AU779507B2 - Use of insulin-like growth factors I and II for inhibition of inflammatory response - Google Patents

Use of insulin-like growth factors I and II for inhibition of inflammatory response Download PDF

Info

Publication number
AU779507B2
AU779507B2 AU29233/02A AU2923302A AU779507B2 AU 779507 B2 AU779507 B2 AU 779507B2 AU 29233/02 A AU29233/02 A AU 29233/02A AU 2923302 A AU2923302 A AU 2923302A AU 779507 B2 AU779507 B2 AU 779507B2
Authority
AU
Australia
Prior art keywords
igf
cells
class
inflammatory response
dose
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.)
Ceased
Application number
AU29233/02A
Other versions
AU2923302A (en
Inventor
Philip Francis Halloran
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.)
Novartis AG
Original Assignee
Novartis AG
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
Priority claimed from AU42376/99A external-priority patent/AU4237699A/en
Application filed by Novartis AG filed Critical Novartis AG
Priority to AU29233/02A priority Critical patent/AU779507B2/en
Publication of AU2923302A publication Critical patent/AU2923302A/en
Application granted granted Critical
Publication of AU779507B2 publication Critical patent/AU779507B2/en
Priority to AU2005201558A priority patent/AU2005201558A1/en
Anticipated expiration legal-status Critical
Ceased legal-status Critical Current

