Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
METHOD FOR DIAGNOSING RHEUMATOID ARTHRITIS VIA ASSAYING MYOFIBROBLAST-LIKE SYNOVIOCYTES FOR FIBROBLAST ACTIVATION PROTEIN
Document Type and Number:
WIPO Patent Application WO/2007/111657
Kind Code:
A3
Abstract:
The invention relates to methods for diagnosing rheumatoid arthritis by assaying for Fibroblast Activation Protein Alpha expression in rheumatoid, myofibrblast like synoviocytes. Therapeutic aspects are also a part of the invention.

Inventors:
RENNER CHRISTOPHER (DE)
BAUER STEFAN (CH)
Application Number:
PCT/US2006/046608
Publication Date:
September 12, 2008
Filing Date:
December 05, 2006
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
LUDWIG INST CANCER RES (US)
RENNER CHRISTOPHER (DE)
BAUER STEFAN (CH)
International Classes:
G01N33/53; G01N33/00; G01N33/543; G01N33/566
Foreign References:
US6455677B12002-09-24
US20050272703A12005-12-08
US5767242A1998-06-16
US5587299A1996-12-24
US20020061839A12002-05-23
Other References:
SCANLAN ET AL.: "Molecular cloning of fibroblast activation protein a, a member of the serine protease family selectively expressed in stromal fibroblasts of epithelial cancers", PROC. NATL. ACAD. SCI. USA, vol. 91, June 1994 (1994-06-01), pages 5657 - 5661
GARIN-CHESA ET AL.: "Cell surface glycoprotein of reactive stromal fibroblasts as a potential antibody target in human epithelial cancers", PROC. NATL. ACAD. SCI. USA, vol. 87, September 1990 (1990-09-01), pages 7235 - 7239, XP002113522
KELLY: "Fibroblast activation protein-a and dipeptidyl peptidase IV (CD26): Cell-surface proteases that activate cell signaling and are potential targets for cancer therapy", DRUG RESISTANCE UPDATES, vol. 8, 2005, pages 51 - 58, XP004919314
Attorney, Agent or Firm:
HANSON, Norman, D. (666 Fifth AvenueNew York, NY, US)
Download PDF:
Claims:
CLAIMS:

1. A method of determining presence of rheumatoid arthritis ("RA") in a patient, comprising assaying a sample of synovial tissue taken from said patient for expression of FAP-α, wherein expression of said FAP-α is indicative of RA in said patient.

2. The method of claim I , further comprising amplifying a nucleic acid molecule which encodes FAP-α as an indication of RA.

3. The method of claim 1 , wherein said amplifying comprises polymerase chain reaction.

4. The method of claim 1, further comprising assaying said sample for a protein expressed by a nucleic acid molecule which encodes FAP-α.

5. The method of claim 4, wherein said assay is an immunoassay.

6. The method of claim 5, wherein said assay is an immunohistochemical assay.

7. The method of claim 4, wherein said immunoassay comprises contacting said sample with a monoclonal antibody which binds specifically for FAP- α.

8. The method of claim 7, wherein said monoclonal antibody is a mouse anti-human antibody.

9. A method for screening for RA in a sample, comprising assaying said sample for mRNA for FAP-α by contacting said sample with at least one oligonucleotide, of which consists of the nucleotide sequence set forth in SEQ ID NO: 1 or SEQ ID NO: 2, and determining hybridization of said oligonucleotide to said mRNA for FAP-α, said hybridization being indicative of RA in said sample.

10. A method for treating a patient with RA 3 comprising administering to said patient an amount of a FAP-α inhibitor sufficient to inhibit activity of FAP-α.

11. The method of claim 10, wherein said activity is enzymatic activity.

12. The method of claim 10, wherein said inhibitor is a monoclonal antibody which specifically binds to and inhibits FAP- α.

