throbber
(19)
`
`(12)
`
`Europäisches Patentamt
`
`European Patent Office
`
`Office européen des brevets
`
`*EP000699686B1*
`EP 0 699 686 B1
`
`(11)
`
`EUROPEAN PATENT SPECIFICATION
`
`(45) Date of publication and mention
`of the grant of the patent:
`08.10.2003 Bulletin 2003/41
`
`(21) Application number: 95305963.1
`
`(22) Date of filing: 25.08.1995
`
`(51) Int Cl.7: C07K 14/605, A61K 38/22
`
`(54) Biologically active fragments of glucagon-like insulinotropic peptide
`
`Biologisch aktive Fragmente des Glucagon ähnlichen, insulinotropen Peptides
`
`Fragments biologiquement actifs de peptide insalinotrope de type glucagon
`
`(84) Designated Contracting States:
`AT BE CH DE DK ES FR GB GR IE IT LI LU NL PT
`SE
`Designated Extension States:
`LT LV SI
`
`(30) Priority: 30.08.1994 US 297731
`
`(43) Date of publication of application:
`06.03.1996 Bulletin 1996/10
`
`(73) Proprietor: ELI LILLY AND COMPANY
`Indianapolis, Indiana 46285 (US)
`
`(72) Inventors:
`• Johnson, William Terry
`Indianapolis, Indiana 46227 (US)
`• Yakubu-Madus, Fatima Emitsela
`Indianapolis, Indiana 46256 (US)
`
`(74) Representative: Denholm, Anna Marie et al
`Eli Lilly and Company Limited,
`Lilli Research Centre,
`Erl Wood Manor
`Windlesham Surrey GU20 6PH (GB)
`
`(56) References cited:
`WO-A-93/25579
`
`• ENDOCRINOLOGY (BALTIMORE) (1989), 125(6),
`3109-14 CODEN: ENDOAO;ISSN: 0013-7227,
`1989, XP000574778 SUZUKI, SEIJI ET AL:
`"Comparison of the effects of various C-terminal
`and N-terminal fragment peptides of
`glucagon-like peptide-1 on insulin and glucagon
`release from the isolated perfused rat pancreas"
`
`• BIOMED. RES. (1988), 9(SUPPL. 3), 213-17
`CODEN: BRESD5;ISSN: 0388-6107, 1988,
`XP000574772 KAWAI, KOICHI ET AL: "The
`biological effects of glucagon-like peptide-1
`(GLP-1) and its structure-activity relationship"
`• INTERNATIONAL JOURNAL OF PEPTIDE AND
`PROTEIN RESEARCH, vol. 40, no. 3 / 04, 1
`September 1992, pages 333-343, XP000311244
`SVETLANA MOJSOV: "STRUCTURAL
`REQUIREMENTS FOR BIOLOGICAL ACTIVITY
`OF GLUCAGON-LIKE PEPTIDE-I"
`• ENDOCRINOLOGY (BALTIMORE) (1990), 126(4),
`2164-8 CODEN: ENDOAO;ISSN: 0013-7227,
`1990, XP000574862 GEFEL, DOV ET AL:
`"Glucagon-like peptide-I analogs: effects on
`insulin secretion and adenosine
`3’,5’-monophosphate formation"
`• J. CLIN. ENDOCRINOL. METAB. (1995), 80(3),
`952-7 CODEN: JCEMAZ;ISSN: 0021-972X, 1995,
`XP000574780 DEACON, CAROLYN F. ET AL:
`"Degradation of glucagon-like peptide-1 by
`human plasma in vitro yields an N-terminally
`truncated peptide that is a major endogenous
`metabolite in vivo"
`• ENDOCRINOLOGY (1995), 136(8), 3585-97
`CODEN: ENDOAO;ISSN: 0013-7227, 1995,
`XP000574863 KIEFFER, TIMOTHY J. ET AL:
`"Degradation of glucose-dependent
`insulinotropic polypeptide and truncated
`glucagon-like peptide 1 in vitro and in vivo by
`dipeptidyl peptidase IV"
`
`Note: Within nine months from the publication of the mention of the grant of the European patent, any person may give
`notice to the European Patent Office of opposition to the European patent granted. Notice of opposition shall be filed in
`a written reasoned statement. It shall not be deemed to have been filed until the opposition fee has been paid. (Art.
`99(1) European Patent Convention).
`
`Printed by Jouve, 75001 PARIS (FR)
`
`EP0 699 686B1
`
`MYLAN INST. EXHIBIT 1044 PAGE 1
`
`MYLAN INST. EXHIBIT 1044 PAGE 1
`
`

