gamma polypeptides employs peptide mapping in combination with mass spectrometry and chemical amino acid sequencing.
  • the interferon gamma polypeptide is degraded with a protease of known specificity (e.g. Asp-N protease) followed by peptide separation using RP-HPLC. Fractions can then by mass analysed either on-line using a protease of known specificity (e.g. Asp-N protease) followed by peptide separation using RP-HPLC. Fractions can then by mass analysed either on-line using
  • the sulfoalkyl ether cyclodextrin derivative is any of the derivatives described in US 5,874,418, US 5,376,645 and US 5,134,127, the contents of which are incorporated herein by reference.
  • the sulfoalkyl ether cyclodextrin derivative is also described in WO 91/11172, the contents of which is incorporated herein by reference.
  • the sulfoalkyl ether cyclodextrin is a compound of the Formula (I):
  • n 4, 5 or 6
  • Ri, R 2 , R 3 , R 4 , R 5 , R 6 , R 7 , Rs, and R 9 are each, independently, -O- or a -0-(C 2 -C 6 alkylene)-S0 3 - group, wherein at least one of Ri and R 2 is independently a -0-(C 2 -C 6 alkylene)-S0 3 - group, and
  • Si, S 2 , S 3 , S , S 5 , S 6 , S 7 , Sg, and Sg are each, independently, a pharmaceutically acceptable cation.
  • n 5
  • n 6
  • At least one of Ri and R 2 is -0-(CH 2 ) m -S0 3 -, and m is 2, 3, 4, 5 or 6.
  • Rj and R 2 is independently selected from -OCH 2 CH 2 CH 2 S0 3 - or -OCH 2 CH 2 CH 2 CH S0 3 -.
  • At least one of ⁇ , R 6 , and R 8 is independently, -0-(C 2 -C 6 alkylene)-S0 3 -; and R 5 , R 7 , and R 9 are all -0-.
  • Si, S 2 , S 3 , S 4 , S 5 , S 6 , S 7 , S 8 , and S are each, independently, a pharmaceutically acceptable cation selected from H + , alkali metals (e.g. Li + , Na + , K + ), alkaline earth metals (e.g., Ca +2 , Mg +2 ), ammonium ions and amine cations such as the cations of (C ⁇ -C 6 ) alkylamines, piperidine, pyrazine, (Ci-C 6 ) alkanolamine and (C 4 -C 8 )cycloalkanolamine.
  • alkali metals e.g. Li + , Na + , K +
  • alkaline earth metals e.g., Ca +2 , Mg +2
  • ammonium ions and amine cations such as the cations of (C ⁇ -C 6 ) alkylamines, piperidine, pyr
  • Si, S 2 , S 3 , S 4 , S 5 , S 6 , S 7 , S 8 , and S are independently selected from alkaline metal cation, alkaline earth metal cation, quaternary ammonium cation, tertiary ammonium cation, and secondary ammonium cation.
  • at least one of P ., R 6 , and R 8 is independently, -0-(C 2 -
  • alkylene and alkyl as used herein (e.g., in the -0-(C 2 -C 6 - alkylene)S0 3 - group or in the alkylamines), include linear, cyclic, and branched, saturated and unsaturated (i.e., containing one double bond) divalent alkylene groups and monovalent alkyl groups, respectively.
  • alkanol in this text likewise includes both linear, cyclic and branched, saturated and unsaturated alkyl components of the alkanol groups, in which the hydroxyl groups may be situated at any position on the alkyl moiety.
  • cycloalkanol includes unsubstituted or substituted (e.g., by methyl or ethyl) cyclic alcohols.
  • the presently preferred sulfoalkyl ether cyclodextrin derivative is a salt of beta cyclodextrin sulfobutyl ether (in particular the sodium salt thereof also termed SBE7- ⁇ -CD which is available as Captisol®) (Cydex, Overland Park, Kansas 66213, US).
  • composition of the invention has a pH in the range of 3-8, such as 4-8, 5-
  • the preferred pH range is 4-8, preferably 5-8, or alternatively 4-7. In a further embodiment the pH range is 5-6, such about 5.5.
  • the pH is normally obtained by use of a suitable amount of a buffering agent as further described below.
  • the composition is about isotonic to blood, i.e. by having an osmolarity of about 240-360 mOsmol/kg, such as 280-320 mOsmol/kg, in particular about
  • the osmolarity is normally obtained by use of a suitable amount of tonicity agent as further described below.
  • the present invention relates to a stabilized composition
  • a stabilized composition comprising an interferon polypeptide and a sulfoalkyl ether cyclodextrin derivative.
  • the interferon polypeptide is typically selected from any one of those mentioned above and in the examples.
  • Preferred embodiments of the interferon polypeptide are interferon beta and interferon gamma, in particular interferon beta variants and interferon gamma variants, such as glycosylated and/or pegylated variants, as described in detail above.
  • the interferon polypeptide is typically present in a concentration of about 1-100 MlU/ml in a liquid preparation or about 1-100 MlU/dose in a solid preparation.
  • Such concentration may be selected from about 1-10 MlU/ml, 1-20 MlU/ml, 1-30 MlU/ml, 1-40 MlU/ml, 1-50 MlU/ml, 1-60 MlU/ml, 1-70 MlU/ml, 1-80 MlU/ml, 1-90 MlU/ml, 10-20 MlU/ml, 20-30 MlU/ml, 30-40 MlU/ml, 40-50 MlU/ml, 50-60 MlU/ml, 60-70 MlU/ml, 70-80 MlU/ml, 80-90 MlU/ml, 90-100 MlU/ml, 5-95 MlU/ml, 15-85 MlU/ml, 25-75 MlU/ml, 35-65 MlU/ml, and 45-55 MlU/ml in a liquid preparation.
  • such concentration may be selected from about 1-10 MlU/dose, 1- 20 MlU/dose, 1-30 MlU/dose, 1-40 MlU/dose, 1-50 MlU/dose, 1-60 MlU/dose, 1-70 MlU/dose, 1-80 MlU/dose, 1-90 MlU/dose, 10-20 MlU/dose, 20-30 MlU/dose, 30-40 MlU/dose, 40-50 MlU/dose, 50-60 MlU/dose, 60-70 MlU/dose, 70-80 MlU/dose, 80-90 MlU/dose, 90-100 MlU/dose, 5-95 MlU/dose, 15-85 MlU/dose, 25-75 MlU/dose, 35-65 MlU/dose, and 45-55 MlU/dose in a solid preparation.
  • the sulfoalkyl ether cyclodextrin derivative is typically present in a concentration of about 1-150 mg/ml. Such concentration may be selected from about 1-10 mg/ml, 1-20 mg/ml, 1- 30 mg/ml, 1-40 mg/ml, 1-50 mg/ml, 1-60 mg/ml, 1-70 mg/ml, 1-80 mg/ml, 1-90 mg/ml, 1-100 mg/ml, 1-110 mg/ml, 1-120 mg/ml, 1-130 mg/ml, 1-140 mg/ml, 10-20 mg/ml, 20-30 mg/ml, 30- 40 mg/ml, 40-50 mg/ml, 50-60 mg/ml, 60-70 mg/ml, 70-80 mg/ml, 80-90 mg/ml, 90-100 mg/ml, 100-110 mg/ml, 110-120 mg/ml, 120-130 mg/ml, 130-140 mg/ml, 140-150 mg/ml, 5- 100 mg/ml,
  • the composition of the invention has a pH in the range of 4-8, and an osmolarity of about 240-360 mOsmol/kg, such as 280-320 mOsmol/kg, in particular about 300 mOsmol/kg.
  • the composition of the invention relates to a liquid solution comprising an interferon polypeptide and a sulfoalkyl ether cyclodextrin derivative.
  • composition of the invention relates to an aqueous solution comprising an interferon polypeptide and a sulfoalkyl ether cyclodextrin derivative.
  • the sulfoalkyl ether cyclodextrin derivative is typically present in a concentration of about 1-150 mg/ml. However, any one of the concentration ranges mentioned above is considered an embodiment of the invention.
  • the interferon polypeptide is typically present in a concentration of about 1-100 MlU/ml in the liquid solution. However, any one of the concentration ranges mentioned above is considered an embodiment of the invention.
  • the liquid solution or aqueous solution is isotonic and has an osmolarity of about 240-360 mOsmol/kg.
  • the liquid solution or aqueous solution is isotonic and has an osmolarity of about 240-360 mOsmol kg, and has a pH in the range of 4-8, such as 5-8, 6-8, 7-8, 4-7, 4-6, or 4-5.
  • the liquid solution or aqueous solution further comprises a tonicity agent providing an osmolarity of about 240-360 mOsmol/kg.
  • the tonicity agent may be any suitable tonicity agent such as any one of those mentioned in the section "parenterals” below.
  • the liquid solution or aqueous solution further comprises a buffering agent present in a concentration up to 100 mM.
  • the concentration of buffering agent may be selected from any one of the concentration ranges mentioned in the section "parenterals” below.
  • the buffering agent may be any suitable buffer such as any one of those mentioned in the section "parenterals” below.
  • interferon polypeptide is typically selected from any one of those mentioned above and in the examples.
  • Preferred embodiments of the interferon polypeptide are interferon beta and interferon gamma, in particular any one of the interferon beta variants and interferon gamma variants, such as any one of the glycosylated and/or pegylated variants, as described in detail above.
  • Other prefened embodiments of the interferon polypeptide are human wild type interferon beta and human wild type interferon gamma, such as glycosylated and/or pegylated molecules, eg. glycosylated interferon beta- la coupled to a polymer, such as a polymer comprising a polyethylene glycol.
  • the composition of the invention is one, wherein the interferon polypeptide has essentially retained its bioactivity during storage at a temperature of 37°C for a period of at least 1 week, preferably at least 2, 3 or 4 weeks (as measured in an accelerated stability test).
  • the interferon polypeptide has essentially retained its bioactivity it means that at least 80% of its bioactivity is retained, preferably at least 90% of its bioactivity.
  • the interferon polypeptide has essentially retained its bioactivity during storage at a temperature of 25°C for at least 4 weeks, such as for at least 5, 6, 7, 8, 9, 10, 11 or 12 weeks.
  • the composition of the invention is one, wherein the interferon polypeptide has essentially retained its bioactivity during storage at a temperature of 37°C for a period of at least 1 week, and during storage at a temperature of 25°C for at least 4 weeks.
  • the bioactivity to be measured is e.g. the antiviral activity.
  • the antiviral activity is determined by a method known in the art, e.g. by use of the assays described in WO 01/15736 (Interferon beta) and WO 01/36001 (Interferon gamma).
  • the assay of WO 01/15736 is as follows:
  • the antiviral bioassay is performed using A549 cells (CCL 185, American tissue culture collection) and Encephalomyocarditis (EMC) virus (VR-129B, American tissue culture collection).
  • the cells are seeded in 96 well tissue culture plates at a concentration of 10,000 cells/well and incubated at 37°C in a 5% C0 2 air atmosphere.
  • a polypeptide or conjugate of the invention is added in concentrations ranging from about 100-0.0001 IU/mL (typically from 100-0.0001 IU/mL) in a total of lOO ⁇ l DMEM medium containing fetal calf serum and antibiotics. After 24 hours the medium is removed and 0.1 mL fresh medium containing EMC virus is added to each well.
  • the EMC virus is added in a concentration that causes 100% cell death in interferon-beta free cell cultures after 24 hours. After another 24 hrs, the antiviral effect of the polypeptide or conjugate is measured using the WST-1 assay.
  • 0.01 mL WST-1 WST-1 cell proliferation agent, Roche Diagnostics GmbH, Mannheim, Germany
  • the cleavage of the tetrazolium salt WST-1 by mitochondrial dehydrogenases in viable cells results in the formation of formazan that is quantified by measuring the absorbance at 450 nm.
  • ISRE Interferon Stimulated Response Element
  • HeLa cells are co-transfected with ISRE-Luc and pCDNA 3.1/hygro and foci (cell clones) are created by selection in DMEM media containing Hygromycin B. Cell clones are screened for luciferase activity in the presence or absence of interferon gamma. Those clones showing the highest ratio of stimulated to unstimulated luciferase activity are used in further assays.
