Molecular targets and compounds, and methods to identify the same, useful in the treatment of joint degenerative and inflammatory diseases

Information

  • Patent Grant
  • 8673268
  • Patent Number
    8,673,268
  • Date Filed
    Thursday, January 5, 2012
    12 years ago
  • Date Issued
    Tuesday, March 18, 2014
    10 years ago
Abstract
The application discloses methods for identifying and using compounds that inhibit extra-cellular matrix (ECM) degradation and inflammation, using a polypeptide sequence including SEQ ID NO: 17-127 (hereinafter “TARGETS”) and fragments thereof, expression inhibitory agents such as antisense polynucleotide, a ribozyme, and a small interfering RNA (siRNA), comprising a nucleic acid sequence complementary to, or engineered from, a naturally occurring polynucleotide sequence encoding a polypeptide of SEQ ID NO: 17-127, useful in pharmaceutical compositions comprising said agent, for the treatment, or prevention, of chronic joint degenerative and/or inflammatory diseases such as rheumatoid arthritis.
Description
FIELD OF INVENTION

The present invention relates to methods for identifying compounds, and expression-inhibition agents, capable of inhibiting the expression of proteins involved in the pathway resulting in the degradation of extra-cellular matrix (ECM), which inhibition is useful in the prevention and treatment of joint degeneration and diseases involving such degradation and/or inflammation.


Diseases involving the degradation of extra-cellular matrix include, but are not limited to, psoriatic arthritis, juvenile arthritis, early arthritis, reactive arthritis, osteoarthritis, ankylosing spondylitis. osteoporosis, muscular skeletal diseases like tendonitis and periodontal disease, cancer metastasis, airway diseases (COPD, asthma), renal and liver fibrosis, cardio-vascular diseases like atherosclerosis and heart failure, and neurological diseases like neuroinflammation and multiple sclerosis. Diseases involving primarily joint degeneration include, but are not limited to, psoriatic arthritis, juvenile arthritis, early arthritis, reactive arthritis, osteoarthritis, ankylosing spondylitis.


Rheumatoid arthritis (RA) is a chronic joint degenerative disease, characterized by inflammation and destruction of the joint structures. When the disease is unchecked, it leads to substantial disability and pain due to loss of joint functionality and even premature death. The aim of an RA therapy, therefore, is not to slow down the disease but to attain remission in order to stop the joint destruction. Besides the severity of the disease outcome, the high prevalence of RA (□ 0.8% of the adults are affected worldwide) means a high socio-economic impact. (For reviews on RA, we refer to Smolen and Steiner (2003); Lee and Weinblatt (2001); Choy and Panayi (2001); O'Dell (2004) and Firestein (2003)).


Although it is widely accepted that RA is an auto-immune disease, there is no consensus concerning the precise mechanisms driving the ‘initiation stage’ of the disease. What is known is that the initial trigger(s) does mediate, in a predisposed host, a cascade of events that leads to the activation of various cell types (B-cells, T-cells, macrophages, fibroblasts, endothelial cells, dendritic cells and others). Concomitantly, an increased production of various cytokines is observed in the joints and tissues surrounding the joint (for example, TNF-α, IL-6, IL-1, IL-15, IL-18 and others). As the disease progresses, the cellular activation and cytokine production cascade becomes self-perpetuating. At this early stage, the destruction of joint structures is already very clear. Thirty percent of the patients have radiographic evidence of bony erosions at the time of diagnosis and this proportion increases to 60 percent-after two years.


Histologic analysis of the joints of RA patients clearly evidences the mechanisms involved in the RA-associated degradative processes. The synovium is a cell, layer, composed of a sublining and a lining region that separates the joint capsule from the synovial cavity. The inflamed synovium is central to the pathophysiology of RA. The synovial joint is =shown as composed of two adjacent bony ends each covered with a layer of cartilage, separated by a joint space and surrounded by the synovial membrane and joint capsule. The synovial membrane is composed of the synovial lining (facing the cartilage and bone), which consists of a thin (1-3 cells) layer of synoviocytes and the sublining connective tissue layer that is highly vascularised. The synovial membrane covers almost all intra-articular structures except for cartilage. Histological differences in the synovium between normal and RA patients are indicated in FIG. 1.


Like many other forms of arthritis, rheumatoid arthritis (RA) is initially characterized by an inflammatory response of the synovial membrane (‘synovitis’) that is characterised by an important influx of various types of mononuclear cells as well as by the activation of the local or infiltrated mononuclear cells. The lining layer becomes hyperplastic (it can have a thickness of >20 cells) and the synovial membrane expands. However, in addition, the hallmark of RA is joint destruction: the joint spaces narrow or disappear as a sign of cartilage degradation and destructions of the adjacent bone, also termed ‘erosions’, have occurred. The destructive portion of the synovial membrane is termed ‘pannus’. Enzymes secreted by synoviocytes lead to cartilage degradation.


This analysis shows that the main effector responsible for RA-associated joint degradation is the pannus, where the synovial fibroblast, by producing diverse proteolytic enzymes, is the prime driver of cartilage and bone erosion. In the advanced RA patient, the pannus mediates the degradation of the adjacent cartilage, leading to the narrowing of the joint space, and has the potential to invade adjacent bone and cartilage. As collagen type I or II are main components of bone and cartilage tissues, respectively, the pannus destructive and invasive properties are mediated by the secretion of collagenolytic proteases, principally the matrix metallo proteinases (MMPs). The erosion of the bone under and adjacent to the cartilage is also part of the RA process, and results principally from the presence of osteoclasts at the interface of bone and pannus. Osteoclasts adhere to the bone tissue and form a closed compartment, within which the osteoclasts secrete proteases (Cathepsin K, MMP9) that degrade the bone tissue. The osteoclast population in the joint is abnormally increased by osteoblast formation from precursor cells induced by the secretion of the receptor activator of NFkB ligand (RANKL) by activated SFs and T-cells.


Various collagen types have a key role in defining the stability of the extra-cellular matrix (ECM). Collagen type I and collagen type II, for example, are main components of bone and cartilage, respectively. Collagen proteins typically organize into multimeric structures referred to as collagen fibrils. Native collagen fibrils are very resistant to proteolytic cleavage. Only a few types of ECM-degrading proteins have been reported to have the capacity to degrade native collagen: matrix-metallo proteases (MMPs) and Cathepsins. Among the Cathepsins, cathepsin K, which is active mainly in osteoclasts, is the best characterised.


Matrix Metallo Proteases (MMPs) possess various physiological roles, for example, they are involved in the maturation of other proteases, growth factors, and the degradation of extra-cellular matrix components. Among the MMPs, MMP1, MMP2, MMP8 MMP13 and MMP14 are known to have collagenolytic properties. MMP1 is a member of the MMP family and is able to degrade native collagen, an important component of bone and cartilage. The correlation between an increased expression of MMP1 by synovial fibroblasts (SFs) and the progression of the arthritic disease is well-established and is predictive for joint erosive processes (Cunnane et al., 2001). In the context of RA, therefore, MMP1 represents a highly relevant collagen degrading protein. In vitro, the treatment of cultured SFs with cytokines relevant in the RA pathology (for example, TNF-α and IL1β) will increase the expression of MMP1 by these cells (Andreakos et al., 2003).


The activity of the ECM-degrading proteins can also be causative or correlate with the progression of various diseases other than RA, which diseases involve also the degradation of the joints. These diseases include, but are not limited to, psoriatic arthritis, juvenile arthritis, early arthritis, reactive arthritis, osteoarthritis, and ankylosing spondylitis. Other diseases that may be treatable with compounds identified according to the present invention and using the targets involved in the expression of MMPs as described herein are osteoporosis, muscular skeletal diseases like tendonitis and periodontal disease (Gapski et al., 2004), cancer metastasis (Coussens et al., 2002), airway diseases (COPD, asthma) (Suzuki et al., 2004), lung, renal fibrosis (Schanstra et al., 2002), liver fibrosis associated with chronic hepatitis C (Reiff et al., 2005), cardio-vascular diseases like atherosclerosis and heart failure (Creemers et al., 2001), and neurological diseases like neuro-inflammation and multiple sclerosis (Rosenberg, 2002). Patients suffering from such diseases may benefit from a therapy that protects the ECM from enzymatic degradation.


Reported Developments

NSAIDS (Non-steroidal anti-inflammatory drugs) are used to reduce the pain associated with RA and improve life quality of the patients. These drugs will not, however, put a brake on the RA-associated joint destruction.


Corticosteroids are found to decrease the progression of RA as detected radiographically and are used at low doses to treat part of the RA patients (30 to 60%). Serious side effects, however, are associated with long corticosteroid use (Skin thinning, osteoporosis, cataracts, hypertension, hyperlipidemia).


Synthetic DMARDs (Disease-Modifying Anti-Rheumatic Drugs) (for example, methotrexate, leflunomide, sulfasalazine) mainly tackle the immuno-inflammatory component of RA. As a main disadvantage, these drugs only have a limited efficacy (joint destruction is only slowed down but not blocked by DMARDs such that disease progression in the long term continues). The lack of efficacy is indicated by the fact that, on average, only 30% of the patients achieve an ACR50 score after 24 months treatment with methotrexate. This means that, according to the American College of Rheumatology, only 30% of the patients do achieve a 50% improvement of their symptoms (O'Dell et al., 1996). In addition, the precise mechanism of action of DMARDs is often unclear.


Biological DMARDs (Infliximab, Etanercept, Adalimumab, Rituximab, CTLA4-Ig) are therapeutic proteins that inactivate cytokines (for example, TNF-α) or cells (for example, T-cells or B-cells) that have an important role in the RA pathophysiology (Kremer et al., 2003; Edwards et al., 2004). Although the TNF-α-blockers (Infliximab, Etanercept, Adalimumab) and methotrexate combination therapy is the most effective RA treatment currently available, it is striking that even this therapy only achieves a 50% improvement (ACR50) in disease symptoms in 50-60% of patients after 12 months therapy (St Clair et al., 2004). Some adverse events warnings for anti-TNF-α drugs exist, shedding a light on the side effects associated to this type of drugs. Increased risk for infections (tuberculosis) hematologic events and demyelinating disorders have been described for the TNF-α blockers. (see also Gomez-Reino et al., 2003). Besides the serious side effects, the TNF-α blockers do also share the general disadvantages of the biological class of therapeutics, which are the unpleasant way of administration (frequent injections accompanied by infusion site reactions) and the high production cost. Newer agents in late development phase target T-cell co-stimulatory molecules and B-cells. The efficacy of these agents is expected to be similar to that of the TNF-a blockers. The fact that a variety of targeted therapies have similar but limited efficacies, suggests that there is a multiplicity of pathogenic factors for RA. This is also indicative for the deficiencies in our understanding of pathogenic events relevant to RA.


The current therapies for RA are not satisfactory due to a limited efficacy (no adequate therapy exists for 30% of the patients). This calls for additional strategies to achieve remission. Remission is required since residual disease bears the risk of progressive joint damage and thus progressive disability. Inhibiting the immuno-inflammatory component of the RA disease, which represents the main target of drugs currently used for RA treatment, does not result in a blockade of joint degradation, the major hallmark of the disease.


The histological analysis of RA patient joints clearly identifies the pannus, as an aggressive, invasive tissue that represents the main culprit in joint degradation. Within the pannus, the synovial fibroblasts represent a link between the initiation of the abnormally triggered immune system that lies at the basis of RA pathogenesis, and the ultimate joint erosion. As no current RA therapy efficiently abolishes the erosive activity of the pannus in the long term, the discovery of novel drugs and/or drug targets that inhibit the generation, and/or the activity, of the pannus would represent an important milestone for the development of novel RA treatments.


The present invention is based on the discovery of that certain proteins function in the pathway that results in the expression of extra-cellular matrix (ECM) degradation proteases, such as MMP1, and that inhibitors of the activity of these proteins, are useful for the treatment of diseases involving the abnormally high expression of such proteases.


SUMMARY OF THE INVENTION

The present invention relates to a method for identifying compounds that inhibit extra-cellular matrix (ECM) degradation, comprising contacting a compound with a polypeptide comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 17-32 (hereinafter “TARGETS”) and fragments thereof, under conditions that allow said polypeptide to bind to said compound, and measuring a compound-polypeptide property related to extra-cellular matrix (ECM) degradation.


Aspects of the present method include the in vitro assay of compounds using polypeptide of a TARGET, or fragments thereof, such fragments including the amino acid sequences described by SEQ ID NO: 33-127, and cellular assays wherein TARGET inhibition is followed by observing indicators of efficacy including, for example, TARGET expression levels, TARGET enzymatic activity and/or Matrix Metallo Proteinase-1 levels.


The present invention also relates to (1) expression inhibitory agents comprising a polynucleotide selected from the group of an antisense polynucleotide, a ribozyme, and a small interfering RNA (siRNA), wherein said polynucleotide comprises a nucleic acid sequence complementary to, or engineered from, a naturally occurring polynucleotide sequence encoding a polypeptide comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 17-127, and (2) pharmaceutical compositions comprising said agent, useful in the treatment, or prevention, of chronic joint degenerative diseases such as rheumatoid arthritis.


Another aspect of the invention is a method of treatment, or prevention, of a condition involving extra-cellular matrix (ECM) degradation, in a subject suffering or susceptible thereto, by administering a pharmaceutical composition comprising an effective TARGET-expression inhibiting amount of a expression-inhibitory agent or an effective TARGET activity inhibiting amount of a activity-inhibitory agent.


A further aspect of the present invention is a method for diagnosis relating to disease conditions characterized by extra-cellular matrix (ECM) degradation comprising measurement of indicators of levels of TARGET expression in a subject.


Another aspect of this invention relates to the use of the present compound in a therapeutic method, a pharmaceutical composition, and the manufacture of such composition, useful for the treatment of a disease involving inflammation, and in particular, a disease characteristic of abnormal matrix metallo protease activity.





BRIEF DESCRIPTION OF THE DRAWINGS


FIG. 1. Schematic view of a normal joint and its changes in rheumatoid arthritis (From Smolen and Steiner, 2003).



FIG. 2. Characterization of the expression of MMP1 by synovial fibroblasts. In panel A, the MMP1 mRNA levels present in the SF lysate are determined by real-time PCR. These MMP1 levels are normalized to the 18S levels that are also determined by real-time PCR for the same samples. Panel B shows the MMP1 signal detected from the supernatant that is subjected to Western blotting for detection of MMP1 protein levels using an MMP1-specific polyclonal antibody. Panel C shows the results of subjecting the supernatant to a commercially available MMP1 “activity ELISA” (Amersham Biosciences). The signal represented is proportional to the MMP1 activity present in the samples tested.



FIG. 3. Increased expression of MMP1 by SFs triggered with various model adenoviruses. The SF supernatant from uninfected SFs and SFs infected with the indicated model recombinant adenoviruses is subjected to the MMP1 ELISA and the MMP1 level measured by using a luminescence generating substrate is shown.



FIG. 4. Increased expression of MMP1 by SFs triggered with various cytokines relevant in rheumatoid arthritis pathology. The SF supernatant is subjected to the MMP1 ELISA and the MMP1 level measured by using a luminescence generating substrate is shown. The white bars show the MMP1 levels of the SF supernatant from untreated SFs and SFs treated with the indicated cytokine or combination of cytokines. The grey bars show the MMP1 levels of the SF supernatant treated with the supernatant of THP1 cells that were either untreated or treated with the indicated cytokine or combination of cytokines.



FIG. 5. Dose-dependent inhibition of the “TNFa-based trigger”-induced expression of MMP1 by SFs by a known anti-inflammatory compound. The SF supernatant from SFs treated with the “TNFα-based trigger” and the anti-inflammatory compound at various concentrations is subjected to the MMP1 ELISA and the percent inhibition of MMP1 expression versus the log concentration of the anti-inflammatory compound is shown.



FIG. 6. Schematic representation of the screening of the SilenceSelect collection using the “reverse MMP1 assay.”



FIG. 7. Layout and performance of the control plate produced for the reverse MMP1 ELISA assay.



FIG. 8. Representative example of the performance of the reverse MMP1 ELISA assay run on a subset of 384 Ad-siRNAs of the SilenceSelect collection that are tested in duplicate in a primary screen (A) and a repeat screen (B).



FIG. 9. All data points obtained in the screening of the SilenceSelect collection against the reverse MMP1 assay are shown. The averaged relative luminescence data obtained from the duplicate samples in the primary screen (Y-axis) is plotted against the averaged relative luminescence data for the corresponding Ad-siRNA obtained in the repeat screen (X-axis).



FIG. 10. Layout of the plates produced for the 3 MOI repeat screen of the reverse MMP1 assay (10A) and the cytotoxicity and secretion repeat screen of the reverse MMP1 assay (10B).



FIG. 11. Reduction of the expression of the TARGETS in primary SFs by Ad-siRNAs inhibits cytokine-induced MMP1 expression in a dose dependent manner. The supernatant from SFs infected with various doses of Ad-siRNAs targeting the expression of control genes (11A) and the genes listed in Table 1 (11B) is subjected to the MMP1 ELISA and the MMP1 level (11A) or MMP1 level relevant to the negative control (11B) is shown.



FIG. 12. Reduction of the expression of the TARGETS in primary SFs by Ad-siRNAs inhibits cytokine-induced native collagen degradation. The SF supernatant from “TNF□-based trigger”-treated SFs infected with the indicated Ad-siRNAs is subjected to the collagen degradation assay. FIG. 12A shows the percent collagen degraded as compared to the control. FIG. 12B shows the raw fluorescence signal.



FIG. 13. Reduction of the expression of the TARGETS in primary SFs by Ad-siRNAs modulates TNFα-induced IL-8 expression. The supernatant from SFs infected with the indicated Ad-siRNAs is subjected to the IL-8 ELISA assay and the percent inhibition of IL-8 expression relative to negative controls is shown.





DETAILED DESCRIPTION

The following terms are intended to have the meanings presented therewith below and are useful in understanding the description and intended scope of the present invention.


The term “agent” means any molecule, including polypeptides, polynucleotides and small molecules.


The term “agonist” refers to a ligand that stimulates the receptor the ligand binds to in the broadest sense.


The term “assay” means any process used to measure a specific property of a compound. A “screening assay” means a process used to characterize or select compounds based upon their activity from a collection of compounds.


The term “binding affinity” is a property that describes how strongly two or more compounds associate with each other in a non-covalent relationship. Binding affinities can be characterized qualitatively, (such as “strong”, “weak”, “high”, or “low”) or quantitatively (such as measuring the KD).


The term “carrier” means a non-toxic material used in the formulation of pharmaceutical compositions to provide a medium, bulk and/or useable form to a pharmaceutical composition. A carrier may comprise one or more of such materials such as an excipient, stabilizer, or an aqueous pH buffered solution. Examples of physiologically acceptable carriers include aqueous or solid buffer ingredients including phosphate, citrate, and other organic acids; antioxidants including ascorbic acid; low molecular weight (less than about 10 residues) polypeptide; proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino acids such as glycine, glutamine, asparagine, arginine or lysine; monosaccharides, disaccharides, and other carbohydrates including glucose, mannose, or dextrins; chelating agents such as EDTA; sugar alcohols such as mannitol or sorbitol; salt-forming counterions such as sodium; and/or nonionic surfactants such as TWEENT™, polyethylene glycol (PEG), and PLURONICS™.


The term “complex” means the entity created when two or more compounds bind to each other.


The term “compound” is used herein in the context of a “test compound” or a “drug candidate compound” described in connection with the assays of the present invention. As such, these compounds comprise organic or inorganic compounds, derived synthetically or from natural sources. The compounds include inorganic or organic compounds such as polynucleotides, lipids or hormone analogs that are characterized by relatively low molecular weights. Other biopolymeric organic test compounds include peptides comprising from about 2 to about 40 amino acids and larger polypeptides comprising from about 40 to about 500 amino acids, such as antibodies or antibody conjugates.


The term “condition” or “disease” means the overt presentation of symptoms (i.e., illness) or the manifestation of abnormal clinical indicators (for example, biochemical indicators). Alternatively, the term “disease” refers to a genetic or environmental risk of or propensity for developing such symptoms or abnormal clinical indicators.


The term “contact” or “contacting” means bringing at least two moieties together, whether in an in vitro system or an in vivo system.


The term “derivatives of a polypeptide” relates to those peptides, oligopeptides, polypeptides, proteins and enzymes that comprise a stretch of contiguous amino acid residues of the polypeptide and that retain the biological activity of the protein, for example, polypeptides that have amino acid mutations compared to the amino acid sequence of a naturally-occurring form of the polypeptide. A derivative may further comprise additional naturally occurring, altered, glycosylated, acylated or non-naturally occurring amino acid residues compared to the amino acid sequence of a naturally occurring form of the polypeptide. It may also contain one or more non-amino acid substituents compared to the amino acid sequence of a naturally occurring form of the polypeptide, for example a reporter molecule or other ligand, covalently or non-covalently bound to the amino acid sequence.


The term “derivatives of a polynucleotide” relates to DNA-molecules, RNA-molecules, and oligonucleotides that comprise a stretch or nucleic acid residues of the polynucleotide, for example, polynucleotides that may have nucleic acid mutations as compared to the nucleic acid sequence of a naturally occurring form of the polynucleotide. A derivative may further comprise nucleic acids with modified backbones such as PNA, polysiloxane, and 2′-O-(2-methoxy)ethyl-phosphorothioate, non-naturally occurring nucleic acid residues, or one or more nuclei acid substituents, such as methyl-, thio-, sulphate, benzoyl-, phenyl-, amino-, propyl-, chloro-, and methanocarbanucleosides, or a reporter molecule to facilitate its detection.


The terms “ECM-degrading protein” and “ECM-degrading activity” refer to a protein and activity, respectively, which are capable of degrading extra-cellular matrixes found in bone and cartilage.


The term “effective amount” or “therapeutically effective amount” means that amount of a compound or agent that will elicit the biological or medical response of a subject that is being sought by a medical doctor or other clinician.


The term “endogenous” shall mean a material that a mammal naturally produces. Endogenous in reference to the term “protease”, “kinase”, or G-Protein Coupled Receptor (“GPCR”) shall mean that which is naturally produced by a mammal (for example, and not limitation, a human). In contrast, the term non-endogenous in this context shall mean that which is not naturally produced by a mammal (for example, and not limitation, a human). Both terms can be utilized to describe both “in vivo” and “in vitro” systems. For example, and not a limitation, in a screening approach, the endogenous or non-endogenous TARGET may be in reference to an in vitro screening system. As a further example and not limitation, where the genome of a mammal has been manipulated to include a non-endogenous TARGET, screening of a candidate compound by means of an in vivo system is viable.


The term “expressible nucleic acid” means a nucleic acid coding for a proteinaceous molecule, an RNA molecule, or a DNA molecule.


The term “expression” comprises both endogenous expression and overexpression by transduction.


The term “expression inhibitory agent” means a polynucleotide designed to interfere selectively with the transcription, translation and/or expression of a specific polypeptide or protein normally expressed within a cell. More particularly, “expression inhibitory agent” comprises a DNA or RNA molecule that contains a nucleotide sequence identical to or complementary to at least about 17 sequential nucleotides within the polyribonucleotide sequence coding for a specific polypeptide or protein. Exemplary expression inhibitory molecules include ribozymes, double stranded siRNA molecules, self-complementary single-stranded siRNA molecules, genetic antisense constructs, and synthetic RNA antisense molecules with modified stabilized backbones.


The term “expressible nucleic acid” means a nucleic acid coding for a proteinaceous molecule, an RNA molecule, or a DNA molecule.


The term “fragment of a polynucleotide” relates to oligonucleotides that comprise a stretch of contiguous nucleic acid residues that exhibit substantially a similar, but not necessarily identical, activity as the complete sequence.


The term “fragment of a polypeptide” relates to peptides, oligopeptides, polypeptides, proteins and enzymes that comprise a stretch of contiguous amino acid residues, and exhibit substantially a similar, but not necessarily identical, functional activity as the complete sequence.


The term “hybridization” means any process by which a strand of nucleic acid binds with a complementary strand through base pairing. The term “hybridization complex” refers to a complex formed between two nucleic acid sequences by virtue of the formation of hydrogen bonds between complementary bases. A hybridization complex may be formed in solution (for example, C0t or R0t analysis) or formed between one nucleic acid sequence present in solution and another nucleic acid sequence immobilized on a solid support (for example, paper, membranes, filters, chips, pins or glass slides, or any other appropriate substrate to which cells or their nucleic acids have been fixed). The term “stringent conditions” refers to conditions that permit hybridization between polynucleotides and the claimed polynucleotides. Stringent conditions can be defined by salt concentration, the concentration of organic solvent, for example, formamide, temperature, and other conditions well known in the art. In particular, reducing the concentration of salt, increasing the concentration of formamide, or raising the hybridization temperature can increase stringency.


The term “inhibit” or “inhibiting”, in relationship to the term “response” means that a response is decreased or prevented in the presence of a compound as opposed to in the absence of the compound.


The term “inhibition” refers to the reduction, down regulation of a process or the elimination of a stimulus for a process that results in the absence or minimization of the expression of a protein or polypeptide.


The term “induction” refers to the inducing, up-regulation, or stimulation of a process that results in the expression of a protein or polypeptide.


The term “ligand” means an endogenous, naturally occurring molecule specific for an endogenous, naturally occurring receptor.


The term “pharmaceutically acceptable salts” refers to the non-toxic, inorganic and organic acid addition salts, and base addition salts, of compounds of the present invention. These salts can be prepared in situ during the final isolation and purification of compounds useful in the present invention.


The term “polypeptide” relates to proteins, proteinaceous molecules, fractions of proteins, peptides, oligopeptides, enzymes (such as kinases, proteases, GCPR's etc.).


The term “polynucleotide” means a polynucleic acid, in single or double stranded form, and in the sense or antisense orientation, complementary polynucleic acids that hybridize to a particular polynucleic acid under stringent conditions, and polynucleotides that are homologous in at least about 60 percent of its base pairs, and more preferably 70 percent of its base pairs are in common, most preferably 90 percent, and in a special embodiment 100 percent of its base pairs. The polynucleotides include polyribonucleic acids, polydeoxyribonucleic acids, and synthetic analogues thereof. It also includes nucleic acids with modified backbones such as peptide nucleic acid (PNA), polysiloxane, and 2′-O-(2-methoxy)ethylphosphorothioate. The polynucleotides are described by sequences that vary in length, that range from about 10 to about 5000 bases, preferably about 100 to about 4000 bases, more preferably about 250 to about 2500 bases. One polynucleotide embodiment comprises from about 10 to about 30 bases in length. A special embodiment of polynucleotide is the polyribonucleotide of from about 17 to about 22 nucleotides, more commonly described as small interfering RNAs (siRNAs). Another special embodiment are nucleic acids with modified backbones such as peptide nucleic acid (PNA), polysiloxane, and 2′-O-(2-methoxy)ethylphosphorothioate, or including non-naturally occurring nucleic acid residues, or one or more nucleic acid substituents, such as methyl-, thio-, sulphate, benzoyl-, phenyl-, amino-, propyl-, chloro-, and methanocarbanucleosides, or a reporter molecule to facilitate its detection.


The term “polypeptide” relates to proteins (such as TARGETS), proteinaceous molecules, fractions of proteins peptides and oligopeptides.


The term “solvate” means a physical association of a compound useful in this invention with one or more solvent molecules. This physical association includes hydrogen bonding. In certain instances the solvate will be capable of isolation, for example when one or more solvent molecules are incorporated in the crystal lattice of the crystalline solid. “Solvate” encompasses both solution-phase and isolable solvates. Representative solvates include hydrates, ethanolates and methanolates.


The term “subject” includes humans and other mammals.


The term “TARGET” or “TARGETS” means the protein(s) identified in accordance with the assays described herein and determined to be involved in the modulation of MMP1 expression levels.


“Therapeutically effective amount” means that amount of a drug or pharmaceutical agent that will elicit the biological or medical response of a subject that is being sought by a medical doctor or other clinician. In particular, with regard to treating an disease condition characterized by the degradation of extracellular matrix, the term “effective matrix metallo-protease inhibiting amount” is intended to mean that effective amount of an compound of the present invention that will bring about a biologically meaningful decrease in the production of MMP-1 in the subject's disease affected tissues such that extracellular matrix degradation is meaningfully reduced. A compound having matrix metallo-protease inhibiting properties or a “matrix metallo-protease inhibiting compound” means a compound that provided to a cell in effective amounts is able to cause a biologically meaningful decrease in the production of MMP-1 in such cells.


The term “treating” means an intervention performed with the intention of preventing the development or altering the pathology of, and thereby alleviating a disorder, disease or condition, including one or more symptoms of such disorder or condition. Accordingly, “treating” refers to both therapeutic treatment and prophylactic or preventative measures. Those in need of treating include those already with the disorder as well as those in which the disorder is to be prevented. The related term “treatment,” as used herein, refers to the act of treating a disorder, symptom, disease or condition, as the term “treating” is defined above.