Links

Description

Our Ref:7687830 P/00/011 Regulation 3:2
AUSTRALIA
Patents Act 1990
ORIGINAL
COMPLETE SPECIFICATION STANDARD PATENT Applicant(s): Address for Service: Novartis AG Schwarzwaldallee 215 CH-4058 Basel Switzerland DAVIES COLLISON CAVE Patent Trade Mark Attorneys Level 10, 10 Barrack Street SYDNEY NSW 2000 Invention Title: Use of insulin-like growth factors I and II for inhibition of inflammatory response The following statement is a full description of this invention, including the best method of performing it known to me:- -1- USE OF INSULIN-LIKE GROWTH FACTORS I AND II FOR INHIBITION OF INFLAMMATORY RESPONSE This invention relates to the use of insulin-like growth factor (IGF), including IGF-I and IGF- II, for inhibition of inflammatory response such as in rheumatoid arthritis, psoriatic arthritis, Reiter's syndrome, and other known inflammatory arthritis, as well as inflammatory bowel disease. This invention also relates to inhibition of ischemic injury and inhibition of organ rejection upon transplantation.
Insulin-like growth factors, IGF-I and IGF-II, each have a molecular weight of about 7,500 daltons. Each of IGF-I and IGF-II possesses A and B domains that are highly homologous to the corresponding domains of proinsulin. A and B domains are connected to each other S: 'by a C domain. A carboxy terminal extension, the D domain, is present in IGF but is not found in proinsulin. Both IGF-I and IGF-II are single-chain polypeptides each with 3 disulfide bridges and have a sequence identity of 49% and 47%, respectively, to human insulin A chain and B chain. Like insulin, IGF stimulate phosphorylation of specific tyrosine residues within the cytoplasmic domain of the receptors to which they bind. The designation "insulinlike growth factor" was chosen to express the insulin-like effects and the insulin-like structure of these polypeptides which act as mitogens on a number of cells. IGF-1 is a amino acid peptide, while IGF-II is a 67 amino acid peptide, as described in Rinderknecht, J Biol Chem, (1978) 253:2769; and Rinderknecht, FEBS Letters, (1978) 89:283. IGF-I and IGF-II have 62% structural homology to each other. Both have been isolated from human serum.
Insulin-like growth factors are also known under the class name somatomedins, and have been identified in various animal species as polypeptides that act to stimulate growth of cells in a variety of tissues and cell types, particularly during development. Growth promoting effects of somatomedins include enhancement of cell multiplication and stimulation of cartilage proliferation, stimulation of transport of amino acids, stimulation of synthesis of RNA, DNA and protein, and stimulation of incorporation of sulfate into proteoglycan and of proline into collagen. Much mammalian postnatal growth is due to stimulation of cartilage growth by somatomedins and growth in utero may also be somatomedin-dependent.
25-11-04; 1:29 612 93645173 6/ -2- Uses of IGF as a known stimulatory and growth promoting agent includes use for bone repair and replacement therapy; as a means to counteract certain harmful side effects of carcinostatic drugs; and as a way to increase lactation and meat production in cattle and other farm animals.
IGF-1 has also been found useful in the treatment of osteoporosis in mammals exhibiting decreased cortical bone mineral density and those exposed to drugs or environmental conditions that result in bone density reduction and potentially to an osteoporosis condition.
IGF-I has been administered with sodium pentosan polysulfate (PPS) to severely osteoarthritic canines with the effect of reducing the severity of the disease by lowering the levels of active neutral metalloproteinase in the cartilage. In the model of mildly osteoarthritic canines, therapeutic Intervention with IGF-i and PPS together appeared to successfully maintain cartilage structure and biochemistry, while IGF alone was ineffective, as described in Rogachefsky, Osteoarthritis and Cartilage, (1993) 1:105-114. It would be advantageous to discover further uses for IGFs.
Summary of the Invention S*o: The present invention seeks to discover and provide for additional uses of IGFs. The present invention also seeks to provide a method for inhibition of an inflammatory response such as in rheumatoid arthritis, psoriatic arthritis, Reiter's syndrome, and other known inflammatory arthritis and inflammatory bowel disease.
In addition, the present invention seeks to provide a method for inhibition of ischemic injury as, for example, in acute renal tubular necrosis (ATN), ischemic injury to the heat, brain or liver.
The present invention also seeks to provide a method for inhibition of organ rejection upon transplantation, including transplantation of kidney, heart, lung, liver and pancreas.
COMS ID No: SBMI-01013331 Received by IP Australia: Time 15:32 Date 2004-11-25 25-11-04: 1:29 612 93645173 7/ -3- Further more, the present invention seeks to provide a method for the treatment of rheumatoid arthritis, psoriatic arthritis, Reiter's syndrome, and other known inflammatory arthritis, as well as inflammatory bowel disease.
According to one aspect the present invention provides a method for inhibition of an inflammatory response by administering an inflammatory response inhibitory dose of IGF.
In accordance with another aspect there is provided a method for inhibiting organ rejection after transplantation by administering an organ refection inhibitory dose of an IGF.
As now claimed, according to one aspect, the present invention provides a method for inhibition of an inflammatory response in a mammal that has an inflammatory condition, wherein said inflammatory condition causes degeneration of cartilage, pancreatic cells, brain cells or gastrointestinal cells, said method comprising administering thereto an inflammatory response inhibitory dose of IGF sufficient to inhibit degeneration of said cartilage, pancreatic cells, brain cells or gastrointestinal cells.
Further, features and advantages of the present invention will become apparent from the following detailed description. It should be understood, however, that the detailed description, while indicating preferred embodiments of the invention, is given by way of illustration only, since various changes and modifications within the spirit and scope of the invention will become apparent to those skilled in the art from this detailed description.
Brief Description of the Drawnoas o Figure 1 shows the effect of recombinant human IGF-.(rhlGF) on MHC Class I and if expression by RIA on ATN induced group day 4, Figure 2 shows the effect of rhlGF MHC Class I and II expression by RIA on ATN induced group day e COMS ID No: SBMI-01013331 Received by IP Australia: Time 15:32 Date 2004-11-25 2 5 -II- 04: I1 93:7 2 6/ 1 612 93645173 8/ 1 C 3a Detailed Description of the Invention This invention relates to a method for inhibiting an inflammatory response, ischemic injury, and organ rejection upon transplantation with the use of IGF-l or IGF-ll. Previously. IGF-l and IGF-11 have been known primarily for their stimulatory and growth-promoting ef fects.
Unexpectedly.. the inventors herein have found that IGF is also capable of inhibitory responses such as inhibiting inflammatory response, ischemic injury, and organ rejection.
COMS ID No: 5BMI-01013331 Received by IP Australia: Time 15:32 Date 2004-11-25 The term "insulin-like growth factor" as used herein encompasses IGF-I and IGF-II in their substantially purified, native, recombinantly produced, or chemically synthesized forms, and includes biologically active fragments, analogues, muteins, including C-terminal deletion muteins, and derivatives thereof that retain IGF activity and/or ability to bind the IGF receptors, as described in, for example, EP 135 094, WO 85/00831, U.S. Patent No.
4,738,921, WO 92/04363, U.S. Patent No. 5,158,875, EP 123 228, and EP 128 733. An analog of IGF or an analog of the fragment includes native IGF that has been modified by one or more amino acid insertion, deletion, or substitution that does not substantially affect its properties. Preferably, the analog has increased activity compared to native IGF. More preferably, at least 2-fold increase, most preferably, at least 7-10 fold increase. For O example, the analog can include conservative amino acid substitutions. An IGF analog also includes peptides having one or more peptide mimics ("peptoids"), such as those described in WO 91/04282.
An IGF mutein is a polypeptide variant with one or more amino acids altered to produce a desired characteristic, such as to replace a cysteine residue with a non-disulfide bond forming amino acid. Muteins, analogues and derivatives may be generated using conventional techniques. For example, PCR mutagenesis can be used. While the following discussion refers to DNA, it is understood that the technique also finds application with RNA. An example of a PCR technique is described in WO 92/22653. Another method for making analogs, muteins, and derivatives, is cassette mutagenesis based on the technique described by Wells, Gene, (1985) 34:315.
