throbber
(12) INTERNATIONAL APPLICATION PUBLISHED UNDER THE PATENT COOPERATION TREATY (PCT)
`
`(19) World Intellectual Property Organization
`International Bureau
`
`Alba. A
`
`0 OO
`
`EOC Et
`
`(43) International Publication Date
`31 January 2008 (31.01.2008)
`
`(10) International Publication Number
`WO 2008/014414 A2
`
`
`
`(51) International Patent Classification:
`AG1K 38/17 (2006.01)
`
`(21) International Application Number:
`PCT/US2007/074514
`
`(22) International Filing Date:
`
`26 July 2007 (26.07.2007)
`
`(25) Filing Language:
`
`(26) Publication Language:
`
`(30) Priority Data:
`60/833,239
`
`English
`
`English
`
`26 July 2006 (26.07.2006)
`
`US
`
`(81) Designated States (unless otherwise indicated, for every
`kind of national protection available): AE, AG, AL, AM,
`AT, AU, AZ, BA, BB, BG, BH, BR, BW, BY, BZ, CA, CH,
`CN, CO, CR, CU, CZ, DE, DK, DM, DO, DZ, EC, EE, EG,
`ES, FI, GB, GD, GE, GH, GM, GT, HN, HR, HU, ID, IL,
`IN, IS, JP, KE, KG, KM, KN, KP, KR, KZ, LA, LC, LK,
`LR, LS, LT, LU, LY, MA, MD, ME, MG, MK, MN, MW,
`MX, MY, MZ, NA, NG, NI, NO, NZ, OM, PG, PH, PL,
`PT, RO, RS, RU, SC, SD, SE, SG, SK, SL, SM, SV, SY,
`TJ, TM, TN, TR, TT, TZ, UA, UG, US, UZ, VC, VN, ZA,
`ZIM, ZW.
`
`US):
`except
`States
`designated
`all
`(for
`(71) Applicant
`BIOMARCK PHARMACEUTICALS, LTD. [US/US];
`7200 Falls of Neuse Road, Suite 202, Raleigh, North
`Carolina 27615 (US).
`
`(72) Inventor; and
`PARIKH, Indu
`(75) Inventor/Applicant (for US only):
`[US/US]; 2558 Booker Creek Road, Chapel Hill, North
`Carolina 27514 (US).
`
`(84) Designated States (unless otherwise indicated, for every
`kind of regional protection available): ARIPO (BW, GH,
`GM, KE, LS, MW, MZ, NA, SD, SL, SZ, TZ, UG, ZM,
`ZW), Eurasian (AM, AZ, BY, KG, KZ, MD, RU, TJ, TM),
`European (AT, BE, BG, CH, CY, CZ, DE, DK, EE, ES, FI,
`FR, GB, GR, HU, IE, IS, IT, LT, LU, LV, MC, MT, NL, PL,
`PT, RO, SE, SI, SK, TR), OAPI (BF, BJ, CF, CG, CI, CM,
`GA, GN, GQ, GW, ML, MR, NE, SN, TD, TG).
`
`(74) Agents: HULEATT, Jayme A. et al.; Cooley Godward
`Kronish LLP, Attn: Patent Group, 1200 19th Street, N.W.,
`Suite 500, Washington, District Of Columbia 20036 (US).
`
`Published:
`without international search report and to be republished
`upon receipt of that report
`
`[Continued on next page]
`
`
`
`(54) Title: METHODS FOR ATTENUATING RELEASE OF INFLAMMATORY MEDIATORS AND PEPTIDES USEFUL
`THEREIN
`
`MPO Secretion from LPS-Primed Human Neutrophils
`
`0.354
`
`0.30
`
`0.254
`
`0.20
`
`0.15
`
`0.104
`
`0.05
`
` 0.00
`
`
`
`Absorbance 450am
`
`10 uM 8-Br-cGMP
`+ PMA (nM)
`Treatments
`
`087014414 A2 | IN IV 0000 OO OO
`
`=
`
`(57) Abstract: The present invention includes methods of inhibiting or suppressing cellular secretory processes. More specifically
`& the present invention relates to inhibiting or reducing the release of inflammatory mediators from inflammatory cells by inhibiting
`the mechanism associated with the release of inflammatory mediators from granules in inflammatory cells. In this regard, the present
`invention discloses an intracellular signaling mechanism that illustrates several novel intracellular targets for pharmacological in-
`tervention in disorders involving secretion of inflammatory mediators from vesicles in inflammatory cells. Peptide fragments and
`variants thereof of MANS peptide as disclosed in the present invention are useful in such methods.
`
`
`
`
`MYLAN - EXHIBIT 1030
`
`