Applicants' Invention Based on TARGET Relationship to Extra-Cellular Matrix Degradation


MMP1 expression by SFs is relevant to ECM degradation as it correlates to the activation of SFs towards an erosive phenotype that, in vivo, is responsible for cartilage degradation. This has been demonstrated by the administration to TNF□ transgenic mice, which spontaneously develop an arthritic phenotype, of adenovirus that overexpress TIMP1, an inhibitor of MMPs, which administration results in the reduction of joint degradation (Shett et al., Arthritis Rheum. (2001) 44:2888-98). Therefore, inhibition of the MMP1 expression by SFs represents a valuable therapeutic approach towards the treatment of RA. Accordingly, if the reduction in expression of a candidate protein in activated SFs leads to a corresponding reduction of MMP1 expression, then such protein is involved in the regulation of MMP1 expression and is a relevant target for the development of therapeutic strategies for the treatment of RA. The present inventors have identified such target proteins by screening recombinant adenoviruses mediating the expression of a library of shRNAs, referred to herein as “Ad-siRNAs.” The collection used herein is further referred to as “adenoviral siRNA library” or “SilenceSelect” collection. These libraries contain recombinant adenoviruses, further referred to as knock-down (KD) viruses or Ad-siRNAs, that mediate the expression in cells of shRNAs which reduce the expression levels of targeted genes by a RNA interference (RNAi)-based mechanism (WO03/020931). The screening work is described below in Example 1.


As noted above, the present invention is based on the present inventors' discovery that the TARGET polypeptides, identified as a result of a variety of screens described below in the Examples, are factors not only in the up-regulation and/or induction of MMP1, but even in the up-regulation and/or induction of extra-cellular matrix degradation. The activity of the ECM-degrading proteins is believed to be causative and to correlate with the progression of various diseases associated with an increased degradation of the extra-cellular matrix, including diseases that involve the degradation of the joint.


In one aspect, the present invention relates to a method for assaying for drug candidate compounds that inhibit extra-cellular matrix degradation, comprising contacting the compound with a polypeptide comprising an amino acid sequence of SEQ ID NO: 17-127, or fragment thereof, under conditions that allow said polypeptide to bind to the compound, and detecting the formation of a complex between the polypeptide and the compound. One preferred means of measuring the complex formation is to determine the binding affinity of said compound to said polypeptide.


More particularly, the invention relates to a method for identifying an agent that inhibits extra-cellular matrix degradation, the method comprising further:

    • (a) contacting a population of mammalian cells with one or more compound that exhibits binding affinity for a TARGET polypeptide, or fragment thereof, and
    • (b) measuring a compound-polypeptide property related to extra-cellular matrix degradation.


The compound-polypeptide property referred to above is related to the expression and/or activity of the TARGET, and is a measurable phenomenon chosen by the person of ordinary skill in the art. The measurable property may be, for example, the binding affinity for a peptide domain of the polypeptide TARGET such as for SEQ ID NO: 33-127, or the level of any one of a number of biochemical marker levels of extra-cellular matrix degradation. Extra-cellular matrix degradation can be measured, for example, by measuring the level of enzymes that are induced during the process, such as expression of a MMP and/or a Cathepsin polypeptide.


In a preferred embodiment of the invention, the TARGET polypeptide comprises an amino acid sequence selected from the group consisting of SEQ ID No: 17-32 as listed in Table 1.


















TABLE 1













SEQ
SEQ





Ref/SEQ
SEQ
REF/SEQ
SEQ

ID KD
ID KD


Hit
Gene

Accession
ID NO
Accession
ID No
Protein
Target
Target


No.
Name
Description
(DNA)
DNA
(Protein)
Protein
Class
19-Mer
21-Mer
























H46-030
KCNF1

Homo sapiens

NM_002236
1
NP_002227
17
Ion
128
231




potassium voltage-




Channel
176-182





gated channel,





219-220





subfamily F, member 1









H46-137
SLC9A8

Homo sapiens solute

NM_015266
2
NP_056081
18
Ion
129
232




carrier family 9




Channel
183-189





(sodium/hydrogen





229-230





exchanger), isoform 8









H46-160
ARAF1

Homo sapiens v-raf

NM_001654
3
NP_001645
19
Kinase
130
233




murine sarcoma 3611





156-162





viral oncogene





204





homolog









H46-010
AXL

Homo sapiens AXL

NM_001699
4
NP_001690
20
Kinase
131
234




receptor tyrosine





142-148





kinase (AXL),





205-206





transcript variant 2









H46-010
AXL

Homo sapiens AXL

NM_021913
5
NP_068713
21
Kinase
131
234




receptor tyrosine





205-206





kinase (AXL),











transcript variant 1









H46-393
FGFR3

Homo sapiens

NM_000142
6
NP_000133
22
Kinase
132
235




fibroblast growth





190-196





factor receptor 3,





211-215





transcript variant 1









H46-189
NR2F6

Homo sapiens nuclear

NM_005234
7
NP_005225
23
NHR
133
236




receptor subfamily 2,





163-168





group F, member 6





225-226



H46-020
SCN9A

Homo sapiens sodium

NM_002977
8
NP_002968
24
Ion
134
237




channel, voltage-




Channel
169-175





gated, type IX, alpha





227-228



H46-359
MAPK13

Homo sapiens

NM_002754
9
NP_002745
25
Kinase
135
238




mitogen-activated





149-155





protein kinase 13





223



H46-343
ILK

Homo sapiens

NM_004517
10
NP_004508
26
Kinase
136
239




integrin-linked kinase





216-218





(ILK), transcript











variant 1









H46-226
CHRNA5

Homo sapiens

M83712
11
AAA58357
27
Ion
137
240




cholinergic receptor,




Channel/
197-203





nicotinic, alpha




Receptor
207-208





polypeptide 5









H46-129
NQO2

Homo sapiens

NM_000904
12
NP_000895
28
Dehydro-
138-139
241-242




NAD(P)H




genase
224





dehydrogenase,











quinone 2









H46-021
DGKB

Homo sapiens

NM_145695
13
NP_663733
29
Kinase
140
243




diacylglycerol kinase,





209





beta 90 kDa (DGKB),











transcript variant 2









H46-021
DGKB

Homo sapiens

NM_004080
14
NP_004071
30
Kinase
140
243




diacylglycerol kinase,





209-210





beta 90 kDa (DGKB),











transcript variant 1









H46-021
INCENP

Homo sapiens inner

NM_020238
15
NP_064623
31
Not
140
243




centromere protein




Classified






antigens 135/155 kDa











(INCENP)









H46-265
MAPK12

Homo sapiens

NM_002969
16
NP_002960
32
Kinase
141
244




mitogen-activated





221-222





protein kinase 12









Another special embodiment of the invention comprises the kinase. TARGETS identified as SEQ ID NOS. 19-22, 25, 26, 29, 30, and 32. Another special embodiment of the invention comprises the ion channel TARGETS identified as SEQ ID NOS. 17, 18, 24, and 27. A further preferred embodiment is the Nuclear Hormone Receptors (NHRs) TARGET identified as SEQ. ID NO. 23.


Depending on the choice of the skilled artisan, the present assay method may be designed to function as a series of measurements, each of which is designed to determine whether the drug candidate compound is indeed acting on the polypeptide to thereby inhibit extra-cellular matrix degradation. For example, an assay designed to determine the binding affinity of a compound to the polypeptide, or fragment thereof, may be necessary, but not sufficient, to ascertain whether the test compound would be useful for inhibiting extra-cellular matrix degradation when administered to a subject.


Such binding information would be useful in identifying a set of test compounds for use in an assay that would measure a different property, further down the biochemical pathway, such as for example MMP-1 expression. Such second assay may be designed to confirm that the test compound, having binding affinity for the polypeptide, actually inhibits extra-cellular matrix degradation. Suitable controls should always be in place to insure against false positive readings.


The order of taking these measurements is not believed to be critical to the practice of the present invention, which may be practiced in any order. For example, one may first perform a screening assay of a set of compounds for which no information is known respecting the compounds' binding affinity for the polypeptide. Alternatively, one may screen a set of compounds identified as having binding affinity for a polypeptide domain, or a class of compounds identified as being an inhibitor of the polypeptide. However, for the present assay to be meaningful to the ultimate use of the drug candidate compounds, a measurement of extra-cellular matrix degradation activity is necessary. Validation studies including controls and measurements of binding affinity to the polypeptides of the invention are nonetheless useful in identifying a compound useful in any therapeutic or diagnostic application.


The present assay method may be practiced in vitro, using one or more of the TARGET proteins, or fragments thereof. The amino acid sequences of exemplary protein domain fragments of selected TARGETS are SEQ ID NO: 33-127, listed in Table 1A below.












TABLE 1A








SEQ ID NO


Accession
Name
Protein Segment
Protein segment


















NM_002236
KCNF1
Intracellular domain
33


NM_002236
KCNF1
Transmembrane domain
34


NM_002236
KCNF1
Extracellular domain
35


NM_002236
KCNF1
Transmembrane domain
36


NM_002236
KCNF1
Intracellular domain
37


NM_002236
KCNF1
Transmembrane domain
38


NM_002236
KCNF1
Extracellular domain
39


NM_002236
KCNF1
Transmembrane domain
40


NM_002236
KCNF1
Intracellular domain
41


NM_015266
SLC9A8
Intracellular domain
42


NM_015266
SLC9A8
Transmembrane domain
43


NM_015266
SLC9A8
Extracellular domain
44


NM_015266
SLC9A8
Transmembrane domain
45


NM_015266
SLC9A8
Intracellular domain
46


NM_015266
SLC9A8
Transmembrane domain
47


NM_015266
SLC9A8
Extracellular domain
48


NM_015266
SLC9A8
Transmembrane domain
49


NM_015266
SLC9A8
Intracellular domain
50


NM_015266
SLC9A8
Transmembrane domain
51


NM_015266
SLC9A8
Extracellular domain
52


NM_015266
SLC9A8
Transmembrane domain
53


NM_015266
SLC9A8
Intracellular domain
54


NM_015266
SLC9A8
Transmembrane domain
55


NM_015266
SLC9A8
Extracellular domain
56


NM_015266
SLC9A8
Transmembrane, domain
57


NM_015266
SLC9A8
Intracellular domain
58


NM_015266
SLC9A8
Transmembrane domain
59


NM_015266
SLC9A8
Extracellular domain
60


NM_015266
SLC9A8
Transmembrane domain
61


NM_015266
SLC9A8
Intracellular domain
62


NM_015266
SLC9A8
Transmembrane domain
63


NM_015266
SLC9A8
Extracellular domain
64


NM_001699
AXL
Extracellular domain
65


NM_001699
AXL
Transmembrane domain
66


NM_001699
AXL
Intracellular domain
67


NM_021913
AXL
Extracellular domain
68


NM_021913
AXL
Transmembrane domain
69


NM_021913
AXL
Intracellular domain
70


NM_000142
FGFR3
Extracellular domain
71


NM_000142
FGFR3
Transmembrane domain
72


NM_000142
FGFR3
Intracellular domain
73


NM_000142
FGFR3
Transmembrane domain
74


NM_000142
FGFR3
Extracellular domain
75


NM_002977
SCN9A
Intracellular domain
76


NM_002977
SCN9A
Transmembrane domain
77


NM_002977
SCN9A
Extracellular domain
78


NM_002977
SCN9A
Transmembrane domain
79


NM_002977
SCN9A
Intracellular domain
80


NM_002977
SCN9A
Transmembrane domain
81


NM_002977
SCN9A
Extracellular domain
82


NM_002977
SCN9A
Transmembrane domain
83


NM_002977
SCN9A
Intracellular domain
84


NM_002977
SCN9A
Transmembrane domain
85


NM_002977
SCN9A
Extracellular domain
86


NM_002977
SCN9A
Transmembrane domain
87


NM_002977
SCN9A
Intracellular domain
88


NM_002977
SCN9A
Transmembrane domain
89


NM_002977
SCN9A
Extracellular domain
90


NM_002977
SCN9A
Transmembrane domain
91


NM_002977
SCN9A
Intracellular domain
92


NM_002977
SCN9A
Transmembrane domain
93


NM_002977
SCN9A
Extracellular domain
94


NM_002977
SCN9A
Transmembrane domain
95


NM_002977
SCN9A
Intracellular domain
96


NM_002977
SCN9A
Transmembrane domain
97


NM_002977
SCN9A
Extracellular domain
98


NM_002977
SCN9A
Transmembrane domain
99


NM_002977
SCN9A
Intracellular domain
100


NM_002977
SCN9A
Transmembrane domain
101


NM_002977
SCN9A
Extracellular domain
102


NM_002977
SCN9A
Transmembrane domain
103


NM_002977
SCN9A
Intracellular domain
104


NM_002977
SCN9A
Transmembrane domain
105


NM_002977
SCN9A
Extracellular domain
106


NM_002977
SCN9A
Transmembrane domain
107


NM_002977
SCN9A
Intracellular domain
108


NM_002977
SCN9A
Transmembrane domain
109


NM_002977
SCN9A
Extracellular domain
110


NM_002977
SCN9A
Transmembrane domain
111


NM_002977
SCN9A
Intracellular domain
112


NM_002977
SCN9A
Transmembrane domain
113


NM_002977
SCN9A
Extracellular domain
114


NM_002977
SCN9A
Transmembrane domain
115


NM_002977
SCN9A
Intracellular domain
116


NM_002977
SCN9A
Transmembrane domain
117


NM_002977
SCN9A
Extracellular domain
118


M83712
CHRNA5
Extracellular domain
119


M83712
CHRNA5
Transmembrane domain
120


M83712
CHRNA5
Intracellular domain
121


M83712
CHRNA5
Transmembrane domain
122


M83712
CHRNA5
Extracellular domain
123


M83712
CHRNA5
Transmembrane domain
124


M83712
CHRNA5
Intracellular domain
125


M83712
CHRNA5
Transmembrane domain
126


M83712
CHRNA5
Extracellular domain
127









The binding affinity of a compound with the polypeptide TARGET can be measured by methods known in the art, such as using surface plasmon resonance biosensors (Biacore), by saturation binding analysis with a labeled compound (for example, Scatchard and Lindmo analysis), by differential UV spectrophotometer, fluorescence polarization assay, Fluorometric Imaging Plate Reader)(FLIPR®) system, Fluorescence resonance energy transfer, and Bioluminescence resonance energy transfer. The binding affinity of compounds can also be expressed in dissociation constant (Kd) or as IC50 or EC50. The IC50 represents the concentration of a compound that is required for 50% inhibition of binding of another ligand to the polypeptide. The EC50 represents the concentration required for obtaining 50% of the maximum effect in any assay that measures TARGET function. The dissociation constant, Kd, is a measure of how well a ligand binds to the polypeptide, it is equivalent to the ligand concentration required to saturate exactly half of the binding-sites on the polypeptide. Compounds with a high affinity binding have low Kd, IC50 and EC50 values, for example, in the range of 100 nM to 1 μM; a moderate- to low-affinity binding relates to high Kd, IC50 and EC50 values, for example in the micromolar range.


The present assay method may also be practiced in a cellular assay, A host cell expressing the TARGET can be a cell with endogenous expression or a cell over-expressing the TARGET, for example, by transduction. When the endogenous expression of the polypeptide is not sufficient to determine a baseline that can easily be measured, one may use using host cells that over-express TARGET. Over-expression has the advantage that the level of the TARGET substrate end-products is higher than the activity level by endogenous expression. Accordingly, measuring such levels using presently available techniques is easier.


One embodiment of the present method for identifying a compound that decreases extra-cellular matrix (ECM) degradation comprises culturing a population of mammalian cells expressing a TARGET polypeptide, or a functional fragment or derivative thereof; determining a first: level of ECM degradation in said population of cells; eventually activating the population of cells; exposing said population of cells to a compound, or a mixture of compounds; determining a second level of ECM degradation in said population of cells during or after exposure of said population of cells to said compound, or the mixture of said compounds; and identifying the compound(s) that decreases ECM degradation. As noted above, ECM degradation may be determined by measuring the expression and/or activity of the TARGET polypeptide and/or a known ECM-degrading protein. In a preferred embodiment, said ECM-degrading protein is able to degrade collagen, and more preferably, is able to degrade collagen type I and/or collagen type II. In another preferred embodiment of the present invention, said ECM-degrading protein is a Matrix Metallo Proteinase (MMP), and more preferably is selected from the group consisting of MMP1, MMP2, MMP3, MMP8, MMP9, MMP13 and MMP14. In this context, the most preferred ECM-degrading protein is Matrix Metalloprotease 1 (MMP1). In yet another preferred embodiment, said ECM-degrading protein is Cathepsin K.


The expression of an ECM-degrading protein can be determined by methods known in the art such as Western blotting using specific antibodies, or an ELISA using antibodies specifically recognizing a particular ECM-degrading protein.


The activity of an ECM-degrading protein can be determined by using fluorogenic small peptide substrates. The specificity of these substrates, however, is often limited. In general, the use of these substrates is limited to the testing of purified proteases in biochemical assays, to avoid interference of other proteases.


The present inventors have developed a protocol allowing the detection, in a high throughput mode, of the activity of collagen degrading enzymes in complex media such as the supernatant of cultured cells. This protocol makes use of native collagen, being labelled with a fluorescent label, as a substrate.


The present inventors identified TARGET genes involved in ECM-degradation by using a ‘knock-down’ library. This type of library is a screen in which siRNA molecules are transduced into cells by recombinant adenoviruses, which siRNA molecules inhibit or repress the expression of a specific gene as well as expression and activity of the corresponding gene product in a cell. Each siRNA in a viral vector corresponds to a specific natural gene. By identifying a siRNA that represses ECM-degradation, a direct correlation can be drawn between the specific gene expression and ECM degradation. The TARGET genes identified using the knock-down library (the protein expression products thereof herein referred to as “TARGET” polypeptides) are then used in the present inventive method for identifying compounds that can be used to prevent ECM-degradation. Indeed, shRNA compounds comprising the sequences listed in Table 2 (SEQ ID NO: 128-141 and 231-244) inhibit the expression and/or activity of these TARGET genes and decrease the ECM-degrading activity of cells, confirming the role of the TARGETS in ECM-degradation.









TABLE 2







List of target sequences selected within the coding sequences of the genes identified as modulators of the


collagenolytic activity of SFs for use in RNAi-based down-regulation of the expression of these genes.












GENE



SEQ ID NO
SEQ ID NO


NAME
ACCESSION
PLASMID NAME
siRNA NAME
19-mer
21-mer





KCNF1
NM_002236
A150100-KCNF1_v2
NM_002236_idx1263
128
231


SLC9A8
NM_015266
A150100-KIAA0939_v4
XM_030524_idx840
129
232


ARAF1
NM_001654
A150100-ARAF1_v1
NM_001654_idx372
130
233


AXL
NM_001699
A150100-AXL_v1
NM_021913_idx2487
131
234


AXL
NM_021913
A150100-AXL_v1
NM_021913_idx2487
131
234


FGFR3
NM_000142
A150100-FGFR3_v2
oKD173
132
235


NR2F6
NM_005234
A150100-NR2F6_v1
NM_005234_idx1269
133
236


SCN9A
NM_002977
A150100-SCN9A_v1
NM_002977_idx3381
134
237


MAPK13
NM_002754
A150100-MAPK13_v6
oKD090
135
238


ILK
NM_004517
A150100-ILK_v2
NM_004517_idx1243
136
239


CHRNA5
M83712
A150100-CHRNA5_v3
NM_000745_idx511
137
240


NQO2
NM_000904
A150100-NQO2_v1
NM_000904_idx598
138-139
241-242


DGKB
NM_145695
A150100-DGKB_v2
NM_004080_idx1064
140
243


DGKB
NM_004080
A150100-DGKB_v2
NM_004080_idx1064
140
243


INCENP
NM_020238
A150100-DGKB_v2
NM_004080_idx1064
140
243


MAPK12
NM_002969
A150100-MAPK12_v6
NM_002969_idx797
141
244









Specific methods to determine the activity of a kinase by measuring the phosphorylation of a substrate by the kinase, which measurements are performed in the presence or absence of a compound, are well known in the art.


Ion channels are membrane protein complexes and their function is to facilitate the diffusion of ions across biological membranes. Membranes, or phospholipid bilayers, build a hydrophobic, low dielectric barrier to hydrophilic and charged molecules. Ion channels provide a high conducting, hydrophilic pathway across the hydrophobic interior of the membrane. The activity of an ion channel can be measured using classical patch clamping. High-throughput fluorescence-based or tracer-based assays are also widely available to measure ion channel activity. These fluorescent-based assays screen compounds on the basis of their ability to either open or close an ion channel thereby changing the concentration of specific fluorescent dyes across a membrane. In the case of the tracer-based assay, the changes in concentration of the tracer within and outside the cell are measured by radioactivity measurement or gas absorption spectrometry.


Nuclear receptor activation is believed to involve a conformational change of the receptor that is induced by ligand binding. The results of protease protection assays have confirmed that nuclear hormone agonists and antagonists cause receptor proteins to adopt different conformations (Keidel et al. Mol. Cell. Biol. 14:287 (1994); Allan et al. J. Biol. Chem. 267:19513 (1992)). Accordingly, a protease protection assay can be used to measure the activity of a NHR. Recruitment of co-activators or repressors is another basis on which assays for assessment of NHR activity are developed.


Specific methods to determine the activity of a dehydrogenase by measuring the oxidation of a substrate by the dehydrogenase, which measurements are performed in the presence or absence of a compound, are well known in the art.


Specific methods to determine the inhibition by the compound by measuring the cleavage of the substrate by the polypeptide, which is a protease, are well known in the art. Classically, substrates are used in which a fluorescent group is linked to a quencher through a peptide sequence that is a substrate that can be cleaved by the target protease. Cleavage of the linker separates the fluorescent group and quencher, giving rise to an increase in fluorescence.


Table 4 lists a large number of other polypeptide types identified using applicant's knock-down library against the MMP1 assay described below. One such polypeptide class are the G-protein coupled receptors (GPCR), which are capable of activating an effector protein, resulting in changes in second messenger levels in the cell. The activity of a GPCR can be measured by measuring the activity level of such second messengers. Two important and useful second messengers in the cell are cyclic AMP (cAMP) and Ca2+. The activity levels can be measured by methods known to persons skilled in the art, either directly by ELISA or radioactive technologies or by using substrates that generate a fluorescent or luminescent signal when contacted with Ca2+ or indirectly by reporter gene analysis. The activity level of the one or more secondary messengers may typically be determined with a reporter gene controlled by a promoter, wherein the promoter is responsive to the second messenger. Promoters known and used in the art for such purposes are the cyclic-AMP responsive promoter that is responsive for the cyclic-AMP levels in the cell, and the NF-AT responsive promoter that is sensitive to cytoplasmic Ca2+-levels in the cell. The reporter gene typically has a gene product that is easily detectable. The reporter gene can either be stably infected or transiently transfected in the host cell. Useful reporter genes are alkaline phosphatase, enhanced green fluorescent protein, destabilized green fluorescent protein, luciferase and □-galactosidase.


It should be understood that the cells expressing the polypeptides, may be cells naturally expressing the polypeptides, or the cells may be may be transfected to express the polypeptides, as described above.


In one embodiment it is preferred that the methods of the present invention further comprise the step of contacting the population of cells with an agonist of the polypeptide. This is useful in methods wherein the expression of the polypeptide in a certain chosen population of cells is too low for a proper detection of its activity. By using an agonist the polypeptide may be triggered, enabling a proper read-out if the compound inhibits the polypeptide. Similar considerations apply to the measurement of ECM degradation. In a preferred embodiment, the cells used in the present method are mammalian synovial fibroblasts and the triggers that may be used to induce the ECM-degrading activity are cytokines relevant in the field of arthritis: for instance TNFalpha, IL1 beta, IL6, OSM, IL17, IL15, IL18 and MIF1 alpha. In another preferred embodiment, the trigger is a mixture of factors generated by contacting cytokine-producing cells relevant in the field of arthritis, such as monocytes, macrophages, T-cells, and B-cells. The cytokine-producing cells will respond to the contact by producing a complex and unbiased mixture of factors. If the cytokine-producing cell used is also found in the pannus, and the cytokine applied to this trigger is found in the synovial fluid of rheumatoid arthritis patients, the mixture of factors ultimately produced will contain factors that are present in the joints of arthritis patients.


The present invention further relates to a method for identifying a compound that inhibits extra-cellular matrix degradation, comprising:

    • (a) contacting a compound with a polypeptide comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 17-127;
    • (b) determining the binding affinity of the compound to the polypeptide;
    • (c) contacting a population of mammalian cells expressing said polypeptide with the compound that exhibits a binding affinity of at least 10 micromolar; and
    • (d) identifying the compound that inhibits extra-cellular matrix degradation.


The population of cells may be exposed to the compound or the mixture of compounds through different means, for instance by direct incubation in the medium, or by nucleic acid transfer into the cells. Such transfer may be achieved by a wide variety of means, for instance by direct transfection of naked isolated DNA, or RNA, or by means of delivery systems, such as recombinant vectors. Other delivery means such as liposomes, or other lipid-based vectors may also be used. Preferably, the nucleic acid compound is delivered by means of a (recombinant) vector such as a recombinant virus.


For high-throughput purposes, libraries of compounds may be used such as antibody fragment libraries, peptide phage display libraries, peptide libraries (for example, LOPAP™, Sigma Aldrich), lipid libraries (BioMol), synthetic compound libraries (for example, LOPAC™, Sigma Aldrich) or natural compound libraries (Specs, TimTec).


Preferred drug candidate compounds are low molecular weight compounds. Low molecular weight compounds, for example, with a molecular weight of 500 Dalton or less, are likely to have good absorption and permeation in biological systems and are consequently more likely to be successful drug candidates than compounds with a molecular weight above 500 Dalton (Lipinski et al. (1997)). Peptides comprise another preferred class of drug candidate compounds. Peptides may be excellent drug candidates and there are multiple examples of commercially valuable peptides such as fertility hormones and platelet aggregation inhibitors. Natural compounds are another preferred class of drug candidate compound. Such compounds are found in and extracted from natural sources, and which may thereafter be synthesized. The lipids are another preferred class of drug candidate compound.


Another preferred class of drug candidate compounds is an antibody. The present invention also provides antibodies directed against a TARGET. These antibodies may be endogenously produced to bind to the TARGET within the cell, or added to the tissue to bind to TARGET polypeptide present outside the cell. These antibodies may be monoclonal antibodies or polyclonal antibodies. The present invention includes chimeric, single chain, and humanized antibodies, as well as FAb fragments and the products of a FAb expression library, and Fv fragments and the products of an Fv expression library. In another embodiment, the compound may be a nanobody, the smallest functional fragment of naturally occurring single-domain antibodies (Cortez-Retamozo et al. 2004).


In certain embodiments, polyclonal antibodies may be used in the practice of the invention. The skilled artisan knows methods of preparing polyclonal antibodies. Polyclonal antibodies can be raised in a mammal, for example, by one or more injections of an immunizing agent and, if desired, an adjuvant. Typically, the immunizing agent and/or adjuvant will be injected in the mammal by multiple subcutaneous or intraperitoneal injections. Antibodies may also be generated against the intact TARGET protein or polypeptide, or against a fragment, derivatives including conjugates, or other epitope of the TARGET protein or polypeptide, such as the TARGET embedded in a cellular membrane, or a library of antibody variable regions, such as a phage display library.


It may be useful to conjugate the immunizing agent to a protein known to be immunogenic in the mammal being immunized. Examples of such immunogenic proteins include but are not limited to keyhole limpet hemocyanin, serum albumin, bovine thyroglobulin, and soybean trypsin inhibitor. Examples of adjuvants that may be employed include Freund's complete adjuvant and MPL-TDM adjuvant (monophosphoryl Lipid A, synthetic trehalose dicorynomycolate). One skilled in the art without undue experimentation may select the immunization protocol.


In some embodiments, the antibodies may be monoclonal antibodies. Monoclonal antibodies may be prepared using methods known in the art. The monoclonal antibodies of the present invention may be “humanized” to prevent the host from mounting an immune response to the antibodies. A “humanized antibody” is one in which the complementarity determining regions (CDRs) and/or other portions of the light and/or heavy variable domain framework are derived from a non-human immunoglobulin, but the remaining portions of the molecule are derived from one or more human immunoglobulins. Humanized antibodies also include antibodies characterized by a humanized heavy chain associated with a donor or acceptor unmodified light chain or a chimeric light chain, or vice versa. The humanization of antibodies may be accomplished by methods known in the art (see, for example, Mark and Padlan, (1994) “Chapter 4. Humanization of Monoclonal Antibodies”, The Handbook of Experimental Pharmacology Vol. 113, Springer-Verlag, New York). Transgenic animals may be used to express humanized antibodies.


Human antibodies can also be produced using various techniques known in the art, including phage display libraries (Hoogenboom and Winter, (1991) J. Mol. Biol. 227:381-8; Marks et al. (1991). J. Mol. Biol. 222:581-97). The techniques of Cole, et al. and Boerner, et al. are also available for the preparation of human monoclonal antibodies (Cole, et al. (1985) Monoclonal Antibodies and Cancer Therapy, Alan R. Liss, p. 77; Boerner, et al (1991). J. Immunol., 147(1):86-95).