An "inflammatory response" is generally characterized by increased blood flow and entry of leukocytes into the tissues, resulting in swelling, redness, elevated temperature and pain.
One indication of an inflammatory response is the expression of Class I or Class II of the major histocompatibility complex Class I MHC is expressed on the surface of essentially all nucleated cells, particularly lymphoid cells. Class II MHC is expressed on macrophages as part of the inflammatory response in mammals. When activated by agents such as g-interferon, capillary endothelia and many epithelial cells express surface Class II and increased expression of Class I. The inventors herein found that administration of IGF in vivo can be effective in inhibiting inflammation. Such inhibition can be monitored, for example, by reduction in the expression of Class I or Class II MHC, or by the amount of antibody binding to Class I or Class II MHC, as described in Geraughty, Curr. Opin.
Immunol., (1993) 5:3; Kara C. Glimcher, Curr. Opin. Immunol. (1991) 3:16; and Kurander, Science, (1992) 257:678.
"Ischemic injury" resulting from decreased blood flow to an organ, includes, for example, arterial narrowing or mechanical obstruction of the blood supply. Ischemic injury in the mammal involves production of cytokines such as IL-6, IL-8 and IFN-y. Other cytokines that may be induced by ischemic injury include the interleukins, such as IL-1, IL-2 and colony stimulating factors (CSF), and those involved in healing and repair such as tumor necrosis factor (TNF) and transforming growth factor-beta Cytokines have been D documented to exert diverse effects both in vitro and in vivo, including damage to surrounding tissues in addition to healing and repair.
9 a. An "inflammatory response inhibitory dose" as used herein is a therapeutically effective amount that is sufficient to reduce or prevent a further inflammatory response. Inhibition of inflammatory response can be monitored by any conventional method. One method, for example, is to monitor the reduction in expression of Class I MHC or Class II MHC by, for example, detecting antibody binding to the MHCs. Inflammatory response is considered to ****have been inhibited if there is at least a 5% reduction of such response, preferably, at least a 10% reduction, more preferably, at least a 15% reduction, and most preferably, at least a reduction or higher.
An "ischemic injury inhibiting dose" as used herein is a therapeutically effective amount that is sufficient to reduce or prevent a response resulting from ischemic injury. Such as response can also be monitored by conventional methods. One method, for example, is to monitor the reduction in the induction of cytokine production, such as those cytokines mentioned above. Ischemic injury is considered to have been inhibited herein if there is at least a 5% reduction of such cytokine production, preferably, at least a 10% reduction, more preferably, at least a 15% reduction, and most preferably, at least a 20% reduction or higher.
-6- An "organ rejection inhibitory dose" as used herein is a therapeutically effective amount that is sufficient to prevent organ rejection after transplantation or to prolong the acceptance of the transplanted organ.
An "arthritic condition" is considered to have been inhibited if there is no deterioration in the cell type concerned. Typically, an arthritis inhibitory dose will further effect regeneration of the cell type, resulting in at least a 2% increase in the cell type, preferably, at least a increase, more preferably, at least a 10% increase, and most preferably, at least a increase or higher.
O A "pharmaceutically acceptable carrier" is a carrier that is conventionally used by persons skilled in the art for administration of a therapeutic composition into a human. Such a S carrier typically minimizes undesirable side effects of, for example, the induction of antibodies production, the induction of fever, etc. Suitable carriers are typically large, slowly metabolized macromolecules that includes molecules such as a protein, a polysaccharide, a polylactic acid, a polyglycolic acid, a polymeric amino acid, amino acid copolymers or an inactive virus particle. Such carriers are well known to those of ordinary skill in the art.
A "therapeutic composition" herein contains as an active ingredient either IGF-I or IGF-II or both and one or more pharmaceutically acceptable carriers, and/or one or more components such as water, saline, glycerol, or ethanol can be included in such a composition. Additionally, auxiliary substances, such as wetting or emulsifying agents, pH S buffering substances and the like, may optionally be present in such compositions. For S: example, carriers that have been used for oral or parenteral administration include Ringers solution, Hank's solution, and solutions of glucose, sucrose, lactose, mannose, dextrose, dextran, mannitol, sorbitol, albumin, polyethylene glycol (PEG), phosphate, acetate, gelatin, collagen, Carbopol@, vegetable oils, and the like. These compositions may optionally include stabilizers, antioxidants, antimicrobials, preservatives, buffering agents, surfactants, and other accessory additives. Cream or ointment bases useful in formulation include lanolin, Silvadene® (Marion), Aquaphor® (Duke Laboratories), and the like. Other topical formulations include aerosols, bandages, sustained-release patches, and the like.
A "therapeutically effective amount" is that amount that is effective for production of a desired result. This amount varies depending upon the health and physical condition of the individual to be treated, the capacity of the individual's immune system to synthesize antibodies, the degree of protection desired, the formulation, the attending physician's assessment of the medical situation, the age, size, and condition of the subject, the nature and severity of the disorder to be treated, and other relevant factors. It is expected that the amount will fall in a relatively broad range that can be determined through routine trials by a qualified physician or veterinarian. Thus, for example, a therapeutically effective amount for an inflammatory response inhibitory dose can be in the range of about 50-500 mg/kg, preferably, about 100-500 mg/kg, more preferably, about 150-450 mg/kg, most preferably, about 200 mg/kg. In general terms, an effective dose of IGF-I and IGF-II will range from about 5-10 mg/kg to about 50 mg/kg to a range of 100 mg/kg to 500 mg/kg/day.
Alternatively, an effective dose of IGF-I and IGF-II can range from 10-50 mg/kg, 10-500 Smg/kg, 50-100 mg/kg, 100-150 mg/kg, 150-200 mg/kg, 200-400 mg/kg, 250-350 mg/kg, and 400-500 mg/kg. Such a regimen can be administered as a single dose or as multiple S. :doses. When IGF is given as multiple doses, for purposes herein, such multiple doses can be given in a day, preferably they are given within a week, more preferably, they are given within 2 weeks, and most preferably, within a month. Optionally, IGF herein can be given until the desired response is achieved.
Alternatively, one may incorporate or encapsulate IGF in a suitable polymer matrix or membrane, thus providing a sustained-release delivery device suitable for implantation on or near the site to be treated locally. Other devices include indwelling catheters and devices such as the Alzet@ minipump. Further, one may provide the compound in solid form, especially as a lyophilized powder. Lyophilized formulations typically contain stabilizing and bulking agents, for example human serum albumin, sucrose, mannitol, and the like. A thorough discussion of pharmaceutically acceptable excipients is available in Remington's Pharmaceutical Sciences 18th edition, 1990 (Mack Pub. Co., Easton, PA).
The compounds of the invention may be administered by a variety of methods as conventionally known in the art suitable to attain the desired result, including for example, subcutaneously, topically,- intravenously, orally, intramuscularly, intraperitoneally, intraluminally, and intra-articularly. The preferred route of administration will depend upon the nature of the formulation and the condition to be treated. The duration of treatment may include, for example, a single dose of 25-100 mg/kg/shot, which may be administered once, or two or three times daily. The number of days of administration may be one to two days.
