CA2444840A1 - Akt and regulation of ra synovial fibroblast apoptosis - Google Patents

Akt and regulation of ra synovial fibroblast apoptosis Download PDF

Info

Publication number
CA2444840A1
CA2444840A1 CA002444840A CA2444840A CA2444840A1 CA 2444840 A1 CA2444840 A1 CA 2444840A1 CA 002444840 A CA002444840 A CA 002444840A CA 2444840 A CA2444840 A CA 2444840A CA 2444840 A1 CA2444840 A1 CA 2444840A1
Authority
CA
Canada
Prior art keywords
akt
inhibitor
rheumatoid arthritis
apoptosis
tnf
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
CA002444840A
Other languages
French (fr)
Inventor
John D. Mountz
Huang-Ge Zhang
Jin-Fu Xie
Xu Liang
Pingar Yang
Hui-Chen Hsu
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.)
UAB Research Foundation
Original Assignee
Individual
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Individual filed Critical Individual
Publication of CA2444840A1 publication Critical patent/CA2444840A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/63Compounds containing para-N-benzenesulfonyl-N-groups, e.g. sulfanilamide, p-nitrobenzenesulfonyl hydrazide
    • A61K31/635Compounds containing para-N-benzenesulfonyl-N-groups, e.g. sulfanilamide, p-nitrobenzenesulfonyl hydrazide having a heterocyclic ring, e.g. sulfadiazine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/365Lactones
    • A61K31/366Lactones having six-membered rings, e.g. delta-lactones
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/4439Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a five-membered ring with nitrogen as a ring hetero atom, e.g. omeprazole
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/506Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/53771,4-Oxazines, e.g. morpholine not condensed and containing further heterocyclic rings, e.g. timolol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/56Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids
    • A61K31/58Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids containing heterocyclic rings, e.g. danazol, stanozolol, pancuronium or digitogenin
    • A61K31/585Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids containing heterocyclic rings, e.g. danazol, stanozolol, pancuronium or digitogenin containing lactone rings, e.g. oxandrolone, bufalin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Epidemiology (AREA)
  • Immunology (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Rheumatology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

The administration of an Akt inhibitor in a suitable carrier to a rheumatoid arthritis synovial fibroblast affords a process for inducing rheumatoid arthritis synovial fibroblast apoptosis. The Akt inhibitor is administered either as an active molecule or as a gene sequence expressible within rheumatoid arthritis synovial fibroblast cells. The gene sequence can be encompassed within a gene vector such as an adenovirus. A process for assaying rheumatoid arthritis drug candidates for apoptosis affect includes exposing a culture of rheumatoid arthritis synovial fibroblast cells to a drug candidate and monitoring apoptosis in the culture in the presence of the drug candidate.
Apoptosis in the culture is compared to apoptosis induced in a duplicate culture in the presence of a known Akt inhibitor.

Description

AKT AND REGiTLATION OF
RA SYNOVIAL FIBROBLAST APOPTOSIS
Field of the Invention The present invention relates to Akt regulation of rheumatoid arthritis synovial fibroblast (R.ASF) apoptosis and, more particularly, to a screening assay for native Akt activity and a method of regulating R.A synovial fibroblast apoptosis.
Background of the Invention Synovial hyperplasia is a hallmark of rheumatoid arthritis (Feldmann et al. Annual Review of Immunology. 1996; 14:397-440; Arend et al. 1995; AYth.
Rheum. 38:151-60). One of the primary cytokines associated with synovial hyperplasia is tumor necrosis factor-alpha (TNF-a). TNF-a is produced primarily by activated macrophages, and is capable of inducing cell proliferation, activation, and apoptosis (Alvaro-Gracia et al. J. Cliya.
Invest.
1 S 1990; 86:1790-8). In synovial fibroblasts, TNF-a drives proliferation (Kim et al. J. Inamunol. 2000; 164:1576-81; Aupperle et al. J. Irnmunol. 1999; 163:427-33; Kobayashi et al. Arth. Rheum. 1999; 42:519-26) and the production of collagenase and stromylysin, as well as other enzymes, that promote invasion of cartilage and bone.
TNF-a activates nuclear translocation of the transcription factor NF-~cB
and the TNFR-associated death domain-containing protein (TRADD) (Hsu et al. Cell 1995; 19;81:495-504). TNFR signaling also has been shown to activate phosphatidylinositol 3-kinase (PI 3-kinase) and protein kinase B
(Akt), in several cell types (Beraud et al. Proc. Natl. Acad. Sci. LISA 1999;
9;96:429-34; Ozes et al. Nature 1999; 2;401:82-5; Pastorino et al. J. Biol. Chem. 1999;
2;274:19411-6; Reddy et al. J. Immunol. 2000; 164:1355-63; Zhou et al. J.
Biol. Claem. 2000; 17;275:8027-8031; Sanz. EMBO J. 1999; 18:3044-53).
Akt is a serine-threonine protein kinase that is regulated by phosphatidylinositol 3,4,5-triphosphate (PIP3) and has been implicated in signaling of survival in a wide variety of cells, including fibroblastic, epithelial, and neuronal cells (Franke et al. Cell 1997; 1;88:435-7; Hemmings et al.

