US20080287366A1 - Parenteral formulations of a peptide for the treatment of systemic lupus erythematosus - Google Patents

Parenteral formulations of a peptide for the treatment of systemic lupus erythematosus Download PDF

Info

Publication number
US20080287366A1
US20080287366A1 US11/985,068 US98506807A US2008287366A1 US 20080287366 A1 US20080287366 A1 US 20080287366A1 US 98506807 A US98506807 A US 98506807A US 2008287366 A1 US2008287366 A1 US 2008287366A1
Authority
US
United States
Prior art keywords
pharmaceutical composition
peptide
cyclodextrin
compound
hours
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US11/985,068
Inventor
Sharon Cohen-Vered
Esmira Naftali
Vera Weinstein
Adrian Gilbert
Ety Klinger
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.)
Teva Pharmaceutical Industries Ltd
Original Assignee
Teva Pharmaceutical Industries 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 Teva Pharmaceutical Industries Ltd filed Critical Teva Pharmaceutical Industries Ltd
Priority to US11/985,068 priority Critical patent/US20080287366A1/en
Publication of US20080287366A1 publication Critical patent/US20080287366A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • BPERFORMING OPERATIONS; TRANSPORTING
    • B82NANOTECHNOLOGY
    • B82YSPECIFIC USES OR APPLICATIONS OF NANOSTRUCTURES; MEASUREMENT OR ANALYSIS OF NANOSTRUCTURES; MANUFACTURE OR TREATMENT OF NANOSTRUCTURES
    • B82Y5/00Nanobiotechnology or nanomedicine, e.g. protein engineering or drug delivery
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/715Polysaccharides, i.e. having more than five saccharide radicals attached to each other by glycosidic linkages; Derivatives thereof, e.g. ethers, esters
    • A61K31/716Glucans
    • A61K31/724Cyclodextrins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/04Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
    • A61K38/10Peptides having 12 to 20 amino acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/30Macromolecular organic or inorganic compounds, e.g. inorganic polyphosphates
    • A61K47/36Polysaccharides; Derivatives thereof, e.g. gums, starch, alginate, dextrin, hyaluronic acid, chitosan, inulin, agar or pectin
    • A61K47/40Cyclodextrins; Derivatives thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/69Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit
    • A61K47/6949Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit inclusion complexes, e.g. clathrates, cavitates or fullerenes
    • A61K47/6951Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit inclusion complexes, e.g. clathrates, cavitates or fullerenes using cyclodextrin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/14Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles
    • A61K9/19Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles lyophilised, i.e. freeze-dried, solutions or dispersions
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection

Definitions

  • SLE Systemic lupus erythematosus
  • lupus is a debilitating autoimmune disease characterized by the presence of an array of autoantibodies, including antibodies to dsDNA, to nuclear antigens and to ribonucleoproteins.
  • SLE affects approximately 1 in 2000 individuals (U.S. 1 in 700 women). The disease primarily affects young women, with a female-to male ratio of approximately 9:1.
  • Systemic lupus can affect almost any organ or system of the body.
  • Systemic lupus may include periods in which few, if any, symptoms are evident (“remission”) and other times when the disease becomes more active (“flare”). Most often when people mention “lupus,” they are referring to the systemic form of the disease.
  • Corticosteroids are the mainstay in treating systemic autoimmune disorders. Life threatening, severely disabling manifestations of SLE are treated with high doses of glucocorticoids (1-2 mg/kg/day). Undesirable effects of chronic glucocorticoids include an array of prominent adverse effects such as cushingoid habitus, central obesity, hypertension, infection, capillary fragility, hirsutism, accelerated osteoporosis, cataracts, diabetes mellitus, myopathy and psychosis. In addition to corticosteroid toxicity, patient compliance to a dosage regimen also poses a serious problem.
  • Cytotoxic agents are also used for controlling active disease, reducing the rate of disease flares, and reducing steroid requirements.
  • Undesirable side effects of the latter include bone marrow depression, increased infections with opportunistic organisms, irreversible ovarian failure, alopecia and increased risk of malignancy.
  • SLE is an inflammatory disease for which to date there is no definitive treatment or cure.
  • the disease results in acute and chronic complications.
  • the only treatments available are palliative, aimed at relieving acute symptoms and preventing chronic complications, often with profound side effects. There is therefore an unmet need in this field, and both physicians and patients would welcome new treatments which could potentially eliminate or reduce the unwanted manifestations of the disease.
  • Peptides based on the complementarity-determining region of the human monoclonal anti-DNA 16/6Id antibody capable of immunomodulating SLE associated responses have been disclosed in PCT International Publication No. WO 02/067848 A2, the entire contents of which are hereby incorporated by reference.
  • region CDR1 was found to inhibit the proliferative response of peripheral blood lymphocytes (PBL) of SLE patients to the human anti-DNA 16/6Id mAB, and to ameliorate disease manifestations of mice afflicted with spontaneous or experimental SLE.
  • Human CDR1 is a synthetic peptide of 19 amino acids based on the complementarity-determining region 1 (CDR1) of the human anti-dsDNA mAb denoted 16/6 Id (Waisman, A., et al. “Modulation of murine systemic lupus erythematosus with peptides based on complementarity determining regions of pathogenic anti-DNA monoclonal antibodies.” Proc. Natl. Acad. Sci. U.S.A. (1997), 94(4): 4620-4625).
  • CDR1 complementarity-determining region 1
  • mice Balb/c mice and SLE-prone mice, i.e. (NZBxNZW)F1 mice—treatment with either mCDR based-peptides or Compound 1 significantly reduced the SLE related findings, notably immune complex deposits (ICD) in the kidney, proteinuria and leukopenia.
  • ICD immune complex deposits
  • the treatment had no effect on the 16/6 Id specific antibody response (Waisman, A., et al. “Modulation of murine systemic lupus erythematosus with peptides based on complementarity determining regions of pathogenic anti-DNA monoclonal antibodies.” Proc. Natl. Acad. Sci. U.S.A .
  • the subject invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising
  • the subject invention also provides a pharmaceutical composition
  • a pharmaceutical composition comprising
  • the subject invention also provides a method of alleviating symptoms of systemic lupus erythematosus (SLE) in a human subject comprising administering to the human subject any of the above pharmaceutical compositions in an amount effective to alleviate the symptoms of SLE in the human subject.
  • SLE systemic lupus erythematosus
  • the subject invention also provides a process for manufacturing the above pharmaceutical composition comprising the steps of:
  • the subject invention also provides a process of lyophilizing the above pharmaceutical composition, comprising the steps of:
  • the subject invention also provides a process of lyophilizing the above pharmaceutical composition, comprising the steps of:
  • FIG. 1 Human CDR1 (Compound 1) as acetate salt—showing the molecular and structural formulas of hCDR1, the amino acid sequence, and physical parameters
  • FIG. 2 IL-2 Secretion from cells taken from mice treated with Compound 1 and Captisol® solution after the cells were subsequently activated with a solution of Compound 1 in PBS.
  • FIG. 3 IFN- ⁇ Secretion from cells taken from mice treated with Compound 1 solution after the cells were subsequently activated with a solution of compound 1 in EM-1 (2.5 ⁇ 10 6 cells/well).
  • FIG. 4 IFN- ⁇ Secretion from cells taken from mice treated with Compound 1 solution after the cells were subsequently activated with a solution of compound 1 in EM-1 (5 ⁇ 10 6 cells/well).
  • FIG. 6 Kidney sections from (NZBxNZW)F1 mice showing intensity of Immune Complex Deposits.
  • the top row sections are from a Captisol®-treated mouse, the mid-row sections are from a mouse treated with 50 ⁇ g/mouse Compound 1 and the bottom row sections are from a mouse treated with 25 ⁇ g/mouse Compound 1.
  • FITC immunohistology Left: ⁇ 100, Right: ⁇ 400.
  • the subject invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising
  • the concentration of the acetate salt of the peptide is at least 0.5 mg/ml.
  • the concentration of the salt of the peptide is from 0.5 mg/ml to 10 mg/ml.
  • the concentration of the salt of the peptide is from 0.5 mg/ml to 2.5 mg/ml.
  • the concentration of the salt of the peptide is from 2.5 mg/ml to 5 mg/ml.
  • the concentration of the salt of the peptide is from 5 mg/ml to 7 mg/ml.
  • the concentration of the salt of the peptide is from 7 mg/ml to 8.5 mg/ml.
  • the concentration of the salt of the peptide is from 8.5 mg/ml to 10 mg/ml.
  • the concentration of the salt of the peptide is from 9 mg/ml to 10 mg/ml.
  • the concentration of the salt of the peptide is from 10 mg/ml to 15 mg/ml.
  • the concentration of the salt of the peptide is from 15 mg/ml to 20 mg/ml.
  • the concentration of the salt of the peptide is 1.0 mg/ml.
  • the concentration of the salt of the peptide is 2.5 mg/ml.
  • the concentration of the salt of the peptide is 5 mg/ml.
  • the concentration of the salt of the peptide is 10 mg/ml.
  • the concentration of the salt of the peptide is 15 mg/ml.
  • the concentration of the salt is from 0.1 mg/ml to 0.5 mg/ml.
  • the concentration of the salt is from 0.1 mg/ml to 0.2 mg/ml.
  • the concentration of the salt is from 0.2 mg/ml to 0.3 mg/ml.
  • the concentration of the salt is from 0.3 mg/ml to 0.4 mg/ml.
  • the concentration of the salt is from 0.4 mg/ml to 0.5 mg/ml.
  • the composition has a pH between 6.5 and 8.5.
  • the composition has a pH between 7.5 and 8.5.
  • the composition has a pH between 4 and 5.
  • the composition has a pH between 5 and 6.
  • the composition has a pH between 6 and 7.
  • the composition has a pH between 7 and 8.
  • the composition has a pH between 8 and 9.
  • the pharmaceutically acceptable salt is an acetate salt.
  • the substituted ⁇ -cyclodextrin is a hydroxypropyl, a sulfobutyl ether, or a sulfopropyl ether substituted ⁇ -cyclodextrin.
  • the substituted ⁇ -cyclodextrin is a sulfobutyl ether substituted ⁇ -cyclodextrin.
  • the pharmaceutically acceptable salt is an acetate salt
  • the substituted ⁇ -cyclodextrin is hepta-(sulfobutyl ether)- ⁇ -cyclodextrin.
  • the composition further comprises a pharmaceutically acceptable buffer in an amount and of a type suitable to make the pH of the pharmaceutical composition in the range of 4-9.
  • the buffer may be acetate buffer, citrate buffer, or sodium carbonate.
  • the subject invention also provides a pharmaceutical composition
  • a pharmaceutical composition comprising
  • the concentration of the acetate salt of the peptide is at least 0.5 mg/ml.
  • the concentration of the acetate salt of the peptide is from 0.5 mg/ml to 10 mg/ml.
  • the concentration of the acetate salt of the peptide is from 0.5 mg/ml to 2.5 mg/ml.
  • the concentration of the salt is from 0.1 mg/ml to 0.5 mg/ml.
  • the concentration of the salt is from 0.1 mg/ml to 0.2 mg/ml.
  • the concentration of the salt is from 0.2 mg/ml to 0.3 mg/ml.
  • the concentration of the salt is from 0.3 mg/ml to 0.4 mg/ml.
  • the concentration of the salt is from 0.4 mg/ml to 0.5 mg/ml.
  • the concentration of the salt of the peptide is from 5 mg/ml to 7 mg/ml.
  • the concentration of the salt of the peptide is from 7 mg/ml to 8.5 mg/ml.
  • the concentration of the salt of the peptide is from 8.5 mg/ml to 10 mg/ml.
  • the concentration of the salt of the peptide is from 9 mg/ml to 10 mg/ml.
  • the concentration of the salt of the peptide is from 10 mg/ml to 15 mg/ml.
  • the concentration of the salt of the peptide is from 15 mg/ml to 20 mg/ml.
  • the concentration of acetate salt of the peptide is 1.0 mg/ml.
  • the concentration of acetate salt of the peptide is 2.5 mg/ml.
  • the concentration of the salt of the peptide is 5 mg/ml.
  • the concentration of the salt of the peptide is 10 mg/ml.
  • the concentration of the salt of the peptide is 15 mg/ml.
  • the concentration of hepta-(sulfobutyl ether)- ⁇ -cyclodextrin is 120 mg/ml and the pH of the composition is between 7.5 and 8.5.
  • the subject invention also provides a method of alleviating symptoms of systemic lupus erythematosus (SLE) in a human subject comprising administering to the human subject any of the above pharmaceutical compositions in an amount effective to alleviate the symptoms of SLE in the human subject.
  • SLE systemic lupus erythematosus
  • the subject invention also provides the above pharmaceutical compositions for use in treating SLE in a human subject.
  • the subject invention also provides a process for manufacturing any of the above pharmaceutical compositions comprising the steps of:
  • the resulting final concentration of the substituted ⁇ -cyclodextrin in the pharmaceutical composition is from 70 mg/ml to 170 mg/ml.
  • the predetermined concentration of the substituted ⁇ -cyclodextrin is such which results in a final concentration of substituted ⁇ -cyclodextrin in the pharmaceutical composition of from 80 mg/ml to 170 mg/ml.
  • the predetermined concentration of the substituted ⁇ -cyclodextrin is such which results in a final concentration of substituted ⁇ -cyclodextrin in the pharmaceutical composition of from 90 mg/ml to 170 mg/ml.
  • the predetermined concentration of the substituted ⁇ -cyclodextrin is such which results in a final concentration of substituted ⁇ -cyclodextrin in the pharmaceutical composition of from 100 mg/ml to 170 mg/ml.
  • the predetermined concentration of the substituted ⁇ -cyclodextrin is such which results in a final concentration of substituted ⁇ -cyclodextrin in the pharmaceutical composition of from 110 mg/ml to 170 mg/ml.
  • the predetermined concentration of the substituted ⁇ -cyclodextrin is such which results in a final concentration of substituted ⁇ -cyclodextrin in the pharmaceutical composition of from 120 mg/ml to 170 mg/ml.
  • the predetermined, concentration of the substituted ⁇ -cyclodextrin is such which results in a final concentration of substituted ⁇ -cyclodextrin in the pharmaceutical composition of from 0.130 mg/ml to 170 mg/ml.
  • the predetermined concentration of the substituted ⁇ -cyclodextrin is such which results in a final concentration of substituted ⁇ -cyclodextrin in the pharmaceutical composition of from 140 mg/ml to 170 mg/ml.
  • the predetermined concentration of the substituted ⁇ -cyclodextrin is such which results in a final concentration of substituted ⁇ -cyclodextrin in the pharmaceutical composition of from 150 mg/ml to 170 mg/ml.
  • the predetermined concentration of the substituted ⁇ -cyclodextrin is such which results in a final concentration of substituted ⁇ -cyclodextrin in the pharmaceutical composition of from 160 mg/ml to 170 mg/ml.
  • the predetermined concentration of the substituted ⁇ -cyclodextrin is such which results in a final concentration of substituted ⁇ -cyclodextrin in the pharmaceutical composition is 120 mg/ml.
  • the predetermined amount of peptide is such which results in a final concentration of peptide in the pharmaceutical composition is at least 0.1 mg/ml.
  • the predetermined amount of peptide is such which results in a final concentration of peptide in the pharmaceutical composition is at least 0.5 mg/ml.
  • the predetermined amount of peptide is such which results in a final concentration of peptide in the pharmaceutical composition is 2.5 mg/ml, 2.0 mg/ml, 11.0 mg/ml, 0.5 mg/ml or 0.1 mg/ml.
  • the predetermined amount of peptide is such which results in a final concentration of peptide in the pharmaceutical composition is 5 mg/ml, 10 mg/ml or 15 mg/ml.
  • step b) further comprises mixing the solution for 1 hour.
  • step c) the pH is adjusted using HCl or NaOH 1.0N.
  • the process further comprises filtering the solution of step d) through a cellulose acetate filter.
  • the subject invention also provides a composition prepared by the above process.
  • the subject invention also provides a process of lyophilizing the above pharmaceutical composition, comprising the steps of:
  • step a) is performed within 2 hours.
  • step b) is performed within 3 hours.
  • step c) is performed over 13 hours.
  • step c) is performed at a pressure of 110 ⁇ bar.
  • step d) is performed over 13 hours.
  • step d) is performed at a pressure of 110 ⁇ bar.
  • step e) the pressure is reduced to 10 ⁇ bar.
  • step e) is performed over 5 hours.
  • the subject invention also provides a lyophilized pharmaceutical composition prepared by the above process.
  • the subject invention also provides a process of lyophilizing the above pharmaceutical composition, comprising the steps of:
  • step a) is performed within 6 hours.
  • step b) is performed within 3 hours.
  • step c) is performed over 19 hours.
  • step c) is performed at a pressure of 150 ⁇ bar.
  • step d) is performed over 13 hours.
  • step d) is performed at a pressure of 150 ⁇ bar.
  • step e) is performed over 8 hours.
  • step e) is performed at a pressure of 150 ⁇ bar.
  • the subject invention also provides a lyophilized pharmaceutical composition prepared by any of the above processes.
  • the water content of the composition is less than 5%.
  • the water content of the composition is less than 4.0%.
  • the water content of the composition is less then 3.5%.
  • the subject invention also provides a lyophilized pharmaceutical composition comprising
  • the subject invention also provides a packaged pharmaceutical composition comprised of:
  • the preparations of the present invention may be given parenterally, topically, or rectally. They are of course given by forms suitable for each administration route. For example, they are administered by injection, inhalation, ointment, suppository, etc. administration by injection, infusion or inhalation; topical by lotion or ointment; and rectal by suppositories.
  • parenteral administration and “administered parenterally” as used herein means modes of administration other than enteral and topical administration, usually by injection, and includes, without limitation, intravenous, intramuscular, intraarterial, intrathecal, intracapsular, intraorbital, intracardiac, intradermal, intraperitoneal, transtracheal, subcutaneous, subcuticular, intraarticular, subcapsular, subarachnoid, intraspinal and intrasternal injection and infusion.
  • systemic administration means the administration of a compound, drug or other material other than directly into the central nervous system, such that it enters the patient's system and, thus, is subject to metabolism and other like processes, for example, subcutaneous administration.
  • the human hCDR1 peptide (Compound 1) is described in PCT International Publication No WO 02/067848, published Sep. 6, 2002, and can be prepared by methods well known in the art, (see, for example, Peptides: Synthesis, Structure and Applications , ed. by B. Gutte, Academic Press, 1995 ; Peptide Synthesis Protocols , ed. By M. Pennington and B. Dunn, Humana Press, 1994; Schnolzer, M. et al., “In situ neutralization in Boc-chemistry solid phase synthesis. Rapid, High yield assembly of difficult sequences.” Int. J. Pept. Protein Res . (1992) 40: 180-193).
  • Compound 1 is a synthetic polypeptide composed of 19 amino acids. It is provided as an acetate salt. The aqueous solubility of the peptide has been determined to be less than 0.5 mg/ml.
  • FIG. 1 shows compound 1 as an acetate salt.
  • Compound 1 was dissolved in the chosen solubility enhancer solution either separately or in combination with other excipients and the solutions were stirred for at least an hour. The pH was adjusted if needed. The solutions were visually examined to estimate the solubility and sent for analytical assay determination. For a few chosen formulations, biological activity was also tested.
  • Table 1 presents the type of solubility enhancers used for the formulation development.
  • Tables 2 and 3 summarize the experiments that were performed with the various solubility enhancers.
  • Table 2 summarizes the initial screening performed with peptide concentrations in the range of 5 to 10 mg/ml. The experimental work that was performed with higher peptide concentration was then repeated with the lower doses (see table 3)
  • Non aqueous solvents such as Ethanol, Glycerin, Propylene glycol, Chremophore and their combinations were tested but did not increase the solubility of Compound 1.
  • a solution of 30% DMA (dimethyl-acetamide) yielded solubility in the desired ranges (5 to 9 mg/ml), but was not suitable for a pharmaceutical formulation due to its toxicity profile. Improved solubility was also observed using 30% (w/w) PEG 400 (5 to 9 mg/ml). This latter formulation was chosen for the toxicology studies, but it has proved to be both inactive in the biological assay, and may have been the cause of some adverse effects in a mouse toxicity study. Thus, it was decided not to further pursue this formulation. In view of the preliminary experiments non-aqueous solvents were not used in the subject formulations.
  • Co-solvents including Polysorbate 20 and Polysorbate 80 were tested alone and in combination with other excipients. While lower concentrations of Polysorbates (up to 6%) did not improve the solubility of the peptide, higher concentrations (up to 10%—see table 2) improved the solubility of the peptide up to 2 mg/ml. However, such high concentrations of Polysorbates were deemed unsuitable for pharmaceutical formulations.
  • cyclodextrins Two types of cyclodextrins, both approved for use in marketed parenteral products, were also tested: Hydroxypropyl- ⁇ -cyclodextrin and Sulfobutylether- ⁇ -cyclodextrin (Captisol®). Both markedly increased the solubility of the peptide (concentrations in the levels of 10 mg/ml for Hydroxypropyl- ⁇ -cyclodextrin and 2.5 for Captisol®). The biological activity of the two cyclodextrin formulations was tested and was found to be equal to the activity of the peptide alone.
  • CAPTISOL® is a commercially available polyanionic ⁇ -cyclodextrin derivative with a sodium sulfonate salt separated from the hydrophobic cavity by a butyl ether spacer group, or sulfobutylether (SBE).
  • SBE sulfobutylether
  • CAPTISOL® is the trade name for CyDex Inc.'s hepta-substituted sulfobutylether ⁇ -cyclodextrin (SBE7- ⁇ -CD) preparation (www.captisol.com).
  • SBE7- ⁇ -CD sulfobutylether
  • CAPTISOL® is hydrophilic, the solubility of the complexed drug molecule is thereby enhanced.
  • cyclodextrins to enhance the solubility of drug molecules is disclosed in U.S. Pat. Nos. 5,134,127 and 5,376,645, the entire contents of which are hereby incorporated by reference.
  • CAPTISOL® is safe when administered parenterally and does not exhibit the nephrotoxicity associated with beta-cyclodextrin. Relative to beta-cyclodextrin, CAPTISOL® provides comparable or higher complexation characteristics and superior water solubility in excess of 90 grams/100 ml—a 50-fold improvement.
  • solubility enhancers were found to match the desired solubility range: DMA, PEG-400, dimethyl-acetamide, polyethylene glycol, polyoxylated castor oil, N-methyl-2-pyrrolidinone, 1-ethenyl-2-pyrrolidinone, Polysorbate 20, Polysorbate 80, Hydroxypropyl- ⁇ -cyclodextrin and Sulfobutylether- ⁇ -cycldextrin (Captisol®).
  • DMA solubility enhancers both cyclodextrins have proven to be superior with respect to solubility, biological activity and stability.
  • Captisol® as the solubility enhancer for use in Example 5 formulations and to further study both cyclodextrin formulations.
  • the final formulation for the Example 5 clinical studies consists of: 120 mg/ml of Captisol® in water with the desired amount of peptide (0.5, 1.0 or 2.5 mg/ml), and HCl and NaOH for pH adjustment.
  • Solubility Enhancers used for Compound 1 formulation development Solubility enhancer classification Solubility Enhancers Solvents Cremophor EL, CMC, Ethanol, DMA, Gycerin, Propylene Glycol, PEG 400, Monotioglycerol Co-solvents Polysorbate 20, Ploysorbate 80 Solubilizing agents Argenine, HSA, Glycine, Creatinine, Glutamic acid, Lysine (acetate salt and free base), Captisol ®, Hydroxypropyl- ⁇ -cyclodextrin, Bulking agents Mannitol, Sorbitol, Dextrose, Lactose Dextran pH Adjustment Agents Citrate buffer, Acetate buffer, Sodium Carbonate
  • Standard dissolution methods such as mixing dry Compound 1 and dry Captisol® into water or adding Compound 1 to a prepared solution of Captisol® and water did not result in complete dissolution at the desired concentrations.
  • concentrations of both Compound 1 and Captisol® were tested at various pH levels. However, the following method for producing a solution of Compound 1 in Captisol® resulted in complete dissolution at the desired concentrations.
  • the current lyophilization process differs from other lyophilization processes in that the percentage of solids in the formulation is high (12%) whereas lyophilized products normally contain between 5 and 10% solids.
  • the freeze drier used was an Edwards lyophilizer Lyoflex 0.6.
  • the equipment IQ/OQ was performed and checked for compliance by quality assurance prior to the process development.
  • Freezing Freezing was from room temperature to ⁇ 40° C. within 2 hours. Shelves were held at ⁇ 40° C. for 3 hours. Drying—Drying was performed at 110 ⁇ bar pressure. Shelf temperature was increased to 20° C. over 13 hours and held at that temperature for additional 13 hours.
  • Drying was performed at 110 ⁇ bar pressure. Shelf temperature was increased to 20° C. over 13 hours and held at that temperature for additional 13 hours (Batch 4) or 8 hours (Batch 5). Pressure was decreased to 10 ⁇ bar for additional 5 hours.
  • the biological activity was monitored by inhibition of IL-2 secretion from Compound 1 reference standard (RS) specific T-cells following subcutaneous (s.c.) treatment with the lyophilized compound solution, i.e. the drug product (DP), at two concentrations.
  • the results of the treatment are compared to those of treating mice with Compound 1 (RS) in phosphate buffered saline (PBS). The results are shown in the tables below and in FIG. 2 .
  • Immunization Day 0 (Compound 1 RS emulsified with CFA, at all four footpads) 2. Treatment Day 0 (s.c. at the back of the neck, in 200 ⁇ l solution) 3. In-vitro activation with: Day 10 a. Compound 1 RS at concentrations of 0; 0.5; 1; 2.5; 5; 10; 25; 50 and 100 ⁇ g/ml b. a peptide with the reverse order of amino acids of Compound 1 (negative control). c. Con A (positive control). 4. Incubation of culture for 20 hrs at 37° C. in a humidified 5% CO 2 incubator. 5. IL-2 measurement by ELISA.
  • mice were immunized with 50 ⁇ g/mouse of Compound 1 RS.
  • the immunized mice were allocated to five treatment groups as follows: placebo, 25, 50, 100 and 200 ⁇ g/mouse of Compound 1 DP (subcutaneous administration).
  • mice 20 female BALB/c mice, supplied by Harlan animals breeding center, Rehovot. Age at immunization (week+days): 10 Average weight of mice included in the experiment: 19.01 gr.
  • 70% ethanol was prepared from 96% ethanol by diluting with purified H 2 O.
  • CFA-Compound 1 RS emulsion 500 ⁇ g/ml, 50 ⁇ g/mouse was prepared as follows:
  • Treatment was by a s.c. injection of 200 ⁇ l solution.
  • Captisol® 1.2 gr were dissolved in 10 ml of WFI to yield a solution of 12% Captisol®.
  • mice were weighed before immunization. Average mice weight: 19.01 ⁇ 0.97 gr
  • the immunization was performed by injecting 100 microliters of the emulsion (50 microliters into each hind footpad).
  • mice were treated by s.c. injection of 200 ⁇ l from the designated Compound 1 DP or 12% Captisol® treatment solutions, at the back of their neck.
  • mice were sacrificed by cervical dislocation. LN were extracted from the hind legs and were transferred to a sterile petri dish containing about 5 mL RPMI. The cells were extracted by gentle squeezing of the tissue against a 200 micrometer mesh stainless steel net. The cells were collected and centrifuged at 300 G for 10 minutes at RT.
  • IFN- ⁇ and TGF- ⁇ secretion of IFN- ⁇ and TGF- ⁇ , as indication of cellular response, were determined by ELISA of culture media (48 hrs for IFN- ⁇ and 72 hrs for TGF- ⁇ ).
  • the 10 ⁇ 10 6 cells/ml suspensions were diluted 1:2 by adding 5 ml EM-1 to 5 ml cells suspension.
  • tissue culture plates were prepared. The following was added to each plate.
  • 96-wells plates were prepared by applying 100 ⁇ l from the cell suspension and 100 ⁇ l from the activation solutions.
  • the culture plates were incubated at 37° C. in a humidified 5% CO 2 incubator, for either 48 or 72 hrs.
  • the cultured plates were centrifuged at 300 g for 10 minutes at RT. Supernatants (850 ⁇ l from each well) were transferred either to mirror plates or to tubes. The supernatant was then divided into working aliquots (two aliquots of 200 and one aliquot of 450 ⁇ l), in order to avoid repeated freeze/thawing of the samples. Each tube was labeled with the following details:
  • the supernatants were stored at ⁇ 20° C. until used for ELISA.
  • Captisol® sulfobutyl ether beta-cyclodextrin sodium
  • mice were followed for anti-dsDNA antibodies and for proteinuria. When the mice were sacrificed, the intensity of ICD was determined in kidneys.
  • Table 13 also shows that the beneficial effect of treatment with Compound 1 could be observed starting from the 5 th injection and it was sustained up to the 10th injection.
  • the mean levels of proteinuria in the Captisol® control group were consistently higher than in the Compound I-treated groups.
  • Table 13 also shows that a reduction in the intensity of ICD was observed in kidneys of both Compound 1 dose groups. There was an overall trend showing that the lower dose (25 ⁇ g/mouse) was more effective than the higher dose (50 ⁇ g/mouse) in reducing the clinical symptoms of SLE in these mice.
  • b The death of one animal with a high level of proteinuria resulted in a lower group mean.
  • c p ⁇ 0.05 (compared to Captisol ®-treated control mice; Mann-Whitney).
  • FIG. 6 shows representative sections of one kidney from each treatment group.
  • the top row sections are from a Captisol®-treated mouse, the mid-row sections are from a mouse treated with 50 ⁇ g/mouse Compound 1 and the bottom row sections are from a mouse treated with 25 ⁇ g/mouse Compound 1. It can be seen that the intensity of immune complex deposits observed in kidney sections of mice treated with Compound 1 (dissolved in Captisol®) at either dose level was much lower than that observed in the control group.
  • SLE patients Thirty-six (36) subjects participated in the study. To be eligible for inclusion in the study, SLE patients must have fulfilled at least four criteria used for the diagnosis of lupus by the American College of Rheumatology. Patients must also have had stable, mild/moderate disease and score less than or equal to 10 on the SLE Disease Activity Index, SLEDAI.
  • a standard battery of safety tests including blood and urine collection for laboratory tests, was performed at screening, during the day of dosing, at 24 hours post-dose and at 2, 4 and 8 weeks following dosing. Prior to dosing, and on scheduled follow-up visits, blood samples were withdrawn for SLE-related immunological tests, anti-Compound 1 antibodies and PBL proliferation assay. The following immunology tests were performed:
  • Compound 1 in Captisol®), Placebo, Water for Injection-Ampoules, Dose and Mode of Administration:
  • Vials of Compound 1 in Captisol® 120 mg/vial were injected subcutaneously as a single dose per subject in the following dosages:
  • Placebo for Compound 1 120 mg Captisol®/vial (identical in appearance to vials of Compound 1 in Captisol®).
  • Blood samples serum and whole blood for safety laboratory tests were withdrawn at Screening, Dosing Day (pre-dose), Day 2 (post dose), at Weeks 2, 4 and 8 (Termination visit). Blood samples for immunological tests were withdrawn at: Screening, Dosing Day (pre-dose) and at Weeks 4 and 8.
  • Eligible subjects for this study were SLE patients who fulfilled at least four diagnostic criteria of the American College of Rheumatology (ACR). Their disease condition had to be stable, mild to moderate with a score equal to or less than 10 on the SLE disease activity index, year 2000 updated (SLEDAI 2K).
  • SLE patients who reported unstable or severe asthma, stroke, acute myocardial infarction, unstable angina, cerebral hemorrhage and pulmonary embolism during the six months prior to study screening. SLE patients who had any clinically significant or unstable medical or surgical conditions, diabetes mellitus, liver disease (cirrhosis, active hepatitis, portal hypertension, and/or ascites), clinically significant hypertension, a medical history of any malignancy, dialysis, or chronic obstructive pulmonary disease (COPD) were also excluded from study participation.
  • COPD chronic obstructive pulmonary disease
  • prednisone 30 mg/day or greater or an equivalent dose of another corticosteroid
  • intravenous corticosteroids or an equivalent dose of another corticosteroid
  • IgG intravenous immunoglobulin G
  • oral anticoagulants e.g. azathioprine, chlorambucil, cyclophosphamide, mycophenolate mofetil, methothrexate, tacrolimus.
  • cytotoxic agents e.g. azathioprine, chlorambucil, cyclophosphamide, mycophenolate mofetil, methothrexate, tacrolimus.
  • corticosteroids more than ⁇ 10 mg/day prednisone, or an equivalent dose of another corticosteroid
  • anti-malarials during the last 3 months prior to screening were excluded from the study.
  • Drug-related immunological responses were followed by using the PBL proliferation assay and anti-Compound 1 antibodies assay at the Dosing Day and at follow-up visits including Termination visit.
  • SLE disease activity index score year 2000 updated (SLEDAI 2K) was assessed at Screening, during the hospitalization and at follow-up visits including Termination visit.
  • Descriptive statistics for vital signs including number of observations, mean, standard deviation, median, minimum and maximum values were determined for Screening, Day 1 (pre and post dose, and at each time point) Day 2, Weeks 2, 4 and 8 are tabulated by the assigned treatment. Changes from baseline to each time point/visit is presented in by visit and treatment assignment.
  • Descriptive statistics including mean, standard deviation, median, minimum and maximum values of SLEDAI 2K are presented.
  • the peripheral blood lymphocytes (PBL) assay showed that 50% of the subjects (18) were classified as responders (SI>2) with similar distribution in all treatment groups.
  • the T cell response was relatively low and no association between Compound 1 treatment dose or concentration used in the assay and responder/non-responder status could be detected, taking into consideration that only a single SC dose of the study drug was administered. Also, no indication of increased incidence, of responder status over time was observed.
  • the tetanus toxoid (TTX) assay that serves as a safety control shows that the response to TTX was preserved throughout the study period in all treatment groups indicating that Compound 1 in Captisol® did not change the immunological response to TTX recall antigen.
  • the immunological findings are the result of the administration of only a single dose of the study drug Compound 1.
  • This study is being performed in order to evaluate the safety and tolerability of repeated Compound 1 sc administration to SLE subjects.
  • the study's secondary objective is to evaluate immunological responses following repeated sc administration of Compound 1 in Captisol® in SLE subjects.
  • Compound 1 is given in doses of 0.5, 1.0 or 2.5 mg in Captisol®.
  • the investigational product is administered every other day (excluding weekends) for a total of 12 sc injections, i.e. 3 doses a week for 4 weeks.
  • Subjects are monitored on planned visits scheduled at 2, 4, 8 and 12 weeks after start of dosing. Safety and tolerability are evaluated using tests similar to those described in the Phase Ia Clinical Study above.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Epidemiology (AREA)
  • Immunology (AREA)
  • Nanotechnology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Molecular Biology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Organic Chemistry (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Biotechnology (AREA)
  • Biophysics (AREA)
  • General Engineering & Computer Science (AREA)
  • Medical Informatics (AREA)
  • Crystallography & Structural Chemistry (AREA)
  • Dermatology (AREA)
  • Transplantation (AREA)
  • Inorganic Chemistry (AREA)
  • Medicinal Preparation (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Peptides Or Proteins (AREA)

