METHODS AND MEANS FOR TREATMENT OF OSTEOARTHRITIS

- GALAPAGOS N.V.

The present invention relates to in vivo and in vitro methods, agents and compound screening assays for inducing anabolic stimulation of chondrocytes, including cartilage formation enhancing pharmaceutical compositions, and the use thereof in treating and/or preventing a disease involving a systemic or local decrease in mean cartilage thickness in a subject.

Skip to: Description  ·  Claims  · Patent History  ·  Patent History
Description
CROSS-REFERENCE TO RELATED APPLICATIONS

This application is a Divisional of U.S. application Ser. No. 13/360,393 filed Jan. 27, 2012, which a Divisional of U.S. application Ser. No. 12/575,220, filed Oct. 7, 2009, which is a Divisional of U.S. application Ser. No. 11/158,252, filed Jun. 21, 2005, which claims the benefit of priority of U.S. Provisional Application No. 60/581,568, filed Jun. 21, 2004, each of which are hereby incorporated by reference in their entirety.

FIELD OF THE INVENTION

The invention relates to the field of medicinal research, cartilage physiology and diseases involving the degeneration of cartilage tissue. More specifically, the invention relates to methods and means for identifying compounds that stimulate anabolic processes in chondrocytes and that typically induce the synthesis of cartilage. The invention also relates to the compounds that are useful in the treatment of osteoarthritis.

Cartilage is an avascular tissue made up largely of cartilage-specific cells, the chondrocytes, proteoglycans and collagen proteins, which are structural proteins that provide structural strength to connective tissue, such as skin, bone and cartilage. Collagen II, together with the protein collagen IX, forms a “biological alloy”, which is molded into a fibril-like structure and is arranged in a precise network, providing cartilage with great mechanical strength. The chondrocytes in normal articular cartilage occupy approximately 5% of the tissue volume, while the extra-cellular matrix makes up the remaining 95% of the tissue. The chondrocytes secrete the components of the matrix, which in turn supplies the chondrocytes with an environment suitable for their survival under mechanical stress.

Breakdown of articular cartilage, which is part of joints and which cushions the ends of the bones, causes the bones to rub against each other leading to pain and loss of movement. Cartilage degradation may also be the result of an imbalance in cartilage synthesizing (anabolic) and cartilage degrading (catabolic) processes. Unlike most tissues, cartilage does not self-repair following injury. The inability of cartilage to self-repair after injury, disease, or surgery is a major limiting factor in rehabilitation of degrading joint surfaces and injury to meniscal cartilage.

There are many diseases involving the degeneration of cartilage. Rheumatoid arthritis and osteoarthritis are among the most prominent. Osteoarthritis (also referred to as OA, or as wear-and-tear arthritis) is the most common form of arthritis and is characterized by loss of articular cartilage, often associated with hypertrophy of the bone. The disease mainly affects hands and weight-bearing joints such as knees, hips and spines. This process thins the cartilage through a phenomenon called apoptosis, or programmed cell death. When the surface area has disappeared due to the thinning, there is a grade I osteoarthritis; when the tangential surface area has disappeared, there is a grade two osteoarthritis. There are other levels of degeneration and destruction, which affect the deep and the calcified layers that border with the subchondral bone.

The clinical manifestations of the development of the osteoarthritis condition are: increased volume of the joint, pain, crepitation and functional disability that, gradually and steadily, first hinders the performance of lengthy walks and forced flexion and extension movements, depending on the affected joint, and then pain and limitation of minimum efforts emerge as well as pain at rest which interrupts sleeping. If the condition persists without correction and/or therapy, the joint is destroyed, leading the patient to major replacement surgery with total prosthesis, or to disability.

Therapeutic methods for the correction of the articular cartilage lesions that appear during the osteoarthritic disease have been developed, but so far none of them have been able to achieve the regeneration of articular cartilage in situ and in vivo.

REPORTED DEVELOPMENTS

Osteoarthritis is difficult to treat. At present, no cure is available and treatment focuses on relieving pain and preventing the affected joint from becoming deformed. Common treatments include the use of non-steroidal anti-inflammatory drugs (NSAID's), which are often used to relieve pain, while specific COX-2 inhibitors are used to relieve severe pain. Medicines such as chondroitin and glucosamine are thought to improve the cartilage itself. These treatments may be relatively successful, but not a substantive amount of research data is available.

In severe cases, joint replacement may be necessary. This is especially true for hips and knees. If a joint is extremely painful and cannot be replaced, it may be fused. This procedure stops the pain, but results in the permanent loss of joint function, making walking and bending difficult.

The treatment that has 74% to 90% effectiveness and produces excellent results is the transplantation of cultured autologous chondrocytes, by taking chondral cellular material from the patient, sending it to a laboratory where it is seeded in a proper medium for its proliferation, and, once enough volume is achieved after a variable period that may last from weeks to months, transporting it in a special container and implanting it in the damaged tissues to cover the tissue's defects.

Another treatment includes the intraarticular instillation of Hylan G-F 20 (Synvisc, Hyalgan, Artz etc.), a substance that improves temporarily the rheology of the synovial fluid, producing an almost immediate sensation of free movement and a marked reduction of pain. The residual effects of this substance act on the synovial receptors causing a pain reduction that lasts several weeks and even months. However, this isolated effect is counterproductive for the course of the disease and for the viability of the cartilage because, as it masks the symptoms, the joint is used with more intensity and its destruction is accelerated as the original problem is not corrected and the damaged articular cartilage is not restored.

Other reported methods include application of tendinous, periosteal, fascial, muscular or perichondral grafts; implantation of fibrin or cultured chondrocytes; implantation of synthetic matrices, such as collagen, carbon fiber; administration of electromagnetic fields. All of these have reported minimal and incomplete results with formation of repair, but not regenerative tissue, resulting in a poor quality tissue that can neither support the weighted load nor allow the restoration of an articular function with normal movement.

Stimulation of the anabolic processes, blocking catabolic processes, or a combination of these two, may result in stabilization of the cartilage, and perhaps even reversion of the damage, and therefore prevent further progression of the disease. Various triggers may stimulate anabolic stimulation of chondrocytes. Insulin-like growth factor-I (IGF-I) is the predominant anabolic growth factor in synovial fluid and stimulates the synthesis of both proteoglycans and collagen. It has also been shown that members of the bone morphogenetic protein (BMP) family, notably BMP2, BMP4, BMP6, and BMP7, and members of the human transforming growth factor-b (TGF-b) family can induce chondrocyte anabolic stimulation (Chubinskaya and Kuettner, 2003). A compound has recently been identified that induces anabolic stimulation of chondrocytes (U.S. Pat. No. 6,500,854; EP 1.391211). However, most of these compounds show severe side effects and, consequently, there is a strong need for compounds that stimulate chondrocyte differentiation without severe side effects.

The present invention relates to the relationship between the function of selected proteins identified by the present inventors (hereinafter referred to as “TARGETS”) and anabolic stimulation of chondrocytes.

SUMMARY OF THE INVENTION

The present invention relates to a method for identifying compounds that induce cartilage-synthesizing processes, which lead to anabolic stimulation of chondrocytes, comprising contacting the compound with a polypeptide comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 55-82 and 198-391 or a functional fragment or derivative thereof, under conditions that allow said polypeptide to bind to the compound, and measuring a compound-polypeptide property related to the anabolic stimulation of chondrocytes.

The present invention also relates to expression inhibitory agents, pharmaceutical compositions comprising the same, methods for the in vitro production of cartilage tissue, and host cells expressing said agents.

Aspects of the present method include the in vitro assay of compounds using polypeptide of a TARGET, and cellular assays wherein TARGET inhibition is followed by observing indicators of efficacy, including collagen type II, alpha-1 (col2α1) and aggrecan levels.

Another aspect of the invention is a method of treatment or prevention of a condition involving de-differentiation of chondrocytes and/or loss of cartilage thickness, in a subject suffering or susceptible thereto, by administering a pharmaceutical composition comprising an effective cartilage formation-enhancing amount of a TARGET inhibitor.

A further aspect of the present invention is a pharmaceutical composition for use in said method wherein said inhibitor comprises a polynucleotide selected from the group of an antisense polynucleotide, a ribozyme, and a small interfering RNA (siRNA), wherein said inhibitor 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: 55-82 and 198-391 or a fragment thereof.

Another further aspect of the present invention is a pharmaceutical composition comprising a therapeutically effective cartilage formation-enhancing amount of a TARGET inhibitor or its pharmaceutically acceptable salt, hydrate, solvate, or prodrug thereof in admixture with a pharmaceutically acceptable carrier. The present polynucleotides and TARGET inhibitor compounds are also useful for the manufacturing of a medicament for the treatment of conditions involving de-differentiation of chondrocytes and/or cartilage thickness loss.

Furthermore, the invention relates also to diagnostic methods.

BRIEF DESCRIPTION OF THE DRAWINGS

FIG. 1. Quantification of Col2α1 expression in primary human chondrocytes 12 days post infection with the indicated viruses.

FIG. 2. Example of duplicate screening results results for Col2α1 expression of part of the SILENCESELECT® library.

FIG. 3 (A-N): Quantification of Col2α1 expression in primary human chondrocytes 12 days post infection with the indicated viruses.

FIG. 4. Quantification of alcian blue staining on human primary chondrocytes, 12 days after infection with the indicated viruses, compared to uninfected cells.

FIG. 5 (A-N): Quantification of aggrecan expression in primary human chondrocytes 12 days post infection with the indicated viruses.

FIG. 6 (A-K): On target analysis with different constructs targeting the indicated genes. The on target analysis is assessed through detection of the Col2α1 expression in primary human chondrocytes 12 days post infection with the indicated viruses. Data are represented as luminescence units. The different thresholds corresponding to the different infection volumes are indicated as a line. Increasing the infection volumes leads to an increased threshold.

FIG. 7 (A-L): GAG analysis on chondrocytes in alginate cell culture 10 days post infection. The data are represented as relative GAG levels to the average of KD control 1 and KD control 2, being Ad-siRNA targeting PTGER4 and GRM7 respectively. Two individual data points are shown for every condition.

FIG. 8 (A-L): Hydroxyproline analysis on chondrocytes in alginate cell culture 10 days post infection. The data are represented as relative hydroxyproline levels to the average of KD control 1 and KD control 2, being Ad-siRNA targeting PTGER4 and GRM7 respectively. Two individual data points are shown for every condition.

FIG. 9 (A-L): mRNA marker analysis on chondrocytes in alginate cell culture 10 days post infection. The cells are infected with either Ad5/ALPL and Ad5/BMP2 overexpressing viruses, and with an Ad-siRNA targeting the indicated gene. The data are represented as relative mRNA levels to the ALPL control.

DETAILED DESCRIPTION

The following terms are used herein in accordance with the following definitions:

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

The term “anabolic stimulation of chondrocytes” should be understood as inducing chondrogenesis or as inducing or enhancing the anabolic activity of chondrocytes. Anabolic stimulation takes place for instance by stimulating the synthesis of cartilage components, or inducing synthesis of components that are required for cartilage synthesis. “Anabolic stimulation of chondrocytes” may also be understood as a process in which the expression of the matrix Gla protein (MGP) is induced. Anabolic stimulation of chondrocytes may furthermore be understood as inducing the expression of cartilage derived retinoic acid sensitive protein (CD-RAP), as inducing the expression of cartilage oligomeric matrix protein (COMP), as inducing the expression of aggrecan 1 (agc1, also termed chondroitin sulfate proteoglycan core protein 1, or CSPG1), or as inducing synthesis of collagen II, also known as collagen, type II, alpha-1 (col2α1), collagen of cartilage, chondrocalcin, and collagen, type xi, alpha-3 (col11α3).

The term ‘antisense nucleic acid’ refers to an oligonucleotide that has a nucleotide sequence that interacts through base pairing with a specific complementary nucleic acid sequence involved in the expression of the target such that the expression of the gene is reduced. Preferably, the specific nucleic acid sequence involved in the expression of the gene is a genomic DNA molecule or mRNA molecule that encodes (a part of) the gene. This genomic DNA molecule can comprise regulatory regions of the gene, or the coding sequence for the mature gene.

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 TWEEN™, 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 ‘complementary to a nucleotide sequence’ in the context of antisense oligonucleotides and methods should be understood as sufficiently complementary to such a sequence as to allow hybridization to that sequence in a cell, i.e., under physiological conditions.

The term “condition” or “disease” means the overt presentation of symptoms (i.e., illness) or the manifestation of abnormal clinical indicators (e.g., 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 “de-differentiation” refers to a general process wherein chondrocytes differentiate away from a cell phenotype that synthesizes cartilage components. Such components include, but are not limited to, collagen II, aggrecan 1, versican, link protein, perlecan, SZP/lubricin, biglycan (DS-PGI), decorin (DS-PGII), epiphycan (DS-PGIII), fibromodulin, lumican, CILP, C-type lectin, fibronectin, PRELP, COMP (thrombospondin-5), thrombospondin-1 and -3, CMP (matrilin-1), matrilin-3, C-type lectin, fibronectin, condroadherin, tenascin-C, fibrillin, elastin, gp-39/YKL-40, matrix Gla protein/MGP, pleiotrophin, chondromodulin-I/SCGP, chondromodulin-II, CD-RAP, chondrocalcin, PARP, lysozyme, and phospholipase A2.

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. In particular, with regard to inducing anabolic stimulation of chondrocytes, the term “effective amount” is intended to mean an effective differentiation-promoting amount of a compound or agent that will bring about a biologically meaningful increase in the levels of chondrocyte markers, representative for the process of an increase in chondrocyte anabolism.

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

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 “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 ‘genetic antisense’ as used herein refers to the incorporation of antisense constructs complementary to sequences of genes into the genome of a cell. Such incorporation allows for the continued synthesis of the antisense molecule.

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 (e.g., C0t or R0t analysis) or formed between one nucleic acid sequence present in solution and another nucleic acid sequence immobilized on a solid support (e.g., 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, e.g., 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, and that may also result in a phenotypical cellular change.

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 “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 per cent, and in a special embodiment 100 percent of its base pairs. The polynucleotides include polyribonucleic acids, polydeoxyribonucleic acids, and synthetic analogues thereof. 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. A preferred polynucleotide embodiment comprises from about 10 to about 30 bases in length. A special embodiment of polynucleotide is the polyribonucleotide of from about 10 to about 22 nucleotides, more commonly described as small interfering RNAs (siRNAs). Another special embodiment are nucleic acids with modified backcartilages 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 ‘ribozymes’ as used herein relates to catalytic RNA molecules capable of cleaving other RNA molecules at phosphodiester bonds in a manner specific to the sequence.

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 “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.

The term ‘vectors’ also relates to plasmids as well as to viral vectors, such as recombinant viruses, or the nucleic acid encoding the recombinant virus.

The term “vertebrate cells” means cells derived from animals having vertera structure, including fish, avian, reptilian, amphibian, marsupial, and mammalian species. Preferred cells are derived from mammalian species, and most preferred cells are human cells. Mammalian cells include feline, canine, bovine, equine, caprine, ovine, porcine murine, such as mice and rats, and rabbits.

Applicants' Invention Based on TARGET Relationship to Anabolic Stimulation of Chondrocytes

As noted above, the present invention is based on the present inventors' discovery that TARGETS are factors in the up-regulation and/or induction of anabolic processes of chondrocytes. The term “TARGET” or “TARGETS” means the proteins identified in accordance with the assay described below to be involved in the induction of the anabolic stimulation of chondrocytes. The present inventors have identified such TARGETS 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 preferred TARGETS are identified as SEQ ID NOS. 55-82 and 198-391 in Table 1A. Table 1A lists the polypeptides, polynucleotides and knock-down target sequences of the present invention. Table 1B lists exemplary fragments of the TARGETS, SEQ ID NOS. 198-391. Table 1C lists exemplary KD target sequences useful in the practice of the present expression-inhibitory agent invention.