13. The method of claim 11 , wherein said inhibitor is a FAP-α antagonist.

14. The method of claim 10, wherein said inhibitor is a peptide.

15. The method of claim 14, wherein said peptide consists of 2-10 amino acids.

16. The method of claim 14, wherein said peptide consists of 2-5 amino acids.

17. The method of claim 14, wherein said peptide consists of 2 amino acids.

18. The method of claim 10, wherein said inhibitor is a modified peptide (Val-boro- Pro).

19. The method of claim 17, wherein said peptide is PT-100.

20. A method for monitoring status of RA in a patient comprising:

(i) assaying a sample of cells taken from a subject with RA to determine expression of mRJMA for FAP-α, wherein a change in said mRNA expression is associated with RA, to determine a value, wherein said value is the level of mRNA expression in said sample and

(ii) comparing the value determined in (i) to a value determined previously for FAP-α obtained the same way the value in (i) was determined; wherein a change in the value obtained in (i) as compared to the value determined previously shows a change in status of said RA.

Description:

METHOD FOR DIAGNOSING RHEUMATOID ARTHRITIS VIA

ASSAYING MYOFIBROBLAST-LIKE SYNOVIOCYTES FOR

FIBROBLAST ACTIVATION PROTEIN

RELATED APPLICATION

[0001] This application claims priority of US provisional application. Serial Number 60/750,173, filed on December 14, 2005, the disclosure of which is incorporated by reference in its entirety.

FIELD OF THE INVENTION

[0002] This invention relates to certain molecules associated with rheumatoid arthritis cells. More particularly, it relates to fibroblast activation protein alpha ("FAP-α" hereafter) and its expression by rheumatoid myofibroblast-like synoviocytes. The molecule is a cell surface-bound type II transmembrane glycoprotein and has a molecular weight of from about 88 to about 95 kilodaltons as determined by SDS-PAGE. FAP-α has previously been identified as being associated with cancer cells and reactive with tumor stroma cells, but has not yet been identified as a marker for rheumatoid arthritis cells, i.e., synovial fibroblasts with a myofϊbroblastic phenotype. This, inter alia, is the subject of the invention. FAP-α is mainly expressed on the surface of mesenchymal cells that arc involved in epithclium-mescnchyme interactions contributing to tissue remodeling. Nucleic acid molecules encoding FAP-α, portions thereof, FAP-α itself, and antibodies specific for FAP-α, such as monoclonal antibodies, or antibody fragments, are all useful in connection with features of the invention. AU journal articles, cited to infra, are incorporated by reference in their entireties.

BACKGROUND AND PRIOR ART

[0003] FAP-α is a Mr 95 kDa cell surface-bound type II transmembrane glycoprotein and is a member of the serine prolyl oligopeptidase family. Comparison of amino acid sequences indicate that FAP-α is essentially identical to seprase (Goldstein LA, et al., Biochim Biophys Acta 1997, 1361(1): 11-19) and is closely related to DPP IV (dipeptidylpeptidase IV), also known as CD26, another type II integral membrane protein. (Morimoto C, et al., Immunol Rev 1998, 161:55-70). These exoproteases cleave NH2- lerminal dipeptides from polypeptides with L-proline or L-alanine immediately following

the N-temiinal amino acid. FAP-α has been found to have collagenase activity in vitro. (Goldstein LA, et al., supra; Jones B 3 et al., Blood 2003, 102(5):1641-1648). In addition to various families of proteolytic enzymes, such as matrix or disintegrin metal loproteases that serve as major collagenases, peptidase activity of FAP-α contributes to extracellular matrix ("ECM") degradation. (Park JE, et al., J Biol Chein 1999, 274(51):36505-36512; Ghersi G 3 ct al., J Biol Chem 2002, 277(32):29231-29241). This is not only a fundamental property of normal tissue repair and remodeling, but is also involved in the pathological processes of invasive growth. This property correlates with the expression of FAP-α in granulation tissue of healing wounds (Grinnell F, J Cell Biol 1994, 124(4):401 -404), desmoplastic reactions (Yen TW, et al., Surgery 2002, 131(2): 129- 134), and in more than 90% of human epithelial carcinomas. (Garin-Chesa P, et al., Proc Natl Acad Sci USA 1990, 87(18):7235-7239). FAP-α is mainly expressed on the surface of mesenchymal cells that are involved in epithelium-mesenchyme interactions contributing to tissue remodeling. Consistent with its mesenchymal origin, FAP-α is also occasionally expressed by bone and soft tissue sarcomas. (Rettig WJ, et al., Proc Natl Acad Sci US A 1988, 85(9):31103114). Immunohistochemical staining of colorectal carcinomas and breast cancer (Park JE, et al., supra; Scanlan MJ, et al., Proc Natl Acad Sci USA 1994, 91 (12):5657-5661) confirmed the specific expression of FAP-α by tumor stroma fibroblasts but not by malignant cells themselves. (See also, Sappino AP, et al., Int J Cancer 1988, 41(5):707-712). Observations made dining a clinical Phase I study, which examined the biodistribution of a humanized anti-FAP-α antibody in patients with advanced or metastatic FAP-α-positive cancer, a minor low-grade uptake in the knees and shoulders of three patients without clinical symptoms of arthritis, could have led to the conclusion that FAP-α might also be present in human joints. (Scott AM, et al., Clin Cancer Res 2003, 9(5): 1639-1647). Unfortunately, no further explanation or discussion of this observation was presented. Further, it is unknown whether these patients were suffering from osteoarthritis. In contrast, resting fibrocytes in normal adult tissue generally lack detectable FAP-α expression. (Rettig WJ, et al., supra; Garin-Chesa P, et al., Proc Natl Acad Sci USA 1990, 87(18):7235-7239).