`

`
`
`WO 2008/014414 A 2
`
`{NII DA 000 000000 0000 0
`
`—
`
`with sequence listing part of description published sepa-
`rately in electronic form and available upon request from
`the International Bureau
`
`For two-letter codes and other abbreviations, refer to the "Guid-
`ance Notes on Codes and Abbreviations" appearing at the begin-
`ning of each regular issue of the PCT Gazette.
`
`

`

`WO 2008/014414
`
`PCT/US2007/074514
`
`METHODS FOR ATTENUATING RELEASE OF INFLAMMATORY
`
`MEDIATORS AND PEPTIDES USEFUL THEREIN
`
`Cross Reference to Related Application
`
`[0001]
`
`The present application claims priority to U.S. Serial No.: 60/833,239 filed on July 26,
`
`20006, which is incorporated in its entirety by reference.
`
`Field of Invention
`
`[0002]
`
`The present invention relates to peptides or peptide compositions and methods of their
`
`use to attenuate (or inhibit or reduce) the stimulated release of mediators of inflammation from
`
`inflammatory cells during inflammation.
`
`The present invention also relates to use of these
`
`peptides or peptide compositions to modulate an intracellular signaling mechanism regulating the
`
`secretion of inflammatory mediators from inflammatory cells.
`
`Background of the Invention
`
`[0003]
`
`Inflammatory leukocytes synthesize a number of inflammatory mediators that
`
`are
`
`isolated intracellularly and stored in cytoplasmic membrane-bound granules. Examples of such
`
`mediators include, but are not limited to, myeloperoxidase [MPO] in neutrophils (see,
`
`for
`
`example, Borregaard N, Cowland JB. Granules of the human neutrophilic polymorphonuclear
`
`leukocyte. Blood 1997; 89:3503-3521), eosinophil peroxidase [EPO] and major basic protein
`
`[MBP] in eosinophils (see,
`
`for example, Gleich G J. Mechanisms of eosinophil-associated
`
`inflammation.
`
`J
`
`Allergy ~~ Clin
`
`Immunol ~~ 2000;
`
` 105:651-663),
`
`lysozyme
`
`in
`
`monocytes/macrophages (see, for example, Hoff T, Spencker T, Emmendoerffer A., Goppelt-
`
`Struebe M. Effects of glucocorticoids on the TPA-induced monocytic differentiation. J Leukoc
`
`Biol
`
`1992;
`
`52:173-182;
`
`and
`
`Balboa
`
`M A,
`
`Saez
`
`Y,
`
`Balsinde
`
`J.
`
`Calcium-independent
`
`phospholipase A2 is required for lysozyme secretion in U937 promonocytes. J Immunol 2003;
`
`170:5276-5280), and granzyme in natural killer (NK) cells and cytotoxic lymphocytes (see, for
`
`example, Bochan MR, Goebel WS, Brahmi Z.
`
`Stably transfected antisense granzyme B and
`
`perforin constructs inhibit human granule-mediated lytic ability. Cell Immunol 1995;164:234-
`
`239; Gong JH., Maki G, Klingemann HG. Characterization of a human cell line (NK-92) with
`
`phenotypical and functional characteristics of activated natural killer cells. Leukemia 1994;
`
`
`
`