TABLE 1A Target DNA KD Hit Gene GenBank or Pro- SEQ ID KD Target Sequence Symbol Accession Name Class mRNA tein NO. H33- CCTGAATGTGACTGTGGAC DGKB- NM_020238 diacylglycerol  Kinase  1 55  84-91 025 (SEQ ID NO: 91) INCENP NM_004080 kinase, beta   2 56 NM_145695 90 kDa/inner  3 57 centromere pro- tein antigens 135/155 kDa H33- GACTGACTGGCCTGAAGGC ICK NM_016513 intestinal  Kinase  4 58  92-99 032 (SEQ ID NO: 92) NM_014920 cell (MAK-  5 59 like) kinase H33- GATCTACACCACCTTCATC GPR103 AF411117 G protein- GPCR  6 60 100-107 034 (SEQ ID NO: 101) NM_198179 coupled re-  7 61 ceptor 103 H33- GGTGTATGGGCTCATGTAC FZD1 NM_003505 frizzled  GPCR  8 62 108-114 041 (SEQ ID NO: 108) homolog 1 (Drosophila) H33- AGAACTGGGTGATGACAGC ELA1 NM_001971 elastase 1,  Protease  9 63 115-122 056 (SEQ ID NO: 116) pancreatic H33- ATGAACTCTGTGATCCAGC USP9Y NM_004654 ubiquitin  Protease 10 64 123-130 061 (SEQ ID NO: 123) specific pro- tease 9, Y- linked (fat facets-like, Drosophila) H33- TTGGAATTCCAGTGTACCC DUSP11 NM_003584 dual speci- Phospha- 11 65 131-138 082 (SEQ ID NO: 132) ficity phos- tase phatase 11 (RNA/RNP complex 1- interacting) H33- GCTAGTTATCGCCTACCTC DUSP3 NM_004090 dual speci- Phospha- 12 66 139-145 083 (SEQ ID NO: 139) ficity phos- tase phatase 3  (vaccinia virus phos- phatase VH1- related) H33- AGATTCCAGATGCAACCCC JAK1 SK185- Janus kinase  Kinase 13 67 146-154 096 (SEQ ID NO: 148) NM_002227 1 (a protein  14 68 tyrosine kinase) H33- CTGAACTACTGGTACAGCC ABCG1 NM_016818 ATP-binding  Trans- 15 69 155-162 107 (SEQ ID NO: 157) NM_004915 cassette, porter 16 70 NM_207174 sub-family  17 71 NM_207627 G (WHITE), 18 72 NM_207628 member 1 19 73 NM_207629 20 74 NM_207630 21 75 H33- GCGAATTCCACCAGCATTC SLC26A NM_052961 solute carrier  Trans- 22 76 163-175 130 (SEQ ID NO: 165) 8 family porter 26, member 8 H33- ACATTGACCAGGAAGTGAC GGTLA4 NM_178312 gamma- Enzyme 23 77 176-182 192 (SEQ ID NO: 178) NM_178311 glutamyltrans- 24 78 NM_080920 ferase-like  25 79 activity 4 H33- GAAGCTGAATTAGGGCTTC PDE1A NM_005019 phosphodi- PDE 26 80 183-190 217 (SEQ ID NO: 184) NM_00100- esterase 1A,  27 81 3683 calmodulin- dependent H33- GAAGCCATCTCCGACAATC SLC15A NM_021082 solute carrier  Trans- 28 82 191-197 279 (SEQ ID NO: 192) 2 family 15 porter (H+/peptide transporter),  member 2

TABLE 1B Seq Accession Name Protein Segment ID protein segment AF411117 GPR103 Extracellular domain 198 AF411117 GPR103 Transmembrane domain 199 AF411117 GPR103 Intracellular domain 200 AF411117 GPR103 Transmembrane domain 201 AF411117 GPR103 Extracellular domain 202 AF411117 GPR103 Transmembrane domain 203 AF411117 GPR103 Intracellular domain 204 AF411117 GPR103 Transmembrane domain 205 AF411117 GPR103 Extracellular domain 206 AF411117 GPR103 Transmembrane domain 207 AF411117 GPR103 Intracellular domain 208 AF411117 GPR103 Transmembrane domain 209 AF411117 GPR103 Extracellular domain 210 NM_198179 GPR103 Extracellular domain 211 NM_198179 GPR103 Transmembrane domain 212 NM_198179 GPR103 Intracellular domain 213 NM_198179 GPR103 Transmembrane domain 214 NM_198179 GPR103 Extracellular domain 215 NM_198179 GPR103 Transmembrane domain 216 NM_198179 GPR103 Intracellular domain 217 NM_198179 GPR103 Transmembrane domain 218 NM_198179 GPR103 Extracellular domain 219 NM_198179 GPR103 Transmembrane domain 220 NM_198179 GPR103 Intracellular domain 221 NM_198179 GPR103 Transmembrane domain 222 NM_198179 GPR103 Extracellular domain 223 NM_198179 GPR103 Transmembrane domain 224 NM_198179 GPR103 Intracellular domain 225 NM_003505 FZD1 Extracellular domain 226 NM_003505 FZD1 Transmembrane domain 227 NM_003505 FZD1 Intracellular domain 228 NM_003505 FZD1 Transmembrane domain 229 NM_003505 FZD1 Extracellular domain 230 NM_003505 FZD1 Transmembrane domain 231 NM_003505 FZD1 Intracellular domain 232 NM_003505 FZD1 Transmembrane domain 233 NM_003505 FZD1 Extracellular domain 234 NM_003505 FZD1 Transmembrane domain 235 NM_003505 FZD1 Intracellular domain 236 NM_003505 FZD1 Transmembrane domain 237 NM_003505 FZD1 Extracellular domain 238 NM_003505 FZD1 Transmembrane domain 239 NM_003505 FZD1 Intracellular domain 240 NM_016818 ABCG1 Extracellular domain 241 NM_016818 ABCG1 Transmembrane domain 242 NM_016818 ABCG1 Intracellular domain 243 NM_016818 ABCG1 Transmembrane domain 244 NM_016818 ABCG1 Extracellular domain 245 NM_016818 ABCG1 Transmembrane domain 246 NM_016818 ABCG1 Intracellular domain 247 NM_016818 ABCG1 Transmembrane domain 248 NM_016818 ABCG1 Extracellular domain 249 NM_016818 ABCG1 Transmembrane domain 250 NM_016818 ABCG1 Intracellular domain 251 NM_016818 ABCG1 Transmembrane domain 252 NM_016818 ABCG1 Extracellular domain 253 NM_016818 ABCG1 Transmembrane domain 254 NM_016818 ABCG1 Intracellular domain 255 NM_004915 ABCG1 Extracellular domain 256 NM_004915 ABCG1 Transmembrane domain 257 NM_004915 ABCG1 Intracellular domain 258 NM_004915 ABCG1 Transmembrane domain 259 NM_004915 ABCG1 Extracellular domain 260 NM_004915 ABCG1 Transmembrane domain 261 NM_004915 ABCG1 Intracellular domain 262 NM_004915 ABCG1 Transmembrane domain 263 NM_004915 ABCG1 Extracellular domain 264 NM_004915 ABCG1 Transmembrane domain 265 NM_004915 ABCG1 Intracellular domain 266 NM_004915 ABCG1 Transmembrane domain 267 NM_004915 ABCG1 Extracellular domain 268 NM_004915 ABCG1 Transmembrane domain 269 NM_004915 ABCG1 Intracellular domain 270 NM_207174 ABCG1 Extracellular domain 271 NM_207174 ABCG1 Transmembrane domain 272 NM_207174 ABCG1 Intracellular domain 273 NM_207174 ABCG1 Transmembrane domain 274 NM_207174 ABCG1 Extracellular domain 275 NM_207174 ABCG1 Transmembrane domain 276 NM_207174 ABCG1 Intracellular domain 277 NM_207174 ABCG1 Transmembrane domain 278 NM_207174 ABCG1 Extracellular domain 279 NM_207174 ABCG1 Transmembrane domain 280 NM_207174 ABCG1 Intracellular domain 281 NM_207174 ABCG1 Transmembrane domain 282 NM_207174 ABCG1 Extracellular domain 283 NM_207174 ABCG1 Transmembrane domain 284 NM_207174 ABCG1 Intracellular domain 285 NM_207627 ABCG1 Extracellular domain 286 NM_207627 ABCG1 Transmembrane domain 287 NM_207627 ABCG1 Intracellular domain 288 NM_207627 ABCG1 Transmembrane domain 289 NM_207627 ABCG1 Extracellular domain 290 NM_207627 ABCG1 Transmembrane domain 291 NM_207627 ABCG1 Intracellular domain 292 NM_207627 ABCG1 Transmembrane domain 293 NM_207627 ABCG1 Extracellular domain 294 NM_207627 ABCG1 Transmembrane domain 295 NM_207627 ABCG1 Intracellular domain 296 NM_207627 ABCG1 Transmembrane domain 297 NM_207627 ABCG1 Extracellular domain 298 NM_207627 ABCG1 Transmembrane domain 299 NM_207627 ABCG1 Intracellular domain 300 NM_207628 ABCG1 Extracellular domain 301 NM_207628 ABCG1 Transmembrane domain 302 NM_207628 ABCG1 Intracellular domain 303 NM_207628 ABCG1 Transmembrane domain 304 NM_207628 ABCG1 Extracellular domain 305 NM_207628 ABCG1 Transmembrane domain 306 NM_207628 ABCG1 Intracellular domain 307 NM_207628 ABCG1 Transmembrane domain 308 NM_207628 ABCG1 Extracellular domain 309 NM_207628 ABCG1 Transmembrane domain 310 NM_207628 ABCG1 Intracellular domain 311 NM_207628 ABCG1 Transmembrane domain 312 NM_207628 ABCG1 Extracellular domain 313 NM_207628 ABCG1 Transmembrane domain 314 NM_207628 ABCG1 Intracellular domain 315 NM_207629 ABCG1 Extracellular domain 316 NM_207629 ABCG1 Transmembrane domain 317 NM_207629 ABCG1 Intracellular domain 318 NM_207629 ABCG1 Transmembrane domain 319 NM_207629 ABCG1 Extracellular domain 320 NM_207629 ABCG1 Transmembrane domain 321 NM_207629 ABCG1 Intracellular domain 322 NM_207629 ABCG1 Transmembrane domain 323 NM_207629 ABCG1 Extracellular domain 324 NM_207629 ABCG1 Transmembrane domain 325 NM_207629 ABCG1 Intracellular domain 326 NM_207629 ABCG1 Transmembrane domain 327 NM_207629 ABCG1 Extracellular domain 328 NM_207629 ABCG1 Transmembrane domain 329 NM_207629 ABCG1 Intracellular domain 330 NM_207630 ABCG1 Extracellular domain 331 NM_207630 ABCG1 Transmembrane domain 332 NM_207630 ABCG1 Intracellular domain 333 NM_207630 ABCG1 Transmembrane domain 334 NM_207630 ABCG1 Extracellular domain 335 NM_207630 ABCG1 Transmembrane domain 336 NM_207630 ABCG1 Intracellular domain 337 NM_207630 ABCG1 Transmembrane domain 338 NM_207630 ABCG1 Extracellular domain 339 NM_207630 ABCG1 Transmembrane domain 340 NM_207630 ABCG1 Intracellular domain 341 NM_207630 ABCG1 Transmembrane domain 342 NM_207630 ABCG1 Extracellular domain 343 NM_207630 ABCG1 Transmembrane domain 344 NM_207630 ABCG1 Intracellular domain 345 NM_052961 SLC26A8 Intracellular domain 346 NM_052961 SLC26A8 Transmembrane domain 347 NM_052961 SLC26A8 Extracellular domain 348 NM_052961 SLC26A8 Transmembrane domain 349 NM_052961 SLC26A8 Intracellular domain 350 NM_052961 SLC26A8 Transmembrane domain 351 NM_052961 SLC26A8 Extracellular domain 352 NM_052961 SLC26A8 Transmembrane domain 353 NM_052961 SLC26A8 Intracellular domain 354 NM_052961 SLC26A8 Transmembrane domain 355 NM_052961 SLC26A8 Extracellular domain 356 NM_052961 SLC26A8 Transmembrane domain 357 NM_052961 SLC26A8 Intracellular domain 358 NM_052961 SLC26A8 Transmembrane domain 359 NM_052961 SLC26A8 Extracellular domain 360 NM_052961 SLC26A8 Transmembrane domain 361 NM_052961 SLC26A8 Intracellular domain 362 NM_052961 SLC26A8 Transmembrane domain 363 NM_052961 SLC26A8 Extracellular domain 364 NM_052961 SLC26A8 Transmembrane domain 365 NM_052961 SLC26A8 Intracellular domain 366 NM_052961 SLC26A8 Transmembrane domain 367 NM_052961 SLC26A8 Extracellular domain 368 NM_021082 SLC15A2 Intracellular domain 369 NM_021082 SLC15A2 Transmembrane domain 370 NM_021082 SLC15A2 Extracellular domain 371 NM_021082 SLC15A2 Transmembrane domain 372 NM_021082 SLC15A2 Intracellular domain 373 NM_021082 SLC15A2 Transmembrane domain 374 NM_021082 SLC15A2 Extracellular domain 375 NM_021082 SLC15A2 Transmembrane domain 376 NM_021082 SLC15A2 Intracellular domain 377 NM_021082 SLC15A2 Transmembrane domain 378 NM_021082 SLC15A2 Extracellular domain 379 NM_021082 SLC15A2 Transmembrane domain 380 NM_021082 SLC15A2 Intracellular domain 381 NM_021082 SLC15A2 Transmembrane domain 382 NM_021082 SLC15A2 Extracellular domain 383 NM_021082 SLC15A2 Transmembrane domain 384 NM_021082 SLC15A2 Intracellular domain 385 NM_021082 SLC15A2 Transmembrane domain 386 NM_021082 SLC15A2 Extracellular domain 387 NM_021082 SLC15A2 Transmembrane domain 388 NM_021082 SLC15A2 Intracellular domain 389 NM_021082 SLC15A2 Transmembrane domain 390 NM_021082 SLC15A2 Extracellular domain 391