`

`EP 0 699 686 B1
`
`Description
`
`Field of Invention
`
`5
`
`10
`
`15
`
`20
`
`25
`
`30
`
`35
`
`40
`
`45
`
`50
`
`55
`
`[0001] The present invention relates to medicinal chemistry and provides novel peptides and compositions thereof
`that are useful for treating diabetes.
`
`Background of the Invention
`
`[0002] Endocrine secretions of pancreatic islets are regulated by complex control mechanisms driven not only by
`blood-borne metabolites such as glucose, amino acids, and catecholamines, but also by local paracrine influences.
`The major pancreatic islet hormones, glucagon, insulin and somatostatin, interact with specific pancreatic cell types
`(A, B, and D cells, respectively) to modulate the secretory response. Although insulin secretion is predominantly con-
`trolled by blood glucose levels, somatostatin inhibits glucose-mediated insulin secretion.
`[0003] The human hormone glucagon is a 29-amino acid hormone produced in pancreatic A-cells. The hormone
`belongs to a multi-gene family of structurally related peptides that include secretin, gastric inhibitory peptide, vasoactive
`intestinal peptide and glicentin. These peptides variously regulate carbohydrate metabolism, gastrointestinal motility
`and secretory processing. However, the principal recognized actions of pancreatic glucagon are to promote hepatic
`glycogenolysis and glyconeogenesis, resulting in an elevation of blood sugar levels. In this regard, the actions of glu-
`cagon are counter regulatory to those of insulin and may contribute to the hyperglycemia that accompanies Diabetes
`mellitus (Lund, P.K., et al., Proc. Natl. Acad. Sci. U.S.A., 79:345-349 (1982)).
`[0004] When glucagon binds to its receptor on insulin producing cells, cAMP production increases which in turn
`stimulates insulin expression (Korman, L.Y., et al., Diabetes, 34:717-722 (1985)). Moreover, high levels of insulin down-
`regulate glucagon synthesis by a feedback inhibition mechanism (Ganong, W.F., Review of Medical Physiology, Lange
`Publications, Los Altos, California, p. 273 (1979)). Thus, the expression of glucagon is carefully regulated by insulin,
`and ultimately by serum glucose levels.
`[0005] Preproglucagon, the zymogen form of glucagon, is translated from a 360 base pair gene and is processed to
`form proglucagon (Lund, et al., Proc. Natl. Acad. Sci. U.S.A. 79:345-349 (1982)). Patzelt, et al. (Nature, 282:260-266
`(1979)) demonstrated that proglucagon is further processed into glucagon and a second peptide. Later experiments
`demonstrated that proglucagon is cleaved carboxyl to Lys-Arg or Arg-Arg residues (Lund, P.K., et al., Lopez L.C., et
`al., Proc. Natl. Acad. Sci. U.S.A., 80:5485-5489 (1983), and Bell, G.I., et al., Nature 302:716-718 (1983)). Bell, G.I., et
`al., also discovered that proglucagon contained three discrete and highly homologous peptide regions which were
`designated glucagon, glucagon-like peptide 1 (GLP-1), and glucagon-like peptide 2 (GLP-2). Lopez, et al., demon-
`strated that GLP-1 was a 37 amino acid peptide and that GLP-2 was a 34 amino acid peptide. Analogous studies on
`the structure of rat preproglucagon revealed a similar pattern of proteolytic cleavage at Lys-Arg or Arg-Arg residues,
`resulting in the formation of glucagon, GLP-1, and GLP-2 (Heinrich, G., et al., Endocrinol., 115:2176-2181 (1984)).
`Finally, human, rat, bovine, and hamster sequences of GLP-1 have been found to be identical (Ghiglione, M., et al.,
`Diabetologia, 27:599-600 (1984)).
`[0006] The conclusion reached by Lopez, et al., regarding the size of GLP-1 was confirmed by studying the molecular
`forms of GLP-1 found in the human pancreas (Uttenthal, L.O., et al. J. Clin. Endocrinol. Metabol., 61:472-479 (1985)).
`Their research showed that GLP-1 and GLP-2 are present in the pancreas as 37 and 34 amino acid peptides respec-
`tively.
`[0007] The similarity between GLP-1 and glucagon suggested to early investigators that GLP-1 might have biological
`activity. Although some investigators found that GLP-1 could induce rat brain cells to synthesize cAMP (Hoosein, N.
`M., et al., Febs Lett. 178:83-86 (1984)), other investigators failed to identify any physiological role for GLP-1 (Lopez,
`L.C., et al. supra). However, GLP-1 is now known to stimulate insulin secretion (insulinotropic action) causing glucose
`uptake by cells which decreases serum glucose levels (see, e g., Mojsov, S., Int. J. Peptide Protein Research, 40:
`333-343 (1992)).
`[0008] Numerous GLP-1 analogs demonstrating insulinotropic action are known in the art. These variants and ana-
`logs include, for example, GLP-1(7-36), Gln9-GLP-1(7-37), D-Gln9-GLP-1(7-37), acetyl-Lys9-GLP-1(7-37), Thr16-
`Lys18-GLP-1(7-37), and Lys18-GLP-1(7-37). Derivatives of GLP-1 include, for example, acid addition salts, carboxylate
`salts, lower alkyl esters, and amides (see, e.g., WO91/11457 (1991)). More importantly, it was demonstrated using
`GLP-1(8-37)OH that the histidine residue at position 7 is very important to insulinotropic activity of GLP-1 (Suzuki, S.,
`et al. Diabetes Res.; Clinical Practice 5 (Supp. 1):S30 (1988).
`[0009] Endocrinology, Volume 125, Number 6, Issued 1989, Suzuki et al., "Comparison of the Effects of Various C
`Terminal and N-Terminal Fragment Peptides of Glucagen-Like Peptide-1 on Insulin and Glucagon Release from the
`Isolated Perfused Rat Pancreas", pages 3109-3114 teaches that truncated glucagon-like peptide-1 (GLP-1) posseses
`a potent stimulatory activity for insulin production. The activities of N and C-terminal fragments were examined.
`
`2
`
`MYLAN INST. EXHIBIT 1044 PAGE 2
`
`MYLAN INST. EXHIBIT 1044 PAGE 2
`
`