Abstract

The subject invention provides a pharmaceutical composition comprising an aqueous carrier; from 0.1 mg/ml to 20 mg/ml of the composition of a pharmaceutically acceptable salt of a peptide having the structural formula

NH 2-Gly Tyr Tyr Trp Ser Trp Ile Arg Gln Pro Pro Gly Lys Gly Glu Glu Trp Ile Gly-COOH;
and a substituted β-cyclodextrin in an amount effective to dissolve the peptide in the aqueous carrier, wherein the composition has a pH between 4 and 9, a process for preparation, and a method of alleviating symptoms of systemic lupus erythematosus (SLE) in a human subject comprising administering to the human subject the pharmaceutical composition.

Description

  • This application claims the benefit of U.S. Provisional Application No. 60/439,950, filed Jan. 14, 2003, the entire contents of which are hereby incorporated by reference.
  • Throughout this application, various publications are referenced by full citations. The disclosures of these publications in their entireties are hereby incorporated by reference into this application in order to more fully describe the state of the art as known to those skilled therein as of the date of the invention described and claimed herein.
  • BACKGROUND OF THE INVENTION
  • Systemic lupus erythematosus (SLE), or lupus, is a debilitating autoimmune disease characterized by the presence of an array of autoantibodies, including antibodies to dsDNA, to nuclear antigens and to ribonucleoproteins. SLE affects approximately 1 in 2000 individuals (U.S. 1 in 700 women). The disease primarily affects young women, with a female-to male ratio of approximately 9:1.
  • Systemic lupus can affect almost any organ or system of the body. Systemic lupus may include periods in which few, if any, symptoms are evident (“remission”) and other times when the disease becomes more active (“flare”). Most often when people mention “lupus,” they are referring to the systemic form of the disease.
  • Corticosteroids are the mainstay in treating systemic autoimmune disorders. Life threatening, severely disabling manifestations of SLE are treated with high doses of glucocorticoids (1-2 mg/kg/day). Undesirable effects of chronic glucocorticoids include an array of prominent adverse effects such as cushingoid habitus, central obesity, hypertension, infection, capillary fragility, hirsutism, accelerated osteoporosis, cataracts, diabetes mellitus, myopathy and psychosis. In addition to corticosteroid toxicity, patient compliance to a dosage regimen also poses a serious problem.
  • Cytotoxic agents are also used for controlling active disease, reducing the rate of disease flares, and reducing steroid requirements. Undesirable side effects of the latter include bone marrow depression, increased infections with opportunistic organisms, irreversible ovarian failure, alopecia and increased risk of malignancy.
  • SLE is an inflammatory disease for which to date there is no definitive treatment or cure. The disease results in acute and chronic complications. The only treatments available are palliative, aimed at relieving acute symptoms and preventing chronic complications, often with profound side effects. There is therefore an unmet need in this field, and both physicians and patients would welcome new treatments which could potentially eliminate or reduce the unwanted manifestations of the disease.
  • Peptides based on the complementarity-determining region of the human monoclonal anti-DNA 16/6Id antibody capable of immunomodulating SLE associated responses have been disclosed in PCT International Publication No. WO 02/067848 A2, the entire contents of which are hereby incorporated by reference. In particular, region CDR1 was found to inhibit the proliferative response of peripheral blood lymphocytes (PBL) of SLE patients to the human anti-DNA 16/6Id mAB, and to ameliorate disease manifestations of mice afflicted with spontaneous or experimental SLE.
  • Human CDR1, Compound 1, shown in FIG. 1, is a synthetic peptide of 19 amino acids based on the complementarity-determining region 1 (CDR1) of the human anti-dsDNA mAb denoted 16/6 Id (Waisman, A., et al. “Modulation of murine systemic lupus erythematosus with peptides based on complementarity determining regions of pathogenic anti-DNA monoclonal antibodies.” Proc. Natl. Acad. Sci. U.S.A. (1997), 94(4): 4620-4625).
  • In experimental SLE models—Balb/c mice and SLE-prone mice, i.e. (NZBxNZW)F1 mice—treatment with either mCDR based-peptides or Compound 1 significantly reduced the SLE related findings, notably immune complex deposits (ICD) in the kidney, proteinuria and leukopenia. The treatment had no effect on the 16/6 Id specific antibody response (Waisman, A., et al. “Modulation of murine systemic lupus erythematosus with peptides based on complementarity determining regions of pathogenic anti-DNA monoclonal antibodies.” Proc. Natl. Acad. Sci. U.S.A. (1997), 94(4): 620; Eilat, E., et al., “Prevention of systemic lupus erythematosus-like disease in (NZBxNZW)F1 mice by treating with CDR1- and CDR3-based peptides of pathogenic autoantibody” J. Clin. Immunol. (2000), 20: 268; Eilat, E., et al., “The mechanism by which a peptide based on complementarity determining region-1 of pathogenic anti-DNA antibody ameliorates experimental SLE” (2001), Proc. Natl. Acad. Sci. U.S.A. 98: 1148).
  • These peptides, like many peptides, are not very soluble. Therefore, formulations that improve the solubility of the peptides are desired.
  • SUMMARY OF INVENTION
  • The subject invention provides a pharmaceutical composition comprising
      • an aqueous carrier;
      • from 0.1 mg/ml to 20 mg/ml of the composition of a pharmaceutically acceptable salt of a peptide having the structural formula
  • (SEQ ID NO:1)
    NH2-Gly Tyr Tyr Trp Ser Trp Ile Arg Gln Pro Pro
    Gly Lys Gly Glu Glu Trp Ile Gly-COOH;
    and
      • a substituted β-cyclodextrin in an amount effective to dissolve the peptide in the aqueous carrier,
      • wherein the composition has a pH between 4 and 9.
  • The subject invention also provides a pharmaceutical composition comprising
      • an aqueous carrier;
      • from 0.1 mg/ml to 20 mg/ml of the composition of an acetate salt of a peptide having the structural formula
  • (SEQ ID NO:1)
    NH2-Gly Tyr Tyr Trp Ser Trp Ile Arg Gln Pro Pro
    Gly Lys Gly Glu Glu Trp Ile Gly-COOH;
      • and
      • from 70 mg/ml to 170 mg/ml of the composition of hepta-(sulfobutyl ether)-β-cyclodextrin,
      • wherein the peptide and the hepta-(sulfobutyl ether)-β-cyclodextrin are dissolved in the aqueous carrier; and
      • wherein the solution has a pH between 6.5 and 8.5.
  • The subject invention also provides a method of alleviating symptoms of systemic lupus erythematosus (SLE) in a human subject comprising administering to the human subject any of the above pharmaceutical compositions in an amount effective to alleviate the symptoms of SLE in the human subject.
  • The subject invention also provides a process for manufacturing the above pharmaceutical composition comprising the steps of:
      • a) preparing a solution of a substituted β-cyclodextrin in an aqueous carrier at a predetermined concentration;
      • b) adding predetermined amount of a pharmaceutically acceptable salt of the peptide NH 2-Gly Tyr Tyr Trp Ser Trp Ile Arg Gln Pro Pro Gly Lys Gly Glu Glu Trp Ile Gly-COOH (SEQ ID NO:1) to the solution of step a);
      • c) adjusting the pH of the solution of step b) until the peptide dissolves in the solution; and
      • d) if necessary, adjusting the pH of the solution of step c) to a pH of 4-9, thereby manufacturing the pharmaceutical composition.
  • The subject invention also provides a process of lyophilizing the above pharmaceutical composition, comprising the steps of:
      • a) lowering the temperature of the pharmaceutical composition to −40° C.;
      • b) holding the temperature at −40° C. for a predetermined time;
      • c) raising the temperature of the solution to 20° C.;
      • d) holding the temperature at 20° C. for a predetermined time; and
      • e) reducing the pressure to 10 μbar, thereby lyophilizing the pharmaceutical composition.
  • The subject invention also provides a process of lyophilizing the above pharmaceutical composition, comprising the steps of:
      • a) lowering the temperature of the pharmaceutical composition to −45° C.;
      • b) holding the temperature at −45° C. for a predetermined time;
      • c) raising the temperature of the solution to −20° C.;
      • d) raising the temperature of the solution to 25° C.; and
      • e) holding the temperature at 25° C. for a predetermined time, thereby lyophilizing the pharmaceutical composition.
    BRIEF DESCRIPTION OF FIGURES
  • FIG. 1. Human CDR1 (Compound 1) as acetate salt—showing the molecular and structural formulas of hCDR1, the amino acid sequence, and physical parameters
  • FIG. 2. IL-2 Secretion from cells taken from mice treated with Compound 1 and Captisol® solution after the cells were subsequently activated with a solution of Compound 1 in PBS.
      • ▪—Compound 1 (RS) 50 μg/mouse
      • ▴—Compound 1 (RS) 200 μg/mouse
      • □— DP 50 μg/mouse
      • Δ—DP 200 μg/mouse
      • —12% Captisol® ampulized
  • FIG. 3. IFN-γ Secretion from cells taken from mice treated with Compound 1 solution after the cells were subsequently activated with a solution of compound 1 in EM-1 (2.5×106 cells/well).
      • ♦—Placebo
      • —Compound 150 μg/mouse (treatment dose)
      • Δ—Compound 1100 μg/mouse (treatment dose)
      • X—Compound 1200 μg/mouse (treatment dose)
  • FIG. 4. IFN-γ Secretion from cells taken from mice treated with Compound 1 solution after the cells were subsequently activated with a solution of compound 1 in EM-1 (5×106 cells/well).
      • ♦—Placebo
      • □—Compound 1 25 μg/mouse
      • Δ—Compound 1 50 μg/mouse
      • X—Compound 1 100 pg/mouse
      • *—Compound 1 200 pg/mouse
  • FIG. 5. Anti-dsDNA antibodies in (NZBxNZW)F1 mice after 10 injections with Compound 1 in Captisol® [OD=Optical Density; Compound 1 (C)=Compound 1 dissolved in Captisol®]
      • □—Placebo
      • ⋄—Compound 150 μg/mouse
      • ∘—Compound 125 μg/mouse
  • FIG. 6. Kidney sections from (NZBxNZW)F1 mice showing intensity of Immune Complex Deposits. The top row sections are from a Captisol®-treated mouse, the mid-row sections are from a mouse treated with 50 μg/mouse Compound 1 and the bottom row sections are from a mouse treated with 25 μg/mouse Compound 1. Magnification: Left: ×100, Right: ×400. FITC immunohistology.
  • DETAILED DESCRIPTION
  • The subject invention provides a pharmaceutical composition comprising
      • an aqueous carrier;
      • from 0.1 mg/ml to 20 mg/ml of the composition of a pharmaceutically acceptable salt of a peptide having the structural formula