  • muteins To screen muteins, 15,000 cells/well are seeded in 96 well culture plates and incubated overnight in DMEM media. The next day muteins as well as a known standard are added to the cells in various concentrations. The plates are incubated for 6 hours at 37°C in a 5% C0 2 air atmosphere LucLite substrate (Packard Bioscience, Groningen The Netherlands) is subsequently added to each well. Plates are sealed and luminescence measured on a TopCount luminometer (Packard) in SPC (single photon counting) mode. Each individual plate contains wells incubated with interferon gamma as a stimulated control and other wells containing normal media as an unstimulated control. The ratio between stimulated and unstimulated luciferase activity serves as an internal standard for both mutein activity and experiment-to-experiment variation.
  • composition of the invention may be in a variety of forms, including liquid, gel, lyophilized, pulmonary dispersion, or any other suitable form, e.g. as a compressed solid.
  • liquid is intended to include the term "aqueous”.
  • the composition is in the form of a dry or liquid formulation. In a further embodiment of the invention the composition is in dry form. In a further embodiment of the invention the composition is in liquid form. In a further embodiment of the invention the composition is an aqueous solution. In a further embodiment of the invention the composition is an aqueous suspension.
  • prefened form will depend upon the particular indication being treated and will be apparent to one of skill in the art. For instance, lyophilization from a solution may be used to further increase stability.
  • the pha ⁇ naceutical composition of the invention may be administered orally, intravenously, intracerebrally, intramuscularly, intraperitoneally, intradermally, subcutaneously, intranasally, pulmonary, or in any other acceptable manner, e.g. using drug delivery systems like PowderJect or ProLease technology.
  • the prefened mode of administration will depend upon the particular indication being treated and will be apparent to one of skill in the art.
  • compositions suitable for specific types of formulations are described. It will be understood that the nature and amount into which various additives are used depend on the interferon polypeptide as well as the type of formulation and adminstration route.
  • the composition of the invention comprises a buffering agent, a tonicity agent, a preservative, a wetting agent, a viscosity increasing agent, and/or one or more stabilizers in addition to the sulfoalkyl ether cyclodextrin derivative. It will be understood that such stabilizers must not adversely affect the stabilizing effects of the sulfoalkyl ether cyclodextrin derivative. Additional constituents of the composition are further described below.
  • composition of the invention may comprise human serum albumin or other human protein serving to stabilize the composition and/or minimizing adsorption to the container in which the composition is stored.
  • the composition is essentially free from human serum albumin or other human protein, since the presence of such proteins may be undesirable from a regulatory point of view.
  • composition of the invention may be prepared by a conventional method for preparing pharmaceutical compositions.
  • the composition is prepared by premixing the stabilizing and buffering agents, and any other additives prior to incorporation of the interferon polypeptide.
  • the composition is prepared with nitrogen purge of aqueous formulation and/or nitrogen purge of void volume of a partly filled product container.
  • the composition is prepared without such nitrogen purge.
  • the amount of interferon polypeptide present in the composition depends on the nature of the interferon polypeptide, of the formulation and of the administration route. For instance, when the composition is an interferon beta containing composition, the interferon beta polypeptide is present in an amount conesponding to 1-100 MlU/ml, typically 1-50 MlU/ml, (when formulated into a liquid formulation) or 1-100 MlU/dose (when formulated into a solid formulation).
  • compositions designed for parenteral administration.
  • parenteral formulations may also be provided in frozen or in lyophilized form.
  • the composition must be thawed prior to use.
  • the latter form is often used to enhance the stability of the active compound contained in the composition under a wider variety of storage conditions, as it is recognized by those skilled in the art that lyophilized preparations are generally more stable than their liquid counterparts.
  • Such lyophilized preparations are reconstituted prior to use by the addition of one or more suitable pharmaceutically acceptable diluents such as sterile water for injection or sterile physiological saline solution.
  • parenterals In case of parenterals, they are prepared for storage as lyophilized formulations or aqueous solutions by mixing, as appropriate, the interferon polypeptide having the desired degree of purity with one or more pharmaceutically acceptable carriers, excipients or stabilizers typically employed in the art (all of which are termed "excipients"), for example buffering agents, stabilizing agents, preservatives, tonicity agents, non-ionic detergents, antioxidants and/or other miscellaneous additives.
  • excipients typically employed in the art
  • the buffering agent is present in a concentration which ensures that the pH is kept at the desired level, e.g. a level which approximates physiological level.
  • the buffering agents have a suitable concentration up to 100 mM for each buffer type. For most buffering agents this concentration is normally in the range of 1-100 mM, such as 1-90 mM, 1-80 mM, 1-70 mM, 1- 60 mM, 1-50 mM, 1-40 mM, 1-30 mM, 1-20 mM, 1-10 mM, 5-15 mM, 15-25 mM, 25-35 mM, 35-45 mM, 45-55 mM, 55-65 mM, 65-75 mM, 75-85 mM, 85-95 mM.
  • the buffering agent is typically a solution of a weak acid, a weak base or a salt of the anion of such acid.
  • Suitable buffering agents for use with the present invention include both organic and inorganic acids and salts thereof such as citrate buffers (e.g., monosodium citrate-disodium citrate mixture, citric acid-trisodium citrate mixture, citric acid-monosodium citrate mixture, etc.), succinate buffers (e.g., succinic acid-monosodium succinate mixture, succinic acid-sodium hydroxide mixture, succinic acid-disodium succinate mixture, etc.), tartrate buffers (e.g., tartaric acid-sodium tartrate mixture, tartaric acid-potassium tartrate mixture, tartaric acid-sodium hydroxide mixture, etc.), fumarate buffers (e.g., fumaric acid-monosodium fumarate mixture, fumaric acid-d
  • phosphate buffers e.g., sodium carbonate
  • histidine buffers e.g., glutamate buffers.
  • Preservatives are added to retard microbial growth, and are typically added in amounts of about 0.2%-l% (w/v).
  • Suitable preservatives for use with the present invention include phenol, benzyl alcohol, meta-cresol, alkyl parabens, such as methyl paraben or propyl paraben, benzalkonium halides (e.g. benzalkonium chloride, bromide or iodide), catechol, resorcinol, 3- pentanol and appropriate mixtures thereof.
  • composition of the invention comprises a buffer, such as any one of the above, or mixtures thereof.
  • composition of the invention further comprises a preservating agent and/or a viscocity increasing agent.
  • composition of the invention is free from a preservating agent.
  • composition of the invention comprises a buffer selected from citrate buffers, succinate buffers, tartrate buffers, fumarate buffers, maleate buffers, gluconate buffers, oxalate buffer, phosphate buffers, carbonate buffers, histidine buffers, glutamate buffers, lactate buffers, and acetate buffers.
  • a buffer selected from citrate buffers, succinate buffers, tartrate buffers, fumarate buffers, maleate buffers, gluconate buffers, oxalate buffer, phosphate buffers, carbonate buffers, histidine buffers, glutamate buffers, lactate buffers, and acetate buffers.
  • the composition of the invention comprises a buffer which is present in a concentration up to 100 mM, such as 1 mM to 100 mM, 1-90 mM, 1-80 mM, 1-70 mM, 1-60 mM, 1-50 mM, 1-40 mM, 1-30 mM, 1-20 mM, 1-10 mM, 5-15 mM, 15-25 mM, 25- 35 mM, 35-45 mM, 45-55 M, 55-65 mM, 65-75 mM, 75-85 mM, or 85-95 mM.
  • a buffer which is present in a concentration up to 100 mM, such as 1 mM to 100 mM, 1-90 mM, 1-80 mM, 1-70 mM, 1-60 mM, 1-50 mM, 1-40 mM, 1-30 mM, 1-20 mM, 1-10 mM, 5-15 mM, 15-25 mM, 25- 35 mM, 35
  • Tonicity agents are added to ensure isotonicity of liquid compositions and include polyhydric sugar alcohols, preferably trihydric or higher sugar alcohols, such as glycerin, erythritol, arabitol, xylitol, sorbitol, and mannitol (mannitol is typically present in a concentration of up to 50 mg/ml), or NaCl (NaCl is typically present in a concentration of up to 9 mg/ml).
  • Polyhydric alcohols can be present in an amount between 0.1 % and 25% by weight, typically 1% to 5%, taking into account the relative amounts of the other ingredients.
  • Stabilizers refer to a broad category of excipients which can range in function from a bulking agent to an additive which solubilizes the therapeutic agent or helps to prevent denaturation or adherence to the container wall.
  • Typical stabilizers can be polyhydric sugar alcohols (enumerated above); amino acids such as arginine, lysine, glycine, glutamine, asparagine, histidine, alanine, ornithine, L-leucine, 2-phenylalanine, glutamic acid, threonine, etc., organic sugars or sugar alcohols, such as lactose, trehalose, stachyose, mannitol, sorbitol, xylitol, ribitol, myoinisitol, galactitol, glycerol and the like, including cyclitols such as inositol; polyethylene glycol; amino acid polymers; sulfur-containing reducing agents, such as
  • proteins such as human serum albumin, bovine serum albumin, gelatin or immunoglobulins
  • hydrophilic polymers such as polyvinylpyrrolidone
  • monosaccharides such as xylose, mannose, fructose and glucose
  • disaccharides such as lactose, maltose and sucrose
  • trisaccharides such as raffinose, and polysaccharides such as dextran.
  • Stabilizers are typically present in the range of from 0.1 to 10,000 parts by weight based on the active protein weight.
  • Non-ionic surfactants or detergents may be present to help solubilize the therapeutic agent as well as to protect the therapeutic polypeptide against agitation-induced aggregation, which also permits the formulation to be exposed to shear surface stress without causing denaturation of the polypeptide.
  • Suitable non-ionic surfactants include polysorbates (20, 80, etc.), polyoxamers (184, 188 etc.), Pluronic® polyols, polyoxyethylene sorbitan monoethers (Tween®-20 (Tween-20 is typically present in a concentration of up to 2 mg/ml), Tween®-80 (Tween-80 is typically present in a concentration of up to 2 mg/ml), etc.).
  • no surfactant such as non-ionic surfactant
  • the composition comprises an interferon beta polypeptide.
  • Additional miscellaneous excipients include bulking agents or fillers (e.g. starch), chelating agents (e.g. EDTA), antioxidants (e.g., ascorbic acid, methionine, vitamin E) and cosolvents.
  • the active ingredient may also be entrapped in microcapsules prepared, for example, by coascervation techniques or by interfacial polymerization, for example hydroxymethylcellulose, gelatin or poly-(methylmethacylate) microcapsules, in colloidal drug delivery systems (for example liposomes, albumin microspheres, microemulsions, nano- particles and nanocapsules) or in sustained release preparations (see next section).