Science 1997; 275:628-30). Akt was first recognized as an anti-apoptosis factor during analysis of signaling by insulin-like growth factor-1 (IGF-1), which promotes the survival of cerebellar neurons (Dudek et al. Science 1997;
275:661-S). IGF-1 was shown to activate PI 3-kinase-triggered activation of S the serine-threonine kinase, Akt. An anti-apoptosis role for Akt also has been identified in NIH 3T3 fibroblasts (Goruppi et al. Mol. Cell Biol. 1997;
17:4442-S3).
The phosphatase and tensin homologue deleted on chromosome 10 gene (PTEN) is a phosphatase and tensin homologue suppressor gene located on human chromosome 10q23 (Li et al. Science 1997; 275:1943-7; Steck et al.
Nat. Genet. 1997; 1S:3S6-62; Olschwang et al. Nat. Genet. 1998; 18:12-4;
Liaw et al. Nat. Genet. 1997; 16:64-7). PTEN is deleted or mutated in a wide range of human cancers, including glioblastoma, melanoma, and prostate, breast, and endometrial cancers. The domains of PTEN share a high degree of 1 S homology with the family of protein-tyrosine phosphatases and the cytoskeletal protein, tensin. PTEN functions as a dual-specificity phosphatase and lipid phosphatase in vitro (Furnari et al. Proc. Natl. Acad. Sci. USA 1997; 94:12479-84). Specific substrates include phosphatidylinositol 3,4,5-trisphosphate (PIP3). PTEN has been shown to increase the sensitivity of the cell death response to several apoptotic stimuli, including LTV irradiation and treatment with TNF-a.
Previous investigators have reported that ceramides exhibit a growth-promoting anti-apoptotic signal in rheumatoid synovial cells, and that treatment with C2-ceramide completely inhibits PDGF-induced cell cycle progression of rheumatoid synovial cells (Romashkova et al. Nature 1999; 401: 86-89).
Increased activity of Akt has been reported in 293 cells and is involved in the activation of NF-~cB by TNF-a, following the activation of PI3-kinase.
Constitutively active Akt induces NF-~cB activity, mediated by phosphorylation of hcBI~-a (Ozes et al. Nature 1999; 2;401:82-S). In contrast, platelet-derived growth factor (PDGF), but not TNF-a, has been reported to activate NF-kB
through activation of Akt in normal human skin fibroblasts and in rat fibroblast-like synoviocytes (Romashkova et al. Nature 1999; 401: 86-89).
RASF exhibit increased proliferation in vitro, an increased response to TNF-a, and increased production of matrix metalloproteinases (Zhang et al.
Cell Dev. Biol. 1997; 33:37-415). It has been shown previously that TNF-a signaling of the TNF receptor results in phosphorylation of IxB and degradation by the proteosome (Ones et al. Nature 1999; 2;401:82-5; Zhang et al. Arth. Rheum. 2000 43(5):1094-105). This leads to nuclear translocation of NF-xB which upregulates anti-apoptosis genes including XIAP (Zhang et al.
Arth. Rheum. 2000; 43(5):1094-105; Reddy et al. J. Immufaol. 2000;
1;164:1355-6). PI3-kinase has recently been shown to induce NF-xB
activation (Reddy et al. J. Immunol. 2000; 1;164:1355-6) and TNF receptor signaling has been shown to activate Akt (Ozes et al. Nature 1999; 2;401:82-5;
Reddy et al. J. Immuuol. 2000; 1;164:1355-6).
While Akt has an anti-apoptosis role in regard to IGF-1 and IVIIi 3T3 fibroblasts, no apoptotic role has been determined in regard to RASFs.
Determination of Akt operation in RASF apoptosis has implications in suppressing synovial hyperplasia.
Summary of the Invention A process of inducing rheumatoid arthritis synovial fibroblast apoptosis includes the step of administering an Akt inhibitor in a suitable carrier to a rheumatoid arthritis synovial fibroblast. An Akt inhibitor in a suitable carrier in contact with a rheumatoid arthritis synovial fibroblast represents a composition useful in inducing fibroblast apoptosis. A vector such as an adenovirus includes a gene expressible within rheumatoid arthritis synovial fibroblasts, the gene encoding a polypeptide having an inhibitory effect on a Akt.
A process for assaying rheumatoid arthritis drug candidate apoptosis affect includes the steps of exposing a culture of rheumatoid arthritis synovial fibroblast to a drug candidate and monitoring apoptosis of the culture in the presence of the drug candidate. A comparison of apoptosis in the culture to apoptosis induced in a duplicate culture in the presence of a controller known Akt inhibitor affords a measure of drug candidate efficacy.
Descri tion of the Drawings Figure 1 (A-F) are micrographs of primary R.ASF (A, C, E) and OASF
(B, D, F) grown to confluence and stimulated with either PBS control (A, B) with TNF-a (10 ng/ml) (C, D) or TNF-a (10 ng/ml) plus wortmannin (50 nM) (E, F). After 6 hours, the cells were washed and stained with an anti-phosphorylated Akt (Thr308) antibody and revealed by DAB substrate. Cells were photographed at 40X.
Figure 2(A) are Western blots of cellular extracts prepared from BASF
and OASF treated either with PBS, TNF-a, or TNF-a plus wortmamlin as described in Figure 1. The levels of phosphorylated-Akt and total Akt were determined by Western blot analysis of lysates from RASF and OASF cell lines. Identical blots were probed with an antibody for: upper panel, phosphorylated Akt; middle panel, Akt; and lower panel, anti-b-actin. Lane 1, RASF treated with PBS; lane 2, R.ASF treated with TNF-a 10 ng/ml); lane 3, RASF treated with TNF-a 10 ng/ml + wortmannin (50 nM); lane 4, OASF
treated with PBS; lane 5, OASF treated with TNFa 110 ng/ml); lane 6, OASF
treated with TNF-a 10 ng/ml) + wortmannin (50 nM). (B) axe Akt-kinase activity in BASF and OASF. Akt from equal amounts of total proteins was immunoprecipitated from RA cell line RA68 or an OA cell line. The Akt kinase activity of cell lysates was revealed using a GSK-3 fusion protein.
Lane 1, unstimulated; lane 2, stimulation with TNF-a (10 ng/ml); lane 3, stimulation with TNF-a (10 ng/ml) + worhnannin (50 nM); lane 4, treatment with SV40 DNA which stimulates Akt-kinase activity.
Figure 3 is a histogram showing apoptosis of BASF cells treated with TNF-a ng/ml or wortmannin alone or TNF-a (10 ng/ml) with different concentrations of wortmannin determined after 18 hours by ATPlite assay.
Each data point represents the mean +/- SEM of 5 replications for each bar.
(*) indicates groups that are statistically significant different (p < 0.05) compared to the other groups.
Figure 4(A-B) are micrographs showing RASF cells transfected with (A) AdGFP or (B) AdAkt-DN (50 pfu/cell) and cultured for 18 hr, and 5 analyzed for expression of phosphorylated Akt by immunohistochemical staiiung at 40x. ' Figure 4(C) is a Western blot of BASF transfected with either 50 pfu/cell of AdGFP (lane 1), or with AdAkt-DN at a concentration of 5 pfu/cell (lane 2) or 50 pfu/cell (lane 3) for 18 hr. The cells were stimulated with TNFa (10 ng/ml) for 18 hr and Western blot analysis was carned out using an anti-phosphorylated Akt antibody or an anti-(3-actin antibody.
Figure 5(A-D) are a histologic analyses of apoptosis of RASF
transfected by AdAkt-DN in the presence of TNF-a. RASF cell lines were transfected with (A) AdGFP (100 pfu/cell) or different amounts of AdAkt-DN
and cultured for 18 hr. (B) 5 pfulcell; (C) 50 pfu/cell; (D) 100 pfu/cell. The cells were then incubated with TNF-a (10 ng/ml) for an additional 18 hr. The apoptosis was determined by microscopic analysis of morphology using an inverted fluorescent microscope.
Figure 6 is a plot of cytotoxicity of synovial fibroblasts as a function of AdAkt-DN transfection quantity (pfu/cell) in the presence of TNF-a for 3 different R.ASF and OASF cell lines. Cytotoxicity was quantitated using the ATPLite assay. There was no significant cytotoxicity of either RASF or OASF
18 hr after transfection with AdGFP in the presence of TNF-a (10 ng/ml) (dashed line).
Figure 7 is a plot of cytotoxicity of synovial fibroblasts as a function of AdPTEN transfection quantity (pfu/cell) in the presence of TNF-a for 3 different R.ASF and OASF cell lines. Cytotoxicity was quantitated using the ATPLite assay. There was no significant cytotoxicity of either BASF or OASF
I8 hr after transfection with AdPTEN followed by incubation with TNF-a (dashed line).
Detailed Description of the Invention The expression and activation of Akt in synovial fibroblasts from patients with rheumatoid arthritis (R.ASF), using synovial fibroblasts obtained from patients with osteoarthritis (OASF) as the controls inoperative according to the present invention as a heretofore unknown method of inducing apoptosis in BASF.