[0004] Further information on FAP-α can be found in, e.g., U.S. Patent Nos. 5,587,299; 5,767,242; 5,965,373; and 6,846,910, incorporated by reference in their entireties.

[0005] Rheumatoid arthritis ("RA") is a chronic inflammatory disease of unknown etiology and characterized by hyperplasia and chronic inflammation of the synovial membranes that invade deeply into the articular cartilage and bone. Activated fibroblast-like synoviocytes ("FLS") in the lining layer of the synovium are one of the dominant cells involved in pannus formation and are key players in joint destruction. (Firestein GS, Arthritis Rheum 1996, 39(1 1): 1781 -1790; Pap T, et al., Arthritis Rheum 2000, 43(l l):2531-2536). Rheumatoid FLS have been shown to proliferate in an anchorage independent manner and express increased proliferation markers and matrix degrading enzymes when compared with FLS from patients with osteoarthritis ("OA"). (Qu Z, et al., Arthritis Rheum 1994, 37(2):212-220; Bucala R, et al., J Exp Med 1991 , 173(3):569-574; Lafyatis R 3 et al., J Clin Invest 1989, 83(4): 12671276). Expression of the CD44v7/8 epitope is linked to the proliferative behavior of FLS obtained from patients with RA, whereas expression of variants containing CD44v3 is linked with, their increased invasive capacity (Croft DR, et al., Eur J Immunol 1997, 27(7): 1680-1684; Wibulswas A, et al., Am J Pathol 2000, 157(6):2037-2044; Wibulswas A, et al., Arthritis Rheum 2002, 46(8):2059-2064). Matrix metalloproteases ("MMP") have been shown to be essential for degradation of the articular matrix, with MMP-I and MMP-13 being considered as important candidates for joint destruction in RA (Tomita T, et al., Arthritis Rheum 2002, 46(2):373-378; Westhoff CS, et al., Arthritis Rheum 1999, 42(7):1517- 1527). Invasion of migratory fibroblasts into connective tissue, however requires cell surface serine proteases, as well as metallocollagenases. (Ghersi G, et al., supra). Among these exoproteases that may cooperate with interstitial collagenase are groups of serine prolyl-peptidases such as DPP 1V/CD26 and FAP-α/seprase. (Park JE, et al., supra; De Meester T, et al., Immunol Today 1999, 20(8):367-375). Fibroblasts with smooth muscle differentiation, termed myofibroblasts, are generally accepted to be the main source for extracellular matrix degrading factors. Furthermore, FLS with a myofibroblast-like molecular phenotype have been identified in the intimal lining layer of inflamed rheumatoid synovium. (Kasperkovitz PV, et al., Arthritis Rheum 2005, 52(2):430-441).

[0006] Despite the elucidation of FLS as a key player in joint inflammation and proteolytic enzymes like MMPs as representative markers for ECM degradation, no anti-fibroblast directed therapy is currently available. Approaches to inhibit the joint destructive process in RA by elimination or inhibition of one proteolytic enzyme did not

produce sufficient results in clinical trials regardless of supportive in-vivo results. (Schedel J 3 et al., Gene Ther 2004, 11(13):1040-1047; Lewis EJ, et al., Br J Pharmacol 1997, 121 (3):540-546; Brown PD, Expert Opin Investig Drugs 2000, 9(9):2167-2177).