  • colloidal drug delivery systems for example liposomes, albumin microspheres, microemulsions, nano- particles and nanocapsules
  • sustained release preparations see next section.
  • Such techniques are disclosed in Remington's Pharmaceutical Sciences, by E.W.Martin, 18 edition, A. R. Gennaro, Ed., Mack Publishing Company [1990]; Pharmaceutical Formulation Development of Peptides and Proteins.
  • Parenteral formulations to be used for in vivo administration must be sterile. This is readily accomplished, for example, by filtration through sterile filtration membranes.
  • sustained-release preparations include semi-permeable matrices of solid hydrophobic polymers and/or hydrophilic polymers containing the interferon polypeptide, the matrices having a suitable form such as a film, a rod, a microcapsule or a microsphere.
  • sustained-release matrices include polyesters, hydrogels (for example, poly(2- hydroxyethyl-methacrylate) or poly(vinylalcohol)), polylactides, copolymers of L-glutamic acid and ethyl-L-glutamate, non-degradable ethylene- vinyl acetate, dextrans, starch, degradable lactic acid-glycolic acid copolymers such as the ProLease® technology or Lupron Depot® (injectable microspheres composed of lactic acid-glycolic acid copolymer and leuprolide acetate), and poly-D-(-)-3-hydroxybutyric acid. While polymers such as ethylene-vinyl acetate and lactic acid-glycolic acid enable release of molecules for long periods such as up to or over 100 days, certain hydrogels release proteins for shorter time periods.
  • polymers such as ethylene-vinyl acetate and lactic acid-glycolic acid enable release of molecules for long periods such
  • Such formulations may be in the form of liquid or solid formulations to be used as is or in the form of a dispersion. All relevant devices for administration and/or generation of a dispersion require the use of formulations suitable for dispensing the interferon polypeptide. Typically, each formulation is specific to the type of device employed and may involve the use of an appropriate propellant material, in addition to the usual diluents, adjuvants and/or carriers useful in therapy. Each formulation is also typically specific to the interferon polypeptide to be delivered as a dispersion. Also, the use of liposomes, microcapsules or microspheres, inclusion complexes, or other types of carriers is contemplated. Formulations of the interferon polypeptide which can be utilized in the most common types of pulmonary dispensing devices to practice this invention are now described.
  • Interferon polypeptide formulations suitable for use with a nebulizer, either jet or ultrasonic will typically comprise the interferon polypeptide dissolved in water at a concentration of, e.g., about 0.01 to 25 mg of interferon polypeptide per ml of solution, preferably about 0.1 to 10 mg/ml.
  • the formulation may also include a buffer and a tonicity agent, e.g. a sugar for protein stabilization and regulation of osmotic pressure, and/or human serum albumin ranging in concentration from 0.1 to 10 mg/ml.
  • buffers which may be used are sodium acetate, citrate and glycine.
  • the buffer will have a composition and molarity suitable to adjust the solution to a pH in the range of 3 to 9.
  • buffer molarities of from 1 mM to 50 mM are suitable for this purpose.
  • sugars which can be utilized are lactose, maltose, mannitol, sorbitol, trehalose, and xylose, usually in amounts ranging from 1% to 10% by weight of the formulation.
  • the nebulizer formulation may also contain a surfactant to reduce or prevent surface induced aggregation of the protein caused by atomization of the solution in forming the aerosol.
  • a surfactant to reduce or prevent surface induced aggregation of the protein caused by atomization of the solution in forming the aerosol.
  • Various conventional surfactants can be employed, such as polyoxyethylene fatty acid esters and alcohols, and polyoxyethylene sorbitan fatty acid esters. Amounts will generally range between 0.001% and 4% by weight of the formulation.
  • An especially prefened surfactant for purposes ofthis invention is polyoxyethylene sorbitan monooleate.
  • nebulizers suitable for the practice of this invention are the Ultravent nebulizer, manufactured by Mallinckrodt, Inc., St. Louis, Mo., the Acorn II nebulizer, manufactured by Marquest Medical Products, Englewood, Colorado, and the AERx pulmonary drug delivery system manufactured by Aradigm Corporation, Hayward, California.
  • Interferon formulations for use with a metered dose inhaler device will generally comprise a finely divided powder of relevant shape, surface and size.
  • This powder may be produced by lyophilizing and, if needed, then milling a solid interferon formulation and may also contain a stabilizer such as human serum albumin (HSA). Typically, more than 0.5%) (w/w) HSA is added.
  • HSA human serum albumin
  • one or more sugars or sugar alcohols may be added to the preparation if necessary. Examples include lactose maltose, mannitol, sorbitol, sorbitose, trehalose, xylitol, and xylose.
  • the amount added to the formulation can range from about 0.01 up to 100% (w/w), preferably from approximately 1 to 50%.
  • Such formulations are then lyophilized and milled to the desired particle size.
  • the properly sized particles are then suspended in a propellant with the aid of a surfactant.
  • the propellant may be any conventional material employed for this purpose, such as a chlorofluorocarbon, a hydrochlorofluorocarbon, a hydrofluorocarbon, or a hydrocarbon, including trichlorofluoromethane, dichlorodifluoromethane, dichlorotetrafluoroethanol, and 1,1,1,2-tetrafluoroethane, or combinations thereof.
  • Suitable surfactants include sorbitan trioleate and soya lecithin. Oleic acid may also be useful as a surfactant. This mixture is then loaded into the delivery device.
  • An example of a commercially available metered dose inhaler suitable for use in the present invention is the Ventolin metered dose inhaler, manufactured by Glaxo Inc., Research Triangle Park, N.C.
  • Such interferon formulations for powder inhalers will comprise a finely divided dry powder containing conjugate and may also include a bulking agent, such as lactose, sorbitol, sucrose, or mannitol in amounts which facilitate dispersal of the powder from the device, e.g., 50% to 90%) by weight of the formulation.
  • the particles of the powder shall have aerodynamic properties in the lung. This typically conesponds to particles with a density of about 1 g/cm having a median diameter less than 10 micrometers, preferably between 0.5 and 5 micrometers, most preferably of between 1.5 and 3.5 micrometers.
  • powder inhaler suitable for use in accordance with the teachings herein is the Spinhaler powder inhaler, manufactured by Fisons Corp., Bedford, Mass.
  • the powders for these devices may be generated and/or delivered by methods disclosed in US 5997848, US 5993783, US 5985248, US 5976574, US 5922354, US 5785049 and US 55654007 which are hereby incorporated by reference.
  • the pharmaceutical composition containing the interferon polypeptide may be administered by a wide range of mechanical devices designed for pulmonary delivery of therapeutic products, including but limited to nebulizers, metered dose inhalers, and powder inhalers, all of which are familiar to those of skill in the art.
  • nebulizers include but limited to nebulizers, metered dose inhalers, and powder inhalers, all of which are familiar to those of skill in the art.
  • Some specific examples of commercially available devices suitable for the practice of this invention are the Ultravent nebulizer, manufactured by Mallinckrodt, Inc., St. Louis,
  • the present invention also provides for a primary product container comprising a composition of the invention.
  • the container may be any type of container suited for the composition in question, e.g. made from stainless steel or glass.
  • the container may be a syringe, such as a prefilled syringe.
  • kits for parental administration of a liquid interferon composition according to the invention may comprise the interferon composition as the only pharmaceutically active ingredient, or may comprise one or more further pharmaceutically active ingredients - either in the same container or a separate container.
  • the invention relates to a method for increasing stability of an interferon polypeptide formulated into a pharmaceutical composition, said method comprising incorporating into said composition a sulfoalkyl ether cyclodextrin derivative and a buffering agent.
  • the method is of particular relevance when the interferon polypeptide exhibits aggregate formation during storage and the sulfoalkyl ether cyclodextrin derivative is incorporated in an amount sufficient to reduce aggregate formation of the interferon polypeptide.
  • the interferon polypeptide is preferably any of those described herein.
  • the sulfoalkyl ether cyclodextrin derivative is preferably any of those described in US 5,376,645, US 5,874,418 or US 5,134,127, in particular a salt of beta cyclodextrin sulfobutyl ether, e.g. the sodium salt available as Captisol®.
  • the invention relates to a method of subjecting a mammal to interferon therapy, which method comprises administrering a therapeutically effective amount of a composition according to the invention. Also, the invention relates to a composition of the invention for use in treatment of diseases as well as to the use of a composition of the invention for the manufacture of a medicament for treatment of diseases. It is clear that when a composition of the invention should be used as a medicine in the treatment of diseases or disorders it is a pharmaceutical composition.
  • the pharmaceutical composition of the invention may be administered in conjunction with other therapeutic agents. These agents maybe incorporated as part of the same pharmaceutical composition or may be administered separately from the interferon polypeptide either concunently or in accordance with any other acceptable treatment schedule. In addition, the pharmaceutical composition of the invention may be used as an adjunct to other therapies.
  • this invention provides compositions and methods for treating most types of viral infections, cancers or tumors or tumour angiogenesis, Chrohn's disease, ulcerative colitis, Guillain-Bane syndrome, glioma, idiopathic pulmonary fibrosis, abnormal cell growth, or for immunomodulation in any suitable animal, preferably mammal, and in particular human.
  • the composition of the invention may be used in the treatment of osteosarcoma, basal cell carcinoma, ovarian carcinoma, cervical dysplasia, cervical carcinoma, laryngeal papillomatosis, mycosis fungoides, glioma, acute myeloid leukemia, multiple myeloma, Hodgk s disease, melanoma, breast carcinoma, non-small cell lung cancer, malignant melanoma (adjuvant, late stage, as well as prophylactic), carcinoid tumour, B-cell lymphoma, T-cell lymphoma, follicular lymphoma, Kaposi's sarcoma, chronic myelogenous leukaemia, renal cell carcinoma, recunent superfiecial bladder cancer, colorectal carcinoma, hairy cell leukaemia, and viral infections such as papilloma virus, viral hepatitis, herpes genitalis, herpes zoster, herpetic keratitis, herpe