The present invention derives from the discovery that the levels of phosphorylated-Akt are higher in rheumatoid arthritis synovial fibroblasts (BASF) than in osteoarthritis synovial fibroblasts (OASF), as demonstrated by immunohistochemical staining, immunoblot analysis and an Akt kinase assay.
The levels of Akt, phosphorylated Akt and Akt kinase activity are increased by stimulation of primary BASF with tumor necrosis factor-alpha (TNF-a).
Treatment of BASF with the PI 3-kinase inhibitor, wortmannin, and a constant dose of TNF-a resulted in apoptosis of greater than half of BASF within 24 hours. This pro-apoptosis effect is specific for Akt, as equivalent levels of apoptosis are observed upon TNF-a treatment of BASF transfected with an adenovirus expressing a dominant negative-Akt (AdAkt-DN). Similarly, apoptosis is induced by TNF-a treatment of RASF transfected with an adenovirus expressing PTEN (AdPTEN), which opposes the action of Akt, indicating that phosphorylated Akt acts as a survival signal in RASF and contributes to the stimulatory effect of TNF-a on these cells by inhibiting the apoptosis response. This effect is not observed in OASF.
In another embodiment, the present invention is operative as a process for assaying RA apoptotic drug candidate efficacy. A drug candidate is exposed to a BASF cell culture prepared as described herein. Apoptosis is monitored in the cell culture as a function of time. It is appreciated that the cell culture is optionally stimulated towards apoptosis with an apoptotic stimulant such as TNF-a or other TNF isoforms. Upon comparison of cell culture apoptosis in the presence of a drug candidate to a control culture exposed to a substance having a known apoptotic effect, the efficacy of the drug candidate is assessed.
It is noted that catalytically active variants and fragments of Akt inhibitors are also operative according to the present invention to induce BASF
apoptosis.
The present invention provides for an Akt inhibitor; wherein the Akt inhibitor is a molecule illustratively including a cyclooxygenase-2 inhibitor, a pyridinyl imidazole inhibitor, a Ber-Abl tyrosine kinase inhibitor and a PI-3 kinase inhibitor. An example of a pyridinyl imidazole is SB203580 conunercially available from Calbiochem-Novabiochem. An example of a Ber-Abl tyrosine kinase inhibitor is CGP57148B, also known as STI-571, made by Novartis Pharma AG. An example of a PI-3 kinase inhibitor is LY294002, also known as 2-(4-morpholinyl)-8-phenyl-4H-1-benzopyran-4-one, commercially available from Calbiochem. An example of a cyclooxygenase~2 inhibitor is celecoxib.
It will be appreciated by those skilled in the art that Akt inhibition is achieved by inhibition of factors that cause an increase in Akt levels, activity or phosphorylation or which are necessary for Akt activation. Factors known to increase Akt or which are necessary for Akt activation illustratively include insulin-like growth factor-l, IL-l, PDGF, focal adhesion kinase, lipoarabinomannan and Syk.
It will be appreciated by those skilled in the art that stimulation of Akt activation is useful in inhibiting apoptosis.
The present invention provides for an Akt inhibitor protein or expressible gene to an BASF through incubation exposure or gene delivery, respectively; wherein the Akt inhibitor polypeptide includes a PI-3 kinase inhibitor such as wortmannin, and the Akt inhibitor expressing gene encodes a phosphatase such as PTEN, or dominant-negative Akt. Certain truncations of these proteins or genes perform the regulatory or enzymatic functions of the full sequence protein or gene. For example, the nucleic acid sequences coding therefor can be altered by substitutions, additions, deletions or multimeric expression that provide for functionally equivalent proteins or genes. Due to the degeneracy of nucleic acid coding sequences, other sequences which encode substantially the same amino acid sequences as those of the naturally occurring proteins may be used in the practice of the present invention. These include, but are not limited to, nucleic acid sequences including all or portions of the nucleic acid sequences encoding the above polypeptides, which are altered by the substitution of different codons that encode a functionally equivalent amino acid residue within the sequence, thus producing a silent change. For example, one or more amino acid residues within a polypeptide sequence can be substituted by another amino acid of a similar polarity which acts as a functional equivalent, resulting in a silent alteration. Substitutes for an amino acid within the sequence may be selected from other members of the class to which the amino acid belongs. For example, the nonpolar (hydrophobic) amino acids include alanine, leucine, isoleucine, valine, proline, phenylalanine, tryptophan and methionine. The polar neutral amino acids include glycine, serine, threonine, cysteine, tyrosine, asparagine, and glutamine. The positively charged (basic) amino acids include axginine, lysine and histidine. The negatively charged (acidic) amino acids include aspartic acid and glutamic acid. Also included within the scope of the present invention are proteins or fragments or derivatives thereof which are differentially modified during or after translation, e.g., by glycosolation, protolytic cleavage, linkage to an antibody molecule or other cellular ligands, etc. In addition, the recombinant vector encoding nucleic acid sequences of the present invention Akt inhibitor may be engineered so as to modify processing or expression of a vector. For example, a signal sequence may be inserted upstream of an inhibitor encoding sequence to permit inhibitor secretion and thereby facilitate apoptosis.
Additionally, an inhibitor encoding nucleic acid sequence can be mutated in vitro or in vivo to create and/or destroy translation, initiation, and/or termination sequences or to create variations in coding regions and/or form new restriction endonuclease sites or destroy pre-existing ones, to facilitate further ih vitro modification. Any technique for mutagenesis known in the art can be used, including but not limited to in vitro site directed mutagenesis, J.
Biol. Chem. 253:6551, use of Tab linkers (Phannacia), and the like.
The present invention further provides for an Akt inhibitor that is an antisense Akt nucleic acid. In order to inhibit Akt by an antisense approach, oligonucleotides (either DNA or RNA) that are complementary to Akt mRNA
are synthesized and administered by methods known to those skilled in the art.
In addition, an anti-Akt antibody is appreciated to act as an AKT inhibitor.
Such an antibody is created by methods common to the art.
An Akt inhibitor polypeptide or gene vector and TNF-a are administered in vivo by modes illustratively including parenterally, intrasynovially and topically with a carrier or diluent suitable for the mode of administration. Suitable carriers and diluents for each mode of administration are known to those skilled in the art. It is appreciated that additional adjuvants known to those skilled in the art are optionally added to a Garner or diluent.
Thus, an Akt inhibitor formulation suitable for injection illustratively includes aqueous and nonaqueous, isotonic sterile inj ection solutions, which can contain antioxidants, buffers, bacteriostats, and solutes that render the formulation isotonic with the blood of the intended recipient, and aqueous and nonaqueous sterile suspensions that can include suspending agents, solubilizers, thickening agents, stabilizers, and preservatives.
The present invention operates to inhibit expression of activated Akt that plays a central role in inhibition of apoptosis of RASF. Thus, apoptosis resistance enables cells that normally occur at a low frequency to become the predominant cell type within a pathologic lesion. The disablement of the resistance mechanism is expected to return normal cell type proportions. The enhanced expression of phosphorylated Akt in early-passage primary RASF is noted as an indicator for utilization of the present invention.
The present invention demonstrates that Akt is constitutively activated, and further activated in response to TNF-a, in early passage synovial fibroblasts from several patients with RA and that inhibition of Akt serves to induce RASF apoptosis. Lead molecules that affect Akt activity are summarized in Table 1.
Table 1. Lead Molecules that Affect Akt Small Com ound Action Cell Reference cyclo-oxygenaseBlock Akt Prostate Hsu et al.
2 cancer inhibitor JBC 275, 11397, (Celebrex) 2000 Celecoxib Pyridinyl Block Akt IL-2 activatedLali et al.