Techniques known in the art for the production of single chain antibodies can be adapted to produce single chain antibodies to the TARGET polypeptides and proteins of the present invention. The antibodies may be monovalent antibodies. Methods for preparing monovalent antibodies are well known in the art. For example, one method involves recombinant expression of immunoglobulin light chain and modified heavy chain. The heavy chain is truncated generally at any point in the Fc region so as to prevent heavy chain cross-linking. Alternatively, the relevant cysteine residues are substituted with another amino acid residue or are deleted so as to prevent cross-linking.


Bispecific antibodies are monoclonal, preferably human or humanized, antibodies that have binding specificities for at least two different antigens and preferably for a cell-surface protein or receptor or receptor subunit. In the present case, one of the binding specificities is for one domain of the TARGET, while the other one is for another domain of the same or different TARGET.


Methods for making bispecific antibodies are known in the art. Traditionally, the recombinant production of bispecific antibodies is based on the co-expression of two immunoglobulin heavy-chain/light-chain pairs, where the two heavy chains have different specificities (Milstein and Cuello, (1983) Nature 305:537-9). Because of the random assortment of immunoglobulin heavy and light chains, these hybridomas (quadromas) produce a potential mixture of ten different antibody molecules, of which only one has the correct bispecific structure. Affinity chromatography steps usually accomplish the purification of the correct molecule. Similar procedures are disclosed in Trauneeker, et al. (1991) EMBO J. 10:3655-9.


According to another preferred embodiment, the assay method uses a drug candidate compound identified as having a binding affinity for a TARGET, and/or has already been identified as having down-regulating activity such as antagonist activity vis-à-vis one or more TARGET.


The present invention further relates to a method for inhibiting extra-cellular matrix degradation comprising contacting mammalian cells with an expression inhibitory agent comprising a polyribonucleotide sequence that complements at least about 17 to about 30 contiguous nucleotides of the nucleotide sequence selected from the group consisting of SEQ ID NO: 1-16.


Another aspect of the present invention relates to a method for inhibiting extra-cellular matrix degradation, comprising by contacting mammalian cells with an expression-inhibiting agent that inhibits the translation in the cell of a polyribonucleotide encoding a TARGET polypeptide. A particular embodiment relates to a composition comprising a polynucleotide including at, least one antisense strand that functions to pair the agent with the TARGET mRNA, and thereby down-regulate or block the expression of TARGET polypeptide. The inhibitory agent preferably comprises antisense polynucleotide, a ribozyme, and a small interfering RNA (siRNA), wherein said agent comprises a nucleic acid sequence complementary to, or engineered from, a naturally-occurring polynucleotide sequence selected from the group consisting of SEQ ID NO: 1-16.


A special embodiment of the present invention relates to a method wherein the expression-inhibiting agent is selected from the group consisting of antisense RNA, antisense oligodeoxynucleotide (ODN), a ribozyme that cleaves the polyribonucleotide coding for SEQ ID NO: 1-16, a small interfering RNA (siRNA, preferably shRNA,) that is sufficiently homologous to a portion of the polyribonucleotide corresponding to SEQ ID NO: 1-16, such that the siRNA, preferably shRNA, interferes with the translation of the TARGET polyribonucleotide to the TARGET polypeptide.


Another embodiment of the present invention relates to a method wherein the expression-inhibiting agent is a nucleic acid expressing the antisense RNA, antisense oligodeoxynucleotide (ODN), a ribozyme that cleaves the polyribonucleotide encoded by SEQ ID NO: 1-16, a small interfering RNA (siRNA, preferably shRNA,) that is sufficiently complementary to a portion of the polyribonucleotide corresponding to SEQ ID NO: 1-16, such that the siRNA, preferably shRNA, interferes with the translation of the TARGET polyribonucleotide to the TARGET polypeptide. Preferably the expression-inhibiting agent is an antisense RNA, ribozyme, antisense oligodeoxynucleotide, or siRNA, preferably shRNA, comprising a polyribonucleotide sequence that complements at least about 17 to about 30 contiguous nucleotides of a nucleotide sequence selected from the group consisting of SEQ. ID NO: 1-16. More preferably, the expression-inhibiting agent is an antisense RNA, ribozyme, antisense oligodeoxynucleotide, or siRNA, preferably shRNA, comprising a polyribonucleotide sequence that complements at least about 17 to about 25 contiguous nucleotides of a nucleotide sequence selected from the group consisting of SEQ ID NO: 1-16. A special embodiment comprises a polyribonucleotide sequence that complements a polynucleotide sequence selected from the group consisting of SEQ ID NO: 128-141 and 231-244.


The down regulation of gene expression using antisense nucleic acids can be achieved at the translational or transcriptional level. Antisense nucleic acids of the invention are preferably nucleic acid fragments capable of specifically hybridizing with all or part of a nucleic acid encoding a TARGET polypeptide or the corresponding messenger RNA. In addition, antisense nucleic acids may be designed which decrease expression of the nucleic acid sequence capable of encoding a TARGET polypeptide by inhibiting splicing of its primary transcript. Any length of antisense sequence is suitable for practice of the invention so long as it is capable of down-regulating or blocking expression of a nucleic acid coding for a TARGET. Preferably, the antisense sequence is at least about 17 nucleotides in length. The preparation and use of antisense nucleic acids, DNA encoding antisense RNAs and the use of oligo and genetic antisense is known in the art.


One embodiment of expression-inhibitory agent is a nucleic acid that is antisense to a nucleic acid comprising SEQ ID NO: 1-16, for example, an antisense nucleic acid (for example, DNA) may be introduced into cells in vitro, or administered to a subject in vivo, as gene therapy to inhibit cellular expression of nucleic acids comprising SEQ ID NO: 1-16. Antisense oligonucleotides comprise preferably a sequence containing from about 17 to about 100 nucleotides and more preferably the antisense oligonucleotides comprise from about 18 to about 30 nucleotides. Antisense nucleic acids may be prepared from about 17 to about 30 contiguous nucleotides selected from the sequences of SEQ ID NO: 1-16, expressed in the opposite orientation.


The antisense nucleic acids are preferably oligonucleotides and may consist entirely of deoxyribo-nucleotides, modified deoxyribonucleotides, or some combination of both. The antisense nucleic acids can be synthetic oligonucleotides. The oligonucleotides may be chemically modified, if desired, to improve stability and/or selectivity. Since oligonucleotides are susceptible to degradation by intracellular nucleases, the modifications can include, for example, the use of a sulfur group to replace the free oxygen of the phosphodiester bond. This modification is called a phosphorothioate linkage. Phosphorothioate antisense oligonucleotides are water soluble, polyanionic, and resistant to endogenous nucleases. In addition, when a phosphorothioate antisense oligonucleotide hybridizes to its TARGET site, the RNA-DNA duplex activates the endogenous enzyme ribonuclease (RNase) H, which cleaves the mRNA component of the hybrid molecule.


In addition, antisense oligonucleotides with phosphoramidite and polyamide (peptide) linkages can be synthesized. These molecules should be very resistant to nuclease degradation. Furthermore, chemical groups can be added to the 2′ carbon of the sugar moiety and the 5 carbon (C-5) of pyrimidines to enhance stability and facilitate the binding of the antisense oligonucleotide to its TARGET site. Modifications may include 2′-deoxy, O-pentoxy, O-propoxy, O-methoxy, fluoro, methoxyethoxy phosphorothioates, modified bases, as well as other modifications known to those of skill in the art.


Another type of expression-inhibitory agent that reduces the levels of TARGETS is the ribozyme. Ribozymes are catalytic RNA molecules (RNA enzymes) that have separate catalytic and substrate binding domains. The substrate binding sequence combines by nucleotide complementarity and, possibly, non-hydrogen bond interactions with its TARGET sequence. The catalytic portion cleaves the TARGET RNA at a specific site. The substrate domain of a ribozyme can be engineered to direct it to a specified mRNA sequence. The ribozyme recognizes and then binds a TARGET mRNA through complementary base pairing. Once it is bound to the correct TARGET site, the ribozyme acts enzymatically to cut the TARGET mRNA. Cleavage of the mRNA by a ribozyme destroys its ability to direct synthesis of the corresponding polypeptide. Once the ribozyme has cleaved its TARGET sequence, it is released and can repeatedly bind and cleave at other mRNAs.


Ribozyme forms include a hammerhead motif, a hairpin motif, a hepatitis delta virus, group I intron or RNaseP RNA (in association with an RNA guide sequence) motif or Neurospora VS RNA motif Ribozymes possessing a hammerhead or hairpin structure are readily prepared since these catalytic RNA molecules can be expressed within cells from eukaryotic promoters (Chen, et al. (1992) Nucleic Acids Res. 20:4581-9). A ribozyme of the present invention can be expressed in eukaryotic cells from the appropriate DNA vector. If desired, the activity of the ribozyme may be augmented by its release from the primary transcript by a second ribozyme (Ventura, et al. (1993) Nucleic Acids Res. 21:3249-55).


Ribozymes may be chemically synthesized by combining an oligodeoxyribonucleotide with a ribozyme catalytic domain (20 nucleotides) flanked by sequences that hybridize to the TARGET mRNA after transcription. The oligodeoxyribonucleotide is amplified by using the substrate binding sequences as primers. The amplification product is cloned into a eukaryotic expression vector.


Ribozymes are expressed from transcription units inserted into DNA, RNA, or viral vectors. Transcription of the ribozyme sequences are driven from a promoter for eukaryotic RNA polymerase I (pol (I), RNA polymerase II (pol II), or RNA polymerase III (pol III). Transcripts from pol II or pol III promoters will be expressed at high levels in all cells; the levels of a given pol II promoter in a given cell type will depend on nearby gene regulatory sequences. Prokaryotic RNA polymerase promoters are also used, providing that the prokaryotic RNA polymerase enzyme is expressed in the appropriate cells (Gao and Huang, (1993) Nucleic Acids Res. 21:2867-72). It has been demonstrated that ribozymes expressed from these promoters can function in mammalian cells (Kashani-Sabet, et al. (1992) Antisense Res. Dev. 2:3-15).


A particularly preferred inhibitory agent is a small interfering RNA (siRNA, preferably small hairpin RNA, “shRNA”). siRNA, preferably shRNA, mediate the post-transcriptional process of gene silencing by double stranded RNA (dsRNA) that is homologous in sequence to the silenced RNA. siRNA according to the present invention comprises a sense strand of 17-25 nucleotides complementary or homologous to a contiguous 17-25 nucleotide sequence selected from the group of sequences described in SEQ ID NO: 1-16, preferably from the group of sequences described in SEQ ID No: 128-141 and 231-244, and an antisense strand of 17-25 nucleotides complementary to the sense strand. The most preferred siRNA comprises sense and anti-sense strands that are 100 percent complementary to each other and the TARGET polynucleotide sequence. Preferably the siRNA further comprises a loop region linking the sense and the antisense strand.


A self-complementing single stranded shRNA molecule polynucleotide according to the present invention comprises a sense portion and an antisense portion connected by a loop region linker. Preferably, the loop region sequence is 4-30 nucleotides long, more preferably 5-15 nucleotides long and most preferably 8 nucleotides long. In a most preferred embodiment the linker sequence is UUGCUAUA (SEQ ID NO: 871) or GUUUGCUAUAAC (SEQ ID NO: 872). Self-complementary single stranded siRNAs form hairpin loops and are more stable than ordinary dsRNA. In addition, they are more easily produced from vectors.


Analogous to antisense RNA, the siRNA can be modified to confirm resistance to nucleolytic degradation, or to enhance activity, or to enhance cellular distribution, or to enhance cellular uptake, such modifications may consist of modified internucleoside linkages, modified nucleic acid bases, modified sugars and/or chemical linkage the siRNA to one or more moieties or conjugates. The nucleotide sequences are selected according to siRNA designing rules that give an improved reduction of the TARGET sequences compared to nucleotide sequences that do not comply with these siRNA designing rules (For a discussion of these rules and examples of the preparation of siRNA, WO2004094636, published Nov. 4, 2004, and UA20030198627, are hereby incorporated by reference).


The present invention also relates to compositions, and methods using said compositions, comprising a DNA expression vector capable of expressing a polynucleotide capable of inhibiting extra-cellular matrix degradation and described hereinabove as an expression inhibition agent.


A special aspect of these compositions and methods relates to the down-regulation or blocking of the expression of a TARGET polypeptide by the induced expression of a polynucleotide encoding an intracellular binding protein that is capable of selectively interacting with the TARGET polypeptide. An intracellular binding protein includes any protein capable of selectively interacting, or binding, with the polypeptide in the cell in which it is expressed and neutralizing the function of the polypeptide. Preferably, the intracellular binding protein is a neutralizing antibody or a fragment of a neutralizing antibody having binding affinity to an epitope of the TARGET polypeptide of SEQ ID NO: 17-32, preferably to a domain of SEQ ID NO: 33-127. More preferably, the intracellular binding protein is a single chain antibody.


A special embodiment of this composition comprises the expression-inhibiting agent selected from the group consisting of antisense RNA, antisense oligodeoxynucleotide (ODN), a ribozyme that cleaves the polyribonucleotide coding for SEQ ID NO: 17-32, and a small interfering RNA (siRNA) that is sufficiently homologous to a portion of the polyribonucleotide corresponding to SEQ ID NO: 1-16, such that the siRNA interferes with the translation of the TARGET polyribonucleotide to the TARGET polypeptide.


The polynucleotide expressing the expression-inhibiting agent is preferably included within a vector. The polynucleic acid is operably linked to signals enabling expression of the nucleic acid sequence and is introduced into a cell utilizing, preferably, recombinant vector constructs, which will express the antisense nucleic acid once the vector is introduced into the cell. A variety of viral-based systems are available, including adenoviral, retroviral, adeno-associated viral, lentiviral, herpes simplex viral or a sendaviral vector systems, and all may be used to introduce and express polynucleotide sequence for the expression-inhibiting agents in TARGET cells.


Preferably, the viral vectors used in the methods of the present invention are replication defective. Such replication defective vectors will usually pack at least one region that is necessary for the replication of the virus in the infected cell. These regions can either be eliminated (in whole or in part), or be rendered non-functional by any technique known to a person skilled in the art. These techniques include the total removal, substitution, partial deletion or addition of one or more bases to an essential (for replication) region. Such techniques may be performed in vitro (on the isolated DNA) or in situ, using the techniques of genetic manipulation or by treatment with mutagenic agents. Preferably, the replication defective virus retains the sequences of its genome, which are necessary for encapsidating, the viral particles.


In a preferred embodiment, the viral element is derived from an adenovirus. Preferably, the vehicle includes an adenoviral vector packaged into an adenoviral capsid, or a functional part, derivative, and/or analogue thereof. Adenovirus biology is also comparatively well known on the molecular level. Many tools for adenoviral vectors have been and continue to be developed, thus making an adenoviral capsid a preferred vehicle for incorporating in a library of the invention. An adenovirus is capable of infecting a wide variety of cells. However, different adenoviral serotypes have different preferences for cells. To combine and widen the TARGET cell population that an adenoviral capsid of the invention can enter in a preferred embodiment, the vehicle includes adenoviral fiber proteins from at least two adenoviruses. Preferred adenoviral fiber protein sequences are serotype 17, 45 and 51. Techniques or construction and expression of these chimeric vectors are disclosed in US Published Patent Applications 20030180258 and 20040071660, hereby incorporated by reference.


In a preferred embodiment, the nucleic acid derived from an adenovirus includes the nucleic acid encoding an adenoviral late protein or a functional part, derivative, and/or analogue thereof. An adenoviral late protein, for instance an adenoviral fiber protein, may be favorably used to TARGET the vehicle to a certain cell or to induce enhanced delivery of the vehicle to the cell. Preferably, the nucleic acid derived from an adenovirus encodes for essentially all adenoviral late proteins, enabling the formation of entire adenoviral capsids or functional parts, analogues, and/or derivatives thereof. Preferably, the nucleic acid derived from an adenovirus includes the nucleic acid encoding adenovirus E2A or a functional part, derivative, and/or analogue thereof. Preferably, the nucleic acid derived from an adenovirus includes the nucleic acid encoding at least one E4-region protein or a functional part, derivative, and/or analogue thereof, which facilitates, at least in part, replication of an adenoviral derived nucleic acid in a cell. The adenoviral vectors used in the examples of this application are exemplary of the vectors useful in the present method of treatment invention.


Certain embodiments of the present invention use retroviral vector systems. Retroviruses are integrating viruses that infect dividing cells, and their construction is known in the art. Retroviral vectors can be constructed from different types of retrovirus, such as, MoMuLV (“murine Moloney leukemia virus” MSV (“murine Moloney sarcoma virus”), HaSV (“Harvey sarcoma virus”); SNV (“spleen necrosis virus”); RSV (“Rous sarcoma virus”) and Friend virus. Lentiviral vector systems may also be used in the practice of the present invention. Retroviral systems and herpes virus system may be preferred vehicles for transfection of neuronal cells.


In other embodiments of the present invention, adeno-associated viruses (“AAV”) are utilized. The AAV viruses are DNA viruses of relatively small size that integrate, in a stable and site-specific manner, into the genome of the infected cells. They are able to infect a wide spectrum of cells without inducing any effects on cellular growth, morphology or differentiation, and they do not appear to be involved in human pathologies.


In the vector construction, the polynucleotide agents of the present invention may be linked to one or more regulatory regions. Selection of the appropriate regulatory region or regions is a routine matter, within the level of ordinary skill in the art. Regulatory regions include promoters, and may include enhancers, suppressors, etc.


Promoters that may be used in the expression vectors of the present invention include both constitutive promoters and regulated (inducible) promoters. The promoters may be prokaryotic or eukaryotic depending on the host. Among the prokaryotic (including bacteriophage) promoters useful for practice of this invention are lac, lacZ, T3, T7, lambda P.sub.r, P.sub.1, and trp promoters. Among the eukaryotic (including viral) promoters useful for practice of this invention are ubiquitous promoters (for example, HPRT, vimentin, actin, tubulin), intermediate filament promoters (for example, desmin, neurofilaments, keratin, GFAP), therapeutic gene promoters (for example, MDR type, CFTR, factor VIII), tissue-specific promoters (for example, actin promoter in smooth muscle cells, or Flt and Flk promoters active in endothelial cells), including animal transcriptional control regions, which exhibit tissue specificity and have been utilized in transgenic animals: elastase I gene control region which is active in pancreatic acinar cells (Swift, et al. (1984) Cell 38:639-46; Ornitz, et al. (1986) Cold Spring Harbor Symp. Quant. Biol. 50:399-409; MacDonald, (1987) Hepatology 7:425-515); insulin gene control region which is active in pancreatic beta cells (Hanahan, (1985) Nature 315:115-22), immunoglobulin gene control region which is active in lymphoid cells (Grosschedl, et al. (1984) Cell 38:647-58; Adames, et al. (1985) Nature 318:533-8; Alexander, et al. (1987) Mol. Cell. Biol. 7:1436-44), mouse mammary tumor virus control region which is active in testicular, breast, lymphoid and mast cells (Leder, et al. (1986) Cell 45:485-95), albumin gene control region which is active in liver (Pinkert, et al. (1987) Genes and Devel. 1:268-76), alpha-fetoprotein gene control region which is active in liver (Krumlauf, et al. (1985) Mol. Cell. Biol., 5:1639-48; Hammer, et al. (1987) Science 235:53-8), alpha 1-antitrypsin gene control region which is active in the liver (Kelsey, et al. (1987) Genes and Devel., 1: 161-71), beta-globin gene control region which is active in myeloid cells (Mogram, et al. (1985) Nature 315:338-40; Kollias, et al. (1986) Cell 46:89-94), myelin basic protein gene control region which is active in oligodendrocyte cells in the brain (Readhead, et al. (1987) Cell 48:703-12), myosin light chain-2 gene control region which is active in skeletal muscle (Sani, (1985) Nature 314.283-6), and gonadotropic releasing hormone gene control region which is active in the hypothalamus (Mason, et al. (1986) Science 234:1372-8).


Other promoters which may be used in the practice of the invention include promoters which are preferentially activated in dividing cells, promoters which respond to a stimulus (for example, steroid hormone receptor, retinoic acid receptor), tetracycline-regulated transcriptional modulators, cytomegalovirus immediate-early, retroviral LTR, metallothionein, SV-40, E1a, and MLP promoters.


Additional vector systems include the non-viral systems that facilitate introduction of polynucleotide agents into a patient, for example, a DNA vector encoding a desired sequence can be introduced in vivo by lipofection. Synthetic cationic lipids designed to limit the difficulties encountered with liposome-mediated transfection can be used to prepare liposomes for in vivo transfection of a gene encoding a marker (Feigner, et. al. (1987) Proc. Natl. Acad. Sci. USA 84:7413-7); see Mackey, et al. (1988) Proc. Natl. Acad. Sci. USA 85:8027-31; Ulmer, et al. (1993) Science 259:1745-8). The use of cationic lipids may promote encapsulation of negatively charged nucleic acids, and also promote fusion with negatively charged cell membranes (Feigner and Ringold, (1989) Nature 337:387-8). Particularly useful lipid compounds and compositions for transfer of nucleic acids are described in International Patent Publications WO 95/18863 and WO 96/17823, and in U.S. Pat. No. 5,459,127. The use of lipofection to introduce exogenous genes into the specific organs in vivo has certain practical advantages and directing transfection to particular cell types would be particularly advantageous in a tissue with cellular heterogeneity, for example, pancreas, liver, kidney, and the brain. Lipids may be chemically coupled to other molecules for the purpose of targeting. Targeted peptides, for example, hormones or neurotransmitters, and proteins, for example, antibodies, or non-peptide molecules could be coupled to liposomes chemically. Other molecules are also useful for facilitating transfection of a nucleic acid in vivo, for example, a cationic oligopeptide (for example, International Patent Publication WO 95/21931), peptides derived from DNA binding proteins (for example, International Patent Publication WO 96/25508), or a cationic polymer (for example, International Patent Publication WO 95/21931).


It is also possible to introduce a DNA vector in vivo as a naked DNA plasmid (see U.S. Pat. Nos. 5,693,622; 5,589,466; and 5,580,859). Naked DNA vectors for therapeutic purposes can be introduced into the desired host cells by methods known in the art, for example, transfection, electroporation, microinjection, transduction, cell fusion, DEAE dextran, calcium phosphate precipitation, use of a gene gun, or use of a DNA vector transporter (see, for example, Wilson, et al. (1992) J. Biol. Chem. 267:963-7; Wu and Wu, (1988) J. Biol. Chem. 263:14621-4; Hartmut, et al. Canadian Patent Application No. 2,012,311, filed Mar. 15, 1990; Williams, et al (1991). Proc. Natl. Acad. Sci. USA 88:2726-30). Receptor-mediated DNA delivery approaches can also be used (Curiel, et al. (1992) Hum. Gene Ther. 3:147-54; Wu and Wu, (1987) J. Biol. Chem. 262:4429-32).


The present invention also provides biologically compatible, extra-cellular matrix degradation inhibiting compositions comprising an effective amount of one or more compounds identified as TARGET inhibitors, and/or the expression-inhibiting agents as described hereinabove.


A biologically compatible composition is a composition, that may be solid, liquid, gel, or other form, in which the compound, polynucleotide, vector, and antibody of the invention is maintained in an active form, for example, in a form able to effect a biological activity. For example, a compound of the invention would have inverse agonist or antagonist activity on the TARGET; a nucleic acid would be able to replicate, translate a message, or hybridize to a complementary mRNA of a TARGET; a vector would be able to transfect a TARGET cell and expression the antisense, antibody, ribozyme or siRNA as described hereinabove; an antibody would bind a TARGET polypeptide domain.


A preferred biologically compatible composition is an aqueous solution that is buffered using, for example, Tris, phosphate, or HEPES buffer, containing salt ions. Usually the concentration of salt ions will be similar to physiological levels. Biologically compatible solutions may include stabilizing agents and preservatives. In a more preferred embodiment, the biocompatible composition is a pharmaceutically acceptable composition. Such compositions can be formulated for administration by topical, oral, parenteral, intranasal, subcutaneous, and intraocular, routes. Parenteral administration is meant to include intravenous injection, intramuscular injection, intraarterial injection or infusion techniques. The composition may be administered parenterally in dosage unit formulations containing standard, well-known non-toxic physiologically acceptable carriers, adjuvants and vehicles as desired.


A particularly preferred embodiment of the present composition invention is a extra-cellular matrix degradation inhibiting pharmaceutical composition comprising a therapeutically effective amount of an expression-inhibiting agent as described hereinabove, in admixture with a pharmaceutically acceptable carrier. Another preferred embodiment is a pharmaceutical composition for the treatment or prevention of a condition involving ECM degradation, or a susceptibility to the condition, comprising an effective extra-cellular matrix degradation inhibiting amount of a TARGET antagonist or inverse agonist, its pharmaceutically acceptable salts, hydrates, solvates, or prodrugs thereof in admixture with a pharmaceutically acceptable carrier.


Pharmaceutical compositions for oral administration can be formulated using pharmaceutically acceptable carriers well known in the art in dosages suitable for oral administration. Such carriers enable the pharmaceutical compositions to be formulated as tablets, pills, dragees, capsules, liquids, gels, syrups, slurries, suspensions, and the like, for ingestion by the patient. Pharmaceutical compositions for oral use can be prepared by combining active compounds with solid excipient, optionally grinding a resulting mixture, and processing the mixture of granules, after adding suitable auxiliaries, if desired, to obtain tablets or dragee cores. Suitable excipients are carbohydrate or protein fillers, such as sugars, including lactose, sucrose, mannitol, or sorbitol; starch from corn, wheat, rice, potato, or other plants; cellulose, such as methyl cellulose, hydroxypropylmethyl-cellulose, or sodium carboxymethyl-cellulose; gums including arabic and tragacanth; and proteins such as gelatin and collagen. If desired, disintegrating or solubilizing agents may be added, such as the cross-linked polyvinyl pyrrolidone, agar, alginic acid, or a salt thereof, such as sodium alginate. Dragee cores may be used in conjunction with suitable coatings, such as concentrated sugar solutions, which may also contain gum arabic, talc, polyvinyl-pyrrolidone, carbopol gel, polyethylene glycol, and/or titanium dioxide, lacquer solutions, and suitable organic solvents or solvent mixtures. Dyestuffs or pigments may be added to the tablets or dragee coatings for product identification or to characterize the quantity of active compound, i.e., dosage.


Pharmaceutical preparations that can be used orally include push-fit capsules made of gelatin, as well as soft, sealed capsules made of gelatin and a coating, such as glycerol or sorbitol. Push-fit capsules can contain active ingredients mixed with filler or binders, such as lactose or starches, lubricants, such as talc or magnesium stearate, and, optionally, stabilizers. In soft capsules, the active compounds may be dissolved or suspended in suitable liquids, such as fatty oils, liquid, or liquid polyethylene glycol with or without stabilizers.


Preferred sterile injectable preparations can be a solution or suspension in a non-toxic parenterally acceptable solvent or diluent. Examples of pharmaceutically acceptable carriers are saline, buffered saline, isotonic saline (for example, monosodium or disodium phosphate, sodium, potassium; calcium or magnesium chloride, or mixtures of such salts), Ringer's solution, dextrose, water, sterile water, glycerol, ethanol, and combinations thereof 1,3-butanediol and sterile fixed oils are conveniently employed as solvents or suspending media. Any bland fixed oil can be employed including synthetic mono- or di-glycerides. Fatty acids such as oleic acid also find use in the preparation of injectables.


The composition medium can also be a hydrogel, which is prepared from any biocompatible or non-cytotoxic homo- or hetero-polymer, such as a hydrophilic polyacrylic acid polymer that can act as a drug absorbing sponge. Certain of them, such as, in particular, those obtained from ethylene and/or propylene oxide are commercially available. A hydrogel can be deposited directly onto the surface of the tissue to be treated, for example during surgical intervention.


Embodiments of pharmaceutical compositions of the present invention comprise a replication defective recombinant viral vector encoding the polynucleotide inhibitory agent of the present invention and a transfection enhancer, such as poloxamer. An example of a poloxamer is Poloxamer 407, which is commercially available (BASF, Parsippany, N.J.) and is a non-toxic, biocompatible polyol. A poloxamer impregnated with recombinant viruses may be deposited directly on the surface of the tissue to be treated, for example during a surgical intervention. Poloxamer possesses essentially the same advantages as hydrogel while having a lower viscosity.


The active expression-inhibiting agents may also be entrapped in microcapsules prepared, for example, by interfacial polymerization, for example, hydroxymethylcellulose or gelatin-microcapsules and poly-(methylmethacylate) microcapsules, respectively, in colloidal drug delivery systems (for example, liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsules) or in macroemulsions. Such techniques are disclosed in Remington's Pharmaceutical Sciences (1980) 16th edition, Osol, A. Ed.