[0007] The difference between the targeting strategies of the approaches described supra, and a FAP-α-specific targeting approach, results from the substantial potential of FAP -α as a specific marker for synovial fibroblasts in RA, as described in the Detailed Description which follows. Thus, an embodiment of the present invention includes a diagnostic methodology for determining presence of RA in a patient, involving assaying a synovial tissue sample taken from the patient for the expression of FAP-α, wherein the expression of FAP-α is indicative of presence of EA in the patient.

[0008] Another embodiment of the present invention includes a therapeutic methodology involving inhibition of FAP-α playing its role in tissue remodeling, as well as focusing antibody mediated cytotoxic activity on this synovial cell type to eradicate FAP -α's role in the joint destructive process. It has now been found that these deleterious cells can be identified via analysis of at least one marker on their cellular surfaces. Identification of this marker has therapeutic ramifications which are set forth in the disclosure which follows.

[0009] These, as well as other features of the invention, will be disclosed in greater detail in the Detailed Description which follows.

DETAILED DESCRIPTION OF THE PREFERRED EMBODIMENTS EXAMPLE l

[0010] This example describes experiments in which synovial tissue samples were collected from the diseased joints of patients. To elaborate, messenger RNA ("mPvNA") was isolated from the collected synovial tissue samples, and reverse transcriptase polymerase chain reaction ("RT-PCR") was performed to amplify mRNA encoding specific proteins.

[0011] Synovial tissue was collected from a total of 20 patients during routine orthopedic surgery, i.e., ten patients with end-stage osteoarthritis ("OA"), and ten patients with refractory destructive rheumatoid arthritis ("RA") who underwent joint replacement surgery. The mRNA extraction and cDNA synthesis was performed immediately after surgery, and results obtained from these patients were compared to results obtained from control samples.

[0012] Specifically, synovial tissue was collected, using well known techniques which need not be elaborated here. A portion of the collected synovial tissue from each patient, as discussed supra, was homogenized immediately after surgery using a tissue tearer, and total RNA was extracted using commercially available products, and well established methods. The control samples included HTl 080 cells transfected with FAP-α, mock-transfected cells (HTl 080 par) (as described in Bauer S, et al., J Immunol 2004, 172(6):3930-3939), or cancer cells, taken from tumor samples from patients with breast cancer. The breast cancer cells were used as a positive control, because immunohistochemical staining of breast cancer confirmed the specific expression of FAP- α by tumor stroma fibroblasts but not by malignant cells themselves. (Park et al., J Biol Chem 1999, 274(51):36505-36512; Scanlan, et al., Proc Natl Acad Sci USA 1994, 91(12):5657-5661; and U.S. Patent No. 5,587,299).

[0013] All samples were analyzed for niRNA transcripts coding for fragments of FAP-α (741bp), MMP-I (fibroblast type collagenase; collagenase-1; 428 bp) and MMP-13 (coIlagenase-3; 390 bp). The total amount of RNA present was quantified by spectrophotometry before first strand cDNA synthesis was performed, using commercially available products, and well established methods.

[0014] The following primers were used to generate specific cDNA-fragments (as described in Konttinen, et al., Ann Rheum Dis 1999, 58(11):691-697):

(a) the FAP-α PCR product was generated with

5 '-GTTATTGCCTATTCCTATTATG- 3' (SEQ ID NO: 1) and 5 '-GTCCATCATGAAGGGTGGAAA- 3' (SEQ ID NO: 2);

(b) the MMP-I PCR product was generated with 5 '-CTGAAGGTGATGAAGCAGCC- 3' (SEQ ID NO: 3) and 5 '-AGTCCAAGAGAATGGCCGAG- 3' (SEQ ID NO: 4);

(c) the MMP-13 PCR product was generated with 5 '-CTATGGTCCAGGAGATGAAGS' (SEQ ID NO: 5) and 5 '-AGAGTCTTGCCTGTATCCTC- 3' (SEQ ID NO: 6);

(d) amplification of a genomic p53 fra9ment with the expected size of 753 bp served as control, and the p53 PCR product was generated with

5'-CGTGAGCGCTTCGAGATGTTCCG-S' (SEQ DD NO: 7) and 5'-CCTAACCAGCTGCCCAACTGTAG-S' (SEQEDNO: 8).