TABLE 1C SEQ ID TARGET Name siRNA_Name KD Target Sequence NO. ABCG1 A150100-ABCG1_v5 NM_004915_idx1797 AGTGGATGTCCTACATCTC 155 A150100-ABCG1_v6 NM_004915_idx500 ATCATGCAGGATGACATGC 156 A150100-ABCG1_v7 NM_004915_idx1481 CTGAACTACTGGTACAGCC 157 A150100-ABCG1_v10 NM_004915_idx872 CAGCTTTACGTCCTGAGTC 158 A150100-ABCG1_v11 NM_004915_idx1067 TCAGACCACAAGAGAGACC 159 A150100-ABCG1_v12 NM_004915_idx1789 GTACCTACAGTGGATGTCC 160 A150100-ABCG1_v8 NM_016818_idx603 TGGTCAAGGAGATACTGAC 161 A150100-ABCG1_v9 NM_016818_idx718 CCCTCCAGTCATGTTCTTC 162 DGKB A150100-DGKB_v1 NM_004080_idx104 TTCCATGGTAATGGTGTGC  84 A150100-DGKB_v2 NM_004080_idx1064 CCTGAATGTGACTGTGGAC  91 A150100-DGKB_v3 NM_004080_idx2398 CCGAAGCAAGGAATAATCC  85 A150100-DGKB_v10 NM_145695_idx466 TATGTTTCGCCTTTATGAC  86 A150100-DGKB_v11 NM_145695_idx654 GGATTCAAGGAGGAATGAC  87 A150100-DGKB_v12 NM_145695_idx870 CTCCCTCTTGCATCAAGAC  88 A150100-DGKB_v13 NM_145695_idx1387 AAATCCTCGTCAGGTTTAC  89 A150100-DGKB_v14 NM_145695_idx1729 AGTGCCTTACAGTATCATC  90 DUSP11 A150100-DUSP11_v1 NM_003584_idx427 CAGAGGATTTGCCAGAAAC 131 A150100-DUSP11_v2 NM_003584_idx743 TTGGAATTCCAGTGTACCC 132 A150100-DUSP11_v3 NM_003584_idx945 CAGAGACACCATCTCCCTC 133 A150100-DUSP11_v4 NM_003584_idx885 ACCCAGACCCAAAGTTTGC 134 A150100-DUSP11_v5 NM_003584_idx221 AAGGTGGAAAGACTATCTC 135 A150100-DUSP11_v6 NM_003584_idx420 TATAAACCAGAGGATTTGC 136 A150100-DUSP11_v7 NM_003584_idx836 GTATAATCTACATCAGATC 137 A150100-DUSP11_v8 NM_003584_idx933 CCACATGTTTACCAGAGAC 138 DUSP3 A150100-DUSP3_v1 NM_004090_idx425 GCTAGTTATCGCCTACCTC 139 A150100-DUSP3_v2 NM_004090_idx300 GACACACAGGAGTTCAACC 140 A150100-DUSP3_v3 NM_004090_idx176 GCTGCAGAAACTAGGCATC 141 A150100-DUSP3_v4 NM_004090_idx248 TGCCAACTTCTACAAGGAC 142 A150100-DUSP3_v5 NM_004090_idx299 CGACACACAGGAGTTCAAC 143 A150100-DUSP3_v6 NM_004090_idx458 GATGGACGTCAAGTCTGCC 144 A150100-DUSP3_v7 NM_004090_idx4305 ACAGGAGTTCAACCTCAGC 145 ELA1 A150100-ELA1_v1 NM_001971_idx754 TAATGTCATCGCCTCCAAC 115 A150100-ELA1_v2 NM_001971_idx162 AGAACTGGGTGATGACAGC 116 A150100-ELA1_v3 NM_001971_idx421 CAACAGTCCCTGCTACATC 117 A150100-ELA1_v4 NM_001971_idx280 GATCGTGGTGCATCCATAC 118 A150100-ELA1_v5 NM_001971_idx230 GCGTGGATTACCAGAAGAC 119 A150100-ELA1_v6 NM_001971_idx459 TAACAACAGTCCCTGCTAC 120 A150100-ELA1_v7 NM_001971_idx669 CCATTGCTTGGTGAATGGC 121 A150100-ELA1_v8 NM_001971_idx692 ATTCTCTCCATGGAGTGAC 122 FZD1 A150100-FZD1_v10 NM_003505_idx1323 GGTGTATGGGCTCATGTAC 108 A150100-FZD1_v11 NM_003505_idx2058 CATCGTCATCGCCTGCTAC 109 A150100-FZD1_v9 NM_003505_idx2007 GCTCATGGTGCGCATTGGC 110 A150100-FZD1_v12 NM_003505_idx2229 GTACCTTATGACGCTGATC 111 A150100-FZD1_v13 NM_003505_idx3317 ACCTGGTATGGGTTTGGCC 112 A150100-FZD1_v14 NM_003505_idx3883 ATGTGTGCAGGTCTACTGC 113 A150100-FZD1_v15 NM_003505_idx2704 TTATTTAGGGCGGTTTAAC 114 GGTLA4 A150100-GGT1_v8 NM_080839_idx451 ACTGGCCATCATCTACAAC 176 A150100-GGTLA4_v5 NM_080920_idx292 TGCTCACCTGTCTGTGGTC 177 A150100-GGTLA4_v6 NM_080920_idx702 ACATTGACCAGGAAGTGAC 178 A150100-GGTLA4_v7 NM_080920_idx411 TGGATGACTTCAGCTCTAC 179 A150100-GGT1_v10 NM_178311_idx629 CTACAACCTCTGGTTCGGC 180 A150100-GGT1_v11 NM_178311_idx707 CACGACAGTGGAGAGAAAC 181 A150100-GGTLA4_v8 NM_178311_idx287 GTTCTACATGCCGGATGAC 182 GPR103 A150100-GPR103_v5 AF411117_idx611 AGGCACCAGGGACTTGTGC 100 A150100-GPR103_v6 AF411117_idx820 GATCTACACCACCTTCATC 101 A150100-GPR103_v7 XM_172359_idx288 TGGTGTTCTACGTGGTGAC 102 A150100-GPR103_v8 AF411117_idx136 TGTTAGGCGCCTGCATTGC 103 A150100-GPR103_v10 AF411117_idx424 CAACATCTTTATCTGCTCC 104 A150100-GPR103_v11 AF411117_idx662 CGAAGGGCTTTCACAATGC 105 A150100-GPR103_v12 AF411117_idx106 GTACTACGTTGTAGCCCAC 106 A150100-GPR103_v9 AF411117_idx186 TGCAGGCGCTTAACATTAC 107 ICK A150100-ICK_v1 NM_016513_idx870 GACTGACTGGCCTGAAGGC  92 A150100-ICK_v2 NM_016513_idx1665 GCAGCACTATTTGAAGCAC  93 A150100-ICK_v3 NM_016513_idx588 GCCTGAGAACCTCCTCTGC  94 A150100-ICK_v10 NM_016513_idx1027 ACAGCTAGTCAGGCACTTC  95 A150100-ICK_v11 NM_016513_idx1707 TATAAGAAATGGCATACTC  96 A150100-ICK_v12 NM_016513_idx1754 CTAATCCATGGTCTAGTTC  97 A150100-ICK_v8 NM_016513_idx504 GTCTGCTATAAGGAATATC  98 A150100-ICK_v9 NM_016513_idx713 AAGTACTCCTGAGGTCTAC  99 JAK1 A150100-JAK1_v1 oKD271 TTGGCATGGAACCAACGAC 146 A150100-JAK1_v2 oKD270 CCTCTTTGCCCTGTATGAC 147 A150100-JAK1_v7 oKD272 AGATTCCAGATGCAACCCC 148 A150100-JAK1_v8 SK185_idx1743 CATGAGCCAGCTGAGTTTC 149 A150100-JAK1_v9 SK185_idx142 GTGGAAGTGATCTTCTATC 150 A150100-JAK1_v12 NM_002227_idx1351 TGGCTGTCATGGTCCAATC 151 A150100-JAK1_v13 NM_002227_idx2512 CCGCTGCATGAACTATGAC 152 A150100-JAK1_v14 NM_002227_idx3093 TTGGAGACTTCGGTTTAAC 153 A150100-JAK1_v15 NM_002227_idx3269 TGTGATTCAGATTCTAGTC 154 PDE1A A150100-PDE1A_v5 NM_005019_idx913 AGGTATCATGCACTGGCTC 183 A150100-PDE1A_v6 NM_005019_idx1382 GAAGCTGAATTAGGGCTTC 184 A150100-PDE1A_v7 NM_005019_idx1709 CTGGTGGACATCATTCAGC 185 A150100-PDE1A_v10 NM_005019_idx1413 TTTGTGATCGGAAGTCAAC 186 A150100-PDE1A_v11 NM_005019_idx1601 ATTGCTGATGCACTAAGAC 187 A150100-PDE1A_v12 NM_005019_idx754 CAGATATGATCTTATCAAC 188 A150100-PDE1A_v13 NM_005019_idx887 ACTGTGCATTACATAATGC 189 A150100-PDE1A_v9 NM_005019_idx1073 CACGTGAGTGCAGCTTATC 190 SLC15A2 A150100-SLC15A2_v1 NM_021082_idx457 AGTCCTATCATTGATCGGC 191 A150100-SLC15A2_v2 NM_021082_idx121 GAAGCCATCTCCGACAATC 192 A150100-SLC15A2_v3 NM_021082_idx1166 ATGGCTGTTGGTATGATCC 193 A150100-SLC15A2_v4 NM_021082_idx1575 CCGTGAGGTTTGTTAACAC 194 A150100-SLC15A2_v5 NM_021082_idx423 TTGGGTGCCTTACCAATAC 195 A150100-SLC15A2_v6 NM_021082_idx1136 CTCCAAGTGTGGAATTAAC 196 A150100-SLC15A2_v7 NM_021082_idx1534 GCATGATGGTAAAGGATAC 197 SLC26A8 A150100- NM_052961_idx1925 TTCTGCAACTGTGATGATC 163 SLC26A8_v10 A150100- NM_052961_idx2288 GTACACTACGTGGATTCAC 164 SLC26A8_v11 A150100-SLC26A8_v2 NM_052961_idx923 GCGAATTCCACCAGCATTC 165 A150100-SLC26A8_v3 NM_052961_idx1761 TCTTCCAGTGCTGCAGCTC 166 A150100-SLC26A8_v4 NM_052961_idx2693 TCAGAACAAGAGGCTGGGC 167 A150100-SLC26A8_v5 NM_052961_idx1228 GAAGATTGCCAGTCTTCAC 168 A150100-SLC26A8_v6 NM_052961_idx457 GATTCCTCCTCTCAACATC 169 A150100-SLC26A8_v7 NM_052961_idx936 GCATTCTAGTATTTCTAAC 170 A150100-SLC26A8_v8 NM_052961_idx1249 TTACAGTGTCAATTCCAAC 171 A150100-SLC26A8_v9 NM_052961_idx1723 TGATTATCGGGAGATCATC 172 A150100- NM_052961_idx338 GAATGGATGTGTATGTATC 173 SLC26A8_v12 A150100- NM_052961_idx1105 TGACATGATTCCTTATAGC 174 SLC26A8_v13 A150100- NM_052961_idx1446 TCTACACACTGCCAAATGC 175 SLC26A8_v14 USP9Y A150100-USP9Y_v1 NM_004654_idx5651 ATGAACTCTGTGATCCAGC 123 A150100-USP9Y_v2 NM_004654_idx1600 AGGTTGGCTAGTGGATCTC 124 A150100-USP9Y_v3 NM_004654_idx2636 AAGTGGGTAATTCCTGCTC 125 A150100-USP9X_v4 NM_021906_idx1189 CGAATGGCAGAATGGATAC 126 A150100-USP9X_v5 NM_004654_idx7911 TCTGGCAGGTTGCATATTC 127 A150100-USP9Y_v4 NM_004654_idx1489 CTGCAAGTTTCATATCTAC 128 A150100-USP9Y_v5 NM_004654_idx2820 ATAGCATCAGATTGTATGC 129 A150100-USP9Y_v6 NM_004654_idx5731 TTTACACGATGATATGTTC 130

The present invention relates to a method for assaying for compounds that induce anabolic stimulation of chondrocytes, comprising contacting the compound with a polypeptide comprising an amino acid sequence of the polypeptides of SEQ ID NO: 55-82 (“TARGETS”) or a functional 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 induces anabolic stimulation of chondrocytes, the method comprising further:

    • (a) contacting a population of chondrocyte cells with one or more of said compound that exhibits binding affinity for said TARGETS, and
    • (b) measuring a compound-polypeptide property related to the anabolic stimulation of chondrocytes.

The compound-polypeptide property referred to above is related to the anabolic stimulation of chondrocytes, and is a measurable phenomenon chosen by the person of ordinary skill in the art. The measurable property may e.g. be the binding affinity for a peptide domain of the polypeptide TARGET or the level of any one of a number of biochemical marker levels of increased chondrocyte anabolism. Anabolic stimulation of chondrocytes can e.g. be measured by measuring the level of proteins and other molecules that are induced during the differentiation process, such as key components of normal cartilage. In particular, the induction of the major protein component of cartilage, collagen II, is measured.

In addition, compound-polypeptide properties related to the anabolic stimulation of chondrocytes are measured in C20/A4; T/C-28a2; T/C-28a4; C-28/12; Ch-4,8,N; Ch-8-OA; TC6; MCT; MC615; IRC; RCS2; Hig82; and D1 ORL UVA (D1) cells. However, such porterties are also measured in non-chondrocyte cell systems. For example, in situ binding assays that determine the affinity of compounds to bind to polypeptides of the invention are performed using any cell type that expresses the polypeptide. Expression of the polypeptide is exogenous or endogenous. Furthermore, when the compound-polypeptide property is activation of a biological pathway, any cell that contains the pathway cellular components is used to measure the compound-polypeptide property. For example, induction of col2α1 or aggrecan in chondrocytes is indicative of anabolic stimulation of chondrocytes. Specifically, non-chondrocyte cells can be engineered to contain a reporter molecule activated by the col2α1 or aggrecan promoters. In this way a non-chondrocyte can be used to measure a property indicative of anabolic stimulation of chondrocytes.

The invention relates to a method for identifying a compound that induces and/or increases anabolic stimulation of chondrocytes, said method comprising the steps of: culturing a population of cells expressing a polypeptide of any one of those listed in Table 1A, or a functional fragment or derivative thereof; determining a first level of chondrogenic differentiation in said population of cells; exposing said population of cells to a compound, or a mixture of compounds; determining the level of chondrogenic differentiation in said population of cells during or after exposure of said population of cells to the compound, or the mixture of compounds; and identifying the compound that induces and/or increases chondrogenic differentiation.

The invention also relates to a method for identifying a compound that decreases the expression and/or activity of any one of the polypeptides listed in Table 1A, said method comprising the steps of: culturing a population of cells expressing said polypeptide, or a fragment, or a derivative thereof; determining a first level of expression and/or activity of said polypeptide; exposing said population of cells to a compound, or a mixture of compounds; determining the level of expression and/or activity of said polypeptide during or after exposure of said population of cells to the compound, or the mixture of compounds; and identifying the compound that decreases the expression and/or activity of said polypeptide. If the polypeptide activity is not readily measurable, the identification of the compound may benefit from an extra step comprising exposing said population of cells to an agonist of said polypeptide. Furthermore, the methods of the present invention may comprise the step of introducing a gene encoding any one of the polypeptides listed in Table 1A, in said population of cells. For high-throughput purposes it may be beneficial to have the gene stably integrated in the genome of said cells.

In a preferred embodiment, the level of chondrocyte (re-)differentiation is determined by measuring the expression level of a marker gene, wherein a preferred marker gene encodes collagen type II, alpha-1 (col2α1) or aggrecan. For proper anabolic stimulation it is preferred that the expression and/or activity of col2α1 or aggrecan is increased.

The present invention provides in one particular embodiment methods for identifying novel compounds, wherein the polypeptide is a GPCR. If so, the expression and/or activity of said GPCR is preferably determined by measuring the level of a second messenger. Preferred second messengers are cyclic AMP, Ca2+ or both. Typically, the level of the second messenger is determined with a reporter gene under the control of a promoter that is responsive to the second messenger, wherein it is preferred that the promoter is a cyclic AMP-responsive promoter, an NF-KB responsive promoter, or a NF-AT responsive promoter, and wherein the reporter gene is selected from the group consisting of: alkaline phosphatase, GFP, eGFP, dGFP, luciferase and β-α galactosidase.

In another particular embodiment, the invention provides methods for identifying novel compounds, wherein the polypeptide is a kinase or a phosphatase. Preferably, the activity of said kinase or phosphatase is determined by measuring the level of phosphorylation of a substrate of said kinase or phosphatase.

In yet another particular embodiment, the invention provides methods for identifying novel compounds, wherein the polypeptide is a protease. Preferably, the activity of said protease is measured by determining the level of cleavage of a substrate of said protease.

Methods for determining second messenger levels, use of the reporter genes and second-messenger responsive promoters as well as phosphatase assays and protease assays are well known in the art and not further elaborated upon herein.

In a preferred embodiment, the compound that inhibits the polypeptide exhibits a binding affinity to the polypeptide of at most 10 micromolar.

In a preferred embodiment of the invention, the polypeptide TARGET comprises an amino acid sequence selected from the group consisting of SEQ ID NO: 55-82 and 198-391 (Tables 1A and 1B). In an especially preferred embodiment of the invention, the polypeptide TARGET comprises an amino acid sequence selected from the group consisting of SEQ ID NO: 55-82 (Table 1A).

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 induce the anabolic stimulation of chondrocytes. 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 increasing mean cartilage thickness when administered to a subject. Nonetheless, 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 up the biochemical pathway, such as cartilage component synthesis, assayed by measuring the amount of collagen II. Such second assay may be designed to confirm that the test compound, having binding affinity for the polypeptide, actually induces the anabolic stimulation of chondrocytes. 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 collagen II levels or aggrecan 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 binding affinity of the 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 (e.g. 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, i.e. in the range of 100 nM to 1 pM; a moderate to low affinity binding relates to a high Kd, IC50 and EC50 values, i.e. in the micromolar range.

The present assay method may also be practiced in a cellular assay, A host cell expressing TARGET can be a cell with endogenous expression or a cell over-expressing the TARGET e.g. 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. In such cellular assay, the biological activity of TARGET may be measured by following the production of cartilage component synthesis.

The present invention further relates to a method for identifying a compound that induces anabolic stimulation of chondrocytes, comprising:

    • (a) contacting a compound with a polypeptide comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 55-82 and 198-391;
    • (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 induces the synthesis of proteins that are a constituent of normal cartilage and/or that are required for the formation of cartilage.

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

Preferred drug candidate compounds are low molecular weight compounds. Low molecular weight compounds, i.e. 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 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, e.g. 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; 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 inducing anabolic stimulation of chondrocytes comprising contacting said cells with an expression inhibitory agent comprising a polynucleotide sequence that complements at least about 17 nucleotides of the polyribonucleotide comprising a nucleotide sequence selected from the group consisting of SEQ ID NO: 1-28. In a preferred embodiment the expression-inhibitory agent comprises a polynucleotide sequence that complements a nucleotide sequence selected from the group consisting of SEQ ID NO: 84-197 and 410.

Another aspect of the present invention relates to a method for inducing the anabolic stimulation of chondrocytes, comprising by contacting said cell 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 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 encoding a portion of a polypeptide comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 55-82. In a preferred embodiment the expression-inhibiting agent is complementary to a polynucleotide sequence selected from the group consisting of SEQ ID NO: 1-28. In an especially preferred embodiment the expression-inhibiting agent is complementary to a polynucleotide sequence selected from the group consisting of SEQ ID NO: 84-197 and 410.

An 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: 55-82, a small interfering RNA (siRNA, preferably shRNA,) that is sufficiently complementary to a portion of the polyribonucleotide coding for SEQ ID NO: 55-82, 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, complementary to a nucleotide sequence selected from the group consisting of SEQ ID NO: 1-28. In an especially preferred embodiment the expression-inhibiting agent is complementary to a polynucleotide sequence selected from the group consisting of SEQ ID NO: 84-197 and 410.

A special 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 coding for SEQ ID NO: 55-82, a small interfering RNA (siRNA, preferably shRNA,) that is sufficiently complementary to a portion of the polyribonucleotide coding for SEQ ID NO: 55-82, such that the siRNA, preferably shRNA, interferes with the translation of the TARGET polyribonucleotide to the TARGET polypeptide. Preferably the nucleotide sequence is complementary to a polynucleotide selected from the group consisting of SEQ ID NO: 1-28. In an especially preferred embodiment nucleotide sequence is complementary to a polynucleotide selected from the group consisting of SEQ ID NO: 84-197 and 410.

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: 1-28. For example, an antisense nucleic acid (e.g. 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: 1-28. Antisense oligonucleotides preferably comprise 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 10 to about 30 contiguous nucleotides complementary to a nucleic acid sequence selected from the sequences of SEQ ID NO: 1-28.

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 RN202-315NA 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 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: 1-28, preferably from the group of sequences described in SEQ ID No: 84-197 or 410, and an antisense strand of 17-23 nucleotides complementary to the sense strand. Exemplary sequences are described as sequences complementary to SEQ ID NO: 84-197 or 410. The most preferred siRNA comprises sense and anti-sense strands that are 100 per cent 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 siRNA 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: 83). 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 inducing anabolic stimulation of chondrocytes 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: 55-82, 198-391. 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: 55-82, and a small interfering RNA (siRNA) that is sufficiently homologous to a portion of the polyribonucleotide coding for SEQ ID NO: 55-82, 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.