Sustained-release preparations may be prepared. Suitable examples of sustained-release preparations include semi-permeable matrices of solid hydrophobic polymers containing the antibody, which matrices are in the form of shaped articles, for example, films, or microcapsules. Examples of sustained-release matrices include polyesters, hydrogels (for example, poly(2-hydroxyethyl-methacrylate), or poly(vinylalcohol)), polylactides (U.S. Pat. No. 3,773,919), copolymers of L-glutamic acid and gamma-ethyl-L-glutamate, non-degradable ethylene-vinyl acetate, degradable lactic acid-glycolic acid copolymers such as the LUPRON DEPOT™. (injectable microspheres composed of lactic acid-glycolic acid copolymer and leuprolide acetate), and poly-D-(−)-3-hydroxybutyric acid. While polymers such as ethylene-vinyl acetate and lactic acid-glycolic acid enable release of molecules for over 100 days, certain hydrogels release proteins for shorter time periods. When encapsulated antibodies remain in the body for a long time, they may denature or aggregate as a result of exposure to moisture at 37° C., resulting in a loss of biological activity and possible changes in immunogenicity. Rational strategies can be devised for stabilization depending on the mechanism involved. For example, if the aggregation mechanism is discovered to be intermolecular S—S bond formation through thio-disulfide interchange, stabilization may, be achieved by modifying sulfhydryl residues, lyophilizing from acidic solutions, controlling moisture content, using appropriate additives, and developing specific polymer matrix compositions.


As defined above, therapeutically effective dose means that amount of protein, polynucleotide, peptide, or its antibodies, agonists or antagonists, which ameliorate the symptoms or condition. Therapeutic efficacy and toxicity of such compounds can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, for example, ED50 (the dose therapeutically effective in 50% of the population) and LD50 (the dose lethal to 50% of the population). The dose ratio of toxic to therapeutic effects is the therapeutic index, and it can be expressed as the ratio, LD50/ED50. Pharmaceutical compositions that exhibit large therapeutic indices are preferred. The data obtained from cell culture assays and animal studies is used in formulating a range of dosage for human use. The dosage of such compounds lies preferably within a range of circulating concentrations that include the ED50 with little or no toxicity. The dosage varies within this range depending upon the dosage form employed, sensitivity of the patient, and the route of administration.


For any compound, the therapeutically effective dose can be estimated initially either in cell culture assays or in animal models, usually mice, rabbits, dogs, or pigs. The animal model is also used to achieve a desirable concentration range and route of administration. Such information can then be used to determine useful doses and routes for administration in humans. The exact dosage is chosen by the individual physician in view of the patient to be treated. Dosage and administration are adjusted to provide sufficient levels of the active moiety or to maintain the desired effect. Additional factors which may be taken into account include the severity of the disease state, age, weight and gender of the patient; diet, desired duration of treatment, method of administration, time and frequency of administration, drug combination(s), reaction sensitivities, and tolerance/response to therapy. Long acting pharmaceutical compositions might be administered every 3 to 4 days, every week, or once every two weeks depending on half-life and clearance rate of the particular formulation.


The pharmaceutical compositions according to this invention may be administered to a subject by a variety of methods. They may be added directly to targeted tissues, complexed with cationic lipids, packaged within liposomes, or delivered to targeted cells by other methods known in the art. Localized administration to the desired tissues may be done by direct injection, transdermal absorption, catheter, infusion pump or stent. The DNA, DNA/vehicle complexes, or the recombinant virus particles are locally administered to the site of treatment. Alternative routes of delivery include, but are not limited to, intravenous injection, intramuscular injection, subcutaneous injection, aerosol inhalation, oral (tablet or pill form), topical, systemic, ocular, intraperitoneal and/or intrathecal delivery. Examples of ribozyme delivery and administration are provided in Sullivan et al. WO 94/02595.


Antibodies according to the invention may be delivered as a bolus only, infused over time or both administered as a bolus and infused over time. Those skilled in the art may employ different formulations for polynucleotides than for proteins. Similarly, delivery of polynucleotides or polypeptides will be specific to particular cells, conditions, locations, etc.


As discussed hereinabove, recombinant viruses may be used to introduce DNA encoding polynucleotide agents useful in the present invention. Recombinant viruses according to the invention are generally formulated and administered in the form of doses of between about 104 and about 1014 pfu. In the case of AAVs and adenoviruses, doses of from about 106 to about 1011 pfu are preferably used. The term pfu (“plaque-forming unit”) corresponds to the infective power of a suspension of virions and is determined by infecting an appropriate cell culture and measuring the number of plaques formed. The techniques for determining the pfu titre of a viral solution are well documented in the prior art.


The present invention also provides methods of inhibiting extra-cellular matrix degradation, comprising administering, to a subject suffering from a disease condition involving extra-cellular matrix degradation, an extra-cellular matrix degradation inhibiting pharmaceutical composition as described herein, preferably a therapeutically effective amount of an expression-inhibiting agent of the present invention. The diseases involving extra-cellular matrix degradation, include psoriatic arthritis, juvenile arthritis, early arthritis, reactive arthritis, osteoarthritis, ankylosing spondylitis, osteoporosis, muscular skeletal diseases such as tendinitis and periodontal disease, cancer metastasis, airway diseases (COPD, asthma), renal and liver fibrosis, cardio-vascular diseases such as atherosclerosis and heart failure, and neurological diseases such as neuroinflammation and multiple sclerosis. More preferred diseases for treatment in accordance with the present invention are the degenerative joint diseases such as psoriatic arthritis, juvenile arthritis, early arthritis, reactive arthritis, osteoarthritis, ankylosing spondylitis. The most preferred degenerative joint disease for treatment in accordance with the present method is rheumatoid arthritis. The present invention also provides methods for treatment of inflammatory diseases.


Administering of the expression-inhibiting agent of the present invention to the subject patient includes both self-administration and administration by another person. The patient may be in need of treatment for an existing disease or medical condition, or may desire prophylactic treatment to prevent or reduce the risk for diseases and medical conditions affected by a disturbance in bone metabolism. The expression-inhibiting agent of the present invention may be delivered to the subject patient orally, transdermally, via inhalation, injection, nasally, rectally or via a sustained release formulation.


A preferred regimen of the present method comprises the administration to a subject in suffering from a disease condition characterized by inflammatory, with an effective inhibiting amount of an expression-inhibiting agent of the present invention for a period of time sufficient to reduce the abnormal levels of extracellular matrix degradation in the patient, and preferably terminate, the self-perpetuating processes responsible for said degradation. A special embodiment of the method comprises administering of an effective matrix metallo-protease inhibiting amount of a expression-inhibiting agent of the present invention to a subject patient suffering from or susceptible to the development of rheumatoid arthritis, for a period of time sufficient to reduce or prevent, respectively, collagen and bone degradation in the joints of said patient, and preferably terminate, the self-perpetuating processes responsible for said degradation.


The invention also relates to the use of an agent as described above for the preparation of a medicament for treating or preventing a disease involving extra-cellular matrix degradation. Preferably the pathological condition is arthritis. More preferably, the pathological condition is rheumatoid arthritis.


The polypeptides and polynucleotides useful in the practice of the present invention described herein may be free in solution, affixed to a solid support, borne on a cell surface, or located intracellularly. To perform the methods it is feasible to immobilize either the TARGET polypeptide or the compound to facilitate separation of complexes from uncomplexed forms of the polypeptide, as well as to accommodate automation of the assay. Interaction (for example, binding of) of the TARGET polypeptide with a compound can be accomplished in any vessel suitable for containing the reactants. Examples of such vessels include microtitre plates, test tubes, and microcentrifuge tubes. In one embodiment, a fusion protein can be provided which adds a domain that allows the polypeptide to be bound to a matrix. For example, the TARGET polypeptide can be “His” tagged, and subsequently adsorbed onto Ni-NTA microtitre plates, or ProtA fusions with the TARGET polypeptides can be adsorbed to IgG, which are then combined with the cell lysates (for example, (35)S-labelled) and the candidate compound, and the mixture incubated under conditions favorable for complex formation (for example, at physiological conditions for salt and pH). Following incubation, the plates are washed to remove any unbound label, and the matrix is immobilized. The amount of radioactivity can be determined directly, or in the supernatant after dissociation of the complexes. Alternatively, the complexes can be dissociated from the matrix, separated by SDS-PAGE, and the level of the protein binding to the TARGET protein quantified from the gel using standard electrophoretic techniques.


Other techniques for immobilizing protein on matrices can also be used in the method of identifying compounds. For example, either the TARGET or the compound can be immobilized utilizing conjugation of biotin and streptavidin. Biotinylated TARGET protein molecules can be prepared from biotin-NHS (N-hydroxy-succinimide) using techniques well known in the art (for example, biotinylation kit, Pierce Chemicals, Rockford, and immobilized in the wells of streptavidin-coated 96 well plates (Pierce Chemical). Alternatively, antibodies reactive with the TARGETS but which do not interfere with binding of the TARGET to the compound can be derivatized to the wells of the plate, and the TARGET can be trapped in the wells by antibody conjugation. As described above, preparations of a labeled candidate compound are incubated in the wells of the plate presenting the TARGETS, and the amount of complex trapped in the well can be quantitated.


The polynucleotides encoding the TARGET polypeptides are identified as SEQ ID NO: 1-16. The present inventors show herein that transfection of mammalian cells with Ad-siRNAs targeting these genes decreases the release of factors that promote extra-cellular matrix degradation.


The present invention also relates to a method for diagnosis of a pathological condition involving ECM degradation, comprising determining the nucleic acid sequence of at least one of the genes of SEQ ID NO: 1-16 within the genomic DNA of a subject; comparing the sequence with the nucleic acid sequence obtained from a database and/or a healthy subject; and identifying any difference(s) related to the onset of the pathological condition.


Still another aspect of the invention relates to a method for diagnosing a pathological condition involving extra-cellular matrix degradation or a susceptibility to the condition in a subject, comprising determining the amount of polypeptide comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 17-32 in a biological sample, and comparing the amount with the amount of the polypeptide in a healthy subject, wherein an increase of the amount of polypeptide compared to the healthy subject is indicative of the presence of the pathological condition.


The invention is further illustrated in the following figures and examples.


EXAMPLES

The following assays, when used in combination with arrayed adenoviral shRNA (small hairpin RNA) expression libraries (the production and use of which are described in WO99/64582), are useful for the discovery of factors that modulate the capacity of synovial fibroblasts (SFs) to produce MMP1 and degrade collagen, an important component of cartilage. Candidate factors are filtered first through a primary followed by a secondary assay.


Example 1 describes the development and setup of the primary assay screen of an adenoviral siRNA library using an ELISA for detection of protein levels of Matrix Metalloprotease 1 (MMP 1), and is referred to herein as the “reverse MMP1 assay”. Example 2 describes the screening and its results. Example 3 describes a further screen that eliminates any targets that inhibit MMP1 expression in a nonspecific way. Example 4 describes the testing of a subset of the targets for the endogenous SF expression. Example 5 describes a dose response experiment that tests a subset of the Ad-siRNAs for the inhibition of cytokine-induced MMP1 expression. Example 6 describes the secondary assay referred to herein as the “collagen degradation assay”, which is more functionally oriented, and which detects collagen degradation in the supernatant of SFs. Example 7 describes the testing of a subset of the Ad-siRNAs for the inhibition of cytokine-induced IL-8 expression.


Control Viruses Used:


The control viruses used in these studies are listed below. dE1/dE2A adenoviruses are generated by co-transfection of adapter plasmids described below with the helper plasmid pWEAd5AflII-rITR.dE2A in PER.E2A packaging cells, as described in WO99/64582.


(A) Negative Control Viruses:




  • Ad5-LacZ: Described as pIPspAdApt6-lacZ in WO02/070744.

  • Ad5-ALPP: The 1.9 kb insert is isolated from pGT65-PLAP (Invitrogen) by digestion with NsiI; blunted; followed by digestion with EcoR1 and cloned into EcoRI and HpaI-digested pIPspAdApt6.

  • Ad5-eGFP: Described as pIPspAdApt6-EGFP in WO02/070744.










(SEQ ID NO: 245)


Ad5-eGFP_KD: Target sequence: GCTGACCCTGAAGTTCATC.








    • Cloned using Sap1-sites into vector and virus generated as described in WO03/020931.














(SEQ ID NO: 246)



Ad5-Luciferase_v13_KD:



Target sequence: GCTGACCCTGAAGTTCATC.








    • Cloned using Sap1-sites into vector and virus generated as described in WO03/020931.














(SEQ ID NO: 247)



Ad5-DCK_v1_KD:



Target sequence ATGAAGAGCAAGGCATTCC.








    • Cloned using Sap1-sites into vector and virus generated as described in WO03/020931.














(SEQ ID NO: 248)



Ad5-PRKCL1_v1_KD:



Target sequence TGCCTGGGACCAGAGCTTC.








    • Cloned using Sap1-sites into vector and virus generated as described in WO03/020931.














(SEQ ID NO: 249)



Ad5-TNFSF15_v1_KD:



Target sequence GGAAGTAATTGGATCATGC.








    • Cloned using Sap1-sites into vector and virus generated as described in WO03/020931.














(SEQ ID NO: 250)



Ad5-M6PR_v1_KD:



Target sequence: GCTGACCCTGAAGTTCATC.








    • Cloned using Sap1-sites into vector and virus generated as described in WO03/020931.


      (B) Positive Control Viruses:



  • Ad5-MMP1: The cDNA encoding MMP1, cloned into the pIPspAdapt6 plasmid, is isolated from a human placenta cDNA library (see WO02/070744) by classical filter colony hybridization strategy. A human placental cDNA library is transformed into bacteria and plated out on agar plates. Thousands of individual colonies are picked (using a Q-pix device (Genetix)) and re-arrayed on agar plates. After growing bacteria up, these plates are overlaid on hybridization filters. These filters are subjected to a classical hybridization procedure with a MMP1 specific probe. This probe is obtained by PCR on a placenta cDNA library using the following primers:












(SEQ ID NO: 873)










upstream:
GTTCTGGGGTGTGGTGTCTCACAGC;



and














(SEQ ID NO: 874)










downstream:
CAAACTGAGCCACATCAGGCACTCC.








    • A bacterial colony, at a position corresponding to that of a positive signal spot on the filter after hybridization, is picked and used for plasmid preparation. 5′ sequence verification confirms that the 5′ sequence of the insert corresponds to NM002421.



  • Ad5-MMP13: The cDNA of MMP13 is isolated from a cDNA preparation from human synovial fibroblasts by PCR. The 1498 by PCR product is cloned into pIPspAdapt6 using a HindIII/EcoRI cloning strategy. Sequence verification confirms that the insert corresponds to by 18 to 1497 of NM002427.

  • Ad5-MYD88: This cDNA is isolated from a human placenta cDNA library constructed in pIPspAdapt6. The virus mediating the expression of MYD88 is identified as a hit in one of the genomic screen run at Galapagos Genomics. Sequence verification of the insert confirms that the insert corresponds to by 40 to 930 of NM002468.












(SEQ ID NO: 251)



Ad5-MMP1_v10_KD:



Target sequence: GCTGACCCTGAAGTTCATC.








    • Cloned using Sap1-sites into vector and virus generated as described in WO03/020931.














(SEQ ID NO: 252)



Ad5-PRKCE_v11_KD:



Target sequence GTCATGTTGGCAGAACTC.








    • Cloned using Sap1-sites into vector and virus generated as described in WO03/020931.





Example 1
Development of the Reverse MMP1 Assay

The MMP1 assay is developed by first testing the capacity of synovial fibroblasts (SFs) to produce MMP1.


To evaluate the capacity of SFs to produce MMP1, these cells were infected with a recombinant adenovirus mediating the expression of the MYD88 adaptor molecule involved in the pro-inflammatory IL1□ signaling pathway. This virus is expected to increase MMP1 expression in these cells (see Vincenti and Brinckerhoff, 2002).


40,000 SFs are seeded per well of a 6-well plate in DMEM+10% FBS and each well is either infected with a recombinant adenovirus mediating the expression of eGFP, MYD88 or MMP1 (with a multiplicity of infection (MOI) of 7500 viral particles per cell (vp/cell)) or left uninfected (blank). The SF expression of MMP1 is first determined at the mRNA level, by means of real-time, quantitative PCR. RNA of the cells infected with the control viruses is prepared 48 hours post infection using the SV RNA isolation kit (Promega), according to the instructions of the manufacturer. cDNA is prepared from this RNA using Multiscribe reverse transcriptase (50 U/μl, Applied Biosystems) and random hexamers. cDNA synthesis is performed in 25 μl total volume consisting of 1× TaqMan buffer A (PE Applied Biosystems), 5 mM MgCl2, dNTPs (500 μM final per dNTP), 2.5 mM random hexamers, 0.4 U/μl RNase Inhibitor, and 1.25 U/μl MultiScribe Reverse Transcriptase. The mixture is incubated for 10 minutes at 25° C., 30 minutes at 48° C., and 5 minutes at 95° C. Specific DNA products are amplified from the resulting cDNA with AmpliTaq Gold DNA polymerase (Applied BioSystems) during 40 PCR cycles using suited primer pairs. Amplification of the specific DNA products is monitored on an ABI PRISM® 7000 Sequence Detection System. The subsequent real time PCR reaction contained 5 μl of the RT reaction product in a total volume of 25 μl consisting of 1×SYBR Green mix (Applied Biosystems), 300 nM forward primer, and 300 nM reverse primer. Each sample is analyzed in duplicate. The PCR reaction is performed using the following program: 10 minutes at 95° C. followed by 40 cycles of (15 sec. 95° C., 1 minutes 60° C.). After each PCR reaction the products are analyzed by measuring the dissociation curve by incubating for 15 sec. 95° C., and 15 sec. at 60° C., followed by increasing the temperature to 95° C. over a 20 minutes time period, ending with 15 sec. at 95° C. The sequences of the primer pairs used for the detection of MMP1 expression are listed in Table 3.









TABLE 3







List of primers and their sequences used herein.









Primer name
Primer sequence
SEQ ID NO





pAdapt_FW
GGTGGGAGGTCTATATAAGC
875





pAdapt_REV
GGACAAACCACAACTAGAATGC
876





MMP1_For
CCGGTTTTTCAAAGGGAATAAGTAC
877





MMP1_Rev
TTCACAGTTCTAGGGAAGCCAAAG
878









MMP1 is detected using the SYBR Green method, whereas the levels of 18S rRNA, used as internal calibrator for the PCR reaction, is measured using a commercially available set of primers and Taqman probe (TaqMan® Ribosomal RNA Control Reagents, Applied Biosystems). The amplification plot and the resulting threshold Ct value are indicators for the amount of specific mRNAs present in the samples. Delta-delta Ct values are presented, meaning the normalized (relative to the 18S calibrator) levels of MMP1 mRNA in the samples infected with the positive control viruses relative to the expression levels in a Ad5-eGFP infected control sample. Results indicate a strong up-regulation of the MMP1 mRNA levels upon expression of MYD88 in SFs as compared to the non-infected or Ad5-eGFP-infected SFs (FIG. 2, panel A).


The level of MMP1 expressed by SFs is also determined at the protein level by Western Blotting. Two days after infection, supernatant of cells, infected with various recombinant adenoviruses as indicated for the Real-time PCR experiment, is collected and concentrated 15 times by classical TCA precipitation. 15 μl of the supernatant are resolved by SDS-PAGE using a 10% polyacrylamide gel. For these experiments, the medium used is M199 medium+1% FBS. For the MMP1 control sample, non-concentrated supernatant of cells infected with Ad5-MMP1 is loaded onto the gel. The resolved proteins are transferred onto a nitrocellulose membrane. The quality of the transfer and equal loading of the samples are verified by Ponceau-S staining of the membrane. Immunodetection is performed using a goat anti-MMP1 polyclonal antibody as primary antibody (R&D Systems, 1/500 dilution) and an HRP-linked rabbit anti-goat antibody (DAKO, 1/10000 dilution) as secondary antibody and ECL plus HRP substrate (Amersham Biosciences). The Western Blotting revealed a strongly increased level of MMP1 protein in the supernatant of the SFs infected with the adenoviruses mediating expression of Ad5-MYD88 as compared to the Ad5-eGFP infected cells. A very strong signal is detected for the supernatant of cells infected with Ad5-MMP1 (FIG. 2, panel B).


The high levels of MMP1 protein present in the supernatant of the Ad5-MYD88 infected SFs are confirmed using a commercially available MMP1 activity ELISA (RPN2629, Amersham Biosciences). In this ELISA, MMP1 is captured by an antibody immobilized in a well and the amount is subsequently quantified based on the conversion of a MMP1 substrate. 50 μl of non-concentrated supernatant of SFs (prepared as indicated for the western blotting experiment) are processed in this ELISA as recommended by the manufacturer (FIG. 2, panel C).


These experiments confirm the capacity of SFs, in general, and of the cell batch used for screening and validation experiments, in particular, to produce MMP1 protein upon triggering of inflammatory pathways.


A 384-well format ELISA for measurement of MMP1 is developed. Various primary antibodies are tested, as well as various ELISA protocols. The following protocol is developed and validated to measure MMP1 levels in SF supernatant in 384 well plates: white Lumitrac 600 384 well plates (Greiner) are coated with 2 μg/ml anti-MMP1 antibody MAB1346 (Chemicon). The antibody is diluted in buffer 40 (1.21 g Tris base (Sigma), 0.58 g NaCl (Calbiochem) and 5 ml 10% NaN3 (Sigma) in 1 L milliQ water and adjusted to pH 8.5). After overnight incubation at 4° C., plates are washed with PBS (80 g NaCl, 2 g KCl (Sigma), 11.5 g Na2HPO4.7H2O and 2 g KH2PO4 in 10 L milliQ; pH 7.4) and blocked with 100 μl/well Casein buffer (2% Casein (VWR International) in PBS). Next day, casein buffer is removed from ELISA plates and replaced by 50 μl/well EC buffer (4 g casein, 2.13 g Na2HPO4 (Sigma), 2 g bovine albumin (Sigma), 0.69 g NaH2PO4.H2O (Sigma), 0.5 g CHAPS (Roche), 23.3 g NaCl, 4 ml 0.5 M EDTA pH 8 (Invitrogen), 5 ml 10% NaN3 in 1 L milliQ and adjusted to pH 7.0). 0.25 mM DTT (Sigma) are added to the thawed samples plates. After removal of the EC buffer, 20 μl of sample are transferred to the ELISA plates. After overnight incubation at 4° C., the plates are washed twice with PBS, once with PBST (PBS with 0.05% Tween-20 (Sigma)), and incubated with 35 μl/well biotinylated anti-MMP1 antibody solution (R&D). This secondary antibody is diluted in buffer C (0.82 g NaH2PO4.H2O, 4.82 g Na2HPO4, 46.6 g NaCl, 20 g bovine albumin and 4 ml 0.5M EDTA pH 8 in 2 L milliQ and adjusted to pH 7.0) at a concentration of 5 μg/ml. After 2 hours of incubation at RT, the plates are washed as described above and incubated with 50 μl/well streptavidin-HRP conjugate (Biosource). Streptavidin-HRP conjugate is diluted in buffer C at a concentration of 0.25 μg/ml. After 45 minutes, the plates are washed as described above and incubated for 5 minutes with 50 μl Uwell BM Chem ELISA Substrate (Roche). Readout is performed on the Luminoscan Ascent Luminometer (Labsystems) with an integration time of 200 msec or with an Envision reader (Perkin Elmer).


Typical results obtained with the MMP1 ELISA developed are shown in FIG. 3. For this experiment, 3000 SFs are seeded in a 96 well plate in DMEM+10% FBS. 24 hours later, SFs are either infected at an MOI of 10000 with adenoviruses mediating the expression of ALPP, MYD88, MMP1, or left uninfected. One day after the infection, the medium of the cells is replaced by M199 medium (Invitrogen) supplemented with 1% FBS. After an incubation time of 48 hours, the supernatant is harvested, transferred to a 384 well plate and subjected to the MMP1 ELISA procedure described above. A robust, more than 3.5-fold up-regulation of the signal is observed. This experiment demonstrated the robustness and specificity of the MMP1 ELISA.


The increase of MMP1 expression by SFs upon treatment with cytokines relevant in the field of RA (TNF□, IL1□ and OSM) or a combination thereof is monitored. Results are shown in FIG. 4 as white bars. For this experiment, SFs are seeded in 96 well plates at 3000 cells/well. 24 hours later, the medium is changed to M199 medium supplemented with 1% FBS. One day after the medium change, cytokines or combinations thereof are added to the cultures, each cytokine is added to a final concentration of 25 ng/ml. 72 hours after cytokine addition, the supernatant is collected and processed in the ELISA, as described for FIG. 3. As shown by the white bars in FIG. 4, TNF□ alone induces an almost 3-fold increase in MMP1 expression over the untreated cells. Triggering of SFs with a combination of TNF□ and OSM and/or IL1□ leads to even higher MMP1 expression levels. This experiment demonstrates that the sensitivity of the MMP1 ELISA developed is sufficient to measure increases in MMP1 expression by SFs induced by cytokines relevant in RA pathogenesis.


In parallel with the above experiment, SFs are triggered, using the same protocol, with the supernatant of THP1 cells (2-fold diluted in M199+1% FBS) that are treated with the same cytokines or combinations of cytokines for 48 hours in M199 medium+1% FBS. MMP1 levels for these samples are shown in FIG. 4 as grey bars. The supernatant of TNF□-treated THP1 cells alone induces a more than 4.5-fold increase in MMP1 expression as compared to the level for untreated supernatant of THP1 cells. The 4.5-fold increase is more than the 3-fold induction with recombinant TNF□ alone, and almost equals the 5-fold induction obtained by a mixture of 3 purified cytokines (TNF□, IL1□, OSM). Thus, the supernatant of TNF□-induced THP1 cells contains, besides TNF□, additional pro-inflammatory factors that activate MMP1 expression in SFs. This TNF□-based trigger mixture (prepared by contacting THP-1 cells with TNF□) will likely contain factors present in the joints of RA patients and therefore is relevant to RA. This TNF□-based complex trigger further referred to as the “TNF□-based trigger,” will be used as a basis for the “reverse MMP1 assay”.


The activation of SFs by the “TNF□-based trigger” can be inhibited in a dose-dependent manner by treating the SFs with dexamethasone, a potent anti-inflammatory agent that also strongly reduces collagen-induced arthritis in rodents (Yang et al., 2004) (FIG. 5). SFs are seeded at a density of 3000 cells/well in 96 well plates. Twenty-four hours after seeding, dexamethasone at various concentrations is added to the cells. Following overnight incubation, the media from each cell is refreshed with the supernatant of THP-1 cells treated with TNF□ (50% diluted in M199+0.5% FBS), and dexamethasone at the same concentration as previously added. Forty-eight hours after treatment, the supernatant is collected and subjected to the MMP1 ELISA described above. MMP1 expression by SFs is reduced in a dose-dependent manner by the addition of dexamethasone, which exhibits an IC50 value of about 1 nM (see FIG. 5). This data shows that MMP1 expression by activated SFs can be reduced by the addition of a physiologically relevant inhibitor and validates the principal for the “reverse MMP1 assay”.


Example 2
Screening of 11,744 “Ad-siRNAs” in a Reverse MMP1 Assay

Primary Screening


An arrayed collection of 11,744 different recombinant adenoviruses mediating the expression of shRNAs in SFs are screened using the reverse MMP1 assay. These shRNAs cause a reduction in expression levels of genes that contain homologous sequences by a mechanism known as RNA interference (RNAi). The 11,744 Ad-siRNAs contained in the arrayed collection target 5046 different transcripts. On average, every transcript is targeted by two to three independent Ad-siRNAs. A schematic representation of the screening process is illustrated in FIG. 6. As discussed in more detail below, SFs are seeded in 384 well plates and infected the day thereafter with the arrayed shRNA library, whereby each well is infected with one individual Ad-siRNA. Five days after infection, the medium is refreshed and cells are triggered with the supernatant of TNFα treated THP-1 cells. Two days after addition of the trigger, supernatant is collected and subjected to the MMP1 ELISA.


A 384 well control plate is generated to assess the quality of the assay during different screening runs. The composition of this plate is shown in FIG. 7A. Wells are filled with control viruses that are produced under the same conditions as the SilenceSelect adenoviral collection. The viruses include three sets of 48 positive control viruses (P1 (Ad5-DCK_v1_KD), P2 (Ad5-PRKCL1_v1_KD), P3 (Ad5-TNFSF15_v1_KD)), arranged in diagonal, interspaced with three sets of 48 negative control viruses (N1 (Ad5-eGFP_v5_KD), N2 (Ad5-Luc_v15_KD), N3 (Ad5-eGFP_v1_KD), B1: blanco, uninfected). Every well of a control plate contained 50 μl of virus crude lysate. Multiple aliquots of this control plate are produced and stored at −80° C.


Optimal screening protocol: RASFs at passage 1 (Cell Applications) are cultured in DMEM medium (Invitrogen), supplemented with 10% fetal calf serum (ICN), 100 units/ml penicillin (Invitrogen) and 100 μg/ml streptomycin (Invitrogen), and incubated at 37° C. and 10% CO2. The cells are passed once a week by a ⅓ split. The maximal passage number for RASFs used in the screening is 11. For screening, the SFs are seeded in transparent 384 well plates (Greiner) coated with 0.1% gelatin (Merck) at a density of 1150 cells/well in 50 μl Synovial Cell growth medium (Cell Applications, Inc.). One day later, 2 μl Ad-siRNA virus from the SilenceSelect collection (WO 03/020931) are transferred to each individual well of the 384 well plates containing the SFs. The average titer of the adenoviral is 1×109 viral particles/ml, representing a MOI of about 1700. The SilenceSelect collection is stored in 384 well plates and transferred to the SFs by using a 96/384-channel dispenser. Five days after infection, the medium is removed and the wells are rinsed once by addition and subsequent removal of 80 μl of M199 medium supplemented with 1% FBS. The wells are then filled with 60 μl of “TNFalpha based trigger” diluted 2-fold in M199 medium containing 1% FBS. Two days after addition of the “TNFalpha based trigger”, the supernatant is collected in 384 well plates (Greiner) and stored at −80° C. until further processing in the MMP1 ELISA. The infection, rinsing, and medium collection steps are performed with a TECAN Freedom pipette (Tecan Freedom 200 equipped with TeMO96, TeMO384 and RoMa, Tecan A G, Switzerland). 25 μl of the collected supernatant is subjected to the MMP1 ELISA. The ELISA step is performed as indicated in Example 1.


