Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
MATERIALS AND METHODS FOR TREATING ESTROGEN RECEPTOR α (ER) POSITIVE CANCER
Document Type and Number:
WIPO Patent Application WO/2012/168720
Kind Code:
A1
Abstract:
Experiments described herein in which the genetic dependency on ADCK2 is shown in endocrine receptor positive cancer. Accordingly, the use of ACDK2 inhibitors for treating individuals having estrogen receptor α (ER) positive cancers, such as ER positive breast cancer, ovarian cancer, prostate cancer or endometrial cancer, are disclosed.

Inventors:
ASHWORTH ALAN (GB)
LORD CHRISTOPHER JAMES (GB)
BROUGH RACHEL (GB)
FRANKUM JESSICA (GB)
Application Number:
PCT/GB2012/051289
Publication Date:
December 13, 2012
Filing Date:
June 08, 2012
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
CANCER RES INST ROYAL (GB)
ASHWORTH ALAN (GB)
LORD CHRISTOPHER JAMES (GB)
BROUGH RACHEL (GB)
FRANKUM JESSICA (GB)
International Classes:
C12N15/113; A61K31/713; A61P35/00
Domestic Patent References:
WO2007117432A22007-10-18
WO2009102469A22009-08-20
WO2010088498A12010-08-05
WO2010130782A12010-11-18
WO1993011161A11993-06-10
WO1994013804A11994-06-23
WO1998050431A21998-11-12
WO2001029058A12001-04-26
WO1999032619A11999-07-01
Foreign References:
US20110059900A12011-03-10
EP0184187A21986-06-11
GB2188638A1987-10-07
EP0239400A21987-09-30
EP0120694A21984-10-03
EP0125023A11984-11-14
Other References:
BROUGH RACHEL ET AL.: "Functional viability profiles of breast cancer", CANCER DISCOVERY, vol. 1, no. 3, August 2011 (2011-08-01), pages 260 - 273 URL, XP002682730
BROUGH ET AL., CURRENT OPINION IN GENETICS & DEVELOPMENT, vol. 21, 2011, pages 34 - 41
WARD, E.S. ET AL., NATURE, vol. 341, 1989, pages 544 - 546
BIRD ET AL., SCIENCE, vol. 242, 1988, pages 423 - 426
HUSTON ET AL., PNAS USA, vol. 85, 1988, pages 5879 - 5883
HOLLIGER ET AL., P.N.A.S. USA, vol. 90, 1993, pages 6444 - 6448
REITER ET AL., NATURE BIOTECH, vol. 14, 1996, pages 1239 - 1245
HU ET AL., CANCER RES., vol. 56, 1996, pages 3055 - 3061
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
"Applied Biosystems 430A Users Manual", ABI INC.
PEYMAN; ULMAN, CHEMICAL REVIEWS, vol. 90, 1990, pages 543 - 584
CROOKE, ANN. REV. PHARMACOL. TOXICOL., vol. 32, 1992, pages 329 - 376
ANGELL; BAULCOMBE, THE EMBO JOURNAL, vol. 16, no. 12, 1997, pages 3675 - 3684
VOINNET; BAULCOMBE, NATURE, vol. 389, 1997, pages 553
FIRE ET AL., NATURE, vol. 391, 1998, pages 806 - 811
FIRE, TRENDS GENET., vol. 15, 1999, pages 358 - 363
SHARP, RNA INTERFERENCE, GENES DEV., vol. 15, 2001, pages 485 - 490
HAMMOND ET AL., NATURE REV. GENET., vol. 2, 2001, pages 110 - 1119
TUSCHL, CHEM. BIOCHEM., vol. 2, 2001, pages 239 - 245
HAMILTON ET AL., SCIENCE, vol. 286, 1999, pages 950 - 952
HAMMOND ET AL., NATURE, vol. 404, 2000, pages 293 - 296
ZAMORE ET AL., CELL, vol. 101, 2000, pages 25 - 33
BERNSTEIN, NATURE, vol. 409, 2001, pages 363 - 366
ELBASHIR ET AL., GENES DEV., vol. 15, 2001, pages 188 - 200
ELBASHIR ET AL., NATURE, vol. 411, 2001, pages 494 - 498
ZAMORE, NATURE STRUCTURAL BIOLOGY, vol. 8, no. 9, 2001, pages 746 - 750
KASHANI-SABET; SCANLON, CANCER GENE THERAPY, vol. 2, no. 3, 1995, pages 213 - 223
MERCOLA; COHEN, CANCER GENE THERAPY, vol. 2, no. 1, 1995, pages 47 - 59
JOHN ET AL., PLOS BIOLOGY, vol. 11, no. 2, 2004, pages 1862 - 1879
MYERS, NATURE BIOTECHNOLOGY, vol. 21, 2003, pages 324 - 328
SHINAGAWA ET AL., GENES AND DEV., vol. 17, 2003, pages 1340 - 5
"Remington's Pharmaceutical Sciences", 2000, LIPPINCOTT, WILLIAMS & WILKINS
ASHWORTH A; LORD CJ; REIS-FILHO JS: "Genetic interactions in cancer progression and treatment", CELL, vol. 145, 2011, pages 30 - 8
BASELGA J; SWAIN SM: "Novel anticancer targets: revisiting ERBB2 and discovering ERBB3", NAT REV CANCER, vol. 9, 2009, pages 463 - 75, XP009139034, DOI: doi:10.1038/nrc2656
IORNS E; LORD CJ; GRIGORIADIS A; MCDONALD S; FENWICK K; MACKAY A ET AL.: "Integrated functional, gene expression and genomic analysis for the identification of cancer targets", PLOS ONE, vol. 4, 2009, pages E5120, XP002637299, DOI: doi:10.1371/journal.pone.0005120
NEVE RM; CHIN K; FRIDLYAND J; YEH J; BAEHNER FL; FEVR T ET AL.: "A collection of breast cancer cell lines for the study of functionally distinct cancer subtypes", CANCER CELL, vol. 10, 2006, pages 515 - 27, XP002580459, DOI: doi:10.1016/J.CCR.2006.10.008
BOUTROS M; BRAS LP; HUBER W: "Analysis of cell-based RNAi screens", GENOME BIOL, vol. 7, 2006, pages R66, XP021021269, DOI: doi:10.1186/gb-2006-7-7-r66
ZHAO L; VOGT PK: "Class I PI3K in oncogenic cellular transformation", ONCOGENE, vol. 27, 2008, pages 5486 - 96
STEMKE-HALE K; GONZALEZ-ANGULO AM; LLUCH A; NEVE RM; KUO WL; DAVIES M ET AL.: "An integrative genomic and proteomic analysis of PIK3CA, PTEN, and AKT mutations in breast cancer", CANCER RES, vol. 68, 2008, pages 6084 - 91, XP002534389, DOI: doi:10.1158/0008-5472.CAN-07-6854
MANNING BD; CANTLEY LC: "AKT/PKB signaling: navigating downstream", CELL, vol. 129, 2007, pages 1261 - 74