`

`WO 2008/014414
`
`PCT/US2007/074514
`
`8:652-658; Maki G, Klingemann HG, Martinson JA, Tam YK. Factors regulating the cytotoxic
`
`activity of the human natural killer cell line, NK-92. J Hematother Stem Cell Res 2001; 10:369-
`
`383; and Takayama H, Trenn G,
`
`Sitkovsky MV. A novel cytotoxic T lymphocyte activation
`
`assay. J Immunol Methods 1987; 104:183-190).
`
`Such mediators are released at sites of injury
`
`and contribute to inflammation and tissue repair such as in the lung and elsewhere.
`
`It is known
`
`that leukocytes release these granules via an exocytotic mechanism (see, for example, Burgoyne
`
`RD, Morgan A. Secretory granule exocytosis. Physiol Rev 2003; 83:581-632; and Logan MR,
`
`Odemuyiwa SO, Mogbel R. Understanding exocytosis in immune and inflammatory cells: the
`
`molecular basis
`
`of mediator secretion. J Allergy Clin Immunol 2003;
`
`111: 923-932), but
`
`regulatory molecules and specific pathways involved in the exocytotic process have not been
`
`fully described.
`
`[0004]
`
`Several exogenous stimuli can provoke degranulation of leukocytes via a pathway that
`
`involves activation of protein kinase C and subsequent phosphorylation events (see, for example,
`
`Burgoyne RD, Morgan A. Secretory granule exocytosis. Physiol Rev 2003; 83:581-632; Logan
`
`MR, Odemuyiwa SO, Mogbel R. Understanding exocytosis in immune and inflammatory cells:
`
`the molecular basis of mediator secretion. J Allergy Clin Immunol 2003; 111: 923-932; Smolen
`
`JE, Sandborg RR. Ca2+-induced secretion by electropermeabilized human neutrophils: the roles
`
`of Ca2+, nucleotides and protein kinase C. Biochim Biophys Acta 1990; 1052:133-142; Niessen
`
`HW,
`
`Verhoeven
`
`AJ.
`
`Role
`
`of
`
`protein
`
`phosphorylation
`
`in
`
`the
`
`degranulation
`
`of
`
`electropermeabilized human neutrophils. Biochim. Biophys. Acta
`
`1994; 1223:267-273; and
`
`Naucler C, Grinstein S, Sundler R., Tapper H. Signaling to localized degranulation in neutrophils
`
`adherent to immune complexes. J Leukoc Biol 2002; 71:701-710).
`
`[0005]
`
`MARCKS protein (where MARCKS as used herein means “Myristoylated Alanine-
`
`Rich C Kinase Substrate™), is a ubiquitous phosphorylation target of protein kinase C (PKC), and
`
`is highly expressed in leukocytes (see, for example, Aderem AA, Albert KA, Keum MM, Wang
`
`JK, Greengard P, Cohn ZA. Stimulus-dependent myristoylation of a major substrate for protein
`
`kinase C. Nature 1988; 332:362-364; Thelen M, Rosen A, Nairn AC, Aderem A. Regulation by
`
`phosphorylation of reversible association of a myristoylated protein kinase C substrate with the
`
`plasma membrane. Nature 1991; 351:320-322; and Hartwig JH, Thelen M, Rosen A, Janmey
`
`PA, Nairn AC, Aderem A. MARCKS is an actin filament crosslinking protein regulated by
`
`
`
`

`

`WO 2008/014414
`
`PCT/US2007/074514
`
`protein kinase C and calcium-calmodulin. Nature 1992; 356:618-622. MARCKS protein is
`
`mechanistically involved in a process of exocytotic secretion of mucin by goblet cells that line
`
`respiratory airways (see, for example, Li et al., J Biol Chem 2001; 276:40982-40990; and Singer
`
`et al.,, Nat Med 2004; 10:193-196). MARCKS is myristoylated via an amide bond at the N-
`
`terminal amino acid in the MARCKS protein’s amino acid sequence at the alpha-amine position
`
`of the glycine which resides at the N-terminus (i.e.,
`
`at position 1) of amino acid sequence. In
`
`airway epithelial cells, the myristoylated N-terminal region of MARCKS appears to be integral
`
`to the secretory process.
`
`By the N-terminus of the MARCKS protein is meant the MANS
`
`peptide which contains Myristoyl-GAQFSKTAAKGEAAAERPGEAAVA (SEQ ID NO: 1),
`
`which are L-amino acids.
`
`Additionally, the peptide fragments of the MANS peptide disclosed
`
`herein, also preferably are composed of L-amino acids.
`
`The mechanism appears to involve
`
`binding of MARCKS, a myristoylated protein, to membranes of intracellular granules.
`
`[0006]
`
`An N-terminal myristoylated peptide from the N-terminus of MARCKS has been
`
`shown to block both mucin secretion and binding of MARCKS to mucin granule membranes in
`
`goblet cells (see, for example, Singer et al., Nat Med 2004; 10:193-196). This peptide contains
`
`24 amino acids of the MARCKS protein beginning with the N-terminal glycine of the MARCKS
`
`protein which is myristoylated via an amide bond and is known as myristoylated alpha-N-
`
`terminal sequence (MANS); i.e., Myristoyl-GAQFSKTAAKGEAAAERPGEAAVA (SEQ ID
`
`NO: 1). Also Vergeres et al., J. Biochem. 1998, 330; 5-11, discloses that the N-terminal glycine
`
`residue of MARCKS proteins is myristoylated via a reaction catalyzed by myristoyl CoA:protein
`
`N-myristoyl transferase (NMT).
`
`[0007]
`
`In inflammatory diseases, such as asthma, COPD and chronic bronchitis; in genetic
`
`diseases such
`
`as
`
`cystic
`
`fibrosis;
`
`in
`
`allergic conditions (atopy,
`
`allergic inflammation); in
`
`bronchiectasis; and in
`
`a number of acute, infectious respiratory illnesses such as pneumonia,
`
`rhinitis, influenza or the common cold, arthritis or auto-immune diseases, inflammatory cells are
`
`usually found in or migrate to areas of injury or infection associated with inflammatory disease
`
`states, especially in or to respiratory passages or airways of patients suffering from such diseases.
`
`These inflammatory cells can contribute greatly to the pathology of diseases via tissue damage
`
`done by inflammatory mediators released from these cells. One example of such tissue damage
`
`or destruction via this chronic inflammation occurs in cystic fibrosis patients where mediators
`
`
`
`