  • the composition may be used for CML monotherapy or in combination with cytarabne, for B-cell lymphoma monotherapy or in combination with doxorubicin-based regimens, for foUicular lymphoma therapy as an adjunct to CHOP -like regimen, for hepatitis C monotherapy or in combination with ribavirin, for multiple myeloma monotherapy or in combination with VBMCP, BCNU or VBMCP + HiCy, or for renal carcinoma monotherapy or in combination with Vinblastine, floxuridine, 5-fluoruouracil or IL- 10.
  • polypeptide or composition of the invention may be used for the treatment of multiple sclerosis (MS), such as any of the generally recognized four types of MS (benign, relapsing remitting MS (RRMS), primary progressive MS (PPMS) and secondary progressive MS (SPMS)) and for monosymptomatic MS), cancer or tumours, hepatitis, e.g. hepatitis B and hepatitis C, or a he ⁇ es infection (the latter treatment optionally being combined with a treatment with IL-10).
  • MS multiple sclerosis
  • RRMS relapsing remitting MS
  • PPMS primary progressive MS
  • SPMS secondary progressive MS
  • cancer or tumours cancer or tumours
  • hepatitis e.g. hepatitis B and hepatitis C
  • a he ⁇ es infection optionally being combined with a treatment with IL-10.
  • the present invention also relates to use of a composition comprising an interferon beta polypeptide and a sulfoalkyl ether cyclodextrin derivative for the manufacture of a medicament for treatment of viral infections, cancers or tumors or tumour angiogenesis, Crohn's disease, ulcerative colitis, Guillain-Bane syndrome, glioma, idiopathic pulmonary fibrosis, abnormal cell growth, or for immunomodulation in any suitable animal, preferably mammal, and in particular human.
  • composition of the invention comprising an interferon beta polypeptide and a sulfoalkyl ether cyclodextrin derivative may be used in the treatment of osteosarcoma, basal cell carcinoma, ovarian carcinoma, cervical dysplasia, cervical carcinoma, laryngeal papillomatosis, mycosis fungoides, glioma, acute myeloid leukemia, multiple myeloma, Hodgkh s disease, melanoma, breast carcinoma, non-small cell lung cancer, malignant melanoma (adjuvant, late stage, as well as prophylactic), carcinoid tumour, B-cell lymphoma, T-cell lymphoma, foUicular lymphoma, Kaposi's sarcoma, chronic myelogenous leukaemia, renal cell carcinoma, recunent superfiecial bladder cancer, colorectal carcinoma, hairy cell leukaemia, and viral infections such as papilloma virus, viral he
  • the present invention relates to use of a composition comprising an interferon beta polypeptide and a sulfoalkyl ether cyclodextrin derivative, wherein the interferon beta polypeptide comprises the substitutions C17S+Q49N+Q51T+D110F+F11 IN+Rl 13T relative to the wildtype human interferon beta shown in SEQ ID NO 1, and has 3 sugar moieties attached to 3 N-glycosylation sites and 1 PEG molecule having a molecular weight of about 20 kDa (a particular example of such an interferon beta polypeptide is eg.
  • the interferon beta variant of example 3 pegylated with a 20kDa-PEG for the manufacture of a medicament for treatment of a cancer selected from any one of osteosarcoma, basal cell carcinoma, ovarian carcinoma, cervical dysplasia, cervical carcinoma, laryngeal papillomatosis, mycosis fungoides, glioma, acute myeloid leukemia, multiple myeloma, Hodgku s disease, melanoma, breast carcinoma, non-small cell lung cancer, malignant melanoma (adjuvant, late stage, as well as prophylactic), carcinoid tumour, B-cell lymphoma, T-cell lymphoma, foUicular lymphoma, Kaposi's sarcoma, chronic myelogenous leukaemia, renal cell carcinoma, recu ⁇ ent superfiecial bladder cancer, colorectal carcinoma, and hairy cell leukaemia.
  • a cancer selected from any one of osteosarcoma, bas
  • the cancer is melanoma.
  • the cancer is breast carcinoma.
  • the cancer is non-small cell lung cancer.
  • the cancer is malignant melanoma.
  • the present invention also relates to use of a composition comprising an interferon alfa polypeptide and a sulfoalkyl ether cyclodextrin derivative for the manufacture of a medicament for treatment of viral infections, cancers or tumors or tumour angiogenesis, Crohn's disease, ulcerative colitis, Guillain-Ba ⁇ e syndrome, glioma, idiopathic pulmonary fibrosis, abnormal cell growth, or for immunomodulation in any suitable animal, preferably mammal, and in particular human.
  • composition of the invention comprising an interferon alfa polypeptide and a sulfoalkyl ether cyclodextrin derivative may be used in the treatment of osteosarcoma, basal cell carcinoma, ovarian carcinoma, cervical dysplasia, cervical carcinoma, laryngeal papillomatosis, mycosis fungoides, glioma, acute myeloid leukemia, multiple myeloma, Hodgkkfs disease, melanoma, breast carcinoma, non-small cell lung cancer, malignant melanoma (adjuvant, late stage, as well as prophylactic), carcinoid tumour, B-cell lymphoma, T-cell lymphoma, foUicular lymphoma, Kaposi's sarcoma, chronic myelogenous leukaemia, renal cell carcinoma, recurrent superfiecial bladder cancer, colorectal carcinoma, hairy cell leukaemia, and viral infections such as papilloma virus, viral
  • the invention when the composition comprises an interferon beta polypeptide the invention relates to a method of treating a mammal having circulating antibodies against interferon beta la, such as AvonexTM or Rebif®, or lb, such as Betaseron®, which method comprises admimstering a composition of the invention comprising an interferon beta polypeptide which has a reduced or no reaction with said antibodies.
  • the compound is administered in an effective amount.
  • the mammal is preferably a human being.
  • the mammals to be treated may suffer from any of the diseases listed above for which interferon ⁇ is a useful treatment.
  • this aspect of the invention is of interest for the treatment of multiple sclerosis (any of the types listed above), hepatitis or cancer.
  • the term "circulating antibodies” is intended to indicate antibodies, in particular neutralizing antibodies, formed in a mammal in response to having been treated with any of the commercially available interferon beta preparations (Rebif, Betaseron, Avonex).
  • the composition of the invention may be used for treatment of any of the medical indications described in WO 01/36001, in particular interstitial pulmonary diseases, most particularly idiopathic pulmonary fibrosis.
  • Interferon gamma has been suggested for treatment of interstitial lung diseases (also known as Interstitial Pulmonary Fibrosis (IPF) (Ziesche et al. (N. Engl. J. Med. 341:1264-1269, 1999 and Chest 110:Suppl:25S, 1996) and EP 795332) for which pu ⁇ ose interferon gamma can be used in combination with prednisolone.
  • IPF Interstitial Pulmonary Fibrosis
  • the present invention also relates to use of a composition comprising an interferon gamma polypeptide and a sulfoalkyl ether cyclodextrin derivative for the manufacture of a medicament for treatment of interstitial pulmonary diseases, most particularly idiopathic pulmonary fibrosis, interstitial lung diseases (also known as Interstitial Pulmonary Fibrosis (IPF)), granulomatous diseases, mycobacterial infections, kidney cancer, osteopetrosis, scleroderma, hepatitis B, hepatitis C, septic shock, and rheumatoid arthritis.
  • interstitial pulmonary diseases most particularly idiopathic pulmonary fibrosis, interstitial lung diseases (also known as Interstitial Pulmonary Fibrosis (IPF)), granulomatous diseases, mycobacterial infections, kidney cancer, osteopetrosis, scleroderma, hepatitis B, hepatitis C, septic shock, and rhe
  • CHO-K1 cells available from American Type Culture Collection (ATCC #CCL-61)).
  • HeLa cells available from American Type Culture Collection (ATCC #CCL-2)). ISRE-Luc was obtained fromStratagene, La Jolla USA. pCDNA 3.1/hygro was obtained from Invitrogen, Carlsbad USA.
  • Restricion enzymes and polymerases were obtained from New England Biolabs Inc.,
  • DMEM medium Dulbecco's Modified Eagle Media (DMEM), 10% fetal bovine serum and Hygromycin B were obtained from Life Technologies A S, Copenhagen, Denmark.
  • LucLite substrate was obtained from Packard Bioscience, Groningen, The Netherlands.
  • TopCount luminometer was obtained from Packard Bioscience, Groningen, The
  • Biotinylated polyclonal anti-human interferon gamma antibody, BAF285, was obtained available from R&D Systems Inc., Minneapolis, USA.
  • TMB blotting reagent was obtained from KEM-EN-TEC, Copenhagen, Denmark.
  • ISRE Interferon Stimulated Response Element
  • HeLa cells are co-transfected with ISRE-Luc and pCDNA 3.1/hygro and foci (cell clones) are created by selection in DMEM media containing Hygromycin B. Cell clones are screened for luciferase activity in the presence or absence of interferon gamma. Those clones showing the highest ratio of stimulated to unstimulated luciferase activity are used in further assays.
  • Each individual plate contains wells incubated with interferon gamma as a stimulated control and other wells containing normal media as an unstimulated control.
  • the ratio between stimulated and unstimulated luciferase activity serves as an internal standard for both interferon gamma activity and experiment-to-experiment variation.
  • ASA Accessible Surface Area
  • the fractional ASA of the side chain atoms is computed by division of the sum of the ASA of the atoms in the side chain with a value representing the ASA of the side chain atoms of that residue type in an extended ALA-x-ALA tripeptide. See Hubbard, Campbell & Thornton (1991) J.Mol.Biol.: 220,507-530.
  • the CA atom is regarded as a part of the side chain of Glycine residues but not for the remaining residues.
  • the following table are used as standard 100% ASA for the side chain:
  • Residues not detected in the structure are defined as having 100% exposure as they are thought to reside in flexible regions.
  • the distance between atoms was determined using molecular graphics software e.g. Insightl! v. 98.0, MSI INC. Determination of receptor binding site:
  • the receptor-binding site is defined as comprising of all residues having their accessible surface area changed upon receptor binding. This is determined by at least two ASA calculations; one on the isolated ligand(s) in the ligand(s)/receptor(s) complex and one on the complete ligand(s)/receptor(s) complex.
  • the X-ray structure used was of an interferon gamma homo-dimer in complex with two molecules of a soluble form of the interferon gamma receptor having a third molecule of the interferon gamma receptor in the structure not making interactions with the interferon gamma homodimer reported by Thiel et.al. Structure 8:927-936 (2000).
  • the structure consists of the interferon gamma homodimer wherein the two molecules are labeled A and B.
  • the M0 is removed from the structure in all the calculations ofthis example.
  • the structure of the two interferon gamma monomers has very weak electron density after residue 120 and residues were only modeled until residue T126. Therefore, residues S121-T126 were removed from the structure prior to the calculations in this example.
  • the two receptor fragments labeled C and D make direct interactions with the interferon gamma homodimer and a third receptor molecule labeled E makes no contact with the interferon gamma homodimer and are not included in these calculations.
