FUNCTION AND REGULATION OF ADAMTS-1

The present invention relates to ADAMTS-1 and uses thereof. The present invention also relates to fragments of ADAMTS-1 and methods of inhibiting cell growth and metastasis. The present invention also provide methods of identifying inhibitors and activators relating to the function of ADAMTS-1.

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

This application is a continuation of U.S. patent application Ser. No. 12/709,296, now abandoned, which is a divisional of U.S. patent application Ser. No. 11/104,075, filed Apr. 12, 2005 and issued as U.S. Pat. No. 7,696,307, all of which claim priority to U.S. Provisional Application Ser. No. 60/561,429, filed Apr. 12, 2004 and U.S. Provisional Application Ser. No. 60/650,027 filed Feb. 4, 2005 each of which is herein incorporated by reference in its entirety.

GOVERNMENT SUPPORT

This invention was made with U.S. Government support (NIH Grants No. RO1HL074117) and the U.S. Government may therefore have certain rights in the invention.

BACKGROUND OF THE INVENTION

The members of ADAMTS (A Disintegrin And Metalloproteinase with ThromboSpondin motifs) family belong to ADAM (A Disintegrin And Metalloproteinase) family of multifunctional proteins that display a significant sequence homology with snake venom metalloproteinases. The amino-terminal half of ADAMTS is similar to that of ADAM, which contains propeptide, metalloproteinase, disintegrin, and cysteine-rich domains; while the C-terminal half of ADAMTS is completely different and contains thrombospondin type I-like (TSP) motifs that are originally found in thrombospondin 1 and 2 and spacer region. At least 18 members of ADAMTS have been identified. ADAMTS-1 is the first member identified and is expressed in many embryonic tissues and in tumors. Disruption of ADAMTS-1 gene results in reduced growth, abnormalities in uteral, adrenal, and adipose tissues, and female infertility.

ADAMTS-1 cleaves aggrecan and versican in vitro, however, physiologic substrates of ADAMTS-1 remain to be identified. In addition, ADAMTS-1 is cleaved at the spacer region by matrix metalloproteinases (MMPs). The role of ADAMTS-1 in tumor growth and metastasis is not well established. ADAMTS-1 was found to display anti-angiogenic and anti-tumor activity, however, increased expression of ADAMTS-1 was correlated to the enhanced metastatic potential of pancreatic cancers, and studies have shown that ADAMTS-1 is one of the genes up-regulated in the breast cancer with elevated metastatic activity.

Thus, there is a need to clarify the biologic role of ADAMTS-1. Furthermore, there is a need to identify compounds and/or compositions that can be used to treat cancer or inhibit cell growth.

SUMMARY OF THE INVENTION

In some embodiments, the present invention provides isolated polypeptide fragments of ADAMTS-1 that inhibits tumor growth and metastasis.

In some embodiments, the present invention provides compositions comprising at least two different polypeptide fragment of ADAMTS-1 that inhibit cell growth and/or metastasis.

In some embodiments, the present invention provides isolated polynucleotides encoding a polypeptide fragment of ADAMTS-1 wherein the fragment inhibits metastasis In some embodiments, the present invention provides methods for identifying an inhibitor or an activator of ADAMTS-1 cleavage.

In some embodiments, the present invention provides methods for identifying a heparin inhibitor.

In some embodiments, the present invention provides methods of identifying an inhibitor of the metalloproteinase activity of ADAMTS-1.

In some embodiments, the present invention provides methods of inhibiting metastasis comprising contacting the cell with a polypeptide fragment of ADAMTS-1 that inhibits metastasis and/or a nucleic acid that encodes a polypeptide fragment of ADAMTS-1 that inhibits cell proliferation or metastasis.

In some embodiments, the present invention provides methods of treating cancer in an individual comprising administering to the individual a therapeutically effective amount of a polypeptide fragment of ADAMTS-1 and/or a nucleic acid that encodes a polypeptide fragment of ADAMTS-1 that inhibits cell proliferation or metastasis.

In some embodiments, the present invention provides methods of treating cancer comprising administering an inhibitor of the metalloproteinase activity of ADAMTS-1.

In some embodiments, the present invention provides methods of treating cancer comprising administering a therapeutically effective amount of a composition comprising a polypeptide fragment of ADAMTS-1 comprising the spacer/Cys-rich domain or the spacer domain of ADAMTS-1 or a nucleic acid molecule encoding a polypeptide fragment of ADAMTS-1 comprising the spacer/Cys-rich domain or the spacer domain of ADAMTS-1.

BRIEF DESCRIPTION OF THE FIGURES

FIG. 1A and FIG. 1B depict how ADAMTS-1 undergoes auto-proteolytic cleavage and the self-cleavage of ADAMTS-1 is regulated. FIG. 1A depicts a diagram of the expression constructs. FIG. 1B depicts how cleavage of ADAMTS-1 is blocked by heparin and HS. TA3ADAMTS-1 cells were cultured in the absence (lane 1) or presence of 100 μg/ml of heparin (lane 2), HS (lane 3), hyaluronan (lane 4), or CS (lane 5) for 48 hours and the cell culture supernatants were analyzed by Western blot with anti-v5 antibody.

FIG. 2A through FIG. 2E depict that ADMATS-1 promotes pulmonary metastasis, while ADAMTS-1NTCF and ADAMTS-1CTCF inhibits the process. FIG. 2A depicts a representative gross pictures of the mouse lungs two-three weeks after i.v. injection of TA3wtb (a-c) or TA3ADAMTS-1 (d-f), or TA3ADAMTS-1NTCF (g-i) cells. FIG. 2B depicts the survival rate of the experimental mice which were injected with the TA3 transfectants intravenously. Total of thirty mice were used for each type of transfectants. FIG. 2C depicts that pulmonary metastatic burden is expressed by weight of the lungs derived from the experimental mice 11 days and 18 days after the i.v. injection of the TA3 transfectants. FIG. 2D depicts representative H&E stained lung sections were derived the experimental mice injected with TA3wtb (a), TA3ADAMTS-1 (b), TA3ADAMTS-1NTCF (c), and TA3ADAMTS-1CTCF (d) cells. Bar, 100 μm. FIG. 2E depicts Western blot analysis of v5-epitope tagged ADAMTS-1 protein expressed by TA3ADAMTS-1 cells in vivo using anti-v5 mAb. The proteins were derived from different pulmonary metastases derived from TA3ADAMTS-1 cells. The arrow indicates the mature proteolytically active ADAMST-1, and the arrowhead marks pro-ADAMTS-1.

FIG. 3A through FIG. 3C depict ADAMTS-1NTCF and ADAMTS-1CTCF blocks pulmonary metastasis by inhibiting proliferation and survival of tumor cells and by inhibiting tumor angiogenesis. A 5-Bromo-2′-deoxy-uridine (Brdu) incorporation assay and in situ detection of apoptotic cells on the sections derived from the experimental mouse lungs (six days after i.v. injection of TA3 transfectants) was performed. Results demonstrated that expression of ADAMTS-1NTCF or ADAMTS-1CTCF, but not that of ADAMTS-1minusTSP, inhibits proliferation and promotes apoptosis of the tumor cells, and inhibits tumor angiogenesis; while overexpression of full-length exogenous ADAMTS-1 on the top of endogenous ADAMTS-1 has weak effect on tumor cell proliferation and apoptosis and promotes tumor angiogenesis in vivo. The quantitative data that reveals the effects of ADAMTS-1 and the fragments of ADAMTS-1 on tumor cell apoptosis and proliferation and on tumor angiogenesis are shown in FIG. 3B.

FIG. 4A through FIG. 4D depict how ADAMTS-1NTCF and ADAMTS-1CTCF block activation of EGFR and ErbB-2 in vivo, while ADAMTS-1 promotes activation of these receptors and shedding of AR and HB-EGF precursors. FIG. 4A. Tumor cell tracking assay was performed to determine the pulmonary extravasation of TA3 transfectants. 24 hours after i.v. injection of the green fluorescein labeled TA3 transfectants, the mice lung were fixed and sectioned. TA3wtb (A-a), TA3ADAMTS-1 (A-b), TA3ADAMTS-NTCF (A-c), and TA3ADAMTS-1CTCF (A-d) cells in the lung parenchyma were shown. FIG. 4B. The pulmonary extravasation rates of the TA3 transfectants were expressed as average number of the tumor cells per microscopic field. FIG. 4C. Activation of EGFR and ErbB-2 in vivo: immunoprecipitation with anti-EGFR(C-a) or anti-ErbB-2 (C-b) antibody was performed using the protein lysates derived from the mouse lungs which were implanted without (lanes 1-3) or with TA3wtb(lanes 4-6), TA3ADAMTS-1 (lanes 7-9), TA3ADAMTS-1NTCF (lane 10-12), and TA3ADAMTS-1CTCF (lanes 13-15) 24 hours prior. To normalize number of the tumor cells that were included in the protein lysates, based on the tumor cell extravasation rates (B), 100 μg of the lung lysates with or without TA3wtb cells, 71 μg of the lung lysates containing TA3ADAMTS-1 cells, and 143 μg of the lung lysates containing TA3ADAMTS-1NTCF cells, and 130 μg of the lysates containing TA3ADAMTS-1CTCF cells have been used. The precipitated proteins were analyzed by Western blotting with anti-phosphotyrosine antibody to detect phosphor-EGFR(C-a, upper panel) or phosphor-ErbB-2 (C-b, upper panel), or with anti-EGFR(C-a, bottom panel) or anti-ErbB-2 (C-b, bottom panel) antibody to detect total amount of EGFR or ErbB-2, respectively. FIG. 4D. ADAMTS-1 promotes shedding of AR (D-a), HB-EGF (D-b), but not epigen (D-c), and the constitutive shedding of AR and HB-EGF is blocked or inhibited by ADAMTS-1E/Q, respectively (lane 4 in D-a and -b). Cos-7 cells were co-transfected with the expression constructs containing cDNA inserts that encode AR, HB-EGF, or epigen precursors without (lane 1) or with ADAMTS-1NTCF (lane 2), ADAMTS-1CTCF (lane 3), ADAMTS-1E/Q (lane 4), or ADAMTS-1 (lane 5), and the concentrated serum-free culture media derived from these co-transfected Cos-7 cells were analyzed using anti-AR, HB-EGF, or epigen antibody.

FIG. 5A through FIG. 5D depict how the ADAMTS-1 fragments block activation of EGFR and ErbB-2; while ADAMTS-1 promotes shedding of AR and HB-EGF. FIG. 5A. Immunoprecipitation with anti EGFR (A-a) or anti-ErbB-2 (A-b) antibody was performed by using the proteins derived from the mouse lungs received TA3wtb (lanes 1-3), TA3ADAMTS-1 (lanes 4-6), TA3ADAMTS-1minusTSP (lanes 7-9), TA3ADAMTS-1NTCF (lanes 10-12), and TA3ADAMTS-1ctcf (lanes 13-15) intravenously 5 days prior. The precipitated proteins were analyzed Western blotting with anti-phospho-tyrosine antibody to detect phosphor-EGFR (A-a, upper panel) and phosphor-ErbB-2 (A-b, upper panel), respectively or with anti-EGFR (A-a, bottom panel) or anti-ErbB-2 (A-b, bottom panel) antibody to detect total amount of EGFR or ErbB-2, respectively. FIG. 5B. ADAMTS-1 promotes shedding of AR (B-a), HB-EGF (B-b), but not epigen (B-c), and the shedding is blocked by ADAMTS-1E/Q (lane 4). Cos-7 cells were co-transfected with the expression constructs containing cDNA inserts that encode the EGF family ligand precursors with or without (lane 1) of TA3ADAMTS-1NTCF (lane 2), TA3ADAMTS-1CTCF (lane 3), ADAMTS-1E/Q (lane 4), and ADAMTS-1 (lane 5). FIG. 5C. The cell culture supernatants derived from the AR-(C-a) or HB-EGF (C-b) transfected Cos-7 cells were applied to serum-starved MCF-10A cells without (lane 5-6) or without prior absorption of the supernatants with blocking antibodies against AR (a, lane 7-8) or HB-EGF (b, lane 7-8) in the presence of 400 ng of ADAMTS-1 (lane 9-10), ADAMTS-1NTCF (lane 11-12), or ADAMTS-1CTCF (lane 13-14). Serum free medium alone (lane 1-2) or containing 5 ng of AR (a, lane 3-4) or 4 ng of HB-EGF (b, lane 3-4) was applied to serum starved MCF-10A cells. Equal amount of the proteins derived from the MCF-10A cells were analyzed Western blotting with anti-phospho-Erk1/2 to detect phosphor-Erk1/2 or with anti-Erk antibody to detect total amount of Erk1/2. FIG. 5D. The cleavage fragments of ADAMTS-1 blocks activation of Erk1/2 in HUVECs induced by VEGF 165 (D-a), TGF-α (D-c), HB-EGF (D-d), and AR (D-e), but not that induced by bFGF (D-b). HUVECs were applied with SFM alone (lane 1) or containing different GFs alone (lane 2) with 400 ng of ADAMTS-1 (lane 3), ADAMTS-1minusTSP (lane 4), ADAMTS-1NTCF (lane 5) or ADAMTS-1CTCF (lane 6). Equal amount of the proteins derived from HUVECs were analyzed Western blotting with anti-phospho-Erk1/2 to detect phosphor-Erk1/2 or with anti-Erk antibody to detect total amount of Erk1/2.

FIG. 6A through FIG. 6D depict how ADAMTS-1NTCF and ADAMTS-1CTCF blocks activation of Erk1/2 kinases in HUVECs induced by AR, HB-EGF, or VEGF165. VEGF165 (A, 15 ng/ml), bFGF (B, 15 ng/ml), HB-EGF (C, 4 ng/ml), or AR (D, 5 ng/ml) was used alone (lane 2) or in the presence of full-length ADAMTS-1 and ADAMTS-1 fragments (lanes 3-6). The serum-starved HUVECs were applied with SFM alone (lane 1), or containing different GFs alone (lane 2) or the GFs plus 400 ng of ADAMTS-1 (lane 3), ADAMTS-1minusTSP(lane 4), ADAMTS-1NTCF (lane 5) or ADAMTS-1CTCF (lane 6). Equal amount of the proteins derived from these HUVECs were analyzed Western blotting with anti-phospho-Erk1/2 to detect phosphor-Erk1/2 (upper panels in A-D) or with anti-Erk antibody to detect total amount of Erk1/2 (bottom panels in A-D).

FIG. 7A through FIG. 7D depict full-length ADMATS-1 promotes pulmonary metastasis of TA3 cells, while ADAMTS-1E/Q, ADAMTS-1NTF, or ADAMTS-1CTF inhibits the process. FIG. 7A depicts a schematic diagram of the domain organization of full-length ADAMTS-1 and the different deletional and point-mutated ADAMTS-1 that were used in FIGS. 1, 2, 3, and 8. FIG. 7B depicts the expression levels of full-length ADAMTS-1 (lane 1), ADAMTS-1E/Q (lane 2), ADAMTS-1NTF (lane 3), ADAMTS-1minusTSP-1 (lane 4), or ADAMTS-1CTF (lane 5) by the pooled populations of TA3 transfectants. FIG. 7C depicts the survival rates of the experimental mice which were injected with the different TA3 transfectants intravenously. A total of 12 mice were used for each type of transfectants. FIG. 7D depicts the pulmonary metastatic burden was expressed by the weight of the tumor bearing mouse lungs derived from the experimental mice 12 and 20 days after the i.v. injection of the TA3 transfectants.

FIG. 8A through FIG. 8C depict the spacer/Cys-rich domain of ADAMTS-1 plays a major role in binding of ADAMTS-1 to the ECM and the cells. Western blotting was performed using anti-v5 antibody to determine the distribution patterns of the v5-epitope tagged full-length ADAMTS-1 (lane 1), ADAMTS-1E/Q (lane 2), ADAMTS-1NTF+spacer/Cys-rich (lane 3), ADAMTS-1NTF (lane 4), ADAMTS-1minusTSP-1 (lane 5), ADAMTS-1CTF (lane 6), ADAMTS-1CTF+spacer (lane 7), and ADAMTS-13TSP-1 (lane 8) in the cell culture supernatants (FIG. 8A), the ECM materials (FIG. 8B), and the EDTA-lifted Cos-7 cells (FIG. 8C) that were transfected with the corresponding expression constructs.

FIG. 9A through FIG. 9D depict how ADAMTS-1NTCF and ADAMTS-1CTCF blocks activation of Erk1/2 kinases in HUVECs induce by AR, HB-EGF, or VEGF165. VEGF165 (A, 15 ng/ml), bFGF (B, 15 ng/ml), HB-EGF (C, 4 ng/ml), or AR (D, 5 ng/ml) was used alone (lane 2) or in the presence of full-length ADAMTS-1 and ADAMTS-1 fragments (lanes 3-6). The serum-starved HUVECs were applied with SFM alone (lane 1), or containing different GFs alone (lane 2) or the GFs plus 400 ng of ADAMTS-1 (lane 3), ADAMTS-1minusTSP(lane 4), ADAMTS-1NTCF (lane 5) or ADAMTS-1CTCF (lane 6). Equal amount of the proteins derived from these HUVECs were analyzed Western blotting with anti-phospho-Erk1/2 to detect phosphor-Erk1/2 (upper panels in A-D) or with anti-Erk antibody to detect total amount of Erk1/2 (bottom panels in A-D).

FIG. 10 depicts expression of ADAMTS-1. Expression of ADAMTS-1 was assessed by RT-PCR using RNAs derived from TA3wt, TA3wt1, Lewis lung carcinoma cells, CMT-93 colon carcinoma cells, B 16F1 and F10 cells, 3T3 fibroblasts, C2C12 myoblasts, and mouse placenta (lanes 2-10). Expression of β-actin by these cells was used as controls. In lane 1, reverse transcriptase was not included in RT reaction with RNA derived from TA3wt1 cells.

FIG. 11A and FIG. 11B depict how ADAMTS-1 promotes tumor growth while the cleavage fragments of ADAMTS-1 inhibit tumor growth. Growth rates of the s.c. tumors derived from different TA3 transfectants are expressed as the means of tumors volumes +/−SD. Total of fifteen mice were used for each type of transfectants.

FIG. 12A and FIG. 12B depict the cleavage fragments of ADAMTS-1 blocks subcutaneous tumor growth by inhibiting proliferation and survival of tumor cells, and inhibiting tumor angiogenesis in vivo. The s.c. tumors were section 12 days after implanting TA3wtb (A, a-d), TA3ADAMTS-1 (A, e-h), TA3ADAMTS-1NTCF (A, i-l), and TA3ADAMTS-1CTCF (A, m-p). These sections were stained with H&E (A-a, e, I, m), or reacted with Apoptag to detect apoptotic tumor cells in situ (A-b, f, j, n), anti-Brdu antibody to detect proliferating tumor cells (A-c, g, k, o), or with anti-vWF antibody to reveal blood vessels with the tumors (a-d, h, l, p). Bar: 120 μm. The quantitative data that reveals the effects of ADAMTS-1 and the fragments of ADAMTS-1 on tumor cell apoptosis and proliferation in vivo and on tumor angiogenesis are shown in panels B-a, b, c, respectively.

FIG. 13A through FIG. 13D depicts ADAMTS-1NTCF and ADAMTS-1CTCF blocks activation of Erk1/2 kinases in HUVECs induce by AR, HB-EGF, or VEGF165. VEGF165 (A, 15 ng/ml), bFGF (B, 15 ng/ml), HB-EGF (C, 4 ng/ml), or AR (D, 5 ng/ml) was used alone (lane 2) or in the presence of full-length ADAMTS-1 and ADAMTS-1 fragments (lanes 3-6). The serum-starved HUVECs were applied with SFM alone (lane 1), or containing different GFs alone (lane 2) or the GFs plus 400 ng of ADAMTS-1 (lane 3), ADAMTS-1minusTSP (lane 4), ADAMTS-1NTCF (lane 5) or ADAMTS-1CTCF (lane 6). Equal amount of the proteins derived from these HUVECs were analyzed Western blotting with anti-phospho-Erk1/2 to detect phosphor-Erk1/2 (upper panels in A-D) or with anti-Erk antibody to detect total amount of Erk1/2 (bottom panels in A-D). The ADAMTS-1 fragments block activation of EGFR and ErbB-2; while ADAMTS-1 promotes shedding of AR and HB-EGF. A. Immunoprecipitation with anti EGFR (A-a) or anti-ErbB-2 (A-b) antibody was performed by using the proteins derived from the mouse lungs received TA3wtb (lanes 1-3), TA3ADAMTS-1 (lanes 4-6), TA3ADAMTS-1minusTSP (lanes 7-9), TA3ADAMTS-1NTCF (lanes 10-12), and TA3ADAMTS-1ctcf (lanes 13-15) intravenously 5 days prior. The precipitated proteins were analyzed Western blotting with anti-phospho-tyrosine antibody to detect phosphor-EGFR (A-a, upper panel) and phosphor-ErbB-2 (A-b, upper panel), respectively or with anti-EGFR (A-a, bottom panel) or anti-ErbB-2 (A-b, bottom panel) antibody to detect total amount of EGFR or ErbB-2, respectively. B. ADAMTS-1 promotes shedding of AR (B-a), HB-EGF (B-b), but not epigen (B-c), and the shedding is blocked by ADAMTS-1E/Q (lane 4). Cos-7 cells were co-transfected with the expression constructs containing cDNA inserts that encode the EGF family ligand precursors with or without (lane 1) of TA3ADAMTS-1NTCF (lane 2), TA3ADAMTS-1CTCF (lane 3), ADAMTS-1E/Q (lane 4), and ADAMTS-1 (lane 5). C. The cell culture supernatants derived from the AR-(C-a) or HB-EGF (C-b) transfected Cos-7 cells were applied to serum-starved MCF-10A cells without (lane 5-6) or without prior absorption of the supernatants with blocking antibodies against AR (a, lane 7-8) or HB-EGF (b, lane 7-8) in the presence of 400 ng of ADAMTS-1 (lane 9-10), ADAMTS-1NTCF (lane 11-12), or ADAMTS-1CTCF (lane 13-14). Serum free medium alone (lane 1-2) or containing 5 ng of AR (a, lane 3-4) or 4 ng of HB-EGF (b, lane 3-4) was applied to serum starved MCF-10A cells. Equal amount of the proteins derived from the MCF-10A cells were analyzed Western blotting with anti-phospho-Erk1/2 to detect phosphor-Erk1/2 or with anti-Erk antibody to detect total amount of Erk1/2. D. The cleavage fragments of ADAMTS-1 blocks activation of Erk1/2 in HUVECs induced by VEGF 165 (D-a), TGF-α (D-c), HB-EGF (D-d), and AR (D-e), but not that induced by bFGF (D-b). HUVECs were applied with SFM alone (lane 1) or containing different GFs alone (lane 2) with 400 ng of ADAMTS-1 (lane 3), ADAMTS-1minusTSP (lane 4), ADAMTS-1NTCF (lane 5) or ADAMTS-1CTCF (lane 6). Equal amount of the proteins derived from HUVECs were analyzed Western blotting with anti-phospho-Erk1/2 to detect phosphor-Erk1/2 or with anti-Erk antibody to detect total amount of Erk1/2.

FIG. 14 depicts the domain organization of ADAMTS-1. The various domains of ADAMTS-1 are shown.

FIG. 15 depicts possible mechanisms of ADAMTS-1 function. 1) Full-length ADAMTS-1 promotes tumor growth and metastasis by enhancing tumor cell proliferation/survival and tumor angiogenesis through shedding/activating HB-EGF and AR transmembrane precursors and by promoting tumor cell invasion through degrading versican; 2) full-length ADAMTS-1 binds to their substrates through its spacer/Cys-rich domain directly or indirectly through binding to HSPGs. Thus, the whole or different parts of the spacer/Cys-rich domain can be used as a dominant negative regulator of the full-length ADAMTS-1 (by regulating the substrate-binding of ADAMTS-1) and to regulate its own cleavage status (to promote proteolytic cleavage of ADAMTS-1, therefore generate anti-tumor fragments); 3) the anti-tumor activity of the ADAMTS-1 fragments resides in the TSP-1 domains, which exerts the anti-tumor activity by inhibiting bioactivity of several soluble heparin binding growth/angiogenic factors including AR and HB-EGF. Thus, the whole or parts of ADAMTS-1NTF (ADANTS-1NTFE/Q) and/or ADAMTS-1CTF can be used to inhibit cancers.

FIG. 16A through FIG. 16D depict full-length ADAMTS-1 and the ADAMTS-1 fragments displayed opposite effects on growth and metastasis of LLC cells. FIG. 16A. The expression level of the v5-epitope tagged ADAMTS-1 (lane 1), ADAMTS-1E/Q (lane 2), ADAMTS-1NTF (lane 3), ADAMTS-1minusTSP-1 (lane 4), ADAMTS-1CTF (lane 5), thrombospondin-1 (lane 6) and thrombospondin-2 (lane 7) by the pooled LLC transfectants. FIG. 16B. The growth rates of the s.c. tumors derived from the different LLC transfectants are expressed as the means of tumors volumes +/−SD. A total of 15 mice were used for each type of transfectants. FIG. 16C. Survival rates of the experimental mice after removal of the s.c. tumors derived from the different LLC transfectants. A total of thirty mice were used for each type of transfectants. FIG. 16D. Pulmonary metastatic burden is expressed by the average weight of the lungs derived from experimental mice three weeks after removal of the s.c. tumors.

FIG. 17A through FIG. 17C depict the metalloproteinase activity of ADAMTS-1NTF is not required for its anti-tumor activity. FIG. 17A. The expression level of the v5-epitope tagged ADAMTS-1NTF (lane 1-3) and ADAMTS-1NTFE/Q(lane 4-6) by the TA3 transfectants. FIG. 17B Survival rates of the experimental mice after i.v. injection of 1×106/mouse TA3 transfectants. A total of 15 mice were used for each type of transfectants. FIG. 17C. Pulmonary metastatic burden is expressed by the average weight of the lungs derived from the experimental mice three weeks after the iv injection.

FIG. 18 depicts the multiple amino acid sequence alignment of the second and third repeats of thrombospondin-1 and m and cTSP-1 domains in ADAMTS-1, and the deletional and peptide generation strategy in the TSP-1 domains of ADAMTS-1. The deletions and generation of three different peptides s in m and cTSP-1 domains of ADAMTS-1 are shown.

DETAILED DESCRIPTION

In the present invention it has been discovered that ADAMTS-1 is expressed by many tumor cells and overexpression of ADAMTS-1 promotes growth and metastasis of TA3 mammary carcinoma cells by promoting survival, proliferation, invasiveness of the tumor cells and tumor angiogenesis in vivo. Additionally, disclosed herein is that ADAMTS-1 undergoes auto-proteolytic cleavage to generate N- and C-terminal cleavage fragments that contain at least one TSP type I motif. Auto-proteolytic cleavage of ADAMTS-1 is blocked by heparin and heparin sulfate (HS). Although not bound by any theory, this indicates that the self-cleavage is regulated by HS and heparin sulfate proteoglycans (HSPGs). Thus, as described herein, ADAMTS-1 expressed by TA3 cells is maintained in the full-length form in vivo to exert pro-tumor growth and metastasis activity. In contrast to the full-length ADAMTS-1, overexpression of the N- or C-terminal fragment of ADAMTS-1 (ADAMTS-1NTCF and/or ADAMTS-1CTCF) inhibits subcutaneous (s.c.) growth of TA3 cells and blocks pulmonary metastasis of the cells by inhibiting proliferation and inducing apoptosis of the tumor cells and by inhibiting tumor angiogenesis. Additionally, the anti-tumor effect of the ADAMTS-1 fragments requires a TSP type-I motif. The direct evidence was provided for the first time that ADAMTS-1 promotes tumor growth and metastasis, and can serve as a target for cancer therapy.

For the first time, it has been demonstrated that unlike full-length ADAMTS-1 which promotes shedding of the EGF family ligands including amphiregulin (AR) and heparin-binding EGF (HB-EGF) and activation of EGF receptor (EGFR) and ErbB-2, the cleavage fragments of ADAMTS-1 inhibits activation of EGFR and ErbB-2 in vivo, and interferes with Erk1/2 kinases activation induced by soluble AR. HB-EGF, and/or VEGF in mammary epithelial cells and endothelial cells. These different effects likely underlie the opposite roles of ADAMTS-1 and its cleavage fragments in tumor growth and metastasis, suggesting the ADAMTS-1 fragments and the inhibitors of ADAMTS-1 can be most successfully used to treat the cancers overexpressing these heparin binding growth and angiogenic factors and with activated erbB-signaling pathways.

The term “ADAMTS-1NTCF” can also be referred to as “ADAMTS-1NTF”. The term “ADAMTS-1CTCF” can also be referred to as “ADAMTS-1CTF”. In some embodiments, ADAMTS-1NTCF comprises SEQ ID NO: 9 and/or 11. In some embodiments, ADAMTS-1CTCF comprises SEQ ID NO: 5 and/or 7.

The discovery that ADAMTS-1 can be cleaved into at least two fragments has led to the following invention. In some embodiments, the present invention provides an isolated polypeptide comprising a fragment of ADAMTS-1 that inhibits cell growth or cell survival and/or metastasis.

As used herein, the term “isolated polypeptide fragment” refers to a polypeptide fragment that is free of the full length protein. In some embodiments, the isolated polypeptide is also free of nucleic acid molecules. In some embodiments, the isolated polypeptide is free of cellular membranes. In some embodiments, the isolated polypeptide has been purified away from cellular components. In some embodiments, the polypeptide comprises a fragment of SEQ ID NO: 1 and/or SEQ ID NO: 3. In some embodiments, the fragment of ADAMTS-1 comprises SEQ ID NO: 5, 7, 9, and/or 11. The fragment of ADAMTS-1 can be any length such that it is not the full-length ADAMTS-1 protein. In some embodiments, the fragment comprises about 100 to about 150, about 100 to about 200, about 100 to about 300, about 100 to about 400, about 100 to about 500, about 100 to about 600, about 100 to about 700, about 100 to about 800, about 100 to about 900, or about 100 to 950 amino acid residues. In some embodiments, the fragments of ADAMTS-1 comprise modifications of the polypeptide sequence. The modification can be any modification including, but not limited to, mutations, insertions, substitutions, deletions, and the like. In some embodiments, the fragment comprises a mutation of Glu to Gln. In some embodiments, the mutation of Glu to Gln occurs at a position corresponding to position 386 (in mouse ADAMTS-1) in the full length protein. One of skill in the art can determine what position in a fragment corresponds to position 386 in the full length protein (e.g. position 385 in human ADAMTS-1). One of skill in the art can do this by, for example, performing an alignment using any alignment software or BLAST software using default settings. Examples of software that can be used include, but are not limited to, BLAST, GCG, and MacVector™. In some embodiments, the polypeptide fragment containing a mutation comprises SEQ ID NO: 33 and/or 35 or a nucleic acid molecule encoding the same. In some embodiments, the nucleic acid molecule encoding the fragment comprises SEQ ID NOs: 34 and/or 36.

In some embodiments the fragments of ADAMTS-1 are linked to a non-ADAMTS-1 molecule. In some embodiments, the non-ADAMTS-1 molecule is a toxin, peptide, polypeptide, small molecule, drug, and the like. In some embodiments, the non-ADAMTS-1 molecule is a 6-His-tag, GST polypeptide, HA tag, the Fc fragment of human IgG and the like. In some embodiments, the proteinase cleavage sites will be put before the tag sequences, so that after purification these tags can be removed by proteolytic cleavage. For example, the HRV 3C (human rhinovirus type 14 3C) protease cleavage site (LEVLFQ↓GP-SEQ ID NO:46) can be located before the COOH-terminal v5 and His epitope tags. The HRV 3C protease specifically claves the sequence LEVLFQ↓GP at 40C and were used to efficiently removal the COOH-terminal tags (Novagen).

In some embodiments, the fragment of ADAMTS-1 is fused to another polypeptide that is derived from a protein that is not ADAMTS-1. In some embodiments two fragments from ADAMTS-1 are fused or linked together. In some embodiments, the two fragments are identical. In some embodiments, the fragments are different from one another. The fragments that can be linked or fused together are ADAMTS-1CTCF (SEQ ID NO: 5 and/or SEQ ID NO:7), ADAMTS-1NTCF (SEQ ID NO: 9 and/or 11), and ADAMTS-1spacer or ADAMTS-1spacer/Cys-rich to achieve maximal anti-tumor efficiency, however any two fragments from ADAMTS-1 can be fused together.

In some embodiments, the present invention provides nucleic acid molecules encoding a fragments of ADAMTS-1. In some embodiments, the fragments of ADAMTS-1 that inhibits cell proliferation or metastasis comprise a TSP type-I motif.

A fragment that inhibits cell proliferation or metastasis can also be referred to as a fragment that inhibits cancer or a fragment can be used to treat cancer.

As used herein, the term “inhibit cell proliferation” refers to any measurement of cell proliferation. A fragment, compound, or composition that causes a cell to undergo necrosis or apoptosis is considered to inhibit cell proliferation. Cell proliferation can also be referred to as cell growth or cell division.

Methods of measuring cell proliferation, division, and metastasis are routine and any method can be used.

For example, one can measure cell invasion using Matrigel in vitro. Metastasis can also be measured and/or observed in vivo by injecting a mouse with a tumor cell and determining if the cell spreads to a different location away from the sight of injection. Metastasis can also be measured by measuring or observing tumor burden or tumor growth in areas that are distinct from the primary tumor location. Cell proliferation can be measured, for example, by counting cells. Cell division can be measured, for example, by monitoring what phase of the cell cycle a cell or a population of cells is in by using flow cytometry or FACS. Determining if a cell or cell population is dividing is routine.

In some embodiments, the present invention provides a fragment of ADAMTS-1 that lacks a TSP motif. In some embodiments, the present invention provides a deletion of ADAMTS-1 that lacks a TSP motif. In some embodiments, a polypeptide of ADAMTS-1 that lacks a TSP motif comprises SEQ ID NO:13 and/or SEQ ID NO:15. The term “ADAMTS-1minus TSP” can also be referred to as “ADAMTS-1minus TSP-1”. In some embodiments, the present invention provides a nucleic acid molecule that encodes for a ADAMTS-1 polypeptide that lacks a TSP motif. In some embodiments the nucleic acid molecule is isolated. In some embodiments the nucleic acid molecule comprises SEQ ID NO: 14 and/or SEQ ID NO: 16.

In some embodiments, the present invention provides an isolated nucleic acid molecule (polynucleotide) encoding a polypeptide fragment of ADAMTS-1.

As used herein the term “isolated nucleic acid molecule encoding a polypeptide fragment of ADAMTS-1” refers to a nucleic acid molecule is free of a nucleic acid molecule encoding full length ADAMTS-1.

In some embodiments, a fragment encoded by the nucleic acid molecule can inhibit cell proliferation and/or metastasis. In some embodiments, the nucleic acid molecule comprises a fragment of a nucleic acid molecule encoding a polypeptide comprising SEQ ID NO:1 and/or SEQ ID NO: 2. In some embodiments the nucleic acid molecule comprises a fragment of SEQ ID NO: 3 and/or SEQ ID NO: 4 In some embodiments, the nucleic acid molecule encodes a polypeptide comprising SEQ ID NOs: 5, 7, 9, and/or 11. In some embodiments, the nucleic acid molecule comprises SEQ ID NOs: 6, 8, 10, and/or 12. In some embodiments, the nucleic acid molecule encoding a fragment of ADAMTS-1 is operably linked to a promoter. In some embodiments, the promoter can facilitate the expression in a prokaryotic cell and/or eukaryotic cell. The promoter can be any promoter that can drive the expression of the nucleic acid molecule. Examples of promoters include, but are not limited to, CMV, SV40, pEF, actin promoter, and the like. In some embodiments, the nucleic acid molecule is DNA or RNA. In some embodiments, the nucleic acid molecule is a virus, vector, or plasmid. In some embodiments, the expression of the nucleic acid molecule is regulated such that it can be turned on or off based on the presence or absence of a regulatory substance. Examples of such a system include, but is not limited to a tetracycline-ON/OFF system.

In some embodiments, the nucleic acid molecule is a recombinant viral vector. “A recombinant viral vector” refers to a construct, based upon the genome of a virus, that can be used as a vehicle for the delivery of nucleic acids encoding proteins, polypeptides, or peptides of interest. Recombinant viral vectors are well known in the art and are widely reported. Recombinant viral vectors include, but are not limited to, retroviral vectors, adenovirus vectors, adeno-associated virus vectors, and lenti-virus vectors, which are prepared using routine methods and starting materials.

Using standard techniques and readily available starting materials, a nucleic acid molecule may be prepared. The nucleic acid molecule may be incorporated into an expression vector which is then incorporated into a host cell. Host cells for use in well known recombinant expression systems for production of proteins are well known and readily available. Examples of host cells include bacteria cells such as E. coli, yeast cells such as S. cerevisiae, insect cells such as S. frugiperda, non-human mammalian tissue culture cells Chinese hamster ovary (CHO) cells or Cos-7 cells, and human tissue culture cells such as 293 cells or HeLa cells.

In some embodiments, for example, one having ordinary skill in the art can, using well known techniques, insert DNA molecules into a commercially available expression vector for use in well known expression systems. For example, the commercially available plasmid pSE420 (Invitrogen, San Diego, Calif.) may be used for production of immunomodulating proteins in E. coli. The commercially available plasmid pYES2 (Invitrogen, San Diego, Calif.) may, for example, be used for production in S. cerevisiae strains of yeast. The commercially available MAXBAC™ complete baculovirus expression system (Invitrogen, San Diego, Calif.) may, for example, be used for production in insect cells. The commercially available plasmid pcDNAI, pcDNA3, or PEF6/v5-His (Invitrogen, San Diego, Calif.) may, for example, be used for production in mammalian cells such as Cos-7 and CHO cells. One having ordinary skill in the art can use these commercial expression vectors and systems or others to produce proteins by routine techniques and readily available starting materials. (See e.g., Sambrook et al., eds., 2001, supra) Thus, the desired proteins or fragments can be prepared in both prokaryotic and eukaryotic systems, resulting in a spectrum of processed forms of the protein or fragments.

One having ordinary skill in the art may use other commercially available expression vectors and systems or produce vectors using well known methods and readily available starting materials. Expression systems containing the requisite control sequences, such as promoters and polyadenylation signals, and preferably enhancers, are readily available and known in the art for a variety of hosts (See e.g., Sambrook et al., eds., 2001, supra).

In some embodiments, the nucleic acid molecules can also be prepared as a genetic construct. “Genetic constructs” include regulatory elements necessary for gene expression of a nucleic acid molecule. The elements include: a promoter, an initiation codon, a stop codon, and a polyadenylation signal. In addition, enhancers can be used for gene expression of the sequence that encodes the protein or fragment. It is necessary that these elements be operably linked to the sequence that encodes the desired polypeptide and that the regulatory elements are operably in the individual or cell to whom they are administered. Initiation codons and stop codon are generally considered to be part of a nucleotide sequence that encodes the desired protein. However, it is necessary that these elements are functional in the individual or cell to which the gene construct is administered. The initiation and termination codons must be in frame with the coding sequence. Promoters and polyadenylation signals used must be functional within the cells. Examples of promoters useful to practice the present invention include but are not limited to promoters from Simian Virus 40 (SV40), Mouse Mammary Tumor Virus (MMTV) promoter, Human Immunodeficiency Virus (HIV) such as the HIV Long Terminal Repeat (LTR) promoter, Moloney virus, ALV, Cytomegalovirus (CMV) such as the CMV immediate early promoter, Epstein Barr Virus (EBV), Rous Sarcoma Virus (RSV) as well as promoters from human genes such as human Actin, human Myosin, human Hemoglobin, human muscle creatine and human metallothionein. Examples of polyadenylation signals useful to practice the present invention include but are not limited to SV40 polyadenylation signals and LTR polyadenylation signals. In some embodiments, the SV40 polyadenylation signal which is inpCEP4 plasmid (Invitrogen, San Diego Calif.), referred to as the SV40 polyadenylation signal, is used. In addition to the regulatory elements required for DNA expression, other elements may also be included in the DNA molecule. Such additional elements include enhancers. The enhancer may be selected from the group including but not limited to: human Actin, human Myosin, human Hemoglobin, human muscle creatine and viral enhancers such as those from CMV, RSV and EBV. Genetic constructs can be provided with mammalian origin of replication in order to maintain the construct extrachromosomally and produce multiple copies of the construct in the cell. Plasmids pCEP4 and pREP4 from Invitrogen (San Diego, Calif.) contain the Epstein Barr virus origin of replication and nuclear antigen EBNA-1 coding region which produces high copy episomal replication without integration. In some embodiments, the nucleic acid molecule is free of infectious particles.

In some embodiments, the present invention provides compositions comprising at least one polypeptide fragment of ADAMTS-1 that inhibits cell proliferation or cell growth or metastasis. In some embodiments, the composition comprises a fragment comprising SEQ ID NO:1 and/or SEQ ID NO:2. In some embodiments, the composition comprises a fragment comprising SEQ ID NO:5, SEQ ID NO:7, SEQ ID NO: 9, and/or SEQ ID NO: 11. In some embodiments, the composition comprises two or at least two polypeptide fragments of ADAMTS-1. In some embodiments, the fragment comprises the TSP-type I motif. In some embodiments, the composition is a pharmaceutical composition.

As used herein, the term “fragment of ADAMTS-1 that inhibits cell proliferation or metastasis” refers to a fragment of ADAMTS-1 that can inhibit cell growth, cell division, or cell proliferation. In some embodiments, the fragment inhibits cell growth, cell division, cell proliferation, or metastasis by 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, or 100%. In some embodiments, the fragment can inhibit cell growth, cell division, cell proliferation, or metastasis by at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, or at least 99%. One of skill in the art can determine the level of inhibition by, for example, comparing the property or properties of the cell or population of cells in the absence of the fragment.

A fragment of ADAMTS-1 that inhibits cell proliferation, cell growth, cell division, or metastasis can be identified using any known growth, proliferation, or division assay. For example, one of skill in the art can contact a fragment of ADAMTS-1 with a cell either in vitro or in vivo and determine whether the cell's growth, proliferation, divisions, or metastasis have been inhibited. One of skill in the art could also use a nucleic acid molecule encoding a fragment of ADAMTS-1 and introduce the nucleic acid molecule into a cell or organism such that the fragment is expressed. The cell's or population of cell's property or properties could then be measured and it could be determined whether the fragment encoded by the nucleic acid molecule can inhibit cell growth (proliferation), cell division, or metastasis.

The fragments of ADAMTS-1 that can be used to inhibit cell proliferation or growth can be produced by a cleavage event of ADAMTS-1. In some embodiments, the cleavage produces fragments of ADAMTS-1 comprising SEQ ID NOs: 5, 7, 9 and/or 11 or encoded by nucleic acid molecules comprising SEQ ID NO: 6, 8, 10, and/or 12.

In some embodiments, the present invention provides methods of inhibiting the cleavage of ADAMTS-1 in a cell comprising contacting the cell with a cleavage inhibiting factor. A “cleavage inhibiting factor” is a compound or composition that can inhibit the cleavage of ADAMTS-1. In some embodiments, the cleavage of ADAMTS-1 is auto-cleavage or cleavage that is facilitated by a protease that is not ADAMTS-1. In some embodiments, the cleavage inhibiting factor is heparin or heparan sulfate proteoglycans (HSPGs). Heparin or derivatives of heparin were found to inhibit the cleavage of ADAMTS-1 as described herein. In some embodiments, the present invention provides methods of promoting cleavage of ADAMTS-1 comprising contacting ADAMTS-1 with a ADAMTS-1 cleavage activating factor. In some embodiments, the cleavage activating factor is a compound that inhibits and/or sequesters heparin. In some embodiments, the factor that inhibits heparin is heparinase, platelet factor 4 (PF4-a), protamine, or polybrene. A “cleavage activating factor” is a compound or composition that enhances, induces, or increases the level of cleavage of ADAMTS-1. In some embodiments, the cleavage of ADAMTS-1 can be auto-cleavage. In some embodiments, the cleavage of ADAMTS-1 can be facilitated by a protease that is not ADAMTS-1.

In some embodiments, the present invention provides methods of inhibiting cell proliferation or metastasis comprising contacting the cell with a fragment of ADAMTS-1 that inhibits cell proliferation or metastasis. In some embodiments, the fragment of ADAMTS-1 comprises a fragment of SEQ ID NO:1 and/or SEQ ID NO:3. In some embodiments, the fragment comprises SEQ ID NOs: 5, 7, 9 and/or 11. One of skill in the art can determine if the fragment inhibits cell proliferation or metastasis of a cell or population of cells by measuring the growth or metastasis in the presence and/or absence of the fragment of ADAMTS-1.

As used herein, the term “cell” refers to any cell. In some embodiments, the cell is a human cell or a mouse cell. In some embodiments, the cell is a tumor cell, inflammatory cells, or keratinocytes. In some embodiments, the cell is a primary tumor cell. As used herein, the term “primary tumor cell” refers to a cell that has been excised from a tumor from an individual or animal and has not been propagated through more than 10 cell divisions.

The discovery that fragments of ADAMTS-1 can inhibit cell growth and/or metastasis demonstrates that in some embodiments, the present invention provides methods of treating cancer in an individual comprising administering to the individual a therapeutically effective amount of a fragment of ADAMTS-1 that is able to inhibit cell proliferation or metastasis. The fragments can also be said to inhibit tumor growth and the like. In some embodiments, the fragment comprises a fragment of SEQ ID NO:1 and/or SEQ ID NO: 3. In some embodiments, the fragment comprises SEQ ID NOs: 5, 7, 9 and/or 11. In some embodiments, the fragment of ADAMTS-1 is co-administered with at least one other cancer treatment. The fragment of ADAMTS-1 can be either administered prior to, subsequently to, or at the same time as the other cancer treatment. The fragment(s) of ADAMTS-1 can be co-administered with any other cancer treatment, including, but not limited to, surgery, chemotherapy, antibodies, small molecules, radiation, and the like. In some embodiments, the fragment of ADAMTS-1 that is used to treat the cancer in an individual is a fragment of ADAMTS-1 that is able to inhibit cell proliferation, metastasis, or angiogenesis. In some embodiments, the fragment inhibits cell proliferation and/or metastasis in vitro.

Since it has been discovered that the full length ADAMTS-1 is pro-cancer while the cleavage fragments of ADAMTS-1 have anti-cancer properties, the present invention provides methods of treating cancer in an individual comprising administering to the individual a composition that induces the cleavage of ADAMTS-1. In some embodiments, the composition that induces the cleavage of ADAMTS-1 is a heparin inhibitor. Examples of heparin inhibitors include, but are not limited to, heparinase, platelet factor 4 (PF4-a), protamine, polybrene, the heparin-binding domain/peptide derived from HSPGs, and the like. In some embodiments, the cleavage of ADAMTS-1 results in the production of at least one fragment of ADAMTS-1 that can inhibit cell proliferation or metastasis. In some embodiments, the cleavage of ADAMTS-1 results in the production of two or at least two fragments of ADAMTS-1 that can inhibit cell proliferation or metastasis. In some embodiments, the fragments that are produced by the cleavage of ADAMTS-1 comprise SEQ ID NOs: 5, 7, 9 and/or 11.

In some embodiments, the present invention provides methods of inhibiting metastasis in an individual comprising administering the individual a fragment or mutant of ADAMTS-1 that inhibits metastasis and/or angiogenesis. In some embodiments, the mutant of ADAMTS-1 is a metalloproteinase defective mutant. In some embodiments, the fragment of ADAMTS-1 that inhibits metastasis comprises SEQ ID NO: 5, 7, 9, and/or 11. In some embodiments, the fragment or mutant of ADAMTS-1 that inhibits metastasis, cell growth or proliferation and/or angiogenesis comprises SEQ ID NO: 5, 7, 9, 11, 17, 19, 21, 23, 25, 27, 29, 31, 33, and/or 35.

In some embodiments, a method of treating cancer can refer to a method of inhibiting cell growth, division, inducing cell death (e.g. apoptosis and/or necrosis), promoting metastasis and angiogenesis, or combinations thereof.

The fragments or mutants of the present invention can also be administered in the form of a nucleic acid molecule that encodes for the fragments or for the mutants. In some embodiments, the nucleic acid molecule comprises SEQ ID NO: 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, 34, and/or 36.

The present invention also provides antibodies or fragments of antibodies that can specifically bind to and block the pro-cancer activity of ADAMTS-1. In some embodiments, the antibody specifically binds to ADAMTS-1 comprising SEQ ID NO: 1, 2, 3, or 4

As used herein, the term “specifically binds to” in reference to an antibody refers to an antibody that will bind to one peptide or protein with higher affinity than another peptide. In some embodiments, the antibody that specifically binds to a peptide or polypeptide will not bind to more than one peptide. In some embodiments, the specific antibody binds with a Kd that is 10×, 100×, 1000× greater to one peptide over another. Methods of making and identifying specific antibodies are routine.

The present invention also provides for antibodies that binds to full-length ADAMTS-1 to inhibit cell proliferation, division, growth, or metastasis. In some embodiments, the polypeptide comprises SEQ ID NO: 1, 2, 3, or 4.

The present invention also provides methods of inducing the cleavage of ADAMTS-1 in a cell comprising contacting the cell with a heparin inhibitor. Examples of heparin inhibitors include, but are not limited to heparinase, platelet factor 4 (PF4-a), protamine, polybrene, and the like.

The present invention also provides for methods for identifying an inhibitor or an activator of ADAMTS-1 cleavage comprising performing a test assay comprising contacting ADAMTS-1 with a test compound; and measuring the cleavage of ADAMTS-1, wherein a decrease in cleavage indicates that the test compound is a cleavage inhibitor or wherein an increase in cleavage indicates that the test compound is a cleavage activator. In some embodiments, the effect of the test compound is compared what occurs in the absence of any test compound. In some embodiments, the compound is contacted with ADAMTS-1 under conditions in which ADAMTS-1 is cleaved. In some embodiments, ADAMTS-1 undergoes auto-cleavage (e.g. where the enzyme cleaves itself). In some embodiments, the method comprising contacting a test compound with ADAMTS-1 under conditions where ADAMTS-1 can be cleaved. These conditions can be any conditions and can be modified such that ADAMTS-1 is able to be cleaved either by itself (auto-cleavage) or by another molecule. Conditions that can be modified include, but are not limited to, pH, ion concentration, metal concentration, and the like.

In some embodiments the methods comprise contacting more than one test compound, in parallel. In some embodiments, the methods comprises contacting 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 40, 50, 100, 1000, at least 2, at least 5, at least 10, at least 50, at least 100, or at least 1000 test compounds in parallel. In some embodiments, the present invention is used in High Throughput Screening of compounds and complete combinatorial libraries can be assayed, e.g., up to thousands of compounds. Methods of how to perform high throughput screenings are well known in the art. The methods can also be automated such that a robot can perform the experiments. The present invention can be adapted for the screening of large numbers of compounds, such as combinatorial libraries of compounds. Indeed, compositions and methods allowing efficient and simple screening of several compounds in short periods of time are provided. The instant methods can be partially or completely automated, thereby allowing efficient and simultaneous screening of large sets of compounds.

In some embodiments, the method of the present invention comprises the step of contacting a cell expressing v5-epitope tagged ADAMTS-1 (such as TA3ADAMTS-1) in the presence of a test compound. The cells can be observed to determine if the test compound inhibits or promotes the cleavage of ADAMTS-1. A control may be provided in which the cell is not contacted with a test compound. A further control may be provided in which test compound is contacted with cells that either do not express ADAMTS-1 or in which ADAMTS-1 cannot be cleaved (the cleavage-resistant ADAMTS-1 mutant). If the cells contacted with the test compound increase the cleavage of ADAMTS-1 then pro-cleavage activity is indicated for the test compound. If the cells contacted with the test compound decrease the cleavage of ADAMTS-1 then anti-cleavage activity is indicated for the test compound.

Positive and negative controls may be performed in which known amounts of test compound and no compound, respectively, are added to the assay. One skilled in the art would have the necessary knowledge to perform the appropriate controls.

The test compound can be any product in isolated form or in mixture with any other material (e.g., any other product(s)). The compound may be defined in terms of structure and/or composition, or it may be undefined. For instance, the compound may be an isolated and structurally-defined product, an isolated product of unknown structure, a mixture of several known and characterized products or an undefined composition comprising one or several products. Examples of such undefined compositions include for instance tissue samples, biological fluids, cell supernatants, vegetal preparations, etc. The test compound may be any organic or inorganic product, including a polypeptide (or a protein or peptide), a nucleic acid, a lipid, a polysaccharide, a chemical product, or any mixture or derivatives thereof. The compounds may be of natural origin or synthetic origin, including libraries of compounds.

In some embodiments, the activity of the test compound(s) is unknown, and the method of this invention is used to identify compounds exhibiting the selected property (e.g., ADAMTS-1 cleavage). However, in some embodiments instances where the activity (or type of activity) of the test compound(s) is known or expected, the method can be used to further characterize the activity (in terms of specificity, efficacy, etc.) and/or to optimize the activity, by assaying derivatives of the test compounds.

The amount (or concentration) of test compound can be adjusted by the user, depending on the type of compound (its toxicity, cell penetration capacity, etc.), the number of cells, the length of incubation period, the amount of ADAMTS-1, etc. In some embodiments, the compound can be contacted in the presence of an agent that facilitates penetration or contact with a cell comprising ADAMTS-1. The test compound is provided, in some embodiments, in solution. Serial dilutions of test compounds may be used in a series of assays. In some embodiments, test compound(s) may be added at concentrations from 0.01 μM to 1M. In some embodiments, a range of final concentrations of a test compound is from 10 μM to 100 μM. One such test compound that is effective to activate cleavage of ADAMTS-1 in a cell is a heparin inhibitor.

In some embodiments, the method comprises measuring ADAMTS-1 cleavage in the presence of the test compound. If the test compound is found to cleave ADAMTS-1 it is indicative that the test compound is pro-cleavage ADAMTS-1 agent. Since the cleavage fragments of ADAMTS-1 agent, a pro-cleavage fragment can also be considered an anti-cancer agent.

ADAMTS-1 cleavage can be measured by any means that demonstrates that the cleavage of ADAMTS-1 has been modulated (increased or decreased) in the presence of the test compound. Examples of how to measure ADAMTS-1 cleavage include measuring an increase or decrease in the cleavage fragments of ADAMTS-1. The cleavage fragments can be measured by any means including, but not limited to, Western Blot, ELISA, Sandwich Assay, and the like. Methods of measuring the levels protein cleavage fragments are routine to one of ordinary skill in the art.

In some embodiments, the test compound activates the cleavage of ADAMTS-1 by at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 100%, at least 200%. In some embodiments, the test compound inhibits the cleavage of ADAMTS-1 by at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, or at least 99%. In some embodiments, the cleavage of ADAMTS-1 is compared to cleavage of ADAMTS-1 observed in the absence of the test compound.

In some embodiments, the methods further comprise performing a control assay. In some embodiments, the control assay comprising contacting a cell with a negative or positive control and measuring, including, but not limited to, ADAMTS-1 cleavage. In some embodiments, the control compound is compared to the test compound. In some embodiments, the control compound is a negative control (e.g. a compound that does not inhibit or activate ADAMTS-1 cleavage). A negative control can also be the absence of a test compound or the vehicle (e.g. solvent) that the test compound is contacted with the cell. In some embodiments, the control compound is a positive control (e.g. a compound that inhibits or activates ADAMTS-1 cleavage).

In some embodiments, the test compound is a small molecule, a peptide (including the peptides from the heparin-binding proteins and HSPGs), an antibody, a cellular fraction, a protease, or a mixture thereof. As discussed above, the test compound can be contacted with a cell comprising ADAMTS-1, but the test compound can be contacted with ADAMTS-1. For example, ADAMTS-1 can be expressed as a protein and either be purified or not be purified, but is isolated from a cell. For the purposes of the screening assays to identify test compounds that can inhibit or activate the cleavage of ADAMTS-1, an isolated protein is a protein that is separated from a cell. The protein can be purified from other components in the cell, but it does not have to be. In some embodiments, an isolated ADAMTS-1 protein results from a cell being lysed which releases all the contents of the cell. The cleavage of ADAMTS-1 can then be measured or monitored in a non-cellular environment. The test compound is then contacted with ADAMTS-1 to determine if the test compound can inhibit or activate the cleavage of ADAMTS-1.

In some embodiments, the methods further comprise performing a negative control assay which comprises contacting a cell that does not comprise ADAMNTS-1 or a cell that comprises a cleavage resistant mutant of ADAMTS-1. In some embodiments, the negative control assay comprises contacting an isolated cleavage resistant mutant of ADAMTS-1.

The present invention also provides methods for identifying an anti-cancer agent comprising performing a test assay comprising contacting a cell comprising ADAMTS-1 with a test compound; and measuring the cleavage of ADAMTS-1, wherein an increase in cleavage indicates that the test compound is an anti-cancer compound. In some embodiments, the cleavage in the presence of the test compound is compared to an assay where the cell is comprising ADAMTS-1 is not contacted with the test compound.

As used herein, “a cell comprising ADAMTS-1” refers to a cell expressing the protein ADAMTS-1. The cell can be either be expressing the protein endogenously (e.g. from within its native genome) or exogenously. An exogenously expressed protein is a protein in a cell that would not normally be present except for some modification. The exogenously expressed protein can be, for example, transfected into a cell either stably or transiently.

The present invention also provides methods of inhibiting angiogenesis in an individual comprising administering to the individual a fragment of ADAMTS-1. In some embodiments, the fragment of ADAMTS-1 comprises ADAMTS-1CTCF or ADAMTS-1NTCF (SEQ ID NOs: 5, 7, 9 and/or 11). In some embodiments a nucleic acid molecule encoding the fragments is administered. In some embodiments, the nucleic acid molecule comprises SEQ ID NOs: 6, 8, 10, and/or 12.

The present invention provides methods of inhibiting the growth or metastasis of a tumor. In some embodiments, the tumor is vascularized or non-vascularized.

The present invention also provides methods of treating cancer comprising inhibiting the metalloproteinase activity of ADAMTS-1. In some embodiments, the metalloproteinase activity of ADAMTS-1 is inhibited by administering a metalloproteinase defective full-length ADAMTS-1 or the ADAMTS-1 fragments containing its substrate-binding domain such as ADAMTS-1spacer/Cys-rich or ADAMTS-1spacer, which can act as the dominant negative mutants of ADAMTS-1 and inhibit the activity of the wild-type protein. In some embodiments, the metalloproteinase defective ADAMTS-1 comprises SEQ ID NO: 29, 31, 33, and/or 35. In some embodiments, the metalloproteinase activity is inhibited by an antibody or a small molecule that binds to ADAMTS-1. In some embodiments, the metalloproteinase activity is inhibited by an antibody or a small molecule that binds to the metalloproteinase active site of ADAMTS-1.

The present invention also provides methods of identifying inhibitors of ADAMTS-1 metalloproteinase activity comprising contacting a fragment of or full-length ADAMTS-1 that has metalloproteinase activity with a test compound and determining if the metalloproteinase activity is inhibited. (In some embodiments, the fragment of ADAMTS-1 comprises SEQ ID NO: 5, 7, 9, and/or 11.) In some embodiments, the activity in the presence of the test compound is compared to the activity in the absence of the test compound. In some embodiments, the method comprises comparing the activity with a positive control assay and/or a negative control assay. In some embodiments, the method comprises comparing the activity of the fragment to a fragment that is defective in metalloproteinase activity. A fragment can be defective in metalloproteinase because of a mutation, substitution, deletion, or insertion. In some embodiments, the fragment is defective in metalloproteinase activity due to a Glu to Gln mutation. In some embodiments, the fragment that lacks metalloproteinase activity comprises SEQ ID NO: 33 and/or 35. In some embodiments, the fragment that lacks metalloproteinase activity is encoded by a nucleic acid molecule comprising SEQ ID NO: 34 and/or 36.

Methods of measuring metalloproteinase activity (e.g. ADAMTS-1 activity) are routine. For example, the cleavage of substrates of ADAMTS-1 can be measured and compared in the absence and presence of a test compound. However, any method or means can be used to measure metalloproteinase activity of ADAMTS-1. Substrates of ADAMTS-1 are known in the art and can be measured. In some embodiments, the substrate of the metalloproteinase is aggrecan or versican.

In some embodiments, the present invention provides methods of treating cancer comprising administering to an individual a compound that is a ADAMTS-1 metalloproteinase activity inhibitor. In some embodiments, the inhibitor is a dominant negative mutant of ADAMTS-1. In some embodiments, the inhibitor is a polypeptide or comprising SEQ ID NO: 33 and/or 35. In some embodiments, the inhibitor is encoded by a nucleic acid molecule comprising SEQ ID NO: 34 and/or 36.

Other fragments or mutants of ADAMTS-1 can also be used to treat cancer because they also act as a dominant negative regulator of ADAMTS-1 and, thus, be able to inhibit the function of ADAMTS-1. Accordingly, the present invention provides methods of treating cancer comprising administering a therapeutically effective amount of a composition comprising a fragment of ADAMTS-1 comprising the spacer/Cys-rich and/or Spacer domain of ADAMTS-1.

The present invention also provides polypeptide fragments of ADAMTS-1 comprising the spacer/Cys-rich and/or spacer domain of ADAMTS-1. In some embodiments, the fragment comprises SEQ ID NO: 17, 19, 21, and/or 23. In some embodiments, the fragments are encoded by nucleic acid molecules comprising 18, 20, 22, and/or 24.

The present invention also provides for fragments of ADAMTS-1 that bind to the extracellular matrix (ECM). According, in some embodiments, the present invention provides an ECM binding fragment of ADAMTS-1. An “ECM binding fragment of ADAMTS-1” is a fragment of ADAMTS-1 that binds to the ECM. In some embodiments, the ECM binding fragment of ADAMTS-1 comprises SEQ ID NO: 17, 19, 21, and/or 23. In some embodiments, a nucleic acid molecule encodes for an ECM binding fragment of ADAMTS-1. In some embodiments, the ECM binding fragment comprises SEQ ID NO: 18, 20, 22, and/or 24.

In some embodiments, the present invention provides nucleic acid molecules encoding any fragment of ADAMTS-1 described herein. In some embodiments, the nucleic acid molecule comprises SEQ ID NOs: 2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 16, 28, 30, 32, 34, 36, or combinations thereof.

In some embodiments, the present inventions provides polypeptides comprising at least a fragment of ADAMTS-1 as described herein. In some embodiments, the polypeptides comprise SEQ ID NO: 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 33, 35, or combinations thereof. In some embodiments, the present invention provides polypeptides that comprise mutations that inactivate the metalloproteinase activity of ADAMTS-1. In some embodiments, the mutation is a Glu to Gln mutation that corresponds to position 386 (mouse) (or 385 in human) of the wild-type ADAMTS-1. In some embodiments, the mutant ADAMTS-1 comprises SEQ ID NO: 29 and/or 31. In some embodiments a nucleic acid molecule encoding such mutants is provided. In some embodiments, the nucleic acid molecule comprises SEQ ID NO: 30 and/or 32.

The present invention also provides methods for identifying a compound that induces the cleavage of ADAMTS-1 comprising performing a test assay comprising identifying a compound that inhibits heparin, wherein if a compound inhibits heparin, the compound would be said to induce the cleavage of ADAMTS-1. Since heparin inhibitors induce the cleavage of ADAMTS-1 heparin inhibitors would be able to activate the cleavage of ADAMTS-1. Thus, a compound that is identified as heparin inhibitor would be said to be able to induce the cleavage of ADAMTS-1. In some embodiments, the effect of heparin is a protective effect.

The present invention also provides methods of identifying a heparin inhibitor comprising contacting heparin and ADAMTS-1 with a test compound under conditions that ADAMTS-1 is cleaved in the absence of heparin and determining if the test compound inhibits heparin. As described herein, heparin inhibits the cleavage of ADAMTS-1. Therefore, a test compound that inhibits heparin will allow ADAMTS-1 to be cleaved by another protein or by itself. A test compound is said to be a heparin inhibitor if ADAMTS-1 is cleaved in the presence of heparin. In some embodiments, the heparin and ADAMTS-1 are free of cellular proteins. In some embodiments, the heparin and ADAMTS-1 is free of extracellular matrix.

A fragment of ADAMTS-1, a nucleic acid molecule encoding a fragment of ADAMTS-1, a compound that inhibits or activates the cleavage of ADAMTS-1 can be administered by any means to the individual whether in the form of a composition or a pharmaceutical composition. Methods of administration are known to one of skill in the art. For example, the agent can be prepared as a pharmaceutical composition. In some embodiments, the pharmaceutical composition comprises a pharmaceutically acceptable carrier or diluent. In some embodiments, the pharmaceutical compositions are sterile and/or pyrogen free. The pharmaceutical composition comprising the molecule and a pharmaceutically acceptable carrier or diluent may be formulated by one having ordinary skill in the art with compositions selected depending upon the chosen mode of administration. Suitable pharmaceutical carriers are described in the most recent edition of Remington's Pharmaceutical Sciences, A. Osol, a standard reference text in this field.

For parenteral administration, the composition can be, for example, formulated as a solution, suspension, emulsion or lyophilized powder in association with a pharmaceutically acceptable parenteral vehicle. Examples of such vehicles are water, saline, Ringer's solution, dextrose solution, and 5% human serum albumin. Liposomes and nonaqueous vehicles such as fixed oils may also be used. The vehicle or lyophilized powder may contain additives that maintain isotonicity (e.g., sodium chloride, mannitol) and chemical stability (e.g., buffers and preservatives). The formulation is sterilized by commonly used techniques. For example, a parenteral composition suitable for administration by injection is prepared by dissolving 1.5% by weight of active ingredient in 0.9% sodium chloride solution.

The pharmaceutical compositions according to the present invention may be administered as a single doses or in multiple doses. The pharmaceutical compositions of the present invention may be administered either as individual therapeutic agents or in combination with other therapeutic agents. The treatments of the present invention may be combined with conventional therapies, which may be administered sequentially or simultaneously.

The pharmaceutical compositions may be administered by any means that enables the agent to reach the agent's site of action in the body of a mammal. Because the compositions may be subject to being digested when administered orally, parenteral administration, i.e., intravenous, subcutaneous, intramuscular, would ordinarily be used to optimize absorption. In addition, the pharmaceutical compositions of the present invention may be injected at a site at or near hyperproliferative growth. For example, administration may be by direct injection into a solid tumor mass or in the tissue directly adjacent thereto. The composition may also be formulated with a pharmaceutically acceptable topical carrier and the formulation may be administered topically as a creme, lotion or ointment for example.

The dosage administered varies depending upon factors such as: pharmacodynamic characteristics; its mode and route of administration; age, health, and weight of the recipient; nature and extent of symptoms; kind of concurrent treatment; and frequency of treatment. Usually, a daily dosage of a composition to treat cancer is used in an amount effect to have an anti-cancer effect. In some embodiments, the daily dosage is used in an amount to cleave ADAMTS-1 into a fragment that can inhibit cell proliferation or cell growth (e.g tumor growth). In some embodiments, the dosage can be about 1 lag to 100 milligrams per kilogram of body weight. Ordinarily 0.5 to 50, and preferably 1 to 10 milligrams per kilogram per day given in divided doses 1 to 6 times a day or in sustained release form is effective to obtain desired results.

EXAMPLES

The invention is now described with reference to the following examples. These examples are provided for the purpose of illustration only and the invention should in no way be construed as being limited to these examples, but rather should be construed to encompass any and all variations which become evident as a result of the teaching provided herein. Those of skill in the art will readily recognize a variety of non-critical parameters that could be changed or modified to yield essentially similar results.

Example 1 Materials and Methods

Cell Lines and Reagents

Human umbilical vein endothelial cells (HUVECs) were obtained from Cambrex (Walkersville, Md.). TA3 transfectants were maintained as described previously (11, 12). Anti-v5 epitope (Invitrogen), -vWF (Dako), -phosphorylated tyrosine (BD Transduction Lab), -EGFR, -ErbB-2, -Erk1/2, and -phospho-Erk1/2 (Santa Cruz) antibodies, and Brdu-cell proliferation kit (Roche) and Apoptag kit (Chemicon) were used in the experiments.

Reverse Transcriptase-Polymerase Chain Reaction (RT-PCR), Mutagenesis, and Expression Constructions

Expression of ADAMTS-1 was assessed by RT-PCR as described (13). Full-length mouse ADAMTS-1 was obtained by RT-PCR with a primer pair consisting of 24 nucleotides corresponding to the 3′ or 5′ extremity of the coding sequence of ADAMTS-1 (accession number NM009621). The stop codon was omitted from the reverse primers to fuse ADAMTS-1 to the C-terminal v5 epitope tag existed in the expression vector (pEF6/v5-HisTOPO, Invitrogen). Various mutation and deletion of ADAMTS-1 were generated as detailed in FIG. 1A using the QuikChange™ site and ExSite PCR-based site-directed mutagenesis kits (Stratagene).

Transfection

Lipofectamine (Invitrogen) was used to transfect TA3wt1 cells with empty expression vector alone or the expression constructs containing cDNA inserts encoding ADAMTS-1 and various mutant or fragments of ADAMTS-1 (FIG. 1A). TA3 transfectants were selected and the expression level of v5-tagged full-length and fragments of ADAMTS-1 was determined by Western blotting with anti-v5 antibody (Invitrogen).

ADAMTS-1 Production and Purification, Proteolytic Cleavage Assay, and Western Blot Analysis

Cell culture supernatants derived from Cos-7 and TA3 transfectants expressing v5-epitope tagged wild type ADAMTS-1 or ADAMTS-1 mutants (FIG. 1A) were collected and purified through Ni+-Probond (Invitrogen) and anti-v5 antibody conjugated affinity columns (Sigma). Auto-proteolytic cleavage capacity of ADAMTS-1 was assessed by in vitro proteolytic cleavage assay using purified ADAMTS-1. In this assay, 100 ng of ADAMTS-1 was incubated in 50 mM Tris-acetate buffer (pH 7.3) containing 5 mM CaCl2 and 0.1M NaCl at 37° C. for 30 min, 1, 2, 4, 8 and 12 hours, and reaction was stopped by addition of 8×SDS sample buffer. The reaction products were analyzed by Western blot with anti-v5 mAb.

To assess ADAMTS-1 cleavage in cellular context and to determine how the cleavage is regulated, Cos-7 or TA3 transfectants expressing ADAMTS-1 or ADAMTS-1E/Q was cultured for 48 hours in the absence or presence of different reagents as detailed in the figure legend, and the cell culture supernatants were collected and analyzed by Western blot with anti-v5 antibody.

Tumor Cell Tracking and Pulmonary Metastasis

To track TA3 transfectants during early pulmonary metastasis, the TA3 transfectants were labeled with green 5-chloromethyl-fluorescein diacetate (CMFDA, Molecular Probes, Inc.) as described (13), and the CMFDA-labeled TA3 transfectants (1×106 cells/mouse) were injected into the tail vein of A/Jax syngenic mice (the Jackson Lab). The mice were sacrificed 24 hours after the injection, and lungs were removed, fixed, and sectioned. The localization of tumor cells in mouse lung parenchyma were revealed under fluorescence microscope, and the extent of tumor cell extravasation was determine by counting number of the tumor cells in five randomly selected 10× microscopic fields.

Experimental pulmonary metastasis was carried out as detailed previously (13), and five independent clonal TA3 transfectants expressing ADAMTS-1, ADAMTS-1CTCF, ADAMTS-1NTCF or ADAMTS-1minusTSP, or transfected with the empty expression vector were used. For each type of the experiment, six mice were injected with each clonal transfectant and two independent experiments were performed. The experimental mice were observed daily after the i.v. injection and duration of mouse survival was recorded. The survival rate of these mice was calculated as the following: survival rate (%)=(number of mice are still alive/total number of the experimental mice)×100%. The mice that are free of symptom 60 days after the i.v. injection were sacrificed and their lungs were examined. In the second set of experiments, 11 and 18 days after i.v. injection, pulmonary metastatic burden was assessed by measuring weight of the mouse lungs.

Histology and Immunohistochemistry

To determine the tumor cell proliferation rate in vivo, 5-Bromo-2′-deoxy-uridine (Brdu) was injected into mice four hours prior to sacrifice of the experimental mice. The mouse lungs were fixed, sectioned, and stained with H&E as described (11). In addition, the sections were reacted with anti-von Willebrand factor (vWF) antibody to assess tumor angiogenesis, with anti-Brdu antibody to detect proliferating cells or with Apoptag kit to detect apoptotic cells in situ. Total number of the tumor cells and number of the tumor cells that are positive for anti-Brdu antibody or TUNEL-staining were counted in five randomly selected 400× microscopic fields within the pulmonary macro- and micro-metastases. More than 2,000 cells were counted in total for each type of transfectants. The proliferation and apoptosis rate was calculated as the following: proliferation or apoptosis rate=(number of the anti-Brdu or TUNEL-positive cells per microscopic field/total number of the tumor cells per microscopic field)×100%. To determine blood vessel number, the vWF-positive blood vessels were countered in six randomly selected 200× microscopic fields within macro- or micro-metastases. The number of blood vessels/microscopic field was expressed as means+/−S.D.

EGFR and ErbB-2 Phosphorylation

RIPA buffer (50 mM Tris-HCl, PH 7.4, 50 mM NaCl, 1% Triton-X100, 2 mM EDTA, 2 mM sodium orthovanadate, 2 mM sodium fluoride, 2 mM phenylmethylsulfonyl fluoride, 1 mM Leupeptin, 1 mM Pepstain A, and 10 μg/ml aprotinin) was used to extract the lung tissues derived from the mice that were injected with or without different TA3 transfectants (1×106/mouse) intravenously 24 hours prior. The proteins were used in the immunoprecipitation reactions to pull-down EGFR and ErbB-2 using the agarose beads conjugated with anti-EGFR or anti-ErbB-2 antibody (Santa Cruz). The precipitated proteins were analyzed by Western blotting with anti-phosphotyrosine antibody (BD Bioscience) to detect phosphor-EGFR and phosphor-ErbB-2, or with anti-EGFR or anti-ErbB-2 antibody (Santa Cruz) to detect total amount of EGFR or ErbB-2, respectively.

Shedding of the EGF Family GFs and Activation of Erk1/2 Kinases

Shedding of the transmembrane precursors of AR, HB-EGF, and epigen by ADAMTS-1, its mutant and fragments were assessed by co-transfection of Cos-7 cells with the expression constructs containing cDNA inserts that encode these EGF family precursors and various ADAMTS-1 constructs as detailed in the figure legend. The concentrated cell culture supernatants of the co-transfected COS-7 cells were analyzed by Western blotting to detect the soluble GFs using the corresponding antibodies (R&D Systems).

Ability of the ADAMTS-1 fragments to inhibit activation of Erk1/2 kinase induced by soluble AR (5 ng/ml) and HB-EGF (4 ng/ml) were assessed by applying the serum starved MCF-10A cells with purified soluble AR or HB-EGF in the absence or presence of their corresponding neutralization antibodies or purified full-length ADAMTS-1 or the ADAMTS-1 fragments (400 ng). MCF-10A cells were then lysed and equal amount of the proteins were analyzed by Western blotting with anti-phospho-Erk1/2 to detect phosphor-Erk1/2 or with anti-Erk antibody to detect total amount of Erk1/2 protein.

HUVECs were cultured until subconfluence and switched to serum-free medium (SFM) for overnight. VEGF165 (10 ng), bFGF (10 ng), AR (5 ng), and HB-EGF (4 ng) were applied to the serum-starved HUVECs in the absence or presence of 400 ng of ADAMTS-1, ADAMTS-1minusTSP, ADAMTS-1NTCF, or ADAMTS-1CTCF for 20 minutes. The HUVECs were lysed and equal amount of the proteins were subjected to Western blotting with anti-phospho-Erk1/2 or anti-Erk (Santa Cruz) to detect phosphor-Erk1/2 or total amount of Erk, respectively.

Example 2 ADAMTS-1 Undergoes Auto-Proteolytic Cleavage and the Self-Cleavage of ADAMTS-1 is Regulated

Previous results have shown that ADAMTS-1 is cleaved within the spacer region and several matrix metalloproteinases (MMPs) are responsible for the cleavage (9). Since several other members of ADAMTS family undergo auto-proteolytic cleavage and ADAMTS-1 is an active metalloproteinase, the possibility that the cleavage of ADAMTS-1 can be mediated by its own metalloproteinase activity was investigated. To achieve that, a protease-dead mutant of ADAMTS-1 was generated by switching E386 to Q (ADAMTS-1E/Q) in the Zinc-binding pocket of the metalloproteinase domain. The expression constructs containing v5-epitope tagged wild type ADAMTS-1 or ADAMTS-1E/Q were used to transfect Cos-7 cells. The cell culture supernatants of the transiently transfected Cos-7 cells were analyzed and the results showed that only wild type ADAMTS-1 but not ADAMTS-1E/Q is cleaved to generate the C-terminal cleavage fragments (FIG. 1B, arrows), suggesting that the metalloproteinase activity of ADAMTS-1 is required for the cleavage.

In order to produce full-length ADMATS-1, the regulation of ADAMTS-1 cleavage was investigated. Different reagents were applied to a stable TA3 transfectant expressing ADAMTS-1, and the cell culture supernatants were analyzed 48 hours late. The result showed that heparin and heparan sulfate (HS) completely block the proteolytic cleavage of ADAMTS-1 (FIG. 1C), while the control glycosaminoglycans (GAGs), chondroitin sulfate (CS) and hyaluronan (HA), and displayed no effect on the cleavage. This result suggests that auto-proteolytic cleavage of ADAMTS-1 is regulated by synthesis and degradation rate of HS/heparan sulfate proteoglycans (HSPGs) in the microenvironment where ADAMTS-1 is produced and HS/HSPGs likely play important role in regulating ADAMTS-1 function.

Full-length ADAMTS-1 protein was produced by Cos-7 cells transfected with the expression construct containing ADAMTS-1 cDNA in the presence of heparin. Cell culture media of the transfected Cos-7 cells were collected and purified through the affinity columns. Purified ADAMTS-1 was used in a proteolytic cleavage assay and the result showed that ADAMTS-1 was auto-proteolytically cleaved to release v5-epitope tagged C-terminal cleavage fragments that have molecular weight similar to that generated in the cell culture condition (FIG. 1E).

ADAMTS-1 Promotes Metastasis, while ADAMTS-1NTCF and ADAMTS-1CTCF Block the Process

ADAMTS-1 was found to inhibit bFGF-induced vascularization in the cornea pocket assay and VEGF-induced angiogenesis in the chorioallantoic membrane assay and tumor growth in vivo. However, a study analyzing clinical pancreatic cancer samples demonstrated that increased expression of ADAMTS-1 is correlated to enhanced metastatic potential and worse prognosis, implying that ADAMTS-1 facilitates tumor metastasis. In addition, studies have shown that ADAMTS-1 is one of the genes up-regulated in the breast cancer with elevated metastatic activity. To determine the exact roles of ADAMTS-1 and its cleavage fragments in tumor metastasis and the underlying mechanism, set to investigate how overexpression of full-length and the fragments of ADAMTS-1 affects metastasis of TA3 mammary carcinoma (TA3) cells. As shown in FIGS. 1B and C, the C-terminal cleavage fragments of ADAMTS-1 are heterogenic in their molecular weight, suggesting that ADAMTS-1 are cleaved at more than one sites within the spacer/Cys-rich region (FIG. 1A, arrows). The molecular weight of the shortest C-terminal cleavage fragments is similar to that of the expressed C-terminal fragment of ADAMTS-1 containing the last two TSP-1 type I motifs (ADAMTS-1CTCF: amino acids 842-951, FIG. 1A, D), suggesting that in addition to the previous identified cleavage site in the spacer region (FIG. 1A, the bigger arrow), there is at least one additional cleavage site at the junction between spacer region and the second TSP-1 type I motif (FIG. 1A, the smaller arrow).

It was difficult to express the N-terminal fragments of ADAMTS-1 containing different parts of the spacer and/or Cys-rich domains (data not shown). In addition, studies have shown that auto-proteolytic cleavage of ADAMTS-4 occurs at the multiple sites within its spacer/Cyr-rich region, and the shortest N-terminal cleavage fragment of ADAMTS-4 is generated by cleavage around the junction between the Cys-rich domain and the TSP-1 type I motif. Thus, two expression constructs containing N-terminal fragments of ADAMTS-1 were made, which expressed well in TA3 cells. These constructs contain the N-terminal domains of ADAMTS-1 until the end of the first TSP-1 type I motif (ADAMTS-1NTCF, amino acids 1-596, FIG. 1A, D) or until the end of the disintegrin domain (ADAMTS-1minusTSP, amino acids 1-545, FIG. 1A, D). ADAMTS-1NTCF likely represents the shortest N-terminal cleavage fragment of ADAMTS-1.

In order to assess the effects of ADAMTS-1 and its fragments on tumor metastasis reliably, the heterogeneity of TA3 cells was eliminated by transfecting the cells with empty expression vector containing neomycin-resistant gene. A clonal TA3 cell (TA3wt1) that undergoes aggressive pulmonary metastasis after intravenous (i.v.) injection was selected (data not shown). Our RT-PCR result showed that like its wild type counterpart, TA3wt1 cells express ADAMTS-1 endogenously (data not shown). TA3wt1 was used to transfect several expression constructs that contain blasticidin-resistant gene and different ADAMTS-1 cDNA inserts (FIG. 1A). Five independent clonal TA3 transfectants that were transfected with the empty expression vector alone (TA3wtb) or expressing the following gene products (FIG. 1D) were identified and used in pulmonary metastasis experiments: wild type ADAMTS-1 (TA3ADAMTS-1), ADAMTS-1NTCF (TA3ADAMTS-1NTCF), ADAMTS-1CTCF (TA3ADAMTS-1CTCF), and ADAMTS-1minusTSP (TA3ADAMTS-1minusTSP). These TA3 transfectants displayed similar growth rate in cell culture condition (data not shown).

Our results showed that overexpression of ADAMTS-1 significantly accelerated pulmonary metastasis and shortened the survival time of the mice (FIG. 2A-C). On the contrary, ADAMTS-1NTCF or ADAMTS-1CTCF, but not ADAMTS-1minusTSP blocks pulmonary metastasis of the TA3 transfectants (FIG. 2A-C), suggesting that the inhibitory effect of these ADAMTS-1 fragments is likely derived from the TSP type I motifs which exist in ADAMTS-1NTCF and ADAMTS-1CTCF, but not in ADAMTS-1minusTSP; and the anti-tumor activity is likely masked in full-length ADAMTS-1.

The metastatic tumors derived from TA3wtb, TA3ADAMTS-1, and TA3ADAMTS-1minusTSP cells are invasive and fused together (FIGS. 2A, D, a-b, and data not shown), which made it difficult to determine accurate number of the metastatic lesions. Thus, metastatic burden of the experimental mice was quantified by average weight of the experimental mouse lungs (FIG. 2C). Because there is a significant difference in survival time of these mice and the mice usually succumb to pulmonary metastasis when metastatic burden causes the lung weight to reach 1-1.2 grams, the metastatic burden of the remaining survival mice at day 11 and day 18 after i.v. injection of the TA3 transfectants was measured. At least fifteen experimental mouse lungs were measured for each type of the transfectants at each time point. Our results showed that overexpression of ADAMTS-1 accelerated time that is need to reach the maximal metastatic burden and shortened the survival time of the mice, while overexpression of ADAMTS-1NTCF or ADAMTS-1CTCF blocked pulmonary metastasis and render most of the experimental mice free of metastatic disease (FIG. 2B-C).

Histologic analysis of the lung sections showed that TA3wtb, TA3ADAMTS-1, and TA3ADAMTS-1minusTSP cells are invasive and fill up the pulmonary space (FIG. 2D-a-b, and data not shown). On the contrary, only micrometastasis were detected in the lungs received TA3ADAMTS-1NTCF or TA3ADAMTS-1CTCF cells (FIG. 2D-c, d, arrows). To assess whether ADAMTS-1 expressed by the transfected TA3 cells is cleaved in vivo, different pulmonary tumors derived from TA3ADAMTS-1 cells were lysed and the proteins were analyzed by Western blotting with anti-v5 antibody, which recognizes the v5-tagged ADAMTS-1. The result showed that ADAMTS-1 protein is maintained in full-length form in vivo and no cleavage fragments of ADAMTS-1 were detected (FIG. 2E). This result suggests that proteolytic cleavage of ADAMTS-1 regulates ADAMTS-1 function and the cleavage status of ADAMTS-1 in vivo determine its effect (stimulatory or inhibitory) on tumor metastasis.

ADAMTS-1NTCF and ADAMTS-1CTCF Blocks Pulmonary Metastasis by Inhibiting Proliferation and Inducing Apoptosis of Tumor Cells, and by Repressing Tumor Angiogenesis

To determine the mechanism underlying the pro-tumor effect of full-length of ADAMTS-1 and the anti-tumor effect of ADAMTS-1NTCF and ADAMTS-1CTCF, proliferation and apoptosis rates of the tumor cells and the extent of tumor angiogenesis during pulmonary metastasis of TA3 transfectants were analyzed. A 5-Bromo-2′-deoxy-uridine (Brdu) incorporation assay and in situ detection of apoptotic cells on the sections derived from the experimental mouse lungs (six days after i.v. injection of TA3 transfectants) was performed. Results demonstrated that expression of ADAMTS-1NTCF or ADAMTS-1CTCF, but not that of ADAMTS-1minusTSP, inhibits proliferation and promotes apoptosis of the tumor cells, and inhibits tumor angiogenesis; while overexpression of full-length exogenous ADAMTS-1 on the top of endogenous ADAMTS-1 has weak effect on tumor cell proliferation and apoptosis and promotes tumor angiogenesis in vivo (FIG. 3). These results imply that ADAMTS-1 plays a role in releasing/activating growth/survival/angiogenesis factors in the microenvironments, while ADAMTS-1NTCF and ADAMTS-1CTCF blocks/interferes activities of the factors that promote tumor cell proliferation and survival and tumor angiogenesis.

ADAMTS-1 Promotes Extravasation of the Tumor Cells and Activation of EGFR And ErbB-2 In Vivo, and Promotes Shedding of AR and HB-EGF

Activation of EGFR and ErbB-2 is known to promote proliferation and survival of breast carcinoma cells and is essential for progression of breast cancers. To determine whether ADAMTS-1 promotes activation of EGFR and/or ErbB-2 in vivo, activity of EGFR and ErbB-2 in the lungs where TA3wtb, TA3ADAMTS-1, TA3ADAMTS-1NTCF, or TA3ADAMTS-1CTCF cells were injected intravenously 24 hours prior was assessed. In order to normalizing amount of the tumor cells that were included in the protein lysates and used in the immunoprecipitation, a tumor cells tracking assay to determine the pulmonary extravasation rate of TA3 transfectants that were injected intravenously into their syngenic mice 24 hours prior was performed. The result showed that overexpression of ADAMTS-1 promotes tumor cell extravasation into lung parenchyma, while expression of TA3ADAMTS-1NTCF, or TA3ADAMTS-1CTCF inhibits the process (FIG. 4A-B).

Normal mouse lungs and the mouse lungs that received TA3 transfectants intravenously 24 hour prior were lysed, and the protein lysates that statistically contain the same amount of the tumor cells were used in immunoprecipitation to pull-down EGFR or ErbB-2 and anti-phosphotyrosine antibody was used to detect phosphor-EGFR or phosphor-ErbB-2. The result showed that overexpression of ADAMTS-1 promotes activation of EGFR and ErbB-2 (FIG. 4C). On the contrary, expression of ADAMTS-1NTCF or ADAMTS-1CTCF blocks activation of EGFR and ErbB-2 (FIG. 4C).

Whether increased activation of EGFR and ErbB-2 induced by ADAMTS-1 is achieved via shedding/activating EGF family GF precursors, the ligands of ErbB receptor tyrosine kinases which include EGFR, ErbB-2, -3, and -4. EGF family GFs include EGF, transforming growth factors-α (TGF-α), HB-EGF, AR, betacellulin, epiregulin, neuregulin, and epigen, and are shed from cell surface was investigated. Increasing amount of data suggests that shedding of the EGF family GF precursors are essential in regulating availability and bioactivity of these factors and in activation of the ErbB signaling pathways. The members of ADAM family, especially ADAM17 have been shown to play major but not sole role in shedding of these factors.

To determine whether ADAMTS-1 play an important role in constitutive shedding EGF family GFs especially the ones that bind to heparin, Cos-7 cells with several EGF family GF precursors that are expressed by TA3 cells (data not shown) including HB-EGF, AR, and epigen with were co-transfected with empty expression vector or the expression constructs containing full-length ADAMTS-1, ADAMTS-1E/Q or various ADAMTS-1 fragments. Serum-free cell culture medium (SFM) of the co-transfected Cos-7 cells were collected, concentrated and analyzed. Cos-7 cells express endogenous ADAMTS-1 (data not shown). Overexpression of exogenous ADAMTS-1 promotes shedding of AR and HB-EGF but not shedding of epigen (FIG. 4D). More importantly, overexpress ADAMTS-1E/Q which acts as a dominant negative regulator of endogenous ADAMTS-1 completely blocks the shedding of AR and inhibits the shedding of HB-EGF, while ADAMTS-1 fragments displayed no significant effect on the shedding (FIG. 4D). These data suggest that ADAMTS-1 promotes activation of EGFR and ErbB-2 by promoting shedding and activation of the EGF family GFs.

ADAMTS-1NTCF and ADAMTS-1CTCF Inhibit Activation of Erk1/2 Kinases Induced by the EGF Family GFs

Since ADAMTS-1NTCF and ADAMTS-1CTCF display no significant inhibitory effect on shedding of AR and HB-EGF, it was investigated as to whether ADAMTS-1NTCF and ADAMTS-1CTCF inhibits activation of EGFR and ErbB-2 by interfering activity of the soluble EGF family GFs. To assess that, purified soluble AR or HB-EGF was applied to MCF-10A mammary epithelial cells in the presence and absence of the naturalizing antibodies to HB-EGF or AR, ADAMTS-1NTCF, ADAMTS-1CTCF, or full-length ADAMTS-1. This result showed that the neutralizing antibodies, ADAMTS-1NTCF and ADAMTS-1CTCF, but not full-length ADAMTS-1 inhibit Erk1/2 kinase activation induced by soluble AR and HB-EGF (FIG. 5). This result suggests that ADAMTS-1NTCF or ADAMTS-1CTCF inhibits activation of EGFR and ErbB-2 by inhibiting their ligand activity likely via interfering the binding between ligands and their receptors and that the different effects of ADAMTS-1 and its cleavage fragments on availability and activity of soluble AR and HB-EGF underlie their opposite roles in tumor metastasis.

To further determine the molecular mechanism underlying the anti-angiogenic activity of ADAMTS-1NTCF and ADAMTS-1CTCF, it was investigated how these fragments affect activities of several important growth/angiogenic factors that are known to regulate angiogenesis. Bioactivity of VEGF165, basic FGF (bFGF), HB-EGF, and AR were revealed by their ability to induce activation of Erk1/2 kinases in HUVECs in the presence or absence of purified ADAMTS-1 or the ADAMTS-1 fragments. Our results showed that ADAMTS-1NTCF and ADAMTS-1CTCF but not full-length ADAMTS-1 or ADAMTS-1minusTSP block activation of Erk1/2 kinases induced by VEGF165, HB-EGF, and AR but not that induced by bFGF (FIG. 6). These results suggest that ADAMTS-1NTCF and ADAMTS-1CTCF block tumor angiogenesis by sequestering the activities of several important heparin binding factors that are essential for endothelial cells proliferation and survival.

ADAMTS-1 was found to inhibit tumor growth by blocking tumor angiogenesis; however, this study did not investigate whether the anti-tumor activity is derived from the full-length ADAMTS-1, its cleavage fragments, or both. On the contrary, increased expression of ADAMTS-1 was correlated to the increased metastatic potential in the clinic tumor samples. The current study was designed to better understand the role of full-length and the cleavage fragments of ADAMTS-1 in tumor metastasis and to elucidate the underlying mechanisms. It was demonstrated that overexpression of ADAMTS-1 promotes tumor metastasis by promoting tumor cell extravasation and tumor angiogenesis. It is well established that tumor cell extravasation is a critical step during tumor metastasis and studies have shown that ADAMTS-1 is capable of degrading aggrecan and versican. The ability of ADAMTS-1 to degrade aggrecan/versican and other not yet identified ECM components is likely responsible for the enhanced extravasation ability of TA3ADAMTS-1 cells. Furthermore, as described herein, ADAMTS-1 promotes shedding of AR and HB-EGF, which in turn promotes activation of EGFR and ErbB-2 and proliferation and survival of the tumor cells in vivo.

In the current study, it was demonstrated that ADAMTS-1 undergoes auto-proteolytic cleavage and overexpression of the cleavage fragments of ADAMTS-1 (ADAMTS-1NTCF and ADAMTS-1CTCF) block metastasis of TA3 cells by inhibiting extravasation, proliferation and survival of the tumor cells, and by repressing tumor angiogenesis via interfering activities of several important heparin binding growth/angiogenic factors. Furthermore, it was demonstrated that auto-proteolytic cleavage of ADAMTS-1 is blocked by HS, which suggests that the level of HS/HSPG in the microenvironment likely regulates which form of ADAMTS-1 (full-length or the cleavage fragments) presents predominantly in the microenvironment to exert pro- or anti-tumor activity, respectively. Thus, the roles of ADAMTS-1 and its cleavage fragments in tumor metastasis, provided the regulatory mechanism of ADAMTS-1 function (by auto-proteolytic cleavage and HS/HSPGs), and revealed the mechanisms underlying the function of ADAMTS-1 and the ADAMTS-1 cleavage fragments (by regulating availability and activity of the EGF family GFs and ErbB signaling pathway).

Shedding EGF Family GFs by ADAMTS-1

Although functional differences between mature soluble EGF family GFs and their transmembrane precursors are not well-established, the phenotype similarity between TGF-α- and ADAM17-null mice and between HB-EGF-null and HB-EGF cleavage resistant mice clearly suggested that shedding of these precursors is essential for availability and activity of these factors. Several members of ADAM family including ADAM 9, 10, 12, 17 have been implicated in shedding of HB-EGF and AR. The studies using the cells derived from ADAM-9, -10, -12, -15, and/or -17 null-mice have suggested that ADAM17 are the major but not the sole sheddase of AR and HB-EGF, and other member(s) of ADAM and/or ADAMTS family is(are) likely play important roles as well, especially in the non-PMA-induced/metalloproteinase inhibitor sensitive/constitutive shedding of these factors.

Several members of EGF family GFs including HB-EGF and AR bind to HS/HSPGs. ADAMTS-1 binds to HS as well through the spacer region and the TSP type I motifs, which brings the proteinase domain of ADAMTS-1 close to the HS/HSPG bound factors and makes ADAMTS-1 as an ideal sheddase to cleave these HS/HSPG binding GF precursors. The present disclosure has provided evidences that ADAMTS-1 promotes shedding of AR and HB-EGF and ADAMTS-1 may be a major sheddase that is responsible for constitutive shedding of AR and HB-EGF. Soluble AR and HB-EGF shed by ADADMTS-1 can in turn promote tumor cell survival and proliferation and tumor angiogenesis in vivo.

As discussed herein, it is shown that ADAMTS-1 but not the ADAMTS-1 fragments promotes shedding of AR and HB-EGF, suggesting that the intact spacer/Cys-rich domain is required for the shedding and the spacer/Cys-rich domain contains substrate recognition/binding site(s) which is(are) destroyed by the auto-proteolytic cleavage in this region. Since all the members of ADAMTS family have similar domain organization, in addition to ADAMTS-1, other members of the ADAMTS family may also involve in regulating availability and activity of HS/HSPG-binding factors.

The Anti-Tumor Activity of the ADAMTS-1 Fragments is Masked in the Full-Length Molecule.

As described herein, it is demonstrated that in contrast to the effect of full-length ADAMTS-1, the ADAMTS-1 cleavage fragments (ADAMTS-1NTCF and ADAMTS-1CTCF) block pulmonary metastasis of TA3 cells. How can auto-proteolytic cleavage convert a pro-tumor factor into anti-tumor ones? The results suggest that auto-proteolytic cleavage destroys the substrate binding domain in the spacer/Cys-rich region that is likely required for binding to AR and HB-EGF precursors. In addition, it is described herein that the N-terminal deletion fragment of ADAMTS-1 in which all the TSP type I motifs were deleted (ADAMTS-1minusTSP) displayed no anti-tumor activity, suggesting that the anti-tumor activity of ADAMTS-1NTCF and ADAMTS-1CTCF is derived from the TSP type I motif. Even though full-length ADAMTS-1 contains all three TSP type I motifs, they are likely masked and unable to exert anti-tumor activity. Auto-proteolytic cleavage of ADAMTS-1 at the spacer/Cys-rich region not only renders the N-terminal cleavage fragment (ADAMTS-1NTCF) that contains the metalloproteinase domain incapable of binding to and shedding AR and HB-EGF precursors (FIG. 4D), but also exposes the cryptic anti-tumor domains in both N- and C-terminal cleavage fragments. In addition to ADAMTS-1, ADAMTS-4, and -12 undergo proteolytic cleavage at their spacer/Cys-rich region as well. The auto-proteolytic cleavage may be a general mechanism that regulates function of many ADAMTS family members, and our results provided the first example of this type of regulatory mechanism.

As described herein, it has been shown that ADAMTS-1NTCF and ADAMTS-1CTCF inhibits activation of Erk1/2 kinase induced by AR, HB-EGF, or VEGF (FIG. 5-6). A recent study has shown that ADAMTS-1 inhibits VEGF activity by blocking the binding between VEGF and their receptor. Although additional study is required to determine the exact biochemical mechanism underlying the inhibitory effect of ADAMTS-1NTCF and ADAMTS-1CTCF, they likely exert their inhibitory effect by sequestering these soluble GFs from their receptors.

The Function of ADAMTS-1 is Regulated by HS/HSPGs

As described herein, heparin/HS blocks auto-proteolytic cleavage of ADAMTS-1, and full-length ADAMTS-1 and the ADAMTS-1 cleavage fragments (ADAMTS-1NTCF and ADAMTS-1CTCF) displayed opposite effects on tumor metastasis. Thus, ability of ADAMTS-1 to inhibit or promote tumor metastasis is dependent on the ability of tumor cells and their surrounding microenvironment to cleave ADAMTS-1. In other words, in a microenvironment that is highly enriched HS and HSPGs, binding of ADAMTS-1 to HS/HSPGs protects the proteolytic cleavage sites in the spacer/Cys-rich region which keeps ADAMTS-1 in the full-length form and in turn binds and cleaves its substrates including transmembrane AR and HB-EGF. In that situation, full-length ADAMTS-1 exerts pro-tumor activity by releasing and activating pro-proliferation, -survival, and -angiogenic factors. In addition, the anti-tumor activity derived from TSP type I motifs is likely masked in full-length ADAMTS-1. On the contrary, in a microenvironment that is lack of or low in HS/HSPGs, ADAMTS-1 is likely cleaved to generate the cleavage fragments that are without the substrate (AR and HB-EGF) binding site(s) and contain unmask the anti-tumor TSP type I motifs.

Example 3 The Pro-Tumor Effect of Full-Length ADAMTS-1 and the Anti-Tumor Effect of the ADAMTS-1 Fragments were Confirmed in Lewis Lung Carcinoma (LLC) Cells

To confirm the effects of full-length ADAMTS-1 and the ADAMTS-1 fragments on tumor growth and metastasis, and compare their effects with that of thrombospondin-1 and -2, LLC transfectants were established that were transfected with empty expression vectors (LLCwtb) or expressing full-length ADAMTS-1, ADAMTS-1E/Q, ADAMTS-1NTF, ADAMTS-1CTF, thrombospondin-1, or -2 (FIG. 16). ADAMTS-1, thrombospondin-1 and -2 are the members of thrombospondin type I repeat superfamily (TRS). Thrombospondin-1 is a 450 kDa homotrimeric ECM protein and is considered as a potent anti-tumor molecule. Studies have shown that systemic injection or overexpression of thrombospondin-1 inhibits the in vivo growth of several tumor cells including LLC cells (70, 78, 79). A subline of LLC cell (LLCwt) that undergoes spontaneous pulmonary metastasis after removal of the primary subcutaneous (s.c.) tumors were used to establish these transfectants. LLCwt cells express a low level of endogenous ADAMTS-1 as assessed by RT-PCR and Western blot analysis (data not shown). Five independent clonal LLC transfectants expressing a high to intermediate level of the same gene products were randomly selected, pooled (FIG. 16), and used in the s.c. tumor growth and spontaneous pulmonary metastasis experiments following the established protocols (80-82, 108, 110).

The results showed that expression of full-length ADAMTS-1 promotes while expression ADAMTS-1NTF or ADAMTS-1CTF and to a less extent that of ADAMTS-1E/Q inhibits s.c. growth and spontaneous pulmonary metastasis of the LLC transfectants (FIG. 16). More importantly, even though the LLC transfectants express a higher level of thrombospondin-1 or -2 comparing to that of ADAMTS-1NTF and ADAMTS-1CTF, the inhibitory effect derived from the ADAMTS-1 fragments is stronger than that derived from thrombospondin-1 or -2 (FIG. 16), suggesting that the ADAMTS-1 fragments and their derivatives have unique features and a great potential to be used as the potent anti-cancer agents.

The Metalloproteinase Activity in ADAMTS-1NTF is not Required for its Anti-Tumor Activity:

To investigate whether metalloproteinase activity in ADAMTS-1NTF is required for the anti-tumor activity of ADAMTS-1NTF, a protease-dead ADAMTS-1NTFE/Q mutant was established, in which E386 is switched to Q in the Zinc-binding pocket of the metalloproteinase domain. The expression constructs were used to transfect TA3 mouse mammary carcinoma cells. Three independent positive colonies that express ADAMTS-1NTFE/Q or ADAMTS-1NTF or transfected with the empty expression vectors (FIG. 17) were used in the pulmonary tumor metastasis experiments. Our results showed that ADAMTS-1NTFE/Q behaved like ADAMTS-1NTF and significantly promoted the survival of the experimental mice and inhibited the pulmonary tumor metastasis (FIG. 17). This result suggests that the metalloproteinase domain of ADAMTS-1 does not contribute to the anti-tumor effect of ADAMTS-1NTF.

Example 4

The anti-tumor and anti-angiogenic activity of thrombospondin-1 has been well established and the anti-tumor activity has been mapped to the several domains including the TSP type I repeats. All the members of the ADAMTS family contain at least one TSP-1 motif and belong to the thrombospondin type I repeat (TSR) superfamily (73). Since identification of ADAMTS-1 (22), several studies have been performed to investigate the role of ADAMTS-1 in tumor growth and metastasis, and the results obtained appeared to contradict each other. In pancreatic cancer samples, a higher ADAMTS-1 mRNA level was correlate to the severe lymph node metastasis or retroperitoneal invasion and worse prognosis, suggesting that ADAMST-1 likely promotes pancreatic cancer invasion and metastasis. However, ADAMTS-1 mRNA is down-regulated in the breast carcinoma samples comparing to the nonneoplastic mammary tissues but with no strong links between the ADAMTS-1 mRNA level and the clinicopathological features of these breast cancer cases studied. These studies have only measured ADAMTS-1 mRNA level but not the protein level and the proteolytic activity of ADAMTS-1, both of which are more relevant to the ADAMTS-1 function.

In addition, ADAMTS-1 was found to inhibit tumor growth by blocking tumor angiogenesis, which is likely achieved by sequestering VEGF165 from its receptor, and the metalloproteinase activity of ADAMTS-1 is required for the observed anti-angiogenesis and anti-tumor growth activity. In contrast to this finding, overexpression of ADAMTS-1 was found to promote subcutaneous growth of the transfected CHO cells but inhibit experimental metastasis of the same transfectants. However, these studies have neither considered the fact that ADAMTS-1 is proteolytically cleaved, nor investigated the cleavage status of ADAMTS-1 in vivo (in subcutaneous and pulmonary microenvironments), and did not consider the possibility that the requirement of the metalloproteinase activity of ADAMTS-1 for its anti-tumor effect may merely reflect to the fact that the anti-tumor effect is actually derived from the auto-proteolytic cleavage fragments but not the full-length ADAMTS-1, and that the metalloproteinase activity of ADAMTS-1 is required for generating these ADAMTS-1 fragments.

To test this possibility, the how and why full-length and the ADAMTS-1 fragments affect tumor growth and metastasis was investigated. As described herein, it is demonstrated that overexpression of full-length ADAMTS-1, which is maintained in the full-length form during metastasis of TA3 mammary carcinoma cells, promotes the tumor metastasis, and that ADAMTS-1 promotes shedding of AR and HB-EGF precursors and activation of EGFR and ErbB-2 in vivo. In addition, for the first time that ADAMTS-1 undergoes auto-proteolytic cleavage to generate the NH2- and COOH-terminal fragments that contain at least one TSP-1 motif is shown. In contrast to that of full-length ADAMTS-1, overexpression of the fragments of ADAMTS-1 (ADAMTS-1NTCF and ADAMTS-1CTCF) that mimic the proteolytic cleavage fragments of ADAMTS-1 blocks pulmonary metastasis of TA3 cells by inhibiting tumor cell extravasation, proliferation and survival, and by repressing tumor angiogenesis. It is demonstrated that the anti-metastatic activity of the ADAMTS-1 fragments requires the TSP-1 motif, which is likely masked in the full-length molecule, and that ADAMTS-1NTF and ADAMTS-1CTF inhibit activation of EGFR and ErbB-2 in vivo and inhibits the Erk1/2 kinase activation induced by soluble AR and HB-EGF.

Furthermore, it is demonstrated that the proteolytic cleavage of ADAMTS-1 is blocked by heparin, and HS, suggesting that the binding of ADAMTS-1 to heparan sulfate proteoglycans (HSPGs) masks the auto-proteolytic cleavage site(s) in the spacer/Cys-rich domain and keep ADAMTS-1 in the full-length form to cleave their substrates. On the other hand, the auto-proteolytic cleavage of ADAMTS-1 in the spacer/Cys-rich domain likely destroys the substrate binding sites and unmasks the anti-tumor TSP-1 domain, which renders the anti-tumor activity to the ADAMTS-1 fragments. Thus, the level of HSPGs in the microenvironment likely regulates the form of ADAMTS-1 (full-length or the cleavage fragments) that exists predominantly in the microenvironment to exert pro- or anti-tumor activity, respectively. It is demonstrated that ADAMTS-1 expressed by TA3 cells is maintained in the full-length form in vivo to exert pro-metastasis activity. Thus, the results have reconciled the apparent contradiction in the previous results and demonstrated that the cleavage status of ADAMTS-1 determines its effect (stimulatory or inhibitory) on tumor growth and metastasis.

The results described herein suggested that ADAMTS-1 plays the multiple roles in tumor growth and metastasis and is a prime target for cancer therapy, and that the ADAMTS-1 fragments have great potential as the potent anti-cancer agents that inhibit not only tumor cell proliferation/survival/invasion, but also tumor angiogenesis.

The Pro-Metastatic Activity of Full-Length ADAMTS-1 Requires its Metalloproteinase Activity

To determine whether the metalloproteinase activity of ADAMTS-1 is required for the pro-metastatic activity of ADAMTS-1, TA3 transfectants expressing the protease-dead mutant of ADAMTS-1 (ADAMTS-1E/Q), which harbors an E386 to Q point mutation in the Zinc-binding pocket of the metalloproteinase domain were generated. The study has shown that this mutant lacks the catalytic activity.

In order to assess the effects of ADAMTS-1E/Q on tumor metastasis reliably, the established clonal TA3 cell line, TA3wt1 was used. Like its parental cells, TA3wt1 cells express ADAMTS-1 endogenously and undergo pulmonary metastasis after intravenous (i.v.) injection. Five independent clonal TA3 transfectants expressing a high to intermediate level of ADAMTS-1E/Q were randomly selected and used as the pooled population (TA3ADAMTS-1E/Q, FIG. 7B) in the pulmonary metastasis experiments. Five independent clonal TA3 transfectants transfected with the empty expression vectors or expressing the following same gene products were used as the pooled population as well: full-length ADAMTS-1 (TA3ADAMTS-1), ADAMTS-1NTF (TA3ADAMTS-1NTF), ADAMTS-1CTF (TA3ADAMTS-1CTF), and ADAMTS-1minusTSP-1 (TA3ADAMTS-1minusTSP-1). These pooled TA3 transfectants express a similar level of the transfected gene products (FIG. 7B) and displayed a similar growth rate in the cell culture condition with 10% FBS (data not shown).

It was confirmed that the expression of full-length ADAMTS-1 promotes the pulmonary metastasis of TA3 cells and shortens the survival time of the mice, while ADAMTS-1NTF or ADAMTS-1CTF, but not ADAMTS-1minusTSP-1 blocks the pulmonary metastasis of the transfectants (FIG. 7C-D). In addition, the expression of ADAMTS-1E/Q inhibits the pulmonary metastasis of the transfectants and extends the survival time of the mice (FIG. 7C-D), suggesting that the metalloproteinase activity is required for the pro-metastatic activity of full-length ADAMTS-1. The metastatic burden was quantified by the average weight of the experimental mouse lungs (FIG. 7D). Because there is a significant difference in the survival time of the experimental mice which succumb to pulmonary metastasis when metastatic burden causes the lung weight to reach 1-1.2 gram, the metastatic burden was measured in the remaining survival mice at day 12 and 20 after i.v. injection of these TA3 transfectants. At least 12 mouse lungs were weighted for each type of the transfectants at each time point. We confirmed that overexpression of full-length ADAMTS-1 accelerated the time that is required to reach the maximal metastatic burden and shortened the survival time of the mice, while overexpression of ADAMTS-1E/Q, ADAMTS-1NTF, or ADAMTS-1CTF but not ADAMTS-1minusTSP-1 reduced the metastatic burden (FIG. 7D). Furthermore, it was demonstrated that the inhibitory effect derived from ADAMTS-1NTF or ADAMTS-1CTF is stronger than that derived from ADAMTS-1E/Q, implying that the underlying mechanisms for their anti-metastatic effects may be different. This hypothesis was supported by the results obtained previously, which indicated that ADAMTS-1E/Q but not ADAMTS-1NTF and ADAMTS-1CTF serves as a dominant negative regulator of full-length endogenous ADAMTS-1 by inhibiting the shedding of HB-EGF and AR transmembrane precursors (FIG. 4). Together, these data suggest that like full-length ADAMTS-1, the anti-tumor TSP-1 domains in ADAMTS-1E/Q are masked, and that the anti-tumor activity is likely derived from the intact spacer/Cys-rich domain, which competes with ADAMTS-1 for the binding to its substrates.

The Spacer/Cys-Rich Domain is Essential for Binding of ADAMTS-1 to the Cell Surface and the ECM

The ADAMTS-1 substrates identified so far are versican, aggrecan, and HB-EGF and AR precursors, which are located on the ECM and the cell surface, respectively. To determine the domain(s) of ADAMTS-1 that mediate(s) the substrate binding, we first assess the ECM and the cell binding capacity of the different deletional mutants of ADAMTS-1 (see FIG. 7A). All the constructs contain the COOH-terminal v5-epitope tags for easy identification and purification, and the constructs were transfected into COS-7 cells. 72 hours after the transfection, the proteins derived from the cell culture supernatants, the ECM materials deposited by the transfected cells, and the lysates of the transfected cells were analyzed by Western blotting with anti-v5 epitope antibody as described (106, 109). The results showed that the spacer/Cys-rich domain is essential for the binding of ADAMTS-1 to the ECM and the cells (FIG. 8), suggesting that the spacer/Cys-rich domain likely mediates the substrate binding of ADAMTS-1.

ADAMTS-1 Promotes Invasion of TA3 Cells Through Matrigel

It is well established that the pericellular proteolysis mediated by MMPs is essential for tumor invasion. As a member of the Zinc2+-dependent metalloproteinase family, ADAMTS-1 plays an important role in degrading versican, an important component of the ECM and the blood vessel walls. We have shown that ADAMTS-1 promotes extravasation of TA3 cells into lung parenchyma (FIG. 4). To determine how full-length ADAMTS-1 and the fragments of ADAMTS-1 affect tumor cell invasion through Matrigel which mimics the basement membrane as the barriers of tumor cell invasion, an invasion assay by using Transwell cell culture chambers with 8-μm pores (Costar) coated with a layer of Matrigel (Collaborative Biomedical) was performed. The DMEM containing 2% FBS was be added into the lower chambers of the Transwells. 2×105 of the different TA3 transfectants were seeded on top of the Transwell in triplicate and incubated for 24 hours. The bottom filters were then be fixed and stained. The cells on the top chambers were removed by wiping with cotton swabs, and the stained cells (blue color) that have migrated through the Matrigel were counted under a microscope. Six randomly selected 100× microscopic fields will be countered. The invasion index of the different TA3 transfectants was calculated as following formula:

Invasion Index = 100 % × Average numbers of cells in lower camber / microscopic field Numbers of cells seeded on upper camber / microscopic field

The results showed that TA3ADAMTS-1 cells displayed approximately two time higher invasion index than TA3wt1 and TA3ADAMTS-1minusTSP-1 cells, and four-eight time higher invasion index compared to TA3ADAMTS-1NTF/TA3ADAMTS-1CTF and TA3ADAMTS-1E/Q cells, respectively (FIG. 9A-B). These results further confirmed that ADAMTS-1 promotes tumor cell invasion, while TA3ADAMTS-1E/Q and to a less extent TA3ADAMTS-1NTF, or TA3ADAMTS-1CTF inhibits the process. To determine whether ADAMTS-1 promotes TA3 cell invasion by degrading versican or inhibiting the pro-migratory effect of soluble HB-EGF and AR, the confluence TA3 transfectants were lifted by the EDTA solution and the ECM materials remained on the cell culture dishes were extracted and analyzed by Western blotting with anti-DP antibody, which detects the cleavage fragments of versican. The result showed that increased expression of exogenous ADAMTS-1 but not ADAMTS-1minusTSP-1 on top of the endogenous ADAMTS-1 promotes degradation of versican, while expression of ADAMTS-1E/Q but not ADAMTS-1NTF, ADAMTS-1CTF inhibits the degradation (FIG. 9C). These data suggest that ADAMTS-1E/Q inhibits TA3 cell invasion by blocking the ADAMTS-1 mediated versican degradation, while the weaker inhibitory effect of the ADAMTS-1 fragments is likely derived from their indirect effect on activity of HB-EGF/AR.

Example 5

Following the experimental procedures described in Example 1, addition data was generated showing additional differences between full length ADAMTS-1 and its cleavage products, ADAMTS-1NTCF and ADAMTS-1CTCF. Overexpression of ADAMTS-1 promotes growth of TA3 mammary carcinoma (TA3) cells while overexpression of the N- or C-terminal fragment of ADAMTS-1 blocks growth of TA3 cells by inhibiting proliferation and inducing apoptosis of the tumor cells and by inhibiting tumor angiogenesis. ADAMTS-1 expressed by TA3 cells maintained in the full-length form in vivo exerted pro-tumor growth and metastasis activity. In contrast to the of full-length ADAMTS-1, overexpression of the N- or C-terminal fragment of ADAMTS-1 (ADAMTS-1NTCF and ADAMTS-1CTCF) inhibits subcutaneous (s.c.) growth of TA3 cells. In addition, unlike full-length ADAMTS-1 which promotes shedding of the EGF family ligands including amphiregulin (AR) and heparin-binding EGH (HB-EGF) and activation of EGF receptor (EGFR) and ErbB-2, the ADAMTS-1 fragments inhibits activation of EGFR and ErbB-2 in vivo.

RT-PCR results showed that like wild type TA3 cells, TA3wt1 cells express ADAMTS-1 endogenously as do several other tumor cell lines (FIG. 10). Growth rates of the s.c. solid tumors derived from different TA3 transfectants were measured and the result showed that overexpression of ADAMTS-1 promotes tumor growth, while overexpression of ADAMTS-1NTCF and ADAMTS-1CTCF, but not that of ADAMTS-1minusTSP significantly inhibits tumor growth (FIG. 11). These results suggest that the inhibitory effect of the ADAMTS-1 fragments is likely derived from the TSP type I motifs, which exist in ADAMTS-1NTCF and ADAMTS-1CTCF, but not in ADAMTS-1minusTSP. The data show that ADAMTS-1NTCF or ADAMTS-1CTCF, blocks pulmonary metastasis of TA3ADAMTS-1NTCF or TA3ADAMTS-1CTCF cells (FIG. 11, B). The metastatic burden of the experimental mice was quantified by average weight of the experimental mouse lungs received different TA3 transfectants (FIG. 11, B). Results showed that overexpression of ADAMTS-1NTCF or ADAMTS-1CTCF dramatically reduced metastatic burden of the mice received the corresponding TA3 transfectants, and render most of the experimental mice free of metastatic disease and significantly extended survival time of these mice (FIG. 11, B). [0180] The ADAMTS-1 fragments blocks tumor growth by inhibiting proliferating and inducing apoptosis of tumor cells, and inhibiting tumor angiogenesis. To determine the cellular basis of the pro-tumor effect of full-length of ADAMTS-1 and the anti-tumor effect of ADAMTS-1 fragments, proliferation and apoptosis rates of the tumor cells and tumor angiogenesis during s.c. growth were analyzed. Brdu (5-Bomo-2′-deoxy-uridine) incorporation assay and in situ detection of apoptotic cells were performed on the sections derived from s.c. solid tumors (twelve days after implanting the TA3 cells). Results demonstrated that expression of ADAMTS-1NTCF and to a less extent that of ADAMTS-1CFCF, but not expression of ADAMTS-1minusTSP, inhibits proliferation and promotes apoptosis of the tumor cells, and inhibits angiogenesis in the subcutaneous space; while expression of exogenous ADAMTS-1 mildly enhances proliferation rate and reduces apoptosis rate of the tumor cells, and promotes tumor angiogenesis in vivo (FIG. 12) These results suggest that ADAMTS-1 may play an important role in releasing/activating growth/survival factors in the microenvironments, while the cleavage fragments of ADAMTS-1 may block the activities of the factors that promote tumor cell proliferation and survival and tumor angiogenesis.

Activation of EGFR and ErbB-2 is known to promote proliferation and survival of breast carcinoma cells and play essential roles in progression of breast cancers. To determine whether activation of EGFR and/or ErbB-2 underlies the pro-tumor activity of ADAMTS-1, we assessed activity of EGFR and ErbB-2 in the lungs where TA3wtb, TA3ADAMTS-1, TA3ADAMTS-1NTCF, TA3ADAMTS-1NTCF, or ADAMTS-1minusTSP cells were injected five days prior. The result showed that expression of ADAMTS-1 by TA3 cells promotes activation of EGFR and ErbB-2 in vivo (FIG. 13, A). On the contrary, expression of ADAMTS-1NTCF or ADAMTS-1CTCF, but not ADAMTS-1minusTSP which lacks TSP type I motifs, blocks activation of EGFR and ErbB-2 in vivo (FIG. 13, A).

Experiments were done to assess whether induces activation of EGFR and ErbB-2 by ADAMTS-1 is achieved via promoting shedding EGF family ligands. EGF family ligands are produced as transmembrane precursors, which are shed and released from cell surface as soluble mature form. Several EGF family ligands are known to be shed-activated by the ADAM family proteinases including ADAM-17. However, studies using the cells derived from ADAM-17 null-mouse suggested that ADAM-17 is not the sole proteinase that is responsible for shedding of TGF-α and other member(s) of ADAM family is (are) likely to play a role as well. To determine whether ADAMTS-1 play a role in shedding EGF family ligands especially the ones that bind to heparin, several EGF family ligands were co-transfected including HB-EGF, TGF-α, AR, and epigen which are expressed by TA3 cells (data not shown) with or without the full-length ADAMTS-1, ADAMTS-1E/Q and the ADAMTS-1 fragments. The serum-free cell culture medium of the transfected cells were collected and concentrated and analyzed. The results showed that ADAMTS-1 promotes shedding of AR and HB-EGF but not shedding of TGF-α and epigen; while ADAMTS-1E/Q blocks the shedding. The ADAMTS-1 fragments displayed no effect on the shedding (FIG. 13, B and data not shown).

To determine whether the ADAMTS-1 fragments affect the signal transduction pathways activated by HB-EGF and AR, the serum-free cell culture media (SFM) derived from the co-transfected cells were applied to MCF-10A mammary epithelial cells to determine their ability to induce Erk1/2 kinase activation. The results showed that soluble HB-EGF and AR in the SFM induces activation of Erk1/2 kinases, which is specifically blocked by the corresponding blocking antibodies or the ADAMTS-1 fragments, but not by the full-length ADAMTS-1 (FIG. 13, C). This result suggests that the ADAMTS-1 fragments inhibit activation of EGFR and ErbB-2 by interfering with their ligand activity; and the effects of ADAMTS-1 and its cleavage fragments on the availability and activity of EGF family ligands likely underlies their roles in tumor growth and metastasis.

The affect the fragments have on activities of several important growth/angiogenic factors that are known to regulate angiogenesis was investigated. Activity of VEGF165, bFGF, HB-EGF, TGF-α, and AR were revealed by their ability to induce activation of Erk1/2 kinases in HUVECs in the presence or absence of different purified ADAMTS-1 proteins. Results showed that ADAMTS-1NTCF and ADAMTS-1CTCF but not full-length ADAMTS-1 or ADAMTS-1minusTSP block activation of Erk1/2 kinase induced by VEGF165, TGF-α, HB-EGF, and AR

ADAMTS-1 is widely expressed by tumor cells and undergoes auto-proteolytic cleavage. In addition, overexpression of ADAMTS-1 promotes tumor growth and metastasis by enhancing tumor cell proliferation and survival and by promoting tumor angiogenesis through shedding transmembrane EGF family ligands, AR and HB-EGF, which in turn promotes activation of EGFR and ErbB-2 in vivo.

The results not only provided a potential important target (full-length ADAMTS-1), potent novel anti-cancer reagents (the ADAMTS-1 fragments), and the regulatory reagents for ADAMTS-1 activity (HS/HSPGs) for the treatment of cancers especially breast cancers in the figure, but also revealed the mechanism underlying the function of ADAMTS-1 and the ADAMTS-1 fragments.

The presence of TSP type I motif is a common feature of all members of ADAMTS family, among them ADAMTS-1, -4, AND -12 undergo proteolytic cleavage at their spacer/Cys-rich region, which have potential to generate ADAMTS fragments containing unmasked TSP type I motifs that may possess anti-tumor activity. In addition, as indicated in this study, the auto-proteolytic cleavage may be a general mechanism that regulates the function of ADAMTS family members. Additional work is required to verify these hypotheses and the results obtained in this study provide general rules that may apply the other ADAMTS family members as well.

For subcutaneous tumor growth experiments, five independent clonal TA3 transfectants expressing ADAMTS-1, ADAMTS-1CTCF, ADAMTS-1NTCF or ADAMTS-1minusTSP, or transfected with the empty expression vector were used in the in vivo experiments. For each type of the experiment, six mice were injected with each clonal transfectants and two independent experiments were performed.

In tumor growth experiments, TA3 transfectants were injected subcutaneously into syngenic A/Jax-mic as described. After solid tumors became visible (7-10 days after the injection), the tumors were measured by a digital caliper every other day for the next two weeks. The largest and shortest diameters of the solid tumors were measured. The tumor volume was calculated by using the following formula: tumor volume=1/2×(shortest diameter)2×longest diameter (mm3)

Example 6 Determining the Exact Amino Acid Segments in the TSP-1 Domains Containing Anti-Cancer Activity

As discussed herein, the ADAMTS-1 fragments that contain either the middle TSP-1 motif (ADAMTS-1NTF) or the two COOH-terminal TSP-1 modules (ADAMTS-1CTF) inhibit growth and/or metastasis of TA3 and LLC, and their inhibitory effect is much stronger than that caused by thrombospondin-1 and -2, suggesting the unique molecular basis underlying the potent inhibitory effect of the ADAMTS-1 fragments is not present in thrombospondin-1 and -2. Also, the TSP-1 domain is required for the anti-tumor activity of ADAMTS-1NTF. The middle TSP-1 domain (mTSP-1, amino acids 546-596) in ADAMTS-1 is similar but not identical to the second and third TSP-1 repeats (WXXWXXW) in thrombospondin-1, which have been shown to contain anti-tumor and anti-angiogenic activity (102, 111-113). Even though the COOH-terminal TSP-1 modules (cTSP-1, amino acid 842-895 and 896-951, FIG. 9) of ADAMTS-1 do not have high homology to the TSP-1 repeats in thrombospondin-1, the ADAMST-1 fragments that contain either the mTSP-1 or cTSP-1 domain exhibited the similar anti-tumor activity, implying that the common unidentified unique amino acid segments or three dimension feature (other than the WXXWXXW) in the m/cTSP-1 domains of ADAMTS-1 may be essential for the potent anti-tumor activity. Accordingly, the molecular basis for the potent anti-tumor activity that is unique to the ADAMTS-1 fragments can be identified using this domain. To achieve that, deletions in the TSP-1 domains of ADAMTS-1 can be made and tumor growth and metastasis assays can be performed using TA3 and LLC transfectants expressing these ADAMTS-1 mutants (as described herein).

To Determine Whether the m and/or c fTSP-1 Domain Displays Anti-Tumor Activity

It was shown that deletion of the middle TSP-1 (mTSP-1) domain from ADAMTS-1NTF (ADAMTS-1minusTSP-1) abolishes the potent anti-tumor activity of the fragment, suggesting the anti-tumor activity resided in the mTSP-1 domain and that the ADAMTS-1CTF is composed of the two COOH-terminal TSP-1 (cTSP-1) domains. Thus, whether the mTSP-1 domain and each of the cTSP-1 domains display as potent anti-tumor activity as the ADAMTS-1NTF and ADAMTS-1NTF fragments need to be determined. To achieve that, three expression constructs can be generated that contain the signal peptide plus the mTSP-1 domain (fADAMTS-1m-TSP-1), the first cTSP-1 (fADAMTS-1cTSP-1-1), or the second cTSP-1 (fADAMTS-1cTSP-1-2) domain in pEF/6/v5-His expression vectors. They can be used to transfect TA3wt1 and LLCwt1 cells. Five independent TA3 or LLC transfectants expressing a high to intermediate level of fADAMTS-1 mTSP-1, fADAMTS-1cTSP-1-1 or fADAMTS-1cTSP-1-2 will be randomly selected and used as the pooled populations together with the established TA3 and LLC transfectants expressing a similar level of ADAMTS-1CTF or ADAMTS-1NTF, or transfected with the expression vector alone in the s.c. tumor growth and metastasis experiments.

mTSP-1 domain inhibits growth and metastasis of TA3 and LLC cells in a similar extent as that of ADAMTS-1NTF; while expression of each of the cTSP-1 domains display a weaker anti-tumor effect compared to that caused by ADAMTS-1CTF, which contains two TSP-1 modules. These small recombinant proteins (53-56 amino acid long) are used as anti-cancers agents.

Deletions and Mutations in the m or cTSP-1 Domains of ADAMTS-1 and Establish TA3 and LLC Transfectants Expressing these ADAMTS-1 Mutants

Within in the TSP type I repeats of thrombospondin-1, in addition to WXXWXXW motif, the CSVTCG motif, which binds to CD36, has been shown to contain anti-tumor and anti-angiogenic activity. The m/cTSP-1 domains of ADAMTS-1 contain the motifs that are similar to WXXWXXW and/or CSVTCG (SEQ ID NO:47) motifs. In addition, the consensus motif search (GCG genomics) has demonstrated that the most consensus motif among the m and cTSP-1 domains of ADAMTS-1 is the WGE/DCSKTC (SEQ ID NO:50) motif (FIG. 18). Thus, three deletions in m/cTSP-1 domains are made: WGPWGPWGD (ADAMTS-1mTSP-1WXXWde1-SEQ ID NO:48) or WV/QI/VE/GE/DWG/S (ADAMTS-1cTSP-1WXXXXWde1), WGDCSRTC (ADAMTS-1mTSP-1WGde1-SEQ ID NO:49) or WG/SE/PCSKTC (ADAMTS-1cTSP-1WG/Sde1-SEQ ID NO:51), CSRTCGGG (ADAMTS-1mTSP-1CSde1-SEQ ID NO:52) or CSKTCGS/KG (ADAMTS-1cTSP-1CSde1-SEQ ID NO:53, FIG. 18).

The deletional mutagenesis are performed as described using fADAMTS-1mTSP-1 and fADAMTS-1cTSP-1-1, or fADAMTS-1cTSP-1-2 (in pEF/6/v5-His expression vectors) as the templates. These expression constructs are used to transfect Cos-7 cells transiently to assess the expression capacity of these v5-epitope tagged fragments. All the deletional mutants are established in cell lines and their proper expression in Cos-7 cells is demonstrated. These deletional constructs are used to transfect TA3wt1 and LLCwt1 cells. Five independent TA3 or LLC transfectants expressing a high to intermediate level of each of the mutants are used as the pooled populations in the s.c. tumor growth and metastasis experiments together with the established TA3 and LLC transfectants expressing ADAMTS-1NTF, ADAMTS-1CTF, fADAMTS-1mTSP-1 and fADAMTS-1cTSP-1-1, or fADAMTS-1cTsp-1-2, or transfected with the expression vector alone (the control).

The disclosures of each and every patent, patent application, publication, and accession number cited herein are hereby incorporated herein by reference in their entirety. The appended sequence listing is hereby incorporated herein by reference in its entirety.

While this invention has been disclosed with reference to specific embodiments, it is apparent that other embodiments and variations of this invention may be devised by others skilled in the art without departing from the true spirit and scope of the invention. The appended claims are intended to be construed to include all such embodiments and equivalent variations.

Claims

1. An isolated polypeptide fragment of ADAMTS-1 that inhibits tumor growth and/or metastasis, wherein the fragment consists of SEQ ID Nos: 6, 7, 9, and or 11.

2. A pharmaceutical composition comprising a polypeptide of claim 1.

3. A composition comprising at least two different polypeptide fragment of ADAMTS-1 that inhibit cell proliferation or metastasis, wherein said fragment comprises SEQ ID Nos: 5, 7, 9, and/or 11.

4. The position of claim 3 wherein said composition is a pharmaceutical composition.

5. An isolated polynucleotide encoding a polypeptide fragment of ADAMTS-1 wherein said fragment inhibits tumor growth and/or metastasis, wherein said polynucleotide comprises SEQ ID NO: 6, 8, 10, or 12.

6. A pharmaceutical composition comprising the isolated polynucleotide of claim 6.

7. The isolated polynucleotide of claim 6 wherein said polynucleotide is a vector or plasmid.

8. A method for identifying an inhibitor or an activator of ADAMTS-1 auto-cleavage comprising performing a test assay comprising:

a) contacting ADAMTS-1 with a test compound under conditions in which ADAMTS-1 undergoes cleavage in the absence of a test compound;
b) measuring cleavage level of ADAMTS-1; and
c) comparing the cleavage level to cleavage level of ADAMTS-1 in the absence of the test compound, wherein a decrease in auto-cleavage indicates that the test compound is a cleavage inhibitor or wherein an increase in auto-cleavage indicates that the test compound is a cleavage activator.

9. The method of claim 8 wherein said the test compound is contacted with a cell comprising ADAMTS-1.

10. The method of claim 9, further comprising performing, a negative control assay which comprises contacting a cell that does not comprise ADAMNTS-1 or a cell that comprises a cleavage resistant mutant of ADAMTS-1.

11. The method of claim 9, further comprising performing a positive control assay which comprises contacting a cell comprising ADAMTS-1 a positive control compound and measuring cleavage.

12. The method of claim 8, further comprising measuring the cleavage of ADAMTS-1 in the absence of the test compound.

13. A method for identifying a heparin inhibitor comprising:

a) contacting a composition comprising heparin and ADAMTS-1 with a test compound wider conditions in which ADAMTS-1 undergoes auto-cleavage and/or proteolytic cleavage in absence of heparin;
b) measuring cleavage level of ADAMTS-1; and
c) comparing cleavage level of ADAMTS-1 in the absence of the test compound;
wherein an increase in the cleavage of ADAMTS-1 indicates that the compound is a heparin inhibitor.

14. A method of identifying a metalloproteinase inhibitor comprising:

a) contacting a ADAMTS-1 polypeptide or fragment thereof comprising metalloproteinase activity with a test compound under conditions which metalloproteinase activity is detected in the absence of the test compound.
b) measuring metalloproteinase activity level of ADAMTS-1; and
c) comparing the metalloproteinase activity level of ADAMTS-1 in the presence or absence of the test compound,
wherein a decrease in metalloproteinase activity indicates the test compound is a metalloproteinase inhibitor.

15. The method of claim 14 wherein said fragment comprises SEQ ID NO: 5, 7, 9, and/or 11.

16. The method of claim 14 wherein the metalloproteinase activity of ADAMTS-1 is compared to a fragment or mutant of ADAMTS-1 that has no metalloproteinase activity.

17. The method of claim 16 wherein said fragment or mutant of ADAMTS-1 that has no metalloproteinase activity comprises SEQ ID NO 31, 33, 35, and/or 36.

18. A method of treating cancer in an individual comprising administering to the individual a therapeutically effective amount of a polypeptide fragment of ADAMTS-1 and/or a nucleic acid that encodes a polypeptide fragment of ADAMTS-1 that inhibits cell proliferation and/or metastasis.

19. The method of claim 18 wherein the polypeptide fragment comprises a TSP type-I motif.

20. The method claim 18 wherein the fragment comprises SEQ ID NO: 5, 7, 9 and/or 11.

21. The method of claim 18 wherein the nucleic acid molecule encoding the polypeptide fragment comprises SEQ ID NO: 6, 8, 10, and/or 11.

22. The method of claim 18 wherein said polypeptide fragment of ADAMTS-1 comprises the spacer/Cys-rich and/or spacer domain of ADAMTS-1 or a nucleic acid molecule encoding a polypeptide fragment of ADAMTS-1 comprising the spacer/Cys-rich and/or spacer domain of ADAMTS-1.

23. The method of claim 22 wherein said fragment comprises SEQ ID NO: 21 and/or 23.

24. The method of claim 22 wherein said nucleic acid molecule comprises SEQ ID NO: 22 and/or 24.

25. A method of mating cancer comprising administering an inhibitor of the metalloproteinase activity of ADAMTS-1.

26. The method of claim 25 wherein the inhibitor is a metalloproteinase defective polypeptide of ADAMTS-1 or a nucleic acid molecule encoding a metalloproteinase defective polypeptide of ADAMTS-1.

27. The method of claim 26 wherein the metalloproteinase defective polypeptide of comprises SEQ ID NO: 29, 31, 33, and/or 35.

28. The method of claim 26 wherein nucleic acid molecule comprises SEQ ID NO: 30, 32, 34, and/or 36.

29. The method of claim 25 wherein said inhibitor is an antibody that binds to ADAMTS-1.

Patent History
Publication number: 20140294846
Type: Application
Filed: Feb 18, 2014
Publication Date: Oct 2, 2014
Applicant: THE TRUSTEES OF THE UNIVERSITY OF PENNSYLVANIA (Philadelphia, PA)
Inventor: Qin Yu (New York, NY)
Application Number: 14/183,341