The “TNFalpha based trigger” is produced from THP-1 cells at passage 8 to 16 grown in suspension cultures in RPMI supplemented with 10% FBS (Invitrogen) and penicillin (100 units/ml) and Streptomycin (100 μg/ml) (Invitrogen). The cultures are diluted weekly to a cell density of 2×105 cells/ml, avoiding densities exceeding 1.5×106 cells/ml. The production of the “TNFalpha based trigger” is initiated by seeding the THP-1 cells in M199 medium supplemented with 1% serum at a density of 1×106 cells/ml. One day after seeding, recombinant human TNFalpha (Sigma) is added to the culture flasks to a final concentration of 25 ng/ml. 48 hours after addition of the cytokine, the supernatant is collected and stored at −80° C. in aliquots until further use. Every new batch of “TNFalpha based trigger” is characterized for its efficacy at inducing MMP1 expression by SFs.


A representative example of the performance of the control plate tested with the screening protocol described above is shown in FIG. 7B. The raw luminescence signal obtained for every well is shown. The positive control samples are selected as the 3 strongest hits picked up in a limited, preliminary screening of one 384 well SilenceSelect plate.


A stringent cutoff is applied, that is, the average of all 144 negative control viruses minus 1.82 times the standard deviation. Samples scoring below this cutoff are considered positive hits. These positive hits are indicated as white numbers on a gray background. As expected, the positive control viruses score very well in the assay with 88%, 81% and 94% of the samples falling below the cutoff for the P1, P2 and P3 positive controls samples, respectively. As non-infected wells display lower signals as compared to infected wells, the blanco samples often score positive. The reason for this is not known. To rule out that a sample would be positive because of the absence of virus, the virus content of all wells is monitored by checking the cytopathic effect (CPE) when propagating hit viruses as well as checking the amount of adenoviral particles per well in a quantitative real-time PCR (Ma et al., 2000). Of the 3 negative control viruses, the N1 virus gives rise to some false positives due to toxicity. During screening, about 5% false positives for the N1 negative control are allowed. In this Example, 2 samples in 48 (+/−4%) score positive for the N1 control.


The complete SilenceSelect collection, consisting of 11,744 Ad-siRNAs targeting 5046 transcripts contained on 30×384 well plates, is screened in the “reverse MMP1 assay” according to the protocol described above. Every Ad-siRNA plate is screened in duplicate in a primary screen and a repeat screen. As such, four data points are obtained for each Ad-siRNA. Ad-siRNA viruses are nominated as hits if at least 1 data point in both the primary screen and repeat screen scored below threshold. A representative example of screening results and of the analysis performed to identify hits is shown in FIG. 8.


To determine the cutoff value for hit calling, the average as well as standard deviation is calculated on all data points per plate. The cutoff value is then defined as the average minus 1.82 times the standard deviation. This cutoff is indicated as a horizontal line in the graphs shown in FIG. 8. The data represented in FIG. 8 are expressed relative to the plate average as follows: the relative signal for a sample=[(raw luminescence signal for the sample)−(average signal over the plate)]/(the standard deviation over the plate). The average of the 2 duplicate signals obtained for all the 384 Ad-siRNAs in the primary screen (FIG. 8A) and in the repeat screen (FIG. 8B) are shown. The data points identified as hits are indicated as circles. A total of 408 hits that scored below the cutoff are isolated for the 3 MOI repeat screen discussed below. Almost all the same Ad-siRNAs score positive in both the primary screen and repeat screen. These data are indicative of the quality of the screening and of the SilenceSelect collection.


In FIG. 9, all data points obtained in the screening of the SilenceSelect collection against the reverse MMP1 assay are shown. The averaged relative luminescence data obtained from the duplicate samples in the primary screen (Y-axis) is plotted against the averaged relative luminescence data for the corresponding Ad-siRNA obtained in the repeat screen X-axis). The cutoff (−1.82 times standard deviation) is indicated by dotted lines. The data points for Ad-siRNAs nominated as hits are indicated as triangles, the data points for the non-hit Ad-siRNAs are indicated as squares. The strong symmetry observed between the data of the primary screen and of the repeat screen (the data points are concentrated around a straight line) is indicative of the quality and reproducibility of the screening.


“3” MOI Repeat Screen


The 408 hits identified above are re-propagated. The crude lysates of the hit Ad-siRNAs samples from the SilenceSelect collection are picked and arranged together with negative controls in 9×96 well plates. As the containers of crude lystates are labeled with a barcode (Screenmates™, Matrix technologies), quality checks are performed on the plates. The general layout of such a “3” MOI repeat screen plate is shown in FIG. 10A. To propagate the viruses, 2.25×104 PerC6.E2A cells are seeded in 200 μl of DMEM containing 10% non-heat inactivated FCS in each well of a 96 well plate and incubated overnight at 39° C. in a humidified incubator at 10% CO2. One μl of crude lysate from each hit Ad-siRNAs, arranged in the 96 well plates as indicated above, is then added to separate wells of PerC6E2A cells using a 96 well dispenser. After 7 to 10 days of incubation in a humidified incubator at 10% CO2, the re-propagation plates are frozen at −20° C., provided that complete CPE could be observed.


For the “3” MOI repeat screen, SFs are seeded in 96 well plates (Greiner, tissue culture treated) at a density of 3000 cells/well in 100 μl Synovial Cell growth medium (Cell Applications, Inc.). One day later, the SFs are infected with 3, 6, or 12 μl of the crude lysate contained in the re-propagation plates described above using a 96 well dispenser (TECAN freedom 200). Five days after infection, medium is removed from the plates, using a VacuSafe device (Integra), and 100 μl of two-fold diluted TNFalpha based trigger is added. After an incubation of 2 days, medium is collected with a 96 well dispenser (TECAN Freedom 200) and stored at −80° C. in 384 well plates until further processed in the reverse MMP1 ELISA according to the protocol described in Example 1.


The data from the MMP1 ELISA is analyzed as follows: To determine the cutoff value for hit calling, the average of the negative controls is calculated for every plate. For every MOI, a percentage is selected such that less than 3% of the negative controls would score as a hit over the 9 plates. The percentages are 23%, 26% and 19% for the 3 μl, 6 μl, and 12 μl infections, respectively. Of the 408 hits tested, 339 scored in duplicate at one MOI and are identified as a hit. 84% of the hits of the primary screen are thus confirmed in this 3 MOI repeat screen using repropagated Ad-siRNA material.


Quality Control of Target Ad-siRNAs


Target Ad-siRNAs are propagated using derivatives of PER.C6® cells (Crucell, Leiden, The Netherlands) in 96-well plates, followed by sequencing the siRNAs encoded by the target Ad-siRNA viruses. PERC6.E2A cells are seeded in 96 well plates at a density of 40,000 cells/well in 180 μl PER.E2A medium. Cells are then incubated overnight at 39° C. in a 10% CO2 humidified incubator. One day later, cells are infected with 1 μl of crude cell lysate from SilenceSelect stocks containing target Ad-siRNAs. Cells are incubated further at 34° C., 10% CO2 until appearance of cytopathic effect (as revealed by the swelling and rounding up of the cells, typically 7 days post infection). The supernatant is collected, and the virus crude lysate is treated with proteinase K by adding to 4 μl Lysis buffer (1× Expand High Fidelity buffer with MgCl2 (Roche Molecular Biochemicals, Cat. No 1332465) supplemented with 1 mg/ml proteinase K (Roche Molecular Biochemicals, Cat No 745 723) and 0.45% Tween-20 (Roche Molecular Biochemicals, Cat No 1335465) to 12 μl crude lysate in sterile PCR tubes. These tubes are incubated at 55° C. for 2 hours followed by a 15 minutes inactivation step at 95° C. For the PCR reaction, 1 μl lysate is added to a PCR master mix composed of 5 μl 10× Expand High Fidelity buffer with MgCl2, 0.5 μl of dNTP mix (10 mM for each dNTP), 1 μl of “Forward primer” (10 mM stock, sequence: 5′ CCG TTT ACG TGG AGA CTC GCC 3′) (SEQ. ID NO. 879), 1 μl of “Reverse Primer” (10 mM stock, sequence: 5′ CCC CCA CCT TAT ATA TAT TCT TTC C) (SEQ. ID NO. 880), 0.2 μl of Expand High Fidelity DNA polymerase (3.5 U/μl, Roche Molecular Biochemicals) and 41.3 μl of H2O. PCR is performed in a PE Biosystems GeneAmp PCR system 9700 as follows: the PCR mixture (50 μl in total) is incubated at 95° C. for 5 minutes; each cycle runs at 95° C. for 15 sec., 55° C. for 30 sec., 68° C. for 4 minutes, and is repeated for 35 cycles. A final incubation at 68° C. is performed for 7 minutes 5 μl of the PCR mixture is mixed with 2 μl of 6× gel loading buffer, loaded on a 0.8% agarose gel containing 0.5 μg/μl ethidium bromide to resolve the amplification products. The size of the amplified fragments is estimated from a standard DNA ladder loaded on the same gel. The expected size is approximately 500 bp. For sequencing analysis, the siRNA constructs expressed by the target adenoviruses are amplified by PCR using primers complementary to vector sequences flanking the SapI site of the pIPspAdapt6-U6 plasmid. The sequence of the PCR fragments is determined and compared with the expected sequence. All sequences are found to be identical to the expected sequence.


Example 3A
Testing of 339 Hits of the “Reverse MMP1 Assay” to Exclude Hits that Influence MMP1 Expression Levels in a Nonspecific Way

To eliminate general toxicity (reduced viability) of SFs by the Ad-siRNAs as the cause of reduced expression of MMP1 in the reverse MMP1 assay, the CellTiter-Glo Luminescent Cell Viability Assay (Promega) is used. This Assay determines the number of viable cells in culture based on the quantity of ATP present, which is proportional to the presence of metabolically active cells. In this assay, cells are grown in a white 96 well plate at a density of 3000 cells/well in 100 μl Synovial Cell growth medium (Cell Applications, Inc.). After overnight incubation, cells are infected with the confirmed hits from the reverse MMP1 assay. Five days after infection, the medium is removed and replaced by 50 μM199 medium supplemented with 1% FBS. The plate and its content are equilibrated to room temperature for 30 minutes. 50 μl celltiter-Glo reagent is added to the wells, followed by an incubation on an orbital shaker (5 minutes) in the dark. The measurement is performed on a luminoskan Ascent (Labsystems) luminometer, 100 ms integration time.


In a primary test, 12 μl of the viruses contained in the 3 MOI repeat screen plates (layout depicted in FIG. 10A) are used to infect the RASFs, and the readout is performed as described above. To determine which hit viruses cause general toxicity, a cutoff is calculated by averaging the signal for all data points contained in the plate (50 hits and 10 controls) and subtracting 1.5 times the standard deviation over all data points. The hits scoring under the cutoff value in the primary screen are retested as follows: The hits are picked, arranged in 96 well plates (according to the layout depicted in FIG. 10B) and used to infect SFs at 3 MOIs (3, 6, and 12 μl) according to the protocol described above. For the repeat screen, a cutoff is calculated by averaging the signal for all the controls and subtracting 2 times the standard deviation over the controls. Hits scoring under the cutoff in duplicate at one MOI are considered as inducing toxicity.


The secretion of TIMP-2 by SFs is determined to eliminate the factor of general suppression of SF secretory activity by the Ad-siRNAs as the cause of reduced expression of MMP1 in the reverse MMP1 assay. TIMP-2 is a protein that SFs express and secret constitutively. Secretion of TIMP-2 into the SF supernatant is blocked by the addition of BrefeldinA, a small molecule inhibitor of secretion. The level of SF TIMP-2 protein secretion is detected by ELISA. RASF cells are seeded in 96-well gelatin coated plates at 3000 cells/well in 100 μl synovial growth medium. After overnight incubation, cells are infected and incubated for 5 days at 37° C. in a 10% CO2 incubator. Virus is removed and 100 μl M199+1% FBS(HI) medium is applied on top of the cells. The supernatant is harvested after another 48 hours incubation and stored at −20° C. TIMP2 levels are determined by a standard ELISA assay, essentially as outlined for the MMP1 ELISA. In brief, 384-well white Greiner plates (Lumitrac 600) are coated overnight at 4° C. with 40 μl of anti-hTIMP2 capture antibody (Cat. No. MAB9711; R&D systems) at 1 μg/ml in PBS. Blocking is done for 4 hours at room temperature with 80 μl 0.1% casein buffer and after a washing step (30 μl EC-buffer), 40 μl of the samples are added to the wells. Plates are incubated overnight at 4° C., and after washing twice with PBST (0.05% Tween-20) and PBS, 40 μl of the biotinylated anti-hTIMP2 detection antibody (100 ng/ml in Buffer C) (Cat. no. BAF971; R&D systems) is added to the wells. After 2 hours of incubation at room temperature, plates are washed again and incubated at room temperature for another 45 minutes after an addition of 40 μl of Streptavidin-HRP conjugate (Cat. No. SNN2004; BioSource) diluted 1/3000 in Buffer C. Finally, plates are washed and 50 μl of BM Chemiluminescence ELISA substrate (POD)(Cat. No. 1582950; Roche Diagnostic) is added to the wells. After 5 minutes incubation at room temperature in the dark, luminescence is quantified on a luminometer (Luminoscan Ascent).


The procedure described above (plate layout, MOI, method for determination of the cutoff) applied for these experiments is identical to the one described for the toxicity measurements (i.e., a primary test with 12 μl infection volume and a retest with 3, 6, 12 μl infection volumes) differing only in that the cutoff applied in the primary test is determined as the average over all samples minus 2 times standard deviation over all samples. In this assay, viruses mediating a reduction of the TIMP2 levels under the cutoff in duplicate for at least one MOI in the retest are considered as influencing the SF general secretion machinery.


From the 339 hits tested in the toxicity and TIMP secretion assay, 16 scored positive in these assays and considered mediating the reduction in cytokine induced MMP1 expression in an a specific way. As such, 323 hits were identified as modulators of cytokine induced MMP1 expression, 16 of which are most preferred hits and listed in Table 1.


We conclude, from this experiment, that the genes associated with these hits and their expressed proteins represent valuable drug targets that are shown by these studies to modulate MMP1 expression in SFs.














TABLE 4





Hit
SEQ KD Targets
SEQ

Protein



No.
19-mer and 21-mer
ID NO
Gene Name
Class
Accession







H46-
TGCCGTGAATGGAGAACAC
253
DGKI
Kinase
NM_004717


001
AATGCCGTGAATGGAGAACAC
254








H46-
TCCTTAGTTCCTCAGAGGC
255
LATS1
Kinase
NM_004690


002
AATCCTTAGTTCCTCAGAGGC
256








H46-
GATTACCTTGCCTGTTGAC
257
PGK1
Kinase
NM_000291


004
AAGATTACCTTGCCTGTTGAC
258








H46-
CTGACTTTGGATTCTGTGC
259
PAK1
Kinase
NM_002576


006
AACTGACTTTGGATTCTGTGC
260








H46-
GACATGCCGCTATTTGAGC
261
PTK7
Kinase
NM_152881; NM_002821;


008
AAGACATGCCGCTATTTGAGC
262


NM_152880; NM_152883;







NM_152882





H46-
AGTGAGACACTGACAGAAC
263
JAK2
Kinase
NM_004972


009
AAAGTGAGACACTGACAGAAC
264








H46-
CTGCATGCTGAATGAGAAC
131
AXL
Kinase
NM_021913; NM_001699;


010
AACTGCATGCTGAATGAGAAC
234


SK044





H46-
GGCCTTGAAAGGACAGTAC
265
UCK1
Kinase
NM_031432


011
AAGGCCTTGAAAGGACAGTAC
266








H46-
GACTGAGTCAAGTGTCTGC
267
STK31
Kinase
NM_031414; NM_032944


012
AAGACTGAGTCAAGTGTCTGC
268











SgK396
Kinase
SK652





H46-
TGGTGGCTACAACCAACTC
269
MAP4K5
Kinase
NM_198794; NM_006575


013
AATGGTGGCTACAACCAACTC
270








H46-
TGTGACTGTGGGAGTGGAC
271
HK1
Kinase
NM_033498; NM_033500;


014
AATGTGACTGTGGGAGTGGAC
272


NM_033496; NM_000188;







NM_033497





H46-
ACTGCTGCTGTATGTGGAC
273
AK1
Kinase
NM_000476


015 
ACACTGCTGCTGTATGTGGAC
274








H46-
CTGTGTTGATTTCTCTGCC
275
DNAH8
Protease
NM_001371


016
AACTGTGTTGATTTCTCTGCC
276








H46-
TCCCTTGTATCCGGACTTC
277
GALK2
Kinase
NM_002044;


017
AATCCCTTGTATCCGGACTTC
278


NM_001001556





H46-
CAAGAGCTGGTACCGCGTC
279
CTK
Kinase
SK418


019
AACAAGAGCTGGTACCGCGTC
280











MATK
Kinase
NM_002378; NM_139354;







NM_139355





H46-
GCTCCTCAGAGTGCAGCAC
134
SCN9A
Ion Channel 
NM_002977


020
AAGCTCCTCAGAGTGCAGCAC
237








H46-
CCTGAATGTGACTGTGGAC
140
DGKB
Kinase
NM_145695; NM_004080


021
AACCTGAATGTGACTGTGGAC
243











INCENP
Not drugable
NM_020238





H46-
TGTGGTGGCCTACATTGGC
281
GCK
Kinase
SK048


022
AATGTGGTGGCCTACATTGGC
282











MAP4K2
Kinase
NM_004579





H46-
GAACCTCACTTCCAGTCAC
283
MPP6
Kinase
NM_016447


024
AAGAACCTCACTTCCAGTCAC
284








H46-
ATCAGTGGGCTGTGGATTC
285
MASTL
Kinase
NM_032844


025
AAATCAGTGGGCTGTGGATTC
286








H46-
CATTGTGGACTTTGCTGGC
287
IRAK1
Kinase
NM_001569


026
AACATTGTGGACTTTGCTGGC
288











AF346607
Kinase
AF346607





H46-
ATGACTGAGCCACACAAAC
289
LOC160848
Kinase
XM_090535


028
ACATGACTGAGCCACACAAAC
290











MAPK6
Kinase
NM_002748





H46-
CATTGTGGACGTGCTGGCC
128
KCNF1
Ion Channel
NM_002236


030
AACATTGTGGACGTGCTGGCC
231








H46-
CTTTGCACCTATCTCTGAC
291
NEK8
Kinase
SK476; NM_178170


033
ACCTTTGCACCTATCTCTGAC
292








H46-
TGCGCTTTGCCAAGATGCC
293
BRD4
Not drugable 
NM_014299; SK763;


034
AATGCGCTTTGCCAAGATGCC
294


NM_058243





H46-
ACATGTGTTCCAGAAGGAC
295
AMACO
Not drugable 
NM_198496


035
GCACATGTGTTCCAGAAGGAC
296











LOC118680
Kinase
XM_061091





H46-
ACCTGGCTTCAACATCAGC
297
LOC163720
Cytochrome
XM_089093


036
AAACCTGGCTTCAACATCAGC
298

P450









CYP4Z1
Cytochrome
NM_178134






P450






H46-
TGGATGACCACAAGCTGTC
299
ATP1A2
Ion Channel 
NM_000702


038
AATGGATGACCACAAGCTGTC
300








H46-
ACATGCACTGCAGAAGGCC
301
CACNA1E
Ion Channel
NM_000721


039
AAACATGCACTGCAGAAGGCC
302








H46-
ATGTGCCATCCAGTTGTGC
303
LOC136062
Kinase
XM_069683


040
AAATGTGCCATCCAGTTGTGC
304








H46-
CTATGTGGACTGGATCAAC
305
HGFAC
Protease
NM_001528


041
AACTATGTGGACTGGATCAAC
306








H46-
TGAAGGCCTCACGTGTGTC
307
CTRL
Protease
NM_001907


042
ACTGAAGGCCTCACGTGTGTC
308








H46-
CATGAAGGCCCGAAACTAC
309
MAPK3
Kinase
NM_002746; XM_055766


043
AACATGAAGGCCCGAAACTAC
310








H46-
AGATGGCCACAAACCTGCC
311
ACVR2
Kinase
NM_001616


044
AAAGATGGCCACAAACCTGCC
312








H46-
ATGAAGAGCAAGGCATTCC
313
DCK
Kinase
NM_000788


045
AAATGAAGAGCAAGGCATTCC
314








H46-
CATCCCACACAAGTTCAGC
315
PRKCH
Kinase
NM_006255


046
AACATCCCACACAAGTTCAGC
316








H46-
CTCATGTTTACCATGGTCC
317
CHRNB1
Ion Channel
NM_000747


047
ACCTCATGTTTACCATGGTCC
318








H46-
ATTGTTCTCCCAGATGGCC
319
CYP4F12
Cytochrome
NM_023944


049
ACATTGTTCTCCCAGATGGCC
320

P450






H46-
TTCATGAGAAGACTGGTGC
321
NRK
Kinase
NM_198465


050
ACTTCATGAGAAGACTGGTGC
322








H46-
TGACTGCACCTATGAGAGC
323
LOC126392
Kinase
XM_065057


051
CCTGACTGCACCTATGAGAGC
324








H46-
TTCAGTGCATTCAAGGACC
325
FAD104
Receptor
NM_022763


053
ACTTCAGTGCATTCAAGGACC
326











LOC205527
Receptor
XM_116009





H46-
ACGTGCCAGTTTCTTCTCC
327
LOC200959
Ion Channel
XM_116036


054
AAACGTGCCAGTTTCTTCTCC
328








H46-
GTGTATCTTATGCCTACAC
329
DOK5L
Receptor
NM_152721


055
ACGTGTATCTTATGCCTACAC
330








H46-
AGCTGCTGGAGATGTTATC
331
UQCRC2
Protease
NM_003366


056
ACAGCTGCTGGAGATGTTATC
332








H46-
CAAGTGACTGTGATTGCCC
333
COL7A1
Not drugable 
NM_000094


058
ACCAAGTGACTGTGATTGCCC
334








H46-
TTCAGTGCCTAACAGTGGC
335
CDC7
Kinase
NM_003503


060
ACTTCAGTGCCTAACAGTGGC
336








H46-
CTTGATCATGATGGCCTTC
337
PPAP2B
Phosphatase
NM_177414; NM_003713


061
ACCTTGATCATGATGGCCTTC
338








H46-
ACCCGGCACCGTGTACTTC
339
CRLF1
Receptor
NM_004750


062
AAACCCGGCACCGTGTACTTC
340








H46-
CTACTGATGTCCCTGATAC
341
PPP1CB
Phosphatase
NM_206877; NM_206876;


063
ACCTACTGATGTCCCTGATAC
342


NM_002709





H46-
TTTCTGTGAGCTGTGAAAC
343
SCYL2
Kinase
SK475


064
ACTTTCTGTGAGCTGTGAAAC
344











FLJ10074
Kinase
NM_017988





H46-
TATACTGGAGCCTGTAACC
345
TNFRSF10D
Drugable/Secreted
NM_003840


066
AATATACTGGAGCCTGTAACC
346








H46-
TTTGTGGACTCCTACGATC
347
RHEB
Enzyme
NM_005614


067
AATTTGTGGACTCCTACGATC
348








H46-
GGAGACAAGTTCGCATGTC
349
RASGRP1
Drugable/Secreted
NM_005739


068
AAGGAGACAAGTTCGCATGTC
350








H46-
CGGCTGGTTCACCATGATC
351
SIAT7D
Enzyme
NM_175039; NM_014403;


070
ACCGGCTGGTTCACCATGATC
352


NM_175040





H46-
GTATTCTGTACACCCTGGC
353
RDH11
Enzyme
NM_016026


071
ACGTATTCTGTACACCCTGGC
354








H46-
GACTGCTAGACAAGACGAC
355
NQO3A2
Enzyme
NM_016243


072
ACGACTGCTAGACAAGACGAC
356








H46-
CCTGTGTTCAGCTCTGGAC
357
IKBKAP
Kinase
NM_003640


073
AACCTGTGTTCAGCTCTGGAC
358








H46-
ACATTCTCCTGGCCAGGTC
359
ARHGAP21
Drugable/Secreted
NM_020824


075
AAACATTCTCCTGGCCAGGTC
360








H46-
GACTGAGAACCTTGATGTC
361
ATP1B2
Ion Channel
NM_001678


078
AAGACTGAGAACCTTGATGTC
362








H46-
TATTGCCCTGTTAGTGTTC
363
LOC201803
Enzyme
XM_116196


079
CCTATTGCCCTGTTAGTGTTC
364








H46-
TGTGGAGTCTTTGTGCTCC
365
LOC254017
Protease
XM_172342


080
ACTGTGGAGTCTTTGTGCTCC
366











SENP5
Protease
NM_152699





H46-
TTCCTGTGGAATGACTGTC
367
LRRK1
Kinase
NM_024652


081
AATTCCTGTGGAATGACTGTC
368








H46-
ACGTGGTTGGAAGCTAACC
369
ABCB1
Transporter
NM_000927


085
ACACGTGGTTGGAAGCTAACC
370








H46-
CAAGTCTACACGGGCTCAC
371
HSD17B12
Enzyme
NM_016142


086
AACAAGTCTACACGGGCTCAC
372








H46-
TCCTGGAGGCTATCCGCAC
373
DNMT3B
Enzyme
NM_175850; NM_175848;


087
AATCCTGGAGGCTATCCGCAC
374


NM_006892; NM_175849





H46-
TCGGCACAACAATCTAGAC
375
IDH3B
Enzyme
NM_006899; NM_174855;


088
ACTCGGCACAACAATCTAGAC
376


NM_174856





H46-
ACGCGCCTTAATTTAGTCC
377
LDHL
Enzyme
NM_033195


089
AAACGCGCCTTAATTTAGTCC
378








H46-
AGCATGATGATAGTTCCTC
379
TOP2B
Enzyme
NM_001068


090
CCAGCATGATGATAGTTCCTC
380








H46-
TGGGCACACATCTCCAGTC
381
LOC219756
Kinase
XM_166676


091
ACTGGGCACACATCTCCAGTC
382








H46-
ACCGTGGAAGGCCTATCGC
383
CYP24A1
Cytochrome
NM_000782


092
AAACCGTGGAAGGCCTATCGC
384

P450






H46-
TGAATCCACAGCTCTACTC
385
LOC400301
Kinase
XM_375150


093
AATGAATCCACAGCTCTACTC
386











LOC256238
Kinase
XM_171744





H46-
CAAGTACAACAAGGACTCC
387
CYP46A1
Cytochrome
NM_006668


094
ACCAAGTACAACAAGGACTCC
388

P450






H46-
CATTCAGTGCTGCCTTCAC
389
MGC16169
Kinase
NM_033115


095
TCCATTCAGTGCTGCCTTCAC
390











TBCK
Kinase
SK664





H46-
GATGGCCATCCTGCAGATC
391
TIF1b
Kinase
SK784


096
AAGATGGCCATCCTGCAGATC
392











TRIM28
Kinase
NM_005762





H46-
TCATTGTGCCATCCCAAAC
393
LOC169505
Kinase
XM_095725


097
AATCATTGTGCCATCCCAAAC
394








H46-
AACCTGGCCCTAATGCTGC
395
LOC220075
Receptor
XM_169246


098
TCAACCTGGCCCTAATGCTGC
396








H46-
CTGACTCACCTGGTACTGC
397
KCTD3
Ion Channel
NM_016121


099
AACTGACTCACCTGGTACTGC
398








H46-
TGCTGTGTGCTGGAGTACC
399
HAT
Protease
NM_004262


100
AATGCTGTGTGCTGGAGTACC
400








H46-
GCATTGCTATTGCTCGGGC
401
ABCB11
Transporter
NM_003742


101
ACGCATTGCTATTGCTCGGGC
402








H46-
TGTGGAGTGTGAGAAGCTC
403
LOC131136
Transporter
XM_067219


102
AATGTGGAGTGTGAGAAGCTC
404








H46-
TCTGTGGGCCTCCTATTCC
405
APRT
Enzyme
NM_000485


104
ACTCTGTGGGCCTCCTATTCC
406








H46-
TGCCGGCATCTCAACGTTC
407
BDH
Enzyme
NM_203315; NM_203314;