ZHAO L; VOGT PK: "Helical domain and kinase domain mutations in pll0alpha of phosphatidylinositol 3-kinase induce gain of function by different mechanisms", PROC NATL ACAD SCI U S A, vol. 105, 2008, pages 2652 - 7, XP002687648, DOI: doi:10.1073/pnas.0712169105
PANG H; FLINN R; PATSIALOU A; WYCKOFF J; ROUSSOS ET; WU H ET AL.: "Differential enhancement of breast cancer cell motility and metastasis by helical and kinase domain mutations of class IA phosphoinositide 3-kinase", CANCER RES, vol. 69, 2009, pages 8868 - 76, XP002687645, DOI: doi:10.1158/0008-5472.CAN-09-1968
NATRAJAN R; WEIGELT B; MACKAY A; GEYER FC; GRIGORIADIS A; TAN DS ET AL.: "An integrative genomic and transcriptomic analysis reveals molecular pathways and networks regulated by copy number aberrations in basal-like, HER2 and luminal cancers", BREAST CANCER RES TREAT, vol. 121, 2010, pages 575 - 89, XP019814351
SANTARIUS T; SHIPLEY J; BREWER D; STRATTON MR; COOPER CS: "A census of amplified and overexpressed human cancer genes", NAT REV CANCER, vol. 10, 2010, pages 59 - 64
SORLIE T; PEROU CM; TIBSHIRANI R; AAS T; GEISLER S; JOHNSEN H ET AL.: "Gene expression patterns of breast carcinomas distinguish tumor subclasses with clinical implications", PROC NATL ACAD SCI U S A, vol. 98, 2001, pages 10869 - 74, XP002215483, DOI: doi:10.1073/pnas.191367098
HOLLESTELLE A; NAGEL JH; SMID M; LAM S; ELSTRODT F; WASIELEWSKI M ET AL.: "Distinct gene mutation profiles among luminal-type and basal-type breast cancer cell lines", BREAST CANCER RES TREAT, vol. 121, pages 53 - 64, XP019787462
HEWITT L; TIGHE A; SANTAGUIDA S; WHITE AM; JONES CD; MUSACCHIO A ET AL.: "Sustained Mpsl activity is required in mitosis to recruit 0-Mad2 to the Madl-C-Mad2 core complex", J CELL BIOL, vol. 190, 2010, pages 25 - 34
LEE C; KIM JS; WALDMAN T: "PTEN gene targeting reveals a radiation-induced size checkpoint in human cancer cells", CANCER RES, vol. 64, 2004, pages 6906 - 14
PUC J; PARSONS R: "PTEN loss inhibits CHK1 to cause double stranded-DNA breaks in cells", CELL CYCLE, vol. 4, 2005, pages 927 - 9
SHEN WH; BALAJEE AS; WANG J; WU H; ENG C; PANDOLFI PP ET AL.: "Essential role for nuclear PTEN in maintaining chromosomal integrity", CELL, vol. 128, 2007, pages 157 - 70
KWIATKOWSKI N; JELLUMA N; FILIPPAKOPOULOS P; SOUNDARARAJAN M; MANAK MS; KWON M ET AL.: "Small-molecule kinase inhibitors provide insight into Mpsl cell cycle function", NAT CHEM BIOL, vol. 6, 2010, pages 359 - 68, XP055281846, DOI: doi:10.1038/nchembio.345
LAGIER-TOURENNE C; TAZIR M; LOPEZ LC; QUINZII CM; ASSOUM M; DROUOT N ET AL.: "ADCK3, an ancestral kinase, is mutated in a form of recessive ataxia associated with coenzyme Q10 deficiency", AM J HUM GENET, vol. 82, 2008, pages 661 - 72
SOTIRIOU C; PUSZTAI L: "Gene-expression signatures in breast cancer", N ENGL J MED, vol. 360, 2009, pages 790 - 800
WANG Y; KLIJN JG; ZHANG Y; SIEUWERTS AM; LOOK MP; YANG F ET AL.: "Geneexpression profiles to predict distant metastasis of lymph-node-negative primary breast cancer", LANCET, vol. 365, 2005, pages 671 - 9
SIMS D; BURSTEINAS B; GAO Q; JAIN E; MACKAY A; MITSOPOULOS C ET AL.: "ROCK: a breast cancer functional genomics resource", BREAST CANCER RES TREAT, 2010
ZHANG JH; CHUNG TD; OLDENBURG KR: "A Simple Statistical Parameter for Use in Evaluation and Validation of High Throughput Screening Assays", J BIOMOL SCREEN, vol. 4, 1999, pages 67 - 73, XP001145867, DOI: doi:10.1177/108705719900400206
IORNS E; TURNER NC; ELLIOTT R; SYED N; GARRONE 0; GASCO M ET AL.: "Identification of CDK10 as an important determinant of resistance to endocrine therapy for breast cancer", CANCER CELL, vol. 13, 2008, pages 91 - 104, XP002520756, DOI: doi:10.1016/J.CCR.2008.01.001
TURNER NC; LORD CJ; IORNS E; BROUGH R; SWIFT S; ELLIOTT R ET AL.: "A synthetic lethal siRNA screen identifying genes mediating sensitivity to a PARP inhibitor", EMBO J, vol. 27, 2008, pages 1368 - 77, XP002498071, DOI: doi:10.1038/emboj.2008.61
FARMER H; MCCABE N; LORD CJ; TUTT AN; JOHNSON DA; RICHARDSON TB ET AL.: "Targeting the DNA repair defect in BRCA mutant cells as a therapeutic strategy", NATURE, vol. 434, 2005, pages 917 - 21
NATRAJAN R; WEIGELT B; MACKAY A; GEYER FC; GRIGORIADIS A; TAN DS ET AL.: "An integrative genomic and transcriptomic analysis reveals molecular pathways and networks regulated by copy number aberrations in basal-like, HER2 and luminal cancers", BREAST CANCER RES TREAT, vol. 121, pages 575 - 89, XP019814351
GEYER FC; WEIGELT B; NATRAJAN R; LAMBROS MB; DE BIASE D; VATCHEVA R ET AL.: "Molecular analysis reveals a genetic basis for the phenotypic diversity of metaplastic breast carcinomas", J PATHOL, vol. 220, pages 562 - 73
HOCHBERG Y; BENJAMINI Y: "More powerful procedures for multiple significance testing", STAT MED, vol. 9, 1990, pages 811 - 8
NATRAJAN R; LAMBROS MB; RODRIGUEZ-PINILLA SM; MORENO-BUENO G; TAN DS; MARCHIO C ET AL.: "Tiling path genomic profiling of grade 3 invasive ductal breast cancers", CLIN CANCER RES, vol. 15, 2009, pages 2711 - 22
Attorney, Agent or Firm:
KIDDLE, Simon J. et al. (33 Gutter Lane, London Greater London EC2V 8AS, GB)
Download PDF:
Claims:
Claims :