imidazole inhibitor T cell JBC 275, 7395, Bcr-Abl specificBlock Akt HL-60 K562 Fang et al.

tyrosine kinase(decrease IAP, Blood 96, 2246, inhibitor Bcl-xL NF~cB) 2000 CGP57148B (STI-Biologics Com ound Action Cell Reference lipo Activates Akt THP=1 cell Maiti et al.

arabinomannin I. Biol. Chem.
Oct.

(LAM) for M. 2000 Tuberculosis Pathways Com ound - Action Cell Reference Insulin-like Activates Akt Many Many growth ' factor-1, IL-1, PDGF

SSby Activates Akt H202 treatedDing et al.
B

cells JBC 275, 30873, Focal adhesion Activates Akt HL-60 Sohoda et al.

kinase JBC 275, 16309, Akt activation is activated by TNF-oc ligation of the TNFR, and this 5 leads to increased NF-oB activation in BASF. It will be appreciated that Akt is activated by multiple members of the TNFR superfamily known to those skilled in the art, illustratively including TNFR-I, TNFR-II and LT-beta R. It will be further appreciated by those skilled in the art that multiple members of the class of TNFR superfamily ligands, illustratively including TNF-a, TNF-[3, 10 TRAIL, LT-beta and heteromers of these or other ligands bind to TNFR

superfamily members to activate Akt. In addition, TNFR-activating antibodies, such as SSR539 commercially available from R and D Systems, activate Akt.
Akt activity is found to be constitutively higher in RASF compared to OASF. This is shown by ailalysis of phosphorylated Akt from RASF and OASF, and also by immunoprecipitation of activated Akt and evaluation of activity using GSK-3 as the substrate. Furthermore, Akt activity is greatly enhanced in BASF in the presence of TNF-a, but this enhancement was not observed in OASF. Thus, there is an intrinsic dysfunction associated with the higher expression of activated Akt in RASF than in OASF as a result of genetic or environmental factors, leading to enhanced activation of Akt in response to TNF-a. Over expression of TNF-a combined with this intrinsic enhanced expression of activated Akt leads to inhibition of apoptosis of RASF and, consequently, a growth advantage for these synovial fibroblasts. Since rheumatoid arthritis is a chronic inflammatory disease that is characterized by a synovial hyperplasia that develops over many years, there is gradual selection for R.ASF that express high levels of phosphorylated Akt in response to TNF-a. This is consistent with the inventive intervention demonstrating that 45%
of RASF constitutively express phosphorylated Akt, and undergo TNF-a mediated apoptosis in response to blocking phosphorylation of Akt with Akt-DN or AdPTEN. In contrast, OASF have not developed an anti-apoptosis pathway that depends upon phosphorylated Akt, and subsequently, blocking of phosphorylated Akt activity does not result in increased susceptibility to apoptosis in response to TNF-a Akt is regulated in BASF by addition of TNF-a according to the present invention. TNF-a is produced at high levels by the macrophages present in rheumatoid synovium and inhibition of TNF-a by soluble TNF-a receptors is one of the most efficacious therapies for rheumatoid arthritis (sTNFRII, Etanercept) (Moreland et al. N. Engl. J. Med. 1997, 337:141-7;
Weinblatt et al. N. Engl. J. Med. 1999, 340:253-9) and sTNFRI (McCabe et al.
Ar~tla. Rheum. 1998, 9(supplement) 558; Edwards CK 3rd. Ann. Rheum. Dis.