`

`WO 2008/014414
`
`PCT/US2007/074514
`
`released from neutrophils (e.g., myeloperoxidase [MPO]) induce the desquamation of the airway
`
`epithelial tissue.
`
`[0008]
`
`MARCKS, a protein of approximately 82 kD, has three evolutionarily-conserved
`
`regions (Aderem et
`
`al., Nature 1988; 332:362-364; Thelen et
`
`al., Nature 1991; 351:320-322;
`
`Hartwig et al., Nature 1992; 356:618-622; Seykora et al., J Biol Chem 1996; 271:18797-18802):
`
`an N-terminus, a phosphorylation site domain (or PSD), and a multiple homology 2 (MH2)
`
`domain. Human MARCKS cDNA and protein is known and reported by Harlan et al.,
`
`J. Biol.
`
`Chem. 1991, 266:14399 (GenBank Accession No. M68956) and also by Sakai er al., Genomics
`
`1992, 14: 175. These sequences are also provided in a WO 00/50062, which is incoporated in its
`
`entirety by reference. The N-terminus, an alpha-amino acid sequence comprising 24 amino acid
`
`residues with a myristic acid moiety attached via an amide bond to the N-terminal glycine
`
`residue is involved in binding of MARCKS to membranes in cells (Seykora et al., J Biol Chem
`
`1996; 271:18797-18802) and possibly to calmodulin (Matsubara et
`
`al.,
`
`J Biol Chem 2003;
`
`278:48898-48902). This 24 amino acid sequence is known as the MANS peptide.
`
`Summary of the Invention
`
`[0009]
`
`Involvement of MARCKS protein in release of inflammatory mediators from the
`
`granules of infiltrating leukocytes is relevant to inflammation in diseases in
`
`all tissues and
`
`organs, including lung diseases characterized by airway inflammation, such as asthma, COPD
`
`and cystic fibrosis. However, inflammation and mucus secretion in the airways are two separate
`
`and independent processes (Li et al., J Biol Chem 2001; 276:40982-40990; Singer et al., Nut Med
`
`2004; 10:193-196). While mucus production and secretion can be provoked by a number of
`
`factors, including mediators released by inflammatory cells, there is no known direct link
`
`whereby excess mucus causes inflammation.
`
`[00010] In one aspect of this invention, the MANS peptide can play a role in the reducing the
`
`rate and/or amount of release of inflammatory mediators granules or vesicles in inflammatory
`
`leukocytes.
`
`[00011]
`
`In another aspect, peptides derived from the MARCKS N-terminus, especially from the
`
`24 amino acid N-terminal sequence, i.e., active contiguous peptide fragments derived from
`
`
`
`

`

`WO 2008/014414
`
`PCT/US2007/074514
`
`within the N-terminal 1-to-24 amino acid sequence of MARCKS having a glycine at position 1,
`
`as well as N-terminal amides of such fragments, such as N-terminal acetic acid amides of such
`
`fragments, and/or as well as C-terminal amides of such fragments, such as C-terminal amides of
`
`ammonia, can inhibit or reduce the rate and/or amount of release of inflammatory mediators
`
`from inflammatory leukocytes. Such inhibition or reduction in release comprises inhibition of a
`
`MARCKS-related release of inflammatory mediators from inflammatory leukocytes.
`
`[00012]
`
`In another aspect, peptides derived from the MARCKS N-terminus, especially from the
`
`1-to-24 amino acid N-terminal sequence, i.e., active contiguous peptide fragments derived from
`
`within the N-terminal 1 to 24 amino acid sequence of MARCKS having a glycine at position 1,
`
`as well as N-terminal amides of such fragments such as N-terminal acetic acid amides of such
`
`fragments, and as well as C-terminal amides of such fragments such as C-terminal amides of
`
`ammonia, can inhibit the rate of release and/or amount of release of inflammatory mediators
`
`such as those identified herein in this invention, by inhibiting the process of degranulation in
`
`inflammatory leucocytes.
`
`[00013]
`
`In another aspect, the MANS peptide and active fragments thereof, and active amides
`
`of such fragments as described herein, can compete for membrane binding in inflammatory cells
`
`with native MARCKS protein to attenuate (lessen or reduce) MARCKS-related release of
`
`mediators of inflammation from granules or vesicles containing such mediators of inflammation
`
`in such inflammatory cells.
`
`[00014] Leukocyte cell types and model cell types that secrete specific granule contents in
`
`response to phorbol ester induced activation of PKC are useful for in vitro demonstration of
`
`efficacy of peptides of this invention and of substituted peptides (e.g., alpha-N-amides, C-
`
`terminal amides and esters) of this invention.
`
`[00015] The attenuation of release of membrane-bound inflammatory mediators by compounds
`
`and compositions of this invention can be demonstrated using human leukocyte cell lines. For
`
`example, neutrophils isolated from human blood can be used to demonstrate attenuation or
`
`inhibition of release of myeloperoxidase (MPO). The human promyelocytic cell
`
`line HL-60
`
`clone 15 can be used to demonstrate attenuation of release or inhibition of release or secretion of
`
`eosinophil peroxidase (EPO) by compounds and compositions of this invention (see,
`
`for
`
`
`
`