For conditions such as rheumatoid arthritis, administration three times weekly may be appropriate, when administered intra-articularly at higher dosages of, for example, 100-500 mg/kg/dose, administered for three months. Chronic inflammatory bowel and other inflammatory diseases or conditions may require, for example, daily administration for one to six weeks, administered locally including intra-luminally to the GI tract. For systemic inflammatory response conditions, subcutaneous administration may be required at lower doses, including, for example 25-100 mg/kg/dose, for a duration depending on the severity )O of the condition and the responsiveness of the individual to treatment. For acute renal failure, and for acute brain or liver failure, for example, the duration of the treatment may require 4-14 days of daily administration, or 4-21 days of daily administration, or 4-28 days of daily administration. For administration to the brain, intra-ventricular administration may be required.
IGF for use herein can be made by a variety of known techniques. Thus, IGF can be S. isolated and purified from serum or plasma or produced by recombinant DNA techniques in microbial hosts such as bacteria or yeast or in cell cultures such as insect cell or animal cell cultures, or chemically synthesized in accordance with conventional techniques. For example, IGF can be isolated in small quantities from large volumes of plasma or serum as described in Phillips, New Engl. J. Med., (1980) 302.371-380.
IGF can also be synthesized by the solid phase method as described in Li, PROC. NATL.
ACAD. SCI. USA, (1983) 80.2216-2220. In this method, the polypeptide sequence for IGF-I can be assembled by coupling the amino acid residues.
IGF can be made by conventional recombinant DNA techniques, as described in Biochem.
and Biophys. Res. Comm., (1990) 169:832-839 (IGF II) and Cell Regulation, (1990) 1:197- 213, (IGF II), and Biotechnology News, (1983) 3:1-3 (IGF-I and II). For example, IGF can be produced in E.colias a fusion protein with the trpE gene under the control of a modified tryptophan operon, as described in U.S. Patent No. 4,738,921. Alternatively, IGF can be synthesized in E. coli under the control of the Vesicular Stomatitis Virus (VSV) promoter and protector sequences, as described in EP 478 333. The E. coliexpression systems used for expression herein can be modified as described in U.S. Patent No. 5,158,875, to include a modified positively charged leader sequence to enable proper folding of the IGF protein.
Moreover, IGF can be produced in methylotrophic yeast transformants with the IGF coding sequence linked to a signal sequence which direct secretion and proteolytic processing of the protein product. The signal sequence suitable herein includes the S. cerevisiae alpha mating factor pre-pro sequence in protease deficient P. pastoris strains, as described in WO 92/04363.
DNA constructs for production of IGF-Il can be made and expressed in E. colias described in WO 89/03423. Synthesis of recombinant IGF-II can also be achieved by following the S' protocol described in EP 434 605, which relates to the production of recombinant IGF-II with a covalently attached foreign moiety and lacking the N-terminal attached methionine.
IGF can also be made in yeast as described in EP 123 228 and U.S. patent application 9.
S.N. 06/922,199. Another method of producing IGF using recombinant DNA techniques that is suitable herein is as described in Biotechnology News, (1983) 10:1-3. IGF-I or IGF-II coding sequences can be inserted into viral or circular plasmid DNA vectors to form hybrid vectors, and the resulting hybrid vectors can be used to transform host microorganisms such as bacteria or yeast cells. The transformed microorganisms can be grown under appropriate nutrient conditions to express IGF, as described in EP 135 094.
In another context, human IGF-I and IGF-II can be expressed and secreted using a leader sequence that contains a portion of the yeast a-factor signal sequence, as described in EP 128 733.
The IGF-I herein can be expressed in any unicellular organism, including, for example, yeast cells, bacterial, insect, and mammalian cells. For example, yeast cells in which IGF-I can be expressed include Saccharomyces cerevisiae (Hinnen, PROC. NATL. ACAD. SCI.
USA, (1978) 75:1929; Ito, J. Bacteriol., (1983) 153:163; Saccharomyces carlsbergeneis; Candida albicans, Kurtz, Mol. Cell. Biol., (1986) 6:.142; Candida maltosa, Kunze, J. Basic Microbiol., (1985) 25:141; Hansenula polymorpha (Gleeson, J. Gen. Microbiol., (1986) 32.3459; Roggenkamp, Mol. Gen. Genet. (1986) 202*302; Kluyveromyces fragilis, Das, J. Bacteriol., (1984) 158:1165; Kluyveromyces lactis, De Louvencourt, J. Bacteriol., (1983) 154:737; Van den Berg, Bio/Technology, (1990) 8:135; Pichia guillerimondii, Kunze, J.
Basic Microbiol., (1985) 25:141; Pichia pastoris, Cregg, Mol. Cell. Biol., (1985) 5:3376; U.S.
Patent Nos. 4,837,148 and 4,929,555; Schizosaccharomyces pombe, Beach, Nature, (1981) 300-706; and Yarrowia lipolytica, Davidow, Curr. Genet., (1985) 10:380471; Gaillardin, Curr. Genet., (1985) 10:49; Neurospora crassa, Case, PROC. NATL. ACAD.
SCI. USA (USA), (1979) 7&.5259; and filamentous fungi such as, Neurospora, Penici/lium, Tolypocladium, WO 91/00357, and Aspergillus hosts such as A. nidulans, Ballance, Biochem. Biophys. Res. Comm., (1983) 112284; Tilbum, Gene, (1983) 26205, Yelton, 0 PROC. NATL. ACAD. SCI. USA (USA), (1984) 1981:1470, and A. Niger, Kelly, EMBOJ., (1985) 4:475-479.
IGF-I herein can also be expressed in bacterial cells including, for example, Streptococcus spp., and Streptomyces spp., eubacteria, such as Gram-negative or Gram-positive organisms, for example, E. coli, Bacilli such as B. subtilis, Pseudomonas species such as P.
aeruginosa, Salmonella typhimurium, or Serratia marcescans.
IGF-I herein can further be expressed in insect cells with a baculovirus expression system, including, for example, numerous baculoviral strains and variants and corresponding permissive insect host cells from hosts such as Spodoptera frugiperda (caterpillar), Aedes aegypti (mosquito), Aedes albopictus (mosquito), Drosophila melanogaster (fruitfly), and Bombyx mor host cells, as described in Luckow, Bio/Technology, (1988) &47; Miller, Genetic Engineering, Plenum Publishing (1986) 8:277; and Maeda, Nature, (1985) 315:592.
The vector may be a baculovirus transcriptional promoter region derived from any of the over 500 baculoviruses generally infecting insects, such as for example the Orders Lepidoptera, Diptera, Orthoptera, Coleoptera and Hymenoptera, including for example the viral DNAs of Autographo califomica MNPV, Bombyx mori NPV, rrichoplusia ni MNPV, Rachlplusia ou MNPV or Galleria mellonella MNPV, Trichoplusia ni, Rachiplusia ou, and Galleria mellonella, and others as described in Smith, Virol., (1978) 89:517.
IGF-I can also be expressed in mammalian host cells including, for example, expression in monkey cells, as described in Reyes., Nature, (1982) 297:598, cultured mouse and rabbit -11 cells, mouse NIH-3T3 cells many immortalized cell lines available from the American Type Culture Collection (ATCC), including, for example, Chinese hamster ovary (CHO) cells, as described in Urlaub, PROC. NATL ACAD. SCI. USA (USA), (1980) 77:4216, HeLa cells, baby hamster kidney (BHK) cells, monkey kidney cells (COS), human hepatocellular carcinoma cells Hep G2), human embryonic cell line as described in Grisham, J. Gen.
Virol., (1977) 36:59, mouse sertoli cells as described in Mather, Biol. Reprod., (1980) 23:243, African green monkey cells, canine kidney cells, buffalo rat liver cells, human lung cells, human liver cells, and mouse mammary tumor cells, among others.
The following examples are given by way of illustration to facilitate a better understanding of f the invention and are not intended to limit the invention in any way.
Examples Example 1: Making IGF-I The rhlGF-I protein is synthesized by Saccharomyces cerevisiae strain JSC417, transformed with plasmid pYLUIGF1-24. The transformed yeast strain, was deposited with the American Type Culture Collection (ATCC) located at 12301 Parklawn Drive, Rockville, Maryland 20852, on August 2, 1994 with ATCC Accession No. 74295. Strain JSC417 was derived from strain AB110. JSC417 has the following genotype: Mata, ura 3-52, leu 2, pep 4-3, his 4-580, [cir Expression of rhlGF-I in S. cerevisiae strain JSC417 is non-constitutive and under the regulation of a hybrid ADH2-GAP promoter derived from promoter sequences of the yeast alcohol dehydrogenase as described in Beier, Nature, (1982) 300:724, and glyceraldehyde- 3-phosphate dehydrogenase as described in EP 120 551. In addition, the rhlGF-l sequences are fused to the yeast a-factor leader which allows for secretion, and to the yeast a-factor terminator both as described in Brake, PROC. NATL. ACAD. SCI. USA, (1984) 81:4642. Induction of rhlGF-I expression is achieved by maintaining low concentration of glucose in the growth medium during fermentation..