  • Residues not determined in the structure are treated as fully surface exposed, i.e. residues S121, P122, A123, A124, K125, T126, G127, K128, R129, K130, R131, S132, Q133, M134, L135, F136, R137, G138, R139, R140, A141, S142, Q143.
  • residues S121, P122, A123, A124, K125, T126, G127, K128, R129, K130, R131, S132, Q133, M134, L135, F136, R137, G138, R139, R140, A141, S142, Q143 are also constitute separate targets for introduction of attachment groups (or may be viewed as belonging to the group of surface exposed amino acid residues, e.g. having more than 25% or more than 50% exposed side chains).
  • GenBank accession number X13274 encompassing a full length cDNA encoding mature human interferon gamma with its native signal peptide, was modified in order to facilitate high expression in CHO cells. Codons of the human interferon gamma nucleotide sequence were modified by making a bias in the codon usage towards the codons frequently used in homo sapiens. Subsequently, certain nucleotides in the sequence were substituted with others in order to introduce recognition sites for DNA restriction endonucleases. Primers were designed such that the gene could be synthesised.
  • the primers were assembled to the synthetic gene by one step PCR using Platinum P/x-polymerase kit (Life Technologies) and standard three-step PCR cycling parameters.
  • the assembled gene was amplified by PCR using the same conditions and has the sequence shown in SEQ ID NO: 5.
  • the synthesised gene was cloned into pcDNA3.1/hygro (InVitrogen) between the BamHI at the 5' end and the Xbal at the 3' end, resulting in pIGY-22.
  • oligonucleotides were designed in such a way that PCR-generated changes could be introduced in the expression plasmid
  • ADJ013 5'-GATGGCTGGCAACTAGAAG-3' (SEQ ID NO: 6), (antisense downstream vector primer) and ADJ014: 5'-TGTACGGTGGGAGGTCTAT-3' (SEQ ID NO: 6), (antisense downstream vector primer) and ADJ014: 5'-TGTACGGTGGGAGGTCTAT-3' (SEQ ID NO: 6), (antisense downstream vector primer) and ADJ014: 5'-TGTACGGTGGGAGGTCTAT-3' (SEQ ID NO: 6), (antisense downstream vector primer) and ADJ014: 5'-TGTACGGTGGGAGGTCTAT-3' (SEQ ID NO: 6), (antisense downstream vector primer) and ADJ014: 5'-TGTACGGTGGGAGGTCTAT-3' (SEQ ID NO: 6), (antisense downstream vector primer) and ADJ014: 5'-TGTACGGTGGGAGGTCTAT-3' (SEQ ID NO: 6
  • the S99T variant was generated by classical two-step PCR, using ADJ013 and
  • ADJ014 as vector primers ADJ093 (5'-GTTCAGGTCTGTCACGGTGTAATTGG-
  • the E38N+S40T+S99T variant was generated by classical two-step PCR, using ADJ013 and ADJ014 as vector primers, ADJ091 (5'-CATGATCTTCCGATCGGTCTC- GTTCTTCCAATT-3') (SEQ ID NO: 10), and ADJ092 (5'-AATTGGAAGAACGA- GACCGATCGGAAGATCATG-3') (SEQ ID NO: 11), as mutation primers, and pIGY- 48 as template.
  • the 447 bp PCR product was subcloned into pcDNA3.1/Hygro (InVitrogen) using BamHI and Xbal, leading to plasmid ⁇ IGY-54.
  • pIGY-54 was transfected into CHO KI cells by use of Lipofectaim2000 (Life Technologies) as transfection agent. 24 hours later the culture medium was harvested and assayed for interferon gamma activity. Using the Primary assay described herein, an activity of 1.3 x 10 7 AU/ml was obtained.
  • C-terminally truncated interferon gamma variants containing a stop codon immediately downstream of the codon for Leul35, were generated by one-step PCR using pIGY-22, pIGY-48 and pIGY-54 as templates, followed by subcloning of the PCR products into pcDNA3.1/Hygro (InVitrogen) using BamHI and Xbal.
  • the primers used for construction of these variants were: ADJ014 (see above, upstream) and: 5'- GAGTCTAGATTACAGCATCTGGCTTCTCTT-3' (downstream).
  • pIGY-72 wild-type interferon gamma truncated after Leul35
  • pIGY-73 wild-type interferon gamma truncated after Leul35
  • pIGY-74 E38N+S40T+ S99T truncated after Leu 135).
  • Interferon gamma variants containing cysteine residues were generated using Stratagene' s QuikChangeTMXL site-directed mutagenesis kit, according to the manufacturer's specifications. Seven interferon gamma variants, each containing one introduced cysteine, were generated using pIGY-48 as template: N10C+S99T, N16C+S99T, E38C+S99T, N59C+S99T, N83C+S99T, K94C+S99T and S99T+N104C.
  • interferon gamma variants each containing one introduced cysteine, were generated using pIGY-54 as template: N10C+E38N+S40T+S99T, N16C+E38N+S40T+S99T, E38N+S40T+ N59C+S99T, E38N+S40T+ N83C+S99T, E38N+S40T+ K94C+S99T and E38N+S40T+S99T+N104C.
  • Example E PEGylation of cvsteine-containing variants All buffers were de-oxidized prior to use. Protein concentrations were estimated by measuring A280.
  • the PEGylated interferon gamma variant was diafiltered into 5 mM sodium succinate, 4% mannitol, pH 6.0 using a Vivaspin 6 column (VivaScience) and Tween 20 was added to 0.01%.
  • the purified PEGylated interferon gamma variant had a specific activity of 1.3 x 10 6 AU/mg as measured in the Primary Assay described herein (15%) of the specific activity of the conesponding non-PEGylated interferon gamma variant).
  • Ammonium sulphate was added to a concentration of 0.9 M and the PEGylated interferon gamma variant was applied onto a 1 ml ResourceTM phenyl column (Pharmacia) equilibrated in buffer B (20 mM sodium phosphate, 0.9 M ammonium sulphate, pH 6.6). The column was washed with 5 column volumes of buffer B before elution of the bound PEGylated interferon gamma variant in a linear gradient from 0- 50% buffer C (20 mM sodium phosphate, pH 6.6) over 30 column volumes. The PEGylated interferon gamma variant eluted around 0.6 M ammonium sulphate.
  • Fractions containing PEGylated interferon gamma variant were pooled and diafiltered into 5 mM sodium succinate, 4% mannitol, pH 6.0 using a Vivaspin 6 column (VivaScience) and Tween 20 was added to 0.01 %>.
  • the purified PEGylated interferon gamma variant had a specific activity of 2.4 x 10 6 AU/mg as measured in the Primary Assay described herein (15% of the specific activity of the conesponding non- PEGylated interferon gamma variant).
  • Example F Expression of interferon gamma polypeptides in mammalian cells
  • cells were grown to 95% confluency in media (Dulbecco's MEM/Nut. -mix F-12 (Ham) L-glutamine, 15 mM Hepes, pyridoxine-HCl (Life Technologies Cat # 31330-038)) containing 1:10 fetal bovine serum (BioWhittaker Cat # 02-70 IF) and 1:100 penicillin and streptomycin (BioWhittaker Cat # 17-602E).
  • interferon gamma-encoding plasmids were transfected into the cells using Lipofectamine 2000 (Life Technologies) according to the manufacturer's specifications. 24 hrs after transfection, culture media were collected and assayed for interferon gamma activity. Furthermore, in order to quantify the relative number of glycosylation sites utilized, Western blotting was performed using harvested culture medium.
  • Stable clones expressing interferon gamma were generated by transfection of CHO KI cells with interferon gamma-encoding plasmids followed by incubation of the cells in media containing 0.36 mg/ml hygromycin. Stably transfected cells were isolated and sub-cloned by limited dilution. Clones producing high levels of interferon gamma were identified by ELISA.
  • Example G Large-scale production
  • Stable cell lines expressing interferon gamma or variants were grown in Dulbecco's MEM/Nut. -mix F-12 (Ham) L-glutamine, 15 mM Hepes, pyridoxine-HCl (Life Technologies Cat # 31330-038), 1:10 fetal bovine serum (BioWhittaker Cat # 02- 701F), 1:100 penicillin and streptomycin (BioWhittaker Cat# 17-602E) in 1700 cm2 roller bottles (Corning, # 431200) until confluence. The media was then changed to 300 ml UltraCHO with L-glutamine (BioWhittaker Cat # 12-724Q) with the addition of 1:500 EX-CYTE VLE (Serological Proteins Inc.
  • the filtrate was microfiltrated (0.22 ⁇ m) before ultrafiltration to approximately 1/15 volume using a Millipore TFF system.
  • the concentrate was diafiltrated using 10 mM Tris, pH 7.6.
  • Ammonium sulphate was added to a concentration of 1.7 M and after stirring the precipitate was removed by centrifugation at 8000 m for 25 minutes in a Sorvall centrifuge using a GS3 rotor.
  • the supernatant was applied onto a 25 ml Phenyl High Performance (Pharmacia) column previously equilibrated in 10 mM Tris, 1.7 M ammonium sulphate, pH 7.6. After application the column was washed with 3 column volumes of 10 mM Tris, 1.7 M ammonium sulphate, pH 7.6 and the bound interferon gamma variant was then eluted in a linear gradient over 10 column volumes to 100%> 10 mM Tris, pH 7.6. The flow- through as well as the eluted interferon gamma variant was fractionated.
  • Fractions enriched in the interferon gamma variant were pooled and buffer exchanged by diafilfration into 10 mM Tris, pH 9.0, using a Vivaflow200 system (VivaScience) with a molecular weight cut-off of 10,000 Da.
  • the interferon gamma variant was then applied onto a 18 ml Q-sepharose Fast Flow (Pharmacia) column previously equilibrated in 10 mM Tris, pH 9.0. After application the column was washed with 3 column volumes of 10 mM Tris, pH 9.0 before eluting the bound interferon gamma variant in a gradient from 0-100%) 10 mM Tris, 0.5 MNaCl, pH 9.0, over 15 column volumes. The flow-through as well as the eluted interferon gamma variant was fractionated.
  • Fractions enriched in the interferon gamma variant were pooled and buffer exchanged into 10 mM sodium phosphate, pH 7.0, by diafiltration using a Vivaspin20 (VivaScience) column with a molecular weight cut-off of 10,000 Da.
  • the interferon gamma variant was applied onto an 8 ml CHT ceramic hydroxyapatite column (Biorad) previously equilibrated in 10 mM sodium phosphate, pH 7.0. After application the column was washed with 5 column volumes of 10 mM sodium phosphate, pH 7.0, before elution of the bound interferon gamma variant in a gradient from 0-60%> 500 mM sodium phosphate, pH 7.0, over 30 column volumes. The flow-through as well as the eluted interferon gamma variant was fractionated.
  • interferon gamma variants may be purified according to the below purification scheme:
  • the filtrate is microfiltrated (0.22 ⁇ m) before ultrafiltration to approximately 1/15 volume using a Millipore TFF system.
  • the concentrate is diafitrated using 10 mM Tris, pH 7.6, after which pH is adjusted to 9.0 and precipitate is removed by microfiltration.
  • the sample is applied onto a Q-sepharose Fast Flow (Pharmacia) column previously equilibrated in 10 mM Tris, pH 9.0. After application the column is washed with 3 column volumes of 10 mM Tris, pH 9.0 before eluting the bound interferon gamma variant in a gradient from 0-100%> 10 mM Tris, 0.5 M NaCl, pH 9.0 over 15 column volumes.
  • the flow-through as well as the eluted interferon gamma variant is fractionated. Fractions enriched in the interferon gamma variant are pooled, and pH is adjusted to 7.6. Ammonium sulphate is added to 1.5 M and after stirring the precipitate is removed by centrifugation.
  • the interferon gamma variant is then applied onto a Phenyl Sepharose High Performance (Pharmacia) previously equilibrated in 10 mM Tris, 1.5 M ammonium sulphate, pH 7.6.
  • the column is washed with 3 column volumes of 10 mM Tris, 1.5 M ammonium sulphate, pH 7.6, and the bound interferon gamma variant is then eluted in a linear gradient over 10 column volumes to 100% 10 mM Tris, pH 7.6.
  • the flow-through as well as the eluted interferon gamma variant is fractionated. Fractions enriched in the interferon gamma variant are pooled and ammonium sulphate is adjusted to 1.7 M.