`

`WO 2008/014414
`
`PCT/US2007/074514
`
`example, Fischkoff SA. Graded increase in probability of eosinophilic differentiation of HL-60
`
`promyelocytic leukemia cells induced by culture under alkaline conditions. Leuk Res 1988;
`
`12:679-686; Rosenberg HF, Ackerman S
`
`J, Tenen DG. Human eosinophil cationic protein:
`
`molecular cloning of a cytotoxin and helminthotoxin with ribonuclease activity. J Exp Med
`
`1989;
`
`170:163-176; Tiffany HL,
`
`Li
`
`F, Rosenberg HF. Hyperglycosylation of eosinophil
`
`ribonucleases in
`
`a promyelocytic leukemia cell
`
`line and in differentiated peripheral blood
`
`progenitor cells. J Leukoc Biol 1995; 58:49-54; and Badewa AP, Hudson CE, Heiman AS.
`
`Regulatory effects
`
`of eotaxin,
`
`eotaxin-2,
`
`and eotaxin-3
`
`on eosinophil degranulation and
`
`superoxide anion generation. Exp Biol Med 2002; 227:645-651). The monocytic leukemia cell
`
`line U937 can be used to demonstrate attenuation of release or inhibition of release or secretion
`
`of lysozyme by compounds and compositions of this invention (see,
`
`for example, Hoff T,
`
`Spencker T, Emmendoerffer A., Goppelt-Struebe M. Effects of glucocorticoids on the TPA-
`
`induced monocytic differentiation. J Leukoc Biol 1992; 52:173-182; Balboa M A, Saez Y,
`
`Balsinde J. Calcium-independent phospholipase A2 is required for lysozyme secretion in U937
`
`promonocytes. J Immunol 2003; 170:5276-5280; and Sundstrom C, Nilsson K. Establishment
`
`and characterization of a human histiocytic lymphoma cell line (U-937). Int J Cancer 1976;
`
`17:565-577). The lymphocyte natural
`
`killer
`
`cell
`
`line NK-92 can be used to demonstrate
`
`attenuation or inhibition of release of granzyme by compounds and compositions of this
`
`invention (see, for example, Gong JH., Maki G, Klingemann HG. Characterization of a human
`
`cell line (NK-92) with phenotypical and functional characteristics of activated natural killer cells.
`
`Leukemia 1994; 8:652-658; Maki G, Klingemann HG, Martinson JA, Tam YK.
`
`Factors
`
`regulating the cytotoxic activity of the human natural killer cell line, NK-92. J Hematother Stem
`
`Cell Res 2001; 10:369-383; and Takayama H, Trenn G,
`
`Sitkovsky MV. A novel cytotoxic T
`
`lymphocyte activation assay. J Immunol Methods 1987; 104:183-190). In an in vitro method to
`
`inhibit or attenuate the release of a mediator of inflammation such as those described herein,
`
`each of the cell types is preincubated with a peptide compound or peptide composition of this
`
`invention over a range of concentrations followed by incubation of these cells by a stimulator of
`
`release of inflammatory mediators, such as phorbol ester. The percent of inhibition of release of
`
`a mediator of inflammation is determined as compared to the release of the mediator in the
`
`absence of the peptide compound or peptide composition, such as in a specrophotometric readout
`
`of a concentration of the mediator released.
`
`
`
`