Plasmid pYLUIGF1-24 is a yeast expression vector which contains the sequence coding for rhlGF-I cloned into the BamHI site of vector pAB24, as described in Barr, Bio/Technology, (1987) 5:486, as well as pBR322 sequences including the ampicillin resistant (ampR) gene, 12- 2-micron (2 m) sequences, and the yeast LEU 2 and URA 3 genes. The expression cassette for rhlGF-I consists of to ADH2 regulatory sequences, a GAP promoter, afactor leader, rhlGF-I synthetic gene and a-factor terminator, as described in EP 123 228.
The rhlGF-I gene cloned into the expression cassette is chemically synthesized using the phosphoramidite procedure as described by Urdea, PROC. NATL. ACAD. SCI. USA, (1983) 80:7461, and according to the Dayhoff amino acid sequences.
S. cerevisiae cells are transformed with plasmid pYLUIGF1-24 following a standard protocol as described in Hinnen, PROC. NATL. ACAD. SCI. USA (USA), (1978) 75:1929. Briefly, O the transformation mixture is plated onto uracil-deficient selective plates that are yeast nitrogen base with amino acids containing 2% glucose and incubated for four days at 30 C.
Transformant colonies are transferred into leucine-deficient, selective media with 8% glucose, and grown at 30 Expression of rhlGF-I is accomplished by growing the yeast transformants in uracil deficient medium with 0.4% glucose at 30 °C for 48 hours.
Expression of rhlGF-I in the medium at 48 hours is analyzed by any of several standard methods including RP-HPLC, SDS-PAGE, RIA, or radioreceptor assay.
Production of rhlGF-I involves successive amplification of the yeast cells contained within the seedstock aliquot The first amplification stage is carried out in shake flasks at a controlled temperature of 30 °C in a rotary shaker-incubator. Approximately 10 7 cells are thawed into 500 ml of uracil- and leucine-deficient media, as described above, containing 8% glucose. After 25-45 hours, preferably 35 2 hours, the flask contents are transferred to a small fermentation vessel for the second stage of cell amplification. This culture is grown for 24 4 hours under controlled temperature (30 aeration (1 wm) and agitation (400-600 rpm) in 10 lof the same media used for stage I. 10-30 I of the stage II culture is transferred to a larger, production-scale fermentation vessel for the final amplication and rhlGF-I expression phase of growth. Stage III utilizes a semi-defined growth media containing casein hydrolyzate, basal salts, vitamins, trace elements, and antifoam. The casein hydrolyzate employed may be any commercial brand with a composition of at least amino-nitrogen, at least 10% total nitrogen, not more than 20% ash, but preferably would have a composition comparable to that of N-Z-Amine HD (Quest). The antifoam employed may be any of several commercially available polyalcohol- or silicon-based -13compounds. The fermentation is carried out at constant 30 pH 6 (by addition of sodium hydroxide or 75% phosphoric acid), aeration (0.8 wm), pressure (5-12 psig), and glucose feed rate with constant agitation. The fermentation is known to those skilled in the art as a fed-batch mode of operation, so-called because the fermentor is initially filled to less than capacity, allowing for the addition of a suitable amount of a glucose feed solution with a concentration of 25-50% w/v. Typically, 800-900 kg glucose is added to the fermentor over the duration of the run at a rate of addition that depends on yeast cell density and residual glucose concentration. Typically, glucose is added at 500 g/min for the first 26 h, at 1000 g/min for the next 24 h, and finally at 500 g/min until completion. Cell growth concomitant with product expression occurs once the media becomes depleted of IO excess glucose, and may continue until the culture reaches the desired cell density of approximately 35 g DCW/I.
rhlGF-I is recovered from the cell cultures using conventional techniques. At the completion of fermentation, the pH of the fermentor culture is raised to a range of pH 9-10, pH 10.1- 10.3, pH 10.4-10.6, pH 10.6-10.8, pH 10.9-10.11, or pH 11-12 for at least 30 minutes. Prior to cell removal 50% sodium hydroxide is added to the fermentor to raise the pH of the i whole culture to with a range of pH 9-10, pH 10.1-10.3, pH 10.4-10.6, pH 10.6-10.8, pH 10.9-10.11, or pH 11-12 for 0.5 to 4 h (preferably pH 10.2 for at least 0.5 Afterwards the whole yeast are separated from the product-containing spent media by continuous centrifugation. After collection the supematant is readjusted to with a range of pH 3-4, pH 5-6 or pH 6-7 (preferably pH 4) with 75% phosphoric acid, and filtered using microporous tangential flow filtration prior to adsorbing on a cation exchange resin. The column is washed with 20 mM acetic acid and 100 mM potassium borate/0.1 mM EDTA buffers, and eluted with a 100 mM potassium borate/0.1 mM EDTA/300 mM potassium chloride buffer at pH 8.7.
Standard hydrophobic interaction chromatography (HIC) is used for primary purification of rhlGF-l. Ammonium sulfate precipitation and microporous tangential flow filtration is used to remove yeast contaminants. After washing the precipitate with ammonium sulfate, sodium acetate and EDTA buffer at pH 4, the material is loaded onto the HIC matrix and eluted with linear decreasing gradient of 0.9 to 0.5 M ammonium sulfate. Eluted protein at pH 4 is concentrated 20-fold and the buffer is exchanged using a 5000 MWCO tangential flow -14ultrafiltration membrane. The diafiltration agents employed are high purity water followed by mM acetic acid.
Standard reverse-phase high performance liquid chromatography (RP-HPLC) is used to remove product-related species from the authentic rhlGF-I. The 5 K concentrate is loaded onto a suitable C8 matrix and eluted with a linear increasing gradient of 10% to acetonitrile in a pH 6.8 ammonium acetate buffer.
Example 2: Use of IGF-I to Reduce Inflammatory Response Associated with Ischemic Injury It is known that ischemic acute tubular necrosis (ATN) induces inflammation, MHC and S cytokine expression. Here we show that IGF-I reduced cytokine and MHC induction after induction of ATN. The human IGF-I (hulGF-l) is administered to mice after ATN induction.
The degree of inflammation is assessed by the degree of major histocompatibility complex S(MHC) expression and cytokine expression. ATN is produced in mice by clamping the left renal pedicle for 60 minutes. MHC expression is quantified by the binding of radiolabelled monoclonal antibodies (mAbs) (RIA). Steady state mRNA levels for IL-2, IL-10, GM-CSF and IFN-y are assessed by RT-PCR and for TGF-3p-1, TNF-a and ppEGF by Northems. In the absence of IGF-I, MHC Class I and Class II mRNA and product are increased, for two weeks for Class I and for 5 weeks for Class II. In the ischemic kidneys, IL-2, IL-10, GM- CSF, IFN-y, TNF-a, TGF-p-1 mRNA are induced, whereas ppEGF mRNA is greatly decreased. Administration of rhlGF-1, 25 pg s.c. daily starting 1 hour after reperfusion, reduced MHC expression and cytokine mRNA induction after ischemic injury, assessed at S. day 5. Results are shown in Tables I-VI.
Male BALB\c and CBA mice obtained from Jackson laboratories (Bar Harbor, ME) are operated on at 6-15 weeks of age. They are anesthetized with 2,2,2-tribromoethanol in tertbutyl alcohol (Avertin) by intraperitoneal injection. The left renal pedicle is identified through a midline incision and occluded with a micro-bulldog clamp for 60 minutes. Before closure the kidney is inspected to ensure reperfusion, and the abdominal cavity is filled with warm saline. Control mice underwent a simple laparotomy under identical conditions. The mice are harvested at 4 and 5 days postoperatively. Each group contains between 4 and 8 animals at harvesting time. No thrombosis is seen, even though neither heparinization nor renal flushing are used. ATN is confirmed in these experiments with tissue staining, using HE and PAS, of two animals randomly selected in each group. In the first two experiments rhlGF I is administered according to the following protocol, being the number of mice in each group: Exp. 1 Exp. 2 (n) 1. CBA/J 0.9% NaCI 1. CBA/J 0.9% NaCI (6) 2. CBA/J rhlGF I 2. ATN day 5 0.9% NaCI (8) 3. ATN day 4 0.9% NaCI 3. ATN day 5 rhIGF I (8) S 4. ATN day 4 rhlGF (4) Human IGF-I diluted in 0.9% NaCI is administered 25 pg at 2, 24, 48, 72 and 96 hours after surgery. The dose is selected based on the dose used in the rat studies in which rhlGF-I is found to accelerate renal recovery, as described in Miller, PROC. NATL. ACAD.
SCI. USA (USA), (1992) 89:11876, and Ding, J Clin Invest, (1993) 91:2281.
Monoclonal antibodies are prepared as ascites tumors or as supematants from hybridomas supplied by American Type Culture Collection (Rockville, MD). They are purified over a protein A column and adjusted to a protein concentration of 1 mg/ml. The anti-class I, anti- H-2K(k), are Ig class IgG(2a) and the anti-class II, anti-l-A(k) are IgG(2b). The rat antimouse H-2 haplotypes (M1) and anti-I-A (b,d,q haplotypes) and I-E(dk) (M5) are IgG(2a) and IgG(2b), respectively. The goat anti-rat and goat anti-mouse peroxidases are polyclonal F(ab')2 fragments of IgG heavy and light chains. Radioiodination with 1251 is performed by lodogen method as described in Salacinski, Anal Biochem, (1981) 117:136.