  • the interferon gamma variant is applied onto a Butyl Sepharose column previously equilibrated in 10 mM sodium phosphate, 1.7 M ammonium sulphate, pH 7.6. After application the column is washed with 10 mM sodium phosphate, 1.7 M ammonium sulphate, pH 7.6, before eluting the bound interferon gamma variant in a step using 10 mM sodium phosphate, pH 6.5. The flow-through as well as the eluted interferon gamma variant is fractionated.
  • Fractions enriched in the interferon gamma variant are then pooled and applied onto a hydroxyapatite column previously equilibrated in 10 mM sodium phosphate, pH 6.5. After application the column is washed with 5 column volumes of 10 mM sodium phosphate, pH 6.5, before eluting the bound interferon gamma variant in a linear gradient from 0-100%) 500 mM sodium phosphate, pH 6.5, over 30 column volumes. The flow-through as well as the eluted interferon gamma variant is fractionated.
  • Fractions containing the interferon gamma variant are pooled and buffer exchanged into a buffer containing 5 mM sodium succinate, 4% mannitol, pH 6.0. Tween 20 is subsequently added to a concentration of 0.01%.
  • the interferon gamma variant is sterile filtered and stored at -80°C.
  • the variant was constructed and expressed as described in Examples 7 and 8 of WO 01/15736 and purified with the following two-step procedure:
  • the harvested media from roller bottles was centrifuged and filtered through a 0.22 um filter (PVDF).
  • the filtrated media was diafiltrated on a Vivaflow 200 system equipped with a polyethersulfon (PES) membrane with cut off 10000 and applied to a S-Sepharose column (Pharmacia) equilibrated with 50 mM sodium acetate, 50 mM sodium chloride, pH 5.5.
  • PES polyethersulfon
  • S-Sepharose column Pharmacia
  • the interferon variant bound to the column was eluted with 50 mM sodium acetate, 0.5 M sodium chloride, pH 5.5.
  • the concentration of sodium chloride in the eluate from the S-Sepharose column was adjusted to 1.0 M and the sample was applied on a Phenyl-Sepharose High Performance column (Pharmacia) equilibrated with 50 mM sodium acetate, 1.0 M sodium chloride, pH 5.5. Following application the column was washed with Milli Q water. The interferon ⁇ variant was eluted with a gradient from Milli Q water to 60% ethylene glycol, 50 mM sodium acetate, pH 5.5 in 30 column volumes. Fractions containing fully glycosylated interferon ⁇ variant were collected.
  • the buffer in the preparation was changed to 50 mM sodium phosphate, pH 7.0 in a Vivaspin 20 ml concentrator equipped with a PES membrane with cut off 10000.
  • the purified IFN-beta variant was formulated into a composition comprising the variant in an initial concentration of 10 MlU/ml within a 50 mM sodium phosphate buffer (finally adjusted to pH 7.0) and holding 35 mg/ml mannitol as well as 2 mg/ml Tween 80.
  • One composition (Composition A) was without addition of Captisol® (available from CyDex Inc.).
  • composition B comprised 10 mg/ml of Captisol®
  • the compositions were stored in 0.5 ml Eppendorf tubes in aliquots of 50 ⁇ l (with no purging with either nitrogen or argon) at -80°C and 35°C, respectively, for a period of 18 days.
  • the antiviral activity was measured using the antiviral assay described in WO 01/15736.
  • Samples of the protein of example 1 were analyzed by Differential Scanning Calorimetry (DSC) to study the unfolding (or denaturation) of the protein and especially determine the unfolding temperature (Tm) for the protein in each run.
  • DSC Differential Scanning Calorimetry
  • the starting material for DSC analysis was a solution of the protein in 50mM sodium acetate buffer (finally adjusted to pH 5.5). A series of solutions was prepared with varying excipients added to give a final protein concentration of 0.4 mg/mL. Water for Injection was used to serve as blanks for DSC, since the focus was only to dete ⁇ nine shifts in Tm values. Prior to being subjected to DSC analysis, all solutions were degassed by using vacuum for a sufficient time period as described by MicroCal Inc.
  • the behavior of the protein was evaluated by using a DSC apparatus from MicroCal Inc (model VP-DSC).
  • the temperature of the solution in question was gradually increased from ambient temperature (25°C) to about 120°C, at a rate of 1.5°C per minute.
  • the first event was an unfolding reaction (endothermic), and was observed as an upward peak in the scans.
  • the second event was a precipitation (exothermic reaction), and was observed as a downward peak in the scans.
  • Tmi is related to the DSC scan of the original solution without additional excipients added and "Tm 2 " is related to each of the DSC scans of the solutions to which either 2.4 mg/ml Tween 80; 5 mg/ml sodium chloride or 40 mg/ml Captisol® is added.
  • a CHOK1 sub-clone (5/G-10) producing [C17S+Q49N+ Q51T+D110F+ Fl 11N+ RI 13T]IFN-beta glycosylation variant was seeded into 6 roller bottles, each with an expanded surface of 1700 cm 2 (Corning, USA), in 200 ml DMEM/F-12 medium (LifeTechnologies; Cat. # 31330) supplemented with 10% FBS and penicillin/streptomycin (P/S). After 2 days the medium was exchanged. After another 2 days the two roller bottles were nearly 100% confluent and the medium was shifted to 300 ml serum-free UltraCHO medium (BioWhittaker; Cat.
  • the harvested media from roller bottles was centrifuged and filtered through a 0.22 ⁇ m filter (PVDF).
  • the filtrated media was diafiltrated on a Vivaflow 200 system equipped with a polyethersulfon membrane with cut off 10000 and applied to a S- Sepharose column (Pharmacia).
  • the S-Sepharose column was equilibrated with 50 mM sodium acetate, 50 mM sodium chloride, pH 5.5 and the interferon variant was eluted with 50 mM sodium acetate, 0.5 M sodium chloride, pH 5.5. The concentration of sodium chloride in the eluate was adjusted to 1.0 M. The eluate from the S-Sepharose column was applied on a Phenyl-Sepharose
  • High Performance column (Pharmacia) equilibrated with 50 mM sodium acetate, 1.0 M sodium chloride, pH 5.5. Following application the column was washed with 50 mM sodium acetate, 50 mM sodium chloride, pH 5.5. The IFN-beta variant was eluted with a gradient from 50 mM sodium acetate, 50 mM sodium chloride, pH 5.5 to 60% ethylene glycol, 50 mM sodium acetate, pH 5.5 in 30 column volumes. Fractions containing fully glycosylated IFN-beta variant were collected and pooled.
  • the ethylene glycol in the eluate from the Phenyl-Sepharose was removed by passing the eluate through a S-Sepharose column equilibrated with 50 mM sodium acetate, 50 mM sodium chloride, pH 5.5.
  • the ethylene glycol was in the flow through where as the interferon variant bound to the column.
  • the column was washed with 20 mM sodium acetate, pH 5.5 and the interferon variant was eluted with 100 mM sodium phosphate, pH 7.5.
  • the phosphate concentration in the eluate was adjusted to 15 mM sodium phosphate buffer, pH 7.2, and applied on a hydroxyapatite column (CHT I , Ceramic hydroxyapatite, Type I, Biorad) equilibrated with 15 mM sodium phosphate, pH 7.2.
  • CHT I Ceramic hydroxyapatite, Type I, Biorad
  • the fully glycosylated form passed through the column where as the underglycosylated form with one extra site used bound to the column and was eluted with a linear sodium phosphate gradient from 15 mM to 200 mM sodium phosphate, pH 6.8 in 20 column volumes.
  • the reaction mixture contained a mixture of mono-, di- and un-pegylated material.
  • Mono-pegylated material was separated from other species using either cation exchange chromatography or size-exclusion chromatography or a combination of both.
  • pH in the PEGylation solution was adjusted to pH 2.7 and the sample was applied on a SP- Sepharose HR (Pharmacia) column equilibrated with 20 mM sodium citrate, pH 2.7.
  • the pegylated protein was eluted from the column with 50 mM sodium acetate containing 1 M sodium chloride and applied on a size-exclusion column, Sephacryl S-100, ((16/60) Pharmacia) equilibrated with 100 mM sodium acetate, 200 mM sodium chloride, pH 5.5. Fractions containing mono-pegylated material were pooled and characterized further.
  • a protein solution of 0.16 mg/ml in 20 mM sodium phosphate, pH 7.0 was PEGylated with SCM-PEG, 12K, with 2 times molar su ⁇ lus of PEG to possible PEGylation sites, i.e. lysines and N-terminus.
  • the reaction was quenched by addition of a su ⁇ lus of 20 mM glycine, pH 8.0.
  • the reaction mixture contained a mixture of mono-, di-, tri-pegylated material together with underivatized material.
  • the pegylated material was separated from the unmodified protein using either cation exchange chromatography or size- exclusion chromatography or a combination of both. pH in the PEGylation solution was adjusted to pH 2.7 and the sample was applied on a SP-Sepharose HR (Pharmacia) column equilibrated with 20 mM sodium citrate, pH 2.7.
  • the pegylated protein was eluted from the column with 50 mM sodium acetate containing 1 M sodium chloride and applied on a size-exclusion column, Sephacryl S-100, ((16/60) Pharmacia) equilibrated with 100 mM sodium acetate, 200 mM sodium chloride, pH 5.5. Fractions containing the mixture of mono-, di- and tri-pegylated protein were pooled and characterized further.
  • Samples of the IFN-beta glycosylation variant (prepared under example 3) were analyzed by Differential Scanning Calorimetry (DSC) to study the unfolding (or denaturation) of the protein and especially determine the unfolding temperature (Tm) for the protein in each run.
  • DSC Differential Scanning Calorimetry
  • the starting materials for DSC analysis were a solution of the protein in a 20mM sodium phosphate (finally adjusted to pH 7.1). Solutions were prepared with varying excipients added to give a final protein concentration of 0.4 mg/mL. Water for Injection was used to serve as blanks for DSC, since the focus was only to determine shifts in Tm values. Prior to being subjected to DSC analysis, all solutions were degassed by using vacuum for a sufficient time period as described by MicroCal Inc.
  • the behavior of the protein was evaluated by using a DSC apparatus from MicroCal Inc (model VP-DSC).
  • the temperature of the solution in question was gradually increased from ambient temperature (25°C) to about 120°C, at a rate of 1.5°C per minute. As the temperature increased an unfolding reaction (endothermic) was observed as an upward peak in the scans.
  • Captisol® revealed an increase in the Tm- value of about 6.4°C for the sample containing Captisol®. These data clearly demonstrate that addition of Captisol® stabilize the protein being in solution.
  • the purified IFN-beta glycosylation variant PEGylated with 12kDa was formulated into the following compositions comprising the variant in an initial concentration of about 5 MlU/ml within the following buffers: a 10 mM sodium acetate buffer (finally adjusted to pH 5.5) and holding 45 mg/ml mannitol as well as 2 mg/ml Tween 80 (-buffer I), or a 50 mM sodium phosphate buffer (finally adjusted to pH 7.0) and holding 30 mg/ml mannitol as well as 2 mg/ml Tween 80 ( ⁇ buffer II).
  • buffer system I one composition (Composition A) was without addition of Captisol®.
  • composition B comprised 10 mg/ml of Captisol®.
  • composition C was without addition of Captisol®.
  • composition D comprised 10 mg/ml of Captisol®.