`

`EP 0 699 686 B1
`
`[0010]
`In view of the above, it was most surprising when the present inventors discovered that administering N-
`terminal deletion mutants of GLP-1 to experimental animals caused an increase in serum glucose uptake in the absence
`of any insulinotropic activity. This discovery suggests that an entirely new mechanism for lowering elevated blood
`glucose levels may exist and directly lead to the present invention.
`[0011] Accordingly, the primary object of this invention is to provide novel, C-terminal GLP-1 fragments having no
`insulinotropic action but which are nonetheless useful for treating diabetes and hyperglycemic conditions. Further ob-
`jects of the present invention are pharmaceutical compositions that contain biologically-active GLP-1 fragments, as
`well as methods for using such compounds to treat diabetes.
`
`5
`
`10
`
`Summary of the Invention
`
`[0012] According to a first aspect of the present invention there is provided a protected, unprotected, or partially
`protected GLP-1 fragment selected from the following formulae:
`
`15
`
`20
`
`25
`
`30
`
`35
`
`40
`
`45
`
`50
`
`55
`
`wherein R1 is selected from NH2, OH, Gly-NH2, and Gly-OH;1 R2 is selected from OH and NH2.
`[0013] Preferably, the formula of the GLP-1 fragment is:
`
`3
`
`MYLAN INST. EXHIBIT 1044 PAGE 3
`
`MYLAN INST. EXHIBIT 1044 PAGE 3
`
`