1. An ACDK2 inhibitor for use in a method of treating an individual having estrogen receptor a (ER) positive cancer. 2. The ACDK2 inhibitor for use in a method of treatment according to claim 1, wherein ER positive cancer is ER positive breast cancer, ovarian cancer, prostate cancer or endometrial cancer . 3. The ACDK2 inhibitor for use in a method of treatment according to claim 2, wherein the breast cancer is luminal A subgroup of estrogen receptor a (ERa) positive breast cancer.

4. The ACDK2 inhibitor for use in a method of treatment according to any one of claims 1 to 3, wherein the inhibitor is a nucleic acid inhibitor, an antibody, a small molecule or a peptide .

5. The ACDK2 inhibitor for use in a method of treatment according to claim 4, wherein the nucleic acid inhibitor is a RNAi molecule or a siRNA molecule or an shRNA molecule.

6. The ACDK2 inhibitor for use in a method of treatment according to any one of the preceding claims, wherein treatment with an ADCK2 inhibitor is combined with a further anti-cancer therapy .

7. The ACDK2 inhibitor for use in a method of treatment according to claim 6, wherein treatment with an ADCK2 inhibitor is used in conjunction with an endocrine therapy such as

treatment with tamoxifen, ICI182870, faslodex and/or an aromatase inhibitor, radiotherapy and/or chemotherapy.

8. The ACDK2 inhibitor for use in a method of treatment according to claim 6, wherein treatment with an ADCK2 inhibitor is used in conjunction with a further chemotherapeutic agent.

9. The ACDK2 inhibitor for use in a method of treatment according to claim 8, wherein the further chemotherapeutic agent is Amsacrine (Amsidine) , Bleomycin, Busulfan, Capecitabine

(Xeloda) , Carboplatin, Carmustine (BCNU) , Chlorambucil (Leukeran) , Cisplatin, Cladribine (Leustat) , Clofarabine (Evoltra) ,

Crisantaspase (Erwinase) , Cyclophosphamide, Cytarabine (ARA-C) , Dacarbazine (DTIC) , Dactinomycin (Actinomycin D) , Daunorubicin, Docetaxel (Taxotere) , Doxorubicin, Epirubicin, Etoposide

(Vepesid, VP-16), Fludarabine (Fludara) , Fluorouracil (5-FU) , Gemcitabine (Gemzar) , Hydroxyurea (Hydroxycarbamide, Hydrea) ,

Idarubicin (Zavedos) . Ifosfamide (Mitoxana) , Irinotecan (CPT-11, Campto) , Leucovorin (folinic acid) , Liposomal doxorubicin

(Caelyx, Myocet) , Liposomal daunorubicin (DaunoXome®) Lomustine, Melphalan, Mercaptopurine, Mesna, Methotrexate, Mitomycin,

Mitoxantrone , Oxaliplatin (Eloxatin) , Paclitaxel (Taxol) ,

Pemetrexed (Alimta) , Pentostatin (Nipent) , Procarbazine,

Raltitrexed (Tomudex®) , Streptozocin (Zanosar®), Tegafur-uracil (Uftoral), Temozolomide (Temodal), Teniposide (Vumon) , Thiotepa, Tioguanine (6-TG) (Lanvis) , Topotecan (Hycamtin) , Treosulfan, Vinblastine (Velbe) , Vincristine (Oncovin) , Vindesine (Eldisine) or Vinorelbine (Navelbine) .

10. The ACDK2 inhibitor for use in a method of treatment according to claim 8 or claim 9, wherein the further

chemotherapeutic agent is 5FU, a BCL-XL inhibitor, a BCR-ABL, cKIT or PDGFR inhibitor, a CDK inhibitor, a CHK inhibitor, a COX2 inhibitor, a EGFR inhibitor, a HER2 targeted agent, a HSP inhibitor, a hTERT inhibitors, an IDO inhibitor, a MDM2 targeted agent, Methotrexate, a mTOR inhibitor, a PARP inhibitor, a PI3K inhibitor, a platinum salt, a proteasome inhibitor, a RAR or RXR inhibitor, a SRC inhibitor, a TGFB2 inhibitor, a topoisomerase inhibitor, a VEGF, RAF, cKIT or PDGFR inhibitor, or a SMC mimetic . 11. The ACDK2 inhibitor for use in a method of treatment according to any one of the preceding claims, wherein the method comprises the step of determining whether the individual has ER positive cancer.

12. The ACDK2 inhibitor for use in a method of treatment according to any one of the preceding claims, wherein determining whether the individual has ER positive cancer comprises

determining ER protein expression in a sample obtained from the individual .

13. The ACDK2 inhibitor for use in a method of treatment according to claim 12, wherein determining ER protein expression comprises one or more of determining ER protein expression in a tumour sample using immunohistochemistry, determining ER protein expression comprises measuring ER protein levels in a cell lysate by ELISA or Western blotting, and/or determining ER protein expression comprises using a binding agent capable of

specifically binding to the ER protein, or a fragment thereof.

14. The ACDK2 inhibitor for use in a method of treatment according to any one of the preceding claims, wherein determining whether the individual has ER positive cancer is performed on genomic nucleic acid extracted from a sample of cells obtained from the breast cancer or from a sample of cancer cells

circulating in blood.

15. The ACDK2 inhibitor for use in a method of treatment according to claim 14, wherein determining the expression of the ER gene comprises extracting RNA from a sample of cancer cells and measuring expression by real time PCR and/or by using a probe capable of hybridising to ER RNA.

16. The ACDK2 inhibitor for use in a method of treatment according to claim 15, wherein the probe is immobilised in a microarray .

17. The ACDK2 inhibitor for use in a method of treatment according any one of the preceding claims, wherein the method comprises initial step of obtaining a sample from said individual .

18. The ACDK2 inhibitor for use in a method of treatment according to any one of the preceding claims, wherein the sample is a tumour sample, a blood sample, a tissue sample or a cell sample.

19. Use of an inhibitor of ADCK2 in the preparation of a medicament for the treatment of an individual having estrogen receptor (ER) positive breast cancer. 20. A method of treating an individual having estrogen receptor a (ER) positive breast cancer, the method comprising

administering to the individual a therapeutically effective amount of an ADCK2 inhibitor. 21. A method of screening for agents useful in the treatment of estrogen receptor a (ER) positive breast cancer, the method comprising :

(a) contacting ADCK2 at least one candidate agent;

(b) determining an effect of the at least one candidate agent on an activity of the ADCK2; and

(c) selecting a candidate agent that inhibits the activity of the ADCK2.

22. The method according to claim 21, further comprising the step of contacting a candidate agent selected in step (c) with an estrogen receptor (ER) positive cancer cell line to determine whether the candidate agent is cytotoxic to the cancer cell line.

23. The method according to claim 21 or claim 22, wherein the candidate agent is a candidate nucleic acid inhibitor, a

candidate antibody or a candidate small molecule or a candidate peptide .

24. The method according to any one of claims 21 to 23, wherein the candidate agent is a compound that is part of a compound library .

25. The method according to claim 24, wherein the candidate compound has a molecular weight of less than 100 Da.

26. The method according to any one of claims 21 to 25, wherein the candidate agent or candidate compounds is a drug approved for use in the treatment of (breast) cancer.

27. The method according to any one of claims 21 to 26, further comprising determining whether a candidate agent is not lethal on normal cells.

28. The method according to any one of claims 21 to 27, which comprises determining the effect of combinations of two or more candidate compounds on the cell lines or protein targets.

28. A method which comprises having identified a candidate agent useful for the treatment of estrogen receptor (ER) positive breast cancer according to a method of any one of claims 21 to 28, the further step of manufacturing the compound in bulk and/or formulating the agent in a pharmaceutical composition.

Description:
Materials and Methods for Treating Estrogen Receptor a (ER)

Positive Cancer

Field of the Invention

The present invention relates to materials and methods for treating estrogen receptor a (ER) positive cancer, and in particular ER positive breast cancer.

Background of the Invention

Central to the design of novel therapeutic strategies for cancer is the identification of genes that are critical to the survival of tumor cells but which are largely redundant in normal cells. Correlating molecular changes with tumorigenesis has provided one route to the identification of potential drug targets and provides the rationale behind efforts to characterise genetic variation and gene expression in tumors. However, the

correlative nature of these data means that it is frequently not possible to determine whether the observations are causative or merely an effect of the disease state. For example, tumor cells generally exhibit anywhere between 10 4 and 10 5 genetic changes compared to germline DNA but theoretical estimates suggest that only a few of these mutations (probably less than 10) are critical drivers of tumor formation and survival (reviewed in (1) ) ·

Brough et al . (Current Opinion in Genetics & Development 2011, 21:34-41) describe how the biological concept of synthetic sickness/lethality (SSL) may be used to identify new therapeutic approaches and targets in models from yeast through to human cells. It is known to target genes that are overexpressed in particular forms of cancer. However, despite these successes, developing drugs that selectively kill cancer cells without harming normal cells remains a considerable challenge. This process is particularly problematic when considering how to target a protein such as a dysfunctional tumour suppressor that is either largely inactive or even completely absent. If one gene in an SSL relationship is a tumour suppressor gene, then its synthetic lethal partner becomes a candidate therapeutic target that could be used in tumour eelIs with a defined tumour

suppressor gene dysfunction. SS L can occur between genes acting in the same biochemical pathway or in distinct but compensatory pathways .