  • NH 2-Gly Tyr Tyr Trp Ser Trp Ile Arg Gln Pro Pro Gly Lys Gly Glu Glu Trp Ile Gly-COOH (SEQ ID NO:1); and
      • a substituted β-cyclodextrin in an amount effective to dissolve the peptide in the aqueous carrier,
      • wherein the composition has a pH between 4 and 9.
  • In one embodiment, the concentration of the acetate salt of the peptide is at least 0.5 mg/ml.
  • In one embodiment, the concentration of the salt of the peptide is from 0.5 mg/ml to 10 mg/ml.
  • In another embodiment, the concentration of the salt of the peptide is from 0.5 mg/ml to 2.5 mg/ml.
  • In another embodiment, the concentration of the salt of the peptide is from 2.5 mg/ml to 5 mg/ml.
  • In another embodiment, the concentration of the salt of the peptide is from 5 mg/ml to 7 mg/ml.
  • In another embodiment, the concentration of the salt of the peptide is from 7 mg/ml to 8.5 mg/ml.
  • In another embodiment, the concentration of the salt of the peptide is from 8.5 mg/ml to 10 mg/ml.
  • In another embodiment, the concentration of the salt of the peptide is from 9 mg/ml to 10 mg/ml.
  • In another embodiment, the concentration of the salt of the peptide is from 10 mg/ml to 15 mg/ml.
  • In another embodiment, the concentration of the salt of the peptide is from 15 mg/ml to 20 mg/ml.
  • In another embodiment, the concentration of the salt of the peptide is 1.0 mg/ml.
  • In another embodiment, the concentration of the salt of the peptide is 2.5 mg/ml.
  • In another embodiment, the concentration of the salt of the peptide is 5 mg/ml.
  • In another embodiment, the concentration of the salt of the peptide is 10 mg/ml.
  • In another embodiment, the concentration of the salt of the peptide is 15 mg/ml.
  • In another embodiment, the concentration of the salt is from 0.1 mg/ml to 0.5 mg/ml.
  • In another embodiment, the concentration of the salt is from 0.1 mg/ml to 0.2 mg/ml.
  • In another embodiment, the concentration of the salt is from 0.2 mg/ml to 0.3 mg/ml.
  • In another embodiment, the concentration of the salt is from 0.3 mg/ml to 0.4 mg/ml.
  • In another embodiment, the concentration of the salt is from 0.4 mg/ml to 0.5 mg/ml.
  • In a further embodiment, the composition has a pH between 6.5 and 8.5.
  • In a further embodiment, the composition has a pH between 7.5 and 8.5.
  • In a further embodiment, the composition has a pH between 4 and 5.
  • In a further embodiment, the composition has a pH between 5 and 6.
  • In a further embodiment, the composition has a pH between 6 and 7.
  • In a further embodiment, the composition has a pH between 7 and 8.
  • In a further embodiment, the composition has a pH between 8 and 9.
  • In another embodiment, the pharmaceutically acceptable salt is an acetate salt.
  • In another embodiment, the substituted β-cyclodextrin is a hydroxypropyl, a sulfobutyl ether, or a sulfopropyl ether substituted β-cyclodextrin.
  • In a further embodiment, the substituted β-cyclodextrin is a sulfobutyl ether substituted β-cyclodextrin.
  • In a further embodiment, the pharmaceutically acceptable salt is an acetate salt, and the substituted β-cyclodextrin is hepta-(sulfobutyl ether)-β-cyclodextrin.
  • In another embodiment, the composition further comprises a pharmaceutically acceptable buffer in an amount and of a type suitable to make the pH of the pharmaceutical composition in the range of 4-9. The buffer may be acetate buffer, citrate buffer, or sodium carbonate.
  • The subject invention also provides a pharmaceutical composition comprising
      • an aqueous carrier;
      • from 0.1 mg/ml to 20 mg/ml of the composition of an acetate salt of a peptide having the structural formula
  • (SEQ ID NO:1)
    NH2-Gly Tyr Tyr Trp Ser Trp Ile Arg Gln Pro Pro
    Gly Lys Gly Glu Glu Trp Ile Gly-COOH;
      • and
      • from 70 mg/ml to 170 mg/ml of the composition of hepta-(sulfobutyl ether)-β-cyclodextrin,
      • wherein the peptide and the hepta-(sulfobutyl ether)-β-cyclodextrin are dissolved in the aqueous carrier; and
      • wherein the composition has a pH between 6.5 and 8.5.
  • In one embodiment, the concentration of the acetate salt of the peptide is at least 0.5 mg/ml.
  • In one embodiment, the concentration of the acetate salt of the peptide is from 0.5 mg/ml to 10 mg/ml.
  • In a further embodiment, the concentration of the acetate salt of the peptide is from 0.5 mg/ml to 2.5 mg/ml.
  • In another embodiment, the concentration of the salt is from 0.1 mg/ml to 0.5 mg/ml.
  • In another embodiment, the concentration of the salt is from 0.1 mg/ml to 0.2 mg/ml.
  • In another embodiment, the concentration of the salt is from 0.2 mg/ml to 0.3 mg/ml.
  • In another embodiment, the concentration of the salt is from 0.3 mg/ml to 0.4 mg/ml.
  • In another embodiment, the concentration of the salt is from 0.4 mg/ml to 0.5 mg/ml.
  • In another embodiment, the concentration of the salt of the peptide is from 5 mg/ml to 7 mg/ml.
  • In another embodiment, the concentration of the salt of the peptide is from 7 mg/ml to 8.5 mg/ml.
  • In another embodiment, the concentration of the salt of the peptide is from 8.5 mg/ml to 10 mg/ml.
  • In another embodiment, the concentration of the salt of the peptide is from 9 mg/ml to 10 mg/ml.
  • In another embodiment, the concentration of the salt of the peptide is from 10 mg/ml to 15 mg/ml.
  • In another embodiment, the concentration of the salt of the peptide is from 15 mg/ml to 20 mg/ml.
  • In a further embodiment, the concentration of acetate salt of the peptide is 1.0 mg/ml.
  • In a further embodiment, the concentration of acetate salt of the peptide is 2.5 mg/ml.
  • In another embodiment, the concentration of the salt of the peptide is 5 mg/ml.
  • In another embodiment, the concentration of the salt of the peptide is 10 mg/ml.
  • In another embodiment, the concentration of the salt of the peptide is 15 mg/ml.
  • In another embodiment, the concentration of hepta-(sulfobutyl ether)-β-cyclodextrin is 120 mg/ml and the pH of the composition is between 7.5 and 8.5.
  • The subject invention also provides a method of alleviating symptoms of systemic lupus erythematosus (SLE) in a human subject comprising administering to the human subject any of the above pharmaceutical compositions in an amount effective to alleviate the symptoms of SLE in the human subject.
  • The subject invention also provides the above pharmaceutical compositions for use in treating SLE in a human subject.
  • The subject invention also provides a process for manufacturing any of the above pharmaceutical compositions comprising the steps of:
      • a) preparing a solution of a substituted β-cyclodextrin in an aqueous carrier at a predetermined concentration;
      • b) adding predetermined amount of a pharmaceutically acceptable salt of the peptide NH 2-Gly Tyr Tyr Trp Ser Trp Ile Arg Gln Pro Pro Gly Lys Gly Glu Glu Trp Ile Gly-COOH (SEQ ID NO:1) to the solution of step a);
      • c) adjusting the pH of the solution of step b) until the peptide dissolves in the solution; and
      • d) if necessary, adjusting the pH of the solution of step c) to a pH of 4-9, thereby manufacturing the pharmaceutical composition.
  • In one embodiment of the process, the resulting final concentration of the substituted β-cyclodextrin in the pharmaceutical composition is from 70 mg/ml to 170 mg/ml.
  • In one embodiment of the process, the predetermined concentration of the substituted β-cyclodextrin is such which results in a final concentration of substituted β-cyclodextrin in the pharmaceutical composition of from 80 mg/ml to 170 mg/ml.
  • In one embodiment of the process, the predetermined concentration of the substituted β-cyclodextrin is such which results in a final concentration of substituted β-cyclodextrin in the pharmaceutical composition of from 90 mg/ml to 170 mg/ml.
  • In one embodiment of the process, the predetermined concentration of the substituted β-cyclodextrin is such which results in a final concentration of substituted β-cyclodextrin in the pharmaceutical composition of from 100 mg/ml to 170 mg/ml.
  • In one embodiment of the process, the predetermined concentration of the substituted β-cyclodextrin is such which results in a final concentration of substituted β-cyclodextrin in the pharmaceutical composition of from 110 mg/ml to 170 mg/ml.
  • In one embodiment of the process, the predetermined concentration of the substituted β-cyclodextrin is such which results in a final concentration of substituted β-cyclodextrin in the pharmaceutical composition of from 120 mg/ml to 170 mg/ml.
  • In one embodiment of the process, the predetermined, concentration of the substituted β-cyclodextrin is such which results in a final concentration of substituted β-cyclodextrin in the pharmaceutical composition of from 0.130 mg/ml to 170 mg/ml.
  • In one embodiment of the process, the predetermined concentration of the substituted β-cyclodextrin is such which results in a final concentration of substituted β-cyclodextrin in the pharmaceutical composition of from 140 mg/ml to 170 mg/ml.
  • In one embodiment of the process, the predetermined concentration of the substituted β-cyclodextrin is such which results in a final concentration of substituted β-cyclodextrin in the pharmaceutical composition of from 150 mg/ml to 170 mg/ml.
  • In one embodiment of the process, the predetermined concentration of the substituted β-cyclodextrin is such which results in a final concentration of substituted β-cyclodextrin in the pharmaceutical composition of from 160 mg/ml to 170 mg/ml.
  • In one embodiment of the process, the predetermined concentration of the substituted β-cyclodextrin is such which results in a final concentration of substituted β-cyclodextrin in the pharmaceutical composition is 120 mg/ml.
  • In another embodiment, the predetermined amount of peptide is such which results in a final concentration of peptide in the pharmaceutical composition is at least 0.1 mg/ml.
  • In another embodiment, the predetermined amount of peptide is such which results in a final concentration of peptide in the pharmaceutical composition is at least 0.5 mg/ml.
  • In another embodiment, the predetermined amount of peptide is such which results in a final concentration of peptide in the pharmaceutical composition is 2.5 mg/ml, 2.0 mg/ml, 11.0 mg/ml, 0.5 mg/ml or 0.1 mg/ml.
  • In another embodiment, the predetermined amount of peptide is such which results in a final concentration of peptide in the pharmaceutical composition is 5 mg/ml, 10 mg/ml or 15 mg/ml.
  • In another embodiment of the process, step b) further comprises mixing the solution for 1 hour.
  • In another embodiment, in step c) the pH is adjusted using HCl or NaOH 1.0N.
  • In another embodiment, the process further comprises filtering the solution of step d) through a cellulose acetate filter.
  • In another embodiment of the above process,
      • the predetermined concentration of the substituted β-cyclodextrin is such which results in a final concentration of substituted β-cyclodextrin in the pharmaceutical composition is 120 mg/ml;
      • the predetermined amount of peptide is such which results in a final concentration of peptide in the pharmaceutical composition is 2.5 mg/ml, 2.0 mg/ml, 11.0 mg/ml, 0.5 mg/ml or 0.1 mg/ml;
      • step b) further comprises mixing the solution for 1 hour; and
      • in step c) the pH is adjusted using HCl or NaOH 1.0N, and the process further comprises filtering the solution of step d) through a cellulose acetate filter.
  • The subject invention also provides a composition prepared by the above process.
  • The subject invention also provides a process of lyophilizing the above pharmaceutical composition, comprising the steps of:
      • a) lowering the temperature of the pharmaceutical composition to −40° C.;
      • b) holding the temperature at −40° C. for a predetermined time;
      • c) raising the temperature of the solution to 20° C.;
      • d) holding the temperature at 20° C. for a predetermined time; and
      • e) holding the temperature at 25° C. for a predetermined time, thereby lyophilizing the pharmaceutical composition.
  • In one embodiment of the process, step a) is performed within 2 hours.
  • In another embodiment, step b) is performed within 3 hours.
  • In a further embodiment, step c) is performed over 13 hours.
  • In a further embodiment, step c) is performed at a pressure of 110 μbar.
  • In a further embodiment, step d) is performed over 13 hours.
  • In a further embodiment, step d) is performed at a pressure of 110 μbar.
  • In a further embodiment, in step e) the pressure is reduced to 10 μbar.
  • In a further embodiment, step e) is performed over 5 hours.
  • In another embodiment of the process,
      • step a) is performed within 2 hours;
      • step b) is performed within 3 hours;
      • step c) is performed over 13 hours and at a pressure of 110 μbar;
      • step d) is performed over 13 hours and at a pressure of 110 μbar; and
      • step e) is performed over 5 hours and the pressure is reduced to 10 μbar.
  • The subject invention also provides a lyophilized pharmaceutical composition prepared by the above process.
  • The subject invention also provides a process of lyophilizing the above pharmaceutical composition, comprising the steps of:
      • a) lowering the temperature of the pharmaceutical composition to −45° C.;
      • b) holding the temperature at −45° C. for a predetermined time;
      • c) raising the temperature of the solution to −20° C.;
      • d) raising the temperature of the solution to 25° C.; and
      • e) holding the temperature at 25° C. for a predetermined time, thereby lyophilizing the pharmaceutical composition.
  • In one embodiment, step a) is performed within 6 hours.
  • In another embodiment, step b) is performed within 3 hours.
  • In another embodiment, step c) is performed over 19 hours.
  • In another embodiment, step c) is performed at a pressure of 150 μbar.
  • In another embodiment, step d) is performed over 13 hours.
  • In another embodiment, step d) is performed at a pressure of 150 μbar.
  • In another embodiment, step e) is performed over 8 hours.
  • In another embodiment, step e) is performed at a pressure of 150 μbar.
  • In another embodiment of the process,
      • step a) is performed within 6 hours;
      • step b) is performed within 3 hours;
      • step c) is performed over 19 hours and at a pressure of 150 μbar;
      • step d) is performed over 13 hours and at a pressure of 150 μbar; and
      • step e) is performed over 8 hours and at a pressure of 150 μbar.
  • The subject invention also provides a lyophilized pharmaceutical composition prepared by any of the above processes.
  • In one embodiment of the above lyophilized pharmaceutical composition, the water content of the composition is less than 5%.
  • In another embodiment, the water content of the composition is less than 4.0%.
  • In another embodiment, the water content of the composition is less then 3.5%.
  • The subject invention also provides a lyophilized pharmaceutical composition comprising
      • a pharmaceutically acceptable salt of a peptide having the structural formula
  • (SEQ ID NO:1)
    NH2-Gly Tyr Tyr Trp Ser Trp Ile Arg Gln Pro Pro
    Gly Lys Gly Glu Glu Trp Ile Gly-COOH;
    and
      • a substituted β-cyclodextrin.
  • The subject invention also provides a packaged pharmaceutical composition comprised of:
      • a packaging material; and
      • a predetermined amount of the above lyophilized pharmaceutical composition.
  • The preparations of the present invention may be given parenterally, topically, or rectally. They are of course given by forms suitable for each administration route. For example, they are administered by injection, inhalation, ointment, suppository, etc. administration by injection, infusion or inhalation; topical by lotion or ointment; and rectal by suppositories.
  • The phrases “parenteral administration” and “administered parenterally” as used herein means modes of administration other than enteral and topical administration, usually by injection, and includes, without limitation, intravenous, intramuscular, intraarterial, intrathecal, intracapsular, intraorbital, intracardiac, intradermal, intraperitoneal, transtracheal, subcutaneous, subcuticular, intraarticular, subcapsular, subarachnoid, intraspinal and intrasternal injection and infusion.
  • The phrases “systemic administration,” “administered systematically,” “peripheral administration” and “administered peripherally” as used herein mean the administration of a compound, drug or other material other than directly into the central nervous system, such that it enters the patient's system and, thus, is subject to metabolism and other like processes, for example, subcutaneous administration.
  • Details of general formulation procedures and information on additional excipients may be found in Remington: The Science and Practice of Pharmacy, 20th Edition.
  • This invention will be better understood from the Experimental Details which follow. However, one skilled in the art will readily appreciate that the specific methods and results discussed are merely illustrative of the invention as described more fully in the claims which follow thereafter.
  • EXPERIMENTAL DETAILS Example 1 Formulation Development for Compound 1
  • The human hCDR1 peptide (Compound 1) is described in PCT International Publication No WO 02/067848, published Sep. 6, 2002, and can be prepared by methods well known in the art, (see, for example, Peptides: Synthesis, Structure and Applications, ed. by B. Gutte, Academic Press, 1995; Peptide Synthesis Protocols, ed. By M. Pennington and B. Dunn, Humana Press, 1994; Schnolzer, M. et al., “In situ neutralization in Boc-chemistry solid phase synthesis. Rapid, High yield assembly of difficult sequences.” Int. J. Pept. Protein Res. (1992) 40: 180-193).