1999, 58 Suppl 1:I73-81; Bendele et al. Clin. Exp. Rheumatol. 1999, 17:553-60; Su et al. Arthritis Rheum. 1998, 41:139-49). The present results suggest that one of the mechanisms contributing to the efficacy of anti-TNF-a therapy in RA is the down-modulation of the activation of Akt thereby indicating a novel mode of inducing R.ASF apoptosis and alleviating RA symptoms.
It has been shown that treatment with TNF-a results in increased proliferation of BASF but not OASF (Migita et al. Biochem. Biophys. Res.
Commun. 2000; 269:70-5). To determine if this TNF-a survival signal involves a PI 3-kinase dependent mechanism, RASF and OASF are treated with TNF-a (10 ng/ml), using PBS as a control, in the presence and absence of the PI 3-kinase inhibitor, wortmannin (50 nM). After 18 hours, the synovial fibroblasts are stained using an antibody specific for phosphorylated-Akt. The constitutive expression of phosphorylated Akt is higher in the PBS-treated R.ASF than in the OASF as shown in Figures 1A and 1B. Treatment with TNF-a resulted in a greatly enhanced production of phosphorylated Akt by BASF, whereas this effect was much less pronounced in OASF as shown in Figures 1C and 1D). On immunohistochemical staining, strong expression of phosphorylated-Akt is detected in 45 ~ 10% of primary RASF compared to 20 ~ 8% of primary OASF. Treatment with the PI 3-kinase inhibitor, wortmannin inhibited this increase in expression of phosphorylated Akt in response to TNF-a in BASF as shown in Figures 1E and 1F. Thus, primary BASF cells exhibit higher expression of phosphorylated Akt than do OASF cells in the absence of exogenous stimulation with TNF-a. Moreover, stimulation with TNF-a greatly increased the expression of phosphorylated Akt in R.ASF and this enhanced expression requires PI 3-kinase activation.
To confirm the results of the histologic analyses, cellular extracts are prepared from RASF and OASF treated either with PBS, TNF-a, or TNF-a plus wortmannin. The levels of phosphorylated-Akt are determined by Western blot analysis of lysates from RASF and OASF cell lines. The levels of phosphorylated Akt are higher in the unstimulated RASF compared to the OASF as shown in Figure 2A, top panel, lane 1,4. TNF-a results in a marked increase in the levels of phosphorylated-Akt in BASF, but did not affect the levels of phosphorylated Akt in OASF as shown in Figure 2A, top pmel, lane 2,5. This increase in phosphorylated Akt in RASF is inhibited by wortmannin, as shown in Figure 2A, top panel, lane 3. There is no significant change in total Akt in RASF as compared to OASF independent of treatment, as shown in Figure 2A, middle panel. Regardless of treatment, the level of [3-actin protein is not changed, which indicates that equal amounts of total protein have been loaded as shown in Figure 2A, lower panel.
To directly determine the Akt-kinase activity in BASF and OASF, Akt from equal amounts of total protein is immunoprecipitated from cell lysates and the kinase activity of Akt revealed using a GSK-3 fusion protein as the substrate in the presence of ATP and kinase buffer. Active Akt phosphorylates GSK-3 at serine 219. The Akt-kinase activity is relatively low in unstimulated RASF line RA68 as shown in Figure 2B, lane 1, top panel. However, 24 hours after stimulation of RASF with TNF-a (10 ng/ml), there is a 20-fold increase in Akt-l~inase activity as shown in Figure 2B, lane 2, top panel, and this increase in activity is blocked by pretreatment with wortmannin as shown in Figure 2B, lane 3, top panel. In contrast, untreated OASF exhibited no detectable Akt kinase activity and treatment with TNF-a (10 ng/ml) did not induce Akt kinase activity in OASF as shown in Figure 2B, lower panel. Cells transfected with an SV40 expression plasmid, which strongly stimulates Akt-kinase activity in cells (Summers et al. Biochem. Biophys. Res. C~mmun. 1998; 246:76-81) were used as a positive control as shown in Figure 2B, lane 4, top and lower panels.
Thus, the intrinsic activation of Akt is higher in R.ASF than OASF.
Furthermore, stimulation with TNF-a greatly enhances Akt activity in RASF, but has no effect on Akt activity in OASF. These results support the histologic evidence that the levels of phosphorylated Akt are higher in RASF than in OASF as shown in Figure 1.

BASF cells are treated with TNF-a alone or TNF-a with wortmannin (50 nM), and the extent of apoptosis monitored by the ATPlite assay. Neither treatment with TNF-a nor wortmannin alone could induce cytotoxicity in BASF as shown in Figure 3, whereas treatment with TNF-a plus wortmannin resulted in extensive apoptosis. This is consistent with the ability of TNF-a to activate PI 3-kinase in these cells and indicates that TNF-a mediated apoptosis of RASF is potentiated by inhibition of PI 3-kinase activity.
The expression of phosphorylated Akt is higher in the primary cultures of cells from patients with RA than in the primary cultures of cell from patients with OA as shown in Figure 1. AdAkt-DN inhibits endogenous phosphorylation of Akt. A control, AdGFP shown in Figure 4A or AdAkt-DN
shown in Figure 4B is transfected (50 pfu/cell) into an BASF cell line. The cells are cultured 18 hr, and analyzed for expression of phosphorylated Akt by immunohistochemical staining. Decreased expression of phosphorylated Akt by BASF transfected with AdAkt-DN is noted compared to BASF transfected with AdGFP.
To determine the optimal amount of AdAkt-DN required to inhibit Akt phosphorylation, BASF is transfected with either 50 pfu/cell of AdGFP or with 5 pfu/cell or with 50 pfu/cell of AdAkt-DN respectively for 18 hr. The cells are then stimulated with TNFa (10 ng/ml) for 18 hr. Phosphorylated Akt in RASF increased 18 hours after stimulation with TNF-a (10 ng/ml) in cells transfected with AdGFP as shown in Figure 4C, Lane 1. AdAkt-DN inhibits TNFa induced phosphorylation of Akt in a dose-dependent fashion as shown in Figure 4C, Lane 2 and 3. The total proteins are loaded in equal amounts as indicated by the levels of (3-actin as shown in Figure 4C.
RASF cell lines are transfected with AdGFP (100 pfu/cell) or different amounts of AdAkt-DN (5, 50, 100 pfu/cell) and cultured for 18 hr. The cells are then incubated with TNF-a (10 ng/ml) for an additional 18 hr. Apoptosis is' determined by microscopic analysis of the morphology of the RASF using an inverted microscope. There is no apoptosis of RASF after treatment with control AdGFP plus TNF-a as shown in Figure SA. There was a dose-dependent increase in apoptosis of R.ASF after treatment with AdAkt-DN at 5, 50 and 100 pfulcell, respectively as shown in Figure 5, B, C, D. Low apoptosis of 1~ASF after transfection with AdAkt-DN is observed in the absence of TNF-5 a. Thus, treatment with AdAkt-DN plus TNF-a converts the TNF-a apoptosis resistant BASF to TNF-a sensitive RASF.
The effect of AdAkt-DN on apoptosis is analyzed using three different primary BASF and primary OASF cell cultures at different pfu/cell of AdAkt-DN using the ATPLite assay. Cells are transfected with AdAkt-DN or AdGFP
10 for 18 hr, and then incubated with TNF-a (10 ng/ml) for an additional 18 hr.
There is a dose-dependent increase in cytotoxicity of BASF with increasing pfu/cell of AdAlct-DN in the presence of TNF-a (10 ng/ml) as shown in Figure 6. In contrast, transfection of OASF with increasing concentrations of AdAkt-DN resulted in only minimal apoptosis after treatment with TNF-a. Neither 15 BASF nor OASF cell lines exhibited significant cytotoxicity after transfection with AdGFP in the presence of TNF-a (10 ng/ml) as demonstrated for exemplary RASF cell line RA2, by the dashed line.
Three different BASF and OASF primary cell cultures are transfected with AdPTEN for 18 hr followed by treatment with TNF-a (10 ng/ml) for 18 hr. Cytotoxicity is analyzed using the ATPLite assay. As a control, cells are treated with AdGFP plus TNF-a as described with respect to Figure 6. There is a dose-dependent increase in cytotoxicity of RA.SF with increasing pfulcell of AdPTEN in the presence of TNF-a (10 ng/ml) as shown in Figure 7) In contrast, transfection of OASF with increasing concentrations of AdPTEN
resulted in only minimal apoptosis after treatment with TNF-a. Neither R.A.SF
nor OASF cell lines exhibited significant cytotoxicity after transfection with AdGFP in the presence of TNF-a (10 ng/ml) as demonstrated for an exemplary R.ASF cell line, by the dashed line.