In breast cancer, defining the critical genes involved in tumor cell survival can in some cases lead to the development of novel therapeutic approaches to the disease, the most notable recent example being the development of agents such as trastuzumab and lapatinib, that target the reliance of some breast tumors upon the oncogene ERBB2 (2) . However, despite the wealth of

molecular, genetic and histological characterization of breast tumors and cell line models, our understanding of the genetic dependencies in this disease is relatively poor and there is accordingly a need in the art for new therapeutic strategies, in particular those which target genetic dependencies of different forms of cancer.

Summary of the Invention

Broadly, the present invention is based on work which attempts to comprehensively define the genetic dependencies for a set of potentially "druggable" genes in a wide range of breast tumor cell line models. In doing so, we not only reaffirm the impact of Pl3-kinase and ERBB2 signalling in the disease but importantly identify novel genetic dependencies for genetically defined subsets of the disease. In particular, the present invention is based on the experiments described herein in which the genetic dependency on ADCK2 is shown in endocrine receptor positive cancer .

In the present invention, references to ADCK2 denote aarF domain containing protein kinase 2 having the HGNC ID: 19039. The HUGO Gene Symbol report for ESR1 can be found at:

http://www.genenames.org/data/hgnc data . php?hgnc id=19039 which provides links to the ADCK2 nucleic acid and amino acid sequences, as well as reference to the homologous murine and rat proteins . In the present invention, references to estrogen receptor (ER) denotes the receptor also known as estrogen receptor 1 ("ESR1") and estrogen receptor a having the HGNC ID HGNC:3467. These terms are used interchangeably herein. The HUGO Gene Symbol report for ESR1 can be found at:

http : / /ww . genenames . org/data/hgnc data . php?hgnc id=3467

which provides links to the ESR1 nucleic acid and amino acid sequences, as well as reference to the homologous murine and rat proteins. The expression of ERa has been implicated in breast cancer, ovarian cancer, colon cancer, prostate cancer and endometrial cancer.

According, in a first aspect the present invention provides an ACDK2 inhibitor for use in a method of treating an individual having estrogen receptor (ER) positive cancer, and in particular for the treatment of ER positive breast cancer.

In a further aspect, the present invention provides the use of an inhibitor of ADCK2 in the preparation of a medicament for the treatment of an individual having estrogen receptor (ER) positive breast cancer.

In a further aspect, the present invention provides a method of treating an individual having estrogen receptor (ER) positive breast cancer, the method comprising administering to the individual a therapeutically effective amount of an ADCK2 inhibitor .

In a further aspect, the present invention provides a method of screening for agents useful in the treatment of estrogen receptor (ER) positive breast cancer, the method comprising:

(a) contacting ADCK2 at least one candidate agent;

(b) determining an effect of the at least one candidate agent on an activity of the ADCK2; and

(c) selecting a candidate agent that inhibits the activity of the ADCK2. In some aspects, the present invention provides a method which comprises having identified a candidate agent useful for the treatment of estrogen receptor (ER) positive breast cancer according to a method as described herein, the further step of manufacturing the compound in bulk and/or formulating the agent in a pharmaceutical composition.

The full names and database accession information for the preferred target genes that can be inhibited to cause a synthetic lethal effect in estrogen receptor a (ERa) positive breast cancer cells .

The present invention includes the combination of the aspects and preferred features described except where such a combination is clearly impermissible or is stated to be expressly avoided.

Embodiments of the present invention will now be described by way of example and not limitation with reference to the accompanying figures . Brief Description of the Figures

Figure 1. PIK3CA and ERBB2 oncogene addiction effects in breast cancer confirmed by functional viability profiling, a. Heat map showing the results of a supervised clustering of siRNA Z scores. Breast tumour cell lines were clustered according to PIK3CA gene mutation status and differential effects between PIK3CA mutant and wild type groups identified using the median permutation test. Statistically significant effects (p<0.05) are shown. For each siRNA, p values and the p<0.05 threshold are shown to the right of the heat map. siRNA targeting PIK3CA is marked by the arrow, b. Waterfall and box/whiskers plots of PIK3CA siRNA Z scores across the breast tumour cell line panel. In the

box/whiskers plot, *p = 0.014 between PIK3CA mutant and wild type groups using the median permutation test. c. Box/whiskers plots of AKT2 and AKT3 siRNA Z scores in PIK3CA mutant and wild type models (*p= 0.047 and p 0.003 respectively using the median permutation test) . d. Heat map showing the results of a supervised clustering of siRNA Z scores according to ERBB2 amplification status in the breast tumour cell line panel.

Statistically significant effects (p<0.05, median permutation test) are shown. ERBB2 and PIK3CA siRNA are marked by arrows, e. Waterfall and box/whiskers plots of ERBB2 siRNA Z scores across the breast tumour cell line panel. In the box/whiskers plot, *p = 0.003 between ERBB2 amplified and non-amplified groups using the median permutation test. Figure 2. Genetic dependency of ER+ve breast tumour cells upon

ADCK2. a. Heat map showing the results of a supervised clustering of siRNA Z scores. Breast tumour cell lines were clustered according to ER status and differential effects between ER positive and negative groups identified using the median

permutation test. Statistically significant effects (p<0.05) are shown. For each siRNA, p values and the p<0.05 threshold are shown to the right of the heat map. siRNA targeting ADCK2 is marked by the arrow, b. Waterfall and box/whiskers plots of ADCK2 siRNA Z scores across the breast tumour cell line panel. In the box/whiskers plot, *p = 0.004 between ER+ve and ER-ve groups using the median permutation test. c. ADCK2 silencing by multiple ADCK2 siRNA species reduces the expression of ER-target gene as shown by western blotting of MCF7 total cell lysates generated after siRNA transfection . d. Western blot of total cell lysates from MCF7 cells treated with either estradiol (E2) or faslodex

(FAS, ICI182780) . An E2-dependent increase in ADCK2 expression is shown, e. Western blot of ERa immunoprecitated material from MCF7 cells, indicating an ERa- ADCK2 interaction, f. Tumour ADCK2 mRNA expression in ER+ve breast tumours classified according to luminal A or B status. Transcript expression data from two independent studies was used 10,21 and luminal A or B status determined according to Sorlie et al 12. p values were calculated using Student's t test. Detailed Description

Inhibitors

Compounds which may be employed or screened for use in the present invention for treating estrogen receptor (ER) positive breast cancer. Some inhibitors of these polypeptides are known and further examples may be found by the application of screening technologies to these targets.

Small molecule inhibitors

In one aspect of the present invention, ADCK2 inhibitors may be identified by the use of high throughput screening strategies Accordingly, the present invention also extends to the use of small molecule inhibitors found in the screening disclosed herein and to Derivatives which are compounds of similar structure and functionality to the compounds found in the high throughput screen, but with one or more modifications, are expected to have similar physiological effects to these compounds and could therefore also be of use in the treatment of estrogen receptor (ER) positive breast cancer. The screening methods of the invention may be used to screen libraries of such derivatives to optimise their activity, if necessary.