`

`5
`
`10
`
`15
`
`20
`
`25
`
`30
`
`35
`
`40
`
`45
`
`50
`
`55
`
`EP 0 699 686 B1
`
`wherein R1 is selected from NH2, OH, Gly-NH2, and Gly-OH;
`
`wherein R2 is selected from NH2 and OH.
`[0014] The GLP-1 fragment is preferably protected or partially protected. In particular, the side chain of the amino
`acid Lys may be protected. The side chain of the amino acid Lys may be protected by acetyl or C1-Z.
`[0015] According to a second aspect of the present invention, there is provided a GLP-1 fragment of the present
`invention, for use in treating diabetes or hyperglycemia in a mammal.
`[0016] According to a third aspect of the present invention, there is provided a pharmaceutical composition compris-
`ing a GLP-1 fragment of the present invention, in combination with a pharmaceutical carrier, diluent or excipient.
`[0017] The composition may be used for use in treating diabetes or hyperglycemia in a mammal.
`[0018] According to a further aspect of the present invention, there is provided the use of a GLP-1 fragment according
`to the present invention in the preparation of a medicament for the treatment of diabetes.
`
`Detailed Description of the Invention
`
`[0019]
`In one embodiment, the present invention provides novel, biologically-active, C-terminal fragments of GLP-
`1. For purposes of this specification, the term "biologically-active" refers to the ability of a substance to lower elevated
`levels of blood glucose in a mammal without stimulating insulin secretion.
`[0020] Given the sequence information herein disclosed and the state of the art in solid phase protein synthesis,
`biologically-active GLP-1 fragments can be obtained via chemical synthesis. However, it also is possible to obtain a
`biologically-active GLP-1 fragment by fragmenting proglucagon using, for example, proteolytic enzymes. Moreover,
`recombinant DNA techniques may be used to express biologically-active GLP-1 fragments.
`[0021] The principles of solid phase chemical synthesis of polypeptides are well known in the art and may be found
`in general texts in the area such as Dugas, H. and Penney, C., Bioorganic Chemistry (1981) Springer-Verlag, New
`York, pgs. 54-92, Merrifield, J.M., Chem. Soc., 85:2149 (1962), and Stewart and Young, Solid Phase Peptide Synthesis,
`pp. 24-66, Freeman (San Francisco, 1969).
`[0022] For example, a biologically-active GLP-1 fragment may be synthesized by solid-phase methodology utilizing
`an Applied Biosystems 430A peptide synthesizer (Applied Biosystems, Inc., 850 Lincoln Center Drive, Foster City, CA
`94404) and synthesis cycles supplied by Applied Biosystems. Boc amino acids and other reagents are commercially
`available from Applied Biosystems and other chemical supply houses. Sequential Boc chemistry using double couple
`protocols are applied to the starting p-methyl benzhydryl amine resins for the production of C-terminal carboxamides.
`For the production of C-terminal acids, the corresponding PAM resin is used. Asp, Gln, and Arg are coupled using
`preformed hydroxy benzotriazole esters. The following side chain protecting groups may be used:
`
`Arg, Tosyl
`Asp, cyclohexyl
`Glu, cyclohexyl
`Ser, Benzyl
`Thr, Benzyl
`Tyr, 4-bromo carbobenzoxy
`
`[0023] Boc deprotection may be accomplished with trifluoroacetic acid in methylene chloride. Following completion
`of the synthesis the peptides may be deprotected and cleaved from the resin with anhydrous hydrogen fluoride (HF)
`containing 10% meta-cresol. Cleavage of the side chain protecting group(s) and of the peptide from the resin is carried
`
`4
`
`MYLAN INST. EXHIBIT 1044 PAGE 4
`
`MYLAN INST. EXHIBIT 1044 PAGE 4
`
`