The RIA technique employed was previously reported in Cockfield, J Immunol, (1989) 142:1120. Accordingly, tissue homogenates are prepared, aliquoted in 5 mg samples, and spun. The tissue pellets are incubated with s 25 1-labelled anti-MHC mAb for 1 hour. After washing, the pellets are counted in a gamma counter to obtain the number of counts remaining bound to the tissue pellet. All samples are tested in triplicate. Raw counts from experimental tissues are corrected for nonspecific binding by subtracting the counts bound to 5 mg of tissue from an irrelevant strain. Specific binding, demonstrated both directly and -16by competitive inhibition, is observed only in mice expressing the particular MHC haplotype and cannot be explained by FcR binding. The differences between left (clamped) and right (control) kidneys are compared by one-way analysis of variance (Anova). All results are expressed as mean SEM.
Total RNA is extracted from pooled samples according to a modification of the method described by Chirgwin, Biochemistry, (1979) 18:5294. Tissues are homogenized with a polytron in 4 M guanidinium isothyocyanate, and the RNA is pelleted through a 5.7 M CsCI 2 cushion as described in Glisin, Biochemistry, (1974) 13:2633. RNA concentrations are determined by absorbance at 260 nm. Northem blots are prepared using 25 to 35 pg of Q total RNA electrophoresed through a 1.5% agarose gel in the presence of 2.2 M formaldehyde, with transfer to nitrocellulose filters. Blots are hybridized with "P-labelled cDNA probes for the HLA-A3 gene (Class a-chain of I-A (Class II), ppEGF, TNF-a and S: TGF-p-1. Kodak X-Omat AR film is exposed to each blot, with an intensifying screen at
S:
0
C.
Total RNA prepared by the guanidinium-cesium chloride method is reverse transcribed in a 10 l reaction containing 5 ng/l IFN-y specific downstream primer, 5 U/l moloney murine leukemia virus reverse transcriptase (Superscript; BRL), standard reaction buffer, and nucleotides. The cDNA product is then amplified in a 50 ld reaction with standard PCR reaction buffer, BSA, the appropriate primers and 0.01 U/l Taq polymerase (BRL), by using a thermocycler. A MgCIz titration curve establishes that 2.5 mM MgCI 2 is the optimal concentration for this primer pair. The cycler program is one of the many standard ones in the art of PCR, with an initial denaturation, annealing and extension times and temperatures. The RT-PCR product is run on a 1.5% agarose gel, transferred to a nylon membrane, and hybridized with a "P-labelled IFN-y cDNA probe. The primers used are based on known sequence information as described in Gray, PROC. NATL. ACAD. SCI.
USA (USA) (1983) 80:5842, and are located on either side of an intron to ensure that the amplified product is derived from RNA and not contaminating genomic DNA. The strategy for RNA reverse-transcription, and DNA amplification for IL-2, IL-10 and GM-CSF is the same as used for IFN-y.
-17- The cDNA probe used for murine IFN-y is a cDNA fragment of Aad inserted at the Psfi site of pBR322, for detecting transcripts of the a chain of I-A; a 1.5kB cDNA fragment of the HLA-A3 gene inserted into pEMBL8 is used to detect Class I transcripts; a .96kB cDNA fragment of murine ppEGF inserted into the Psfl site of pBR322 is used to probe for ppEGF; a 2.13 kB fragment of the human TGF-p-1 mRNA; the GM-CSF probe is a cDNA fragment of the gene; the IL-10 probe is an oligomer of +213 to +253 of the gene. All probes are 3Plabelled by the random priming method of Feinberg and Volgelstein, Anal Biochem, (1983) 132.6.
It is established that unilateral ischemia in the mouse is well tolerated and is followed by 7 complete parenchymal recovery as described in Shoskes, Transplantation, (1990) 49:201, and Shoskes, Transplant Proc, (1991) 23:599. Here, histological evidence of acute tubular necrosis (ATN) is seen in the ischemic kidney by 12 hours and persisted in severe form for S. 3-5 days. Interstitial hypercellularity is readily apparent in the left kidneys around day 7, with frequent progression to an interstitial mononuclear cell infiltrate by days 14 to 35. Right (control) kidneys are histologically normal. Serum creatinine and blood urea levels at the time of harvesting in selected animals from control and experimental groups show no detectable elevation.
Treatment with rhlGF-I reduced MHC induction after induction of ATN. Northem blot analysis performed with ATN or normal kidneys, using total RNA extracted from a pool of three kidneys, show increased Class I and II mRNA expression in ATN kidneys. A reduction of MHC gene induction after rhIGF-l treatment is found (Table MHC Class I and II gene induction is also studied at the protein level using radiolabelled monoclonal antibodies (RIA) against Class I and II (Table II and III and Graphs 1 and 2 shown in Figures 1 and 2, respectively). Administration of rhIGF-I reduces, by approximately 60-70%, Class I and II product expression. Class I is detected mainly in the basolateral aspect of tubular cells at the outer stripe of the outer medulla with a focal and segmental distribution and is reduced by treatment with rhlGF-I (Tables IV and Class II expression, as previously described, is not detected by staining at days 4 and rhlGF-l also reduces cytokine gene induction after induction of ATN. IL-2, IL-10, GM-CSF, IFN-y, TNF-a and TGF-p-1 and a marked reduction of ppEGF genes are found in ATN 18kidneys. Administration of rhlGF-l reduces the induction of all cytokine genes and reduces the extent of ppEGF mRNA decrease induced by ATN (Table VI). The ability of rhlGF to reduce the cytokine and inflammatory response may reflect reduction in the extent of injury; increased spead of recovery from injury; a direct inhibition of the cytokine and inflammatory response; and a combinantion of these mechanisms.
Table I MHC Gene Induction After Ischemic Injury Effect of rhlGF-I Class I Class II Normal ATN day 5 SATN day 5 rhlGF-I Results in Table I are expressed as the difference between left (ischemic) and right kidney (from to using Northern blot analysis.
Table II Ischemia-Induced MHC Expression Effect of rhlGF-I Class I Class II Normal -222.3 412.6 -61.2 258.9 SATN day 5 2156 618.2 1577.3 407.8 S. ATN day 5 rhlGF-I 1258.4 429.2 498.7 ±536.9 Results in Table II are expressed as the difference between left (ischemic) and right kidney using radiolabelled mAbs against MHC Class I and II products (RIA) (Mean cpm SEM).
19- Table Ill lschemia-lInduced MHC Expression Effect of rhlGF-l Class I Class 11 Normal ATN day 4 ATN day 4 rhlGF-l 41.5 ±164.7 1447.8 177 883.6 ±460 35.7 757.2 204.8 97.7±132.3 Results in Table IlI are expressed as the difference between left (ischemic) and right kidney using radiolabelled mAbs against NHC Class I and 11 products (RIA) (Mean cpm SEM".
0
S.
S.
S
S.
S.
Table IV lschemia-lnduced Renal MHC Expression Effect of rhlGF-l Class I Class 11 Tubular Interstitial Tubular Interstitial ATN day 4R i-- ATN day 4 +rhIGF-l 4+± Results in Table VI are expressed as intensity of immunoperoxidase straining (from to a i using anti-Class I and 11 mAbs.
I Table V Ischemia- Induced Renal MHC Expression Effect of rhIGF-l Class I Class 11 Tubular Interstitial Tubular Interstitial ATN day ATN day 5 rhIGF-l Results in Table V are expressed as intensity of immunoperoxidase straining (from to using anti-Class I and 11 mAbs.
Table VI Cytokine Gene Induction After Ischemic Injury Effect of rhlGF-I GM-CSF IFN-y TNF -a TGF-p-1 ppEGF Normal ATN day 5 ATN day rhGF-I Results in Table VI are expressed as the difference between left (ischemic) and right (normal) kidney (from to using RT-PCR for IL-10, GM-CSF, IFNy and Northern blot analysis for TNF-a, TGF-p-1, and ppEGF.
o 9 The patents, patent applications and publications cited herein are incorporated by reference.
Deposition of Microorganisms The yeast strain JSC417 transformed with plasmid pYLUIGF1-24 is deposited with the American Type Culture Collection (ATCC) located at 12301 Parklawr Drive, Rockville, Maryland 20852, on August 2, 1994 with ATCC Accession No. 74295 o Throughout this specification and the claims which follow, unless the context requires otherwise, the word "comprise", or variations such as "comprises" or "comprising", will be understood to imply the inclusion of a stated integer or step or group of integers or steps but not the exclusion of any other integer or step or group of integers or steps.
The reference to any prior art in this specification is not, and should not be taken as, an acknowledgment or any form of suggestion that that prior art forms part of the common general knowledge in Australia.