  • Compound 1 is a synthetic polypeptide composed of 19 amino acids. It is provided as an acetate salt. The aqueous solubility of the peptide has been determined to be less than 0.5 mg/ml. FIG. 1 shows compound 1 as an acetate salt.
  • In order to develop a formulation with peptide concentration exceeding 2 mg/ml, preferably up to 10 mg/ml, experiments with several solubility enhancers were performed. The preliminary experiments indicated that a concentration of 2 mg/ml cannot be easily attained. In order to develop a formulation for sub-cutaneous injection, it is also desirable that the pH be in the range of 4 to 9 and that the solution be iso-osmotic.
  • Based on an extensive literature survey, a few principal approaches were adopted in order to produce a formulation with maximal solubility. The factors considered were:
  • pH adjustment and buffers
  • Solvents Co-solvents
  • Solubilizing agents
  • Methods
  • Compound 1 was dissolved in the chosen solubility enhancer solution either separately or in combination with other excipients and the solutions were stirred for at least an hour. The pH was adjusted if needed. The solutions were visually examined to estimate the solubility and sent for analytical assay determination. For a few chosen formulations, biological activity was also tested.
  • Results
  • Table 1 presents the type of solubility enhancers used for the formulation development. Tables 2 and 3 summarize the experiments that were performed with the various solubility enhancers. Table 2 summarizes the initial screening performed with peptide concentrations in the range of 5 to 10 mg/ml. The experimental work that was performed with higher peptide concentration was then repeated with the lower doses (see table 3)
  • Initial tests indicated that Compound 1 was more soluble at the limits of the desired pH levels, both acidic and basic, but was less stable at the basic pH range. Thus, several buffers and pH adjustment agents were tested, including acetate buffer, citrate buffer and sodium carbonate. None of the initially tested buffers achieved the desired peptide solubility level. Only above pH 9.2 and below pH 3.0 were solubility levels of 2 mg/ml observed. Nevertheless, at the initial stage, formulations with acetate buffer and citrate buffer (with Mannitol as a tonicity agent) were selected for initial toxicology studies. These formulations were tested for biological activity and proven active.
  • Non aqueous solvents (see table 1) such as Ethanol, Glycerin, Propylene glycol, Chremophore and their combinations were tested but did not increase the solubility of Compound 1. A solution of 30% DMA (dimethyl-acetamide) yielded solubility in the desired ranges (5 to 9 mg/ml), but was not suitable for a pharmaceutical formulation due to its toxicity profile. Improved solubility was also observed using 30% (w/w) PEG 400 (5 to 9 mg/ml). This latter formulation was chosen for the toxicology studies, but it has proved to be both inactive in the biological assay, and may have been the cause of some adverse effects in a mouse toxicity study. Thus, it was decided not to further pursue this formulation. In view of the preliminary experiments non-aqueous solvents were not used in the subject formulations.
  • Several amino acids (see table 1) including L-Arginine, L-Glutamic acid, L-Glycine and L-Lysine were tested to improve the protein solubility. The solubility of the peptide in L-Arginine was at the desired level but the resulting pH was above 9. An attempt to decrease the pH or use an Arginine HCl salt resulted in precipitation of the peptide. Human Serum Albumin was also tested and improved the solubility of the peptide at low peptide concentrations (1 mg/ml) (see table 3). However, due to its potential immunogenicity and the low peptide solubility, it was not utilized in further experiments.
  • Bulking agents (see table 1) including Mannitol, Sorbitol and Dextran were tested alone and in combination with other excipients, but did not improve the solubility of the peptide in solution.
  • Co-solvents (see table 1), including Polysorbate 20 and Polysorbate 80 were tested alone and in combination with other excipients. While lower concentrations of Polysorbates (up to 6%) did not improve the solubility of the peptide, higher concentrations (up to 10%—see table 2) improved the solubility of the peptide up to 2 mg/ml. However, such high concentrations of Polysorbates were deemed unsuitable for pharmaceutical formulations.
  • Two types of cyclodextrins, both approved for use in marketed parenteral products, were also tested: Hydroxypropyl-β-cyclodextrin and Sulfobutylether-β-cyclodextrin (Captisol®). Both markedly increased the solubility of the peptide (concentrations in the levels of 10 mg/ml for Hydroxypropyl-β-cyclodextrin and 2.5 for Captisol®). The biological activity of the two cyclodextrin formulations was tested and was found to be equal to the activity of the peptide alone.
  • CAPTISOL® is a commercially available polyanionic β-cyclodextrin derivative with a sodium sulfonate salt separated from the hydrophobic cavity by a butyl ether spacer group, or sulfobutylether (SBE). CAPTISOL® is the trade name for CyDex Inc.'s hepta-substituted sulfobutylether β-cyclodextrin (SBE7-β-CD) preparation (www.captisol.com). The structure of CAPTISOL® allows drug molecules to fit in the hydrophobic cavity, thereby isolating the drug molecule from the aqueous solvent. Because the outer surface of CAPTISOL® is hydrophilic, the solubility of the complexed drug molecule is thereby enhanced. The use of cyclodextrins to enhance the solubility of drug molecules is disclosed in U.S. Pat. Nos. 5,134,127 and 5,376,645, the entire contents of which are hereby incorporated by reference.
  • According to the literature of CyDex Inc., CAPTISOL® is safe when administered parenterally and does not exhibit the nephrotoxicity associated with beta-cyclodextrin. Relative to beta-cyclodextrin, CAPTISOL® provides comparable or higher complexation characteristics and superior water solubility in excess of 90 grams/100 ml—a 50-fold improvement.
  • Conclusions
  • Several solubility enhancers were found to match the desired solubility range: DMA, PEG-400, dimethyl-acetamide, polyethylene glycol, polyoxylated castor oil, N-methyl-2-pyrrolidinone, 1-ethenyl-2-pyrrolidinone, Polysorbate 20, Polysorbate 80, Hydroxypropyl-β-cyclodextrin and Sulfobutylether-β-cycldextrin (Captisol®). Of these solubility enhancers both cyclodextrins have proven to be superior with respect to solubility, biological activity and stability. Thus, it was decided to select Captisol® as the solubility enhancer for use in Example 5 formulations and to further study both cyclodextrin formulations. The final formulation for the Example 5 clinical studies consists of: 120 mg/ml of Captisol® in water with the desired amount of peptide (0.5, 1.0 or 2.5 mg/ml), and HCl and NaOH for pH adjustment.
  • TABLE 1
    Solubility enhancers used for Compound 1 formulation
    development
    Solubility enhancer
    classification Solubility Enhancers
    Solvents Cremophor EL, CMC, Ethanol, DMA,
    Gycerin, Propylene Glycol, PEG 400,
    Monotioglycerol
    Co-solvents Polysorbate
    20, Ploysorbate 80
    Solubilizing agents Argenine, HSA, Glycine, Creatinine,
    Glutamic acid, Lysine (acetate salt
    and free base), Captisol ®,
    Hydroxypropyl-β-cyclodextrin,
    Bulking agents Mannitol, Sorbitol, Dextrose,
    Lactose Dextran
    pH Adjustment Agents Citrate buffer, Acetate buffer,
    Sodium Carbonate
  • TABLE 2
    List of Cosolvents and Stabilizers evaluated in Compound 1 Peptide Formulations.
    % of Standard Amount of
    amount from peptide
    Solubility Enhancer* % Used the literature added (mg/ml) Assay, % pH of formulation Remarks
    Albumin (HSA), 1.5 0.4-5.0 5 6.0 Insoluble
    Dextrose 1.5 Adjust. to 4.1
    Albumin (HSA), 1.0 0.4-5.0 5 5.8 Insoluble
    Polysorbate 80, 0.6 0.8-4.0 Adjust. to 4.1
    Glycine 2.0 0.2-2.1
    Arginine 1.5 0.8-1.6 15 93 9.8 Clear solution
    Arginine HCl 2.0 0.8-1.6 5 3.5 Insoluble
    Arginine 1.5 0.8-1.6 15 93 9.8 When the pH was lowered
    Lactose 1.5 below 8.5 the peptide
    precipitated and the solution
    turned into gel
    Captisol ® 10.0 Up to 30.0 10 86 4.9 Turbid solution
    20.0 89 Adjust. to 4.4
    CMC (carboxy methyl 0.2 5 90 5.0 For toxicology studies
    cellulose) in acetate 0.05M
    Creatanine 0.8 Up to 0.6  5 6.1 Insoluble
    Adjust. to 4.1
    Cremophor EL 15.0 −10.0  5 4.0 Very turbid
    Ethanol 10.0  0.6-32.9
    Dimethylacetamide 6.0 0.012-6.0 
    Dextran 4.0 to 15.0  3.0-30.0 5 3.9 Insoluble
    Dimethylacetamide (DMA) 6.0-20.0 0.012-6.0  5 4.6 Insoluble
    Dimethylacetamide (DMA) 25.0 0.012-6.0  5 87 5.1 Clear solution
    Dimethylacetamide (DMA) 30.0 0.012-6.0  10 93 5.1 Clear solution
    Ethanol 10.0  0.6-32.9 5 Insoluble
    Glutamic acid 2.0 5 3.7 When the pH was increased
    above 4 the peptide
    precipitated and the
    solution turned into gel
    Glycerin 1.5  1.6-32.5 5 37 4.5 Insoluble
    Glycerin 30.0  1.6-32.5 5 3.7 Insoluble
    Glycerin, 10.0  1.6-32.5 5 12 6.5 Insoluble
    Polysorbate 80 0.6 0.8-4   Adjust. to 4.5
    Glycine 0.4 0.2-2.1 5 4.6 Insoluble
    Hydroxypropyl β-cyclodextrin 20.0 Up to 30.0 10 99 4.6 Clear solution
    Lysine Acetate Salt 2.0 5 3.8 Insoluble
    Lysine Free base 2.0 5 9.2 When the pH was lowered
    below 8 the peptide
    precipitated and the
    solution turned into gel
    Mannitol in Citrate buffer 4.0  2.0-10.0 5 38 3.4 For toxicology studies
    0.035M 
    Mannitol 4.0  2.0-10.0 5 32 4.3 For toxicology studies
    in acetate buffer 0.05M
    Mannitol, 20.0  2.0-10.0 5 14 6.4 Insoluble
    Glycine 0.4 0.2-2.1 Adjust. to 4.5
    Mannitol, 20.0  2.0-10.0 5 22 6.5 Insoluble
    Polysorbate 20 0.6 Adjust. to 4.5
    Monothioglycerol 1.0  0.1-10.0 5 4.5 Turbid solution
    PEG 400 30.0 Up to 30.0 5 88 4.2 Slightly opalescent
    PEG 400 30.0 Up to 30.0 10 89 4.2 Turbid solution
    PEG 400 30.0 Up to 30.0 5 94 4.2 Clear solution
    with DMA 6.0 0.012-0.6 
    PEG 400 10.0  6.0-18.0 5 58 4.2 Insoluble
    PEG 400 10.0 Up to 30.0 5 4.3 Insoluble
    DMA 10.0 0.012-0.6 
    PEG 400 10.0 Up to 30.0 5 4.1 Insoluble
    Propylene glycol PG 10.0 10.0
    PEG 400 18.0 Up to 30.0 5 100  4.2 Clear solution
    Propylene glycol 50.0 10.0
    Polysorbate 80 1.6 0.8-4.0 5 24 7.2 Insoluble
    Adjust. to 4.5
    Polysorbate 80 6.0 0.8-4.0 5 3.9 Insoluble
    Polysorbate 80 6.0 0.8-4.0 5 4.0 Insoluble
    Creatanine 0.6 up to 0.6 
    Propylene glycol PG 10.0 10.0 5 4.2 Insoluble
    DMA 10.0 0.012-0.6 
    Propylene glycol PG 10.0, 30.0 10.0 5 4.2 Insoluble
    Sodium Carbonate 1.5 5 11.4  When the pH was lowered
    below 8.5 the peptide
    precipitated and the
    solution turned into gel
    Sorbitol 5.0 10.0-25.0 5 6.9 Turbid solution
    Adjust. to 4.5
  • TABLE 3
    Compound 1 formulations at low peptide concentrations
    % of Standard Amount of
    amount from the peptide added pH of
    Solubility Enhancer* % Used literature (mg/ml) Assay, % formulation Remarks
    Albumin (HSA), 5.0 0.4-5.0 1.0 90 6.9 Clear solution
    2.5 Adjust. to 4.5 Turbid solution
    Arginine 1.5 0.8-1.6 1.0 24 10.6  When the pH was lowered below 8.5 the peptide
    Adjust. to 8.5 precipitated and the solution turned into gel
    Captisol ® 12.0 Up to 30.0 1.0 106 5.3 Clear solution
    2.5 100 6.5 to 8.5
    Dextran 20.0  3.0-30.0 1.0 69 4.8 Turbid solution
    Adjust. to 4.0
    Glycerin 30.0  1.6-32.5 1.0 4.8 Turbid solution
    Adjust. to 4.0
    Mannitol 4.0 0.8-4.0 1.0 64 4.7 Turbid solution
    Adjust. to 4.0
    Polysorbate 20 10.0 1.0 95 5.8 Clear solution
    2.5 88 Adjust. to 4.8 Clear with small amount of precipitation
    Polysorbate
    20 10.0 2.5 115 5.1 Clear solution
    Mannitol 2.0  2.0-10.0 Adjust. to 4.3
    Polysorbate 80 4.0 0.8-4.0 1.0 91 5.5 Clear solution
    6.0-10.0 2.5 89 Adjust. to 5.0 Slightly turbid solution
    Polysorbate
    80 4.0 0.8-4.0 2.5 88 5.1 Slightly turbid solution
    Mannitol 2.0  2.0-10.0 Adjust. to 4.4
    Propylene glycol PG 10.0 10.0 1.0 78 5.0 Turbid solution
    Adjust. to 4.4
    Sorbitol 20.0 10.0-25   1.0 52 4.5 Turbid solution
  • Example 2 Preparation Protocol for Solution of Compound 1 in Captisol®
  • Standard dissolution methods, such as mixing dry Compound 1 and dry Captisol® into water or adding Compound 1 to a prepared solution of Captisol® and water did not result in complete dissolution at the desired concentrations. Several different concentrations of both Compound 1 and Captisol® were tested at various pH levels. However, the following method for producing a solution of Compound 1 in Captisol® resulted in complete dissolution at the desired concentrations.
  • Materials: Captisol®, Compound 1 and water
  • Method:
      • 1. Weigh the appropriate amount of Captisol® to give a final concentration of 120 mg/ml.
      • 2. Add 80% of the final amount of water and mix for 10 minutes with a magnetic stirrer.
      • 3. Weigh Compound 1 to give a final concentration of 2.5 mg/ml, 2.0 mg/ml, 1.0 mg/ml, 0.5 mg/ml or 0.1 mg/ml.
      • 4. Add the peptide to the Captisol® solution. Mix for 1 hour.
      • 5. Raise the pH to obtain clear solution (in the 2.0 mg/ml formulation there might be a need to raise the pH slightly above 9). pH should be adjusted using HCl 1.0 N and NaOH 1.0 N. Mix for 10 minutes.
      • 6. Correct the pH to the range of 7.5 to 8.5 if needed (using either HCl or NaOH 1.0 N).
      • 7. Add water to final volume.
      • 8. Filter the solution through a 0.2μ cellulose acetate filter.
      • 9. Record final pH.
      • 10. Dispense into aliquots and store at the proper temperature.
    Example 3 Lyophilization of Compound 1 and Captisol® Solution
  • The current lyophilization process differs from other lyophilization processes in that the percentage of solids in the formulation is high (12%) whereas lyophilized products normally contain between 5 and 10% solids.
  • Equipment
  • The freeze drier used was an Edwards lyophilizer Lyoflex 0.6. The equipment IQ/OQ was performed and checked for compliance by quality assurance prior to the process development.
  • Solutions of Compound 1 and Captisol® at concentrations of 0.5 mg/ml, 11.0 mg/ml and 2.5 mg/ml of Compound 1 were prepared. The fill-volume was adjusted 1 ml (1.05 gr).
  • Main Process Steps: 1. Freezing
  • 2. Holding (at low temperature)
    3. Drying under vacuum in two stages:
      • 3.1. Primary drying—shelf warming to an upper hold temperature, controlling shelf temperature at the upper hold level.
      • 3.2. Secondary drying—Pressure reduction to a minimal value at the upper hold shelf temperature.
    Batches 1-3
  • Freezing—Freezing was from room temperature to −40° C. within 2 hours. Shelves were held at −40° C. for 3 hours.
    Drying—Drying was performed at 110 μbar pressure. Shelf temperature was increased to 20° C. over 13 hours and held at that temperature for additional 13 hours.
  • Total process time was 31 hours.
  • Results:
  • Water content results were:
  • Batch no. 1: 3.8% Batch no. 2: 4.0% and Batch no. 3: 4.9% Batches 4 and 5
  • Since the water content results of the processes leading to batches 1, 2 and 3 were higher then the desired value, it was decided to add a secondary drying step at the same temperature and at low pressure.
  • Drying—Drying was performed at 110 μbar pressure. Shelf temperature was increased to 20° C. over 13 hours and held at that temperature for additional 13 hours (Batch 4) or 8 hours (Batch 5). Pressure was decreased to 10 μbar for additional 5 hours.
  • Total process time was 36 hours.
  • Results:
  • Water content results were
  • Batch 4: Placebo: 3.0%,
      • 1 mg/ml: 3.9%.
    Batch 5: Placebo: 4.1% Conclusions
  • As shown, a satisfactory lyophilization process for Compound 1 with Captisol® was developed. Due to the high percentage of solids and hence the condensed cake, the developed process is longer then the currently available lyophilization cycles for peptides and it exhibits an additional secondary drying stage. Table 4 summarizes the developed process.
  • TABLE 4
    Compound 1
    (Peptide) with
    Step Captisol ®
    Loading C.
    Freezing
    2 hours to −40° C.
    Hold at low temp. 3 hours to −40° C.
    Primary Drying:
    Warm to 20° C. 13 hours pressure
    110 μbar
    Hold at 20° C. 13 hours pressure
    110 μbar
    Secondary drying:
    Hold at 20° C. 5 hours pressure
    10 μbar
    Storage at −20° C.
    Process time 36 hours
  • Example 4 Examination of the In-Vivo Biological Activity of the Lyophilized Compound Solution (DP, 1 Mg/Vial, 12% Captisol®)
  • The biological activity was monitored by inhibition of IL-2 secretion from Compound 1 reference standard (RS) specific T-cells following subcutaneous (s.c.) treatment with the lyophilized compound solution, i.e. the drug product (DP), at two concentrations. The results of the treatment are compared to those of treating mice with Compound 1 (RS) in phosphate buffered saline (PBS). The results are shown in the tables below and in FIG. 2.
  • Experimental Design:
  • 1. Immunization Day 0
    (Compound 1 RS emulsified with CFA,
    at all four footpads)
    2. Treatment Day 0
    (s.c. at the back of the neck,
    in 200 μl solution)
    3. In-vitro activation with: Day 10
    a. Compound 1 RS at concentrations of 0; 0.5;
    1; 2.5; 5; 10; 25; 50 and 100 μg/ml
    b. a peptide with the reverse order of amino
    acids of Compound 1 (negative control).
    c. Con A (positive control).
    4. Incubation of culture for 20 hrs at 37° C.
    in a humidified 5% CO2 incubator.
    5. IL-2 measurement by ELISA.
  • Table of experimental Groups:
    Immunization
    Group with Treatment
    A
    50 μg  50 μg Compd. 1 RS in PBS
    B Compound
    1 RS 200 μg Compd. 1 RS in PBS
    C
     50 μg DP(batch 2)
    D 200 μg DP (batch 2)
    F Placebo (12% captisol ®)