Derivatives may be designed, based on a lead compound, by modifying one or more substituents or functional groups compared to the lead compound, for example by replacing these with alternative substituents or groups which are expected to have the same or improved physiological effect. The use of derivatives having such modifications is well known to those in the art.

Antibodies

Antibodies may be employed in the present invention as an example of a class of inhibitor useful for treating estrogen receptor

(ER) positive breast cancer, and more particularly as inhibitors of Uncharacterized aarF domain containing protein kinase 2

(ADCK2) . They may also be used in the methods disclosed herein for assessing an individual having cancer or predicting the response of an individual having cancer, in particular for determining whether the individual has estrogen receptor (ER) positive breast cancer that might be treatable according to the present invention. As used herein, the term "antibody" includes an immunoglobulin whether natural or partly or wholly synthetically produced. The term also covers any polypeptide or protein comprising an antibody binding domain. Antibody fragments which comprise an antigen binding domain are such as Fab, scFv, Fv, dAb, Fd; and diabodies. It is possible to take monoclonal and other

antibodies and use techniques of recombinant DNA technology to produce other antibodies or chimeric molecules which retain the specificity of the original antibody. Such techniques may involve introducing DNA encoding the immunoglobulin variable region, or the complementarity determining regions (CDRs), of an antibody to the constant regions, or constant regions plus framework regions, of a different immunoglobulin. See, for instance, EP 0 184 187 A, GB 2,188,638 A or EP 0 239 400 A.

Antibodies can be modified in a number of ways and the term "antibody molecule" should be construed as covering any specific binding member or substance having an antibody antigen-binding domain with the required specificity. Thus, this term covers antibody fragments and derivatives, including any polypeptide comprising an immunoglobulin binding domain, whether natural or wholly or partially synthetic. Chimeric molecules comprising an immunoglobulin binding domain, or equivalent, fused to another polypeptide are therefore included. Cloning and expression of chimeric antibodies are described in EP 0 120 694 A and EP 0 125 023 A.

It has been shown that fragments of a whole antibody can perform the function of binding antigens. Examples of binding fragments are (i) the Fab fragment consisting of VL, VH, CL and CHI domains; (ii) the Fd fragment consisting of the VH and CHI domains; (iii) the Fv fragment consisting of the VL and VH domains of a single antibody; (iv) the dAb fragment (Ward, E.S. et al., Nature 341, 544-546 (1989)) which consists of a VH domain; (v) isolated CDR regions; (vi) F(ab')2 fragments, a bivalent fragment comprising two linked Fab fragments (vii) single chain Fv molecules (scFv) , wherein a VH domain and a VL domain are linked by a peptide linker which allows the two domains to associate to form an antigen binding site (Bird et al, Science, 242; 423-426, 1988; Huston et al, PNAS USA, 85: 5879- 5883, 1988); (viii) bispecific single chain Fv dimers (WO

93/11161) and (ix) "diabodies", multivalent or multispecific fragments constructed by gene fusion (WO 94/13804; Holliger et al, P.N.A.S. USA, 90: 6444-6448, 1993); (x) immunoadhesins (WO 98/50431) . Fv, scFv or diabody molecules may be stabilised by the incorporation of disulphide bridges linking the VH and VL domains (Reiter et al, Nature Biotech, 14: 1239-1245, 1996) .

Minibodies comprising a scFv joined to a CH3 domain may also be made (Hu et al, Cancer Res., 56: 3055-3061, 1996) . Preferred antibodies used in accordance with the present

invention are isolated, in the sense of being free from

contaminants such as antibodies able to bind other polypeptides and/or free of serum components. Monoclonal antibodies are preferred for some purposes, though polyclonal antibodies are within the scope of the present invention.

The reactivities of antibodies on a sample may be determined by any appropriate means. Tagging with individual reporter

molecules is one possibility. The reporter molecules may directly or indirectly generate detectable, and preferably measurable, signals. The linkage of reporter molecules may be directly or indirectly, covalently, e.g. via a peptide bond or non-covalently . Linkage via a peptide bond may be as a result of recombinant expression of a gene fusion encoding antibody and reporter molecule. One favoured mode is by covalent linkage of each antibody with an individual fluorochrome, phosphor or laser exciting dye with spectrally isolated absorption or emission characteristics. Suitable fluorochromes include fluorescein, rhodamine, phycoerythrin and Texas Red. Suitable chromogenic dyes include diaminobenzidine .

Other reporters include macromolecular colloidal particles or particulate material such as latex beads that are coloured, magnetic or paramagnetic, and biologically or chemically active agents that can directly or indirectly cause detectable signals to be visually observed, electronically detected or otherwise recorded. These molecules may be enzymes which catalyse

reactions that develop or change colours or cause changes in electrical properties, for example. They may be molecularly excitable, such that electronic transitions between energy states result in characteristic spectral absorptions or emissions. They may include chemical entities used in conjunction with

biosensors. Biotin/avidin or biotin/streptavidin and alkaline phosphatase detection systems may be employed.

Antibodies according to the present invention may be used in screening for the presence of a polypeptide, for example in a test sample containing cells or cell lysate as discussed, and may be used in purifying and/or isolating a polypeptide according to the present invention, for instance following production of the polypeptide by expression from encoding nucleic acid. Antibodies may modulate the activity of the polypeptide to which they bind and so, if that polypeptide has a deleterious effect in an individual, may be useful in a therapeutic context (which may include prophylaxis) . Peptide fragments

Another class of inhibitors useful for treating estrogen receptor (ER) positive breast cancer includes peptide fragments that interfere with the activity of ADCK2. Peptide fragments may be generated wholly or partly by chemical synthesis that block the catalytic sites of ADCK2. Peptide fragments can be readily prepared according to well-established, standard liquid or, preferably, solid-phase peptide synthesis methods, general descriptions of which are broadly available (see, for example, in 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); and Applied Biosystems 430A Users Manual, ABI Inc., Foster City, California), or they may be prepared in solution, by the liquid phase method or by any combination of solid-phase, liquid phase and solution chemistry, e.g. by first completing the respective peptide portion and then, if desired and appropriate, after removal of any protecting groups being present, by introduction of the residue X by reaction of the respective carbonic or sulfonic acid or a reactive derivative thereof.

Other candidate compounds for inhibiting ADCK2 may be based on modelling the 3-dimensional structure of these enzymes and using rational drug design to provide candidate compounds with

particular molecular shape, size and charge characteristics. A candidate inhibitor, for example, may be a "functional analogue" of a peptide fragment or other compound which inhibits the component. A functional analogue has the same functional activity as the peptide or other compound in question. Examples of such analogues include chemical compounds which are modelled to resemble the three dimensional structure of the component in an area which contacts another component, and in particular the arrangement of the key amino acid residues as they appear.

Nucleic acid inhibitors

Another class of inhibitors useful for treatment of estrogen receptor (ER) positive breast cancer includes nucleic acid inhibitors of ADCK2, or the complements thereof, which inhibit activity or function by down-regulating production of active polypeptide. This can be monitored using conventional methods well known in the art, for example by screening using real time PCR as described in the examples. may be

of th

:ll-e St

Anti-sense oligonucleotides may be designed to hybridise to the complementary sequence of nucleic acid, pre-mRNA or mature mRNA, interfering with the production of the base excision repair pathway component so that its expression is reduced or completely or substantially completely prevented. In addition to targeting coding sequence, anti-sense techniques may be used to target control sequences of a gene, e.g. in the 5' flanking sequence, whereby the anti-sense oligonucleotides can interfere with expression control sequences. The construction of anti-sense sequences and their use is described for example in Peyman & Ulman, Chemical Reviews, 90:543-584, 1990 and Crooke, Ann. Rev. Pharmacol. Toxicol., 32:329-376, 1992.