105
AATGCCGGCATCTCAACGTTC
408


NM_004051





H46-
TGTGACTGTCCTAGTTGCC
409
UGT2A1
Enzyme
NM_006798


106
AATGTGACTGTCCTAGTTGCC
410








H46-
AGCTGTGTTCATCAACATC
411
GRHPR
Enzyme
NM_012203


107
ACAGCTGTGTTCATCAACATC
412








H46-
TGGGCAGAATGTCCTGGAC
413
TTPR3
Ion Channel
NM_002224


108
AATGGGCAGAATGTCCTGGAC
414








H46-
CACATGCTCAACATGGTGC
415
RASGRP4
Drugable/Secreted
NM_170602; NM_170604;


110
ACCACATGCTCAACATGGTGC
416


NM_052949; NM_170603





H46-
CAGTGACTAACCAGAACTC
417
LOC402692
Enzyme
XM_380039


112
ACCAGTGACTAACCAGAACTC
418








H46-
TACATGTGTCGGAGGTTTC
419
GNPNAT1
Enzyme
NM_198066


113
ACTACATGTGTCGGAGGTTTC
420








H46-
GATGTAGATACCACAAGGC
421
LOC160469
Enzyme
XM_090320


114
ACGATGTAGATACCACAAGGC
422








H46-
TGGTGGCAAGTCCATCTAC
423
LOC341457
Enzyme
XM_292085


115
ACTGGTGGCAAGTCCATCTAC
424











LOC159692
Enzyme
XM_089755








LOC399780
Enzyme
XM_374813








LOC401706
Enzyme
XM_377240








LOC122552
Enzyme
XM_063182








LOC402644
Enzyme
XM_379998








LOC391807
Enzyme
XM_373092








LOC139318
Enzyme
XM_066617








LOC402252
Enzyme
XM_377934








LOC133419
Enzyme
XM_068341








LOC165317
Enzyme
XM_092514








LOC222940
Enzyme
XM_167261








LOC401859
Enzyme
XM_377444





H46-
ACATGGTCCGAGTAGAAGC
425
LOC167127
Enzyme
NM_174914; XM_094300


116
AAACATGGTCCGAGTAGAAGC
426








H46-
ATGTGTTGTGGAGTCCGCC
427
LOC254142
Enzyme
XM_171406


117
AAATGTGTTGTGGAGTCCGCC
428








H46-
ACTTGGGCTGCAGTGCATC
429
CYP19A1
Cytochrome
NM_000103; NM_031226


118
AAACTTGGGCTGCAGTGCATC
430

P450






H46-
GTGGCCTAACCCGGAGAAC
431
CYP1B1
Cytochrome
NM_000104


119
AAGTGGCCTAACCCGGAGAAC
432

P450






H46-
AGTTCCTGGTGAACATTCC
433
TJP3
Kinase
NM_014428


120
GCAGTTCCTGGTGAACATTCC
434








H46-
GTTGTGTTCCCGAGACTAC
435
FLJ10986
Kinase
NM_018291


121
AAGTTGTGTTCCCGAGACTAC
436








H46-
AGAGCTGTGTCTGAACATC
437
PTHLH
Drugable/Secreted 
NM_002820; NM_198966;


122
AAAGAGCTGTGTCTGAACATC
438


NM_198965; NM_198964





H46-
TGCAAGGTGATCGGCTTCC
439
RASGRF1
Drugable/Secreted
NM_153815; NM_002891


123
AATGCAAGGTGATCGGCTTCC
440








H46-
CGACACCAGAAATTCACTC
441
FGF16
Drugable/Secreted 
NM_003868


125
AACGACACCAGAAATTCACTC
442








H46-
TGTTCTCCATCTTCTTCCC
443
SLC12A3
Transporter
NM_000339


126
AATGTTCTCCATCTTCTTCCC
444








H46-
ATCTGTGTGGCAGGTTACC
445
MTHFR
Enzyme
NM_005957


127
ACATCTGTGTGGCAGGTTACC
446








H46-
TAGCGCTCCTTTCCGTAAC
138
NQO2
Enzyme
NM_000904


129
ACTAGCGCTCCTTTCCGTAAC
241








H46-
ACTAGTCCAGTGGATCTCACC
447
LOC222593
Enzyme
XM_167104


130
ACTAGTCCAGTGGATCTCACC
448








H46-
TTCCAGGGATTACTCCAAC
449
CYP20A1
Cytochrome
NM_020674; NM_177538


131
AATTCCAGGGATTACTCCAAC
450

P450






H46-
ACTGACTCCAGTTTCTGCC
451
CYP20A1
Cytochrome
NM_020674; NM_177538


132
AAACTGACTCCAGTTTCTGCC
452

P450






H46-
TGGATGACTTCAGCTCTAC
453
GGTLA4
Enzyme
NM_080920; NM_178312;


133
AATGGATGACTTCAGCTCTAC
454


NM_178311





H46-
GCAGTCGACTCCAGAAGAC
455
USP47
Protease
NM_017944


135
AAGCAGTCGACTCCAGAAGAC
456








H46-
TGACTCTGCTAGGACGGTC
457
IL1R2
Receptor
NM_004633; NM_173343


136
AATGACTCTGCTAGGACGGTC
458








H46-
TGTTCTTTGGCTCTGCAGC
129
SLC9A8
Ion Channel
NM_015266


137
AATGTTCTTTGGCTCTGCAGC
232








H46-
AGAATCTGCAGAAAGCCAC
459
LOC159948
GPCR
XM_089955


139
CCAGAATCTGCAGAAAGCCAC
460








H46-
TTCCACAACGTGTGCCTGC
461
DUSP14
Phosphatase
NM_007026


140
AATTCCACAACGTGTGCCTGC
462








H46-
TGGGCGGGTAGATGGAATC
463
NAP1
Protease
NM_004851


141
ACTGGGCGGGTAGATGGAATC
464








H46-
GTGACTCATGATCTAGCCC
465
GGTL3
Protease
NM_052830; NM_178026


142
ACGTGACTCATGATCTAGCCC
466








H46-
GGCTGAAGCAGACACAGAC
467
RNPEPL1
Protease
NM_018226


144
AAGGCTGAAGCAGACACAGAC
468








H46-
GGACATTCAGATAGCGCTC
469
DUT
Enzyme
NM_001948


146
ACGGACATTCAGATAGCGCTC
470








H46-
GATGTTCTGTGTGGGCTTC
471
TRY6
Protease
NM_139000


147
AAGATGTTCTGTGTGGGCTTC
472








H46-
TTTGTGGACCTGAGCTTCC
473
XPNPEP1
Protease
NM_006523; NM_020383


148
ACTTTGTGGACCTGAGCTTCC
474








H46-
TACTGGCTTCGCTTTCATC
475
LOC119714
Protease
XM_061638


149
ACTACTGGCTTCGCTTTCATC
476








H46-
TGGCTCTTACGTGATTCAC
477
LOC150426
Protease
XM_086914


150
AATGGCTCTTACGTGATTCAC
478








H46-
CTATGTTCAGCCCAAGCGC
479
LOC147221 
Protease
XM_102809


151
AACTATGTTCAGCCCAAGCGC
480








H46-
GGGAAGACAGAGCTGTTTC
481
LOC124739 
Protease
XM_058840


152
AAGGGAAGACAGAGCTGTTTC
482











USP43
Protease
XM_371015





H46-
CCTGAGTTCCCAGCATGTC
483
BAA03370
Protease
3392317CA2


154
ACCCTGAGTTCCCAGCATGTC
484











LOC205016 
Protease
XM_114823








LOC388743 
Protease
XM_371344





H46-
TGGCCTTGGAATCTCTTCC
485
LOC206841
Protease
XM_116753


155
ACTGGCCTTGGAATCTCTTCC
486








H46-
TGCTGTATCCTTCCTGGAC
487
LOC256618 
Protease
XM_171760


157
AATGCTGTATCCTTCCTGGAC
488








H46-
TAACCCTGCCTACTACGTC
489
INPPL1
Phosphatase
NM_001567


159
AATAACCCTGCCTACTACGTC
490








H46-
TGCTGTGTGGTCTACCGAC
130
ARAF1
Kinase
NM_001654


160
ACTGCTGTGTGGTCTACCGAC
233








H46-
TCGGGCGGATCTGCTTATC
491
SLC26A6
Transporter
NM_134263; NM_134426;


161
AATCGGGCGGATCTGCTTATC
492


NM_022911





H46-
TGCTGCTGTCAACCCTTTC
493
DHRS4
Enzyme
NM_021004


163
AATGCTGCTGTCAACCCTTTC
494











DHRS4L2
Enzyme
NM_198083





H46-
TCGTGGAAGGACTCATGGC
495
LOC125850 
Enzyme
XM_064826


164
AATCGTGGAAGGACTCATGGC
496








H46-
ACCTGGTAACCAAGCCTGC
497
LOC133486 
Enzyme
XM_068376


166
ACACCTGGTAACCAAGCCTGC
498








H46-
TGCTCCATCTCCCTGAGTC
499
DRD3
GPCR
NM_033660


167
TCTGCTCCATCTCCCTGAGTC
500








H46-
GTGGAACACACTTCAGTCC
501
BLP1
GPCR
NM_031940; NM_078473


168
ACGTGGAACACACTTCAGTCC
502








H46-
TGGAAGAGTCTGTTGATCC
503
GPR139
GPCR
NM_001002911


169
ACTGGAAGAGTCTGTTGATCC
504








H46-
TTCTTGACCAGTGATGGGC
505
LOC163107
GPCR
XM_092005


170
ACTTCTTGACCAGTGATGGGC
506








H46-
TGGGCAAGAAACTAAGCCC
507
LOC255993 
Enzyme
XM_171939


171
ACTGGGCAAGAAACTAAGCCC
508








H46-
CAGATGTATCTGCCAGGAC
509
LOC254796 
Kinase
XM_172160


172
TCCAGATGTATCTGCCAGGAC
510








H46-
GGCCTGCCAATTTAAGCGC
511
ATP1B4
Ion Channel
NM_012069


174
AAGGCCTGCCAATTTAAGCGC
512








H46-
GCGTGTCCTGACTTCTGTC
513
KSR
Kinase
XM_290793; XM_034172


175
AAGCGTGTCCTGACTTCTGTC
514











KSR1
Kinase
SK205





H46-
CACATACCCGGGACACTAC
515
PDHX
Enzyme
NM_003477


176
AACACATACCCGGGACACTAC
516








H46-
TGTAGTTGCAAACCCAGGC
517
VNIR1
GPCR
NM_020633


177
ACTGTAGTTGCAAACCCAGGC
518








H46-
ATGGGCACCATGTTTGAAC
519
NR1T3
NIIR
NM_005122


178
ACATGGGCACCATGTTTGAAC
520








H46-
TCTGCTGGTCATAGCAGCC
521
EDG4
GPCR
NM_004720


179
AATCTGCTGGTCATAGCAGCC
522








H46-
CATGACTGCTAGTGCTCAC
523
TGFBR2
Kinase
NM_003242


180
AACATGACTGCTAGTGCTCAC
524








H46-
TTGTGCCATCGTGGGCAAC
525
SIAT8B
Enzyme
NM_006011


182
ACTTGTGCCATCGTGGGCAAC
526








H46-
ATCCACAGGAAATGAAGAC
527
FOLH1
Protease
NM_004476


183
ACATCCACAGGAAATGAAGAC
528











PSMAL/GCP
Protease
NM_I53696





III







H46-
CGTCATGATCGCGCTCACC
529
HTR5A
GPCR
NM_024012


185
AACGTCATGATCGCGCTCACC
530








H46-
CCAGAAATCACTCCACTGC
531
F2RL2
GPCR
NM_004101


186
AACCAGAAATCACTCCACTGC
532








H46-
GCTGCTCACCAGGAACTAC
533
GPR14
GPCR
NM_018949


187
ACGCTGCTCACCAGGAACTAC
534








H46-
CATCCCACTCAAGATGCAC
535
GPR80
GPCR
NM_080818


188
AACATCCCACTCAAGATGCAC
536








H46-
GCCCATTGAGACACTGATC
133
NR2F6
NHR
XM_373407; NM_005234


189
ACGCCCATTGAGACACTGATC
236








H46-
CACACTAGATGCCATGATC
537
AGA
Protease
NM_000027


190
ACCACACTAGATGCCATGATC
538








H46-
GCCGAAGACCTGTTCTATC
539
ACE2
Protease
3699373CA2; NM_021804


191
AAGCCGAAGACCTGTTCTATC
540








H46-
GCAAGACGGATATGCATGC
541
ADAM23
Protease
NM_003812


192
AAGCAAGACGGATATGCATGC
542








H46-
GAGCAGATGTGGACCATGC
543
MMP1
Protease
NM_002421


193
AAGAGCAGATGTGGACCATGC
544








H46-
TGGGCTTGAAGCTGCTTAC
545
MMP1
Protease
NM_002421


194
AATGGGCTTGAAGCTGCTTAC
546








H46-
GGTATTGGAGGAGATGCTC
547
MMP8
Protease
NM_002424


195
AAGGTATTGGAGGAGATGCTC
548








H46-
CGTGAATGCCGAGTTGGGC
549
MGC13186
Protease
NM_032324


196
AACGTGAATGCCGAGTTGGGC
550








H46-
TCACTGTGGAGACATTTGC
551
ADAM9
Protease
NM_003816


197
AATCACTGTGGAGACATTTGC
552








H46-
GTGGGCTTCATCAACTACC
553
SLC7A8
Transporter
NM_012244; NM_182728


199 
ACGTGGGCTTCATCAACTACC
554








H46-
CCTGACCCTGGAGCACATC
555
LCAT
Enzyme
NM_000229


200
AACCTGACCCTGGAGCACATC
556








H46-
GGACAACATAACGATGCAC
557
GPD2
Enzyme
NM_000408


201
ACGGACAACATAACGATGCAC
558








H46-
ATCAGTGAGGCATTTGACC
559
ADH4
Enzyme
NM_000670


202
AAATCAGTGAGGCATTTGACC
560








H46-
TGGCTGGAGGACAAGTTCC
561
GSTT2
Enzyme
NM_000854


203
AATGGCTGGAGGACAAGTTCC
562








H46-
ATGGCCATTGCCATGGCTC
563
IMPDH1
Enzyme
NM_183243


204
ACATGGCCATTGCCATGGCTC
564











IMPDH1
Enzyme
NM_000883





H46-
CTGGATGCTGCGCAAACAC
565
GCNT1
Enzymes for
NM_001490


205
AACTGGATGCTGCGCAAACAC
566

KD






H46-
TGCTGCTTATCAACAACGC
567
SPR
Enzyme
NM_003124


206
ACTGCTGCTTATCAACAACGC
568








H46-
ACATGCCACAGGAAACTAC
569
DPM1
Enzyme
NM_003859


207 
AAACATGCCACAGGAAACTAC
570








H46-
CGAGTCCTAGGCTACATCC
571
ALDH1B1
Enzyme
NM_000692


208
AACGAGTCCTAGGCTACATCC
572








H46-
TGTGCCCAACGCCACCATC
573
ABCA3
Transporter
NM_001089


209
AATGTGCCCAACGCCACCATC
574








H46-
GCCTCGGACTTTCATCATC
575
ABCD2
Transporter 
NM_005164


210
AAGCCTCGGACTTTCATCATC
576








H46-
TTGAACCCATCAGGCACTC
577
ADAM21
Protease
NM_003813


211
AATTGAACCCATCAGGCACTC
578








H46-
TGAACCCTGTTATCTACAC
579
ADRA2B
GPCR
NM_000682


213
ACTGAACCCTGTTATCTACAC
580








H46-
CTTCCTGAAGACCAGGTTC
581
AF200815
Kinase
AF200815


214
AACTTCCTGAAGACCAGGTTC
582











STK36
Kinase
XM_050803; NM_015690





H46-
ATGGACATTGACGTGATCC
583
ALPI
Phosphatase 
NM_001631


217
ACATGGACATTGACGTGATCC
584











ALPP
Phosphatase 
NM_001632





H46-
TTTCCCTTCAAGGCCCTGC
585
CAD35089
Drugable/Secreted
3663102CA2


219
ACTTTCCCTTCAAGGCCCTGC
586











ART5
Enzyme
NM_053017





H46-
AAGTCTCAAGAGTCACAGC
587
BAP1
Protease
NM_004656


220
ACAAGTCTCAAGAGTCACAGC
588








H46-
AATAGCAAGAATGTGTGCC
589
PANK2
Kinase
NM_153641; NM_153638;


222
TCAATAGCAAGAATGTGTGCC
590


NM_153637; NM_024960;







NM_153639; NM_153640





H46-
AGAAGAAGGTGGTGTGGAC
591
CARD14
Kinase
NM_024110


223
GCAGAAGAAGGTGGTGTGGAC
592








H46-
TGCCGTGGTGCACTATAGC
593
CELSR2
GPCR
NM_001408


224
AATGCCGTGGTGCACTATAGC
594








H46-
AGTGTCCACTCAGGAACTC
595
CHEK2
Kinase
NM_145862; NM_007194


225
ACAGTGTCCACTCAGGAACTC
596








H46-
GTGTTCCTTCAGACTCTTC
137
CHRNA5
Ion Channel
NM_000745


226
ACGTGTTCCTTCAGACTCTTC
240








H46-
TGCATGATGTCGGTCACCC
597
COX10
Enzyme
NM_0121303


227
ACTGCATGATGTCGGTCACCC
598








H46-
CTGTGCCTGCCATTACAAC
599
CTGF
Drugable/Secreted
NM_001901


228
ACCTGTGCCTGCCATTACAAC
600








H46-
GACTACAGTGATTGTCGGC
601
CYP17A1
Cytochrome
NM_000102


229
AAGACTACAGTGATTGTCGGC
602

P450






H46-
CTCTGTGTTCCACTTCGGC
603
DPYD
Enzyme
NM_000110


232
AACTCTGTGTTCCACTTCGGC
604








H46-
GGAGATCGTGCTGGAGAAC
605
FGF17
Drugable/Secreted
NM_003867


233
ACGGAGATCGTGCTGGAGAAC
606








H46-
CGTGGCCTACATCATCATC
607
GPR108
GPCR
XM_290854


235
AACGTGGCCTACATCATCATC
608








H46-
TGCAGCCAGTGGAATGTCC
609
GPRC5D
GPCR
NM_018654


236
ACTGCAGCCAGTGGAATGTCC
610








H46-
GTATGGCATGCAGCTGTAC
611
GRB14
Drugable/Secreted
NM_004490


237
AAGTATGGCATGCAGCTGTAC
612








H46-
GAATGGCTTTGCTGTGGTC
613
HDAC4
Enzyme
NM_006037


238
AAGAATGGCTTTGCTGTGGTC
614








H46-
ATGTTCCAGGAGATCGTCC
615
CNDP1
Protease
NM_032649


239
AAATGTTCCAGGAGATCGTCC
616








H46-
ACATTCAGCTGTGAACTCC
617
HSD11B2
Enzyme
NM_000196


240
ACACATTCAGCTGTGAACTCC
618








H46-
CAAGCCCTTCCGTGTACTC
619
INPP5A
Phosphatase 
NM_005539


241
AACAAGCCCTTCCGTGTACTC
620








H46-
CCAGCATCCTTTGCATTAC
621
ITGAE
Drugable/Secreted
NM_002208


242
ACCCAGCATCCTTTGCATTAC
622








H46-
TGCAGTCAGTTGTCCATAC
623
KCNA4
Ion Channel 
NM_002233


243
AATGCAGTCAGTTGTCCATAC
624








H46-
AGGCCAATCCTGGTAGCAC
625
KIAA0669
Receptor
NM_014779


245
ACAGGCCAATCCTGGTAGCAC
626








H46-
ATCCTGGGCTATTGGACTC
627
LDHA
Enzyme
NM_005566


246
ACATCCTGGGCTATTGGACTC
628








H46-
CATGGCCTGTGCAATTATC
629
LOC343066 
Enzyme
XM_291392


247
ACCATGGCCTGTGCAATTATC
630








H46-
GGCTGGTATACAGGAACAC
631
LOC128183
Enzyme
XM_060863


248
AAGGCTGGTATACAGGAACAC
632








H46-
ACATGCCATTACCAGCATC
633
FLJ16046
Protease
NM_207407


249
AAACATGCCATTACCAGCATC
634











LOC389208
Protease
XM_371695








LOC133177
Protease
XM_068225





H46-
TTGCTGCTATGTCAGATCC
635
LOC138967
Cytochrome
XM_071222


250
AATTGCTGCTATGTCAGATCC
636

P450






H46-
ACGTGGACCAAGTCATGCC
637
DUSP18
Phosphatase
NM_152511


251
ACACGTGGACCAAGTCATGCC
638








H46-
TTGGATCCTAATGAGCTGC
639
IMP5
Protease
NM_175882


253
ACTTGGATCCTAATGAGCTGC
640








H46-
GTCTTGTGTCAGAATTTCC
641
LOC163107
GPCR
XM_092005


254
ACGTCTTGTGTCAGAATTTCC
642








H46-
AGTAGGCAACGACAGCAGC
643
LOC222656
GPCR
XM_167080


256
AAAGTAGGCAACGACAGCAGC
644








H46-
CTAAGGAGGCTCGGAAATC
645
LOC256669
Kinase
XM_171416


259
GCCTAAGGAGGCTCGGAAATC
646








H46-
GGCTCTGTCAAGGCCATTC
647
LOC257260
Protease
XM_174353


260
TCGGCTCTGTCAAGGCCATTC
648








H46-
TAATGACTTTGGCGCTGTC
649
ADCK1
Kinase
SK401; NM_020421


261
CCTAATGACTTTGGCGCTGTC
650








H46-
CTACATGGACCGCTTCACC
651
SPINL
Transporter
NM_032038


262
AACTACATGGACCGCTTCACC
652








H46-
TAGCCAAGAGTTCAGCCCC
653
MAP3K14
Kinase
NM_003954


263
AATAGCCAAGAGTTCAGCCCC
654








H46-
CTCCACAAACTGATCAGCC
655
MAP3K14
Kinase
NM_003954


264
AACTCCACAAACTGATCAGCC
656








H46-
GACTGTGAGCTGAAGATCC
141
MAPK12
Kinase
NM_002969


265
AAGACTGTGAGCTGAAGATCC
244








H46-
ACCTGAAGAAAGGGAGAGC
657
MIDORI
Kinase
NM_020778; XM_057651


266
ACACCTGAAGAAAGGGAGAGC
658








H46-
AACCCTATGCTGCCTATGC
659
NAALADL1
Protease
NM_005468


269
ACAACCCTATGCTGCCTATGC
660








H46-
TGGCACTTCGGGCAATAAC
139
NQO2
Enzyme
NM_000904


271
ACTGGCACTTCGGGCAATAAC
242








H46-
GAAGTTCATCAGCGCCATC
661
NTSR1
GPCR
NM_002531


272
AAGAAGTTCATCAGCGCCATC
662








H46-
AGCTGCCTGGAAGCATTAC
663
PDK1
Kinase
NM_002610


273
AAAGCTGCCTGGAAGCATTAC
664








H46-
CAGTGTTACACGGCTTTCC
665
PIK3C3
Kinase
NM_002647


275
AACAGTGTTACACGGCTTTCC
666








H46-
GACTGAATCAGGCCTTCCC
667
PPIH
Enzyme
NM_006347


278
AAGACTGAATCAGGCCTTCCC
668








H46-
TGCCTGGGACCAGAGCTTC
669
PKN1
Kinase
SK317


279
AATGCCTGGGACCAGAGCTTC
670











PRKCL1
Kinase
NM_213560; NM_002741





H46-
GTCCAAGATGGAGCTACAC
671
PSMB10
Protease
NM_002801


280
GCGTCCAAGATGGAGCTACAC
672








H46-
ATATCATGTGAACCTCCTC
673
PSMB2
Protease
NM_002794


281
CCATATCATGTGAACCTCCTC
674








H46-
CGTCGTCCAAAGCAGAAGC
675
PTGIR
GPCR
NM_000960


282
AACGTCGTCCAAAGCAGAAGC
676








H46-
TGTAGTGCAGGCATTGGGC
677
PTPN2
Phosphatase 
NM_080423; NM_002828;


283
ACTGTAGTGCAGGCATTGGGC
678


NM_080422





H46-
TGTGGGAGAACTGAAGTCC
679
PTPN4
Phosphatase 
NM_002830


284
ACTGTGGGAGAACTGAAGTCC
680








H46-
GAAGAACAGCAGCCTGGAC
681
RASD1
Enzyme
NM_016084


285
AAGAAGAACAGCAGCCTGGAC
682








H46-
GATGGACTCAGGTGGACTC
683
SENP3
Protease
NM_015670


286
AAGATGGACTCAGGTGGACTC
684








H46-
TGCTGCATCCGACAGATCC
685
DustyPK
Kinase
NM_015375; NM_199462


287
AATGCTGCATCCGACAGATCC
686








H46-
TTTCCAGGTCATCTGCTCC
687
SLC28A2
Transporter 
NM_004212


288
ACTTTCCAGGTCATCTGCTCC
688








H46-
TTCCAGGTCCTGAAGCGAC
689
SLC6A2
Transporter 
NM_001043


289
ACTTCCAGGTCCTGAAGCGAC
690








H46-
TTGTGGAGAGCTCGAATTC
691
SLC8A1
Ion Channel 
NM_021097


290
ACTTGTGGAGAGCTCGAATTC
692








H46-
ATGGCCAGCAACCTGATGC
693
GPR124
GPCR
NM_032777


291
ACATGGCCAGCAACCTGATGC
694








H46-
TTCTCAGGCACCCTCTACC
695
TLK1
Kinase
XM_002626; AB004885;


292
AATTCTCAGGCACCCTCTACC
696


SK373; NM_012290





H46-
GTGCTGGATTCTGCCATGC
697
TPST1
Enzyme
NM_003596


293
AAGTGCTGGATTCTGCCATGC
698








H46-
GTGTGTTAGCACGACTTTC
699
TPTE
Ion Channel 
NM_013315; NM_199259


294
AAGTGTGTTAGCACGACTTTC
700








H46-
CATCCTGCTGTCCAACCCC
701
ULK1
Kinase
NM_003565


296
AACATCCTGCTGTCCAACCCC
702








H46-
TGGTGGCAGACATCCCTTC
703
XDH
Enzyme
NM_000379


297
ACTGGTGGCAGACATCCCTTC
704








H46-
GCACAGCACTTCCACAAGC
705
GDF10
Drugable/Secreted
NM_004962


299
ACGCACAGCACTTCCACAAGC
706








H46-
AGGAGTTTGGGAACCAGAC
707
DO
Enzyme
NM_021071


300
ACAGGAGTTTGGGAACCAGAC
708








H46-
CACTGGCATCATCTGTACC
709
LOC339395
Kinase
XM_290872


301
AACACTGGCATCATCTGTACC
710











PKM2
Kinase
NM_182471; NM_002654;







NM_182470





H46-
GTGACTACACAAGGACTCC
711
CCR2
GPCR
NM_000647


322
AAGTGACTACACAAGGACTCC
712








H46-
CTTCTCCTTTGGTGGCTGC
713
HTR4
GPCR
NM_000870


323
AACTTCTCCTTTGGTGGCTGC
714








H46-
AGTGGGTAAAGCCAATGGC
715
RPE
Enzyme
XM_030834; NM_199229;


324
ACAGTGGGTAAAGCCAATGGC
716


NM_006916








LOC90470
Enzyme
XM_031975





H46-
GCTGCTCAGAAACGTTCTC
717
RPE
Enzyme
XM_030834; NM_199229;


325
AAGCTGCTCAGAAACGTTCTC
718


NM_006916








LOC90470
Enzyme
XM_031975





H46-
ACCACACTCACGCAGTATC
719
RHOBTB1
Enzyme
NM_198225; NM_014836


326
ACACCACACTCACGCAGTATC
720








H46-
GTCTGTCTGTAAGAACACC
721
USP31
Protease
NM_020718


328
AAGTCTGTCTGTAAGAACACC
722











K1AA1203
Protease
XM_049683





H46-
GATGTAGAGCTGGCCTACC
723
LOC150537
Kinase
XM_086946


329
AAGATGTAGAGCTGGCCTACC
724











LOC389069
Not drugable
XM_371588








SEPHS1
Kinase
NM_012247





H46-
GGTGAGCGTGGACATCTTC
725
ABCA2
Transporter 
NM_001606; NM_212533


330
AAGGTGAGCGTGGACATCTTC
726








H46-
GTGGAACAAGAGGTACAAC
727
ABCA6
Transporter 
NM_080284; NM_172346


331
AAGTGGAACAAGAGGTACAAC
728








H46-
CGATGGCTTCCACGTCTAC
729
ACVR1
Kinase
NM_001105


332
AACGATGGCTTCCACGTCTAC
730








H46-
GTGGGCAATGAATATGGCC
731
ADORA2B
GPCR
NM_000676


333
AAGTGGGCAATGAATATGGCC
732








H46-
TTCTGTGGTGGTTCTGGTC
733
CCR4
GPCR
NM_005508


334
AATTCTGTGGTGGTTCTGGTC
734








H46-
CTTCATTATCCACAGGGAC
735
CDK10
Kinase
NM_003674; NM_052987;