`

`WO 2008/014414
`
`PCT/US2007/074514
`
`[00016] To demonstrate the importance of the relative amino acid sequence positioning in the
`
`peptides of the invention, the relative ability to inhibit or reduce the amount of inflammatory
`
`mediator released by a peptide which is identical to the 24 amino acid sequence of the MARCKS
`
`protein N-terminus region (i.e., the MANS- myristoylated alpha-N-terminal sequence peptide)
`
`was compared to the ability to inhibit or reduce the amount of inflammatory mediator released
`
`by a peptide containing the same 24 amino acid residues present in MANS but which are
`
`sequenced in
`
`a random order (i.e., an RNS peptide, otherwise referred to
`
`as a “Random N-
`
`terminal sequence peptide”) with respect to the sequence order in MANS. In each of the cell
`
`types examined, the MANS peptide, but not the RNS peptide, attenuated release of inflammatory
`
`mediators in a concentration-dependent manner over a time course of 0.5-3.0 hrs. These results
`
`suggest that the relative amino acid sequence positioning in the peptides of the invention which
`
`are in the order found in the MARCKS protein, specifically its N-terminal region, and more
`
`specifically its 24 amino acid residue N-terminal region are involved in at least one intracellular
`
`pathway dealing with the inhibition of leukocyte degranulation.
`
`[00017] The invention relates to a new use for the 24 amino acid peptide sequence, and to the
`
`alpha-N-terminal acetylated peptide sequence, the myristoylated polypeptide, also known as the
`
`MANS peptide, and to active fragments thereof, which active fragments can be selected from the
`
`group of peptides having from 4 to 23 contiguous amino acid residues of the MANS peptide
`
`amino acid sequence, and which fragments may be N-terminal-myristoylated if they do not begin
`
`with the N-terminal glycine at position 1 in SEQ ID NO: 1, or which may be N-terminal-acylated
`
`with C2 to C12 acyl groups, including N-terminal-acetylated, and/or C-terminal amidated with
`
`an NH2 group.
`
`[00018] The invention also relates to
`
`a new method for blocking MARCKS-related cellular
`
`secretory processes, especially those that involve the MARCKS-related release of inflammatory
`
`mediators from inflammatory cells, whose stimulatory pathways involve the protein kinase C
`
`(PKC) substrate MARCKS protein
`
`and release of contents from intracellular vesicles or
`
`granules.
`
`[00019] The present invention is directed to a method of inhibiting the exocytotic release of at
`
`least one inflammatory mediator from at least one inflammatory cell comprising contacting the at
`
`
`
`

`

`WO 2008/014414
`
`PCT/US2007/074514
`
`least one inflammatory cell, which cell comprises at least one inflammatory mediator contained
`
`within a vesicle inside the cell, with at least one peptide selected from the group consisting of a
`
`MANS peptide and an active fragment thereof as described herein in an effective amount to
`
`reduce the release of the inflammatory mediator from the inflammatory cell as compared to the
`
`release of the inflammatory mediator from the same type of inflammatory cell that would occur
`
`in the absence of the at least one peptide.
`
`[00020] The present invention is further directed to a method of inhibiting the release of at least
`
`one inflammatory mediator from at least one inflammatory cell in a tissue or fluid of a subject
`
`comprising the administration to the subject’s tissue and/or fluid, which comprises at least one
`
`inflammatory cell comprising at least one inflammatory mediator contained within a vesicle
`
`inside the cell, a therapeutically effective amount of a pharmaceutical composition comprising at
`
`least one peptide selected from the group consisting of a MANS peptide and an active fragment
`
`thereof in a therapeutically effective amount to reduce the release of the inflammatory mediator
`
`from at least one inflammatory cell as compared to release of the inflammatory mediator from at
`
`least one of the same type of inflammatory cell that would occur in the absence of the at least
`
`one peptide. More specifically, inhibiting the release of an inflammatory mediator comprises
`
`blocking or reducing the release of an inflammatory mediator from the inflammatory cell.
`
`[00021]
`
`More particularly, the present invention includes a method of reducing inflammation
`
`in
`
`a
`
`subject comprising the
`
`administration
`
`of
`
`a therapeutically
`
`effective amount of
`
`a
`
`pharmaceutical ~~ composition
`
`comprising
`
`a
`
`MANS
`
`peptide
`
`(i.e,
`
`N-myristoyl-
`
`GAQFSKTAAKGEAAAERPGEAAVA (SEQ ID NO: 1)) or an active fragment thereof. The
`
`active fragment is at least four and preferably at least six amino acids in length. As used herein,
`
`an "active fragment" of a MARCKS protein is one that affects (inhibits or reduces) MARCKS
`
`protein-mediated release, such as MARCKS protein-mediated release of an inflammatory
`
`mediator.
`
`An
`
`active
`
`fragment
`
`can
`
`be
`
`selected
`
`from
`
`the
`
`group
`
`consisting
`
`of
`
`GAQFSKTAAKGEAAAERPGEAAV (SEQ ID NO: 2); GAQFSKTAAKGEAAAERPGEAA
`
`(SEQ
`
`ID
`
`NO:
`
`4);
`
`GAQFSKTAAKGEAAAERPGEA
`
`(SEQ
`
`ID
`
`NO:
`
`7);
`
`GAQFSKTAAKGEAAAERPGE (SEQ ID NO: 11); GAQFSKTAAKGEAAAERPG (SEQ ID
`
`NO: 16); GAQFSKTAAKGEAAAERP (SEQ ID NO: 22); GAQFSKTAAKGEAAAER (SEQ
`
`ID NO: 29); GAQFSKTAAKGEAAAE (SEQ ID NO: 37); GAQFSKTAAKGEAAA (SEQ ID
`
`
`
`
`
`