`

`EP 0 699 686 B1
`
`out at zero degrees centigrade or below, preferably -20°C for thirty minutes followed by thirty minutes at 0°C. After
`removal of the HF, the peptide/resin is washed with ether, and the peptide extracted with glacial acetic acid and lyophi-
`lized.
`[0024] The preparation of protected, unprotected, and partially protected GLP-1 has been described in the art. See
`U.S. Pat. No. 5,120,712 and 5,118,666, herein incorporated by reference, and Orskov, C., et al., J. Biol. Chem., 264
`(22):12826-12829 (1989) and WO 91/11457 (Buckley, D.I., et al., published August 8, 1991).
`[0025]
`Likewise, the state of the art in molecular biology provides the ordinarily skilled artisan another means by
`which biologically-active GLP-1 fragments can be obtained. Although GLP-1 fragments may be produced by solid
`phase peptide synthesis, recombinant methods, or by fragmenting glucagon, recombinant methods may be preferable
`because higher yields are possible. The basic steps in the recombinant production of a biologically-active GLP-1 frag-
`ment are:
`
`a) isolating a natural DNA sequence encoding GLP-1 or constructing a synthetic or semi-synthetic DNA coding
`sequence for GLP-1,
`
`b) placing the coding sequence into an expression vector in a manner suitable for expressing proteins either alone
`or as a fusion proteins,
`
`c) transforming an appropriate eukaryotic or prokaryotic host cell with the expression vector,
`
`d) culturing the transformed host cell under conditions that will permit expression of a GLP-1 intermediate, and
`
`e) recovering and purifying the recombinantly produced protein.
`
`[0026] As previously stated, the coding sequences for GLP-1 fragments may be wholly synthetic or the result of
`modifications to the larger, native glucagon-encoding DNA. A DNA sequence that encodes preproglucagon is presented
`in Lund, et al., Proc. Natl. Acad. Sci. U.S.A. 79:345-349 (1982) and may be used as starting material in the recombinant
`production of a biologically-active GLP-1 fragment by altering the native sequence to achieve the desired results.
`[0027] Synthetic genes, the in vitro or in vivo transcription and translation of which results in the production of a
`biologically-active GLP-1 fragment, may be constructed by techniques well known in the art. Owing to the natural
`degeneracy of the genetic code, the skilled artisan will recognize that a sizable yet definite number of DNA sequences
`may be constructed which encode GLP-1 intermediates.
`[0028] The methodology of synthetic gene construction is well known in the art. See Brown, et al. (1979) Methods
`in Enzymology, Academic Press, N.Y., Vol. 68, pgs. 109-151. DNA sequences that encode GLP-1 intermediates can
`be designed based on the amino acid sequences herein disclosed. Once designed, the sequence itself may be gen-
`erated using conventional DNA synthesizing apparatus such as the Applied Biosystems Model 380A or 380B DNA
`synthesizers (Applied Biosystems, Inc., 850 Lincoln Center Drive, Foster City, CA 94404).
`[0029] To effect the expression of a biologically-active GPL-1 fragment, one inserts the engineered synthetic DNA
`sequence in any one of many appropriate recombinant DNA expression vectors through the use of appropriate restric-
`tion endonucleases. See generally Maniatis et al. (1989) Molecular Cloning; A Laboratory Manual, Cold Springs Harbor
`Laboratory Press, N.Y., Vol. 1-3. Restriction endonuclease cleavage sites are engineered into either end of the DNA
`encoding the GLP-1 fragment to facilitate isolation from, and integration into, known amplification and expression vec-
`tors. The particular endonucleases employed will be dictated by the restriction endonuclease cleavage pattern of the
`parent expression vector to be employed. The choice of restriction sites are chosen so as to properly orient the coding
`sequence with control sequences to achieve proper in-frame reading and expression of the protein of interest. The
`coding sequence must be positioned so as to be in proper reading frame with the promoter and ribosome binding site
`of the expression vector, both of which are functional in the host cell in which the protein is to be expressed.
`[0030] To achieve efficient transcription of the coding region, it must be operably associated with a promoter-operator
`region. Therefore, the promoter-operator region of the gene is placed in the same sequential orientation with respect
`to the ATG start codon of the coding region.
`[0031] A variety of expression vectors useful for transforming prokaryotic and eukaryotic cells are well known in the
`art. See The Promega Biological Research Products Catalogue (1992) (Promega Corp., 2800 Woods Hollow Road,
`Madison, WI, 53711-5399); and The Stratagene Cloning Systems Catalogue (1992) (Stratagene Corp., 11011 North
`Torrey Pines Road, La Jolla, CA, 92037). Also, U.S. Patent No. 4,710,473 describes circular DNA plasmid transfor-
`mation vectors useful for expression of exogenous genes in E. coli at high levels. These plasmids are useful as trans-
`formation vectors in recombinant DNA procedures and:
`
`(a) confer on the plasmid the capacity for autonomous replication in a host cell;
`
`5
`
`10
`
`15
`
`20
`
`25
`
`30
`
`35
`
`40
`
`45
`
`50
`
`55
`
`5
`
`MYLAN INST. EXHIBIT 1044 PAGE 5
`
`MYLAN INST. EXHIBIT 1044 PAGE 5
`
`