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 Pr, P1, and trp promoters. Among the eukaryotic (including viral) promoters useful for practice of this invention are ubiquitous promoters (e.g. HPRT, vimentin, actin, tubulin), intermediate filament promoters (e.g. desmin, neurofilaments, keratin, GFAP), therapeutic gene promoters (e.g. MDR type, CFTR, factor VIII), tissue-specific promoters (e.g. 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 (e.g. steroid hormone receptor, retinoic acid receptor), tetracycline-regulated transcriptional modulators, cytomegalovirus immediate-early, retroviral LTR, metallothionein, SV-40, Ela, 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 (Felgner, 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 (Felgner 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, e.g., 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 (e.g., International Patent Publication WO 95/21931), peptides derived from DNA binding proteins (e.g., International Patent Publication WO 96/25508), or a cationic polymer (e.g., 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, e.g., transfection, electroporation, microinjection, transduction, cell fusion, DEAE dextran, calcium phosphate precipitation, use of a gene gun, or use of a DNA vector transporter (see, e.g., 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, cartilage formation-enhancing 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, e.g., 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, e.g., 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 cartilage formation-enhancing 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 a systemic or local decrease in mean cartilage thickness, or a susceptibility to the condition, comprising an effective cartilage formation-enhancing 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 (e.g. 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, e.g. 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, e.g., 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 TARGET tissues, complexed with cationic lipids, packaged within liposomes, or delivered to TARGET 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 enhancing cartilage formation, which comprise the administration to said subject a therapeutically effective amount of an expression-inhibiting agent of the invention. A further aspect of the invention relates to a method of treating or preventing a disease involving chondrocyte anabolic stimulation, comprising administering to said subject a cartilage formation-enhancing pharmaceutical composition as described herein.

Examples of diseases involving anabolic stimulation of chondrocytes that are treatable using the means and methods of the present invention include, but are not limited to osteoarthritis, rheumatoid arthritis, psoriatic arthritis, juvenile rheumatoid arthritis, gouty arthritis, septic or infectious arthritis, reactive arthritis, reflex sympathetic dystrophy, algodystrophy, Tietze syndrome or costal chondritis, fibromyalgia, osteochondritis, neurogenic or neuropathic arthritis, arthropathy, endemic forms of arthritis like osteoarthritis deformans endemica, Mseleni disease, and Handigodu disease; degeneration resulting from fibromyalgia, systemic lupus erythematosus, scleroderma, and ankylosing spondylitis. Furthermore, people suffering from congenital cartilage malformations, including hereditary chondrolysis, chondrodysplasias and pseudoachondrodysplasias, are likely to benefit from programs that result in anabolic stimulation of chondrocytes, and these diseases therefore may also be treated by using the methods and means of the present invention. Non-limiting examples of congenital cartilage malformation related diseases are microtia, anotia, and metaphyseal chondrodysplasia.

The polypeptides or the polynucleotides employed in the methods of the present invention 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 polypeptide of the present invention or the compound to facilitate separation of complexes from uncomplexed forms of the polypeptide, as well as to accommodate automation of the assay. Interaction (e.g., binding of) of the polypeptide of the present invention 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 polypeptide of the present invention can be “His” tagged, and subsequently adsorbed onto Ni-NTA microtitre plates, or ProtA fusions with the polypeptides of the present invention can be adsorbed to IgG, which are then combined with the cell lysates (e.g., (35S-labelled) and the candidate compound, and the mixture incubated under conditions favorable for complex formation (e.g., 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 protein of the present invention quantitated 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 polypeptide of the present invention or the compound can be immobilized utilizing conjugation of biotin and streptavidin. Biotinylated protein molecules of the present invention can be prepared from biotin-NHS (N-hydroxy-succinimide) using techniques well known in the art (e.g., biotinylation kit, Pierce Chemicals, Rockford, Ill.), and immobilized in the wells of streptavidin-coated 96 well plates (Pierce Chemical). Alternatively, antibodies reactive with the polypeptides of the present invention but which do not interfere with binding of the polypeptide to the compound can be derivatized to the wells of the plate, and the polypeptide of the present invention 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 polypeptide of the present invention, and the amount of complex trapped in the well can be quantitated.

Another embodiment of the present invention relates to a method for in vitro production of cartilage tissue, comprising the steps of contacting chondrocyte cells with a polynucleotide sequence comprising a sequence selected from the group consisting of sequences complementary to SEQ ID No: 1-28, preferably selected from the group consisting of sequences complementary to SEQ ID NO: 84-197 for a time sufficient to re-differentiate the chondrocytes thereby producing a cartilaginous matrix.

In a preferred embodiment, the method comprises the steps of:

    • (a) applying chondrocyte cells on a substrate to form a cellular substrate,
    • (b) introducing a polynucleotide comprising a nucleotide sequence selected from the group consisting of sequences complementary to SEQ ID No: 1-28, preferably selected from the group consisting of sequences complementary to SEQ ID NO: 84-197, for a time sufficient to re-differentiate the chondrocyte cells, thereby producing a cartilaginous matrix.

The invention thus provides a method for producing a substrate with a matrix grown thereon, which matrix may be used for the provision of load-bearing implants, including joint prostheses, such as artificial knee joints and finger joints, and maxillofacial implants. It can also be used for special surgery devices, such as spacers, or cartilage fillers, and for use in augmentation, obliteration or reconstitution of cartilage defects and damaged or lost cartilage.

The present invention also relates to a combination of a load-bearing implant (preferably coated with a matrix as described above) with a cartilage filler comprising a matrix as described.

The method of the invention is also very suitable in relation to revision surgery, i.e., when previous surgical devices require replacement.

Suitable cells are stem cells cells, including mesenchymal stem cells cells and in particular chondrosyte precursor cells. The mesenchymal stem cells, and especially the chondrosyte precursor cells are found to be very effective in the cartilage producing process when taken from their original environment. In addition, cells derived from cartilage biopsies of a subject may be cultured and utilized with the present invention.

The mesenchymal stem cells can be directly applied on the substrate or they can advantageously be multiplied in the absence of the substrate before being applied on the substrate. In the latter mode, the cells are still largely multipotent after multiplication and, for the purpose of the invention, they are still referred to as undifferentiated. Subsequently, the cells are allowed to differentiate. Differentiation can be induced or enhanced by the presence of suitable inductors, such as bone morphogenic proteins (BMP2; BM4; BMP7), transforming growth factor beta (TGFbeta), CDMP1 and CDMP2. Especially suitable inductors of differentiation are the expression inhibitory agents of the present invention.

The use of mesenchymal stem cells provides several advantages. Firstly, their lower differentiation implies a higher proliferation rate and allows the eventual functionality to be better directed and controlled. Moreover, culturing these cells not only produces the required cartilage matrix containing organic and inorganic components, but also results in the presence, in the culture medium and in the matrix, of several factors which are essential for growth of the tissue and for adaptation to existing living tissue. Also, the culture medium can be a source of active factors such as growth factors, to be used in connection with the implanting process. Furthermore, such undifferentiated cells are often available in large quantities and more conveniently than e.g., mature cartilage cells, and exhibit a lower morbidity during recovery. Moreover, the undifferentiated cells can be obtained from the patient for whom the implant is intended. The cartilage resulting from these cells is autologous to the patient and thus no immune response will be induced. Matrices as thick as 100 μm can be produced as a result of the use of undifferentiated cells.

The substrate on which the undifferentiated cells can be applied and cultured can be a metal, such as titanium, cobalt/chromium alloy or stainless steel, a bioactive surface such as a calcium phosphate, polymer surfaces such as polyethylene, and the like. Although less preferred, siliceous material such as glass ceramics, can also be used as a substrate. Most preferred are metals, such as titanium, and calcium phosphates, even though calcium phosphate is not an indispensable component of the substrate. The substrate may be porous or non-porous. The cells can be applied at a rate of e.g., 103-106 per cm2, in particular 104-2×105 cells per cm2.

The culture medium to be used in the method according to the invention can be a commonly known culture medium such as MEM (minimum essential medium). Advantageously, the medium can be a conditioned medium. In this context, a conditioned medium is understood to be a medium wherein similar cells have previously been incubated, causing the medium to contain factors such as polypeptides, secreted by the cells which are important for cell growth and cell differentiation.

The cells are cultured for a time sufficient to produce a matrix layer, e.g., a matrix layer having a thickness of at least 0.5 um, in particular from 1 up to 100 um, more in particular of 10-50 um. The cells may be contacted with the culture medium for e.g. 2-15 weeks, in particular 4-10 weeks.

The production of the matrix, when applied on a substrate, results in a continuous or quasi-continuous coating covering the substrate for at least 50%, in particular at least 80% of its surface area.

In yet another aspect of the invention, the invention provides a method for diagnosing a pathological condition involving chondrocyte de-differentiation, said method comprising the steps of: determining the nucleic acid sequence of any one of the genes encoding the polypeptides listed in Table 1A in a genomic DNA sample; comparing the sequence from step (a) with the nucleic acid sequence of a healthy subject; and identifying any difference(s) related to the pathological condition. Such differences may be further checked in in vitro assays applying similar marker genes as disclosed herein. Such assays will reveal the role of the gene or its encoded polypeptide in anabolic stimulation processes of chondrocytes. If such mutations are identified this knowledge can be further exploited in test-kits for diagnosis of similar diseases.

Still another aspect or the invention relates to a method for diagnosing a pathological condition involving chondrocyte anabolic stimulation 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: 55-82, 198-391 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. Clearly, the activity and/or expression levels of the target genes as disclosed herein may have an effect on anabolic stimulation of chondrocytes. It remains to be determined to what level the activity should be elevated to diagnose for the disease. However, by comparing levels found in patients, individuals without symptoms and clearly healthy individuals the skilled person may easily determine these relevant levels. Since the skilled person is now aware which polypeptides should be monitored, the present invention provides novel tools for test assays for such diagnostics. A prominent disease that may be controlled, checked and diagnosed by using the knowledge provided by the present invention is osteoarthritis.

The rate of chondrocyte anabolic stimulation can typically be measured by determining the deposition of cartilage, or cartilage components, or cartilage-containing extra-cellular matrix produced by the chondrocytes, in the medium. A cell-based ELISA, enzymatic assays, or other general techniques known in the art can be used to measure cartilage components, like the ones described in Walsh G., Proteins: Biotechnology and Biochemistry. John Wiley and Sons, 2001.

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

EXAMPLES Example 1 Development of a High-Throughput Screening Method for the Detection of Endogenous Collagen Type II, Alpha-1 (Col2α1) Principle of the Assay:

Normal human articular chondrocytes (NHAC's) that are grown in two-dimensional cultures become dedifferentiated and gradually cease to synthesize cartilage. They can be re-differentiated into anabolic, active chondrocytes in the presence of appropriate factors (e.g. BMP2). An assay to screen for such factors was developed by monitoring the levels of collagen type ii, alpha-1 (col2α1), a major constituent of normal cartilage. NHAC's are seeded in 384 well plates and 1 day after plating infected with individual siRNA adenoviruses (Ad-siRNA) from the SILENCESELECT® collection (see WO03/020931). Col2α1 deposition is determined at 14 days after the start of the infection (14 dpi.

Control Viruses

Ad-BMP2; described in WO 03/018799

BMP4; Ad5 dE1/E2A adenoviruses that mediate the expression of full length bone morphogenetic protein 4 pre-protein (see NP570912).

Ad-LacZ; referred to as pIPspAdApt6-lacZ in WO 02/070744

Ad-eGFP; referred to as pIPspAdApt6-eGFP in WO 02/070744

Ad-Empty; described in WO 02/070744

Development of the Assay

NHAC's were isolated from donors who died from unrelated causes, and were obtained after informed consent (Cambrex, Verviers, Belgium).

In a series of experiments, carried out in 384-well plates, several parameters are optimized: cell seeding density, multiplicities of infection (MOI) of control viruses (Ad-BMP2 or Ad-eGFP), duration of infection, toxicity, infection efficiency (using Ad-eGFP) and the day of readout.

Using Ad-BMP2 (BMP2 over-expression) as a positive control for assay development, the following protocol resulted in the highest dynamic range for the assay with the lowest standard deviation on the background signal:

NHAC's are seeded on day 0 at 1500 cells/well of a 384-well plate in 60 μl of DMEM/F12 (InVitrogen), containing 10% heat-inactivated fetal calf serum (FBS-HI) and Pen/Strep) and infected the next day with 2.5 μl of Ad-control-virus (Ad-BMP2 or Ad-eGFP; this corresponds to an assumed MOI of 2000). After 7 days, 10 μl of a 50 μg/ml 2-Phospho-L-ascorbic acid in assay culture medium is added to each well. Up-regulation of Col2α1 is read at 10 dpi: The medium is removed with a VacuSafe; 50 μl ice-cold MeOH is added with a multidrop and removed immediately with a VacuSafe; 80 μl of ice-cold MeOH is added with a multidrop to fixate the cells, and plates are incubated for 20 min at −20° C.; MeOH is removed with a VacuSafe; plates are air-dried for 20 min, followed by 2× washing with 80 μl of phosphate buffered saline (PBS); 75 μl of blocking buffer (0.1% casein in PBS) is added and plates are incubated for at least 2 h at room temperature (RT); blocking buffer is removed; cells are washed with 25 μl of EC buffer (20 mM sodium phosphate, 2 mM EDTA, 400 mM NaCl, 0.2% BSA, 0.05% CHAPS, 0.4% casein, 0.05% NaN3, pH 7) and 35 μl of the primary antibody (Collagen II Ab-2 Neomarkers. Catalogus number MS-235-P) diluted 1/450, 1/225 in buffer C (20 mM sodium phosphate, 2 mM EDTA, 400 mM NaCl, 1% BSA, pH 7)) is added with a multichannel pipette; plates are incubated overnight at 4° C.; primary antibody is removed; cells are washed twice with 80 μl of PBST (0.5% Tween 20 in PBS) and once with 80 μl PBS; 35 μl of the secondary antibody (Goat-anti-mouse Immunoglobulins/HRP. DAKO. Catalogus number PO477; diluted 1/2000 in buffer C) is added with multichannel pipette; plates are incubated at RT for 1 h; secondary antibody is removed and cells are washed twice with 80 μl PBST and once with 80 μl PBS; 50 μl of luminol substrate is added and after 5 minutes read-out is determined on a luminometer.

After optimization of the assay (see FIG. 1), a 384 well control plate is prepared that contains positive control viruses (BMP2 and BMP4) and neutral viruses (eGFP; lacZ and empty, see FIG. 1). Aliquots of the control plate are prepared and frozen at −20° C. A control plate is thawed and taken along in every screening batch.

Example 2 Screening of 9216 Adenoviral siRNA Vectors in the Chondrogenesis Assay

The optimized protocol for screening the SILENCESELECT® library runs as follows: on day 0, propagated human primary chondrocytes are seeded in Greiner white, flat bottom, TC-treated 384 well plates with clear bottom (Catalogue number 781080) in 60 μl medium at a density of 1500 cells per well. One day later, 2.5 μl Ad-siRNA virus from each well of the SILENCESELECT® collection (WO 03/020931), stored in 384 well plates (estimated titer of 1×109 viral particles per ml) is transferred with the aid of a 96/384 channel dispenser (Tecan Freedom 200 equipped with TeMO96, TeMO384 and RoMa, Tecan AG, Switzerland) to individual wells of the 384 well plates containing chondrocytes. The control plate is run under the same conditions as the aliquot plates from the SILENCESELECT® collection. All Ad-siRNA viruses are screened in duplicate on independent assay plates. After infection, plates are incubated at 37° C. Seven days post infection the medium containing the adenoviruses is replaced by fresh medium. Thirteen days post infection, the amounts of col2α1 depositions per well is determined with the cELISA method. A typical result of a 384 well screening plate is depicted in FIG. 2.

The duplicate screen is repeated once. Ad-siRNA viruses are nominated as hits if at least 2 data points of the four tested (two times screened in duplicate) score above threshold. Threshold is set at average plus 2.5 times standard deviation of all data points per plate.

A total of 282 hits are isolated that scored above the threshold, representing 274 independent genes. A representative example is provided in FIG. 2, in which the “times standard deviation” of duplicate data points are indicated on the X-axis and Y-axis. The threshold (2.5 times standard deviation) is indicated by dotted lines. Negative values indicate data points that scored below average.

The results for some of the genes are shown in FIG. 3. A clear induction of the collagen II levels is observed upon infection of the Ad-siRNA targeting the indicated gene. The data are represented as relative light units (rlu) correlating to collagen II levels.

Example 3 Propagation of Hits

The 282 Ad-siRNA hits are subjected to further analysis to establish their therapeutic potential to induce chondrocyte anabolic stimulation. A first step entails a quality control on the Ad-siRNA selected for further analysis (this example). Next steps are the screening of the targets in other assays to validate their role in chondrocyte anabolic stimulation such as the induction of aggrecan, another main constituent of cartilage besides collagen II (Example 4), the ability to induce chondrocyte anabolic stimulation in chondrocytes from other donors (Example 5), the induction of a correct marker profile in three-dimensional chondrocyte cultures (example 11), the presence of posttranslational modifications on aggrecan (example 9) and collagen II (example 10) in three-dimensional chondrocyte cultures, the development of additional Ad-siRNAs targeting the identified transcripts (example 7), and confirmation that the corresponding genes are indeed expressed in residing chondrocytes (example 12).