`

`WO 2008/014414
`
`PCT/US2007/074514
`
`NO: 46); GAQFSKTAAKGEAA (SEQ ID NO: 56); GAQFSKTAAKGEA (SEQ ID NO: 67);
`
`GAQFSKTAAKGE (SEQ ID NO: 79); GAQFSKTAAKG (SEQ ID NO: 92); GAQFSKTAAK
`
`(SEQ ID NO: 106); GAQFSKTAA (SEQ ID NO: 121); GAQFSKTA (SEQ ID NO: 137);
`
`GAQFSKT (SEQ ID NO: 154); GAQFSK (SEQ ID NO: 172); GAQFS (SEQ ID NO: 191) and
`
`GAQF (SEQ ID NO: 211). These peptides, instead of containing a myristoyl moiety at the N-
`
`terminal amino acid, either contain no chemical moiety or a non-myristoyl chemical moiety at
`
`the N-terminal amino acid and/or a chemical moiety at the C-terminal amino acid, such as an N-
`
`terminal acetyl group and/or a C-terminal amide group as described herein. The presence of the
`
`hydrophobic N-terminal myristoyl moiety in the MANS peptides and N-terminal myristoylated
`
`fragments thereof can enhance their compatibility with and presumably their permeability to
`
`plasma membranes, and potentially enable the peptides to be taken up by cells. The hydrophobic
`
`insertion of a myristoyl group into a membrane lipid bilayer can provide a partition coefficient or
`
`apparent association constant with lipids of up to 10* M™! or a unitary Gibbs free binding energy
`
`of about 8 kcal/mol (see, for example, Peitzsch, R.M., and McLaughlin, S. 1993, Binding of
`
`acylated peptides and fatty acids to phospholipid vesicles: pertinence to myristoylated proteins.
`
`Biochemistry. 32: 10436-10443) which is sufficient, at least in part, to permit a partitioning of
`
`the MANS peptide and of myristoylated MANS peptide fragments into the plasma membrane of
`
`a cell while additional functional groups and their interactions within the MANS peptide (which
`
`is myristoylated) and within myristoylated MANS peptide fragments can potentiate their relative
`
`membrane permeabilities. The fragments can each exhibit partition coefficients and membrane
`
`affinities that are representative of their respective structure. The fragments can be prepared by
`
`methods of peptide synthesis known in the art, such as by solid phase peptide synthesis (see, for
`
`example, the methods described in Chan, Weng C. and White, Peter D.Eds., Fmoc Solid Phase
`
`Peptide Synthesis: A Practical Approach, Oxford University Press, New York, New York
`
`(2000); and Lloyd-Williams, P.
`
`et
`
`al. Chemical Approaches to the Synthesis of Peptides and
`
`Proteins (1997)) and purified by methods known in the art, such as by high pressure liquid
`
`chromatography. Molecular weight of each peptide can be confirmed by mass spectroscopy with
`
`each showing a peak with an appropriate molecular mass. Efficacy of the individual peptides and
`
`of combinations of individual peptides (for example, combinations of 2
`
`of the peptides,
`
`combinations of 3 of the peptides, combinations of 4 of the peptides) in the methods of this
`
`disclosure can be readily determined without undue experimentation using the procedures
`
`
`
`
`
`