Oligonucleotides may be generated in vitro or ex vivo for administration or anti-sense RNA may be generated in vivo within cells in which down-regulation is desired. Thus, double-stranded DNA may be placed under the control of a promoter in a "reverse orientation" such that transcription of the anti-sense strand of the DNA yields RNA which is complementary to normal mRNA

transcribed from the sense strand of the target gene. The complementary anti-sense RNA sequence is thought then to bind with mRNA to form a duplex, inhibiting translation of the endogenous mRNA from the target gene into protein. Whether or not this is the actual mode of action is still uncertain.

However, it is established fact that the technique works. The complete sequence corresponding to the coding sequence in reverse orientation need not be used. For example fragments of sufficient length may be used. It is a routine matter for the person skilled in the art to screen fragments of various sizes and from various parts of the coding or flanking sequences of a gene to optimise the level of anti-sense inhibition. It may be advantageous to include the initiating methionine ATG codon, and perhaps one or more nucleotides upstream of the initiating codon. A suitable fragment may have about 14-23 nucleotides, e.g., about 15, 16 or 17 nucleotides.

An alternative to anti-sense is to use a copy of all or part of the target gene inserted in sense, that is the same, orientation as the target gene, to achieve reduction in expression of the target gene by co-suppression (Angell & Baulcombe, The EMBO Journal 16(12) :3675-3684, 1997 and Voinnet & Baulcombe, Nature, 389: 553, 1997) . Double stranded RNA (dsRNA) has been found to be even more effective in gene silencing than both sense or antisense strands alone (Fire et al, Nature 391, 806-811, 1998) . dsRNA mediated silencing is gene specific and is often termed RNA interference (RNAi) . Methods relating to the use of RNAi to silence genes in C. elegans , Drosophila, plants, and mammals are known in the art (Fire, Trends Genet., 15: 358-363, 19999; Sharp, RNA interference, Genes Dev. 15: 485-490 2001; Hammond et al . , Nature Rev. Genet. 2: 110-1119, 2001; Tuschl, Chem. Biochem. 2: 239-245, 2001; Hamilton et al . , Science 286: 950-952, 1999;

Hammond, et al . , Nature 404: 293-296, 2000; Zamore et al . , Cell, 101: 25-33, 2000; Bernstein, Nature, 409: 363-366, 2001; Elbashir et al, Genes Dev., 15: 188-200, 2001; WO01/29058; W099/32619, and Elbashir et al, Nature, 411: 494-498, 2001) .

RNA interference is a two-step process. First, dsRNA is cleaved within the cell to yield short interfering RNAs (siRNAs) of about 21-23nt length with 5' terminal phosphate and 3' short overhangs (~2nt) . The siRNAs target the corresponding mRNA sequence specifically for destruction (Zamore, Nature Structural Biology, 8, 9, 746-750, 2001.

RNAi may also be efficiently induced using chemically synthesized siRNA duplexes of the same structure with 3 ' -overhang ends

(Zamore et al, Cell, 101: 25-33, 2000) . Synthetic siRNA duplexes have been shown to specifically suppress expression of endogenous and heterologeous genes in a wide range of mammalian cell lines

(Elbashir et al, Nature, 411: 494-498, 2001) .

Another possibility is that nucleic acid is used which on transcription produces a ribozyme, able to cut nucleic acid at a specific site and therefore also useful in influencing gene expression, e.g., see Kashani-Sabet & Scanlon, Cancer Gene

Therapy, 2(3) : 213-223, 1995 and Mercola & Cohen, Cancer Gene Therapy, 2(1) : 47-59, 1995.

Small RNA molecules may be employed to regulate gene expression. These include targeted degradation of mRNAs by small interfering RNAs (siRNAs), post transcriptional gene silencing (PTGs), developmentally regulated sequence-specific translational repression of mRNA by micro-RNAs (miRNAs) and targeted

transcriptional gene silencing.

A role for the RNAi machinery and small RNAs in targeting of heterochromatin complexes and epigenetic gene silencing at specific chromosomal loci has also been demonstrated. Double- stranded RNA (dsRNA) -dependent post transcriptional silencing, also known as RNA interference (RNAi), is a phenomenon in which dsRNA complexes can target specific genes of homology for silencing in a short period of time. It acts as a signal to promote degradation of mRNA with sequence identity. A 20-nt siRNA is generally long enough to induce gene-specific silencing, but short enough to evade host response. The decrease in expression of targeted gene products can be extensive with 90% silencing induced by a few molecules of siRNA.

In the art, these RNA sequences are termed "short or small interfering RNAs" (siRNAs) or "microRNAs" (miRNAs) depending on their origin. Both types of sequence may be used to down- regulate gene expression by binding to complimentary RNAs and either triggering mRNA elimination (RNAi) or arresting mRNA translation into protein. siRNA are derived by processing of long double stranded RNAs and when found in nature are typically of exogenous origin. Micro-interfering RNAs (miRNA) are

endogenously encoded small non-coding RNAs, derived by processing of short hairpins. Both siRNA and miRNA can inhibit the

translation of mRNAs bearing partially complimentary target sequences without RNA cleavage and degrade mRNAs bearing fully complementary sequences. The siRNA ligands are typically double stranded and, in order to optimise the effectiveness of RNA mediated down-regulation of the function of a target gene, it is preferred that the length of the siRNA molecule is chosen to ensure correct recognition of the siRNA by the RISC complex that mediates the recognition by the siRNA of the mRNA target and so that the siRNA is short enough to reduce a host response. miRNA ligands are typically single stranded and have regions that are partially complementary enabling the ligands to form a hairpin. miRNAs are RNA genes which are transcribed from DNA, but are not translated into protein. A DNA sequence that codes for a miRNA gene is longer than the miRNA. This DNA sequence includes the miRNA sequence and an approximate reverse

complement. When this DNA sequence is transcribed into a single- stranded RNA molecule, the miRNA sequence and its reverse- complement base pair to form a partially double stranded RNA segment. The design of microRNA sequences is discussed in John et al, PLoS Biology, 11(2), 1862-1879, 2004.

Typically, the RNA ligands intended to mimic the effects of siRNA or miRNA have between 10 and 40 ribonucleotides (or synthetic analogues thereof) , more preferably between 17 and 30

ribonucleotides, more preferably between 19 and 25

ribonucleotides and most preferably between 21 and 23

ribonucleotides. In some embodiments of the invention employing double-stranded siRNA, the molecule may have symmetric 3 ' overhangs, e.g. of one or two (ribo) nucleotides, typically a UU of dTdT 3' overhang. Based on the disclosure provided herein, the skilled person can readily design suitable siRNA and miRNA sequences, for example using resources such as Ambion's siRNA finder, see http://www.ambion.com/techlib/misc/siRNA finder.html. siRNA and miRNA sequences can be synthetically produced and added exogenously to cause gene downregulation or produced using expression systems (e.g. vectors) . In a preferred embodiment the siRNA is synthesized synthetically. Longer double stranded RNAs may be processed in the cell to produce siRNAs (e.g. see Myers, Nature Biotechnology, 21: 324- 328, 2003) . The longer dsRNA molecule may have symmetric 3' or 5' overhangs, e.g. of one or two ( ribo ) nucleotides , or may have blunt ends. The longer dsRNA molecules may be 25 nucleotides or longer. Preferably, the longer dsRNA molecules are between 25 and 30 nucleotides long. More preferably, the longer dsRNA molecules are between 25 and 27 nucleotides long. Most

preferably, the longer dsRNA molecules are 27 nucleotides in length. dsRNAs 30 nucleotides or more in length may be expressed using the vector pDECAP (Shinagawa et al . , Genes and Dev., 17: 1340-5, 2003) .