[0015] 1 μl of any cDNA fragments prochiced were amplified with 1.25 unit of Taq DNA polymerase with 2.5 ml of 1Ox standard Taq buffer in a final volume of 25 μl containing 0.5 μl of nucleosidtriphosphate (10 mM), 0.5 of μl of each primer (10 μM), and distilled water.

[0016] PCR amplification was carried out using a thermocycler and the following cycling protocol: 95°C for 5 rain, annealing for 60 seconds, and extension at 72°C for 1 min. The number of cycles and the annealing temperature depended on the primers used: (a) FAP-α (55°C, 30 cycles); (b) MMP-I, (c) MMP- 13 (62°C, 40 cycles); and (d) p53 (65°C, 25 cycles). Aliquots (20 μl) of each PCR product were separated on a 1.5% agarose gel and visualized by ethidium bromide staining.

[0017] The results showed that positive PCR controls were obtained from cDNA derived from FAP-α-transfected HTl 080 cells or pooled breast cancer tumor samples (MMP-I and MMP-13). Amplification of cDNA derived from mock transfected HT 1080 cells was negative for the specific FAP-α fragments discussed above. Analysis of synovial tissue samples from patients with OA or RA demonstrate ubiquitous expression of all of collagenases-1 and -3 and FAP-α, but were found to be positive in RA samples to a much higher extent.

EXAMPLE II

[0018] This next example describes an experiment that was conducted to show differences in the translation of enzymes from mRNA, in synovial tissue samples from patients with OA as compared to patients with RA.

[0019] Imniunohistochemical characterization of fibroblast populations (100- fold optical magnification) was performed. Collected synovial tissue samples, as discussed supra, were snap-frozen and sequential sections were stained for expression of a panel of activation markers including enzymes previously determined to be present by RT-PCR (e.g., Thy-1, FAP-α or SMA), in experiments not discussed herein.

[0020] To elaborate, the tissue samples referred to supra, were embedded in medium, snap-frozen in liquid nitrogen and then stored at — 80 0 C for immunohistochemical analysis. Sequential 5-μM sections were placed on microscope

slides, and the slides were fixed in cold acetone (4°C for lOmin), air dried, relαydrated in PBS and then blocked using a biotin blocking system.

[0021] Before adding a primary antibody (discussed infra), slides were blocked with rabbit serum using commercially available products, and well established methods. Following blocking, one of several different primary antibodies were added. These were murine F19 (mouse anti-human FAP-α mAb), anti-human CD90 (Thy-1; clone AS02), biotin-SP-conjugated rabbit anti-mouse IgG (irrelevant isotype matched IgG), 1 :200, mouse anti-human CD44v3 (clone VFF-327), 1:50, mouse anti-human CD44v7/8 (clone VFF- 17), 1:200, mouse anti-human smooth muscle actin (SMA; clone 1A4), 1 :50, goat anti-human MMP-I (C-18), goat anti human MMP-13 (D-17), and biotinylated rabbit anti-goat IgG (irrelevant isotype matched IgG), 1 :100. The murine Fl 9, anti-human CD90 and biotin-SP-conjugated rabbit anti-mouse IgG were incubated for 60 minutes at room temperature, while the others were incubated at 4 0 C overnight. A biotinylated secondary antibody (rabbit anti-mouse IgG, 1:500 or rabbit anti-goat IgG, 2g/ml) and a preformed avidin-biotinylated horse radish peroxidase P-complex (ABC reagent) were used in accordance with standard protocols. The resulting antibody-ABC complex was visualized with a 3-amino 9 ethylcarbazole based chromogen (using commercially available products, and well established methods), resulting in pink-brown coloration of antigen-positive cells. All slides were counterstained for about 5 mill with Meyer's hematoxylin. Final slide adjustment was perfoπned using commercially available products, and well established methods.

[0022] The results showed that the tissue samples from the OA and RA patients, as discussed supra, show distinct synovial fibroblast populations. Associated expression of SMA and FAP-α in the myofibroblasts of the intimal synovial lining is distinguished from Thy-1 positive fibroblasts, and from perivascular smooth muscle cells that only express SMA.

[0023] With respect to OA synovial fibroblasts, the results showed that these cells expressed CD90 (Thy-1) on their surface. The results also showed, however, a distinct OA synovial fibroblast population which completely lacked Thy-1 surface expression, but was clearly characterized by cell surface expression of FAP-α.

[0024] With respect to RA synovial fibroblasts, the results showed that the cell surface expression of FAP-α and SMA, i.e., the staining of anti-human FAP-α and anti-human SMA, was more intensive in this tissue as compared to the OA synovial

fibroblasts, as discussed supra. Also, as shown in the OA synovial fibroblasts supra, the surface expression of FAP-α in the lining layer of the synovium is distinguished from non-proliferating and Thy-1 expressing fibroblasts of the structural scaffold.

EXAMPLE III

[0025] This example describes the visual scoring of the density of FAP-α - specific PCR-bands and of the intensity of histological FAP-α -detection, in the OA and RA synovial tissue samples, after RT-PCR analysis and immunohistochemistry was performed as described supra.

[0026] The visual scoring analysis was done by an independent observer, who was not made aware of the sample source being asserted. Comparison between synovial tissue samples taken from patients with OA and RA was performed using commercially available products, and well established methods. The scoring parameters were as follows: 1+: weak; 2+: modest; 3+: strong; 4+: very strong. P values <0.05 were considered statistically significant.

[0027] The results showed that the difference in density of PCR-bands, and intensity of histological FAP-α detection, between synovial tissue samples taken from patients with OA as compared to patients with RA, was significant (p < 0.0002). The results are summarized in Table 1 , which follows:

[0028] Tn follow up, visual observation work, FAP-α positive FLS were analyzed to determine what activation molecules are associated with these cells. Using the same scoring system referred to, supra, SMA, CD44v33, CD44v7/8, MMP-I, MMP- 13 expression levels were analyzed, in samples taken from 5 RA patients. The results are summarized in Table 2, which follows:

EXAMPLE IV

[0029] The preceding examples demonstrate FAP-α expression in the synovial tissue samples studied. Further histomorphological analysis was carried out, using the methods and materials of example 2, supra, to further define the cell types in the samples analyzed. MMP-I 5 MMP-13, CD44v3, and CD44v7/8 were studied, as was SMA (400- fold optical magnification). The first four of these are known to be instrumental in ECM alteration in malignancies, and to be expressed in the synovial membranes of diseased joints, while SMA is a reliable means to identify the subpopulation of cells that are fibroblasts with myofibrobastic phenotype. This cell type is proteolytic.

[0030] Staining of perivasuclar smooth muscle cells showed that SMA was expressed by FAP-α positive, FLS cells, in the intimal lining area. This so-called "expression signature" is characteristic of FAP-α expressing myofϊbroblastic cells, which are the center of high inflammation activity in the rheumatoid synovium. This expression of FAP-α and SMA in synovial tissue samples of patients with RA was accompanied by accumulation of activation markers MMP-I and MMP-13, and splice variants CD44v3 and CD44v7/8.

[0031] On the other hand, immunohistochemical analysis of the activation markers, referred to supra, in synovial tissue samples of patients with OA, showed limited staining for the MMPs and the CD44 variants. Specifically, the results showed only minor expression of MMP-I, MMP-13, and CD44v7/8 in areas that were slightly FAP-α positive.

[0032] There was a clear difference between OA and RA samples with respect to the expression pattern and staining intensity of the activation markers, referred to supra. Specifically, the RA samples showed a homogenous expression pattern and a homogenous staining intensity, which was not the case for the OA samples. The results

also showed that the expression pattern and staining intensity of the RA tissue samples represented a stronger intensity of synovial inflammation when compared to the OA tissue samples.

[0033] Since FAP-α expression has been shown to be much more pronounced in RA tissue, as compared to OA tissue, this expression could be related to the degree of synovial inflammation (as discussed supra), as has already been demonstrated for MMPs and FAP-α in collagen induced arthritis ("ClA"). In mice, analysis of CIA gene expression profiles revealed a seven-fold increase in either FAP-α or MMP gene expression in inflamed when compared to non-inflamed paws. (Mclndoe RA, et al., Proc Natl Acad Sci U S A 1999, 96(5):2210-2214).

[0034] The fact that FLS in the rheumatoid synovium express FAP-α intensively, permits the artisan to screen for therapeutic agents, such as, but not being limited to, enzymatic substrates. The probability of enzymatic activity is supported by immunohistochemistry, as described supra. As shown by the results described herein, characterization of the synovial lining layer in RA patients revealed that FAP-α expression is accompanied by accumulation of other degradation markers that are predominantly found in this area, e.g., MMP-I, MMP-13 as well as CD44v3 and CD44v7/S. These markers are instrumental in extracellular matrix remodeling in malignancies and are also already known to be present in the synovial membranes of diseased joints (Croft DR 3 et al., supra; Wibulswas A, et al., Am J Pathol 2000, 157(6):2037-2044; Wibulswas A, et al., Arthritis Rheum 2002, 46(8):2059-2064); Tomita T, et al., supra; Westhoff CS, et al., Arthritis Rheum 1999, 42(7):1517-1527). In addition, fibroblasts of the myofibroblastic phenotype were shown to be the major cell type expressing FAP-α in RA patients, as discussed supra. These fibroblasts are also the major source of other proteolytic enzymes and are generally thought to be responsible for ECM degradation. (Kasperkovitz PV, et al., supra; Sundarrajan M, et al.; Arthritis Rheum 2003, 48(9):2450-2460; Yamamoto N, et al., J Clin Invest 2003, 112(2):181-188).

[0035] As discussed supra, a reliable marker to identify fibroblasts of the myofibroblastic phenotype is SMA, and immunohistochemistry revealed the expression of SMA by FAP-α -positive FLS in the intimal lining layer of the rheumatoid synovium. This expression signature characterizes the area of FAP-α-expressing myofibroblastic cells as the centre of high-inflammation activity in the rheumatoid synovium and

discriminates from Thy-1 positive non-proliferating fibroblasts in the synovial matrix. (Seemayer CA, et al., Am J Pathol 2003, 162(5):1549-1557).

[0036] The value of FAP-α as a therapeutic target in RA and OA is twofold: First, since the proteolytic activity of FAP-α contributes to ECM degradation, inhibition thereof is an attractive goal. In this context, it needs to be noted that suppression of FAP- α's enzymatic activity by PT-IOO (which is a modified dipeptide, i.e., the boronic dipeptide Val-boro-Pro) resulted in potent antitumor effects and augmented antibody- dependent cell-mediated cytotoxicity in murine tumor models. (Adams S, et al., Cancer Res 2004, 64(15):5471-5480; Cheng JD, et al., MoI Cancer Ther 2005, 4(3):351-360) (The structure of PT- 100 is provided in Figure 1 of Adams). Second, FAP-α has now been shown to be a marker for the identification of the subset of FLS cells with the highest proteolytic activity. The genetically stable and restricted expression of FAP-α (Garin-Chesa P, et al., suprci) has lead to the establishment of several preclinical strategies for tumor therapy. (Bauer S, et al., supra; Cheng JD, et al., Cancer Res 2002, 62(16):4767-4772; Samel D, et al., J Biol Chem 2003, 278(34):32077-32082). The use of aπti-FAP-α antibody-based cancer immunotargeting has even been proven in a clinical Phase f dose-escalation study (Scott AM, et al., supra).

[0037] Therefore, since FAP-α has been shown to be a valuable therapeutic marker in cancer, e.g., antitumor effects, and FAP-α appears to be a valuable marker for the identification of FLS, it is seen that different pathological conditions, such as rheumatoid arthritis, which are characterized by abnormal expression of FAP-α, are amenable to treatment with anti-FAP-α agents, such as antagonists, including peptides, modified peptides, and so foith.

[0038] Other aspects of the invention will be clear to the skilled artisan, and need not be set forth here.

[0039] The terms and expressions which have been employed are used as terms of description and not of limitation, and there is no intention in the use of such terms and expressions of excluding any equivalents of the features shown and described or portions thereof, it being recognized that various modifications are possible within the scope of " the invention.