The following examples are given for the purpose of illustrating various embodiments of the invention and are not meant to limit the present invention in any fashion.
Exafuple 1- Patient selection.
Patients were recruited who were undergoing total knee replacement related to rheumatoid arthritis or osteoarthritis. All patients met the 1987 ACR
Criteria for rheumatoid arthritis of the knee (Arnett et al. Arthritis Rheum.
1988; 31:315-24). The diagnosis of OA was based on clinical and radiological findings. The patients ranged in age from 43-77 with a mean age of 63.
Synovial cell lines were established from 4 female and 1 male patients. All patients were receiving non-steroidal anti-inflammatory drugs (NSAIDs), but none were receiving steroids or prednisone at the time of surgery.
Example 2 - Reagents.
TNF-a was purchased from R&D Systems (Minneapolis, MN) and used at 10 ng/ml. This is optimal based on previous data (Zhang et al. Cell Dev. Biol. 1997;33:37-41). Wortmannin was purchased from Sigma (St. Louis, MO) and dissolved in DMSO, aliquoted, stored at -80°C until use.
The SV40 plasmid which expresses the polyoma T antigens was purchased from New England Biolab (Beverly, MA) and was used as a positive control for the Akt-kinase assay (Summers et al. Biochem. Biophys. Res. Commun. 1998; 246:76-81).
Example 3 - Primary syuovial cell culture.
Primary synovial cell lines are established as described previously (Zhang et al. Cell Dev. Biol. 1997; 33:37-41). The synovial tissue is minced into small pieces (~lmm3) in prewarmed DMEM then incubated for 2 hours at 37°C in the presence of 1 mg/ml of type I collagenase (Sigma, St.
Louis, MO).
After dissociation of the f broblasts, the cells are harvested by centrifugation at 1,000 rpm for 5 min and plated in 25 cm2 flasks in 8 ml of DMEM
supplemented with IO% fetal bovine serum (FBS). The cells are cultured to 80% confluency before use in experiments. All in vitro experiments are carried out using primary synovial cell cultures at passage numbers between 4-10.

Example 4 - Construction of AdAkt DN and au AdPTEN expression vector.
Akt constructs encoding catalytically inactive Akt (K179M) inserted into pCMV6 vectors and pCMV6 vectors are utilized as previously described (Gu et al. Proc. Natl. Acad. Sci. USA 1997; 94:11345-SO). The Akt (K179M) S is cloned into the ElA deletion site of adenovirus of pAdCMV (He et al.
Proc.
Natl. Acad. Sci. USA 1998; 95:2509-2514) in which green fluorescence protein (GFP) is co-expressed with the mutated Akt driven by the CMV promoter, respectively. A recombinant adenovirus is produced by co-transfection of pAdCMVAkt-DN with pJMl7 in the 293 cell line. The recombinant adenovirus expressing mutant Akt (AdAkt-DN) is selected and purified using standard procedures (McGrory et al. Virology 1988; 163:614-7). The correct orientation and cloning of the Akt-DN in recombinant AdAkt-DN is confirmed by PCR sequence analysis. To obtain a large quantity of recombinant AdAkt-DN, the 293 cells are infected and grown for 48 hours at 37°C prior to harvest 1 S and centrifugation using a tabletop centrifuge at 4,000 rpm for 20 min.
The infected cells are resuspended in PBS buffer, then lysed using three freeze-thaw cycles. The released virus is purified through two CsCI gradients and then the purified recombinant AdAkt-DN is titrated by plaque assay (McGrory et al. Tjirology 1988; 163:614-7), aliquoted, and stored at -80°C until use.
The PTEN fragment is constructed as described previously (Ghosh et al. Gene 1999; 23S:8S-91). The fragment is subsequently cloned into the E1 deleted site of an adenovirus shuttle pAdCMV in which the PTEN gene is driven by the CMV promoter. A recombinant adenovirus is then constructed and characterized as described (Ghosh et al. Gerae 1999; 23S:8S-91).
2S Example S - Immuuolaistochemical analysis of primary syuovial cell cultures.
The in-situ expression of phosphorylated-Akt is determined by staining fixed cells with an antibody specific for Thr308 phosphorylated Akt (New England Biolabs, Beverly, MA). Quenching of endogenous peroxidase is performed by incubating tissue sections with 3% HZOZ at RT for 10 min in a humidified chamber. After washing with PBS, tissue sections are incubated with 0.1% trypsin at 37°C for 10 min to reveal fixed antigen epitopes.
Tissue sections are then treated with denaturing solution at 20°C for 30 min and blocking solution at RT for 10 min followed by incubation with a horseradish peroxidase (HRP) conjugated antibody specific for phosphorylated-Akt antibody (New England Biolabs, Beverly, MA). Slides are incubated with S DAB staining kit (DAKO) for color development then counterstained with methyl green. At least 5 areas from each specimen are chosen randomly for assessment of the percentage of Akt-positive cells.
Example 6 - Evaluation of Akt activatiofZ afad immuhoblot analysis.
For evaluation of the phosphorylated form of Akt, the synovial cells are washed twice with ice-cold PBS followed by incubation with 200 ~1 of cell lysis buffer (20 mmol/L Tris [pH 7.4], 150 mmol/L NaCI, 1 mmol/L EDTA, 1 mmollL EGTA, 1% Triton, 2.5 mmol/L sodium pyrophosphate, 1 mmol/L 13-glycerophosphate, 1 mmol/L Na3V04, 1 ~g/mL leupeptin, and 1 mmol/L
PMSF) for S min on ice. The cells are then harvested by scraping and sonicated using a Branson Sonicator (3x 5 seconds, output control 2, duty cycle 100%) on ice. After centrifugation for 10 min at 20,OOOg at 4°C, the protein concentration of the supernatant is determined using the Bio-Rad protein assay kit ~(Bio-Rad, Hercules, CA). Fifty micrograms of total protein extracted from synovial fibroblasts are loaded on 10% SDS-PAGE mini gel (Bio-Rad, Hercules, CA), and electrophoresed at 100 V for 2 hours, followed by electro-transfer to nitrocellulose membrane. The membrane is then blotted with an antibody specific for Akt phosphorylated at Ser473 (New England Biolabs, Beverly, MA) overnight at 4°C. Phosphorylated Akt is detected using the PhosphoPlus Akt antibody kit (New England Biolabs, Beverly, MA). To monitor the amount of total proteins loaded on the gel, the membrane is restripped, and probed with mouse anti-human [3-actin antibody (clone AC-15, Sigma, St., Louis, MO), and the signal is amplified using HRP-conjugated goat anti-mouse antibody, and detected by the LumiGLO chemiluminescent reagent.
Example 7 - A~aalysis of AKT activity.
Akt kinase assay of R.A synovial fibroblasts is performed using Akt kinase assay kit according to the protocol provided by manufacturer (New England Biolabs, Beverly, MA). Briefly, three hundreds micrograms of total proteins from RA synovial fibroblasts are added into Akt antibody coated beads, incubated at 4°C for 3 hours followed by washing.
Phosphorylation of GSK-3 is used as an indicator of phosphorylated Akt, since Akt negatively regulates GSK-3a/b kinase activity via phosphorylation of GSK-3 at Ser219.
After the kinase reaction, the reaction mixture is electrophoresed on a 12%
SDS-PAGE gel and western blotted. The blots are probed with an anti-phospho-GSK-3a/b (Ser219) antibody (rabbit polyclonal IgG, affinity purified).
The blot is developed using an HRP-conjugated goat anti-rabbit antibody, and I O detected by the LumiGLO chemiluminescent reagent.
Example 8 - ATPlite M assay to analysis synovial fibvoblast cytotoxicity.
Cytotoxicity is determined by the ATPLite-M assay. ATP is a marker for cell viability because it is present in all metabolically active cells and the concentration declines very rapidly when the cells undergo apoptosis. The ATPLite-M assay system is based on the production of light caused by the reaction of ATP with added luciferase and D-luciferin (Crouch et al. J.
Imrnunol. Methods 1993; 160, 81-88). Briefly, R.ASF and OASF are transfected with different amounts of control Ad, AdAkt-DN or AdPTEN.
Eighteen hr later, the cells are stimulated in a 96 well plate in the presence or absence of TNF-a (10 ng/ml). The cytotoxicity is analyzed by the ATPlite-M
assay as described in manual (Packard Instrument Company, Meriden, CT).
Example 9 - Statistical Analysis.
Data are expressed as mean ~ SEM from at least 3 independent experiments. Statistical analysis was performed with ANOVA followed by a modified least significant difference test (SPSS Software).
Patent applications and publications mentioned in the specification are indicative of the levels of those skilled in the art to which the invention pertains. These applications and publications are incorporated herein by reference to the same extent as if each individual application or publication was specifically and individually incorporated herein by reference.