Another alternative is the expression of a short hairpin RNA molecule (shRNA) in the cell. shRNAs are more stable than synthetic siRNAs. A shRNA consists of short inverted repeats separated by a small loop sequence. One inverted repeat is complimentary to the gene target. In the cell the shRNA is processed by DICER into a siRNA which degrades the target gene mRNA and suppresses expression. In a preferred embodiment the shRNA is produced endogenously (within a cell) by transcription from a vector. shRNAs may be produced within a cell by

transfecting the cell with a vector encoding the shRNA sequence under control of a RNA polymerase III promoter such as the human HI or 7SK promoter or a RNA polymerase II promoter.

Alternatively, the shRNA may be synthesised exogenously (in vitro) by transcription from a vector. The shRNA may then be introduced directly into the cell. Preferably, the shRNA sequence is between 40 and 100 bases in length, more preferably between 40 and 70 bases in length. The stem of the hairpin is preferably between 19 and 30 base pairs in length. The stem may contain G-U pairings to stabilise the hairpin structure.

In one embodiment, the siRNA, longer dsRNA or miRNA is produced endogenously (within a cell) by transcription from a vector. The vector may be introduced into the cell in any of the ways known in the art. Optionally, expression of the RNA sequence can be regulated using a tissue specific promoter. In a further embodiment, the siRNA, longer dsRNA or miRNA is produced

exogenously (in vitro) by transcription from a vector. Alternatively, siRNA molecules may be synthesized using standard solid or solution phase synthesis techniques, which are known in the art. Linkages between nucleotides may be phosphodiester bonds or alternatives, e.g., linking groups of the formula P(0)S, (thioate); P(S)S, (dithioate) ; P(0)NR'2; P(0)R'; P(0)OR6; CO; or CONR'2 wherein R is H (or a salt) or alkyl (1-12C) and R6 is alkyl (1-9C) is joined to adjacent nucleotides through-O-or-S- .

Modified nucleotide bases can be used in addition to the

naturally occurring bases, and may confer advantageous properties on siRNA molecules containing them.

For example, modified bases may increase the stability of the siRNA molecule, thereby reducing the amount required for

silencing. The provision of modified bases may also provide siRNA molecules, which are more, or less, stable than unmodified siRNA.

The term Modified nucleotide base' encompasses nucleotides with a covalently modified base and/or sugar. For example, modified nucleotides include nucleotides having sugars, which are

covalently attached to low molecular weight organic groups other than a hydroxyl group at the 3 'position and other than a

phosphate group at the 5 'position. Thus modified nucleotides may also include 2 ' substituted sugars such as 2 ' -O-methyl- ; 2-0- alkyl ; 2-0-allyl ; 2'-S-alkyl; 2'-S-allyl; 2 ' -fluoro- ; 2 ' -halo or 2; azido-ribose , carbocyclic sugar analogues a-anomeric sugars; epimeric sugars such as arabinose, xyloses or lyxoses, pyranose sugars, furanose sugars and sedoheptulose . Modified nucleotides are known in the art and include alkylated purines and pyrimidines, acylated purines and pyrimidines, and other heterocycles . These classes of pyrimidines and purines are known in the art and include pseudoisocytosine, N4,N4- ethanocytosine , 8-hydroxy-N6-methyladenine , 4-acetylcytosine , 5- (carboxyhydroxylmethyl ) uracil, 5 fluorouracil , 5-bromouracil , 5- carboxymethylaminomethyl-2-thiouracil , 5-carboxymethylaminomethyl uracil, dihydrouracil , inosine, N6-isopentyl-adenine, 1- methyladenine, 1-methylpseudouracil, 1-methylguanine, 2,2- dimethylguanine, 2methyladenine, 2-methylguanine, 3- methylcytosine , 5-methylcytosine , N6-methyladenine , 7- methylguanine, 5-methylaminomethyl uracil, 5-methoxy amino methyl-2-thiouracil, -D-mannosylqueosine , 5- methoxycarbonylmethyluracil , 5methoxyuracil , 2 methylthio-N6- isopentenyladenine , uracil-5-oxyacetic acid methyl ester, psueouracil, 2-thiocytosine, 5-methyl-2 thiouracil, 2-thiouracil ,

4-thiouracil , 5methyluracil , N-uracil-5-oxyacetic acid

methylester, uracil 5-oxyacetic acid, queosine, 2-thiocytosine,

5-propyluracil , 5-propylcytosine , 5-ethyluracil , 5ethylcytosine, 5-butyluracil , 5-pentyluracil , 5-pentylcytosine, and

2 , 6, diaminopurine, methylpsuedouracil , 1-methylguanine, 1- methylcytosine .

Methods of screening

In some aspects, the present invention is concerned with methods of screening candidate compounds to determine whether one or more candidate agents are likely to be useful for the treatment of estrogen receptor (ER) positive cancer. As described herein, there are three preferred general approaches that may be used for these methods of screening, either alone or in any combination or order .

The present invention also includes methods of screening that employ ADCK2 as protein targets for the screening of candidate compounds to find ADCK2 inhibitors. Accordingly, methods of screening may be carried out for identifying candidate agents that are capable of inhibiting an activity of ADCK2, for

subsequent use of development as agents for the treatment of estrogen receptor (ER) positive breast cancer. Conveniently, this may be done in an assay buffer to help the components of the assay interact, and in a multiple well format to test a plurality of candidate agents. An activity of ADCK2, for instance a kinase activity, can then be determined in the presence and absence of the one or more candidate compounds to determine whether a given candidate is an ADCK2 inhibitor.

By way of example, the candidate agent may be a known inhibitor of one of the protein targets di sclosed herein, an antibody, a peptide, a nucleic acid molecule or an organic or inorganic compound, e.g. molecular weight of less than 100 Da. In some instances the use of candidate agents that are compounds is preferred. However, for any type of candidate agent,

combinatorial library technology provides an efficient way of testing a potentially vast number of different substances for ability to modulate activity of a target protein. Such libraries and their use are known in the art . The present invention also specifically envisages screening candidate agents known for the treatment of other conditions, and especially other forms of cancer. This has the advantage that the patient or disease profile of known therapeutic agents might be expanded or modified using the screening techniques disclosed herein, or for

therapeutic agents in development, patient or disease profiles established that are relevant for the treatment of estrogen receptor (ER) positive cancer.

Following identification of a candidate agent for further investigation, the agent in question may be tested to determine whether it is not lethal to normal cells or otherwise is suited to therapeutic use. Following these studies, the agent may be manufactured and/or used in the preparation of a medicament, pharmaceutical composition or dosage form.

The development of lead agents or compounds from an initial hit in screening assays might be desirable where the agent in question is difficult or expensive to synthesise or where it is unsuitable for a particular method of administration, e.g.

peptides are unsuitable active agents for oral compositions as they tend to be quickly degraded by proteases in the alimentary canal. Mimetic design, synthesis and testing is generally used to avoid randomly screening large number of molecules for a target property.