335
AACTTCATTATCCACAGGGAC
736


NM_052988





H46-
GGTGGAGCACTACCGCATC
737
CSK
Kinase
NM_004383


336
AAGGTGGAGCACTACCGCATC
738








H46-
AAGTTCCCGAACGATCACC
739
ENS000000117094
Protease
ENSG00000117094


338
ACAAGTTCCCGAACGATCACC
740








H46-
CTCTGTGTGCCTGTCGTTC
741
KAZALD1
Drugable/Secreted
NM_030929


339
ACCTCTGTGTGCCTGTCGTTC
742








H46-
TGGCACCTTAACTGGAGTC
743
GPR64
GPCR
NM_005756


341
AATGGCACCTTAACTGGAGTC
744








H46-
GAAGCCTGAAGACACAAAC
136
ILK
Kinase
NM_004517


343
AAGAAGCCTGAAGACACAAAC
239








H46-
TCTTCTCCCAGAGGAAGGC
745
DPP10
Protease
NM_020868


345
AATCTTCTCCCAGAGGAAGGC
746











KIAA1492
Protease
XM_035312





H46-
CTGCTCCAGCATCACTATC
747
KLK10
Protease
NM_002776; NM_145888


346
ACCTGCTCCAGCATCACTATC
748








H46-
ATTCTGTGGGCTCATCACC
749
LIFR
Receptor
NM_002310


347
AAATTCTGTGGGCTCATCACC
750








H46-
GCAGGACTTCAGAACACAC
751
LOC118461
Transporter
XM_060969


348
AAGCAGGACTTCAGAACACAC
752








H46-
AOCCTGGTTCATTCTAAAC
753
LOC150287
Transporter 
XM_086889


349
ACAGCCTGGTTCATTCTAAAC
754








H46-
ACTGTGGGATTGACCAGGC
755
LOC151234
Enzyme
XM_087136


351
ACACTGTGGGATTGACCAGGC
756








H46-
TTCAGAATTCAGGCAGCTC
757
C9orf52
Protease
NM_152574


352
ACTTCAGAATTCAGGCAGCTC
758











LOC158219 
Protease
XM_088514





H46-
ACTGGGCATTTCCTACTGC
759
IMP5
Protease
NM_175882


353
ACACTGGGCATTTCCTACTGC
760








H46-
GCTGTGGGAGAACTATCCC
761
LOC205678
Protease
XM_120320


354
AAGCTGTGGGAGAACTATCCC
762








H46-
ATGGACCTGACCTGCATTC
763
LOC255782
Enzyme
XM_172181


356
TCATGGACCTGACCTGCATTC
764








H46-
TCTTCTCACATGGAAATGC
765
C9orf77
Protease
NM_016014


357
ACTCTTCTCACATGGAAATGC
766








H46-
CCATTCCATGGTGTTTACC
767
MALT1
Protease
NM_006785; NM_173844


358
ACCCATTCCATGGTGTTTACC
768


SEQ ID NOS: 881 & 882





H46-
GCACATGCAGCATGAGAAC
135
MAPK13
Kinase
NM_002754


359
AAGCACATGCAGCATGAGAAC
238








H46-
ACATGGGCTATCTCAAGCC
769
MC2R
GPCR
NM_000529


360
AAACATGGGCTATCTCAAGCC
770








H46-
CTTCCTGTCTCCCTTCTAC
771
SLC22A13
Transporter 
NM_004256


362 
AACTTCCTGTCTCCCTTCTAC
772








H46-
CGTCTCACAGTATGCATTC
773
7050585CA2 
Drugable/Secreted
7050585CA2


363
AACGTCTCACAGTATGCATTC
774











PCTK2
Kinase
NM_002595





H46-
AGCTGCTATCCAACTCACC
775
TP5313
Enzyme
NM_147184; NM_004881


364
ACAGCTGCTATCCAACTCACC
776








H46-
CAACGTGGAGGAGGAGTTC
777
PP1665
PDE
NM_030792


365 
ACCAACGTGGAGGAGGAGTTC
778








H46-
CAGGCCTGTGGAAACATAC
779
PPP2R2A
Phosphatase
NM_002717


366
AACAGGCCTGTGGAAACATAC
780








H46-
TGATGGCCTTTCCCTGTGC
781
PLA1A
Enzyme
NM_015900


368
ACTGATGGCCTTTCCCTGTGC
782








H46-
GACTCTGGGCTGCTCTATC
783
PTPRN
Phosphatase 
NM_002846


369
AAGACTCTGGGCTGCTCTATC
784








H46-
TTCCGTGGCCTGTTCAATC
785
SIAT7B
Enzyme
NM_006456


371
ACTTCCGTGGCCTGTTCAATC
786








H46-
GGGCAACAATGACTGTGAC
787
TEX14
Kinase
NM_031272; NM_198393


372
ACGGGCAACAATGACTGTGAC
788








H46-
ACCTGGCTTCCCTTCCTTC
789
TFR2
Protease
NM_003227


373
ACACCTGGCTTCCCTTCCTTC
790








H46-
CATGCTCAAGGCCATGTTC
791
INFAIP1
Ion Channel
NM_021137


374
ACCATGCTCAAGGCCATGTTC
792








H46-
TCAGTTCCCAGCTCTGCAC
793
TNFSF15
Drugable/Secreted
NM_005118


375
AATCAGTTCCCAGCTCTGCAC
794








H46-
CATCACAATTGGCCATCAC
795
TOP2B
Enzyme
NM_001068


376
TCCATCACAATTGGCCATCAC
796








H46-
TGGAAGATTATCCTGTGTC
797
TRPM8
Ion Channel 
NM_024080


377
ACTGGAAGATTATCCTGTGTC
798








H46-
TACATTCACCCTGTGTGTC
799
F2
Protease
NM_000506


379
ACTACATTCACCCTGTGTGTC
800








H46-
GATGTGCCTGTCCTGTGTC
801
IL1RN
Receptor
NM_000577; NM_173843;


380
AAGATGTGCCTGTCCTGTGTC
802


NM_173842; NM_173841





H46-
ATTTGTGGGCAACTCAGCC
803
LOC133179
Protease
XM__068227


381
AAATTTGTGGGCAACTCAGCC
804








H46-
ATCAGAAGAAAGCCATGAC
805
ATAD1
Enzyme
NM_032810


382
ACATCAGAAGAAAGCCATGAC
806








H46-
CATGTGTGGGAAGTTGTTC
807
ADAM28
Protease
NM_014265; NM_021778


385
ACCATGTGTGGGAAGTTGTTC
808








H46-
TTGCTGAGATGTGTTAGGC
809
LOC145624
Protease
XM_096824


387
CCTTGCTGAGATGTGTTAGGC
810








H46-
CCACCATGCAGACAAGTCC
811
ADAMTS6 
Protease
NM_014273


389
AACCACCATGCAGACAAGTCC
812








H46-
ACTGACCTCAGAGTACCAC
813
ADORA3
GPCR
NM_000677


390
AAACTGACCTCAGAGTACCAC
814








H46-
CGACACAGTGGTGCTCTAC
815
DUSP6
Phosphatase 
NM_022652; NM_001946


392
ACCGACACAGTGGTGCTCTAC
816








H46-
GATCTCCCGCTTCCCGCTC
132
FGFR3
Kinase
NM_000142


393
AAGATCTCCCGCTTCCCGCTC
235








H46-
CGTCTACTCGCTGGCCTTC
817
FZD9
GPCR
NM_003508


394
AACGTCTACTCGCTGGCCTTC
818








H46-
TGCTGGTGCCATTGTTGTC
819
GLS2
Enzyme
NM_138566; NM_013267


395
AATGCTGGTGCCATTGTTGTC
820








H46-
ACAACTCAGAGGGACCTTC
821
GALNT6
Enzyme
NM_007210


396
ACACAACTCAGAGGGACCTTC
822








H46-
AGGTAATGTGGAACACAGC
823
GSR
Enzyme
NM_000637


398
AAAGGTAATGTGGAACACAGC
824








H46-
CAACTCCACACTGGACTTC
825
LOC138685
Drugable/Secreted 
XM_071038


399
ACCAACTCCACACTGGACTTC
826











LOC158017
Drugable/Secreted 
XM_095763








IFNW1
Drugable/Secreted
NM_002177





H46-
TGCAGATGGTTGTGCTCCC
827
IL24
Drugable/Secreted 
NM_006850


400
AATGCAGATGGTTGTGCTCCC
828








H46-
AACATGATATGTGCTGGAC
829
KLK10
Protease
NM_002776; NM_145888


401
ACAACATGATATGTGCTGGAC
830








H46-
AACACGGTGGAGCTGCTGC
831
FLJ37478
Enzyme
NM_178557


404
ACAACACGGTGGAGCTGCTGC
832








H46-
TTCCACAGCATGAACTGGC
833
LOC221757
Kinase
XM_167231


406
AATTCCACAGCATGAACTGGC
834








H46-
AGATGCATCTTCCCTCCAC
835
LOC255449
Protease
XM_171993


407
AAAGATGCATCTTCCCTCCAC
836











LOC442045
Protease
XM_497874





H46-
ATAAGCGGTTATCACTGCC
837
PCTK2
Kinase
NM_002595


409
AAATAAGCGGTTATCACTGCC
838








H46-
AGTCACAATGTCAAGTGAC
839
PDHX
Enzyme
NM_003477


410
ACAGTCACAATGTCAAGTGAC
840








H46-
CTCAACCCAGAACCTGAGC
841
PPP1R12C
Phosphatase
NM_017607


411
ACCTCAACCCAGAACCTGAGC
842








H46-
AGTATCAGGAGCCAATACC
843
PSMA4
Protease
NM_002789


412
ACAGTATCAGGAGCCAATACC
844








H46-
AATCCTGTATTCAAGGCGC
845
PSMB1
Protease
NM_002793


413
ACAATCCTGTATTCAAGGCGC
846








H46-
GAGCGCATCTACTGTGCAC
847
PSMB9
Protease
NM_148954; NM_002800


414
ACGAGCGCATCTACTGTGCAC
848








H46-
ACATGGACGAGTGTCTCAC
849
RYR2
Ion Channel
NM_001035


415
ACACATGGACGAGTGTCTCAC
850








H46-
TCCGAGCGATTTAAGGAAC
851
SHH
Protease
NM_000193


416
ACTCCGAGCGATTTAAGGAAC
852








H46-
TGGAGTCCTTCAAGGCTAC
853
SRD5A2
Enzyme
NM_000348


417
AATGGAGTCCTTCAAGGCTAC
854








H46-
CATCATGGATGAGTGTGGC
855
SV2B
Transporter
NM_014848


418
ACCATCATGGATGAGTGTGGC
856








H46-
ATTCAGCAGAAGCCCAGAC
857
UGT1A6
Enzyme
NM_001072


419
ACATTCAGCAGAAGCCCAGAC
858








H46-
TGAGCCACGGGAATGTGCC
859
ULK2
Kinase
NM_014683


420
AATGAGCCACGGGAATGTGCC
860








H46-
TTCCACCTACCAGTCCACC
861
PGA5
Protease
NM_014224


423
TCTTCCACCTACCAGTCCACC
862








H46-
CAGCACATTCAGCTGCAGC
863
FGF1
Drugable/Secreted 
NM_000800


424
ACCAGCACATTCAGCTGCAGC
864








H46-
ATCATGGCTGTGACCACAC
865
NDUFS2
Enzyme
NM_004550


426
ACATCATGGCTGTGACCACAC
866








H46-
AGTGGCCTTCCTCAGGAAC
867
NR0B2
NHR
NM_021969


427
ACAGTGGCCTTCCTCAGGAAC
868








H46-
AGTTCTACGACTCCAACAC
869
PAK2
Kinase
NM_002577


429
AAAGTTCTACGACTCCAACAC
870









Example 3B
Application of Additional Assays and Exclusion Criteria

The 323 hits identified through the primary “reverse MMP1 assay” are further prioritized using the “reverse collagen degradation assay”, which is based on the observation that the supernatant of RA SFs treated with “TNF□-based trigger” has collagenolytic potential. As degradation of native collagen is mediated by multiple proteases (collagenases), the reduction in the degradation of collagen is a more stringent readout compared to reading out only MMP1 expression. The effect of TARGET expression reduction in RA SFs (by means of KD-viruses) on the cytokine-induced collagen degradation by RA SFs is tested using an assay developed in house. In this assay, the degradation of FITC-labeled native collagen gives rise to an increase in fluorescence signal. (The protocol of this assay is detailed in the description of Example 6 (FIG. 12).).


The 323 hit KD viruses identified in the “reverse MMP1 assay” are arrayed in 96 well plates (“Hit plates”) such that every plate contained 60 hit viruses along with 4 positive control viruses and 16 negative control viruses. RA SF, seeded in 96 well plates at a density of 3000 cells/well in complete synovial growth medium (Cell Applications) are infected, one day later, with 5 or 10 μl of the viruses contained in the “hit plates”. The virus load is completed by addition 6 μl of the neutral virus Ad5-Luciferase-v13_KD. This correction guarantees that the effects observed do not result from differences in the virus load applied to the cells. 5 days after infection, the activation step is performed. This step involves the replacement, in every well, of the growth medium by 45 μl of M199 medium supplemented with 15 μl of “TNF□-based trigger”. 4 days later, the supernatant is collected and subjected to the miniaturized collagen type I degradation assay according to the protocol described for the experiment depicted in FIG. 12. Two independent propagation materials for every target KD virus are tested twice at both MOIs, giving rise to 4 data points for each MOI. The results are analysed as follows:


For every plate, the samples with corresponding fluorescence value lower then the average over all negative controls minus 2.1 times (5 μl infection) or 1.6 times (10 μl infection) the standard deviation over all negative controls are considered as giving a positive response in the assay. Every Target KD virus giving rise to 3 out of 4 data points giving a positive readout in the assay is considered as significantly reducing “TNF-based trigger”-induced collagen degradation by RA SFs. Alternatively, every Target KD virus generating 2 positive responses in the assay (out of 4 data points) for each MOI tested are considered as significantly reducing collagen degradation. Of the 323 target KD viruses tested, 192 significantly reduced the “TNF-based trigger”-induced collagen degradation by RA SFs.


To select for the preferred hits, we used a second assay that tests for the effect of the target expression reduction in RA SFs (by means of KD-viruses) on the LPS or TNFE-induced IL8 expression by RA SFs. This assay (1) confirms that the target KD viruses selected are not inhibiting the LPS signaling in RA SFs too much, as LPS signaling is required for the response of patients to pathogens and its complete inhibition would compromise the patients' innate immunity; and (2) confirms that the targets do not enhance the TNF□-induced expression of an inflammation marker. The chemokine IL8 is selected as a readout in this assay. This chemokine plays a role in the recruitment of cells of the immune system (e.g. monocytes, neutrophils) to the site of inflammation (the joint in the case of RA), further increasing the local inflammatory events. Increase of IL8 expression is not a desired feature for a RA therapy.


The target KD viruses identified in the “reverse MMP1 assay” and giving a positive readout in the “reverse collagen degradation assay” are tested in the LPS/TNF□-induced IL8 assay as follows: 192 target KD viruses are arrayed in 96 well plates (“Hit plates”) such that every plate contained 60 target viruses along with 20 negative control viruses. Day 1, SFs (passage 9 to 12) are seeded in 96 well plates at a density of 3000 cells per well in complete synovial growth medium (Cell Applications). One day later, the cells are infected with two amounts (6 or 12 μl) of the target KD viruses arrayed in 96 well plates as indicated above. The cells are then incubated for 5 days before the activation step, which involves the replacement, in every well, of the growth medium by 150 μl of DMEM supplemented with 10% Fetal bovine serum (heat inactivated) and the trigger (2 ng/ml recombinant human TNF□ or 1 μg/ml LPS). 48 hrs after the activation step, 80 μl of the supernatant is collected and a dilution from this is used for the IL-8 ELISA as described for the experiment depicted in FIG. 13. For the samples triggered with TNF□ a 40-fold dilution is made in PBS+1% BSA, and for LPS samples, a 16 fold dilution is performed. 35 μl from these dilutions is subjected to the IL-8 ELISA. Two independent preparations of the target KD viruses are tested at 2 MOI in 2 independent experiments giving rise to 4 data points per MOI for every target KD virus tested.


Data are analysed as follows: the 96 well plates containing the KD viruses designed to reduce the expression of the targets also contained 20 negative controls. The percentage inhibition of IL8 expression is calculated as percentage relative to the negative controls as follows: First, the background signal of the ELISA (in the absence of IL8) is subtracted from all values for all samples. Then the following formula is applied for every plate: % inhibition=[100×(((Average value all negative controls)−value Target sample)/(Average value all negative controls))]. All the Target KD viruses are ranked depending on their performance in the TNF□ and LPS induced IL8 assay. 10% of the target. KD viruses giving the strongest inhibition of LPS-induced IL8 are considered less preferred as well as 5% of the target KD viruses giving rise to the strongest increase of the TNF□-induced IL8 expression by RA SFs. As such, 40 out of the 192 Target KD viruses tested in this assay are considered to be less preferred. Further exclusion criteria used to define the present invention with more particularity are described below.


The TARGETS identified in this invention represent the basis for the identification of small molecule inhibitors developed for the treatment of RA. As such, the testing of such new candidate RA therapies in experimental models of arthritis is required before running experiments in human. The collagen-induced arthritis model in rat or mice is generally used as experimental model of arthritis. Consequently, targets, for which no ortholog can be found in rat or mice, are less preferred. Analysis of the 152 targets (identified through the experimental cascade (the “reverse MMP1” primary assay, the “reverse collagen degradation” secondary assay and the “TNF□/LPS IL8 induction assay”) revealed that 27 had no ortholog in rat or mice, leaving only 125 preferred targets. Further exclusion criteria involved the following in silico analyses:


“Drugability”.


Targets were excluded if it could be determined that the development of a small molecule inhibitor would be predicted to be the least successful within a short timeframe. This analysis involved the assessment of the general drugability of the gene class to which a certain Target belongs based on pharmacology precedents. In addition, the existence of assays allowing the discovery of small molecule inhibitors of the Targets is evaluated. We found assays available for 69 out of the 125 Targets subjected to this analysis.


“Risk Profile”.


Target genes for which the corresponding “knock-out mice” phenotype is diseased or lethal are considered as having lower priority, as inhibition of the product of these genes by means of a small molecule is expected to cause part of this phenotype. The Targets that play an important role in basal metabolic functions of the cells or of the organism are also given lower priority. The risk profile is considered high for 18 out of the 69 targets analysed.


“Disease relevance.”


Targets that are already linked to inflammatory processes or autoimmune processes are considered as more preferred.


The forgoing set of experiments and analyses permitted the narrowing of the present set of 16 most preferred TARGETs from the list of 323 targets.


Example 4
Analysis of the Expression Levels for Certain Targets Identified in Human Primary Synovial Fibroblasts Derived from Synovium of RA Patients

Expression levels for certain identified targets are determined in at least two different isolates of primary human synovial fibroblasts.


The RASFs isolates are obtained as cryo-preserved passage 2 cells from Cell Applications Inc. (Cat. No. 404-05). These cells are cultured and propagated in DMEM (Invitrogen) supplemented with 10% (v/v) heat-inactivated FBS (ICN) and 1×Pen/Strep (Invitrogen). For expression analysis, cells are cultured to passage 11.


For RNA preparation, the primary human synovial fibroblasts are seeded in 10-cm Petri dishes (500,000 cells/dish) in 6-well plates. After overnight incubation, medium is refreshed with 6 ml of M199 medium supplemented with 1% (v/v) heat-inactivated FBS containing 1×Pen/Strep. 24 hours later, total RNA is extracted using the “SV Total RNA Isolation kit” (Promega).


The concentration of RNA in each sample is fluorimetrically quantified using the “Ribogreen RNA quantitation kit” (Molecular Probes). A similar amount of RNA from each preparation is reverse transcribed into first strand cDNA with the “Taqman reverse transcription kit” from Applied Biosystems. Briefly, 40 ng RNA is included per 20 μl reaction mix containing 50 μmol of random hexamers, 10 U Rnase inhibitor, 25 U Multiscribe reverse transcriptase, 5 mM MgCl2 and 0.5 mM of each dNTP. The reaction mixture is incubated at 25° C. for 10 minutes, followed by 30 minutes incubation at 48° C. and heat inactivation (5 minutes 95° C.) of the reverse transcriptase in a thermocycler (Dyad, MJ Research). Reactions are immediately chilled to 4° C. at the end of the program. To avoid multiple freeze/thaw cycles of the obtained cDNA, the different samples are pooled in 96-well plates, aliquoted and stored at −20° C.


Real-time PCR reactions are performed and monitored using the “ABI PRISM 7000 Sequence Detection System Instrument” (Applied Biosystems). Pre-designed, gene-specific Taqman probe and primer sets for quantitative gene expression are purchased from Applied Biosystems as part of the “Assays on Demand” Gene expression products. These commercially available kits are quality checked by the supplier and allow quantitative determination of the amount of target cDNA in the sample. The “Assays on Demand” gene expression products are used according to the protocol delivered by the supplier. The PCR mixture consisted of 1דTaqman Universal PCR Mastermix no AmpErase UNG” and 1× “Taqman Gene Expression Assay on Demand mix” and 5 ul of the retro-transcription reaction product (1-100 ng of RNA converted into cDNA) in a total volume of 25 ul. After an initial denaturation step at 95° C. for 10 minutes, the cDNA products are amplified with 40 cycles consisting of 95° C. for 15 sec, and 60° C. for 1 minutes To normalize for variability in the initial quantities of cDNA between different samples, amplification reactions with the same cDNA are performed for the housekeeping gene □-actin using the predeveloped □-actin “Assays on demand” primer set and Taqman probe mix and “Taqman Universal PCR Mastermix” (all Applied Biosystems) according to the manufacturer's instructions. To identify any contamination resulting from residual genomic DNA, real-time PCR reactions with product from a control (—RT) reverse transcription reaction that is performed under the same conditions but without the addition of the reverse transcriptase are included for each sample. Threshold cycle values (Ct), for example, the cycle number at which the amount of amplified gene of interest reached a fixed threshold are determined for each sample. For each sample, the □Ct value is determined by subtracting the Ct value of the endogenous control (□-actin) from the Ct value obtained for the target gene. A gene is considered as expressed in primary human SFs if the □Ct value obtained for this hit is lower than 13.3 in at least one of the available 2 synovial isolates, activated or not. Genes with a □Ct value below 9.9 are considered highly expressed in RASFs. The results of the expression profiling experiments are summarized in Table 5. The □Ct value relative to □-actin obtained for various targets in 2 isolates of untriggered SFs are given in this Table 5.









TABLE 5







Determination of the Relative Expression Levels of the TARGETS


in Primary Synovial Fibroblasts by Real-Time PCR









Applied Biosystems










Target gene
Assay on demand,
RA SF batch 2
RA SF batch 3












symbol
Catalog number
Ct
DCt
Ct
DCt















ARAF1
Hs00176427_m1
27
2.8
25.3
2.2


AXL
Hs00242357_m1
25.9
1.6
23.6
0.5


KCNF1
Hs00266908_s1
40
14.86
29.3
4.46


MAPK12

NT
NT
NT
NT


MAPK13
Hs00559623_m1
29.9
6.2
29
6


NR2F6
Hs00172870_ml
27.2
2.9
24.2
1.83


SCN9A
Hs00161567_m1
37.02
11.88
34.54
9.7


SLC9A8
Hs00392302_m1
30.76
5.62
28.56
3.72


CHRNA5
Hs00181248_m1
34.8
11.1
32.2
9.1


DGKB

NT
NT
NT
NT


FGFR3
Hs00179829_m1
36.41
13.08
34.48
11.74


ILK
Hs00177914_m1
26.8
3.2
24.4
1.3


NQO2
Hs00168552_m1
28.05
4.72
25.36
2.62









Example 5
Ad-siRNA Reduction of RASF Expression of ILK, CHRNA5, NQO2, KCNF1, SLC9A8, ARAF1, AXL, FGFR3, NR2F6, SCN9A, MAPK13 and DGKB Inhibit Cytokine-Induced MMP1 Expression in a Dose Dependent Manner

As described above, the screening of the SilenceSelect collection identifies a set of Ad5-siRNAs that exhibit the capacity to reduce the cytokine-induced RASF MMP1 expression. A subset of these viruses is tested in the “reverse MMP1 assay” at various concentrations in a dose response experiment.


SFs (passage 9 to 10) are seeded in 96 well plates at a density of 3000 cells/well in complete synovial growth medium (Cell Applications). One day later, the cells are infected with increasing amounts (3, 7.5, 12, or 15 μl) of the Ad5-MMP1_v10_KD (positive control) or Ad5-Luciferase_v13_KD (negative control). The virus load is corrected by addition of the neutral virus Ad5-Luciferase_v13_KD to bring the final virus volume on the cells to 15 μl in every well. This correction guarantees that the effects observed do not result from differences in the virus load applied to the cells. The cells are then incubated for 5 days before the activation step. This step involves the replacement, in every well, of the growth medium by 75 μl of M199 medium supplemented with 25 μl of “complex trigger”. 48 hours after the activation step, the supernatant is collected and subjected to the MMP1 ELISA as described previously. FIG. 11A shows the average of duplicate measurements.


This experiment clearly demonstrates that the application of increased amounts of Ad5-siRNA to RASFs will lead to an increased reduction of the expression of the target in the RASFs. In this case, applying increasing amounts of Ad5-MMP1_v10_KD to the cells leads To higher reduction in MMP1 protein expression by the cells. (FIG. 11A) Taken together, as increasing the amounts of Ad5-siRNA applied to RASFs will lead to greater reduction of the expression of the target in the RASFs, increasing, the amounts of Ad5-siRNA targeting the genes listed in Table 1 is expected also to result in greater reduction of the cytokine induced MMP1 expression. The results of such dose response experiments are illustrated in FIG. 11B.


The Ad-siRNAs used in these dose response experiments are generated according to the procedure described in WO03/020931. The target sequences of these genes, on which the siRNAs are designed and used to generate the recombinant adenoviruses, are listed in Table 1. The Ad5-siRNA efficacy in the MMP1 ELISA is tested as described above with respect to the positive and negative controls with the only difference being that multiple Ad-siRNAs targeting the genes listed in Table 1 are used to infect the cells. The viruses used to infect the cells are shown on. FIG. 11B with Ad5-eGFP-v5_KD, Ad5-Luciferase-v13_KD and Ad-M6PR-v1_KD used as negative controls, and Ad5-MMP1-v10_KD used as a positive control. The results of the experiment are shown in FIG. 11B.


The data shown in FIG. 11B is calculated as follows: For each plate analyzed, the raw signal (RLU) for Ad5-MMP1_v10-KD (positive control) at 15 μl is considered as the lowest possible signal (i.e., the strongest reduction of MMP1 expression that can be obtained), and therefore, is set as the background of the assay. All other signals for this plate are adjusted to account for this background signal. For each MOI, the signal obtained for a particular Ad5-Target-KD virus is then normalized to (divided by) the signal of the negative control virus obtained on the same plate (Ad5-eGFP-v5_KD, Ad5-Luciferase-v13_KD or Ad-M6PR-v1_KD). FIG. 11B shows that the Ad-siRNA viruses mediating the reduction in expression of ILK, CHRNA5, NQO2, KCNF1, SLC9A8, ARAF1, AXL, FGFR3, NR2F6, SCN9A, MAPK13 and DGKB in RASFs cause a significant reduction of cytokine-induced MMP1 expression by these cells. Clearly, greater amounts of Ad5-siRNAs result in greater reduction in MMP1 expression levels. These results indicate that the amount of Ad5-siRNA applied to the cells determines the strength of the reduction in MMP1 expression observed.


We conclude, from this experiment, that these genes represent valuable drug targets that are shown to modulate MMP1 expression in SFs, and that the inhibition of the activity of the protein product of these genes by an inhibitory agent, such as a small molecule compound, is expected to reduce “complex cytokine” induced MMP1 expression in the “reverse MMP1 assay”. We also believe that the inhibition of the activity of the protein products of these genes by such agents would also reduce the joint degradation observed in RA patients.


Table 1 lists multiple KD sequences produced for every TARGET gene identified in Table 1. Different Ad-siRNAs are generated from different target sequences within the TARGET mRNA. Although some of the Ad-siRNA viruses exhibit efficacy in the “reverse MMP1 assay,” we observe that some of these Ad-siRNA viruses do not exhibit efficacy in the “reverse MMP1 assay”. It should be understood, however, that not all shRNAs are as efficient in reducing target expression. There may be many cause of this difference in knockdown efficacy, including the physical availability of the target RNA to the siRNA degradation machinery (for example, in a three dimensional folding of any TARGET mRNA which is in close proximity with and/or binding to other cellular proteins, only a fraction of the mRNA may be “exposed” so as to provide a binding site for siRNA association), the potency of the target protein, degradation processes affecting the particular shRNA itself, and others. More particularly, the low efficacy of certain Ad-siRNAs in the “reverse MMP1 assay” may be explained by the level of reduction in target gene expression required to reduce cytokine-induced MMP1 expression by the cells. Some targets may require only 50% reduction to shut down the MMP1 pathway, while others may require 90% to achieve the same effect. Some targets may participate in metabolic pathways that compensate for target depletion, while others do not. As a consequence, testing these viruses at higher doses might lead to higher efficacy in the reverse MMP1 assay.