Claims (1)

  1. 25-11-04; 1:29 612 93645173 9/ -21- THE CLAIMS DEFINING THE INVENTION ARE AS FOLLOWS: 1. A method for inhibition of an inflammatory response in a mammal that has an inflammatory condition, wherein said inflammatory condition causes degeneration of cartilage, pancreatic cells, brain cells or gastrointestinal cells, said method comprising administering thereto an inflammatory response inhibitory dose of IGF sufficient to inhibit degeneration of said cartilage, pancreatic cells, brain cells or gastrointestinal cells. i 2. The method of claim 1, wherein the IGF is IGF-I. 3. The method of claim 1, wherein the dose of IGF is sufficient to inhibit the expression of class I or class II antigens of MHC. 4. The method of claim 1, wherein the dose of IGF is further sufficient for stimulation of regeneration of cartilage, pancreatic cells, brain cells, and S: gastrointestinal cells. 5. The method of claim 4, wherein the IGF is administered in one dose. 6. The method of claim 1, wherein IGF is administered in at least two doses. The method of claim 5, wherein the dose of IGF is in a range selected from the group consisting of about 10-50 pg/kg, 10-500 pg/kg, about 25-50 /jg/kg, about 25-100 pg/kg, 50-100 pg/kg, 60-75 pg/kg, 100-150 pg/kg, 150- 200 pg/kg, 175-200 pg/kg, 250-350 pg/kg, 200-300 pg/kg, 300-400 pg/kg, *and 400-500 pg/kg. 8. The method of claim 1, wherein IGF is administered either intravenously, subcutaneously, intramuscularly, intra-luminally, intra-articularly, and intrave ntricularty. COMS ID No: SBMI-01013331 Received by IP Australia: Time 15:32 Date 2004-11-25 25-11-04: 1:29 25- 1-4: 612 93645173 10/ 22 9. A method for inhibition of an inflammatory response in a mammalI substantially as hereinbefore described with reference to the examples. DATED THIS 25th day of November, 2004. NOVARTIS AG By Its Patent Attorneys DAVIES COLLISON CAVE COMS IDNo: SBMI-01O1 3331 Received by IP Australia: Time 15:32 Date 2004-11-25
AU29233/02A 1994-09-08 2002-03-28 Use of insulin-like growth factors I and II for inhibition of inflammatory response Ceased AU779507B2 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
AU29233/02A AU779507B2 (en) 1994-09-08 2002-03-28 Use of insulin-like growth factors I and II for inhibition of inflammatory response
AU2005201558A AU2005201558A1 (en) 1994-09-08 2005-04-13 Use of insulin-like growth factors I and II for inhibition of inflammatory response