To propagate the 282 hits of the chondrogenesis assay, 2.25×104 PerC6.E2A cells are seeded in 200 μl of DMEM containing 10% non-heat inactivated FCS into each well of a 96 well plate and incubated overnight at 39° C. in a humidified incubator at 10% CO2. Subsequently, 1 μl of crude lysate from the siRNA adenovirus stocks in matrix tubes is added and incubation proceeds at 34° C. in a humidified incubator at 10% CO2 for 7 days. All hits are propagated in duplicate on two independent plates. The two lysates are pooled and aliquots are frozen at −20° C.

The propagated Ad-siRNAs are re-screened at three MOI's in the chondrogenesis assay in duplicate (see Example 1). The Ad-siRNAs have to score at least once above threshold (average+2.5 times standard deviation) to pass this quality control step.

Example 4 Aggrecan Induction

A second assay to screen for chondrocyte anabolic factors is developed by monitoring the levels of aggrecan, another major constituent of cartilage. In this assay, glycosaminoglycans on aggrecan are stained by Alcian blue. NHACs are seeded in 384 well plates and 1 day after plating infected with individual Ad-siRNA from the SILENCESELECT® collection. Aggrecan deposition is determined at 14 days post infection. Using Ad-BMP2 as a positive control, we confirm in a series of experiments that several parameters optimized for the ColII cELISA assay are also applicable for the Alcian blue stain assay for aggrecan. These parameters include cell seeding density, MOIs of control viruses, duration of infection, and the day of readout.

NHAC's are seeded on day 0 at 1500 cells/well of a 384 well black-plate with clear bottom in 60 μl of DMEM/F12, containing 10% FBS-HI and Pen/Strep and infected the next day with 2.5 μl of Ad-BMP2 or Ad-eGFP; at an MOI of 2000. After 7 days, 10 μl of a 50 μg/ml 2-Phospho-L-ascorbic acid in assay culture medium is added to each well. Up-regulation of aggrecan is read at 10 dpi with Alcian blue staining: The medium is removed with a VacuSafe; 50 μl ice-cold MeOH is added with a multidrop and removed immediately with a VacuSafe; 80 μl of ice-cold MeOH is added with a multidrop to fixate the cells, and plates are incubated for 20 min at −20° C.; MeOH is removed with a VacuSafe; plates are air dried for 20 min. After washing once with 80 μl of PBS; 80 μl of 0.05% Alcian blue stain buffer (0.05% Alcian blue, Sigma, catalog number S-2889; 0.4 M MgCl2/25 mM sodium acetate, pH5.5) is added and plates are incubated overnight at RT. The next day cells are washed subsequently in 80 μl of 3% acetic acid, 25% ethanol/3% acetic acid, and 50% ethanol/3% acetic acid. Solutions are added with a multidrop and removed with a Vacusafe. After replacing 50% ethanol/3% acetic acid with 70% ethanol/3% acetic acid, each individual well is photographed with a SONY CCD camera, images are analyzed using a Galapagos Alcian blue quantification algorithm based on the separation of the blue signal through a color threshold procedure after a noise reduction filter. The amount of blue staining, which is proportional to aggrecan content, is expressed in pixel unit (see FIG. 4).

The propagated hits from Example 3 are used to transduce NHAc cells at three MOI's in duplicate in the chondrogenesis assay (see Example 1). The Ad-siRNAs have to score in duplicate in at least one MOI above threshold (average+2.5× standard deviation) to pass this secondary assay. In total, 101 out of 282 hits passed the Alcian blue assay for aggrecan (see Table 1). The results for some of the genes are shown in FIG. 5. Values represent the numerical output of the described algorithm and are correlated to Alcian blue staining levels. A clear induction of the aggrecan levels is observed upon infection of the Ad-siRNA targeting the indicated gene. The Ad-siRNA targeting FZD1 does not seem to induce Alcian blue staining.

TABLE 1 Overview of the 101 target sequences and their respective KD target sequences corresponding to the genes encoding the different polypeptides involved in chondrogenic differentiation. The GenBank numbers for the polypeptides and target gene symbols (general names) are also given. KD Target Sequence Hit KD Target Gene GenBank SEQ ID ID Sequence Symbol Accession Name Class NO H33- ATAAGCGGTTATCA PCTK2 NM_002595 PCTAIRE protein Kinase 392 006 CTGCC kinase 2 H33- GCTGGGATTCCAAG RYK SK340- RYK receptor-like Kinase 393 007 TGGAC NM_002958 tyrosine kinase H33- AACTGTGCAGGGC NTRK1 NM_002529 neurotrophic Kinase 394 008 CTCTCC tyrosine kinase, receptor, type 1 H33- GCTGCTGGATGTCA CDK2 NM_052827- cyclin-dependent Kinase 395 009 TTCAC NM_001798 kinase 2 H33- AGAGACACAGTGC PCK1 NM_002591 phosphoenolpyru- Kinase 396 010 CCATCC vate carboxykinase 1 (soluble) H33- ACTGAACCTCCGAA NEK4 SK256- NIMA (never in Kinase 397 011 ATGCC NM_003157 mitosis gene a)- related kinase 4 H33- GTGCTGGAGTGCTT MAPK NM_004635 mitogen-activated Kinase 398 013 CCATC APK3 protein kinase- activated protein kinase 3 H33- TTCAGACCTACCTT UMP- NM_016308 UMP-CMP kinase Kinase 399 020 CAGTC CMPK H33- CCTGAATGTGACTG DGKB- NM_020238- diacylglycerol Kinase  91 025 TGGAC INCEN NM_004080- kinase, beta P NM_145695 90 kDa/inner centromere protein antigens 135/155 kDa H33- GAGTCACACAGAG ROCK1 NM_005406 Rho-associated, Kinase 400 027 ATGAGC coiled-coil containing protein kinase 1 H33- CGATGTGCCTTCAA PRKCN SK489- protein kinase C, Kinase 401 028 GATTC NM_005813 nu H33- CAGTGGTTTGGGAA PLK4- SK341- serine/threonine Kinase 402 031 TCTGC STK18 NM_014264 kinase 18 (STK18)/polo- like kinase 4 (Drosophila) H33- GACTGACTGGCCTG ICK NM_016513- intestinal cell Kinase  92 032 AAGGC NM_014920 (MAK-like) kinase H33- GATCTACACCACCT GPR10 AF411117- G protein-coupled GPCR 101 034 TCATC 3 NM_198179 receptor 103 H33- GTGACTACACAAG CCR2 NM_000647 chemokine (C-C GPCR 403 036 GACTCC motif) receptor 2 H33- GACTGATTCGCTCT FPRL2 NM_002030 formyl peptide GPCR 404 040 TTGCC receptor-like 2 H33- GGTGTATGGGCTCA FZD1 NM_003505 frizzled homolog 1 GPCR 108 041 TGTAC (Drosophila) H33- AGTGCAGCCTTGTG P2RY1 NM_198333- purinergic GPCR 405 042 GGTTC 0 NM_014499 receptor P2Y, G- protein coupled, 10 H33- TAACACTCACTGCA EMR3 NM_152939- egf-like module GPCR 406 044 CCTGC NM_032571 containing, mucin- like, hormone receptor-like 3 H33- TAACTGAAACTCAG PROZ NM_003891 protein Z, vitamin Protease 407 049 CTAGC K-dependent plasma glycoprotein H33- ACTGAAGTAGCCCT THRB NM_000461 thyroid hormone NHR 408 054 CCTTC receptor, beta (erythroblastic leukemia viral (v- erb-a) oncogene homolog 2, avian) H33- AGAACTGGGTGAT ELA1 NM_001971 elastase 1, Protease 116 056 GACAGC pancreatic H33- AGTGCAGTACAGC COL7A NM_000094 collagen, type VII, Not  409 058 GATGAC 1 alpha 1 classified (epidermolysis bullosa, dystrophic, dominant and recessive) H33- TTCACATCGCTGAG CPZ NM_003652 carboxypeptidase Z Protease 410 059 CACCC H33- ATGAACTCTGTGAT USP9Y NM_004654 ubiquitin specific Protease 123 061 CCAGC protease 9, Y- linked (fat facets- like, Drosophila) H33- AGCCAGCAACGAC CST3 NM_000099 cystatin C Not  411 063 ATGTAC (amyloid classified angiopathy and cerebral hemorrhage) H33- GCTGCTGGGCATGT LNPEP NM_005575 leucyl/cystinyl Protease 412 065 CCTTC aminopeptidase H33- TGTGATCGTCATCA NLGN1 NM_014932 neuroligin 1 Enzyme 413 066 CAGTC H33- AACATGATATGTGC KLK10 NM_145888- kallikrein 10 Protease 414 068 TGGAC NM_002776 H33- CTGAGAAGGCTTCC LOC11 XM_061692 similar to Protease 415 069 ACTGC 9795 glutamyl aminopeptidase (aminopeptidase A); gp160 H33- TGATACGTGGATCC LOC12 NM_178453 similar to distal Protease 416 070 AGGCC 4221- intestinal serine MGC52 protease 282 H33- CTACAGTGACAAG OVTN- XM_089945- (similar to) Protease 417 072 GCTAAC LOC15 oviductin protease 9938 NM_198185 H33- GAACTGGATAGCC LOC20 XM_116274- similar to Protease 418 073 CTCATC 6008- XM_371709 KIAA1214 protein/ RNF15 ring finger 0 protein 150 H33- CCCTGGTAAAGCTG LOC22 XM_166659 similar to Protease 419 074 CATTC 0213 evidence: NAS/ pothetical hyprotein/putative H33- GATGAAGGCTTCG XYLB NM_005108 xylulokinase Kinase 420 076 GGCTTC homolog (H. influenzae) H33- TGTAAAGCTGGAA PTEN NM_000314- putative protein Phosphatase 421 080 AGGGAC BC038293- tyrosine AF017999 phosphatase homologue H33- CTGAAGAAGCTGG PTPN2 NM_015466 protein tyrosine Phosphatase 422 081 AGTTGC 3 phosphatase, non- receptor type 23/ protein tyrosine phosphatase TD14 H33- TTGGAATTCCAGTG DUSP1 NM_003584 dual specificity Phosphatase 132 082 TACCC 1 phosphatase 11 (RNA/RNP complex 1- interacting) H33- GCTAGTTATCGCCT DUSP3 NM_004090 dual specificity Phosphatase 139 083 ACCTC phosphatase 3 (vaccinia virus phosphatase VH1- related) H33- TCCTTGCAGCAGGC SLC24 NM_004727 solute carrier Ion Channel 423 084 ACATC A1 family 24 (sodium/potassium/ calcium exchanger), member 1 H33- TCTGTGCGTGGACT GABRP NM_014211 gamma- Ion Channel 424 092 GGAAC aminobutyric acid (GABA) A receptor, pi H33- CTTTGCTCGGAAGA RAF1 NM_002880 v-raf-1 murine Kinase 425 095 CGTTC leukemia viral oncogene homolog 1 H33- AGATTCCAGATGCA JAK1 SK185- Janus kinase 1 (a Kinase 148 096 ACCCC NM_002227 protein tyrosine kinase) H33- GAAGGCTTTGGAA LOC16 XM_094437- hypothetical Kinase 426 098 AGTGTC 7359- NM_153361 protein MGC42 MGC42105 105 H33- GTGAACTCTGCTGC PKD1L XM_091397- similar to GPCR 427 102 GACTC 3- NM_181536 KIAA1879 protein/ LOC16 polycystic 2163 kidney disease 1- like 3 H33- GACAAGGCTATGA RPS6K NM_004586 ribosomal protein Kinase 428 104 TGCTGC A3 S6 kinase, 90 kDa, polypeptide 3 H33- GGATGTGTGGTGCT RBKS NM_022128 ribokinase Kinase 429 105 GTCAC H33- CTGAACTACTGGTA ABCG1 NM_016818- ATP-binding Transporter 157 107 CAGCC NM_004915 cassette, sub- family G (WHITE), member 1 H33- CTCTGTGTTCCACT DPYD NM_000110 dihydropyrimidine Enzyme 430 108 TCGGC dehydrogenase H33- CAGCAATGCAGAG TNFRS NM_001561 tumor necrosis Other 431 110 TGTGAC F9 factor receptor drugable or superfamily, secreted member 9 H33- CAAAGCTGGCTACT TNFSF NM_172014- tumor necrosis Other 432 114 ACTAC 14 NM_003807 factor (ligand) drugable or superfamily, secreted member 14 H33- CAGTGCAAAGAGC GAPDS NM_014364 glyceraldehyde-3- Enzyme 433 117 CCAAAC phosphate dehydrogenase, testis-specific H33- GTATTCTGTACACC RDH11 NM_016026 retinol Enzyme 434 118 CTGGC dehydrogenase 11 (all-trans and 9- cis) H33- GTGATCGACAGGA PRKAG NM_017431 protein kinase, Kinase 435 120 TTGCTC 3 AMP-activated, gamma 3 non- catalytic subunit H33- GCGAATTCCACCAG SLC26 NM_052961 solute carrier Transporter 165 130 CATTC A8 family 26, member 8 H33- CACAGTGAAACCTT B4GAL NM_004776 UDP- Enzyme 436 138 CCTGC T5 Gal: betaGlcNAc beta 1,4- galactosyl- transferase, polypeptide 5 H33- ATCTGTGACACTGG LOC12 XM_064820 similar to Aldose Enzyme 437 145 ATCGC 5836 Reductase (E.C.1.1.1.21) H33- AGAGACTGGAGTT GNPN NM_198066 glucosamine- Enzyme 438 147 GTCAGC AT1 phosphate N- acetyltransferase 1 H33- CCTGAGTTGAATGT CYP17 NM_000102 cytochrome P450, Cytochrome  39 152 CATAC A1 family 17, P450 subfamily A, polypeptide 1 H33- CTGAACTAGTGACT MAGI- NM_152900- membrane- Kinase 440 158 ATCCC 3 NM_020965 associated guanylate kinase- related H33- ATAAGCACCGTGA LOC13 XM_071222 similar to Cytochrome 441 161 GCGACC 8967 cytochrome P450 P450 1A1 H33- CATTGGGCCACAG ADOR NM_000674 adenosine A1 GPCR 442 167 ACCTAC A1 receptor H33- GATGAAGACAGCA OPRK1 NM_000912 opioid receptor, GPCR 443 168 ACCAAC kappa 1 H33- AGCATATGATGACC CTSC NM_148170- cathepsin C Protease 444 175 TTGGC NM_001814 H33- ATTCCACTACTACA H105E3 NM_015922 NAD(P) Enzyme 445 180 GCTGC dependent steroid dehydrogenase- like H33- GAAACTGTGGCAG LOC25 XM_171056 similar to Putative Kinase 446 182 GCTAAC 6519 serine/threonine- protein kinase D1044.3 in chromosome III H33- CTGATGAAGGCCTT LOC12 XM_063593 similar to NADH- Enzyme 447 186 CGACC 3326 ubiquinone oxidoreductase PDSW subunit (Complex I- PDSW) (CI- PDSW) H33- TTGAAACAAGAGG ACYP1 XM_370768- (similar to) Phosphatase 448 188 AAGTCC NM_203488- acylphosphatase 1, NM_001107 erythrocyte (common) type H33- TGAACTTGCTCTGA KCNJ1 NM_170720- potassium Ion Channel 449 190 GCTGC 4 NM_013348 inwardly- rectifying channel, subfamily J, member 14 H33- ATCTGTAACCTCAG PPP3C S46622- protein Phosphatase 450 191 CACAC C NM_005605 phosphatase 3 (formerly 2B), catalytic subunit, gamma isoform (calcineurin A gamma) H33- ACATTGACCAGGA GGTLA NM_178312- gamma- Enzyme 178 192 AGTGAC 4 NM_178311- glutamyltransferase- NM080920 like activity 4 H33- GAAGCTAAGCCTC PIK4C NM_058004 phosphatidylinositol Kinase 451 202 GGTTAC A 4-kinase, catalytic, alpha polypeptide H33- TAACCGTGGCATCT TPP2 NM_003291 tripeptidyl Protease 452 204 ACCTC peptidase II H33- TGACCACCTGGAGT CST11 NM_130794 cystatin 11 Not 453 205 ATCAC classified H33- GTGGACATCTTTGA GRIK4 NM_014619 glutamate Ion Channel 454 208 GCTTC receptor, ionotropic, kainate 4 H33- GCTGAGAAGTACTT ARHG NM_014448 Rho guanine Other 455 209 CCACC EF16 exchange factor drugable or (GEF) 16 secreted H33- AGACTACTGCAAG STK23 NM_014370 serine/threonine Kinase 456 210 GGCGGC kinase 23 H33- GAGTATTTGCTGGC SLCO1 XM_372282- solute carrier Protease 457 213 ATTCC A2- NM_134431- organic anion CRLF2 NM_022148- transporter family, NM_021094 member 1A2/ cytokine receptor- like factor 2 H33- GAAGCTGAATTAG PDE1A NM_005019 phosphodiesterase PDE 184 217 GGCTTC 1A, calmodulin- dependent H33- GGAGACACGGAAT PPP1R1 NM_032105- protein Phosphatase 458 219 AAACTC 2B NM_002481 phosphatase 1, regulatory (inhibitor) subunit 12B H33- CCGAGACCACCTCA ACAD8 NM_014384 acyl-Coenzyme A Enzyme 459 222 ATGTC dehydrogenase family, member 8 H33- ATGGACATCTCCAC PTPRN NM_002846 protein tyrosine Phosphatase 460 223 GGGAC phosphatase, receptor type, N H33- TATCCTGACCTTCC KCNG1 NM_172318- potassium voltage- Ion Channel 461 230 TGCGC NM_002237 gated channel, subfamily G, member 1 H33- CACATGATCAAGCT LOC22 XM_055551 similar to Heat Kinase 462 236 AGGTC 0763 shock protein HSP 90-beta (HSP 84) (HSP 90) H33- GAAGCCAGGCATC SPOCK NM_014767 sparc/osteonectin, Enzyme 463 237 TTCATC 2 cwcv and kazal- like domains proteoglycan (testican) 2 H33- GCTGAAGTTATCCA PTPN1 NM_080685- protein tyrosine Phosphatase 464 238 GTCTC 3 NM_080684- phosphatase, non- NM_080683- receptor type 13 NM_006264 (APO-1/CD95 (Fas)-associated phosphatase) H33- AGCATTGGACCAGT GABR NM_173536 gamma- Ion Channel 465 239 TGATC G1 aminobutyric acid (GABA) A receptor, gamma 1 H33- GTGATCTACGTGAA DPP3 NM_005700 dipeptidylpeptidase Protease 466 243 CTGGC 3 H33- GCCGACAGTGGTG LYPLA NM_012320 lysophospholipase Enzyme 467 245 CACTAC 3 3 (lysosomal phospholipase A2) H33- AACATGATGGCTCA CTSE NM_148964- cathepsin E Protease 468 251 GAACC NM_001910 H33- TACAGTGATGGATC SULT1 NM_014465 sulfotransferase Enzyme 469 253 ATAGC B1 family, cytosolic, 1B, member 1 H33- ACCAATATGCCTAC KLKB1 NM_000892 kallikrein B, Protease 470 255 CTTCC plasma (Fletcher factor) 1 H33- ACTGTATCCCAGCA SENP7 NM_020654 sentrin/SUMO- Protease 471 258 GTCCC specific protease H33- AAGCTGAACATAA PTPRR NM_002849 protein tyrosine Phosphatase 472 259 CCTTGC phosphatase, receptor type, R H33- TTGAATAGCTCGGT LOC16 XM_095455 similar to Kinase 473 261 GTCCC 9014 Mitogen-activated protein kinase 6 (Extracellular signal-regulated kinase 3) (ERK-3) (MAP kinase isoform p97) (p97-MAPK) H33- GTGGAAGGCAAGA ABCD1- XM_372940- ATP-binding Transporter 474 263 TCTTCC LOC38 XM_370972- cassette, sub- 8253- NM_000033 family D (ALD), LOC39 member 1/similar 1403 to Adrenoleukodys- trophy protein (ALDP) H33- TGTATGGCTGGTCG ABCA7 NM_033308- ATP-binding Transporter 475 264 ATCAC NM_019112 cassette, sub- family A (ABC1), member 7 H33- GCTGCGACAACTTC GPR11 NM_153840 G protein-coupled GPCR 476 269 TGTTC 0 receptor 110 H33- GCCCACGGTCTTCC ACPT NM_080791- acid phosphatase, Phosphatase 477 276 ACTAC NM_080789- testicular NM_033068 H33- GAAGCCATCTCCGA SLC15 NM_021082 solute carrier Transporter 192 279 CAATC A2 family 15 (H+/peptide transporter), member 2 H33- GACTGAATCAGGC PPIH NM_006347 peptidyl prolyl Enzyme 478 295 CTTCCC isomerase H (cyclophilin H)