    IL-2 Secretion from Compound 1 (DP) Treated Mice Following In-Vitro Activation with Compound 1 RS (pg/ml)
  • Treated with:
    Group
    A B
    Compd. 1 Compd. 1 C D
    F RS RS DP DP
    Concentration of 12% captisol ® 50 μg/ % 200 μg/ % 50 μg/ % 200 μg/ %
    Activator activator (μg/ml) Ampulized mouse inhib. mouse inhib. mouse inhib. mouse inhib.
    Con A 2.5 5,825 6,215 5,403 3,537 4,069
    Compd. 1 RS 0 BQL BQL BQL BQL BQL
    Compd. 1 RS 0.5 11 9 10 8 BQL
    Compd. 1 RS 1 10 BQL BQL BQL BQL
    Compd. 1 RS 2.5 15 8 51 BQL NA 6 61 6 62
    Compd. 1 RS 5 20 9 55 8 60 10 48 7 63
    Compd. 1 RS 10 25 16 38 11 58 13 48 8 67
    Compd. 1 RS 25 29 15 48 11 63 16 45 13 56
    Compd. 1 RS 50 40 21 47 15 62 20 50 12 69
    Compd. 1 RS 100 42 25 41 18 58 24 43 15 64
    Average inhibition (%) 45.6 60.4 46.8 63.7
    (at the range of 5-100 μg/ml)
    BQL = Below Quantitation Limit
    Rows 1-4 were not included in the curve
    NA = Not Applicable
  • Example 5 Evaluation of Optimal Dose for Treatment
  • The following abbreviations are used in the following description:
      • CFA Complete Freund's adjuvant
      • Con A Concanavalin A
      • DP Drug Product
      • DS Drug Substance
      • EM-1 Enriched DCCM-1 Medium
      • EM-3 Enriched RPMI-1640+ fetal calf serum medium
      • FCS Fetal Calf Serum
      • IFN-γ Interferon-gamma
      • LN Lymph Node
      • PBS Phosphate Buffered Saline
      • RS Reference Standard
      • s.c. Subcutaneous
      • TB Trypan Blue
      • TGF-β Transforming Growth Factor-beta
      • WFI Water for Injection
    Introduction
  • A group of 20 mice were immunized with 50 μg/mouse of Compound 1 RS. The immunized mice were allocated to five treatment groups as follows: placebo, 25, 50, 100 and 200 μg/mouse of Compound 1 DP (subcutaneous administration). Ten days post immunization and treatment, LN was extracted and single cell suspension was prepared. The in-vitro secretion of IFN-γ and TGF-β by the cultured cells in response to activation with several concentrations of Compound 1 RS was then measured.
  • Experimental Design
  • 1. Immunization Day 0
    2. Treatment with Compound 1 DP Day 0
    3. In-vitro activation of LN cells Day 10
    from treated mice
    4. Collection of culture media −Day 12
    (for IFN-γ determination)
    5. Collection of culture media −Day 13
    (for TGF-β determination)
    6. ELISA for IFN-γ
    7. ELISA for TGF-β
  • TABLE 7
    Experimental Groups
    In-vitro activation
    Exp. Treatment Compound 1 RS
    Group Article Mice/group Cells/well concentration
    A1 Control 4 2.5 × 106 Compound 1 RS
    12%   5 × 106 0-100 μg/ml
    Captisol ®
    A2  25 μg/mouse 4 2.5 × 106
      5 × 106
    A3  50 μg/mouse 4 2.5 × 106
      5 × 106
    A4 100 μg/mouse 4 2.5 × 106
      5 × 106
    A5 200 μg/mouse 4 2.5 × 106
      5 × 106
  • Materials and Reagents Animals
  • Mice: 20 female BALB/c mice, supplied by Harlan animals breeding center, Rehovot.
    Age at immunization (week+days): 10
    Average weight of mice included in the experiment: 19.01 gr.
  • Materials General Reagents
  • 70% ethanol was prepared from 96% ethanol by diluting with purified H2O.
  • Preparation of Compound 1 Solutions for Immunization
  • CFA-Compound 1 RS emulsion (500 μg/ml, 50 μg/mouse) was prepared as follows:
    • 1. 1.874 mg of Compound 1 was dissolved in 1.87 ml of WFI to yield a solution of 1 mg/ml.
    • 2. The solution was tested with a pH indicator strip and found to have a pH of 5.
    • 3. 1.5 ml of the solution were emulsified with 1.5 ml CFA resulting in a final concentration of 500 μg/ml.
    Preparation of Solutions for Treatment
  • Treatment was by a s.c. injection of 200 μl solution.
  • Preparation of 12% Captisol® Solution
  • 1.2 gr of Captisol® were dissolved in 10 ml of WFI to yield a solution of 12% Captisol®.
  • Experimental Procedure Mice Weighing
  • Mice were weighed before immunization. Average mice weight: 19.01±0.97 gr
  • Immunization
  • The immunization was performed by injecting 100 microliters of the emulsion (50 microliters into each hind footpad).
  • Treatment
  • Following the immunization step the mice were treated by s.c. injection of 200 μl from the designated Compound 1 DP or 12% Captisol® treatment solutions, at the back of their neck.
  • In-Vitro Culture
  • Mice were sacrificed by cervical dislocation. LN were extracted from the hind legs and were transferred to a sterile petri dish containing about 5 mL RPMI. The cells were extracted by gentle squeezing of the tissue against a 200 micrometer mesh stainless steel net. The cells were collected and centrifuged at 300 G for 10 minutes at RT.
  • Single cell suspensions were prepared from pooled LN of each experimental group.
  • 2.5 and 5.0 million cells/ml/well suspensions were cultured with Compound 1 RS (0-100 μg/ml) in EM-1.
  • Secretion of IFN-γ and TGF-β, as indication of cellular response, were determined by ELISA of culture media (48 hrs for IFN-γ and 72 hrs for TGF-β).
  • TABLE 8
    The in-vitro experimental groups
    In-vitro activation
    Experimental Treatment Activation substance
    Group Article Cells/well concentration
    A1-2.5 Control 2.5 × 106 Compound 1 RS
    A1-5 12%   5 × 106 0; 3.125; 6.25; 12.5; 50 and
    Captisol ® 100 μg/ml
    A2-2.5 DP 2.5 × 106 Con A 2.5 μg/ml
    A2-5  25 μg/mouse   5 × 106
    A3-2.5 DP 2.5 × 106
    A3-5  50 μg/mouse   5 × 106
    A4-2.5 DP 2.5 × 106
    A4-5 100 μg/mouse   5 × 106
    A5-2.5 DP 2.5 × 106
    A5-5 200 μg/mouse   5 × 106
  • Preparation of Cell Suspensions
  • TABLE 9
    Results of cell counting and preparation of
    cell suspensions (10 × 106/ml)
    EM-1 to
    add (ml)
    Total for
    Average viable suspension
    Vol. Dilutn. Viable Dead % Viable % Dead viable cells of
    Grp (ml) factor cells cells cells cells cells (×106) 10 × 106 cells/ml
    A1
    10 16 115 100 112 179.2 17.9
    109 100
    A2 10 16 50 4 92.6 7.4 47.5 76 7.6
    45 2 95.7 4.3
    A3 10 16 80 4 95.2 4.8 80.5 128.8 12.8
    81 5 94.2 5.8
    A4 10 16 87 100 89 142 14.2
    91 100
    A5 10 16 120 2 98.4 1.6 112.5 180 18
    105 2 98.1 1.9
  • Preparation of Cell Suspensions (5×106/ml)
  • The 10×106 cells/ml suspensions were diluted 1:2 by adding 5 ml EM-1 to 5 ml cells suspension.
  • Incubation of LN Cells Cultures in 48 Wells Plates
  • 3 tissue culture plates were prepared. The following was added to each plate.
  • Background Control (Cells Incubated with Culture Media)
    0.5 ml of cells suspension
    0.5 ml of culture media (EM-1)
    System Positive Control (Cells Stimulated with Con A)
    0.5 ml of cells suspension
    0.5 ml of Con A 5 μg/ml in EM-1 (final conc. 2.5 μg/well)
    Cells Incubated with Compound 1 Activation Solutions (Samples)
    0.5 ml of cells suspension
    0.5 ml of Compound 1 RS 6.25-200 μg/ml (final conc. 3.125-100 μg/ml/well)
  • Incubation of LN Cells Cultures in 96 Wells Plates
  • After the 48-wells plates were prepared, 96-wells plates were prepared by applying 100 μl from the cell suspension and 100 μl from the activation solutions.
  • The culture plates were incubated at 37° C. in a humidified 5% CO2 incubator, for either 48 or 72 hrs.
  • Supernatants Collection
  • The cultured plates were centrifuged at 300 g for 10 minutes at RT. Supernatants (850 μl from each well) were transferred either to mirror plates or to tubes. The supernatant was then divided into working aliquots (two aliquots of 200 and one aliquot of 450 μl), in order to avoid repeated freeze/thawing of the samples. Each tube was labeled with the following details:
      • 1. Experimental code and time post incubation.
      • 2. Group and sample number
      • 3. Activator and concentration.
      • 4. Date of sup collection
  • The supernatants were stored at −20° C. until used for ELISA.
  • Results
  • TABLE 10
    Summary of Groups
    Experimental Groups:
    Immunization Treatment
    Exp. Immunization Sub In-vitro
    Groups dose group Article activation
    A
    50 μg/mouse A1 12% Captisol ® Compound 1
    Placebo control RS
    A2 Compound
    1 3.125-100 μg/ml
    25 μg/M
    A3 Compound
    1
    50 μg/M
    A4 Compound
    1
    100 μg/M
    A5 Compound
    1
    200 μg/M
  • TABLE 11-A
    Final cytokine concentrations
    Final cytokine (pg/ml) (2.5 million cells/well)
    Compound 1
    concentration Placebo 50 μg/M 100 μg/M 200 μg/M
    3.125 μg/ml  321.3 54.1 64.5 103.9
    6.25 μg/ml 238.6 81.8 116.1 126.1
    12.5 μg/ml 397.1 123.1 180.9 129.0
      25 μ/ml 655.5 215.1 262.8 240.3
      50 μg/ml 573.9 292.5 518.3 378.1
     100 μg/ml 926.0 531.8 582.7 524.1
    Con A 322.6 356.2 337.4 BQL
  • TABLE 11-B
    Final cytokine concentrations
    Final cytokine (pg/ml) (5 million cells/well)
    Compound 1
    concentration Placebo 25 μg/M 50 μg/M 100 μg/M 200 μg/M
    3.125 μg/ml 522.3 BQL 76.2 90.8 204.4
     6.25 μg/ml 634.8 BQL 109.2 157.8 244.1
     12.5 μg/ml 962.8 41.9 179.5 257.1 466.1
      25 μ/ml 967.4 70.0 277.9 421.7 660.5
      50 μg/ml 1338.8 104.2 373.4 739.7 922.5
      100 μg/ml 2010.2 185.2 547.0 995.5 1006.2
    Con A 6839.8 2995.3 4837.0 10126.8 7722.8
  • The results are also presented in FIGS. 3-4.
  • Observations IFN-γ Secretion
      • 1. In the placebo group, a linear dose response upon Compound 1 activation in-vitro was shown. This graph resembles the graph obtained for the Ex-vivo model with the same immunization dose (50 μg/mouse) and culturing medium (EM-1).
      • 2. There was a dose response upon Compound 1 activation in vitro within all the tested groups.
      • 3. Significant inhibition of IFN-γ secretion was seen with all the doses used for treatment (an average of 95% inhibition with treated dose of 25 μg/mouse). A reverse correlation between the dose served for treatment and % inhibition can be found, mainly when 5×106 cells/well were used. When 2.5×106 cells/well were used, treatment of animals with 50 μg/mouse gave better inhibition than 100 or 200 μg. The point of 25 μg is missing (lack of cells).
      • 4. A better inhibition was seen when 5×106 cells/well were used instead of 2.5×106 cells/well.
      • 5. In the linear range of the graphs, SD of % inhibition was low.
      • 6. A technical problem with Con A is apparent when 2.5×106 cells/well were used.
    TGF-β Secretion
      • 1. In the placebo group, no dose response upon in vitro activation with compound 1 was seen. TGF-β secreted level was below the detection limit of the ELISA in all other treatment groups.
    Example 6 Optimizations of Freeze Drying Cycle with Compound 1 and Captisol® for Injection (0, 0.5, 1.0 and 2.5 mg/vial) Purpose
  • The purpose of this study was to optimize the freeze drying cycle for Compound 1 with Captisol® for injection to improve the shape of the lyophilization cake and avoid collapse and cracking. Thus it was decided to improve and optimize the lyophilization cycle. This cycle is transferred to the production lyophilizers for the manufacturing of the phase I batches.
  • Process Optimization
  • Batches of peptide at concentrations of 0.5 mg/ml 11.0 mg/ml, 2.5 mg/ml and Placebo were prepared and several freeze drying cycles were performed. The freeze drier used was an Edwards lyophilizer Lyoflex 0.6.
  • Solubility, water content and cake appearance were tested. According to the obtained results a new lyophilization cycle for Compound 1 was selected. Due to the high percentage of solids (12%) and hence the condensed cake, the new process is longer than the lyophilization cycle in Example 3 and exhibits an additional primary drying stage. Table 12 summarizes the differences between the processes.
  • TABLE 12
    Lyoph. cycle for New Lyoph. cycle for
    Compound 1 and Compound 1 and
    Step Captisol ® of Example 3 Captisol ®
    Loading 5° C. C.
    Freezing
    2 hours to −40° C. 6 hours to −45° C.
    Hold at low 3 hours to −40° C. 3 hours to −45° C.
    temp.
    Primary
    Drying:
    Stage I to 20° C. to −20° C.
    13 hours pressure 110 μbar 19 hours pressure 150 μbar
    Stage II to 25° C.
    13 hours pressure 150 μbar
    Hold at 20° C. 13 hours pressure 110 μbar 8 hours pressure 150 μbar
    (25° C.)
    Secondary
    drying:
    Hold at 20° C. 5 hours pressure 10 μbar
    Storage at 5° C. 5° C.
    Process time 36 hours 49 hours
  • Example 7 Effect of Compound 1 (Administered in Captisol®) on Lupus Symptoms in the SLE-Prone (NZBxNZW)F1 Female Mouse
  • Patients participating in clinical trials are to be treated with Compound 1 using Captisol® (sulfobutyl ether beta-cyclodextrin sodium) as the excipient. For this reason, it was important to determine whether treatment of (NZBxNZW)F1 mice with the formulation of Compound 1 given in Captisol® would have the same beneficial effects on lupus symptoms as observed when this strain of mice was treated with Compound 1 in PBS.
  • To this end, (NZBxNZW)F1 female mice (about 8 months old) were divided into 3 groups that were treated subcutaneously once a week for 10 weeks either with Captisol® alone (n=8) or with 25 or 50 μg/mouse Compound 1 in Captisol® (n=9 and 10, respectively). These doses were selected since prior studies indicated that doses in this range were more effective in ameliorating SLE symptoms than the higher doses tested (100 and 200 μg/mouse). The same batch of drug substance was used in this study and in the first Phase I clinical trial with Compound 1.
  • The mice were followed for anti-dsDNA antibodies and for proteinuria. When the mice were sacrificed, the intensity of ICD was determined in kidneys.
  • As can be seen in FIG. 5, no significant differences between groups could be observed in the levels of dsDNA-specific antibodies after 10 treatment injections.
  • Table 13 also shows that the beneficial effect of treatment with Compound 1 could be observed starting from the 5th injection and it was sustained up to the 10th injection. The mean levels of proteinuria in the Captisol® control group were consistently higher than in the Compound I-treated groups. Table 13 also shows that a reduction in the intensity of ICD was observed in kidneys of both Compound 1 dose groups. There was an overall trend showing that the lower dose (25 μg/mouse) was more effective than the higher dose (50 μg/mouse) in reducing the clinical symptoms of SLE in these mice.
  • TABLE 13
    Clinical Symptoms of SLE in (NZBxNZW)F1 Mice Treated with 25 or
    50 μg/mouse Compound 1 (in Captisol ®)
    Mean Proteinuria ± SEM (g/L) ICDa
    Number of Weeks Following Treatment Initiation (Mean ±
    Study Group 5 7 8 10 SEM)
    Captisol ® 1.81 ± 1.22 5.74 ± 3.13 4.5 ± 2.92  4.46 ± 2.93 2.29 ± 0.28
    (n = 8) (n = 8) (n = 7)b (n = 7)b (n = 7)
    Compound 1 0.75 ± 0.3  0.81 ± 0.3  1.09 ± 0.4  1.29 ± 0.3 1.90 ± 0.23
    (50 μg/mouse)  (n = 10)  (n = 10)  (n = 10)  (n = 10)  (n = 10)
    Compound 1 0.16 ± 0.05 1.26 ± 1.09 0.5 ± 0.31 0.56 ± 0.3 1.22c ± 0.32
    (25 μg/mouse) (n = 9) (n = 9) (n = 9) (n = 9) (n = 9)
    aICD = Immune Complex Deposits. ICD intensity scale: 0 = none; 1 = moderate; 2 = severe; 3 = severe/extremely intense.
    bThe death of one animal with a high level of proteinuria resulted in a lower group mean.
    cp < 0.05 (compared to Captisol ®-treated control mice; Mann-Whitney).
  • FIG. 6 shows representative sections of one kidney from each treatment group. The top row sections are from a Captisol®-treated mouse, the mid-row sections are from a mouse treated with 50 μg/mouse Compound 1 and the bottom row sections are from a mouse treated with 25 μg/mouse Compound 1. It can be seen that the intensity of immune complex deposits observed in kidney sections of mice treated with Compound 1 (dissolved in Captisol®) at either dose level was much lower than that observed in the control group.
  • Example 8 Phase I Clinical Study A Phase I, Multicenter, Randomised, Double-Blind, Placebo Controlled, Single Dose, Four-Arm Study to Assess the Tolerability and Safety of Compound 1 in Captisol® Subcutaneous Injection in SLE Subjects.
  • This was the first clinical study with Compound 1 in Captisol® in humans, conducted in France. Its main objective was to evaluate tolerability and safety of Compound 1 in Captisol®, administered as a single sc injection to SLE subjects. Its secondary objective was to evaluate immunological responses following a single sc dose of Compound 1 in Captisol® in these subjects.
  • Thirty-six (36) subjects participated in the study. To be eligible for inclusion in the study, SLE patients must have fulfilled at least four criteria used for the diagnosis of lupus by the American College of Rheumatology. Patients must also have had stable, mild/moderate disease and score less than or equal to 10 on the SLE Disease Activity Index, SLEDAI.
  • Each patient received a single sc injection of reconstituted Compound 1 for injection or its matching placebo (Captisol®) according to the following group assignment:
      • Group A: Placebo (Captisol®)
      • Group B: 0.5 mg Compound 1 in Captisol®
      • Group C: 1 mg Compound 1 in Captisol®
      • Group D: 2.5 mg Compound 1 in Captisol®
  • A standard battery of safety tests, including blood and urine collection for laboratory tests, was performed at screening, during the day of dosing, at 24 hours post-dose and at 2, 4 and 8 weeks following dosing. Prior to dosing, and on scheduled follow-up visits, blood samples were withdrawn for SLE-related immunological tests, anti-Compound 1 antibodies and PBL proliferation assay. The following immunology tests were performed:
      • Coomb's (direct and indirect)
      • C3, C4 and CH50
      • Total IgG, IgM and IgA
      • ANA, anti-dsDNA (Farr assay), anti-ssDNA
      • Anti-ENA (including anti-La, anti-Ro, anti-RNP, anti-Sm)
      • Anti-cardiolipin antibodies
      • VDRL
      • FTA antibodies
      • Rheumatoid factor
  • The safety and tolerability of Compound 1 in Captisol® in the subject population was evaluated on the basis of the following criteria:
      • Occurrence of AEs, including SLE flare
      • Vital signs
      • 12-lead ECG
      • Changes in physical examination
      • Routine clinical laboratory tests
      • SLEDAI score
      • Immunological test results
    Phase Ia Clinical Study Details
  • Study Principal Investigators and Respective Study Sites: Six (6) study centers in France: Prof. Jean Charles Piette (Hopital La Pitie Salpetriere, Paris), Prof Oliver Meyer (Hopital Bichat, Paris), Prof. Jean Revuz (Hopital Henri Mondor, Creteil), Prof. Loic Guillevin (Hopital Avicenne, Bobigny), Prof. Eric Hachulla (Hopital Claude Huriez, Lille Cedex), Prof. Xavier Mariette (Hopital Bicetre, Kremlin Bicetre).
  • Compound 1 (in Captisol®), Placebo, Water for Injection-Ampoules, Dose and Mode of Administration:
  • Vials of Compound 1 in Captisol® (120 mg/vial) were injected subcutaneously as a single dose per subject in the following dosages:
  • 0.5 mg Compound 1/vial in Captisol®, 1 mg Compound 1/vial in Captisol® and 2.5 mg Compound 1/vial in Captisol®.
  • Placebo for Compound 1: 120 mg Captisol®/vial (identical in appearance to vials of Compound 1 in Captisol®).
  • Methodology
  • This was a multi-center, randomized; double blind, placebo-controlled, four-arm study, using a single subcutaneous injection of Compound 1 or placebo. SLE patients were screened up to 21 days prior to baseline procedures. Each eligible subject was randomized to one of the 4 treatment groups: subcutaneous injection of either 0.5, 1 or 2.5 mg Compound 1 or its matching placebo. All subjects were admitted to the clinic on pre-dosing day. Each subject received a single dose of one of the above listed treatments. Subjects were discharged from the clinic 24 hours after dosing. Subjects were further monitored at weeks 2, 4 and 8 following dosing. Blood samples (serum and whole blood) for safety laboratory tests were withdrawn at Screening, Dosing Day (pre-dose), Day 2 (post dose), at Weeks 2, 4 and 8 (Termination visit). Blood samples for immunological tests were withdrawn at: Screening, Dosing Day (pre-dose) and at Weeks 4 and 8. Peripheral blood lymphocytes (PBL) proliferation was evaluated at Dosing Day (pre-dose) and at Weeks 2, 4 and 8.
  • Number of Subjects (Total and for Each Treatment):
  • Thirty six (36) subjects were randomized into this study as follows; 9 subjects into the 0.5 mg treatment group, 9 subjects into 1 mg treatment group, 10 subjects into the 2.5 mg treatment group, and 8 subjects into the placebo treatment group.
  • Diagnosis and Main Criteria for Inclusion:
  • Eligible subjects for this study were SLE patients who fulfilled at least four diagnostic criteria of the American College of Rheumatology (ACR). Their disease condition had to be stable, mild to moderate with a score equal to or less than 10 on the SLE disease activity index, year 2000 updated (SLEDAI 2K).
  • Excluded from participation were SLE patients who reported unstable or severe asthma, stroke, acute myocardial infarction, unstable angina, cerebral hemorrhage and pulmonary embolism during the six months prior to study screening. SLE patients who had any clinically significant or unstable medical or surgical conditions, diabetes mellitus, liver disease (cirrhosis, active hepatitis, portal hypertension, and/or ascites), clinically significant hypertension, a medical history of any malignancy, dialysis, or chronic obstructive pulmonary disease (COPD) were also excluded from study participation.
  • Also excluded from study participation were SLE patients who underwent plasmapheresis or were treated during the three months prior to screening with one of the drugs listed below: prednisone 30 mg/day or greater (or an equivalent dose of another corticosteroid), intravenous corticosteroids, intravenous immunoglobulin G (IgG), oral anticoagulants and any cytotoxic agents (e.g. azathioprine, chlorambucil, cyclophosphamide, mycophenolate mofetil, methothrexate, tacrolimus.
  • In addition, SLE patients initiating treatment with corticosteroids (more than ±10 mg/day prednisone, or an equivalent dose of another corticosteroid) and/or anti-malarials, during the last 3 months prior to screening were excluded from the study.
  • While an effort was made to retain baseline SLE medical treatments throughout the course of the study, investigators could nevertheless change participant medical treatment at any time during the study to maintain and optimize patient welfare.
  • Criteria for Evaluation Safety:
  • The following safety parameters were assessed at Screening, during the hospitalization and at follow-up visits including Termination visit: vital signs (systolic blood pressure, diastolic blood pressure, pulse, oxygen saturation, temperature and weight), 12-lead ECG, change in physical examination and clinical routine laboratory safety tests. Adverse events were recorded at the Dosing Day and at each visit thereafter.
  • Immunology:
  • SLE-related immunological tests were performed at Screening, during the hospitalization and at follow-up visits including Termination visit.
  • Drug-related immunological responses were followed by using the PBL proliferation assay and anti-Compound 1 antibodies assay at the Dosing Day and at follow-up visits including Termination visit.
  • Disease Activity:
  • Disease activity assessment using the SLE disease activity index score, year 2000 updated (SLEDAI 2K) was assessed at Screening, during the hospitalization and at follow-up visits including Termination visit.
  • Statistical Methods:
  • SAS® version 9.0 software was used to analyze and present data collected during this study. No power calculation was performed and no formal hypothesis testing was conducted for this Phase Ia study.
  • Adverse Experiences
  • The incidence and the frequency of adverse experiences was presented by System Organ Class and preferred terminology according to MedDRA dictionary version 5.0. The data is tabulated by treatment group.
  • Clinical Laboratory Data
  • Descriptive statistics of laboratory values including number of observations, mean, standard deviation, minimum and maximum were determined for Screening, Day 1 (pre dose), Day 2, Week 2, 4 and 8 are presented by treatment group. Changes from baseline to each time point/visit are also presented for each visit by treatment assignment. Percent of abnormal results (low and high, where applicable) are presented on a parameter basis, by treatment group and visit/time point. Shift analyses from baseline to 24-hours post dose and from baseline to termination visit were performed.
  • Vital Signs
  • Descriptive statistics for vital signs including number of observations, mean, standard deviation, median, minimum and maximum values were determined for Screening, Day 1 (pre and post dose, and at each time point) Day 2, Weeks 2, 4 and 8 are tabulated by the assigned treatment. Changes from baseline to each time point/visit is presented in by visit and treatment assignment.
  • Weight
  • Descriptive Statistics of Weight (kg) at baseline, termination and change from baseline is presented by treatment group.
  • ECG
  • Descriptive statistics of ECG parameters at baseline, termination and changes from baseline are presented. Shift analysis is presented as tables of shift from baseline to termination between normal/abnormal or present/absent ECG parameters. Potentially clinically significant (PCS) QTc (Bazett) measurements were identified according to the predefined criteria. Tables of shift analysis between PCS and non-PCS Absolute QTc (Bazett) and incidence table of PCS change in QTc (Bazett) from baseline to any visit are presented.
  • Physical Examination
  • Physical examination results are analyzed by incidence of subjects with abnormal or normal findings for each body system at Baseline and Termination visit. Shift analysis between normal to abnormal and vice versa was also applied. When no change from baseline occurred, it was defined as “other”.
  • Compound 1 Related Immunological Tests
  • For immunological parameters, descriptive statistics, including number of observations, mean, standard deviation, median, minimum and maximum values were calculated and are presented by treatment group and visit. Change from baseline to each follow-up visit is also presented by treatment group. Where applicable, number and percent of subjects with negative/positive results is presented by treatment group and visit.
  • SLEDAI 2K
  • Descriptive statistics, including mean, standard deviation, median, minimum and maximum values of SLEDAI 2K are presented.
  • Results of Phase Ia Clinical Study: Subject Disposition and SLE Characteristics
  • Thirty six (36) study subjects entered and completed this study per protocol. The majority of subjects (34) in all treatment groups were female (94.4%) and Caucasian (30, 83.3%). The mean age for all treatment groups was 35.6 years (range of means from 32 to 39 years). Most of the subjects (91.7%) had between 4 to 6 American College of Rheumatology (ACR) diagnostic criteria and a mean group SLEDAI 2K score ranged from 2.1 to 4.1.
  • Safety Results
  • There was no prominent difference between study drug treatment groups and the placebo group in the incidence of AEs. The most common AEs in all groups were headache, classified as mild or moderate in nature and injection site reaction classified as mild in nature. Dose response was not seen. No serious adverse event (SAE) or AE classified as severe occurred during the study.
  • No clinically significant effect attributable to study drug was seen for hematology, biochemistry or urinalysis values.
  • No clinically significant effect attributable to the study drug was seen for vital signs parameters (systolic blood pressure, diastolic blood pressure, pulse, oxygen saturation).
  • No clinically significant effect attributable to the study drug was seen for temperature and weight.
  • No differences of clinical significance were seen between Compound 1-treated groups and placebo for categorical ECG measurements and digitized ECG parameters. No PCS QTc absolute value and no QTc change from baseline >60 msec was recorded. A similar number of subjects in Compound I-treated and placebo groups had QTcB change from baseline between 30 and 60 msec.
  • No clinically significant effects of Compound 1 on physical exam were noted.
  • Immunology Results
  • Evaluation of serum samples from all subjects indicated that a single subcutaneous administration of Compound 1 at the dose levels of 0.5, 1 and 2.5 mg/patient did not induce the development of anti-Compound 1 specific antibodies. Seven subjects had a response to Compound 1 above the cut-off. These elevated levels of antibodies were already present prior to dosing. No increase in the levels of antibodies was observed in the follow up period (two months) of the study. The sera of these subjects were analyzed for the isotype of the reactive antibodies. The response in two of the subjects was associated with the IgM isotype and with the IgG isotype in two others. None of the seven had specific IgE antibodies.
  • The peripheral blood lymphocytes (PBL) assay showed that 50% of the subjects (18) were classified as responders (SI>2) with similar distribution in all treatment groups. The T cell response was relatively low and no association between Compound 1 treatment dose or concentration used in the assay and responder/non-responder status could be detected, taking into consideration that only a single SC dose of the study drug was administered. Also, no indication of increased incidence, of responder status over time was observed. The tetanus toxoid (TTX) assay that serves as a safety control shows that the response to TTX was preserved throughout the study period in all treatment groups indicating that Compound 1 in Captisol® did not change the immunological response to TTX recall antigen.
  • The immunological findings are the result of the administration of only a single dose of the study drug Compound 1.
  • Disease Activity Results
  • No clinically significant effects of Compound 1 on the SLEDAI score (change of ≧3, ≦12 points) were noted during the study except for one subject in the 0.5 mg treatment group for whom a change in the SLEDAI score of 2 to 10 points was recorded between baseline and week 4 on the basis of an urinalysis showing pyuria. This urinalysis finding was not confirmed by the investigator as a lupus flare per protocol definition and was resolved with no treatment change.
  • Conclusions
  • This Phase Ia study showed that a single subcutaneous injected dose of Compound 1 of 0.5, 1 or 2.5 mg in 120 mg Captisol® was safe and well tolerated and allows continuation to a phase Ib multiple dose study.
  • Example 9 Phase Ib Clinical Study A Phase I, Multicenter, Bi-National, Randomized, Double-Blind, Four-Arm, Placebo Controlled, Multiple Dose Study to Assess the Tolerability and Safety of Compound 1 in Captisol® Subcutaneous Injections in SLE Subjects
  • This study is being performed in order to evaluate the safety and tolerability of repeated Compound 1 sc administration to SLE subjects. The study's secondary objective is to evaluate immunological responses following repeated sc administration of Compound 1 in Captisol® in SLE subjects.
  • Compound 1 is given in doses of 0.5, 1.0 or 2.5 mg in Captisol®. The investigational product is administered every other day (excluding weekends) for a total of 12 sc injections, i.e. 3 doses a week for 4 weeks. Subjects are monitored on planned visits scheduled at 2, 4, 8 and 12 weeks after start of dosing. Safety and tolerability are evaluated using tests similar to those described in the Phase Ia Clinical Study above.
  • Results
  • This Phase Ib study shows that multiple subcutaneous injected doses of Compound 1 of 0.5, 1 or 2.5 mg in 120 mg Captisol® are safe and well tolerated.