  • compositions were stored for a varying length of time period in 0.5 ml Eppendorf tubes in aliquots of 50 ⁇ l (with no purging with either nitrogen or argon) at -80°C. Samples of at least 0.4 mL were stored in siliconized glass vials (Type I glass) at 5, 25 and 35°C (purged with nitrogen prior to closure).
  • the antiviral activity was measured using the antiviral assay described in WO 01/15736.
  • the results of the antiviral activity assay are shown in the table below as "mean percent activity for samples stored at the given temperature, as a function of activity of samples stored at -80°C (analyzed at the same day)" :
  • compositions containing Captisol® either prevents or delays the precipitation of the pegylated IFN-beta variant.
  • composition A and B stored for 34 days at 35°C were inspected to be "highly turbid” and "a clear solution", respectively.
  • the purified IFN-beta variant PEGylated with 20kDa (prepared under example 3) was formulated into the following compositions comprising the variant in an initial concentration of 11-14 MlU/ml within the following buffers: a 10 mM sodium acetate buffer (finally adjusted to pH 5.5) and holding 45 mg/ml mannitol as well as both 2 mg/ml Tween 80 and 6.7 mg/ml
  • Captisol® ( ⁇ Composition A), or a 50 mM sodium phosphate buffer (finally adjusted to pH 7.0) and holding 30 mg/ml mannitol as well as 2 mg/ml Tween 80 and 10 mg/ml Captisol® ( ⁇ Composition B).
  • compositions were stored for a varying length of time period in 0.5 ml Eppendorf tubes in aliquots of 50 ⁇ l (with no purging with either nitrogen or argon) at -80°C. Samples of at least 0.4 L were stored in siliconized glass vials (Type I glass) at 5, 25 and 35°C (purged with nitrogen prior to closure).
  • the antiviral activity was measured using the antiviral assay described in WO 01/15736.
  • the results of the antiviral activity assay are shown in the table below as "mean percent activity for samples stored at the given temperature, as a function of activity of samples stored at -80°C (analyzed at the same day)" :
  • a CHOK1 sub-clone (5/G-10) producing [C17S+K19R+ K33R+K45R+Q49N+ Q51T+D110F+ Fl 11N+ RI 13T]IFN-beta glycosylation variant was produced in 6 roller bottles as described in example 3 and purified according to the protocol used in example 3. The purity of the fully glycosylated variant [C17S+K19R+ K33R+K45R+Q49N+ Q51T+D110F+ Fl 11N+ RI 13T]IFN-beta was judged to be higher than 95% based on SDS-PAGE. Following purification the variant was PEGylated. A fresh stock solution of
  • SCM-PEG succinimidyl ester of carboxymethylated PEG from Shearwater, Alabama, 12 kD or 20 kD
  • a protein solution of 0.1 mg/ml in 20 mM sodium phosphate, pH 7.0 was PEGylated with SCM-PEG, 20K, with 3 times molar su ⁇ lus of PEG to possible PEGylation sites, i.e. lysines and N-terminus. After incubation for 30 min at room temperature, the reaction was quenched by addition of a su ⁇ lus of 20 mM glycine, pH 8.0. The reaction mixture contained a mixture of mono-, di- and un-pegylated material. Mono-pegylated material was separated from other species using either cation exchange chromatography or size-exclusion chromatography or a combination of both. pH in the PEGylation solution was adjusted to pH 2.7 and the sample was applied on a SP-
  • the pegylated protein was eluted from the column with 50 mM sodium acetate containing 1 M sodium chloride and applied on a size-exclusion column, Sephacryl S-100, ((16/60) Pharmacia) equilibrated with 100 mM sodium acetate, 200 mM sodium chloride, pH 5.5. Fractions containing mono-pegylated material was pooled and characterized further.
  • pH in the PEGylation solution was adjusted to pH 2.7 and the sample was applied on a SP-Sepharose HR (Pharmacia) column equilibrated with 20 mM sodium citrate, pH 2.7.
  • the pegylated protein was eluted from the column with 50 mM sodium acetate containing 1 M sodium chloride and applied on a size-exclusion column, Sephacryl S-100, ((16/60) Pharmacia) equilibrated with 100 mM sodium acetate, 200 mM sodium chloride, pH 5.5. Fractions containing the mixture of mono-, di- and tri-pegylated protein were pooled and characterized further.
  • the purified IFN-beta variant PEGylated with 20kDa was formulated into the following compositions comprising the variant in an initial concentration of 5-10 MlU/ml within the following buffers: a 10 mM sodium acetate buffer (finally adjusted to pH 5.0, ⁇ buffer A): a 10 mM sodium acetate buffer (finally adjusted to pH 5.5, ⁇ buffer B), a 10 mM sodium succinate buffer (finally adjusted to pH 5.5, ⁇ buffer C), a 10 mM sodium succinate buffer (finally adjusted to pH 6.0, ⁇ buffer D) and a 10 mM sodium citrate buffer (finally adjusted to pH 6.0, ⁇ buffer E).
  • a 10 mM sodium acetate buffer finally adjusted to pH 5.0, ⁇ buffer A
  • a 10 mM sodium acetate buffer finally adjusted to pH 5.5, ⁇ buffer B
  • a 10 mM sodium succinate buffer finally adjusted to pH 5.5, ⁇ buffer C
  • the amount of mannitol was adjusted to ensure isotonic solutions suitable for parenteral administration.
  • compositions were stored for a varying length of time period in 0.5 ml Eppendorf tubes in aliquots of 25 ⁇ l (with no purging with either nitrogen or argon) at -80°C and 5°C. Samples of at least 0.4 ml were stored in siliconized glass vials (Type I glass) at 25 (purged with nitrogen prior to closure).
  • the antiviral activity was measured using the antiviral assay described in WO 01/15736.
  • results of the antiviral activity assay are shown in the following tables as "mean percent activity for samples stored at the given temperature, as a function of activity of samples stored at -80°C (analyzed at the same day)".
  • compositions that do not contain Captisol® have been stored for a considerably shorter time period than the remaining compositions, the data clearly show that addition of Captisol® to a pharmaceutical acceptable buffer system can delay or even prevent the loss of bioactivity at certain pH values - even when stored at elevated temperatures for an extended time period.
  • Tween 20 up to 2 mg/ml
  • Tween 80 up to 2 mg/ml
  • mannitol up to 50 mg/ml
  • sodium chloride up to 9 mg/ml
  • Formulations containing [C 17 S+Q49N+ Q51T+D110F+ F111N+ R113T] IFN-beta glycosylation variant PEGylated with 20kDa Prior to formulation the purified IFN-beta variant PEGylated with 20kDa (prepared under example 3) was equilibrated with a solution consisting of 10 mM sodium acetate buffer (finally adjusted to pH 5.5) holding 25 mg/ml mannitol.
  • This material was formulated into the following compositions comprising the variant in an initial concentration of lOO ⁇ g/ml within the following buffers: a 10 mM sodium acetate buffer (finally adjusted to pH 5.5) holding mannitol, sodium chloride (NaCl), Tween 80 and Captisol® in the following combinations.
  • compositions were sterilized by filtration, filled under aseptic conditions into sterilized containers and stored for a varying length of time period. Aliquots of 20 ⁇ l were filled into 0.5 ml Eppendorf tubes and stored at -80°C. Aliquots of at least 0.3 ml were filled into siliconized glass vials (Type I glass) and stored at 5, 25 and 35°C.
  • the antiviral activity was measured using the antiviral assay described in WO 01/15736.
  • Example 6 Compared to Example 6, significantly lot less Tween 80 is used in this example, which may explain the observed improved stability at elevated temperatures of the polypeptide in question.
  • the IFN-beta bulk preparation was purified as the variant mentioned in Example 3 with the differences that the final step consisted of a gel filtration through a Superdex 75 column. This material was then equilibrated with a solution consisting of 50 mM sodium acetate (adjusted to pH 5.5) holding both 0.1 M sodium chloride and 0.2 M mannitol.
  • This material formulated into the following compositions comprising the IFN-beta in an initial concentration of about 5 MlU/ml within the following buffers: a 50 mM sodium acetate buffer (finally adjusted to pH 5.5) holding 28 mg/ml mannitol, 1.3 mg/ml sodium chloride, and 2 mg/ml Tween 80 as well as no Captisol® (Formulation A) or 10 mg/ml Captisol® (Formulation B).
  • compositions were filled into Eppendorf tubes and stored for a varying length of time period in aliquots of 50 ⁇ l at -80°C, -20 and 5°C.
  • the antiviral activity was measured using the antiviral assay described in WO 01/15736.
  • the results of the antiviral activity assay after 354 days storage are shown in the table below as "mean percent activity for samples stored at the given temperature, as a function of activity of samples stored at -80°C (analyzed at the same day)" :
  • IFN-gamma wild-type IFN-gamma
  • IFN-gamma bulk preparation was conducted as described in Example H within the section "Materials and methods for preparing interferon gamma".
  • This preparation was formulated into the following compositions comprising the IFN-gamma in an initial concentration of 0.5 mg/ml within the following buffers: a 5 mM sodium succinate buffer (finally adjusted to pH 6.0) holding 40 mg/ml mannitol and 0.01% Tween 20 as well as no Captisol® (Formulation A) or 50 mg/ml Captisol® (Formulation B).
  • compositions were sterilized by filtration, filled under aseptic conditions into sterilized containers and stored for a varying length of time period. Aliquots of 20 ⁇ l were filled into 0.5 ml Eppendorf tubes and stored at -80°C. Aliquots of at least 0.15 ml were filled into siliconized glass vials (Type I glass) and stored at 5, 25, 35 and 40°C. The activity was measured using the luciferase assay described in the section
  • the IFN-gamma variant was prepared as described in Example D described in the section "Materials and methods for preparing interferon gamma”. This material was purified as described in Example H described in the section "Materials and methods for preparing interferon gamma”.
  • the purified material formulated into the following compositions comprising the variant in an initial concentration of 0.5 mg/ml within the following buffers: a 5 mM sodium succinate buffer (finally adjusted to pH 6.0) holding 40 mg/ml mannitol and 0.01 %> Tween 20;as well as either no Captisol® (Formulation A) or 50 mg/ml Captisol® (Formulation B).
  • compositions were sterilized by filtration, filled under aseptic conditions into sterilized containers and stored for a varying length of time period. Aliquots of 20 ⁇ l were filled into 0.5 ml Eppendorf tubes and stored at -80°C. Aliquots of at least 0.15 ml were filled into siliconized glass vials (Type I glass) and stored at 5 and 25.
  • the activity was measured using the luciferase assay described in the section "Materials and methods for preparing interferon gamma”.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Virology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Animal Behavior & Ethology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Public Health (AREA)
  • Oncology (AREA)
  • Engineering & Computer Science (AREA)
  • Molecular Biology (AREA)
  • Communicable Diseases (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biotechnology (AREA)
  • Pulmonology (AREA)
  • Zoology (AREA)
  • Toxicology (AREA)
  • Hematology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Medicinal Preparation (AREA)
EP02748632A 2001-06-29 2002-06-28 Stabilized formulations of interferons with sulfoalkyl ether cyclodextrins Withdrawn EP1414498A2 (en)