Example 5 Donor Dependency

The 282 hits identified by the ColII cELISA assay are further subject to a donor dependency test to demonstrate that the induction of ColII production by a given hit is not restricted to a single donor. In addition to the 11-year-old donor (donor I) previously used, NHAC's from multiple donors with ages of 24 (donor II), 41 (donor III), and 50 (donor IV), are obtained after informed consent (Cambrex, Verviers, Belgium). Cells are seeded as described in Examples 1 and 3. The propagated Ad-siRNAs are used to transduce NHAc cells from these different donors at three MOI's in duplicate in the chondrogenesis assay (see Examples 1 and 3). The Ad-siRNAs have to score at least once above threshold (average+2.5 times standard deviation) to pass this donor dependency test.

Out of 101 hits that passed the Alcian blue assay for aggrecan, 97 score positive in Donor IV. Of the remaining 4, 1 score positive in Donor II. The other 3 did not score in the other donors tested. In addition, 40 of the 101 hits score positive in all three additional donors. These results demonstrate that 98 out of these 101 hits function in a non-donor-dependent manner. Exceptions are H33-145; H33-182; and H33-263 (see Table 1). Indicated in this Table 1 are the Target Gene Symbol, Gene Bank Accession Number, and drugability class of the genes that correspond to the target sequences. The results for some of the genes are shown in Table 2. These data show that knocking down the RNA levels of the indicated genes induces collagen II levels in at least 2 donors. The values represent times standard deviation of the background.

TABLE 2 overview donor dependency data. N = 2 for every condition. All data points are represented as fold standard deviation of the background. All values above 2.5 are considered to be positive and are shaded grey.

Example 6 Quality Control of Target Ad-siRNAs

Target Ad-siRNAs are propagated using derivatives of PER.C6© cells (Crucell, Leiden, The Netherlands) at a 96-well plate level, followed by re-screening these viruses at several MOI's in the primary assay (see Example 1) and by sequencing the siRNAs encoded by the target Ad-siRNA viruses. PER.E2A cells are seeded in 96 well plates at a density of 40,000 cells per 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: 12 μl crude lysate is added 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) in sterile PCR tubes. These are incubated at 55° C. for 2 h followed by a 15 min 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, SEQ ID NO: 29), 1 μl of ‘Reverse Primer’ (10 mM stock, sequence: 5′ CCC CCA CCT TAT ATA TAT TCT TTC C, SEQ ID NO: 30), 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 min; each cycle runs at 95° C. for 15 sec, 55° C. for 30 sec, 68° C. for 4 min, and is repeated for 35 cycles. A final incubation at 68° C. is performed for 7 min. 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 ˜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 7 Evaluation of the on Target Effect of the Identified siRNA Sequences

To evaluate whether the identified siRNA sequences really increase collagen II and aggrecan levels through the knock down of the target mRNA, a second siRNA sequence is identified that exerts the same effect.

A number of additional siRNA sequences targeting the HIT sequences are designed and incorporated in adenoviruses according to WO 03/020931. After production of these adenoviruses, the Ad-siRNAs are infected at different volumes (1.5 μl, 5 μl and 15 μl) in the chondrocytes and their effect on collagen II is assessed as described in example 1. The threshold (average+2.5 standard deviation) is calculated for every volume. If a virus scores above the threshold for one or more of the different infection volumes, it is considered to be positive. The on target results are shown in FIG. 6. These results indicate that at least one additional siRNA, which targets the HIT sequence, can be identified. This underscores the on target effect of the siRNA sequences identified during the screening of the Silence Select library.

Example 8 Development of a Three-Dimensional-Alginate Culture System for a Quantitative Marker Analysis for Stable Cartilage and the Assessment of Glycosaminoglycans (GAG) and Hydroxyprolines (Hyp) Synthesis Principle of the Assay:

Normal human articular chondrocytes (NHAC's) grown in three-dimensional cultures are able to maintain a “differentiated state” as measured by the expression of Collagen type II and aggrecan. De-differentiated chondrocytes cultured in a two-dimensional system for a limited amount of passages can revert to a differentiated state when transferred into a three-dimensional culture system. This system was established to test the capability of siRNA adenoviruses (Ad-siRNA) 1) to induce a mRNA expression pattern that correlates with anabolic active chondrocytes and 2) to induce protein modification of collagen type II (hydroxyprolines) and aggrecan (glycosaminoglycans) involved in the stability of cartilage. Normal cartilage NHAC's are cultured in a two-dimensional culture system for two or three passages for cell expansion purposes. Cells are transduced with individual siRNA adenoviruses (Ad-siRNA) in the two-dimensional culture system and three days later are transferred into the three-dimensional alginate culture system. After 10 days in the alginate culture, various parameters can be assessed (e.g. mRNA marker analysis and protein modifications).

Assay Procedure

Using Ad-BMP2 (BMP2: “strong” collagen II inducer) and Ad-BMP7 (BMP7: “weak” collagen II inducer) as positive controls and Ad-ALPL as negative control the following protocol is set up (for both mRNA marker analysis and protein modification assessment): After two or three passages in monolayer culture conditions NHAC's are seeded at 2.10E+06 cells/T175 flask in 30 ml of chondrocyte growing medium (Cambrex) and transduced the following day with control-viruses (Ad-BMP2, Ad-BMP4, Ad-BMP7, Ad-ALPL) using an MOI of 2000. After three days, cells are trypsinised using the chondrocyte reagent pack (Cambrex) and washed once with 155 mM sodium chloride/20 mM Hepes ph 7.4 (Cambrex). Cells are re-suspended at a density of 2×106 cells/ml in 1.2% sodium alginate (Cambrex). The cell suspension is transferred into a syringe attached to a 21-22 gauge needle and expelled in a drop-wise fashion into 102 mM calcium chloride/5 mM Hepes pH 7.4 (1 ml and 5 ml respectively in 24-well and 6-well plate) Five and 50 beads per well are produced respectively in the 24-well and 6-well plates. Plates containing the alginate beads are incubated for 10 minutes with gentle shaking every 2 minutes. The calcium chloride solution is aspirated with a Vacusafe system and beads are washed three times with the sodium chloride solution using the Vacusafe and once with DMEMF/12 supplemented with 10% heat inactivated fetal bovine serum (FBS-HI) and 1% penicillin/streptomycin. Alginate beads are finally re-suspended into 0.5 ml and 3 ml (respectively in 24-well and 6-well plate) of DMEMF/12 supplemented with 10% heat inactivated fetal calf serum (FBS-HI), 1% penicillin/streptomycin and 25 μg/ml of ascorbic acid (Fluka, Sigm101-128 and 401-594ldrich). The alginate cultures are incubated in a humidified incubator at 37° C. and 5% CO2 during 10 days with a medium refreshment every 48/72 h.

For each adenoviral transduction, 60 alginate beads are generated: 2×5 beads were cultured in 24-well plates for the GAGs/Hyps assessments and 50 beads are cultured in a single well of a 6-well plate for the mRNA expression pattern determination.

Control Viruses

Ad-BMP2; described in WO 03/018799

Ad-BMP7; Ad5 dE1/E2A adenoviruses that mediate the expression of full length bone morphogenetic protein 7 pre-protein (NP001710).

Ad-BMP4; Ad5 dE1/E2A adenoviruses that mediate the expression of full length bone morphogenetic protein 4 pre-protein (see NP570912).

Ad-ALPL; Ad5 dE1/E2A adenoviruses that mediate the expression of full length liver/bone/kidney alkaline phosphatase (NP000469).

Example 9 Effect of Knock Down of Target Genes in Chondrocytes Embedded in Alginate Beads, on the Glycosaminoglycans (GAGs) Levels

Chondrocytes are infected and embedded in alginate beads according to example 8. After 10 days in culture the alginate beads are treated with papain in order to solubilise the glycosaminoglycans prior to quantification: Beads cultured in the 24-well plates are washed once with a 50 mM Phosphate buffer pH 6.5 and incubated for 3 to 4 h at 65° C. with 250 μl/well of the same buffer containing 2 mM EDTA, 2 mM L-cystein and 126 μg/ml papain (Sigma). Complete digestion of the beads is assessed by microscopic observation. Papain digests are frozen at −20° C. until glycosaminoglycans quantification is performed. The GAGs produced by the primary chondrocytes in the alginate culture system are measured using the Blyscan™ assay (Biocolor Ltd, Newtownabbey, Northern Ireland).

Principle of the Blyscan Assay

The Blyscan Assay is a quantitative dye-binding method for the analysis of sulfated GAGs. The dye label used in the assay is 1,9-dimethylmethylene blue employed under conditions that produce a specific label for the sulfated polysaccharides component of proteoglycans and/or the protein-free sulfated glycosaminoglycan chains. Aggrecan is the predomninant proteoglycan in articular cartilage, representing ±90% of the cartilage proteoglycans. It is composed of a central core protein attached to ±50 keratan sulfate and ±100 chondroitin sulfate chains known as GAGs required for the biological function and the stability of aggrecan.

Assay Description

The papain digests generated after culturing the chondrocytes for 10 days in alginate beads (Example 7) are diluted in water 1:100, 1:200 if cells were originally transduced with the positives controls and 1:50, 1:100 if transduced with the negative controls. This dilution step allows readout values within the standard range of the assay. The GAG standard provided by the manufacturer contains 100 μg/ml of chondroitin 4-sulfate purified from bovine trachea. This standard is run in duplicate at four concentrations corresponding to 1, 2, 3 and 5 μg of GAGs. Standard and controls are individually diluted in 100 μl final volume in eppendorf tubes. One milliliter of the Blyscan Dye Reagent is added to each tube and incubated for 30 minutes at room temperature with continuous shaking. When formed, the GAG-dye complex becomes insoluble and is then separated from the remaining excess soluble unbound dye by centrifugation (10000×g for 10 minutes). Supernatant are discarded by inverting and careful draining of the tube contents. One milliliter of Blyscan Dissociation Reagent is added to each tube and incubated for one hour to one and a half hour with continuous shaking. This reagent brings the GAG-bound dye back into solution. The GAG content of the assayed samples is spectrophotometrically determined by the amount of dye recovered from the GAGs in the test sample. Two hundred microliters of the dissociation dye solutions are added to the wells of a 96-well plate and reading is performed on an automatic plate reader set at a dual wavelength (656 and 450 nm).

The GAGs concentrations measured in the Blyscan assay are normalized to DNA content by performing a fluorimetric Hoechst assay on the same papain digests. Hoechst 33342 dye reagent (Molecular Probes), a bisbenzimidasole dye that binds to adenine/thymine rich regions on DNA. Papain digests were diluted 1:1.7 and 1:3.3 in TE buffer, this dilution step allows readout values within the standard range of the assay. Purified calf thymus DNA (Sigm101-128 and 401-594ldrich) is used as standard DNA: the initial stock solution (2 μg/ml) is sequentially diluted in TE buffer in order to obtain the following concentration range: 1.5, 1.0, 0.75, 0.50, 0.2 μg/ml. Standard and test samples are diluted in TE to a final volume of 100 μl and added to a 96-well plate. One hundred microliters of Hoechst 33342 dye reagent are added to the wells, reading is performed on a multifunctional microplate reader (Fluostar Galaxy, BMG Labtechnologies GmbH) with an excitation wavelength set at 360 nm and an emission wavelength set at 440 nm.

When GAG concentrations are normalized to DNA content the resulting values expressed as the ratio GAG concentration (μg/ml)/DNA concentration (μg/ml) are used to calculate the assay window. This window is calculated as the ratio normalized GAG Ad-BMP2 (or Ad-BMP4)/normalized GAG Ad-ALPL.

The effect of the knock down of the 14 target genes on the GAG levels is assessed as described. The results are shown in FIG. 7. The results are expressed as fold induction of the GAG levels compared to the average of the normalized GAG values obtained for two negative control knock-down adenoviruses (Ad-PTGER4 and Ad-GRM7). Knock down of the mRNA of the respective genes results in an increase of the GAG levels.

Control Viruses

Ad-BMP2: described in WO 03/018799

Ad-BMP4: Ad5 dE1/E2A adenoviruses that mediate the expression of full length bone morphogenetic protein 4 pre-protein (see NP570912).

Ad-ALPL: Ad5 dE1/E2A adenoviruses that mediate the expression of full length liver/bone/kidney alkaline phosphatase (NP000469).

    • Ad-PTGER4: Ad5 dE1/E2A adenoviruses that comprise the siRNA sequence CCATGCCTATTTCTACAGC (SEQ ID NO: 31) to knock down the prostaglandin E receptor 4 mRNA.
    • Ad-GRM7: Ad5 dE1/E2A adenoviruses that comprise the siRNA sequence TCAGTAACAGCTCCCAGAC (SEQ ID NO: 32) to knock down the metabotropic glutamate receptor 7 mRNA.

Example 10 Quantitative Analysis of Hydroxyprolines (Hyps)

In articular cartilage, approximately 95% of the collagen is type II collagen. Its polymers are the fibrils that form the basic cohesive framework of the tissue. The collagen biosynthesis involves several unique posttranslational modifications including hydroxylation of proline and lysine residues. These modifications are crucial for collagen stability and resistance to proteolytic enzymes.

Chondrocytes are infected and embedded in alginate beads according to example 8. After 10 days in culture the alginate beads are treated with papain: Beads cultured in the 24-well plates are washed once with a 50 mM Phosphate buffer pH 6.5 and incubated for 3 to 4 h at 65° C. with 250 μl/well of the same buffer containing 2 mM EDTA, 2 mM L-cystein and 126 μg/ml papain (Sigma). Complete digestion of the beads is assessed by microscopic observation. Papain digests are frozen at −20° C. until hydroxyproline quantification is performed.