`

`(b) control autonomous plasmid replication in relation to the temperature at which host cell cultures are maintained;
`
`EP 0 699 686 B1
`
`(c) stabilize maintenance of the plasmid in host cell populations;
`
`5
`
`(d) direct synthesis of a protein prod. indicative of plasmid maintenance in a host cell population;
`
`(e) provide in series restriction endonuclease recognition sites unique to the plasmid; and
`
`(f) terminate mRNA transcription.
`
`10
`
`15
`
`20
`
`25
`
`30
`
`35
`
`40
`
`45
`
`50
`
`55
`
`These circular DNA plasmids are useful as vectors in recombinant DNA procedures for securing high levels of expres-
`sion of exogenous genes.
`[0032] Having constructed an expression vector for a biologically-active GLP-1 fragment, the next step is to place
`the vector into a suitable cell and thereby construct a recombinant host cell useful for expressing a biologically-active
`GLP-1 fragment. Techniques for transforming cells with recombinant DNA vectors are well known in the art and may
`be found in such general references as Maniatis, et al. supra. Host cells made be constructed from either eukaryotic
`or prokaryotic cells. Eukaryotic host cells are capable of carrying out post-translational glycosylations on expressed
`proteins and some are capable of secreting the desired protein into the culture medium.
`[0033] Prokaryotic host cells generally produce the protein at higher rates, are easier to culture but are not capable
`of glycosylating the final protein. Proteins which are expressed in high-level bacterial expression systems may aggre-
`gate in granules or inclusion bodies which contain high levels of the overexpressed protein. Such protein aggregates
`must be solubilized, denatured and refolded using techniques well known in the art. See Kreuger, et al. (1990) in Protein
`Folding, Gierasch and King, eds., pgs 136-142, American Association for the Advancement of Science Publication No.
`89-18S, Washington, D.C.; and U.S. Patent No. 4,923,967.
`[0034] Regardless of the methods used to produce a biologically-active GLP-1 fragment, purification of the protein
`generally will be required. Methods for purifying proteins are well known in the art and include conventional chroma-
`tography, including ion and cation exchange, hydrophobic interaction, and immuno-affinity chromatographic media.
`The amino acid sequences herein disclosed in conjunction with well known protein purification methods will enable
`the ordinarily skilled artisan to purify biologically-active GLP-1 fragments claimed herein.
`[0035] The present invention also includes salt forms of biologically-active GLP-1 fragments. A biologically-active
`GLP-1 fragment of the invention may be sufficiently acidic or sufficiently basic to react with any of a number of inorganic
`bases, and inorganic and organic acids, to form a salt. Acids commonly employed to form acid addition salts are
`inorganic acids such as hydrochloric acid, hydrobromic acid, hydroiodic acid, sulfuric acid, phosphoric acid, and the
`like, and organic acids such as p-toluenesulfonic acid, methanesulfonic acid, oxalic acid, p-bromophenyl-sulfonic acid,
`carbonic acid, succinic acid, citric acid, benzoic acid, acetic acid, and the like. Examples of such salts include the
`sulfate, pyrosulfate, bisulfate, sulfite, bisulfite, phosphate, monohydrogenphosphate, dihydrogenphosphate, meta-
`phosphate, pyrophosphate, chloride, bromide, iodide, acetate, propionate, decanoate, caprylate, acrylate, formate,
`isobutyrate, caproate, heptanoate, propiolate, oxalate, malonate, succinate, suberate, sebacate, fumarate, maleate,
`butyne-1,4-dioate, hexyne-1,6-dioate, benzoate, chlorobenzoate, methylbenzoate, dinitrobenzoate, hydroxybenzoate,
`methoxybenzoate, phthalate, sulfonate, xylenesulfonate, phenylacetate, phenylpropionate, phenylbutyrate, citrate, lac-
`tate, gamma-hydroxybutyrate, glycolate,
`tartrate, methanesulfonate, propanesulfonate, naphthalene-1-sulfonate,
`naphthalene-2-sulfonate, mandelate, and the like. Preferred acid addition salts are those formed with mineral acids
`such as hydrochloric acid and hydrobromic acid, and, especially, hydrochloric acid.
`[0036] Base addition salts include those derived from inorganic bases, such as ammonium or alkali or alkaline earth
`metal hydroxides, carbonates, bicarbonates, and the like. Such bases useful in preparing the salts of this invention
`thus include sodium hydroxide, potassium hydroxide, ammonium hydroxide, potassium carbonate, and the like. Salt
`forms of GLP-1 analogs are particularly preferred. Of course, when the compounds of this invention are used for
`therapeutic purposes, those compounds may also be in the form of a salt, but the salt must be pharmaceutically ac-
`ceptable.
`[0037] The inability of a GLP-1 fragment to stimulate insulin secretion may be determined by providing a GLP-1
`fragment to cultured animal cells, such as the RIN-38 rat insulinoma cell line, and monitoring the release of immuno-
`reactive insulin (IRI) into the media. Alternatively one can inject a GLP-1 fragment into an animal and monitor plasma
`levels of immunoreactive insulin (IRI).
`[0038] The presence of IRI is detected through the use of a radioimmunoassay which can specifically detect insulin.
`Any radioimmunoassay capable of detecting the presence of IRI may be employed; one such assay is a modification
`of the method of Albano, J.D.M., et al., Acta Endocrinol., 70:487-509 (1972). In this modification, a phosphate/albumin
`buffer with a pH of 7.4 is employed. The incubation is prepared with the consecutive addition of 500 µl of phosphate
`buffer, 50 µl of perfusate sample or rat insulin standard in perfusate, 100 µl of anti-insulin antiserum (Wellcome Labo-
`
`6
`
`MYLAN INST. EXHIBIT 1044 PAGE 6
`
`MYLAN INST. EXHIBIT 1044 PAGE 6
`
`