Claims (30)

1. A pharmaceutical composition comprising
an aqueous carrier;
from 0.1 mg/ml to 20 mg/ml of the composition of a pharmaceutically acceptable salt of a peptide having the structural formula

NH2-Gly Tyr Tyr Trp Ser Trp Ile Arg Gln Pro Pro Gly Lys Gly Glu Glu Trp Ile Gly-COOH (SEQ ID NO:1); and
a substituted β-cyclodextrin in an amount effective to dissolve the peptide in the aqueous carrier,
wherein the composition has a pH between 4 and 9.
2. The pharmaceutical composition of claim 1, wherein the concentration of the salt of the peptide is at least 0.5 mg/ml, from 0.5 mg/ml to 10 mg/ml, or from 0.5 mg/ml to 2.5 mg/ml.
3. (canceled)
4. (canceled)
5. The pharmaceutical composition of claim 1 wherein the composition has a pH between 6.5 and 8.5.
6. The pharmaceutical composition of claim 5, wherein the composition has a pH between 7.5 and 8.5.
7. The pharmaceutical composition claim 1 wherein the pharmaceutically acceptable salt is an acetate salt.
8. The pharmaceutical composition of claim 1 wherein the substituted β-cyclodextrin is a hydroxypropyl, a sulfobutyl ether, or a sulfopropyl ether substituted β-cyclodextrin.
9. The pharmaceutical composition of claim 8, wherein the substituted β-cyclodextrin is a sulfobutyl ether substituted β-cyclodextrin.
10. The pharmaceutical composition of claim 7, wherein the substituted β-cyclodextrin is hepta-(sulfobutyl ether)-β-cyclodextrin.
11. The pharmaceutical composition of claim 1 further comprising a pharmaceutically acceptable buffer in an amount and of a type suitable to make the pH of the pharmaceutical composition in the range of 4-9.
12. A pharmaceutical composition comprising
an aqueous carrier;
from 0.1 mg/ml to 20 mg/ml of the composition of an acetate salt of a peptide having the structural formula