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US08/302494 1994-09-08
AU42376/99A AU4237699A (en) 1994-09-08 1999-07-30 Use of insulin-like growth factors I and II for inhibition of inflammatory response
AU29233/02A AU779507B2 (en) 1994-09-08 2002-03-28 Use of insulin-like growth factors I and II for inhibition of inflammatory response

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
AU42376/99A Division AU4237699A (en) 1994-09-08 1999-07-30 Use of insulin-like growth factors I and II for inhibition of inflammatory response

Related Child Applications (1)

Application Number Title Priority Date Filing Date
AU2005201558A Division AU2005201558A1 (en) 1994-09-08 2005-04-13 Use of insulin-like growth factors I and II for inhibition of inflammatory response

Publications (2)

Publication Number Publication Date
AU2923302A AU2923302A (en) 2002-05-16
AU779507B2 true AU779507B2 (en) 2005-01-27

Family

ID=34427342

Family Applications (1)

Application Number Title Priority Date Filing Date
AU29233/02A Ceased AU779507B2 (en) 1994-09-08 2002-03-28 Use of insulin-like growth factors I and II for inhibition of inflammatory response

Country Status (1)

Country Link
AU (1) AU779507B2 (en)

Also Published As

Publication number Publication date
AU2923302A (en) 2002-05-16

Similar Documents

Publication Publication Date Title
AU710303B2 (en) Use of insulin-like growth factors (I) and (II) for inhibition of inflammatory response
JP2662390B2 (en) Insiulin analog
TWI642682B (en) Glucagon analogues
RU2529952C2 (en) Novel insulin derivatives with strongly slowed profile time/action
JP4219404B2 (en) Novel agouti-related genes
US7696158B2 (en) Use of IL-22 for the treatment of conditions of metabolic disorders
JP5907970B2 (en) Application of interleukin-22 in the treatment of viral hepatitis
KR20130018965A (en) Peptide capable of extending half-life of interest in plasma
WO1995027495A2 (en) Treatment of partial growth hormone insensitivity syndrome
AU740735B2 (en) Method for stabilizing useful proteins and useful protein compositions
KR100520723B1 (en) Osteoclast formation inhibitors
US5824642A (en) Treatment of partial growth hormone insensitivity syndrome
CN114901680A (en) Long-acting GLP-1 compounds
AU779507B2 (en) Use of insulin-like growth factors I and II for inhibition of inflammatory response
DE69723982T2 (en) TREATMENT OF PARTIAL GROWTH HORMONE SENSITIVITY SYNDROME
AU615194B2 (en) Bovine growth hormone analogs
AU4237699A (en) Use of insulin-like growth factors I and II for inhibition of inflammatory response
AU2005201558A1 (en) Use of insulin-like growth factors I and II for inhibition of inflammatory response
EP0366532B1 (en) Medical preparations containing glycosylated interleukin-2
WO2022261299A1 (en) Engineered nrg-1 variants with improved selectivity toward erbb4 but not against erbb3
JP2000506517A (en) Obesity protein compounds and formulations thereof
JP2001192396A (en) METHOD FOR PRODUCING HIGH-PURITY PROTEIN HAVING INTERFERON-gamma ACTIVITY, HIGH-PURITY PROTEIN HAVING DOG INTERFERON-gamma ACTIVITY, THERAPEUTIC AGENT FOR DOG DISEASE AND METHOD FOR PRODUCING DOG DISEASE