`

`EP 0 699 686 B1
`
`ratories; 1:40,000 dilution), and 100 µl of [125I) insulin, giving a total volume of 750 µl in a 10x75 mm disposable glass
`tube. After incubation for 2-3 days at 4° C, free insulin is separated from antibody-bound insulin by charcoal separation.
`The assay sensitivity is 1-2 uU/mL. In order to measure the release of IRI into the cell culture medium of cells grown
`in tissue culture, one preferably incorporates radioactive label into proinsulin. Although any radioactive label capable
`of labeling a polypeptide can be used, it is preferable to use 3H leucine in order to obtain labeled proinsulin.
`[0039] To determine whether a GLP-1 fragment has insulinotropic properties may also be determined by pancreatic
`infusion. The in situ isolated perfused rat pancreas assay is a modification of the method of Penhos, J.C., et al., Dia-
`betes, 18:733-738 (1969). Fasted male Charles River strain albino rats, weighing 350-600 g, are anesthetized with an
`intraperitoneal injection of Amytal Sodium (Eli Lilly and Co.: 160 ng/kg). Renal, adrenal, gastric, and lower colonic
`blood vessels are ligated. The entire intestine is resected except for about four cm of duodenum and the descending
`colon and rectum. Therefore, only a small part of the intestine is perfused, minimizing possible interference by enteric
`substances with glucagon-like immunoreactivity. The perfusate is a modified Krebs-Ringer bicarbonate buffer with 4%
`dextran T70 and 0.2% bovine serum albumin (fraction V), and is bubbled with 95% O2 and 5% CO2. A nonpulsatile
`flow, 4-channel roller bearing pump (Buchler polystatic, Buchler Instruments Division, Nuclear-Chicago Corp.) is used,
`and a switch from one perfusate source to another is accomplished by switching a 3-way stopcock. The manner in
`which perfusion is performed, monitored, and analyzed follow the method of Weir, G.C., et al., J. Clin. Inestigat. 54:
`1403-1412 (1974), which is hereby incorporated by reference.
`[0040] The present invention also provides pharmaceutical compositions comprising a GLP-1 fragment of the present
`invention in combination with a pharmaceutically acceptable carrier, diluent, or excipient. Such pharmaceutical com-
`positions are prepared in a manner well known in the pharmaceutical art, and are administered individually or in com-
`bination with other therapeutic agents, preferably via parenteral routes. Especially preferred routes include intramus-
`cular and subcutaneous administration.
`[0041] Parenteral daily dosages, preferably a single, daily dose, are in the range from about 1 pg/kg to about 1,000
`µg/kg of body weight, although lower or higher dosages may be administered. The required dosage will depend upon
`the severity of the condition of the patient and upon such criteria as the patient's height, weight, sex, age, and medical
`history.
`[0042]
`In making the compositions of the present invention, the active ingredient, which comprises at least one protein
`of the present invention, is usually mixed with an excipient or diluted by an excipient. When an excipient is used as a
`diluent, it may be a solid, semi-solid, or liquid material which acts as a vehicle, carrier, or medium for the active ingre-
`dient.
`[0043]
`In preparing a formulation, it may be necessary to mill the active compound to provide the appropriate particle
`size prior to combining with the other ingredients. If the active protein is substantially insoluble, it ordinarily is milled to
`particle size of less than about 200 mesh. If the active compound is substantially water soluble, the particle size is
`normally adjusted by milling to provide a substantially uniform distribution in the formulation, e.g., about 40 mesh.
`[0044] Some examples of suitable excipients include lactose, dextrose, sucrose, trehalose, sorbitol, mannitol, starch-
`es, gum acacia, calcium silicate, microcrystalline cellulose, polyvinylpyrrolidone, cellulose, water, syrup, and methyl
`cellulose. The formulations can additionally include lubricating agents such as talc, magnesium stearate and mineral
`oil, wetting agents, emulsifying and suspending agents, preserving agents such as methyl- and propylhydroxyben-
`zoates, sweetening agents or flavoring agents. The compositions of the invention can be formulated so as to provide
`quick, sustained or delayed release of the active ingredient after administration to the patient by employing procedures
`well known in the art.
`[0045] The compositions are preferably formulated in a unit dosage form with each dosage normally containing from
`about 50 µg to about 100 mg, more usually from about 1 mg to about 10 mg of the active ingredient. The term "unit
`dosage form" refers to physically discrete units suitable as unitary dosages for human subjects and other mammals,
`each unit containing a predetermined quantity of active material calculated to produce the desired therapeutic effect
`in association with a suitable pharmaceutical excipient.
`[0046] For the purpose of parenteral administration, compositions containing a protein of the present invention pref-
`erably are combined with distilled water and the pH is adjusted to about 6.0 to about 9.0.
`[0047] Additional pharmaceutical methods may be employed to control the duration of action. Controlled release
`preparations may be achieved by the use of polymers to complex or absorb a compound of the present invention. The
`controlled delivery may be exercised by selecting appropriate macromolecules (for example, polyesters, polyamino
`acids, polyvinylpyrrolidone, ethylenevinyl acetate, methylcellulose, carboxymethylcellulose, and protamine sulfate) and
`the concentration of macromolecules as well as the methods of incorporation in order to control release.
`[0048] Another possible method to control the duration of action by controlled release preparations is to incorporate
`a protein of the present invention into particles of a polymeric material such as polyesters, polyamino acids, hydrogels,
`poly (lactic acid) or ethylene vinylacetate copolymers.
`[0049] Alternatively, instead of incorporating a compound into these polymeric particles, it is possible to entrap a
`compound of the present invention in microcapsules prepared, for example, by coacervation techniques or by interfacial
`
`5
`
`10
`
`15
`
`20
`
`25
`
`30
`
`35
`
`40
`
`45
`
`50
`
`55
`
`7
`
`MYLAN INST. EXHIBIT 1044 PAGE 7
`
`MYLAN INST. EXHIBIT 1044 PAGE 7
`
`