NH2-Gly Tyr Tyr Trp Ser Trp Ile Arg Gln Pro Pro Gly Lys Gly Glu Glu Trp Ile Gly-COOH (SEQ ID NO:1); and
from 70 mg/ml to 170 mg/ml of the composition of hepta-(sulfobutyl ether)-β-cyclodextrin,
wherein the peptide and the hepta-(sulfobutyl ether)-β-cyclodextrin are dissolved in the aqueous carrier; and
wherein the composition has a pH between 6.5 and 8.5.
13. The pharmaceutical composition of claim 12, wherein the concentration of the acetate salt of the peptide is at least 0.5 mg/ml, from 0.5 mg/ml to 10 mg/ml, or from 0.5 to 2.5 mg/ml.
14. (canceled)
15. (canceled)
16. The pharmaceutical composition of claim 13, wherein the concentration of hepta-(sulfobutyl ether)-β-cyclodextrin is 120 mg/ml, and wherein the pH of the composition is between 7.5 and 8.5.
17. The pharmaceutical composition of claim 16, wherein the concentration of the acetate salt of the peptide is 1.0 mg/ml or 2.5 mg/ml.
18. (canceled)
19. A method of alleviating symptoms of systemic lupus erythematosus (SLE) in a human subject comprising administering to the human subject the pharmaceutical composition of claim 1 in an amount effective to alleviate the symptoms of SLE in the human subject.
20. (canceled)
21. A process for manufacturing the pharmaceutical composition of claim 1 comprising the steps of:
a) preparing a solution of a substituted β-cyclodextrin in an aqueous carrier at a predetermined concentration;
b) adding a predetermined amount of a pharmaceutically acceptable salt of the peptide NH2-Gly Tyr Tyr Trp Ser Trp Ile Arg Gln Pro Pro Gly Lys Gly Glu Glu Trp Ile Gly-COOH (SEQ ID NO:1) to the solution of step a);
c) adjusting the pH of the solution of step b) until the peptide dissolves in the solution; and
d) if necessary, adjusting the pH of the solution of step c) to a pH of 4-9, thereby manufacturing the pharmaceutical composition.
22-31. (canceled)
32. A process of lyophilizing the pharmaceutical composition of claim 2, comprising the steps of:
a) lowering the temperature of the pharmaceutical composition to −40° C.;
b) holding the temperature at −40° C. for a predetermined time;
c) raising the temperature of the solution to 20° C.;
d) holding the temperature at 20° C. for a predetermined time; and
e) reducing the pressure and holding the temperature at 20° C. for a predetermined time, thereby lyophilizing the pharmaceutical composition,
or
comprising the steps of:
i) lowering the temperature of the pharmaceutical composition to −45° C.;
ii) holding the temperature at −45° C. for a predetermined time;
iii) raising the temperature of the solution to −20° C.;
iv) raising the temperature of the solution to 25° C.; and
v) holding the temperature at 25° C. for a predetermined time, thereby lyophilizing the pharmaceutical composition.
33-40. (canceled)
41. The process of claim 32, wherein the process comprises steps a)-e), and
step a) is performed within 2 hours;
step b) is performed within 3 hours;
step c) is performed over 13 hours and at a pressure of 110 μbar;
step d) is performed over 13 hours and at a pressure of 110 μbar; and
step e) is performed over 5 hours and the pressure is reduced to 10 μbar.
42-51. (canceled)
52. The process of claim 32, wherein the process comprises steps i)-v), and
step i) is performed within 6 hours;
step ii) is performed within 3 hours;
step iii) is performed over 19 hours and at a pressure of 150 μbar;
step iv) is performed over 13 hours and at a pressure of 150 μbar; and
step v) is performed over 8 hours and at a pressure of 150 μbar.
53-56. (canceled)
57. A lyophilized pharmaceutical composition comprising
a pharmaceutically acceptable salt of a peptide having the structural formula

NH2-Gly Tyr Tyr Trp Ser Trp Ile Arg Gln Pro Pro Gly Lys Gly Glu Glu Trp Ile Gly-COOH (SEQ ID NO:1); and
a substituted β-cyclodextrin.
58. A packaged pharmaceutical composition comprised of:
a packaging material; and
a predetermined amount of the lyophilized pharmaceutical composition of claim 57.
US11/985,068 2003-01-14 2007-11-12 Parenteral formulations of a peptide for the treatment of systemic lupus erythematosus Abandoned US20080287366A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US11/985,068 US20080287366A1 (en) 2003-01-14 2007-11-12 Parenteral formulations of a peptide for the treatment of systemic lupus erythematosus

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US43995003P 2003-01-14 2003-01-14
US10/758,572 US7294687B2 (en) 2003-01-14 2004-01-14 Parenteral formulations of a peptide for the treatment of systemic lupus erythematosus
US11/985,068 US20080287366A1 (en) 2003-01-14 2007-11-12 Parenteral formulations of a peptide for the treatment of systemic lupus erythematosus

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US10/758,572 Continuation US7294687B2 (en) 2003-01-14 2004-01-14 Parenteral formulations of a peptide for the treatment of systemic lupus erythematosus

Publications (1)

Publication Number Publication Date
US20080287366A1 true US20080287366A1 (en) 2008-11-20

Family

ID=32771763

Family Applications (2)

Application Number Title Priority Date Filing Date
US10/758,572 Expired - Lifetime US7294687B2 (en) 2003-01-14 2004-01-14 Parenteral formulations of a peptide for the treatment of systemic lupus erythematosus
US11/985,068 Abandoned US20080287366A1 (en) 2003-01-14 2007-11-12 Parenteral formulations of a peptide for the treatment of systemic lupus erythematosus

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US10/758,572 Expired - Lifetime US7294687B2 (en) 2003-01-14 2004-01-14 Parenteral formulations of a peptide for the treatment of systemic lupus erythematosus

Country Status (18)

Country Link
US (2) US7294687B2 (en)
EP (1) EP1587525A4 (en)
JP (1) JP2006516034A (en)
KR (1) KR20050100616A (en)
CN (1) CN1761477A (en)
AU (1) AU2004206844A1 (en)
BR (1) BRPI0406737A (en)
CA (1) CA2513331A1 (en)
CO (1) CO5640149A2 (en)
CR (1) CR7936A (en)
EA (1) EA008438B1 (en)
EC (1) ECSP055960A (en)
MX (1) MXPA05007552A (en)
NO (1) NO20053761L (en)
NZ (1) NZ541659A (en)
UA (1) UA83816C2 (en)
WO (1) WO2004064788A2 (en)
ZA (1) ZA200506205B (en)

Families Citing this family (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
IL141647A0 (en) 2001-02-26 2002-03-10 Yeda Res & Dev Synthetic human peptides and pharmaceutical compositions comprising them for the treatment of systemic lupus erythematosus
BRPI0406737A (en) * 2003-01-14 2005-12-20 Teva Pharma Parenteral formulations of a peptide for the treatment of systemic lupus erythematosis
JP4817068B2 (en) * 2003-01-14 2011-11-16 イエダ・リサーチ・アンド・ディベロップメント・カンパニー・リミテッド Parenteral formulation of peptides for the treatment of systemic lupus erythematosus
DE102004043750A1 (en) * 2004-09-10 2006-03-30 Sanofi-Aventis Deutschland Gmbh Formulations of the peptide p277 or its variants with optimized stability
US20100160442A1 (en) * 2006-07-18 2010-06-24 Ossovskaya Valeria S Formulations for cancer treatment
EP2124883A2 (en) * 2006-12-12 2009-12-02 Amylin Pharmaceuticals, Inc. Pharmaceutical formulations and methods for making the same
UA100852C2 (en) * 2007-01-16 2013-02-11 Байпар Сайенсиз, Инк. Composition for use in treating cancer
US7635773B2 (en) 2008-04-28 2009-12-22 Cydex Pharmaceuticals, Inc. Sulfoalkyl ether cyclodextrin compositions
MX2011000847A (en) 2008-08-06 2011-02-25 Novo Nordisk Healthcare Ag Conjugated proteins with prolonged in vivo efficacy.
PL3702374T3 (en) 2012-02-15 2022-11-21 Cydex Pharmaceuticals, Inc. Manufacturing process for cyclodextrin derivatives
CN105073785B (en) 2012-10-22 2018-08-21 锡德克斯药物公司 Alkylated cyclodextrin composition and its preparation and application
EP3183295B1 (en) 2014-08-22 2023-08-02 CyDex Pharmaceuticals, Inc. Fractionated alkylated cyclodextrin compositions and processes for preparing and using the same
JP7242059B2 (en) 2017-01-05 2023-03-20 イエダ リサーチ アンド ディベロプメント カンパニー リミテッド Peptides for the treatment of Sjögren's syndrome
CN110960442B (en) * 2019-12-17 2023-03-31 珠海冀百康生物科技有限公司 Preparation method of polypeptide encapsulate

Citations (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4656158A (en) * 1984-03-02 1987-04-07 Suntory Limited Peptide, and production and use thereof
US5126249A (en) * 1989-05-09 1992-06-30 Eli Lilly And Company Enzymatic removal of a protein amino-terminal sequence
US5134127A (en) * 1990-01-23 1992-07-28 University Of Kansas Derivatives of cyclodextrins exhibiting enhanced aqueous solubility and the use thereof
US5376645A (en) * 1990-01-23 1994-12-27 University Of Kansas Derivatives of cyclodextrins exhibiting enhanced aqueous solubility and the use thereof
US5646131A (en) * 1994-02-22 1997-07-08 The Arab Company For Drug Industries And Medical Applicances (Acdima) Method for solubilizing drugs using cyclodextrins and carboxylic acids
US5730969A (en) * 1988-10-05 1998-03-24 Chiron Corporation Method and compositions for solubilization and stabilization of polypeptides, especially proteins
US6228363B1 (en) * 1997-03-20 2001-05-08 Hadasit Medical Research Services & Development Company Ltd. Peptides for the treatment of systemic lupus erythematosus
US20020054872A1 (en) * 1997-03-20 2002-05-09 Yaakov Naparstek Peptides for the treatment of systemic lupus erythematosus and methods of treating systemic lupus erythematosus
US6407079B1 (en) * 1985-07-03 2002-06-18 Janssen Pharmaceutica N.V. Pharmaceutical compositions containing drugs which are instable or sparingly soluble in water and methods for their preparation
US6613536B1 (en) * 1995-03-28 2003-09-02 Yeda Research And Development Co. Ltd. Synthetic peptides and pharmaceutical compositions comprising them for the treatment of systemic lupus erythematosus
US20040127408A1 (en) * 2001-02-26 2004-07-01 Edna Mozes Synthetic human peptides and pharmaceutical compositions comprising them for the treatment of systemic lupus erythematosus
US20040180059A1 (en) * 2003-01-14 2004-09-16 Sharon Cohen-Vered Parenteral formulations of a peptide for the treatment of systemic lupus erythematosus
US20050008634A1 (en) * 2003-01-14 2005-01-13 Sharon Cohen-Vered Parenteral formulations of peptides for the treatment of systemic lupus erythematosus

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB9017008D0 (en) 1990-08-02 1990-09-19 Erba Carlo Spa Process for the enzymatic preparation of basic fibroblast growth factor
IL113159A0 (en) 1995-03-28 1995-06-29 Yeda Res & Dev Synthetic peptides and pharmaceutical compositions comprising them
WO1999031066A1 (en) 1997-12-18 1999-06-24 Boehringer Ingelheim Pharmaceuticals, Inc. Pyridones as src family sh2 domain inhibitors
US7114005B2 (en) 2002-02-05 2006-09-26 Cisco Technology, Inc. Address hopping of packet-based communications

Patent Citations (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4656158A (en) * 1984-03-02 1987-04-07 Suntory Limited Peptide, and production and use thereof
US6407079B1 (en) * 1985-07-03 2002-06-18 Janssen Pharmaceutica N.V. Pharmaceutical compositions containing drugs which are instable or sparingly soluble in water and methods for their preparation
US5997856A (en) * 1988-10-05 1999-12-07 Chiron Corporation Method and compositions for solubilization and stabilization of polypeptides, especially proteins
US5730969A (en) * 1988-10-05 1998-03-24 Chiron Corporation Method and compositions for solubilization and stabilization of polypeptides, especially proteins
US5126249A (en) * 1989-05-09 1992-06-30 Eli Lilly And Company Enzymatic removal of a protein amino-terminal sequence
US5376645A (en) * 1990-01-23 1994-12-27 University Of Kansas Derivatives of cyclodextrins exhibiting enhanced aqueous solubility and the use thereof
US5134127A (en) * 1990-01-23 1992-07-28 University Of Kansas Derivatives of cyclodextrins exhibiting enhanced aqueous solubility and the use thereof
US5646131A (en) * 1994-02-22 1997-07-08 The Arab Company For Drug Industries And Medical Applicances (Acdima) Method for solubilizing drugs using cyclodextrins and carboxylic acids
US6613536B1 (en) * 1995-03-28 2003-09-02 Yeda Research And Development Co. Ltd. Synthetic peptides and pharmaceutical compositions comprising them for the treatment of systemic lupus erythematosus
US6228363B1 (en) * 1997-03-20 2001-05-08 Hadasit Medical Research Services & Development Company Ltd. Peptides for the treatment of systemic lupus erythematosus
US20020054872A1 (en) * 1997-03-20 2002-05-09 Yaakov Naparstek Peptides for the treatment of systemic lupus erythematosus and methods of treating systemic lupus erythematosus
US20040127408A1 (en) * 2001-02-26 2004-07-01 Edna Mozes Synthetic human peptides and pharmaceutical compositions comprising them for the treatment of systemic lupus erythematosus
US20040180059A1 (en) * 2003-01-14 2004-09-16 Sharon Cohen-Vered Parenteral formulations of a peptide for the treatment of systemic lupus erythematosus
US20050008634A1 (en) * 2003-01-14 2005-01-13 Sharon Cohen-Vered Parenteral formulations of peptides for the treatment of systemic lupus erythematosus
US7294687B2 (en) * 2003-01-14 2007-11-13 Teva Pharmaceutical Industries, Ltd. Parenteral formulations of a peptide for the treatment of systemic lupus erythematosus

Also Published As

Publication number Publication date
CA2513331A1 (en) 2004-08-05
US7294687B2 (en) 2007-11-13
ZA200506205B (en) 2006-12-27
CR7936A (en) 2006-05-31
EP1587525A4 (en) 2008-09-10
ECSP055960A (en) 2006-04-19
JP2006516034A (en) 2006-06-15
EA200501129A1 (en) 2006-02-24
BRPI0406737A (en) 2005-12-20
AU2004206844A1 (en) 2004-08-05
EP1587525A2 (en) 2005-10-26
NO20053761L (en) 2005-10-12
WO2004064788A2 (en) 2004-08-05
NZ541659A (en) 2007-05-31
CO5640149A2 (en) 2006-05-31
NO20053761D0 (en) 2005-08-08
US20040180059A1 (en) 2004-09-16
EA008438B1 (en) 2007-06-29
KR20050100616A (en) 2005-10-19
MXPA05007552A (en) 2006-05-19
WO2004064788A3 (en) 2005-03-24
UA83816C2 (en) 2008-08-26
CN1761477A (en) 2006-04-19

Similar Documents

Publication Publication Date Title
US20080287366A1 (en) Parenteral formulations of a peptide for the treatment of systemic lupus erythematosus
US20130023485A1 (en) Parenteral formulations of peptides for the treatment of systematic lupus erythematosus
AU620926B2 (en) Cyclodextrin-peptide complexes
Conceicao et al. Cyclodextrins as drug carriers in pharmaceutical technology: the state of the art
Irie et al. Cyclodextrins in peptide and protein delivery
US7618944B2 (en) Encapsulated amyloid-beta peptides
US20110097306A1 (en) Oral formulations of cladribine
CN107028897B (en) Immune regulator thymopentin powder injection medicine composition and quality control method
US20140170225A1 (en) Immunotherapy for treatment of amyloid-related disorders using encapsulated beta-amyloid peptides
Kulkarni et al. Cyclodextrins for Delivery of Biotechnology Based Drugs

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

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