`

`WO 2008/014414
`
`PCT/US2007/074514
`
`described in the examples disclosed herein. A preferred combination will comprise two of the
`
`peptides; a preferred molar ratio of the peptides can be from 50:50 (i.e., 1:1) to 99.99 to 0.01,
`
`which ratio can be readily determined using the procedures described in the examples disclosed
`
`herein.
`
`[00022]
`
`Preferably
`
`the MANS peptide
`
`or
`
`active
`
`fragment
`
`thereof is
`
`contained
`
`in
`
`a
`
`pharmaceutical composition which is useful to block inflammation, The present invention also
`
`includes methods for inhibiting a cellular secretory process in
`
`a subject comprising the
`
`administration of a therapeutically effective amount of a compound comprising a MANS peptide
`
`or
`
`an
`
`active fragment thereof,
`
`that inhibits
`
`an inflammatory mediator in
`
`a subject. The
`
`administration is generally selected from the group consisting of topical
`
`administration,
`
`parenteral administration, rectal administration, pulmonary administration, inhalation and nasal
`
`or oral administration, wherein pulmonary administration generally includes either an aerosol, a
`
`dry powder inhaler, a metered dose inhaler, or a nebulizer.
`
`[00023] Administration of a composition comprising a degranulation-inhibiting amount of the
`
`MANS peptide or a degranulation-inhibiting amount of an active fragment thereof, such as a
`
`pharmaceutical composition of the MANS peptide or an active fragment thereof, for human or
`
`animal use provides the MANS peptide or active fragment thereof at least to the site in or on a
`
`tissue or to a fluid-containing layer in contact with the surface of a tissue where an inflammatory
`
`granulocytic cell resides or into which an inflammatory granulocytic cell
`
`will invade, thus
`
`enabling the MANS peptide or
`
`an
`
`active fragment thereof to contact the inflammatory
`
`granulocytic cell. In one aspect, administration of such a composition can be made at the first
`
`onset or first detection of inflammation or first perception of inflammation by the human or
`
`animal or at the first perceptible change in the level of inflammation in a human or animal to
`
`reduce the amount of inflammation that would otherwise occur in the absence of the MANS
`
`peptide or active fragment thereof. In another aspect, administration can be made during an
`
`ongoing inflammation of a tissue in the human or animal to reduce the amount of additional
`
`inflammation that would otherwise occur in the absence of the MANS peptide or active fragment
`
`thereof. While the amount and frequency of dose can be determined by clinical evaluation and be
`
`a function of the disease or source of inflammation and the extent of tissue involved and the age
`
`and size of the patient, it
`
`is anticipated that dosing of a pharmaceutical composition can be
`
`10
`
`
`
`

`

`WO 2008/014414
`
`PCT/US2007/074514
`
`repeated after 3
`
`to
`
`§ hours, preferably after 6
`
`to
`
`8 hours after the first administration of the
`
`pharmaceutical composition.
`
`[00024] The present invention also includes methods of reducing inflammation in
`
`a subject
`
`comprising the administration of a therapeutically effective amount of a compound that inhibits
`
`the MARCKS-related release of inflammatory mediators, whereby the release of at least one
`
`inflammatory mediator in the subject is reduced compared to that which would occur in the
`
`absence of said treatment. As used herein "reducing" generally means a lessening of the effects
`
`of inflammation. Preferably, release of inflammatory mediators are inhibited or blocked by the
`
`methods disclosed.
`
`[00025] Another embodiment of
`
`the
`
`present
`
`invention
`
`includes
`
`methods
`
`of reducing
`
`inflammation in
`
`a subject comprising administering a therapeutically effective amount of a
`
`compound that inhibits the MARCKS-related release of inflammatory mediators, whereby the
`
`inflammation in the subject is reduced compared to that which would occur in the absence of
`
`said
`
`treatment.
`
`The present
`
`invention
`
`also
`
`discloses methods of reducing or
`
`inhibiting
`
`inflammation in a subject comprising the administration of a therapeutically effective amount of
`
`a MANS peptide or an active fragment thereof effective to inhibit an inflammatory mediator at
`
`the inflammation site. The term "inhibiting" means a reduction in the amount of inflammatory
`
`mediator secretion. The term “completely inhibiting” means a reduction to zero in the amount of
`
`inflammatory mediator secretion. Again, as stated above, the active fragment is at least four and
`
`preferably at least six amino acids in length. The term “exocytotic process” means exocytosis,
`
`1.e., a process of cellular secretion or excretion in which substances contained in a vesicle, which
`
`vesicle resides inside a cell, are discharged from the cell by fusion of the vesicular membrane of
`
`the vesicle with the outer cell membrane. “Degranulation” means the release of cellular granule
`
`contents.
`
`The term “degranulation-

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