Hydroxyproline assessment in the papain digests is performed by HPLC after acid hydrolysis and FMOC (9-fluorenylmethyl chloroformate) derivatisation of the samples. This method is described in “Bank R A, Jansen E J, Beekman B and Te Koppele J M. (1996) Amino acid analysis by reverse-phase high performance liquid chromatography: Improved derivatisation and detection conditions with 9-fluorenylmethyl chloroformate. Anal Biochem. 240 (2): 167-176.

The effect of the knock down of the 14 target genes on the hydroxyproline levels is assessed as described. The results are shown in FIG. 8. The results are expressed as fold induction compared to the average of Hyp concentrations measured for the two KD controls (Ad-PTGER4 and AD-GRM7). Knock down of the mRNA of the respective genes results in an increase of the hydroxyproline levels.

Control Viruses

Ad-BMP2; described in WO 03/018799

Ad-BMP4; Ad5 dE1/E2A adenoviruses that mediate the expression of full length bone morphogenetic protein 4 pre-protein (see NP570912).

Ad-ALPL: Ad5 dE1/E2A adenoviruses that mediate the expression of full length liver/bone/kidney alkaline phosphatase (NP000469).

Ad-PTGER4: Ad5 dE1/E2A adenoviruses that comprise the siRNA sequence CCATGCCTATTTCTACAGC (SEQ ID NO: 33) to knock down the prostaglandin E receptor 4 mRNA.

Ad-GRM7: Ad5 dE1/E2A adenoviruses that comprise the siRNA sequence TCAGTAACAGCTCCCAGAC (SEQ ID NO: 34) to knock down the metabotropic glutamate receptor 7 mRNA.

Example 11 Quantitative Analysis of Markers for Stable Cartilage Assay General Principle:

Chondrocyte phenotypes can be categorized by characteristic patterns of gene expression. Quantitative RT-PCR techniques are used to monitor the expression pattern of a set of key marker molecules to define which phenotype is induced on the chondrocytes. Positive markers included collagen type II and FGFR3, typically expressed by cartilage chondrocytes. Negative makers included (1) collagens types I and III for dedifferentiated or fibroblast-like chondrocytes, a phenotype that can also be induced by retinoic acid or interleukin-1; (2) collagen X, PTHLH and ALK-1 for hypertrophic chondrocytes that are found in the calcified zone of adult cartilage and the lower hypertrophic zone of the fetal growth-plate cartilage; and (3) MMP13 as proteolytic enzyme involved in cartilage degradation. Functional cartilage chondrocytes should express high levels of positive markers but low or not detectable levels of negative markers.

Assay Description

Chondrocytes are infected and embedded in alginate beads according to example 8. After 10 days in culture the alginate beads are treated with 55 mM sodium citrate (Cambrex) in order to recover the chondrocytes from the alginate beads and harvest RNA: Incubation medium was removed from the 6-well plates with the Vacusafe and 5 ml of 55 mM sodium citrate are added to each well and incubated for 15 minutes at room temperature. The partially solubilised beads are gently mixed and transferred to a FALCON tube, wells are rinsed once with 2 ml of sodium citrate solution to collect the remaining beads and released cells. Tubes are laid on their side and gently mixed every 2-3 minutes until beads have completely solubilised (+/−15 minutes). Tubes are centrifuged at 1000 rpm for 10 minutes. Supernatant is discarded and the cell pellet is re-suspended in 2 ml of sodium citrate solution and left for 5 minutes. Six milliliters of 155 mM sodium chloride/20 mM Hepes ph 7.4 (Cambrex) are added to each tube prior to a 5 minutes centrifugation at 210 g. Cell pellet is lysed in 180 μl SV40 lysing buffer (Promega SV40 total RNA extraction kit) and frozen at −20° C. until RNA isolation is performed with the Promega SV40 total RNA extraction kit according to the manufacturer's instructions. Purified RNA is quantified using ribogreen reagent (Molecular Probes) and yeast RNA (Ambion) as standard RNA.

Purified RNA from chondrocytes transduced with control adenoviruses and cultured for 10 days in the alginate 3-dimensional culture system is used in a reverse transcription (RT) reaction. RNA is first diluted in water (Life Technologies-Invitrogen, Breda, The Netherlands) depending on the initial concentration of the sample: For RNA concentration below 25 ng/ml, samples are used undiluted, for RNA concentration between 25 and 50 ng/ml samples are diluted 1:2.5, For RNA concentration between 50 and 100 ng/ml samples are diluted 1:5 and for RNA concentration between 100 and 160 ng/ml samples are diluted 1:10. Two microliters of diluted/undiluted RNA are added to 5 μl of a reaction mix consisting of: 1× Taqman RT buffer, 5 mM MgCl2, 500 μM dNTPs (2.5 mM each), 2.5 μM Random hexamers, 0.4 U/μl of RNAse inhibitor and 1.25 U/μl of MultiScribe Reverse Transcriptase (all reagents purchased from Applied Biosystems). The PCR reaction is performed in a Peltier Thermal Cycler-200 (BIOzym, Landgraaf, The Netherlands) as followed: the PCR total mixture (60 μl) is incubated 10 minutes at 25° C. followed by 30 minutes at 48° C., followed by 5 minutes at 95° C. Each reaction is run in parallel with a control that does not contain any RNAse inhibitor or Reverse Transcriptase.

Reverse transcription is followed by a quantitative PCR for specific amplification of the selected positive and negative marker genes, GAPDH is included as the endogenous control. Five microliters of cDNA (from RT reaction) are added to 20 μl of a PCR reaction mix consisting of: 1× Brilliant® SYBR® Green QPCR Master Mix (Sratagene Europe, Amsterdam, The Netherlands), 300 μM of each forward and reverse primers (table 3) (except for GAPDH and ALK-1, used at 100 μM)(Life Technologies-Invitrogen, Breda, The Netherlands) and 300 nM of reference dye (Stratagene) (diluted 1:100 in H2O). The PCR mixture (25 μl) is incubated for 10 minutes at 95° C. followed by 40 cycles: 15 seconds at 95° C. followed by 1 minute at 60° C. in the ABI PRISM® 7000 Sequence Detection System (Applied Biosystems, Nieuwerkerk A/D Ijssel, The Netherlands).

TABLE 3 primer sequences for the positive and  negative markers: SEQ ID Primer Gene Primer sequence NO Forward Collagen  GGCAATAGCAGGTTCACGTACA 35 2a1 L10347 Reverse Collagen  CGATAACAGTCTTGCCCCACTT 36 2a1 L10347 Forward FGFR3  ACGGCACACCCTACGTTACC 37 NM_000142 Reverse FGFR3  TGTGCAAGGAGAGAACCTCTAGCT 38 NM_000142 Forward BMP-2  CCAACACTGTGCGCAGCTT 39 NM_001200 Reverse BMP-2  AAGAATCTCCGGGTTGTTTTCC 40 NM_001200 Forward ALK-1  CAGTCTCATCCTGAAAGCATCTGA 41 NM_000020 Reverse ALK-1  TTTCCCACACACTCCACCAA 42 NM_000020 Forward collagen  TGGAGTGTTTTACGCTGAACGAT 43 10a1 NM_000493 Reverse collagen  CCTCTTACTGCTATACCTTTACTCTT 44 10a1 TATGG NM_000493 Forward Collagen  TGCCATCAAAGTCTTCTGCAA 45 1A1 NM_000088 Reverse Collagen  CGCCATACTCGAACTGGAATC 46 1A1 NM_000088 Forward Collagen  CACTATTATTTTGGCACAACAGGAA 47 3a1 NM_000090 Reverse Collagen  AGACACATATTTGGCATGGTTCTG 48 3a1 NM_000090 Forward MMP13  CAAGGGATCCAGTCTCTCTATGGT 49 NM_002427 Reverse MMP13  GGATAAGGAAGGGTCACATTTGTC 50 NM_002427 Forward PTHLH  GCTCGGTGGAGGGTCTCA 51 NM_002820 Reverse PTHLH  CTGTGTGGATTTCTGCGATCA 52 NM_002820 Forward GAPDH  CATCCATGACAACTTTGGTATCG 53 NM_002046 Reverse GAPDH  AGTCTTCTGGGTGGCAGTGAT 54 NM_002046

Results are expressed for each tested marker gene as the relative expression in the sample (transduction with tested adenovirus) versus control (knock-down control adenoviruses) (Relative expression=2ddCt, where ddCt=dCtKD sample−dCtKD control and dCt=Ctsample−CtGAPDH). The results for the 14 genes are shown in FIG. 9. The knock down of the mRNA of the respective genes results in expression of the positive markers, while the levels of the negative markers are either low or not detectable.

Control Viruses

Ad-BMP2; described in WO 03/018799

Ad-ALPL: Ad5 dE1/E2A adenoviruses that mediate the expression of full length liver/bone/kidney alkaline phosphatase (NP000469).

Example 12 Expression of cDNA's in Human Cartilage

Upon identification of a modulator of cartilage synthesis, it is of the highest importance to evaluate whether the modulator is expressed in the tissue and the cells of interest. This can be achieved by measuring the RNA and/or protein levels. In recent years, RNA levels are being quantified through real time PCR technologies, whereby the RNA is first transcribed to cDNA and then the amplification of the cDNA of interest is monitored during a quantitative PCR reaction. The amplification plot and the resulting Ct value are indicators for the amount of a specific RNA transcript present in the sample. Ct values are determined in the presence or absence of the reverse transcriptase step (+RT versus −RT). An amplification signal in the −RT condition indicates the occurrence of non-specific PCR products originating from the genomic DNA. If the +RT Ct value is 3 Ct values higher than the −RT Ct value, then the investigated RNA is present in the sample.

To assess whether the polypeptides of the genes identified in the above assays are expressed in human cartilage, real time PCR with specific primers for the polynucleotides (“Assay on Demand” Applied Biosystems) is performed on human cartilage total RNA (Clinomics Biosciences). 2 samples of non-osteoarthritis and 2 of osteoarthritis patients are analyzed.

In short, 40 ng of RNA is transcribed to DNA using the MultiScribe Reverse Transcriptase (50 U/μl) enzyme (Applied BioSystems). The resulting cDNA is amplified with AmpliTaq Gold DNA polymerase (Applied BioSystems) during 40 cycles using an ABI PRISM® 7000 Sequence Detection System. Amplification of the transcript is detected via SybrGreen which results in a fluorescent signal upon intercalation in double stranded DNA.

Total RNA isolated from human cartilage is analyzed for the presence of transcripts listed in Table 4 via quantitative real time PCR.

For the genes listed in Table 4 the obtained Ct values indicate that they are detected in all RNA samples. ELA1 RNA is not detected in the real time PCR analysis, underscoring the need for additional patient analysis.

TABLE 4 Ct values Normal Cartilage OA Cartilage TARGET Ct Sample 1 Ct Sample 2 Ct Sample 1 Ct Sample 2 PDE1A 30.8 28.5 30.7 30.2 GPR103 33.1 33.3 38.4 37.4 JAK1 26.2 24.1 27 26 ICK 36.3 33.8 36.1 35.5 DGKB 28.5 26.7 29.7 28.7 DUSP3 27.1 24.2 27 27.1 DUSP11 28.8 27.5 29.9 29.2 SLC26A8 36.13 34.04 37.12 37.22 SLC15A2 32.5 28.1 34.7 32.9 ABCG1 29.7 28 31.1 29.5 FZD1 28.1 25.4 34.1 27.1 ELA1 40 40 40 39.1

Example 13 Identification of Small Molecules that Inhibit TARGET Kinase Activity

Compounds are screened for inhibition of the activity of the TARGETS that are kinase polypeptides. The affinity of the compounds to the polypeptides is determined in an experiment detecting changed reaction conditions after phosphorylation. The TARGET kinase polypeptides are incubated with its substrate and ATP in an appropriate buffer. The combination of these components results in the in vitro phosphorylation of the substrate. Sources of compounds include commercially available screening library, peptides in a phage display library or an antibody fragment library, and compounds that have been demonstrated to have binding affinity for a TARGET kinase.

The TARGET kinase polypeptides can be prepared in a number of ways depending on whether the assay will be run using cells, cell fractions or biochemically, on purified proteins. The polypeptides can be applied as complete polypeptides or as polypeptide fragments, which still comprise TARGET kinase catalytic activity.

Identification of small molecules inhibiting the activity of the TARGET kinase polypeptides is performed by measuring changes in levels of phosphorylated substrate or ATP. Since ATP is consumed during the phosphorylation of the substrate, its levels correlate with the kinase activity. Measuring ATP levels via chemiluminescent reactions therefore represents a method to measure kinase activity in vitro (Perkin Elmer). In a second type of assay, changes in the levels of phosphorylated substrate are detected with phosphospecific agents and are correlated to kinase activity. These levels are detected in solution or after immobilization of the substrate on a microtiter plate or other carrier. In solution, the phosphorylated substrate is detected via fluorescence resonance energy transfer (FRET) between the Eu labeled substrate and an APC labeled phosphospecific antibody (Perkin Elmer), via fluorescence polarization (FP) after binding of a phosphospecific antibody to the fluorescently labeled phosphorylated substrate (Panvera), via an Amplified Luminescent Proximity Homogeneous Assay (ALPHA) using the phosphorylated substrate and phosphospecific antibody, both coupled to ALPHA beads (Perkin Elmer) or using the IMAP binding reagent that specifically detects phosphate groups and thus alleviates the use of the phosphospecific antibody (Molecular Devices). Alternatively, the substrate is immobilized directly or by using biotin-streptavidin on a microtiter plate. After immobilization, the level of phosphorylated substrate is detected using a classical ELISA where binding of the phosphospecific antibody is either monitored via an enzyme such as horseradish peroxidase (HRP) or alkaline phospahtase (AP) which are either directly coupled to the phosphospecific antibody or are coupled to a secondary antibody. Enzymatic activity correlates to phosphorylated substrate levels. Alternatively, binding of the Eu-labeled phosphospecific antibody to the immobilized phosphorylated substrate is determined via time resolved fluorescence energy (TRF) (Perkin Elmer). In addition, the substrate can be coated on FLASH plates (Perkin Elmer) and phosphorylation of the substrate is detected using 33P labeled ATP or 125I labeled phosphospecific antibody.

Small molecules are randomly screened or are preselected based upon drug class, (i.e. known kinase inhibitors), or upon virtual ligand screening (VLS) results. VLS uses virtual docking technology to test large numbers of small molecules in silico for their binding to the polypeptide of the invention. Small molecules are added to the kinase reaction and their effect on levels of phosphorylated substrate is measured with one or more of the above-described technologies.

Small molecules that inhibit the kinase activity are identified and are subsequently tested at different concentrations. IC50 values are calculated from these dose response curves. Strong binders have an IC50 in the nanomolar and even picomolar range. Compounds that have an IC50 of at least 10 micromol or better (nmol to pmol) are applied in collagen II assay to check for their effect on the induction of chondrocyte anabolic stimulation.

Example 14 Ligand Screens for TARGET GPCRs Example 14 A Reporter Gene Screen

Mammalian cells such as Hek293 or CHO-K1 cells are either stably transfected with a plasmid harboring the luciferase gene under the control of a cAMP dependent promoter (CRE elements) or transduced with an adenovirus harboring a luciferase gene under the control of a cAMP dependent promoter. In addition reporter constructs can be used with the luciferase gene under the control of a Ca2+ dependent promoter (NF-AT elements) or a promoter that is controlled by activated NF-κB. These cells, expressing the reporter construct, are then transduced with an adenovirus harboring the cDNA of a TARGET GPCR. Forty (40) hours after transduction the cells are treated with the following:

a) an agonist for the receptor and screened against a large collection of reference compounds comprising peptides (LOPAP, Sigma Aldrich), lipids (Biomol, TimTech), carbohydrates (Specs), natural compounds (Specs, TimTech), small chemical compounds (Tocris), commercially available screening libraries, and compounds that have been demonstrated to have binding affinity for a polypeptide comprising an amino acid sequence selected from the group consisting of the SEQ ID NOs of the TARGET GPCRs; or

b) a large collection of reference compounds comprising peptides (LOPAP, Sigma Aldrich), lipids (Biomol, TimTech), carbohydrates (Specs), natural compounds (Specs, TimTech), small chemical compounds (Tocris), commercially available screening libraries, and compounds that have been demonstrated to have binding affinity for a polypeptide comprising an amino acid sequence selected from the group consisting of SEQ ID NOs of the TARGET GPCRs.

Compounds, which decrease the agonist induced increase in luciferase activity or the constitutive activity, are considered to be antagonists or inverse agonists for a TARGET GPCR. These compounds are screened again for verification and screened against their effect on anabolic stimulation of chondrocytes. The compounds are also screened to verify binding to the GPCR. The binding and reporter activity assays can be performed in essentially any order to screen compounds.

In addition, cells expressing the NF-AT reporter gene can be transduced with an adenovirus harboring the cDNA encoding the α-subunit of G15 or chimerical Gα subunits. G15 is a promiscuous G protein of the Gq class that couples to many different GPCRs and as such re-directs their signaling towards the release of intracellular Ca2+ stores. The chimerical G alpha subunits are members of the Gs and Gi/o family by which the last 5 C-terminal residues are replaced by those of Gαq, these chimerical G-proteins also redirect cAMP signaling to Ca2+ signaling.