Applications Claiming Priority (7)

Application Number Priority Date Filing Date Title
DK200101040 2001-06-29
DKPA200101040 2001-06-29
DKPA200101277 2001-08-30
DK200101277 2001-08-30
DKPA200200257 2002-02-19
DK200200257 2002-02-19
PCT/DK2002/000444 WO2003002152A2 (en) 2001-06-29 2002-06-28 Stabilized formulations of interferons with sulfoalkyl ether cyclodextrins

Publications (1)

Publication Number Publication Date
EP1414498A2 true EP1414498A2 (en) 2004-05-06

Family

ID=27222516

Family Applications (1)

Application Number Title Priority Date Filing Date
EP02748632A Withdrawn EP1414498A2 (en) 2001-06-29 2002-06-28 Stabilized formulations of interferons with sulfoalkyl ether cyclodextrins

Country Status (7)

Country Link
EP (1) EP1414498A2 (ja)
JP (1) JP2004522803A (ja)
CN (1) CN1522159A (ja)
CA (1) CA2452364A1 (ja)
MX (1) MXPA03011793A (ja)
RU (1) RU2004102500A (ja)
WO (1) WO2003002152A2 (ja)

Families Citing this family (32)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7959910B2 (en) 2000-07-31 2011-06-14 Biolex Therapeutics, Inc. C-terminally truncated interferon alpha variants
AU2003254641A1 (en) * 2002-08-28 2004-03-19 Maxygen Aps Interferon beta-like molecules for treatment of cancer
US20040126360A1 (en) * 2002-10-09 2004-07-01 Manning Mark C. Oral formulations for proteins and polypeptides
AU2003303635B2 (en) 2002-12-26 2009-07-23 Mountain View Pharmaceuticals, Inc. Polymer conjugates of interferon-beta with enhanced biological potency
AU2004298781B2 (en) 2003-12-11 2010-04-01 Ares Trading S.A. Stabilized interferon liquid formulations
CA2567309A1 (en) * 2004-06-01 2005-12-15 Ares Trading S.A. Method of stabilizing proteins
CN1984674A (zh) * 2004-06-07 2007-06-20 纳斯泰克制药公司 不含蛋白质或多肽类稳定剂的干扰素β鼻内制剂
US20060024272A1 (en) 2004-07-29 2006-02-02 Large Scale Biology Corporation C-terminally truncated interferon
SI1917276T1 (en) * 2005-08-26 2018-05-31 Ares Trading S.A. PROCEDURE FOR PREPARATION OF GLYCOSYLATED INTERFERONE BETA
WO2007098106A2 (en) * 2006-02-17 2007-08-30 Pepgen Coporation Respiratory tract delivery of interferon-tau
CA2644127A1 (fr) * 2006-03-08 2007-09-13 Biomethodes Variants ameliores de l'interferon-gamma humain (ifny)
JP2008069073A (ja) * 2006-09-12 2008-03-27 Yokohama Tlo Co Ltd ラクトフェリン複合体及びその製造方法
US8617531B2 (en) 2006-12-14 2013-12-31 Bolder Biotechnology, Inc. Methods of making proteins and peptides containing a single free cysteine
CA2685331C (en) 2007-04-27 2016-07-05 Cydex Pharmaceuticals, Inc. Formulations containing clopidogrel and sulfoalkyl ether cyclodextrin and methods of use
US7625555B2 (en) 2007-06-18 2009-12-01 Novagen Holding Corporation Recombinant human interferon-like proteins
EP2234645B1 (en) * 2007-12-20 2012-05-02 Merck Serono S.A. Peg-interferon-beta formulations
WO2010053487A1 (en) 2008-11-07 2010-05-14 Cydex Pharmaceuticals, Inc. Composition containing sulfoalkyl ether cyclodextrin and latanoprost
EP2358395A4 (en) * 2008-11-17 2013-11-20 Hoffmann La Roche METHOD AND FORMULATION FOR REDUCING THE AGGREGATION OF A MACROMOLECULUM UNDER PHYSIOLOGICAL CONDITIONS
KR20190071006A (ko) 2009-05-13 2019-06-21 사이덱스 파마슈티칼스, 인크. 프라수그렐 및 사이클로덱스트린 유도체를 포함하는 약학 조성물 및 그의 제조 및 사용 방법
RU2482871C2 (ru) * 2010-06-07 2013-05-27 Общество с ограниченной ответственностью "Центр Инновационных Технологий "БиоФарм" (ООО "ЦИТ "БиоФарм") Композиция для лечения вирусных заболеваний животных
US8492386B2 (en) 2011-10-21 2013-07-23 Abbvie Inc. Methods for treating HCV
WO2013059638A1 (en) 2011-10-21 2013-04-25 Abbvie Inc. Combination treatment (eg. with abt-072 or abt -333) of daas for use in treating hcv
CN103826627B (zh) 2011-10-21 2016-02-24 艾伯维公司 包含至少两种直接抗病毒剂和利巴韦林的组合物在制备治疗hcv的药物中的用途
US8466159B2 (en) 2011-10-21 2013-06-18 Abbvie Inc. Methods for treating HCV
US10012064B2 (en) 2015-04-09 2018-07-03 Highlands Natural Resources, Plc Gas diverter for well and reservoir stimulation
US10344204B2 (en) 2015-04-09 2019-07-09 Diversion Technologies, LLC Gas diverter for well and reservoir stimulation
SG11201804961UA (en) 2015-12-30 2018-07-30 Genentech Inc Formulations with reduced degradation of polysorbate
US10982520B2 (en) 2016-04-27 2021-04-20 Highland Natural Resources, PLC Gas diverter for well and reservoir stimulation
JP7129703B2 (ja) 2016-04-28 2022-09-02 エモリー ユニバーシティー アルキン含有ヌクレオチド及びヌクレオシド治療組成物並びにそれらに関連した使用
JP7105200B2 (ja) * 2016-05-13 2022-07-22 オリオニス バイオサイエンシズ ビーブイ 標的突然変異体インターフェロン-ベータおよびその使用
CN111358938B (zh) * 2020-03-30 2023-08-11 温州肯恩大学(Wenzhou-KeanUniversity) 人干扰素-ε与干扰素-γ组合药及用途
CN113797317B (zh) * 2021-10-26 2024-01-09 科兴生物制药股份有限公司 一种组合物及其制备方法和应用

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5997856A (en) * 1988-10-05 1999-12-07 Chiron Corporation Method and compositions for solubilization and stabilization of polypeptides, especially proteins
US5376645A (en) * 1990-01-23 1994-12-27 University Of Kansas Derivatives of cyclodextrins exhibiting enhanced aqueous solubility and the use thereof
US6046177A (en) * 1997-05-05 2000-04-04 Cydex, Inc. Sulfoalkyl ether cyclodextrin based controlled release solid pharmaceutical formulations
WO1998056422A1 (en) * 1997-06-13 1998-12-17 The University Of Kansas Polar drug or prodrug compositions with extended shelf-life storage and a method of making thereof

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of WO03002152A2 *

Also Published As

Publication number Publication date
CA2452364A1 (en) 2003-01-09
MXPA03011793A (es) 2004-04-02
CN1522159A (zh) 2004-08-18
RU2004102500A (ru) 2005-04-10
WO2003002152A3 (en) 2003-05-01
JP2004522803A (ja) 2004-07-29
WO2003002152A2 (en) 2003-01-09

Similar Documents

Publication Publication Date Title
US20030138403A1 (en) Interferon formulations
EP1414498A2 (en) Stabilized formulations of interferons with sulfoalkyl ether cyclodextrins
EP1366075B1 (en) New interferon beta-like molecules
US7338788B2 (en) Interferon-β variants and conjugates
IL149267A (en) Couplings of interferon gamma
US20090030183A1 (en) Interferon Beta-Like Molecules
US7390638B2 (en) S99T C-11 Truncated polynucleotides encoding interferon gamma polypeptide variants
KR20020034181A (ko) 새로운 인터페론 베타-유사 분자
EP1334127A1 (en) Single-chain multimeric polypeptides
KR20030094336A (ko) 인터페론 감마 폴리펩티드 변이체
US7524931B2 (en) Full-length interferon gamma polypeptide variants
US20020169290A1 (en) New multimeric interferon beta polypeptides
AU2002319109A1 (en) Stabilized formulations of interferons with sulfoalkyl ether cyclodextrins
EP1334128A2 (en) New multimeric interferon beta polypeptides
ZA200303964B (en) New interferon beta-like molecules.
AU2002235727A1 (en) New interferon beta-like molecules
ZA200200337B (en) New interferon beta-like molecules.
AU2002252971A1 (en) Interferon gamma polypeptide variants

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20040129

AK Designated contracting states

Kind code of ref document: A2

Designated state(s): AT BE CH CY DE DK ES FI FR GB GR IE IT LI LU MC NL PT SE TR

AX Request for extension of the european patent

Extension state: AL LT LV MK RO SI

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20060711