`

`EP 0 699 686 B1
`
`polymerization, for example, hydroxymethylcellulose or gelatin-microcapsules, respectively, or in colloidal drug delivery
`systems, for example, liposomes, albumin microspheres, microemulsions, nanoparticles, and nanocapsules, or in mac-
`roemulsions. Such teachings are disclosed in Remington's Pharmaceutical Sciences (1980).
`[0050] Similarly, the present invention provides a method for treating diabetes or hyperglycemia in a mammal, pref-
`erably a human, in need of such treatment comprising administering an effective amount of a GLP-1 fragment or
`composition of the present invention, to such a mammal.
`[0051] By way of illustration, the following examples are provided to help describe how to make and practice the
`various embodiments of the invention. These example are in no way meant to limit the scope of the invention.
`
`5
`
`10
`
`Example 1
`
`Synthesis of GLP-1(9-36)NH2
`
`[0052] GLP-1(9-36)NH2 (SEQ ID NO:9) was produced by solid phase peptide chemistry on an Applied Biosystems
`(ABI) 460A peptide synthesizer using a MBHA resin (Applied Biosystems Inc., lot # AlA023, 0.77 mmol/g). All amino
`acids had their α-amino groups protected by the tert-butyloxycarbonyl (t-Boc) group. Those with reactive side chains
`had them protected as follows: Arg(Tos); Lys(Cl-Z); Trp(CHO); Glu(CHex); Tyr(Br-Z); Ser(Bzl); Asp(OBzl); Thr(Bzl).
`[0053] The protected amino acids were activated in dichloromethane (DCM) with one half an equivalent of dicy-
`clohexylcarbodiimide (DCC) per equivalent of amino acid to give the symmetric anhydride of the amino acid. However,
`arginine, glutamine, and glycine residues were activated by forming the 1-hydroxybenzotriazole (HOBt) esters of these
`amino acids (1:1:1 equivalents of amino acid, HOBt, and DCC in dimtethylformamid

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket