Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
ADAM22 FOR USE AS A PROGNOSTIC VARIABLE, AND TARGET FOR THERAPY, OF A METASTATIC BREAST CANCER DISEASE
Document Type and Number:
WIPO Patent Application WO/2013/064699
Kind Code:
A1
Abstract:
A method of diagnosing metastatic potential of a breast cancer in an individual with breast cancer is described. The method comprises a step of assaying a breast cancer tumour sample from the patient for expression of ADAM22, wherein expression of ADAM22 correlates with increased potential for metastasis compared with a patient who is ADAM22 negative. The invention also describes an agent for use in the treatment of metastatic breast cancer in a patient, in which the agent is selected from LGI1 protein (SEQ ID NO:1) and an LGI1 peptide mimic capable of mimicking the ADAM22 binding activity of LGI1 by binding to the LGI1 binding domain of ADAM22 (SEQ ID NO: 4) and which is capable of inhibiting migration of endocrine resistant breast cancer cells.

Inventors:
YOUNG LEONIE (IE)
MCCARTAN DAMIAN (IE)
BYRNE CHRISTOPHER (IE)
Application Number:
PCT/EP2012/071864
Publication Date:
May 10, 2013
Filing Date:
November 05, 2012
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
ROYAL COLLEGE OF SURGEONS IE (IE)
International Classes:
G01N33/574
Foreign References:
US6057105A2000-05-02
US20050221376A12005-10-06
US20050221376A12005-10-06
US20050001006A12005-01-06
Other References:
P. KUNAPULI: "LGI1, a Putative Tumor Metastasis Suppressor Gene, Controls in Vitro Invasiveness and Expression of Matrix Metalloproteinases in Glioma Cells through the ERK1/2 Pathway", JOURNAL OF BIOLOGICAL CHEMISTRY, vol. 279, no. 22, 29 February 2004 (2004-02-29), pages 23151 - 23157, XP055021536, ISSN: 0021-9258, DOI: 10.1074/jbc.M314192200
NADIA. GABELLINI ET AL: "Increased expression ofLGI1 gene triggers growth inhibition and apoptosis of neuroblastoma cells", JOURNAL OF CELLULAR PHYSIOLOGY, vol. 207, no. 3, 1 June 2006 (2006-06-01), pages 711 - 721, XP055021481, ISSN: 0021-9541, DOI: 10.1002/jcp.20627
Y. FUKATA ET AL: "Epilepsy-Related Ligand/Receptor Complex LGI1 and ADAM22 Regulate Synaptic Transmission", SCIENCE, vol. 313, no. 5794, 22 September 2006 (2006-09-22), pages 1792 - 1795, XP055021485, ISSN: 0036-8075, DOI: 10.1126/science.1129947
D. MCCARTAN ET AL: "Global Characterization of the SRC-1 Transcriptome Identifies ADAM22 as an ER-Independent Mediator of Endocrine-Resistant Breast Cancer", CANCER RESEARCH, vol. 72, no. 1, 9 November 2011 (2011-11-09), pages 220 - 229, XP055021430, ISSN: 0008-5472, DOI: 10.1158/0008-5472.CAN-11-1976
DATABASE ArrayExpress [online] 1 November 2011 (2011-11-01), "Experiment E-GEOD-28987", XP002671266, retrieved from EBI Database accession no. E-GEOD-28987
SETUBAL; MEIDANIS ET AL.: "Introduction to Computational Biology Methods", 1997, PWS PUBLISHING COMPANY
"Computational Methods in Molecular Biology", 1998, ELSEVIER
RASHIDI; BUEHLER: "Bioinformatics Basics: Application in Biological Science and Medicine", 2000, CRC PRESS
OUELETTE; BZEVANIS: "Bioinformatics: A Practical Guide for Analysis of Gene and Proteins", 2001, WILEY & SONS, INC.
MCCARTAN D; BOLGER J; FAGAN A. ET AL.: "Global characterization of the SRC-1 transcriptome identifies ADAM22 as an ER-independent mediator of endocrine resistant breast cancer", CANCER RES., vol. 72, 2012, pages 220 - 9
DILLON MF; STAFFORD AT; KELLY G; REDMOND AM; MCILROY M; CROTTY TB ET AL.: "Cyclooxygenase-2 predicts adverse effects of tamoxifen: a possible mechanism of role for nuclear HER2 in breast cancer patients", ENDOCR RELAT CANCER, vol. 15, 2008, pages 745 - 53
HARVEY JM; CLARK GM; OSBORNE CK; ALLRED DC.: "Estrogen receptor status by immunohistochemistry is superior to the ligand-binding assay for predicting response to adjuvant endocrine therapy in breast cancer", J CLIN ONCOL., vol. 17, 1999, pages 1474 - 81
FUKATA Y; ADESNIK H; IWANAGA T; BREDT DS; NICOLL RA; FUKATA M.: "Epilepsy-related ligand receptor complex LG11 and ADAM22 regulate synaptic transmission", SCIENCE, vol. 313, 2006, pages 1792 - 95
J. M. STEWART; J. D. YOUNG: "Solid Phase Peptide Synthesis", 1984, PIERCE CHEMICAL COMPANY
M. BODANZSKY; A. BODANZSKY: "The Practice of Peptide Synthesis", 1984, SPRINGER VERLAG
GABELLINI N; MASOLA V; QUARTESAN S ET AL.: "Increased expression of LGl1 gene triggers growth inhibition and apoptosis of neuroblastoma cells", J CELL PHYSIOL, vol. 207, 2006, pages 711 - 721
GABELLINI N; MASOLA V.: "Expression of LGl1 impairs proliferation and survival of HeLa cells", INT J CELL BIOL, 2009
KUNAPULI P; KASYAPA CS; HAWTHORN L; COWELL JK: "LG11, a putative tumour metastasis suppressor gene, controls in vitro invasiveness and expression of matrix metalloproteinase in glioma cells through the ERK1/2 pathway", J BIOL CHEM, vol. 279, 2004, pages 23151 - 23157
KUNAPULI P; KASYAPA CS; HAWTHORN L; COWELL JK.: "LGl1, a putative tumor metastasis suppressor gene, controls in vitro invasiveness and expression of matrix metalloproteinases in glioma cells through the ERK1/2 pathway", J BIOL CHEM., vol. 279, no. 22, 26 March 2004 (2004-03-26), pages 23151 - 7
FUKATA Y; ADESNIK H; IWANAGA T; BREDT DS; NICOLL RA; FUKATA M.: "Epilepsy-related ligand/receptor complex LGl1 and ADAM22 regulate synaptic transmission", SCIENCE, vol. 313, no. 5794, 22 September 2006 (2006-09-22), pages 1792 - 5
SAGANE K, INT J BIOL SCI, vol. 4, no. 6, 2008, pages 387 - 396
Attorney, Agent or Firm:
PURDY, Hugh Barry (6-7 Harcourt Terrace, Dublin 2, IE)
Download PDF:
Claims:
Claims

1. A method of diagnosing metastatic potential of a breast cancer in an individual with breast cancer, the method comprising a step of assaying a breast cancer tumour sample from the patient for expression of ADAM22, wherein expression of ADAM22 correlates with increased potential for metastasis compared with a patient who is ADAM22 negative.

2. A method as claimed in Claim 1 in which the patient is undergoing treatment for a primary breast cancer.

3. An ADAM22 inhibitor for use in the treatment or prevention of a metastatic breast cancer in a patient.

4. An agent for use in the treatment or prevention of metastatic breast cancer in a patient, in which the agent is selected from:

LGI1 protein (SEQ ID NO: l); and

an LGI1 peptide mimic capable of mimicking the ADAM22 binding activity of LGI1 by binding to the LGI1 binding domain of ADAM22 and which is capable of inhibiting migration of endocrine resistant breast cancer cells.

5. Use of Claim 4 in which the ADAM22 inhibitor comprises or consists of an LGI1 peptide mimic selected from the ADAM22 binding domain of LGI1 (SEQ ID NO: 3), or a functional fragment thereof having least 15 amino acids, or functional variants of the fragment having at least 90% sequence identity with the fragment.

6. Use of Claim 5 in which the fragment is selected from the group consisting of the SEQ ID NO:s 5 to 12.

7. Use of Claim 5 in which the ADAM22 inhibitor is an LGIl peptide mimic comprising the peptide of SEQ ID NO: 5, or a functional variant thereof having at least 90% sequence identity with the fragment.

8. Use of any of Claims 3 to 6 in which the patient is ADAM22 positive.

9. A kit comprising (a) means for detecting whether a patient is ADAM22 positive or ADAM22 negative and (b) a pharmaceutical composition comprising a therapeutically effective amount of an ADAM22 inhibitor.

10. A kit comprising (a) means for detecting whether a patient is ADAM22 positive or ADAM22 negative and (b) a pharmaceutical composition comprising a therapeutically effective amount of an agent selected from:

LGIl protein (SEQ ID NO: l); and

an LGIl peptide mimic capable of mimicking the ADAM22 binding activity of LGIl by binding to the LGIl binding domain of ADAM22 and which is capable of inhibiting migration of endocrine resistant breast cancer cells.

11. A kit of Claim 10 in which the agent comprises or consists of an LGIl peptide mimic selected from the ADAM22 binding domain of LGIl (SEQ ID NO: 3), or a functional fragment thereof having at least 15 amino acids, or functional variants of the fragment having at least 90% sequence identity with the fragment.

12. A kit of Claim 10 in which the agent is an LGIl peptide mimic comprising the peptide of SEQ ID NO: 5, or a functional variant of the fragment having at least 90% sequence identity with the fragment.

13. A method of identifying a breast cancer patient suitable for treatment of metastatic breast cancer with an ADAM22 inhibitor, the method comprising a step of assaying a breast cancer tumour sample from the patient for expression of ADAM22, wherein expression of ADAM22 indicates that the patient is suitable for treatment with an ADAM22 inhibitor.

14. An agent for use as a medicament, the agent selected from the group consisting of:

LGI1 protein (SEQ ID NO: 1); and

an LGI1 peptide mimic capable of mimicking the ADAM22 binding activity of LGI1 by binding to the LGI1 binding domain of ADAM22 (SEQ ID NO: 4) and which is capable of inhibiting migration of endocrine resistant breast cancer cells. 15. A pharmaceutical composition comprising an agent in combination with a pharmaceutically acceptable carrier, the agent selected from the group consisting of:

LGI1 protein (SEQ ID NO: 1); and

an LGI1 peptide mimic capable of mimicking the ADAM22 binding activity of LGI1 by binding to the LGI1 binding domain of ADAM22 (SEQ ID NO: 4) and which is capable of inhibiting migration of endocrine resistant breast cancer cells.

16. A pharmaceutical composition of Claim 15 in which the agent comprises an LGI1 peptide mimic selected from the ADAM22 binding domain of LGI1 (SEQ ID NO: 3), or a functional fragment thereof having least 15 amino acids, or functional variants of the fragment having at least 90% sequence identity with the fragment.

17. A pharmaceutical composition of Claim 15 in which the agent is an LGI1 peptide mimic comprising the peptide of SEQ ID NO: 5, or a functional variant of the fragment having at least 90% sequence identity with the fragment.

18. A system for obtaining data from at least one test sample obtained from at least one individual, the system comprising:

a determination module configured to receive at least one test sample and perform at least one test analysis on the test sample to assay for the expression of ADAM22; optionally, a storage system for ADAM22 expression data generated by the determination system; and

a display module for displaying a content based in part on the data output from said determination module, wherein the content comprises a signal indicative of the expression of ADAM22.

19. A system as claimed in Claim 18 for diagnosing metastatic potential of a breast cancer in an individual with breast cancer, in which the system comprises a correlation module for correlating ADAM22 expression data from the determination module with metastatic potential of breast cancer, wherein the expression of ADAM22 correlates with increased potential for metastasis compared with a patient who is ADAM22 negative, and wherein the display module displays a content based in part on the data from the correlation system, the content optionally comprising a signal indicative of the metastatic potential of the breast cancer.

20. A system as claimed in Claim 18 for identifying a breast cancer patient suitable for treatment of metastatic breast cancer with an ADAM22 inhibitor, in which the system comprises a correlation module for correlating ADAM22 expression data from the determination module with suitability for treatment of metastatic breast cancer with an ADAM22 inhibitor, wherein the expression of ADAM22indicates that the patient is suitable for treatment with an ADAM22 inhibitor, and wherein the display module displays a content based in part on the data from the correlation system, the content optionally comprising a signal indicative of the suitability for treatment of metastatic breast cancer with an ADAM22 inhibitor.

21. A system as claimed in any of Claims 18 to 20 in which the determination system comprises an immunohistochemical detection apparatus.

22. An assay for identifying an agent capable of treating or preventing metastasis of a breast cancer, the method comprising the step of providing a sample of endocrine-resistant breast cancer cells, incubating the sample with a test agent, and detecting binding between the test agent and an LGIl binding site of ADAM22 expressed by the cells, wherein a test agent that binds to the LGIl binding site of ADAM22 is an agent capable of treating or preventing metastasis of a breast cancer.

Description:
ADAM22 FOR USE AS A PROGNOSTIC VARIABLE, AND TARGET FOR THERAPY, OF A METASTATIC BREAST CANCER DISEASE

Technical Field

The invention relates to a method of assessing the metastatic status of a breast cancer in a breast cancer patient. Furthermore the invention relates to the treatment or prevention of metastatic breast cancer. The invention also encompasses a companion diagnostic for the directed use of the therapy to treat metastatic breast cancer.

Background to the Invention

Current endocrine treatment in breast cancer -

Breast cancer continues to affect one woman in ten in the western world, and despite significant treatment advances in recent years, the mortality rate still remains at around 35%. Current endocrine therapies are based on manipulating the estrogen receptor (ER) either directly with ER modulators such as tamoxifen, or by reducing levels of circulating estrogen with aromatase inhibitors (AIs). The development of third-generation aromatase inhibitors (AIs) has changed therapy in post-menopausal patients with hormone-sensitive breast cancer. There are two main types of AIs: irreversible steroidal inhibitors exemplified by exemestane, and non-steroidal inhibitors such as anastrozole and letrozole which are competitive reversible inhibitors. Randomized clinical trials, including several led by our group, have demonstrated prolongation of disease- free survival in patients with early stage breast cancer.

Regardless of the age of the patient, adjuvant endocrine therapy offers substantial benefit in terms of reduction in risk of tumour recurrence in women with ER-positive tumours. However, while most patients initially respond to tamoxifen, in 30-40% of cases these tumours have metastatic disease within 5 years. This common clinical scenario precipitates cessation of the regime and the initiation of second line therapy. Uncovering the key mechanisms involved in the development of metastasis provide predictive markers of disease recurrence which will enable the tailoring of existing therapies and the development of new drugs in this class, to improve outcome in specific patient groups. Biomarkers of endocrine sensitivity in breast cancer patients -

Classic pathologic parameters, such as tumour size, grade, ER and HER2 status have been invaluable in informing the clinical management of breast cancer; however, a significant number of patients with a good prognostic profile will have a tumour recurrence. More recently, with the advent of gene expression profiling research, additional mechanisms to predict disease recurrence are now available. Technologies provided by Agendia (MammaPrint) and Genomic Health (OncotypeDx) which involve detection of a panel of genes performed by the providing companies have reported independent hazard ratios of 2.23 and 3.21 respectively. The predictive value of these tests is greater than any of the currently employed pathological parameters including ER and HER2 (hazard ratios 0.665 and 1.505, respectively). Elucidation of the mechanisms of how a tumour can adapt to endocrine therapy and consequently reoccur will provide mechanistically anchored predictive biomarkers, which can easily be detected and used in the clinic to select appropriate treatments.

New therapeutics for the treatment of metastatic disease

There are no targeted therapies currently available that act to specifically inhibit metastasis in breast cancer. Most patients are treated with chemotherapy or radiotherapy which does not specifically target cancer cells. Classic DNA-damaging cytotoxic drugs (eg. cyclophosphamide, doxorubicin, 5-fluorouracil) are still the mainstay of treatment regimens, and, although crude, are effective. Current directed therapies against metastatic cancers only inhibit angiogenesis. In doing so, they cut off nutrient supply and inhibit access of metastatic cells to the blood stream. The anti-angiogenic, Avastin (Roche) is used to treat a range of cancers, including colorectal, lung, and kidney cancer. Its use in metastatic breast cancer however is controversial. The FDA recently withdrew approval for its use in the metastatic setting and the European Commission has only approved it in combination with the chemotherapy drugs Paclitaxel or Xeloda. There is therefore a pressing need in terms of healthcare and economics to develop new directed therapies to treat metastatic breast cancer. It is an object of the invention to overcome at least one of the above-referenced problems. Statements of Invention

The Applicant has identified a marker of breast tumour metastasis, ADAM22 (SEQ ID NO: 2), that can be detected in patient tumour tissue and which functions as a prognostic variable of breast cancer metastatic disease (i.e. the presence of the marker independently correlates with an increased risk of metastasis, and reduced disease free survival, compared with an individual who is negative for the marker). The Applicant has also discovered that ADAM22 is involved in the development and pathology of a metastatic phenotype and that the protein can therefore be targeted in treatment of a metastatic phenotype of breast cancer.

Thus, in a first aspect, the invention relates to a method or assay of diagnosing metastatic potential of a breast cancer in an individual with breast cancer, the method comprising a step of assaying a sample (i.e. of breast tumour tissue) from the patient for expression of ADAM22, wherein expression of ADAM22 correlates with increased metastatic potential (or reduced disease free survival) compared with a patient who is ADAM22 negative.

In a second aspect, the invention relates to a method of treatment or prevention of metastatic breast cancer in a patient, the method comprising a step of administering to the individual a therapeutically effective amount of an ADAM22 inhibitor. Preferably, the ADAM22 inhibitor is selected from an oligonucleotide capable of attenuating the function of ADAM22, LGI1 protein (SEQ ID NO: 1) or a functional variant thereof, or a LGI1 peptide mimic.

In a third aspect, the invention relates to a method of treatment or prevention of metastatic breast cancer in a breast cancer patient, the method comprising a step of administering to the individual a therapeutically effective amount of an ADAM22 inhibitor, wherein the breast cancer patient is ADAM22 positive. Preferably, the ADAM22 inhibitor is selected from an oligonucleotide capable of attenuating the function or expression of ADAM22, LGI1 protein (SEQ ID NO: 1) or a functional variant thereof, or a LGI1 peptide mimic. The invention also relates to a kit for detection of ADAM22 as a companion diagnostic in the treatment/prevention of metastatic breast cancer by inhibition of ADAM22. Thus, in a fourth aspect, the invention relates to a kit comprising means for detecting whether a patient is ADAM22 positive or ADAM22 negative and a pharmaceutical composition comprising a therapeutically effective amount of an ADAM22 inhibitor. Preferably, the ADAM22 inhibitor is selected from an oligonucleotide capable of attenuating the function or expression of ADAM22, LGI1 protein (SEQ ID NO: 1) or a functional variant thereof, or a LGI1 peptide mimic.

The invention relates to a method of or assay for of identifying a breast cancer patient suitable for treatment of metastatic breast cancer with an ADAM22 inhibitor, the method comprising a step of assaying a sample (i.e. of breast tumour tissue) from the patient for expression of ADAM22, wherein expression of ADAM22 indicates that the patient is suitable for treatment with an ADAM22 inhibitor. The invention also relates to an ADAM22 inhibitor for use as a medicament. Preferably, the ADAM22 inhibitor is selected from an oligonucleotide capable of attenuating the function or expression of ADAM22, LGI1 protein (SEQ ID NO: 1) or a functional variant thereof, or a LGI1 peptide mimic. The invention also relates to a pharmaceutical composition comprising an ADAM22 inhibitor and a pharmaceutically acceptable carrier. Preferably, the ADAM22 inhibitor is selected from an oligonucleotide capable of attenuating the function or expression of ADAM22, LGI1 protein (SEQ ID NO: 1) or a functional variant thereof, or a LGI1 peptide mimic.

In the assays of the invention, the step of assaying a sample of breast tumour tissue from the patient for expression of ADAM22 is preferably at least partly performed with a non-human machine, for example a computer. The invention also provides a computer program comprising program instructions for causing a computer to perform the method of the invention.

The invention also relates to a system for obtaining data from at least one test sample obtained from at least one individual, the system comprising:

- a determination module configured to receive at least one test sample and perform at least one test analysis on the test sample to assay for the expression of ADAM22; optionally, a storage system for ADAM22 expression data generated by the determination system; and

a display module for displaying a content based in part on the data output from said determination module, wherein the content comprises a signal indicative of the expression of ADAM22.

The invention also relates to a system according to the invention for diagnosing metastatic potential of a breast cancer in an individual with breast cancer, in which the system comprises a correlation module for correlating ADAM22 expression data from the determination module with metastatic potential of breast cancer, wherein the expression of ADAM22 correlates with increased potential for metastasis compared with a patient who is ADAM22 negative, and wherein the display module displays a content based in part on the data from the correlation system, the content optionally comprising a signal indicative of the metastatic potential of the breast cancer.

The invention also relates to a system according to the invention for identifying a breast cancer patient suitable for treatment of metastatic breast cancer with an ADAM22 inhibitor, in which the system comprises a correlation module for correlating ADAM22 expression data from the determination module with suitability for treatment of metastatic breast cancer with an ADAM22 inhibitor, wherein the expression of ADAM22indicates that the patient is suitable for treatment with an ADAM22 inhibitor, and wherein the display module displays a content based in part on the data from the correlation system, the content optionally comprising a signal indicative of the suitability for treatment of metastatic breast cancer with an ADAM22 inhibitor. Typically, the determination system comprises an immunohistochemical detection apparatus.

The invention also relates to an assay for identifying an agent capable of treating or preventing metastasis of a breast cancer, the method comprising the step of providing a sample of endocrine-resistant breast cancer cells, incubating the sample with a test agent, and detecting binding between the test agent and an LGIl binding site of ADAM22 expressed by the cells, wherein a test agent that binds to the LGIl binding site of ADAM22 is an agent capable of treating or preventing metastasis of a breast cancer. Brief Description of the Figures

Figure 1 : ADAM22 promotes tumour progression in endocrine resistant breast cancer.

(A) A histogram representing the migration pattern of MCF7, LY2 and letrozole resistant (LetR) cells following transient transfection with scrambled (scr) siRNA or ADAM22 siRNA. Average migration is given as a ratio to scr-siRNA in MCF-7 cells. Results are expressed as mean + SD, n=3.

(B) Endocrine resistant cells (LY2 andLetR) have reduced cellular differentiation as exemplified by their inability to form acni in 3D culture in comparison to endocrine sensitive MCF-7 cells, this was restored by knockdown of ADAM22 with siRNA (n=3).

(C) Treatment of endocrine resistant LY2 and LetR cells with recombinant LGIl (5nM) significantly reduced cellular migration. There was no discernable effect of LGIl on migration of

MCF7 cells. Average migration is given as a ratio to MCF-7 cells. Results are expressed as mean + SD, n=3.

(D) Treatment of LetR cells with LGIl restored cellular differentiation (n=3).

(E) The LGIl mimetic (SEQ ID NO: 5) (5nM) inhibits cell migration of endocrine resistant LY2 and LetR cells.

(F) Toxicity study of LGIl peptide mimetic in vivo. LGIl mimetic was delivered by daily intraperitoneal injection to C57/bl6 mice at high (lOOug/kg/day), medium (lOug/kg/day) and low dose (lug/kg/day) for 30 days. No effect was seen on animal health or weight.

(G) Daily inject of LGIl inhibits cell proliferation and metastasis of endocrine resistant LetR cells in vivo. Detailed Description of the Invention

"ADAM22": This refers to any isoform of the transmembrane glycoprotein ADAM22 (A Disintegrin And Metalloproteinase 22) which has been identified as a SRC-1 transcriptional target specific to the resistant phenotype (Fig. 1A). The amino acid sequence of isoform 4 of the protein is provided in SEQ ID NO: 2:

Homo sapiens ADAM metallopeptidase domain 22 (ADAM22), transcript variant 4 Accession number: NM_004194 (SEQ ID NO: 2)

MQAAVAVSVPFLLLCVLGTCPPARCGQAGDASLMELEKRKENRFVERQSIVPLRLIY RS

GGEDESRHDALDTRVRGDLGGPQLTHVDQASFQVDAFGTSFILDVVLNHDLLSSEYI ER

HIEHGGKTVEVKGGEHCYYQGHIRGNPDSFVALSTCHGLHGMFYDGNHTYLIEPEEN DT

TQEDFHFHSVYKSRLFEFSLDDLPSEFQQVNITPSKFILKPRPKRSKRQLRRYPRNV EEET

KYIELMIVNDHLMFKKHRLSVVHTNTYAKSVVNMADLIYKDQLKTRIVLVAMETWAT

DNKFAISENPLITLREFMKYRRDFIKEKSDAVHLFSGSQFESSRSGAAYIGGICSLL KGGG

VNEFGKTDLMAVTLAQSLAHNIGIISDKRKLASGECKCEDTWSGCIMGDTGYYLPKK FT

QCNIEEYHDFLNSGGGACLFNKPSKLLDPPECGNGFIETGEECDCGTPAECVLEGAE CCK

KCTLTQDSQCSDGLCCKKCKFQPMGTVCREAVNDCDIRETCSGNSSQCAPNIHKMDG Y

SCDGVQGICFGGRCKTRDRQCKYIWGQKVTASDKYCYEKLNIEGTEKGNCGKDKDTW I

QCNKRDVLCGYLLCTNIGNIPRLGELDGEITSTLVVQQGRTLNCSGGHVKLEEDVDL GY

VEDGTPCGPQMMCLEHRCLPVASFNFSTCLSSKEGTICSGNGVCSNELKCVCNRHWI GS

DCNTYFPHNDDAKTGITLSGNGVAGTNIIIGIIAGTILVLALILGITAWGYKNYREQ RQLP

QGDYVKKPGDGDSFYSDIPPGVSTNSASSSKKRSNGLSHSWSERIPDTKHISDICEN GRPR

SNSWQGNLGGNKKKIRGKRFRPRSNSTE

The DNA sequence of the ADAM22-encoding genes (several isoform variants exist) include: NM_021723: ADAM22 (uc003uip.l) at chr7:87564071-87811339 - ADAM metallopeptidase domain 22 isoform 4

NM_016351: ADAM22 (ucQ03uio.2) at chr7:87563702-87826447 - ADAM metallopeptidase domain 22 isoform 3

NM_021723: ADAM22 (uc()03ujn.2) at chr7:87563702-87826447 - ADAM metallopeptidase domain 22 isoform 1

NM_021722: ADAM22 (uc003ujm.2) at chr7: 87563702-87826447 - ADAM metallopeptidase domain 22 isoform 2 NM_021721: ADAM22 (ucOOBujll) at chr7:87363702-8781 1428 - ADAM metallopeptidase domain 22 isoform 5

NM_004194: ADAM22 (uc003uik.l) at chr7:87563702-8781 1428 - ADAM metallopeptidase domain 22 isoform 4

NM_021721: ADAM22 (uc003uii.l) at chr7: 87563702-87762113 - ADAM metallopeptidase domain 22 isoform 5

NM_021721: ADAM22 fucCM uiU) at chr7: 87563702-87757991 - ADAM metallopeptidase domain 22 isoform 5 "Metastatic potential": This term should be understood to mean the potential of primary breast cancer to develop a metastatic phenotype leading to recurrence of the breast cancer.

"Expression of ADAM22": This term should be understood to mean positive detection of ADAM22 in breast cancer tumour tissue. Detection may be performed by means of full face tissue section or breast tumour sample tissue microarray (TMA) constructed and data recorded as described previously 1 . Tissue is immunostained using mouse anti ADAM22 (H00053616-B01).

Positive detection is taken to be an Alfred Score of greater than 3, as within the meaning of the assays of the invention . Other methods of detecting expression of ADAM22 will be apparent to those skilled in the art.

"ADAM22 negative": This term means that ADAM22 expression cannot be positively detected in the breast cancer tumour sample using the immunostained tissue described and referenced above. "ADAM22 positive": This term means that ADAM22 expression can be positively detected in the breast cancer tumour sample using the immunostained tissue described and referenced above.

"Treatment": This term means its generally accepted meaning which encompasses prohibiting, preventing, restraining, and slowing, stopping or reversing progression or severity of a metastatic breast cancer phenotype. "Breast cancer patient" or "patient": This term means a patient who has a primary breast cancer tumour and awaits treatment for the cancer or has already undergone or is undergoing treatment for the primary tumour. The term should also be understood to include a patient who has had a primary breast cancer and is in remission, for example remission following treatment including one or more of tumour resection, first line chemotherapy, or both. Usually, the patient will be a breast cancer patient who has, or is undergoing, treatment for a primary tumour and who has been identified as having potential for developing a metastatic phenotype. In one embodiment, the patient has an ER-positive breast cancer. "Administering": ADAM22 inhibitors, or pharmaceutical compositions of the invention, can be administered orally, parenterally, topically, rectally, nasally, buccally, via an implanted reservoir, or via inhalation spray. The term "parenteral" as used herein includes subcutaneous, intracutaneous, intravenous, intramuscular, intraarticular, intraarterial, intrasynovial, intrasternal, intrathecal, intralesional, and intracranial injection or infusion techniques. A sterile injectable composition, e.g., a sterile injectable aqueous or oleaginous suspension, can be formulated according to techniques known in the art using suitable dispersing or wetting agents (such as Tween 80) or suspending agents. The sterile injectable preparation can also be a sterile injectable solution or suspension in a non-toxic parenterally acceptable diluent or solvent, for example, as a solution in 1,3-butanediol. Among the acceptable vehicles and solvents that can be employed are mannitol, water, Ringer's solution and isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium (e.g., synthetic mono- or diglycerides). Fatty acids, such as oleic acid and its glyceride derivatives are useful in the preparation of injectables, as are natural pharmaceutically-acceptable oils, such as olive oil or castor oil, especially in their polyoxyethylated versions. These oil solutions or suspensions can also contain a long-chain alcohol diluent or dispersant, or carboxymethyl cellulose or similar dispersing agents. Other commonly used surfactants such as Tweens or Spans or other similar emulsifying agents or bioavailability enhancers which are commonly used in the manufacture of pharmaceutically acceptable solid, liquid, or other dosage forms can also be used for the purposes of formulation. A composition for oral administration can be any orally acceptable dosage form including, but not limited to, capsules, tablets, emulsions and aqueous suspensions, dispersions and solutions. In the case of tablets/capsules for oral use, carriers which are commonly used include lactose and corn starch. Lubricating agents, such as magnesium stearate, are also typically added. For oral administration in a capsule form, useful diluents include lactose and dried corn starch. When aqueous suspensions or emulsions are administered orally, the active ingredient can be suspended or dissolved in an oily phase combined with emulsifying or suspending agents. If desired, certain sweetening, flavoring, or coloring agents can be added. A nasal aerosol or inhalation composition can be prepared according to techniques well known in the art of pharmaceutical formulation.

"Therapeutically effective amount": This term refers to the amount or dose of the inhibitor or agent, upon single or multiple dose administration to the patient, which provides the desired effect in the patient under treatment. An effective amount can be readily determined by the attending diagnostician, as one skilled in the art, by the use of known techniques and by observing results obtained under analogous circumstances. In determining the effective amount or dose of inhibitor or agent administered, a number of factors are considered by the attending diagnostician, including, but not limited to: the species of mammal; its size, age, and general health; the degree of or involvement or the severity of the disease; the response of the individual patient; the particular inhibitor or agent administered; the mode of administration; the bioavailabilty characteristics of the preparation administered; the dose regimen selected; the use of concomitant medication; and other relevant circumstances. In particular, the term should be understood to mean a sufficient amount of ADAM22 inhibitor or agent to prevent or slow the development of a metastatic phenotype in a breast cancer patient.

"ADAM22 inhibitor": This term refers to a molecule capable of reducing cell migration and/or restoring differentiation in an endocrine resistant cell line selected from LY2 or LetR. The term should be understood to include an agent that attenuates the expression of ADAM22 protein (i.e. interferes with expression of the ADAM22 gene), including suppression of transcription or translation, and/or a molecule that directly inhibits ADAM22 activity, for example by binding to the ADAM22 protein (ADAM22 inhibitor ligand), especially the LGI1 binding site of ADAM22. Thus, in embodiment, the ADAM22 inhibitor is an ADAM22 ligand, typically capable of binding to the LGI1 binding site of ADAM22. The ADAM22 inhibitor ligand may be an ADAM22 antibody or peptide capable of binding to ADAM22 and inhibiting ADAM22 activity. Inhibitors which attenuate the expression of ADAM22 protein are known in the technical field and may comprise any of the group comprising siRNA (i.e. Ambion 4390824), miRNA, shRNA, antisense oligonucleotides, and ribozymes. Expression of ADAM22 may be suppressed by means of RNA interference (RNAi). RNA interference (RNAi) is an evolutionarily highly conserved process of post-transcriptional gene silencing (PTGS) by which double stranded RNA (known as siRNA molecules), when introduced into a cell, causes sequence-specific degradation of mRNA sequences. The RNAi machinery, once it finds a double-stranded RNA molecule, cuts it up, separates the two strands, and then proceeds to destroy RNA molecules that are complementary to one of those segments, or prevent their translation into proteins. Thus, suppression of ADAM22 expression may be achieved by treating an individual with siRNA molecules designed to target ADAM22 mRNA, preferably human ADAM22 and preferably a sequence in the human ADAM22 mRNA sense strand 1 . Five transcript variants of human ADAM22 mRNA are described , namely transcript variant 3, mRNA, NM_016351.4, transcript variant 2, mRNA, NM_021722.4, transcript variant 4, mRNA, NM_004194.3, transcript variant 1, mRNA, NM_021723.3, and transcript variant 5, mRNA, NM_021721.3.

Other types of gene knockdown tools will be well known to the person skilled in the field of molecular biology. For example, micro RNA's (miRNAs) are small (~22nt) non-coding RNAs (ncRNAs) that regulate gene expression at the level of translation. Each miRNA apparently regulates multiple genes and hundreds of miRNA genes are predicted to be present in mammals. Recently miRNAs have been found to be critical for development, cell proliferation and cell development, apoptosis and fat metabolism, and cell differentiation. Alternatively, small hairpin RNA (shRNA) molecules are short RNA molecules having a small hairpin loop in their tertiary structure that may be employed to silence genes. The design of miRNA or shRNA molecules capable of silencing ADAM22 will be apparent to those skilled in the field of miRNA or shRNA molecule design. As an alternative, the level of ADAM22 expression can be modulated using antisense or ribozyme approaches to inhibit or prevent translation of ADAM22 mRNA transcripts or triple helix approaches to inhibit transcription of the ADAM22 gene. Antisense approaches involve the design of oligonucleotides (either DNA or RNA) that are complementary to ADAM mRNA. The antisense oligonucleotides will bind to the complementary mRNA transcripts and prevent translation.

Proteins that bind to ADAM22 and inhibit ADAM22 activity are also known in the field, and include the neuronal protein and known ADAM22 ligand LGIl (SEQ ID NO: 1) and variants thereof described in the literature, for example LGI4 10 .

LGIl GenBank number: AAQ89244

(SEQ ID NO: \_

meserskrmg naciplkria yflcllsall ltegkkpakp kcpavctctk dnalcenars iprtvppdvi slsfvrsgft eisegsflft pslqlllfts nsfdvisdda figlphleyl fiennniksi srhtfrglks lihlslannn Iqtlpkdifk gldsltnvdl rgnsfncdck lkwlvewlgh tnatvediyc egppeykkrk inslsskdfd ciitefaksq dlpyqslsid tfsylndeyv viaqpftgkc iflewdhvek tfrnydnitg tstvvckpiv ietqlyviva qlfggshiyk rdsfankfik iqdieilkir kpndietfki ennwyfvvad sskagfttiy kwngngfysh qslhawyrdt dveyleivrt pqtlrtphli lssssqrpvi yqwnkatqlf tnqtdipnme dvyavkhfsv kgdvyicltr figdskvmkw ggssfqdiqr mpsrgsmvfq plqinnyqya ilgsdysftq vynwdaekak fvkfqelnvq aprsfthvsi nkrnflfass fkgntqiykh vivdlsa

LGIl serves as a specific extracellular ligand for ADAM22 . The ADAM22 binding domain of LGIl is known and consists of the sequence:

KGD V YICLTRFIGD S KVMKWGGS S FQDIQRMPSR (SEQ ID NO: 3). This domain of LGIl binds to ADAM22 (SEQ ID NO: 2) at the LGIl binding domain which is described in the literature (CREAVNDCDIRETCSGNSSQCAPNIHKMDGYSCD (SEQ ID NO: 4)). Thus, ADAM22 inhibitors include LGIl protein (and variants thereof capable of inhibiting migration of endocrine resistant breast cancer cells as determined in the motility assay described below) including LGI4 10 , and LGIl peptide mimics. As employed herein, the term "Inhibiting migration" should be understood as a significant reduction in migration, where significance is taken as p<0.05, as assessed using standard unpaired Student t-test'.

"LGIl peptide mimic": This term refers to a polymer composed of up to 100 amino acid monomers via peptide bond linkage, which is capable of mimicking the ADAM22 binding activity of LGIl by binding to an LGIl binding domain of ADAM22 and which is capable of inhibiting migration of endocrine resistant breast cancer cells as determined in the Motility Assay described below. The term includes poly amino acids comprising the ADAM22 binding domain of LGIl (SEQUENCE ID NO: 3), functional fragments of the ADAM22 binding domain of LGIl (SEQUENCE ID NO: 3), and polyamino acids comprising functional variants of SEQUENCE ID NO: 3. Methods of determining if a peptide is capable of mimicking the ADAM22 binding activity of LGIl will be apparent to a person skilled in the art, and include conventional peptide binding assay.

The term "in-vitro model of breast cancer cell migration" should be understood to mean the Motility Assay described below.

"Functional fragments of ADAM22 binding domain of LGIl": This term means a fragment of the ADAM22 binding domain of LGIl (SEQ ID NO: 3) having at least 15, preferably 20, amino acids, which is capable of binding to the LGIl binding domain of ADAM22 and capable of inhibiting migration of endocrine resistant breast cancer cells as determined in the Motility Assay described below. Examples of functional fragments include the following fragments of SEQ ID NO: 3:

KGDVYICLTRFIGDSKVMKWGG (SEQ ID NO: 5);

KGDVYISLTRFIGDSKVMKWGG (SEQ ID NO: 6);

D V YICLTRFIGD S KVMKWGGS S (SEQ ID NO: 7);

YICLTRFIGD S KVMKWGGS S FQ (SEQ ID NO: 8);

CLTRFIGDSKVMKWGGSSFQDI (SEQ ID NO: 9);

TRFIGDSKV MKWGGS SFQDIQR (SEQ ID NO: 10);

FIGDS KVMKWGGS S FQDIQRMP (SEQ ID NO: 11) ; and

GDSKVMKWGGSSFQDIQRMPSR_(SEQ ID NO: 12).

"Functional variants": This term means a variant of SEQUENCE ID NO: 3 (or a variant of a functional fragment) having at least 90% sequence homology with the SEQ ID NO: 3 or the fragment and which is capable of binding to the LGIl binding domain of ADAM22 and capable of inhibiting migration of endocrine resistant breast cancer cells as determined in the motility assay described below. It shall be taken to mean peptides having amino acid sequences which are substantially identical to the fragment, and include peptides that are altered in respect of one or more amino acid residues compared to the fragment. An example is SEQ ID NO: 6, which is a function variant of the fragment of SEQ ID NO: 5. Preferably such alterations involve the insertion, addition, deletion and/or substitution of 5 or fewer amino acids, more preferably of 4 or fewer, even more preferably of 3 or fewer, most preferably of 1 or 2 amino acids only. Insertion, addition and substitution with natural and modified amino acids are envisaged. The variant may have conservative amino acid changes, wherein the amino acid being introduced is similar structurally, chemically, or functionally to that being substituted. The variant will have at least 90% sequence homology, and ideally at least 95%, 96%, 97%, 98% or 99% sequence homology with the fragment. In this context, sequence homology comprises both sequence identity and similarity, i.e. a polypeptide sequence that shares 90% sequence homology with a specific LGI1 fragment is one in which any 90% of aligned residues are either identical to, or conservative substitutions of, the corresponding residues in the LGI1 fragment.

Peptides (including variants and fragments thereof) of and for use in the invention may be generated wholly or partly by chemical synthesis or by expression from nucleic acid. The peptides of and for use in the present invention can be readily prepared according to well- established, standard liquid or, preferably, solid-phase peptide synthesis methods known in the art (see, for example Young 4 ). When necessary, any of the LGI1 peptide mimics described herein can be chemically modified, for example to increase their stability. A chemically modified peptide (a peptide analog) includes any functional chemical equivalent of the peptide characterized by its increased stability and/or efficacy in vivo or in vitro in respect of the practice of the invention. The term peptide analog also refers to any amino acid derivative of a peptide as described herein. A peptide analog can be produced by procedures that include, but are not limited to, modifications to side chains, incorporation of unnatural amino acids and/or their derivatives during peptide synthesis and the use of cross-linkers and other methods that impose conformational constraint on the peptides or their analogs. Examples of side chain modifications include modification of amino groups, such as by reductive alkylation by reaction with an aldehyde followed by reduction with NaBH 4 ; amidation with methylacetimidate; acetylation with acetic anhydride; carbamylation of amino groups with cyanate; trinitrobenzylation of amino groups with 2, 4, 6, trinitrobenzene sulfonic acid (TNBS); alkylation of amino groups with succinic anhydride and tetrahydrophthalic anhydride; and pyridoxylation of lysine with pyridoxa-5'-phosphate followed by reduction with NABH 4 . The guanidino group of arginine residues may be modified by the formation of heterocyclic condensation products with reagents such as 2,3-butanedione, phenylglyoxal and glyoxal. The carboxyl group may be modified by carbodiimide activation via o-acylisourea formation followed by subsequent derivatization, for example, to a corresponding amide. Sulfhydryl groups may be modified by methods, such as carboxymethylation with iodoacetic acid or iodoacetamide; performic acid oxidation to cysteic acid; formation of mixed disulphides with other thiol compounds; reaction with maleimide; maleic anhydride or other substituted maleimide; formation of mercurial derivatives using 4- chloromercuribenzoate, 4-chloromercuriphenylsulfonic acid, phenylmercury chloride, 2- chloromercuric-4-nitrophenol and other mercurials; carbamylation with cyanate at alkaline pH. Tryptophan residues may be modified by, for example, oxidation with N-bromosuccinimide or alkylation of the indole ring with 2-hydroxy-5-nitrobenzyl bromide or sulphonyl halides. Tryosine residues may be altered by nitration with tetranitromethane to form a 3 -nitro tyrosine derivative. Modification of the imidazole ring of a histidine residue may be accomplished by alkylation with iodoacetic acid derivatives or N-carbethoxylation with diethylpyrocarbonate. Examples of incorporating unnatural amino acids and derivatives during peptide synthesis include, but are not limited to, use of norleucine, 4-amino butyric acid, 4-amino-3-hydroxy-5- phenylpentanoic acid, 6-aminohexanoic acid, t-butyl glycine, norvaline, phenylglycine, ornithine, sarcosine, 4-amino-3-hydroxy-6-methylheptanoic acid, 2-thienyl alanine and/or D-isomers of amino acids.

"Pharmaceutically acceptable carrier": Any of the breast cancer treatments described herein can be mixed with a pharmaceutically acceptable carrier to form a pharmaceutical composition. "Acceptable" means that the carrier must be compatible with the active ingredient of the composition (and preferably, capable of stabilizing the active ingredient) and not deleterious to the subject to be treated. Suitable carriers include microcrystalline cellulose, mannitol, glucose, defatted milk powder, polyvinylpyrrolidone, and starch, or a combination thereof.

Embodiments of the invention also provide for systems (and computer readable media for causing computer systems) to perform a method for diagnosing metastatic potential of a breast cancer in an individual or to perform a method for identifying a breast cancer patient suitable for treatment or prevention of metastatic breast cancer with an ADAM22 inhibitor.

Embodiments of the invention can be described through functional modules, which are defined by computer executable instructions recorded on computer readable media and which cause a computer to perform method steps when executed. The modules are segregated by function for the sake of clarity. However, it should be understood that the modules/systems need not correspond to discreet blocks of code and the described functions can be carried out by the execution of various code portions stored on various media and executed at various times. Furthermore, it should be appreciated that the modules may perform other functions, thus the modules are not limited to having any particular functions or set of functions. The computer readable storage media can be any available tangible media that can be accessed by a computer. Computer readable storage media includes volatile and nonvolatile, removable and non-removable tangible media implemented in any method or technology for storage of information such as computer readable instructions, data structures, program modules or other data. Computer readable storage media includes, but is not limited to, RAM (random access memory), ROM (read only memory), EPROM (erasable programmable read only memory), EEPROM (electrically erasable programmable read only memory), flash memory or other memory technology, CD-ROM (compact disc read only memory), DVDs (digital versatile disks) or other optical storage media, magnetic cassettes, magnetic tape, magnetic disk storage or other magnetic storage media, other types of volatile and non-volatile memory, and any other tangible medium which can be used to store the desired information and which can accessed by a computer including and any suitable combination of the foregoing.

Computer-readable data embodied on one or more computer-readable storage media may define instructions, for example, as part of one or more programs, that, as a result of being executed by a computer, instruct the computer to perform one or more of the functions described herein, and/or various embodiments, variations and combinations thereof. Such instructions may be written in any of a plurality of programming languages, for example, Java, J#, Visual Basic, C, C#, C++, Fortran, Pascal, Eiffel, Basic, COBOL assembly language, and the like, or any of a variety of combinations thereof. The computer-readable storage media on which such instructions are embodied may reside on one or more of the components of either of a system, or a computer readable storage medium described herein, may be distributed across one or more of such components.

The computer-readable storage media may be transportable such that the instructions stored thereon can be loaded onto any computer resource to implement the aspects of the present invention discussed herein. In addition, it should be appreciated that the instructions stored on the computer-readable medium, described above, are not limited to instructions embodied as part of an application program running on a host computer. Rather, the instructions may be embodied as any type of computer code (e.g., software or microcode) that can be employed to program a computer to implement aspects of the present invention. The computer executable instructions may be written in a suitable computer language or combination of several languages. Basic computational biology methods are known to those of ordinary skill in the art and are described in, for example, Setubal and Meidanis et al., Introduction to Computational Biology Methods (PWS Publishing Company, Boston, 1997); Salzberg, Searles, Kasif, (Ed.), Computational Methods in Molecular Biology, (Elsevier, Amsterdam, 1998); Rashidi and Buehler, Bioinformatics Basics: Application in Biological Science and Medicine (CRC Press, London, 2000) and Ouelette and Bzevanis Bioinformatics: A Practical Guide for Analysis of Gene and Proteins (Wiley & Sons, Inc., 2nd ed., 2001).

The functional modules of certain embodiments of the invention include at minimum a determination system, a storage device, optionally a comparison module, and a display module. The functional modules can be executed on one, or multiple, computers, or by using one, or multiple, computer networks. The determination system has computer executable instructions to provide e.g., ADA22 expression levels in computer readable form. The determination system, can comprise any system for assaying a breast cancer tumor sample for expression of ADAM22. Standard procedures such as immunohistochemistry, may be employed.

The information determined in the determination system can be read by the storage device. As used herein the "storage device" is intended to include any suitable computing or processing apparatus or other device configured or adapted for storing data or information. Examples of an electronic apparatus suitable for use with the present invention include a stand-alone computing apparatus, data telecommunications networks, including local area networks (LAN), wide area networks (WAN), Internet, Intranet, and Extranet, and local and distributed computer processing systems. Storage devices also include, but are not limited to: magnetic storage media, such as floppy discs, hard disc storage media, magnetic tape, optical storage media such as CD-ROM, DVD, electronic storage media such as RAM, ROM, EPROM, EEPROM and the like, general hard disks and hybrids of these categories such as magnetic/optical storage media. The storage device is adapted or configured for having recorded thereon nucleic acid sequence information. Such information may be provided in digital form that can be transmitted and read electronically, e.g., via the Internet, on diskette, via USB (universal serial bus) or via any other suitable mode of communication.

As used herein, "stored" refers to a process for encoding information on the storage device. Those skilled in the art can readily adopt any of the presently known methods for recording information on known media to generate manufactures comprising information relating to ADAM22 expression in a sample.

In one embodiment the reference data stored in the storage device to be read by the comparison module is compared.

The "comparison module" can use a variety of available software programs and formats for the comparison operative to compare ADAM22 expression information data determined in the determination system to reference samples and/or stored reference data. In one embodiment, the comparison module is configured to use pattern recognition techniques to compare information from one or more entries to one or more reference data patterns. The comparison module may be configured using existing commercially-available or freely-available software for comparing patterns, staining, and may be optimized for particular data comparisons that are conducted. The comparison module provides computer readable information related to the ADAM22 expression levels of the sample. The comparison module, or any other module of the invention, may include an operating system (e.g., UNIX) on which runs a relational database management system, a World Wide Web application, and a World Wide Web server. World Wide Web application includes the executable code necessary for generation of database language statements (e.g., Structured Query Language (SQL) statements). Generally, the executables will include embedded SQL statements. In addition, the World Wide Web application may include a configuration file which contains pointers and addresses to the various software entities that comprise the server as well as the various external and internal databases which must be accessed to service user requests. The Configuration file also directs requests for server resources to the appropriate hardware— as may be necessary should the server be distributed over two or more separate computers. In one embodiment, the World Wide Web server supports a TCP/IP protocol. Local networks such as this are sometimes referred to as "Intranets." An advantage of such Intranets is that they allow easy communication with public domain databases residing on the World Wide Web (e.g., the GenBank or Swiss Pro World Wide Web site). Thus, in a particular preferred embodiment of the present invention, users can directly access data (via Hypertext links for example) residing on Internet databases using a HTML interface provided by Web browsers and Web servers.

The comparison module provides a computer readable comparison result that can be processed in computer readable form by predefined criteria, or criteria defined by a user, to provide a content based in part on the comparison result that may be stored and output as requested by a user using a display module. In one embodiment of the invention, the content based on the comparison result or the determination system is displayed on a computer monitor. In one embodiment of the invention, the content based on the comparison result or determination system is displayed through printable media. The display module can be any suitable device configured to receive from a computer and display computer readable information to a user. Non-limiting examples include, for example, general-purpose computers such as those based on Intel PENTIUM-type processor, Motorola PowerPC, Sun UltraSPARC, Hewlett-Packard PA-RISC processors, any of a variety of processors available from Advanced Micro Devices (AMD) of Sunnyvale, California, or any other type of processor, visual display devices such as flat panel displays, cathode ray tubes and the like, as well as computer printers of various types.

In one embodiment, a World Wide Web browser is used for providing a user interface for display of the content based on the comparison result. It should be understood that other modules of the invention can be adapted to have a web browser interface. Through the Web browser, a user may construct requests for retrieving data from the comparison module. Thus, the user will typically point and click to user interface elements such as buttons, pull down menus, scroll bars and the like conventionally employed in graphical user interfaces.

The methods described herein therefore provide for systems (and computer readable media for causing computer systems) to perform methods as described in the Statements of Invention above, for example methods for diagnosing metastatic potential of a breast cancer in an individual or methods for identifying a breast cancer patient suitable for treatment or prevention of metastatic breast cancer with an ADAM22 inhibitor.

Systems and computer readable media described herein are merely illustrative embodiments of the invention for performing methods of diagnosis in an individual, and are not intended to limit the scope of the invention. Variations of the systems and computer readable media described herein are possible and are intended to fall within the scope of the invention.

The modules of the machine, or those used in the computer readable medium, may assume numerous configurations. For example, function may be provided on a single machine or distributed over multiple machines. Experimental The invention identifies the disintegrin ADAM22, as a prognostic biomarker of breast cancer metastatic disease. The invention also relates to the use of LGI1 (the natural ligand of ADAM22) or a peptide mimetic thereof, which is functional and can inhibit migration in breast cancer cells. The invention also relates to the use of LGI1 or a peptide mimetic of LGI1 as a potential therapy for the treatment or prevention of metastatic breast cancer optionally in combination with a companion diagnostic for ADAM22, providing directed personalised treatment for patients with recurrent disease.

Endocrine therapies, including tamoxifen and aromatase inhibitors, are first-line treatments for estrogen receptor-positive breast cancer. Although most patients initially respond, approximately 30% develop metastatic disease. There is no effective treatment for endocrine-related tumour metastasis, hence a social and economic need exists to identify patients likely to develop resistance and to discover new drug targets. Studies by our group and others have established that aberrant expression of the SRC-1, is central to the resistant phenotype and development of metastasis. The applicant used SRC-1 ChlP-Seq, expression arrays and bioinformatic analysis to generate a list of SRC-1 target genes representing novel potential druggable targets relevant to treating metastatic disease.

The transmembrane glycoprotein ADAM22 (A Disintegrin And Metalloproteinase 22) was identified as a SRC-1 transcriptional target specific to the resistant phenotype \ ADAM22 was found to be expressed in both endocrine resistant (LY2 and LetR) and insensitive (MDA-MB- 231) cells. SRC-1 can regulate ADAM22 in endocrine sensitive and resistant cells. Furthermore, it has been established that ADAM22 is a significant independent prognostic indicator of poor disease-free survival in breast cancer patients 1 . The Applicant has demonstrated that knockdown of ADAM22 with siRNA reduced cell migration and restored differentiation in endocrine resistant cell 1 (Fig 1A and B). Additionally, in xenograft studies, expression of ADAM22 was absent from both the 4-OHT treated and untreated sensitive tumours and from the untreated resistant tumours 1 . Data from these studies firmly implicate ADAM22 in the development of metastatic breast disease and suggest ADAM22 as a viable drug target for the treatment of endocrine resistant breast cancer. As ADAM22 expression has previously only been described in the central nervous system, a therapeutic directed against systemic ADAM22 has the potential to be highly specific for the breast and have low toxicity. The neuronal protein, LGIl serves as a specific extracellular ligand for ADAM22 4 . It functions as a tumour suppressor of glioblastoma and neuroblastoma and recently has been shown to impair proliferation in HeLa cells 5- " 8. The LGI1/ADAM22 ligand/receptor complex has been suggested as a therapeutic target for synaptic disorders. The Applicant has shown that treatment with recombinant LGIl reduced cellular migration and restored differentiation in endocrine resistant cells in a similar manner to that observed with knockdown of ADAM22 (Fig. 1C and D). A 22 amino acid peptide mimetic of LGIl with a simple hairpin structure against the active binding domain of ADAM22 has been developed. The Applicant demonstrates that this peptide mimetic of LGIl can reduce cell migration in metastatic and endocrine resistant breast cancer cells, in a similar manner to full length LGIl and siADAM22 (Fig. IE). In vivo studies have been undertaken to assess the efficacy of the LGIl mimetic in reducing breast cancer burden in endocrine resistant breast cancer. In preliminary studies the LGIl peptide was shown to be well tolerated with no toxicity observed (Fig IF). In a xenograft model of endocrine resistance, LGIl significantly reduced tumour burden (Fig 1G). These data demonstrate the potential of a mimetic of LGIl as a new directed therapy to treat endocrine related tumour metastasis with its receptor ADAM22 acting as a companion diagnostic. This has significant advantage over previous methods described of detecting metastatic disease 9 .

Motility Assay A. Cell Preparation

• For cell culture use Minimum Essential Medium-Eagle (EMEM; BioWhittaker, Product No. 12-611Q) containing thefoll owing supplements (=EMEM Complete Medium): 10% fetal calf serum (BioWhittaker, Product No. 14-503F), IX penicillin/streptomycin solution (BioWhittaker, Product No. 17-602E), 1% L-glutamine (BioWhittaker, Product No. 17-605E), 1% non-essential amino acids (BioWhittaker, Product No. 13-114E) and 1% sodium pyruvate (BioWhittaker, Product No. 13-115E).

• Split cells when they reach 60-80% confluency (every 2-3 days), diluting 1:5 to 1: 10.

• For cell motility, harvest cells with trypsin-versene mixture (BioWhittaker, Product No. 17- 161F) and dilute into EMEM Complete Medium (warmed to 37°C). For cells cultured in a T-75 flask, wash cells with 3 ml of trypsin. Add another 3 ml of trypsin and aspirate 2 ml. Allow cells to detach by incubating flask at 37 °C for 5 minutes and then add 9 ml of EMEM Complete Medium and centrifuge cells at 400 x g for 5 minutes at room temperature.