Example 14 B FLIPR Screen

Mammalian cells such as Hek293 or CHO-K1 cells are stably transfected with an expression plasmid construct harboring the cDNA of a TARGET GPCR. Cells are seeded, grown, and selected until sufficient stable cells can be obtained. Cells are loaded with a Ca2+ dependent fluorophore such as Fura3 or Fura4. After washing away the excess of fluorophore the cells are screened against a large collection of reference compounds comprising peptides (LOPAP, Sigma Aldrich), lipids (Biomol, TimTech), carbohydrates (Specs), natural compounds (Specs, TimTech), small chemical compounds (Tocris), commercially available screening libraries, and compounds that have been demonstrated to have binding affinity for a polypeptide comprising an amino acid sequence selected from the group consisting of SEQ ID NOs of the TARGET GPCRs, by simultaneously adding an agonist (alternatively no agonist need be added if the constitutive activity of the receptor is used) and a compound to the cells. Activation of the receptor is measured as an almost instantaneously increase in fluorescence due to the interaction of the fluorophore and the Ca2+ that is released. Compounds that reduce or inhibit the agonist induced increase in fluorescence (or constitutive fluorescence) are considered to be antagonists or inverse agonists for the receptor they are screened against. These compounds are screened again to measure the amount of anabolic stimulation of chondrocytes as well as binding to a TARGET GPCR.

Example 14 C AequoScreen

CHO cells, stably expressing Apoaequorin are stably transfected with a plasmid construct harboring the cDNA of a TARGET GPCR. Cells are seeded, grown, and selected until sufficient stable cells can be obtained. The cells are loaded with coelenterazine, a cofactor for apoaequorin. Upon receptor activation intracellular Ca2+ stores are emptied and the aequorin will react with the coelenterazine in a light emitting process. The emitted light is a measure for receptor activation. The CHO, stable expressing both the apoaequorin and the receptor are screened against a large collection of reference compounds comprising peptides (LOPAP, Sigma Aldrich), lipids (Biomol, TimTech), carbohydrates (Specs), natural compounds (Specs, TimTech), small chemical compounds (Tocris), commercially available screening libraries, and compounds that have been demonstrated to have binding affinity for a polypeptide comprising an amino acid sequence selected from the group consisting of SEQ ID NOs of the TARGET GPCRs, by simultaneously adding an agonist (alternatively no agonist need be added if the constitutive activity of the receptor is used) and a compound to the cells. Activation of the receptor is measured as an almost instantaneously light flash due to the interaction of the apoaequorin, coelenterazine, and the Ca2+ that is released. Compounds that reduce or inhibit the agonist induced increase in light or the constitutive activity are considered to be antagonists or inverse agonists for the receptor they are screened against. These compounds are screened again to measure the amount of anabolic stimulation of chondrocytes as well as binding to a TARGET GPCR.

In addition, CHO cells stable expressing the apoaequorin gene are stably transfected with a plasmid construct harboring the cDNA encoding the α-subunit of G15 or chimerical Gα subunits. G15 is a promiscuous G protein of the Gq class that couples to many different GPCRs and as such redirects their signaling towards the release of intracellular Ca2+ stores. The chimerical G alpha subunits are members of the Gs and Gi/o family by which the last 5 C-terminal residues are replaced by those of Gαq, these chimerical G-proteins also redirect cAMP signaling to Ca2+ signaling.

Example 14 D Screening for Compounds that Bind to the GPCR Polypeptides (Displacement Experiment)

Compounds are screened for binding to the TARGET GPCR polypeptides. The affinity of the compounds to the polypeptides is determined in a displacement experiment. In brief, the GPCR polypeptides are incubated with a labeled (radiolabeled, fluorescent labeled) ligand that is known to bind to the polypeptide and with an unlabeled compound. The displacement of the labeled ligand from the polypeptide is determined by measuring the amount of labeled ligand that is still associated with the polypeptide. The amount associated with the polypeptide is plotted against the concentration of the compound to calculate IC50 values. This value reflects the binding affinity of the compound to its TARGET, i.e. the TARGET GPCR polypeptides. Strong binders have an IC50 in the nanomolar and even picomolar range. Compounds that have an IC50 of at least 10 micromol or better (nmol to pmol) are applied an anabolic stimulation of chondrocytes assay to check for their effect on osteogenesis. The TARGET GPCR polypeptides can be prepared in a number of ways depending on whether the assay are run on cells, cell fractions or biochemically, on purified proteins.

Example 14 E Screening for Compounds that Bind to a TARGET GPCR (Generic GPCR Screening Assay)

When a G protein receptor becomes constitutively active, it binds to a G protein (Gq, Gs, Gi, Go) and stimulates the binding of GTP to the G protein. The G protein then acts as a GTPase and slowly hydrolyses the GTP to GDP, whereby the receptor, under normal conditions, becomes deactivated. However, constitutively activated receptors continue to exchange GDP to GTP. A non-hydrolyzable analog of GTP, [35S]GTPγS, can be used to monitor enhanced binding to membranes which express constitutively activated receptors. It is reported that [35S]GTPγS can be used to monitor G protein coupling to membranes in the absence and presence of ligand. Moreover, a preferred approach is the use of a GPCR-G protein fusion protein. The strategy to generate a TARGET GPCR-G protein fusion protein is well known for those known in the art. Membranes expressing TARGET GPCR-G protein fusion protein are prepared for use in the direct identification of candidate compounds such as inverse agonist. Homogenized membranes with TARGET GPCR-G protein fusion protein are transferred in a 96-well plate. A pin-tool is used to transfer a candidate compound in each well plus [35S]GTPγS, followed by incubation on a shaker for 60 minutes at room temperature. The assay is stopped by spinning of the plates at 4000 RPM for 15 minutes at 22° C. The plates are then aspirated and radioactivity is then read.

Example 14 F Receptor Ligand Binding Study on Cell Surface

The receptor is expressed in mammalian cells (Hek293, CHO, COS7) by adenoviral transducing the cells (see U.S. Pat. No. 6,340,595). The cells are incubated with both labeled ligand (iodinated, tritiated, or fluorescent) and the unlabeled compound at various concentrations, ranging from 10 pM to 10 μM (3 hours at 4° C.: 25 mM HEPES, 140 mM NaCl, 1 mM CaCl2, 5 mM MgCl2 and 0.2% BSA, adjusted to pH 7.4). Reactions mixtures are aspirated onto PEI-treated GF/B glass filters using a cell harvester (Packard). The filters are washed twice with ice cold wash buffer (25 mM HEPES, 500 mM NaCl, 1 mM CaCl2, 5 mM MgCl2, adjusted to pH 7.4). Scintillant (MicroScint-10; 35 μl) is added to dried filters and the filters counted in a (Packard Topcount) scintillation counter. Data are analyzed and plotted using Prism software (GraphPad Software, San Diego, Calif.). Competition curves are analyzed and IC50 values calculated. If one or more data points do not fall within the sigmoidal range of the competition curve or close to the sigmoidal range the assay is repeated and concentrations of labeled ligand and unlabeled compound adapted to have more data points close to or in the sigmoidal range of the curve.

Example 14 G Receptor Ligand Binding Studies on Membrane Preparations

Membranes preparations are isolated from mammalian cells (Hek293, CHO, COS7) cells over expressing the receptor is done as follows: Medium is aspirated from the transduced cells and cells are harvested in 1×PBS by gentle scraping. Cells are pelleted (2500 rpm 5 min) and resuspended in 50 mM Tris pH 7.4 (10×106 cells/ml). The cell pellet is homogenized by sonicating 3×5 sec (UP50H; sonotrode MS1; max amplitude: 140 um; max Sonic Power Thickness: 125 W/cm2). Membrane fractions are prepared by centrifuging 20 min at maximal speed (13,000 rpm˜15,000 to 20,000 g or rcf). The resulting pellet is resuspended in 500 μl 50 mM Tris pH 7.4 and sonicated again for 3×5 sec. The membrane fraction is isolated by centrifugation and finally resuspended in PBS. Binding competition and derivation of IC50 values are determined as described above.

Example 14 H Internalization Screen (1)

Activation of a GPCR-associated signal transduction pathway commonly leads to translocation of specific signal transduction molecules from the cytoplasm to the plasma membrane or from the cytoplasm to the nucleus. Norak has developed their transfluor assay based on agonist-induced translocation of receptor-β-arrestin-GFP complex from the cytosol to the plasma membrane and subsequent internalization of this complex, which occurs during receptor desensitization. A similar assay uses GFP tagged receptor instead of β-arrestin. Hek293 cells are transduced with a TARGET GPCR vector that translates for a TARGET GPCR-eGFP fusion protein. 48 hours after transduction, the cells are set to fresh serum-free medium for 60 minutes and treated with a ligand for 15, 30, 60 or 120 minutes at 37° C. and 5% CO2. After indicated exposure times, cells are washed with PBS and fixed with 5% paraformaldehyde for 20 minutes at RT. GFP fluorescence is visualized with a Zeiss microscope with a digital camera. This method aims for the identification of compounds that inhibit a ligand-mediated (constitutive activity-mediated) translocation of the fusion protein to intracellular compartments.

Example 14 I Internalization Screen (2)

Various variations on translocation assays exists using β-arrestin and β-galactosidase enzyme complementation and BRET based assays with receptor as energy donor and β-arrestin as energy acceptor. Also the use of specific receptor antibodies labeled with pH sensitive dyes are used to detect agonist induced receptor translocation to acidic lysosomes. All of the translocation assays are used for screening for both agonistic and antagonistic acting ligands.

Example 14 J Melanophore Assay (Arena Pharmaceutical)

The melanophore assay is based on the ability of GPCRs to alter the distribution of melanin containing melanosomes in Xenopus melanophores. The distribution of the melanosomes depends on the exogenous receptor that is either Gi/o or Gs/q coupled. The distribution of the melanosomes (dispersed or aggregated) is easily detected by measuring light absorption. This type of assay is used for both agonist as well as antagonist compound screens.

Example 15 Identification of Small Molecules that Inhibit Protease Activity

Compounds are screened for inhibition of the activity of the polypeptides of the present invention. The affinity of the compounds to the polypeptides is determined in an experiment detecting changes in levels of cleaved substrate. In brief, the polypeptides of the present invention are incubated with its substrate in an appropriate buffer. The combination of these components results in the cleavage of the substrate.

The polypeptides can be applied as complete polypeptides or as polypeptide fragments, which still comprise the catalytic activity of the polypeptide of the invention.

Cleavage of the substrate can be followed in several ways. In a first method, the substrate protein is heavily labeled with a fluorescent dye, like fluorescein, resulting in a complete quenching of the fluorescent signal. Cleavage of the substrate however, releases individual fragments, which contain less fluorescent labels. This results in the loss of quenching and the generation of a fluorescent signal, which correlates to the levels of cleaved substrate. Cleavage of the protein, which results in smaller peptide fragments, can also be measured using fluorescent polarization (FP). Alternatively, cleavage of the substrate can also be detected using fluorescence resonance energy transfer (FRET): a peptide substrate is labeled on both sides with either a quencher and fluorescent molecule, like DABCYL and EDANS. Upon cleavage of the substrate both molecules are separated resulting in fluorescent signal correlating to the levels of cleaved substrate. In addition, cleavage of a peptide substrate can also generate a new substrate for another enzymatic reaction, which is then detected via a fluorescent, chemiluminescent or colorimetric method.

Small molecules are randomly screened or are preselected based upon drug class, i.e. protease, or upon virtual ligand screening (VLS) results. VLS uses virtual docking technology to test large numbers of small molecules in silico for their binding to the polypeptide of the invention. Small molecules are added to the proteolytic reaction and their effect on levels of cleaved substrate is measured with the described technologies.

Small molecules that inhibit the protease activity are identified and are subsequently tested at different concentrations. IC50 values are calculated from these dose response curves. Strong binders have an IC50 in the nanomolar and even picomolar range. Compounds that have an IC50 of at least 10 micromol or better (nmol to pmol) are applied in amyloid beta secretion assay to check for their effect on the beta amyloid secretion and processing.

Example 16 Identification of Small Molecules that Inhibit Phosphodiesterase Activity

Compounds are screened for inhibition of the activity of the polypeptides of the present invention. The affinity of the compounds to the polypeptides is determined in an experiment detecting changes in levels of substrate or product. In brief, the polypeptides of the present invention are incubated with its substrate in an appropriate buffer. The combination of these components results in the conversion of the substrate into its product.

The polypeptides can be applied as complete polypeptides or as polypeptide fragments, which still comprise the catalytic activity of the polypeptide of the invention.

Conversion of cAMP or cGMP in AMP or GMP can be followed 1) by determining the cAMP or cGMP levels using e.g. ELISA. Alpha screen technology, Time resolved fluorescent technology, IMAP 2) by determining the levels of the products AMP and GMP using a colorimetric assay. The basis for the latter assay is the cleavage of cAMP or

cGMP by a cyclic nucleotide phosphodiesterase. The 5′-nucleotide released is further cleaved into the nucleoside and phosphate by the enzyme 5′-nucleotidase. The phosphate released due to enzymatic cleavage is quantified using BIOMOL GREEN™ reagent in a modified Malachite Green assay.

Small molecules are randomly screened or are preselected based upon drug class, i.e. PDE, or upon virtual ligand screening (VLS) results. VLS uses virtual docking technology to test large numbers of small molecules in silico for their binding to the polypeptide of the invention. Small molecules are added to the PDE reaction and their effect on cyclic nucleotide levels is measured with the described technologies.

Small molecules that inhibit the PDE activity are identified and are subsequently tested at different concentrations. IC50 values are calculated from these dose response curves. Strong binders have an IC50 in the nanomolar and even picomolar range. Compounds that have an IC50 of at least 10 micromol or better (nmol to pmol) are applied in assays evaluating the anabolic activity of chondrocytes. This can be achieved by determining col2α1 and aggrecan levels produced by the chondrocytes.

Claims

1. Method for identifying a compound that induces chondrocyte anabolic stimulation, comprising:

contacting a compound with a polypeptide comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 55-82 and 198-391, and 480; and
measuring a compound-polypeptide property related to the anabolic stimulation of chondrocytes.

2. The method according to claim 1, wherein said polypeptide is in an in vitro cell-free preparation.

3. The method according to claim 2, wherein said polypeptide is present in a mammalian cell.

4. The method of claim 1, wherein said property is a binding affinity of said compound to said polypeptide.

5. The method of claim 3, wherein said property is activation of a biological pathway producing a biochemical marker indicative of the anabolic stimulation of chondrocytes.

6. The method of claim 5 wherein said indicator is selected from the group consisting of collagen type II, alpha-1 (col2a1) and aggrecan.

7. The method of claim 6 wherein said polypeptide comprises an amino acid sequence selected from the group consisting of SEQ ID NO: 55-82.

8. The method according to claim 1, wherein said compound is selected from the group consisting of compounds of a commercially available screening library and compounds having binding affinity for a polypeptide comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 55-82 and 198-391, and 480.

9. The method according to claim 2, wherein said compound is a peptide in a phage display library or an antibody fragment library.

10. An agent for inducing the anabolic stimulation of chondrocytes, selected from the group consisting of an 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 encoding a polypeptide comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 55-82 and 198-391, and 480.

11. The agent according to claim 10, wherein polypeptide comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 55-82.

12. The agent according to claim 11, wherein a vector in a mammalian cell expresses said agent.

13. The agent according to claim 12, wherein said vector is an adenoviral, retroviral, adeno-associated viral, lentiviral, a herpes simplex viral or a sendaiviral vector.

14. The agent according to claim 10, wherein said antisense polynucleotide and said siRNA comprise an antisense strand of 17-25 nucleotides complementary to a sense strand, wherein said sense strand is selected from 17-25 continuous nucleotides of a nucleic acid sequence encoding a polypeptide comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 55-82.

15. The agent according to claim 14, wherein said siRNA further comprises said sense strand.

16. The agent according to claim 15, wherein said sense strand is selected from 17-25 continuous nucleotides of a nucleic acid sequence selected from the group consisting of SEQ ID NO: 1-28 and 479.

17. The agent according to claim 16, wherein said siRNA further comprises a loop region connecting said sense and said antisense strand.

18. The agent according to claim 17, wherein said loop region comprises a nucleic acid sequence defined of SEQ ID NO: 83.

19. The agent according to claim 18, wherein said agent is an antisense polynucleotide, ribozyme, or siRNA comprising a nucleic acid sequence complementary to a polynucleotide selected from the group consisting of SEQ ID NO: 84-197 and 410.

20. A chondrocyte anabolic stimulation enhancing pharmaceutical composition comprising a therapeutically effective amount of an agent of claim 10 in admixture with a pharmaceutically acceptable carrier.

21-32. (canceled)

Patent History
Publication number: 20140107181
Type: Application
Filed: Oct 17, 2013
Publication Date: Apr 17, 2014
Applicant: GALAPAGOS N.V. (MECHELEN)
Inventors: Nick Vandeghinste (Duffel), Peter Herwig Maria Tomme (Gent), Frits Michiels (Leiderdorp), Libin Ma (Oegstgeest), Blandine Mille-Baker (Oegstgeest), Helmuth H.G. van Es (Haarlem)
Application Number: 14/055,955