The foregoing description is illustrative of particular embodiments of the invention, but is not meant to be a limitation upon the practice thereof.
The following claims, including all equivalents thereof, are intended to define the scope of the invention.
5 References Hsu et al., The Cyclooxygenase-2 Inhibitor Celecoxib Induces Apoptosis by Blocking Akt Activation in Human Prostate Cancer Cells Independently of Bcl-2 , The Journal of Biological Chemistry, 275: 11397, 2000.
Lali et al., The Pyridinyl Imidazole Inhibitor SB203580 Blocks 10 Phosphoinositide-dependent Protein Kinase Activity, Protein Kinase B
Phosphorylation, and Retinoblastoma Hyperphosphorylation in Interleukin-2-stimulated T Cells Independently of p38 Mitogen-activated Protein Kinase, The Journal of Biological Chemistry, 275: 7395, 2000.
Fang et al., Blood, 96: 2246, 2000.
15 Maiti et al., Lipoarabinomannan from Mycobacterium tuberca~losis Promotes Macrophage Survival by Phosphorylating Bad through a Phosphatidylinositol 3-Kinase/Akt Pathway The Journal of Biological Chemistry 2000 276: 329-333.
Ding et al., Syk Is Required for the Activation of Akt Survival Pathway in B
20 Cells Exposed to Oxidative Stress, The Journal of Biological Chemistry, 275:
30873, 2000.
Sohoda et al., Anti-apoptotic Role of Focal Adhesion Kinase (FAK).
INDUCTION OF INHIBITOR-OF-APOPTOSIS PROTEINS AND APOPTOSIS
SUPPRESSION BY THE OVEREXPRESSION OF FAK IN A HUMAN LEUKEMIC CELL
LINE, HL-6o, The Journal of Biological Chemistry, 275: 16309, 2000.

Claims (31)

Claims
1. A process of inducing rheumatoid arthritis synovial fibroblast apoptosis comprising the steps of: administering an Akt inhibitor in a suitable carrier to a rheumatoid arthritis synovial fibroblast.
2. The process of claim I further comprising administering tumor necrosis factor to said rheumatoid arthritis synovial fibroblast.
3. The process of claim 1 wherein Akt inhibitor and carrier are administered intrasynovially.
4. The process of claim 1 wherein Akt inhibitor and carrier are administered parenterally.
5. The process of claim 1 wherein Akt inhibitor and carrier are administered topically.
6. The process of claim 1 wherein said Akt inhibitor is selected from the group consisting of wortmannin, a fragment thereof, and a polymorph thereof.
7. The process of claim 1 wherein said Akt inhibitor is administered as a gene sequence expressible within rheumatoid arthritis synovial fibroblast.
8. The process of claim 7 wherein said gene sequence is encompassed within a gene vector.
9. The process of claim 8 wherein said gene vector is an adenovirus.
10. A composition comprising: an Akt inhibitor in a suitable carrier in contact with rheumatoid arthritis synovial fibroblast.
11. The composition of claim 10 further comprising tumor necrosis factor in said carrier.
12. The composition of claim 10 wherein said Akt inhibitor is a PI
3-kinase inhibitor.
13. The composition of claim 10 wherein said Akt inhibitor is wortmannin.
14. The composition of claim 10 wherein said Akt inhibitor is a vector comprising a gene sequence expressible within the rheumatoid arthritis synovial fibroblast encoding a polypeptide selected from the group consisting of: wortmannin, anti-Akt and dominant negative Akt.
15. The composition of claim 14 wherein said vector is an adenovirus vector.
16. A vector comprising: a gene expressible within rheumatoid arthritis synovial fibroblasts encoding a polypeptide having an inhibitory effect on Akt.
17. The vector of claim 16 wherein said polypeptide is selected from the group consisting of: wortmannin, anti-Akt and dominant negative Akt.
18. Use of an Akt inhibitor or a fragment thereof for regulation of rheumatoid arthritis synovial fibroblast apoptosis.
19. A process for assaying rheumatoid arthritis drug candidate apoptosis comprising the steps of:
exposing a culture of rheumatoid arthritis synovial fibroblast to a drug candidate;
monitoring apoptosis of said culture in the presence of said drug candidate; and comparing apoptosis of said culture to apoptosis induced in a duplicate culture by a control Akt inhibitor.
20. The process of claim 19 wherein said control Akt inhibitor is selected from the group consisting of: wortmannin, celecoxib, SB203580, CGP57148B, and LY294002.
21. Use of an Akt inhibitor for the preparation of a composition for the treatment of rheumatoid arthritis.
22. The use of an Akt inhibitor in the manufacture of a medicament for the treatment of rheumatoid arthritis.
23. A composition as claimed in any of claims 10 to 15 as a rheumatoid arthritis synovial fibroblast apoptosis agent.
24. A process according to claim 1 substantially as described herein with reference to and/or as illustrated in the accompanying drawings.
25. The process of claim 1 wherein said Akt inhibitor is selected from the group consisting of a cyclooxygenase-2 inhibitor, a pyridinyl imidazole, a Ber-Abl tyrosine kinase inhibitor and a PI-3 kinase inhibitor.
26. The process of claim 25 wherein said PI-3 kinase inhibitor is LY294002.
27. The process of claim 25 wherein said pyridinyl imidazole is SB203580.
28. The process of claim 25 wherein said Ber-Abl tyrosine kinase inhibitor is CGP57148B.
29. The process of claim 25 wherein said cyclooxygenase-2 inhibitor inhibitor is celecoxib.
30. The process of claim 1 wherein said Akt inhibitor is a phosphatase.
31. The process of claim 40 wherein said phosphatase is selected from the group consisting of: PTEN, a fragment thereof, and a polymorph thereof.
CA002444840A 2001-04-16 2002-04-16 Akt and regulation of ra synovial fibroblast apoptosis Abandoned CA2444840A1 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US28396601P 2001-04-16 2001-04-16
US60/283,966 2001-04-16
PCT/US2002/011820 WO2002083075A2 (en) 2001-04-16 2002-04-16 Akt and regulation of ra synovial fibroblast apoptosis