Example 6
Ad-siRNA-Induced Reduction of the RASF Expression of ILK, CHRNA5, NQO2, KCNF1, SLC9A8, ARAF1, AXL, FGFR3, NR2F6, SCN9A, MAPK12, MAPK13 and DGKB Inhibit Cytokine-Induced Collagen Degradation

Certain MMP subtypes (for example, MMP1, MMP13) are collagenases and have the remarkable capacity of degrading native collagen. Native collagen is organized in stable fibrils that are quite resistant to proteolytic events. As such, the degradation of native collagen can be used to monitor not only MMP1 but also the complete complement of collagenases produced by RASFs. Described herein is an assay developed to detect the cytokine-induced degradation of native collagen in the supernatant of RASF cultures.


“Miniaturized Collagen Type I Degradation Assay” protocol: (Reagents are from Chondrex, Redmond, Wash. (Collagenase assay kit) unless specified differently). A 96 well plate (V-bottom, Greiner) is filled with 9 μl of solution B and 1 μl of trypsin solution per well. 10 μl of sample (RASF supernatant) is added per well, followed by incubation for 15 minutes at 34° C. After incubation, 1 μl SBTI is added. 20 μl of FITC-Collagen mix (10 μl FITC-labeled collagen type I+10 μl solution A) are added to the activated sample followed by incubation for 24 hours at 34° C. 1 μl of 1.10 Phenantroline (Sigma) is added to the reaction mixture. One μl of enhancer solution (elastase) is added, followed by incubation for 30 minutes at 34° C. When the reaction mixture is at room temperature, 40 μl extraction buffer are added and the plate is sealed (Nunc seals) and vortexed. After centrifugation for 25 minutes at 4000 rpm (Beckman centrifuge), 50 μl of the supernatant are transferred into a black F-bottom plate (Greiner) and fluorescence is measured on a Fluostar reader (BMG), 480 nm excitation wavelength, 520 nm emission wavelength).


Ad5-siRNA efficacy in the “miniaturized collagen type I degradation assay” is tested as follows: SFs (passage 9 to 10) are seeded in 96 well plates at a density of 3000 cells/well in complete synovial growth medium (Cell Applications). One day later, the cells are infected with 5 or 10 μl of the Ad-siRNAs indicated on FIG. 12A. These KD viruses are arrayed on a 96 well plate along with 12 negative control viruses. The virus load is corrected by the addition of the neutral virus Ad5-Luciferase-v13_KD to bring the final virus volume on the cells to 15 μl in every well. This correction guarantees that the effects observed do not result from differences in the virus load applied to the cells. The cells are then incubated for 5 days before the activation step. This step involves the replacement, in every well, of the growth medium by 45 μl of M199 medium supplemented with 15 μl of “complex trigger”. Four days later, the supernatant is collected and subjected to the miniaturized collagen type I degradation assay according to the protocol described above. The results are analyzed as follows: The fluorescence signal obtained for the 12 negative controls is averaged for every plate. For each plate and each MOI (5 μl or 10 μl), the fluorescence signal observed for the samples emanating from cells infected with the KD viruses targeting the genes listed in Table 1 is expressed as a percent of the negative control average (set at 100%). Data shown in FIG. 12A are the average of duplicate measurements. The Ad-siRNAs targeting the genes listed in Table 2 do mediate a clear reduction of the complex trigger-induced collagen degradation by primary human SFs. We conclude, from this experiment, that these genes represent valuable drug targets that are shown to modulate not only MMP1 expression by RASFs but also collagen degradation by RASFs. Similarly, the inhibition of the activity of the protein product of these genes by an inhibitory agent, such as a small molecule compound, is expected to reduce the “complex cytokine” induced collagen degradation by SFs. We also believe that the inhibition of the activity of the protein products of these genes by such agents would also reduce the joint degradation observed in RA patients.


In similar experiments (FIG. 12B), the Ad5-MMP1 v10-KD virus is shown to strongly reduce the cytokine induced collagen degradation by SFs, which suggests that MMP1 is the main collagenase responsible for the cytokine induced collagen degradation by SFs. This experiment is run according to the protocol described for the experiment depicted in FIG. 12A. Data shown in FIG. 12B are the average and standard deviation over the raw fluorescence signal emanating from eight data points. As such, this experiment demonstrates that modulation of SF expression of MMP1 is sufficient to reduce cartilage degradation associated with RA. As such, the reduction of MMP1 expression by RASFs is predictive for a reduced degradation of native collagen, one of the main components of bone and cartilage.


Example 7
Reduction of the Expression in RASFs of ARAF1, CHRNA5, DGKB, FGFR3, ILK, KCNF1, MAPK12, MAPK13, NQO2, NR2F6, and SCN9A by Ad-siRNAs Inhibit Cytokine-Induced IL-8 Expression

To better understand the profile of the TARGETS (identified according to the experiments described in previous examples), and to probe for the specificity of TARGET expression reduction effects, IL8, another inflammation marker different from MMP1, is used in the following example. This example measures the effect of TARGET expression reduction in RA SFs on TNF□-induced IL8 expression. IL8 expression is increased by TNF□, and plays a role in inflammatory events. Additionally, a small molecule inhibitor of the IL8 receptor has been shown to have efficacy in an animal model of arthritis (See Weidner-Wells M A et al., Bioorg Med Chem. Lett. (2004)14:4307-11, “Synthesis and structure-activity relationships of 3,5-diarylisoxazoles and 3,5-diaryl-1,2,4-oxadiazoles, novel classes of small molecule interleukin-8 (IL-8) receptor antagonists. As such, achieving inhibition of IL8 expression is a desired feature for an RA therapeutic.


“IL-8 assay” protocol: SFs (passage 9 to 12) are seeded in 96 well plates at a density of 3000 cells/well in complete synovial growth medium (Cell Applications). One day later, the cells are infected with two amounts (6 or 12 μl) of the KD viruses. The cells are then incubated for 5 days before the activation step. The activation step involves the replacement, in every well, of the growth medium by 150 μl of DMEM supplemented with 10% Fetal Bovine serum (heat inactivated) and the trigger (2 ng/ml recombinant human TNFalpha). 48 hours after the activation step, 80 μl of the supernatant is collected and diluted for use in the IL-8 ELISA described below. For the samples triggered with TNFalpha, a 40 fold dilution is made in PBS+1% BSA. 35 μl from these dilutions are added to the IL-8 ELISA plates.


White Lumitrac 600 384 well plates (Greiner) are coated with 0.5 μg/ml anti-IL8 antibody MAB208(R&D). The antibody is diluted in 1×PBS (Gibco). After overnight incubation at 4° C., plates are washed once with 1×PBST (80 g NaCl, 2 g KCl (Sigma), 11.5 g Na2HPO4.7H2O and 2 g KH2PO4 in 10 L milliQ; pH 7.4=10×PBS solution+0.05% tween−20 (sigma)), once with PBS 1×, and blocked with 80 μl blocking buffer (1% BSA+5% sucrose+0.05% NaN3). One day later, the blocking buffer is removed from the ELISA plates by inverting the plate and tapping it on an absorbent paper. The plate is washed with 90 μl PBST and 90 μl PBS 1×. Immediately thereafter, the plate is further processed and 35 μl of the diluted supernatant is added. After overnight incubation at 4° C., the plates are washed once with PBST and once with PBS (as described above) and incubated with 35 μl/well biotinylated anti-IL 8 antibody solution BAF208 (R&D) 50 ng/ml in PBS 1×+1% BSA. After 2 hours of incubation at room temperature, plates are washed as described above and incubated with 50 μl/well streptavidin-HRP conjugate (Biosource). Streptavidin-HRP conjugate is diluted 1/2000 times in PBS 1×+1% BSA. After 45 minutes, plates are washed as described above and incubated for 5 minutes with 50 μl/well BM Chem ELISA Substrate (Roche). Readout is performed on the Luminoscan Ascent Luminometer (Labsystems) with an integration time of 100 msec.


Twenty negative controls are included in the 96 well plates. The percentage inhibition of IL8 expression is calculated as follows. First, the background signal of the ELISA (in the absence of IL8) is subtracted from all values for all samples. Then the following formula is applied: % inhibition=[100×(((Average value all negative controls)−value TARGET sample)/(Average value all negative controls))]. From the results depicted in FIG. 13, we conclude that expression for most TARGETS mediated a reduced TNF□-driven IL8 expression, with strongest effects seen for MAPK12, MAPK13 and ARAF1 and weakest effects observed for SLC9A8 and AXL.









TABLE 6







Summary of the Features of the TARGET Genes









Knock down data














Inhibition of



Gene
Expres-
MMP1
collagen
IL-8


symbol
sion *
Inhibition **
degradation ***
Inhibition ****





ARAF1
SP
SP
SP
SP


AXL
SP
P
SP
N


CHRNA5
P
P
SP
P


DGKB
NT
SP
SP
P


FGFR3
P
P
SP
P


ILK
SP
SP
SP
P


KCNF1
SP
P
SP
P


MAPK12
NT
N
SP
SP


MAPK13
SP
SP
SP
SP


NQO2
SP
SP
SP
P


NR2F6
SP
SP
SP
P


SCN9A
P
P
SP
P


SLC9A8
SP
P
SP
N





P: positive response in the assay


SP: Strong positive response in the assay


NT: not tested


N: negative response in the assay


* Expression in primary RASFs: Genes with DCt <13 are considered expressed (P), genes with DCt >9.9 are considered strongly expressed (SP)


** Inhibition of cytokine mixture induced MMP1 expression by RASFs >50% reduction in MMP1 expression is considered a strong positive response in the assay


*** Inhibition of cytokine induced collagen degradation by RASFs >50% reduction in cytokine induced collagen degradation is considered a strong positive response in the assay


**** Inhibition of TNF□ induced IL-8 expression by RASFs >20% inhibition of TNF□ induced IL-8 expression is considered a significant response in the assay (P), >50 inhibition is considered a strong positive response in the assay





Claims
  • 1. A method for identifying a compound that inhibits extra-cellular matrix degradation by proteolytic enzymes produced from synovial fibroblasts, comprising (a) contacting a compound with a polypeptide comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 881 or 882; and (b) measuring (i) the binding affinity of said compound to said polypeptide, and one or more steps of (ii) the inactivation of a biological pathway producing a biochemical marker indicative of extra-cellular matrix degradation by proteolytic enzymes produced from synovial fibroblasts, wherein said marker is selected from the group consisting of MMP1, and Cathepsin, or (iii) collagen degradation.
  • 2. The method according to claim 1, wherein said measuring the binding affinity of said compound to said polypeptide is in an in vitro cell-free preparation.
  • 3. The method according to claim 1, wherein said measuring of one or more steps (ii) or (iii) is present in a mammalian synovial fibroblast cell.
  • 4. The method of claim 1 wherein said marker is MMP1.
  • 5. A method for identifying a compound that inhibits extra-cellular matrix degradation in a mammalian synovial fibroblast cell that expresses an extra-cellular matrix degrading protein selected from the group consisting of MMP1 and Cathepsin, and that expresses a polypeptide comprising the amino acid sequence of SEQ ID NO: 881 or 882, said method comprising (a) contacting a compound with a polypeptide comprising the amino acid sequence of SEQ ID NO: 881 or 882 in an in vitro cell-free preparation;(b) determining the binding affinity of said compound in said in vitro preparation;(c) contacting said compound having binding affinity to, and capable of forming a complex said polypeptide of SEQ ID NO: 881 or 882 with a mammalian synovial fibroblast cell, in which said polypeptide comprising the amino acid sequence of SEQ ID NO: 881 or 882 is expressed, and which expresses an extra-cellular matrix degrading protein; and(d) measuring (i) one or more extra-cellular matrix degrading protein expressed by said mammalian synovial fibroblast cell and selected from the group consisting of MMP1 and Cathepsin, or (ii) collagen degradation, or (i) and (ii); and(e) determining if said one or more extra-cellular matrix degrading protein expression or collagen degradation, or both are decreased as compared to of said extra-cellular matrix degrading protein expressed, or collagen degradation, respectively, in said mammalian synovial fibroblast cell that is not contacted with said compound.
  • 6. The method according to claim 5 wherein said compound having binding affinity to said polypeptide of SEQ ID NO: 881 or 882 exhibits a binding affinity of at least 10 micromolar.
  • 7. The method according to claim 6, wherein said extra-cellular matrix degrading protein is MMP1.
  • 8. The method according to claim 7, wherein each of said mammalian synovial fibroblasts that is contacted with said compound and that is not contacted with said compound is contacted with a cytokine agonist or with cytokine-producing cells or a TNFα-based trigger.
CROSS-REFERENCE TO RELATED APPLICATIONS

This application claims priority to U.S. application Ser. No. 12/360,332, filed Jan. 27, 2009, which is a divisional of U.S. application Ser. No. 11/251,465, filed Oct. 14, 2005, now U.S. Pat. No. 7,485,468, and Ser. No. 60/619,384, filed Oct. 15, 2004, the disclosures of which are incorporated by reference.

US Referenced Citations (26)
Number Name Date Kind
5459036 Lechner et al. Oct 1995 A
6030822 Lechner et al. Feb 2000 A
6162613 Su et al. Dec 2000 A
6387641 Bellon et al. May 2002 B1
6579972 Lechner et al. Jun 2003 B1
6699983 Dedhar et al. Mar 2004 B1
6753314 Giot et al. Jun 2004 B1
6849716 Su et al. Feb 2005 B1
7202033 Prescott et al. Apr 2007 B2
20030096303 Bellon et al. May 2003 A1
20030165899 Su et al. Sep 2003 A1
20030198627 Arts et al. Oct 2003 A1
20030229209 Lechner et al. Dec 2003 A1
20030232391 Prescott et al. Dec 2003 A1
20040072192 Young et al. Apr 2004 A1
20040096836 Freier et al. May 2004 A1
20040110145 Bennett et al. Jun 2004 A1
20040259166 Su et al. Dec 2004 A1
20050084905 Prescott et al. Apr 2005 A1
20060210552 Demopulos et al. Sep 2006 A1
20060216722 Betsholtz et al. Sep 2006 A1
20070224662 Luo et al. Sep 2007 A1
20070237777 DiMauro et al. Oct 2007 A1
20090075831 Russwurm et al. Mar 2009 A1
20090081228 Lau et al. Mar 2009 A1
20090325152 Russwurm et al. Dec 2009 A1
Foreign Referenced Citations (35)
Number Date Country
2012311 Sep 1999 CA
0689588 Mar 1996 EP
1054976 Nov 2000 EP
1165772 Jan 2002 EP
1399584 Mar 2004 EP
1579186 Sep 2005 EP
1799848 Jun 2007 EP
1863928 Dec 2007 EP
2011887 Jan 2009 EP
08-510897 Nov 1996 JP
2002-504399 Feb 2002 JP
2003-515525 May 2003 JP
2003-524400 Aug 2003 JP
2004-533206 Nov 2004 JP
2005-534286 Nov 2005 JP
9421781 Sep 1994 WO
9942592 Aug 1999 WO
0036096 Jun 2000 WO
0060066 Oct 2000 WO
0231198 Apr 2002 WO
0240541 May 2002 WO
03025148 Mar 2003 WO
03076651 Sep 2003 WO
03081210 Oct 2003 WO
03083046 Oct 2003 WO
2004006838 Jan 2004 WO
2004039955 May 2004 WO
2004058805 Jul 2004 WO
2004094636 Nov 2004 WO
WO 2005016962 Feb 2005 WO
2005124342 Dec 2005 WO
2006042581 Apr 2006 WO
2006063856 Jun 2006 WO
2006100066 Sep 2006 WO
2006100203 Sep 2006 WO
Non-Patent Literature Citations (70)
Entry
Cunnane et al., Arthritis & Rheumatism, vol. 44, No. 10, pates 2263-2274 (2001).
Pinkert et al. “An albumin enhancer located 10 kb upstream functions along with its promoter to direct efficient, liver-specific expression in transgenic mice.” Genes and Development, vol. 1, 1987, pp. 268-276.
Redhead et al. Expression of a myelin basic protein gene in transgenic shiverer mice: correction of the dysmyelinating phenotype. Cell, vol. 48, 1987, pp. 703-712.
Reiff et al. “Matrix Metallproteinases 2 and 9 are markers of inflammation but not of the degree of fibrosis in chronic Hepatitis C.” Digestion, Internal Journal of Gastronterology, vol. 71(2), 2005, pp. 124-130.
Rosenberg. “Matrix metalloproteinase in neuroinflammation.” GLIA, vol. 39(3), 2002, pp. 279-291.
Schanstra et al. “In vivo brandykinin B2 receptor activation reduces renal fibrosis.” The Journal of Clinical Investigation, vol. 110, 2002, pp. 371-379.
Schett et al. “Sdenovirus-based overexpression of the tissue inhibitor of metalloproteinases 1 reduces tissue damage in the joints of tumor necrosis factor alpha transgenic mice/” Arthritis and Rheumatism, vol. 44(12), 2001, pp. 2888-2896.
Shani. “Tissue-specific expression of rat myosin light-chain 2 gene in transgenic mice.” Nature, vol., 314, 1985, pp. 283-286.
Smolen et al. “Therapeutic strategies for rheumatoid arthritis.” Nature Reviews Drug Discovery, vol. 2(6), 2003, pp. 473-488.
St. Clair et al. “Combination of Inflizimab and Methotrexate Therapy for Early Rheumatoid Arthritis: A Randomized, Controlled Trial.” Arthritis Rheumatism, vol. 50(11), 2004, pp. 3432-3443.
Suzuki et al. “Metrix metalloproteinases in the pathogenesis of asthma in COPD: Implications for therapy.” Treatments in respitory Medicine, vol. 3(1—, Nov. 1, 2004, pp. 17-27.
Swift et al. “Tissue-specific expression of the rat [ancreatic elastace I gene in transgenic mice.” Cell, vol. 38, 1984, pp. 639-346.
Adams et al. “The c-myc oncogene driven by immunoglobulin enhancers induces lymphoid malignancy in transgenic mice.” Nature, vol. 318, 1985, pp. 533-538.
Agrawal et al. “Antisense therapeutics: is it as simple as complementary base recognition?”, Molecular Medicine Today, vol. 6, Feb. 2000, pp. 72-81.
Alexander et al. “Expression of the c-myc ocogene under control of an immunoglobulin enhancer in E mu-myc transgenic mice.” Molecular and Cellular Biology, vol. 7, 1987, pp. 1436-1444.
Allan et al., “Hormone and antihormone induce distinct conformational changes which are central to steroid receptor activation.” Journal of Biological Chemistry, vol. 267, 1992, pp. 19513-19520.
Andreakos et al. “Heterogeneous Requirement of IkappaB Kinase 2 for Inflammatory Ctyokine and Matrix Metalloproteinase Production in Rheumatoid Arthritis: Implication for Therapy.” Arthritis & Rheumatism, vol. 48(7), 2003, pp. 1901-1912.
Boerner et al. “Production of antigen-specific human monoclonal antibodies from in vitro-primed human splenocytes.” The Journal of Immunology, vol. 147(1), 1991, pp. 86-95.
Branch “A good antisense molecule is hard to find” Trends Biochem Sci., vol. 23(2), Feb. 1998, pp. 45-50.
Chen et al. “Multitarget-ribozyme directed to cleave at up to nine highly conserved HIV-1 env RNA regions inhibits HIV-1 replication-potential effectiveness against most presently sequenced HIV-1 isolates.” Nucleic Acids Research, vol. 20, 1992, pp. 4581-4589.
Chirila et al. “The use of synthetic polymers for delivery of therapeutic antisense oligodeoxynucleotides.” Biomaterials vol. 23(2), 2002, pp. 321-342.
Choy et al. “Cytokine pathways and joint inflamation in rheumatoid arthritis.” The New England Journal of Medicine, vol. 344(12), 2001, pp. 907-916.
Cole et al. “The EBV-hybridoma technique and its application to human lung cancer.” Monoclonal Antibodies and Cancer Therapy, Reisfeld & Sells, eds. 1985, pp. 77-96.
Cortez-Retamozo et al. “Efficient Cancer Therapy with a Nanobody-Based Conjugate.” Cancer Research, vol. 64, Apr. 15, 2004, pp. 2853-2857.
Coussens et al. “Matrix metalloproteinase inhibitors and cancer: trials and tribulations.” Science. vol. 295, 2002, pp. 2387-2392.
Creemers et al. “Matric Metalloproteinase Inhibition after Myocardial Infarction: A New Approach to Prevent Heart Failure.” Curculation Research, vol. 89(3), 2001, pp. 201-210.
Crooke et al. eds. Antisense Research and Applications. 1993, Chapter 1, pp. 1-50. S Crook.
Cunnane et al. “Early Joint Erosion adn Derum Levels of Matrix Metalloproteinase 1, Matrix Metalloproteinase 3 and Tissue Inhibitor Metalloproteniase 1 in Rheumatoid Arthritis.” Arthritis & Rheumatism, vol. 44(10), 2001, pp. 2263-2274.
Curiel et al. “Higg-efficency gene transfer mediated by adenovirus coupled to DNA-polysine complexes.” Human Gene Therapy, vol. 3, 1992, pp. 147-154.
Edwards et al. “Efficacy of B-Cell-Targeted Therapy with Rituximab in patients with Rehumatoid Arthritis.” New England Journal of Medicine, vol. 350(25), 2004, pp. 2572-2581.
Fearns et al. “Coordinate activation of endogenous p38α, β, γ and δ by inflammatory stimuli.” Journal of Lwukocyte Biology, vol. 67(5), 2000, pp. 507-711.
Keidel et al. “Different Agonist- and antagonist-induced conformational changes in retitonic acid receptors analyzed by protease mapping.” Molecular and Cellular Biology, vol. 14, 1994, pp. 287-298.
Kelsey et al. Species- and tissue-specific expression of human alpha 1-antitrypsin in transgenic mice. Genes and Development, vol. 1, 1987, pp. 161-171.
Kollias et al. “Regulated expression of human a gamma-, beta-, and hybrid gamma beta-globin genes in transgenic mice: manipulation of the developmental expression patterns.” Cell, vol. 46, 1986, pp. 89-94.
Kramer et al. “Etanercept Added to Background Methotrexate Therapy in Patients with Rheumatoid Atrhritis.” Arthritis & Rheumatism, vol. 48(6), 2003, pp. 1493-1499.
Krunlauf et al. “Developmental regulation of alpha-getoprotein genes in transgenic mice.” Molecular and Cellular Biology, vol. 5, 1985, pp. 1639-1648.
Leder et al. “Consequences of widespread deregulation of the c-myc gene in transgenic mice: Multiple neoplasms and normal development.” Cell, vil. 45, 1986, pp. 485-495.
Lee et al. “Rheumatiod Atrhritis.” The Lancet, vol. 358, 2001, pp. 903-911.
Lipinski et al. “Experimental and computational approaches ro estimate solubility and premeability in drug discovery and development settings.” Advanced Drug Delivery Reviews, vol. 23, 1997, pp. 3-25.
MacDonald. “Expression of the pancreatic elastase I gene in transgenic mice.” Hepatology, vol. 7, 1987, pp. 425-515.
MacKey et al. Gene transfer from targeted liposomes to specific lymphiod cells by electroporation/ Proceedings of the National Academy of Sciences, USA, vol. 85, 1988, pp. 8027-8031.
Magram et al. “Developmental regulation of a cloned adult beta-globin gene in transgenic mice.” Nature, vol. 315, 1985, pp. 338-340.
Traunecer et al. “Bispecific signal chain molecules (Janusins) target cytotoxic lumphocytes on HIV infected cells.” EMBO Journal, vol. 10, 1991, pp. 3655-3659.
Ulmer et al. “Heterogous protection against influenza by injection of DAN encoding a viral protein.” Csicence. vol. 259, 1993, pp. 1745-1749.
Ventura et al. “Activation of HIV-specific ribozyme activity by self-cleavage.” Nucleic Acids Research, vol. 251, 1993, pp. 3249-3255.
Vincenti et al. “Transcriptional regulation of collagenase (MMP-1, MMP-13) genes in arthritis: Integration of complex signaling pathways for the recruitment of gene-specific transcription factor.” Arthritis Research, vol. 4(3), 2002, pp. 157-164.
Williams et al. “Introduction to foreign genes into tissues of living mice by DNA coated microprojectiles.”Proceedings of the National Academy of Sciences, USA, vol. 88, 1991, pp. 2726-2730.
Wilson et al. “Hepatocyte-directed gene transfer in vivo leads to transient improvement of hypercholesterolema in low density lipoprotein receptor-deficient rabbits.” Journal of Biological Chemistry, vol. 267, 1993,[pp. 963-667.
Wu et al. “Receptor-mediated in vitro gene transformation by a soluble DNA carrier system.” Journal of Biological Chemistry, vol. 262, 1987, pp. 4429-4432.
Wu et al. Receptor-mediated gene delivery and expression in vivo. Journal of Biological Chemistry, vol. 263, 17988, pp. 14621-14624, (1988).
Yang et al. “Modulation o Inflamation and Response to Dexamethasone by Annexin 1 in Antigen-Induced Arthritis.” Arthritis Rheumatism, col. 50(3), pp. 976-984, (2004).
Felgner et al. “Lipofection: a highly efficient, lipid-mediated DNA-transfection procedure.” Proceedings of the National Academy of Sciences, USA, vol. 84, 1987, pp. 7413-7417.
Felgner et al. “Cationic liposome-mediated transfection.” Nature, vol. 337, 1989, pp. 387-388.
Firestein “Evolving Concepts of Rheumatoid Arthritis.” Nature, vol. 423, 2003, pp. 356-2361.
Gao et al. “Cytoplasmic expression of a reporter gene by co-delivery of T7 RNA polymerase and T7 promoter sequence with cationic liposimes.” Nucleic Acids Research, vol. 21, 1993, pp. 2867-2872.
Gapski et al. “Effect of Systemic Matrix Meralloproteinase Inhibition on Peridontal Wound Repair: A Proof of Concept Trial.” Journal of Peridontology, vol. 75(3), 2004, pp. 441-452.
Gomez-Rino et al. “Treatment of Rheumatoid Arthritis with Tumor Necrosis Factor Inhibitors may Predispose to Significant Increase in Tuberculosis Risk: A Multicenter Active-Surveillance Report.” Arthritis & Rheumatism, vol. 48(8), 2003, pp. 2122-2127.
Grosschedl et al. “Introduction to μ immunoglobulin gene into the mouse germ line: Specific expression in lymphoid cells and synthesis of functional antibody.” Cell, vol. 38,. 1984, pp. 647-658.
Hammer et al. “Diversity of the alpha-fetoprotein genes in transgenic mice is generated by a combination of separate enhancer elements.” Science, vol. 235, 1987, pp. 53-58.
Hanahan. “Heritable formation of pancreatic beta-cell tumours in transgenic mice expressing recombinant insulin/ simian virus 40 oncogenes.” Nature, vol. 315, pp. 115-122, (1985).
Hoogenboom et al. “By-passing immunisation. Human antibodies from synthetic repertoires of germline VH gene segments rearranged in vitro.” Journal of Molecular Biology, vol. 227, 1991, pp. 381-388.
Kashani-Sabet et al. “Reversal of the malignant phenotype by an anti-ras ribozyme.” Antisense Research and Development, vol. 2, 1992, pp. 3-15.
Marks et al. “By-passing immunization. Human antibodies from V-gene libraries displayed on phage.” Journal of Molecular Biology, vol. 222, 1991, pp. 1581-1597.
Mason et al. “The hypogonadal mouse: reproductive funations restored by gene therapy/” Science, vol. 234, 1986, pp. 1372-1378.
Milstein et al. “Hybrid Hybridomas and their use in immunohistochemistry.” Nature, vol. 305, 1983, pp. 537-540.
O'Dell “Efficacy of Triple DMARD Therapy in Patients with RA With Suboptimal Response to Methotrexate.” Journal of Rheumatology, vol. 23, Suppl. 44, 1996, pp. 72-74.
O'Dell. “Therapeutic strategies for rheumatoid arthritis.” The New England Journal of Medicine, vol. 350(25), 2004, pp. 2591-2602.
Omitz et al. “Elastase I Promotor Directs Expression of Human Growth Hormone and SV40 T Antigen Genes to Pancreatic Acinar Cells in Transgenic Mice.” Cold Springs Harbor Symposia in Quantative Biology, vol. 50, 1985, pp. 399-409.
Opalinska et al. “Nucleic-acid therapeutics: basic principles and recent applications” Nature Reviews Drug Discovery, vol. 1, pp. 503-514, (2002).
Perachi et al. “Prospects for antiviral ribozymes and deoxyribozymes.” Rev Med Virol., vol. 14(1), 2004, pp. 47-64.
Related Publications (1)
Number Date Country
20120190570 A1 Jul 2012 US
Provisional Applications (1)
Number Date Country
60619384 Oct 2004 US
Divisions (2)
Number Date Country
Parent 12360332 Jan 2009 US
Child 13344303 US
Parent 11251465 Oct 2005 US
Child 12360332 US