• Wash 3X with Serum-Free Culture Medium (warmed to 37°C) (see Procedure section).

· Adjust cell density to 10 4 cells/ml in EMEM serum-free medium and add 50 μΐ of the cell suspension to each well of a 96-well microplate (= 500 cells/well). Cell motility is stimulated with 50 μΐ of 10% fetal bovine serum (FBS) diluted in EMEM serum-free medium. See Procedure section.

• Incubate for desired time at 37 °C in 5% C02 (22 hours for MCF-7 derived cells)

B. Procedure

Note: Use 100 μΐ per well unless indicated otherwise. The protocol requires -2.5 hours post- compound incubation to perform.

1. Suspend the Blue Fluorescent Microspheres (1 tube per 96-well plate) by vortexing for 30 seconds. Centrifuge the microspheres for 1 minute at 20,000 x g. The highest speed on a standard microcentrifuge is generally sufficient.

2. Aspirate supernatant, add 0.5 ml IX Wash Buffer and completely suspend beads by vortexing on maximum speed for at least 30 seconds.

3. Centrifuge for 1 minute at 20,000 x g.

4. Aspirate supernatant, add 0.5 ml IX Wash Buffer and completely resuspend beads by vortexing on maximum speed forat least 30 seconds.

5. Transfer the Blue Fluorescent Microsphere solution to a tube containing 7.5 ml IX Wash Buffer, vortex on maximumspeed for 60 seconds and immediately add 75 μΐ to each well of a 96- well plate coated with the desired substrate (For MCF-7 derived cells, type 1 collagen).

6. Incubate plate for 1 hour at 37°C in the dark. 7. Wash plate five times with 200 μΐ of IX Wash Buffer, removing each wash by aspiration. Leave buffer from last wash in wells. Plates may be stored in this format at 4°C for up to 1 week.

8. Harvest L929 cells as described in Cell Preparation Section.

9. Centrifuge cells at 400 x g for 5 minutes at room temperature. Resuspend cells in 10 ml Serum-Free Culture Medium, pre-warmed to 37°C. Washing the cells in the absence of serum is essential to prevent stimulation of cell motility.

10. Repeat step 9 two more times and dilute cells to 104 cells/ml in Serum-Free Culture Medium. Use 6 ml of the cell suspension per plate.

11. Warm the Blue Fluorescent Microsphere plates to 37°C. Aspirate Wash Buffer from plates (as prepared in steps 1-7), and gently add 50 μΐ (5 x 102 cells) of the cell suspension to each well. Low cell seeding densities are important for minimizing overlapping of tracks. If test compound is known to prevent cell attachment, for best results pre-incubate cells at 37°C to let cells attach before motility stimulation/inhibition.

12. Add 50 μΐ of Serum-Free Culture Medium containing supplements for negative controls or 50 μΐ of 10% FBS in EMEM Serum-Free Medium for positive controls (for a final concentration of 5% serum) to each well. Serum is a stimulator of cell motility. When screening for cell motility antagonists, add test compounds before or during serum addition.

13. Incubate for desired time (e.g., 22 hours) at 37°C in 5% C02.

14. Add 200 μΐ of 5.5% warmed Fixation Solution to each well and incubate in fume hood at room temperature for 60 minutes. Do not remove medium before adding the Fixation Solution.

Using warm Fixation Solution is critical to maintaining cell integrity.

15. Aspirate Fixation Solution and wash plate three times with 100 μΐ of IX Wash Buffer.

16. Aspirate Wash Buffer and add 100 μΐ of IX Permeabilization Buffer and incubate for 15 minutes at room temperature.

17. Aspirate Permeabilization Buffer, add 100 μΐ Staining Solution and incubate for 30 minutes at room temperature.

18. Aspirate Staining Solution and wash wells three times with 100 μΐ IX Wash Buffer. Aspirate the last wash and fill wells with 200 μΐ IX Wash Buffer.

19. Seal plate and evaluate on the ArrayScan HCS Reader. Store sealed plates in the dark at 4°C. Inimiinohistochemistry methods

Immunohistochemisiry is the localization of antigens in tissue sections by the use of labelled antibodies as specific reagentsthrough antigen-antibody interactions that are visualized by a marker such as an enzyme or a fluorescent label. An unlabelled primary antibody is incubated on the tissue section, binding the antigen of interest. A biotinylated secondary antibody directed against the primary antibody is then applied. A strepavidinbiotin complex (ABC) which possesses biotin binding sites is then added, cross reacts with the biotin molecules on the secondary antibody, amplifying the signal intensity. A slide comprising the tissue section is evaluated by light microscopy using the Allred System described in Harvey et ul\ First, a proportion score was assigned, which represented the estimated proportion of tumour cells staining positive for ADAM 22 [3 (0, none; 1 , < 1/100; 2, 1/100 to 1/10; 3, 1/10 to 1/3; 4, 1/3 to 2/3; and 5, >2/3). Next, an intensity score was assigned which represented the average intensity of positive tumour cells (0, none; I , weak, 2, intermediate; and 3, strong). The proportion and intensity scores were then added to obtain a total score, which ranged from 0 to 8. A score of 3 or greater represents a positive detection within the meaning of the assays of the invention.

The invention pertains to a marker of breast tumour metastasis that can be detected in patient tissue that can also be used as a drug target. A routine immunohistochemical based test for ADAM22 will significantly predict the propensity of the tumour to metastasise and is a potential source of revenue. The test is typically performed on paraffin embedded patient tissue taken at the time of initial surgery for the removal of the tumour. A positive test result indicates poor disease free survival.

References:

1. McCartan D, Bolger J, Fagan A. et al. Global characterization of the SRC-1 transcriptome identifies ADAM22 as an ER-independent mediator of endocrine resistant breast cancer. Cancer Res.

2012;72:220-9

2. Dillon MF, Stafford AT, Kelly G, Redmond AM, Mcllroy M, Crotty TB, et al. Cyclooxygenase-2 predicts adverse effects of tamoxifen: a possible mechanism of role for nuclear HER2 in breast cancer patients. Endocr Relat Cancer.2008, 15: 745-53

Harvey JM, Clark GM, Osborne CK, Allred DC. Estrogen receptor status by immunohistochemistry is superior to the ligand-binding assay for predicting response to adjuvant endocrine therapy in breast cancer. J Clin Oncol. 1999, 17:1474-81. 3.Fukata Y, Adesnik H, Iwanaga T, Bredt DS, Nicoll RA, Fukata

M. Epilepsy-related ligand receptor complex LGI1 and ADAM22 regulate synaptic transmission. Science 2006; 313, 1792-95.

J. M. Stewart and J. D. Young, Solid Phase Peptide Synthesis, 2nd edition, Pierce Chemical Company, Rockford, Illinois (1984), in M. Bodanzsky and A. Bodanzsky, The Practice of Peptide Synthesis, Springer Verlag, New York (1984)

Gabellini N, Masola V, Quartesan S et al. Increased expression of LGI1 gene triggers growth inhibition and apoptosis of neuroblastoma cells. J Cell Physiol 2006; 207, 711-721.

Gabellini N, Masola V. Expression of LGI1 impairs proliferation and survival of HeLa cells. Int J Cell Biol 2009, 417197. 7.Kunapuli P, Kasyapa CS, Hawthorn L, Cowell JK. LGI1, a putative tumour metastasis suppressor gene, controls in vitro invasiveness and expression of matrix

metalloproteinase in glioma cells through the ERKl/2 pathway. J Biol Chem 2004; 279, 23151-23157. Kunapuli P, Kasyapa CS, Hawthorn L, Cowell JK. LGI1, a putative tumor metastasis suppressor gene, controls in vitro invasiveness and expression of matrix metalloproteinases in glioma cells through the ERKl/2 pathway. J Biol Chem. 2004 May 28;279(22):23151-7. Epub 2004 Mar 26.

Fukata Y, Adesnik H, Iwanaga T, Bredt DS, Nicoll RA, Fukata M. Epilepsy-related ligand/receptor complex LGI1 and ADAM22 regulate synaptic transmission. Science. 2006 Sep 22;313(5794):1792-5 US 2005/221376 Al (Young Paul E [US] ET AL) 6 October 2005 (2005-10-06).

Sagane K, Int J Biol Sci 2008; 4(6): 387-396