Publications (1)

Publication Number Publication Date
CA2444840A1 true CA2444840A1 (en) 2002-10-24

Family

ID=23088331

Family Applications (1)

Application Number Title Priority Date Filing Date
CA002444840A Abandoned CA2444840A1 (en) 2001-04-16 2002-04-16 Akt and regulation of ra synovial fibroblast apoptosis

Country Status (3)

Country Link
EP (1) EP1414500A4 (en)
CA (1) CA2444840A1 (en)
WO (1) WO2002083075A2 (en)

Families Citing this family (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB0201882D0 (en) * 2002-01-28 2002-03-13 Novartis Ag Organic compounds
AU2003295320A1 (en) * 2002-06-26 2004-04-08 The Ohio State University Research Foundation The method for reducing inflammation using sti-571 or its salt
GR20050100526A (en) * 2005-10-19 2007-05-23 B.S.R.C. "Alexander Fleming" Deregelated genes and/or processes in inflamatory arthritis.
EP2177510A1 (en) 2008-10-17 2010-04-21 Universität des Saarlandes Allosteric protein kinase modulators

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5476851A (en) * 1994-09-08 1995-12-19 Rhone-Poulenc Rorer Pharmaceuticals, Inc. Pyrazolo[3,4-g]quinoxaline compounds which inhibit PDGF receptor protein tyrosine kinase

Also Published As

Publication number Publication date
WO2002083075A3 (en) 2004-03-04
WO2002083075A2 (en) 2002-10-24
EP1414500A2 (en) 2004-05-06
EP1414500A4 (en) 2005-01-12

Similar Documents

Publication Publication Date Title
Radbill et al. Loss of matrix metalloproteinase-2 amplifies murine toxin-induced liver fibrosis by upregulating collagen I expression
Hulley et al. Hypoxia‐inducible factor 1‐alpha does not regulate osteoclastogenesis but enhances bone resorption activity via prolyl‐4‐hydroxylase 2
Spirli et al. Cyclic AMP/PKA‐dependent paradoxical activation of Raf/MEK/ERK signaling in polycystin‐2 defective mice treated with sorafenib
Idris et al. Small molecule inhibitors of IκB kinase signaling inhibit osteoclast formation in vitro and prevent ovariectomy‐induced bone loss in vivo
Zhao et al. Naringin protects against cartilage destruction in osteoarthritis through repression of NF-κB signaling pathway
Yan et al. Breviscapine protects against cardiac hypertrophy through blocking PKC‐α‐dependent signaling
CA2384982A1 (en) Use of hymenialdisine or derivatives thereof in the manufacture of medicaments
Manoharan et al. Thioredoxin inhibits MPK38-induced ASK1, TGF‐β, and p53 function in a phosphorylation-dependent manner
Lee et al. Growth inhibitory and anti-tumour activities of OSU-03012, a novel PDK-1 inhibitor, on vestibular schwannoma and malignant schwannoma cells
US20110053860A1 (en) COX-2 function and wound healing
Liu et al. Vaccarin prevents titanium particle‐induced osteolysis and inhibits RANKL‐induced osteoclastogenesis by blocking NF‐κB and MAPK signaling pathways
Dong et al. NRF2 is a critical regulator and therapeutic target of metal implant particle-incurred bone damage
Guo et al. Tyloxapol inhibits RANKL-stimulated osteoclastogenesis and ovariectomized-induced bone loss by restraining NF-κB and MAPK activation
JP2020516689A (en) Compounds, compositions and methods for treating or preventing acute lung injury
EP3972608A1 (en) Oxathiazin compounds for inhibiting gapdh
Pan et al. Progranulin regulation of autophagy contributes to its chondroprotective effect in osteoarthritis
Fei et al. Magnolol prevents ovariectomy‑induced bone loss by suppressing osteoclastogenesis via inhibition of the nuclear factor‑κB and mitogen‑activated protein kinase pathways
WO2021184059A1 (en) Treatment methods
CA2444840A1 (en) Akt and regulation of ra synovial fibroblast apoptosis
Sugiura et al. Screening of a panel of low molecular weight compounds that inhibit synovial fibroblast invasion in rheumatoid arthritis
JP2007145745A (en) Lung cancer medicine containing src family tyrosine kinase inhibitor for inhibiting variant egfr downstream signal and utilization thereof
Yang et al. DNA damage-inducible transcript 3 restrains osteoclast differentiation and function
US20060104951A1 (en) Akt and regulation of RA synovial fibroblast apoptosis
AU2002303354A1 (en) Akt and regulation of RA synovial fibroblast apoptosis
Akca et al. Suppression of TNF-α mediated apoptosis by EGF in TNF-α sensitive human cervical carcinoma cell line

Legal Events

Date Code Title Description
FZDE Dead