There are several steps commonly taken in the design of a mimetic from a compound having a given target property. Firstly, the particular parts of the compound that are critical and/or important in determining the target property are determined. In the case of a peptide, this can be done by systematically varying the amino acid residues in the peptide, e.g. by substituting each residue in turn. These parts or residues constituting the active region of the compound are known as its "pharmacophore".

Once the pharmacophore has been found, its structure is modelled to according its physical properties, e.g. stereochemistry, bonding, size and/or charge, using data from a range of sources, e.g. spectroscopic techniques, X-ray diffraction data and NMR. Computational analysis, similarity mapping (which models the charge and/or volume of a pharmacophore, rather than the bonding between atoms) and other techniques can be used in this modelling process. In a variant of this approach, the three-dimensional structure of the ligand and its binding partner are modelled. This can be especially useful where the ligand and/or binding partner change conformation on binding, allowing the model to take account of this in the design of the mimetic.

A template molecule is then selected onto which chemical groups which mimic the pharmacophore can be grafted. The template molecule and the chemical groups grafted on to it can

conveniently be selected so that the mimetic is easy to

synthesise, is likely to be pharmacologically acceptable, and does not degrade in vivo, while retaining the biological activity of the lead compound. The mimetics found by this approach can then be screened to see whether they have the target property, or to what extent they exhibit it. Further optimisation or modification can then be carried out to arrive at one or more final mimetics for in vivo or clinical testing.

Treatment of cancer

The present invention provides methods and medical uses for the treatment of estrogen receptor (ER) positive cancer. A cancer may be identified as estrogen receptor (ER) positive cancer by carrying out one or more assays or tests on a sample of cells from an individual. The sample will generally be a sample of cancer cells.. Activity may be determined relative to a control, for example in the case of defects in cancer cells, relative to non-cancerous cells, preferably from the same tissue. Examples of known types of ER positive cancer include breast cancer, ovarian cancer, prostate cancer, and endometrial cancer.

The determination of the ER positive status of a patient may be carried out using a number of approaches known in the art. By way of example, the presence of ER protein may be determined by using techniques well known in the art such as Western blot analysis, immunohistology, chromosomal abnormalities, enzymatic or DNA binding assays and plasmid-based assays.

Alternatively or additionally, rhe determination of ER gene expression may involve determining the presence or amount of ER mRNA in a sample. Methods for doing this are well known to the skilled person. By way of example, they include determining the presence of ER mRNA; and/or (ii) using PCR involving one or more primers based on an ER nucleic acid sequence to determine whether the ER transcript is present in a sample. The probe may also be immobilised as a sequence included in a microarray.

Preferably, detecting ER mRNA is carried out by extracting RNA from a sample of the tumour and measuring ER expression

specifically using quantitative real time RT-PCR. Alternatively or additionally, the expression of ER could be assessed using RNA extracted from a tumour sample using microarray analysis, which measures the levels of mRNA for a group of genes using a

plurality of probes immobilised on a substrate to form the array.

Alternatively or additionally, the present invention the

determination of endocrine receptor status can be carried out by analysis of ER protein expression. In particular, the results provided herein demonstrate that reduced levels of ERa protein expression are correlated with an increased likelihood of developing resistance to endocrine therapy for breast cancer.

Preferably, the presence or amount of ERa protein may be

determined using a binding agent capable of specifically binding to the ER protein, or fragments thereof. A preferred type of ER protein binding agent is an antibody capable of specifically binding the ER or fragment thereof. The antibody may be labelled to enable it to be detected or capable of detection following reaction with one or more further species, for example using a secondary antibody that is labelled or capable of producing a detectable result, e.g. in an ELISA type assay. As an

alternative a labelled binding agent may be employed in a western blot to detect ER protein.

Alternatively, or additionally, the method for determining the presence of ER protein may be carried out on tumour samples, for example using immunohistochemical (IHC) analysis. IHC analysis can be carried out using paraffin fixed samples or frozen tissue samples, and generally involves staining the samples to highlight the presence and location of ER protein. Pharmaceutical compositions

The active agents disclosed herein for the treatment of estrogen receptor (ER) positive cancer, such as breast cancer, may be administered alone, but it is generally preferable to provide them in pharmaceutical compositions that additionally comprise with one or more pharmaceutically acceptable carriers, adjuvants, excipients, diluents, fillers, buffers, stabilisers,

preservatives, lubricants, or other materials well known to those skilled in the art and optionally other therapeutic or

prophylactic agents. Examples of components of pharmaceutical compositions are provided in Remington's Pharmaceutical Sciences, 20th Edition, 2000, pub. Lippincott, Williams & Wilkins.

These compounds or derivatives of them may be used in the present invention for the treatment of estrogen receptor (ER) positive breast cancer. As used herein "derivatives" of the therapeutic agents includes salts, coordination complexes, esters such as in vivo hydrolysable esters, free acids or bases, hydrates, prodrugs or lipids, coupling partners.

Salts of the compounds of the invention are preferably

physiologically well tolerated and non toxic. Many examples of salts are known to those skilled in the art. Compounds having acidic groups, such as phosphates or sulfates, can form salts with alkaline or alkaline earth metals such as Na, K, Mg and Ca, and with organic amines such as triethylamine and Tris (2- hydroxyethyl ) amine . Salts can be formed between compounds with basic groups, e.g., amines, with inorganic acids such as

hydrochloric acid, phosphoric acid or sulfuric acid, or organic acids such as acetic acid, citric acid, benzoic acid, fumaric acid, or tartaric acid. Compounds having both acidic and basic groups can form internal salts .

Esters can be formed between hydroxyl or carboxylic acid groups present in the compound and an appropriate carboxylic acid or alcohol reaction partner, using techniques well known in the art. Derivatives which as prodrugs of the compounds are convertible in vivo or in vitro into one of the parent compounds. Typically, at least one of the biological activities of compound will be reduced in the prodrug form of the compound, and can be activated by conversion of the prodrug to release the compound or a metabolite of it.

Other derivatives include coupling partners of the compounds in which the compounds is linked to a coupling partner, e.g. by being chemically coupled to the compound or physically

associated with it. Examples of coupling partners include a label or reporter molecule, a supporting substrate, a carrier or transport molecule, an effector, a drug, an antibody or an inhibitor. Coupling partners can be covalently linked to compounds of the invention via an appropriate functional group on the compound such as a hydroxyl group, a carboxyl group or an amino group. Other derivatives include formulating the compounds with liposomes.

The term "pharmaceutically acceptable" as used herein includes compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgement, suitable for use in contact with the tissues of a subject (e.g. human) without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable

benefit/risk ratio. Each carrier, excipient, etc. must also be "acceptable" in the sense of being compatible with the other ingredients of the formulation.

The active agents disclosed herein for the treatment of estrogen receptor a (ERa) positive breast cancer according to the present invention are preferably for administration to an individual in a "prophylactically effective amount" or a "therapeutically effective amount" (as the case may be, although prophylaxis may be considered therapy) , this being sufficient to show benefit to the individual. The actual amount administered, and rate and time-course of administration, will depend on the nature and severity of what is being treated. Prescription of treatment, e.g. decisions on dosage etc., is within the responsibility of general practitioners and other medical doctors, and typically takes account of the disorder to be treated, the condition of the individual patient, the site of delivery, the method of

administration and other factors known to practitioners.

Examples of the techniques and protocols mentioned above can be found in Remington's Pharmaceutical Sciences, 20th Edition, 2000, Lippincott, Williams & Wilkins. A composition may be administered alone or in combination with other treatments, either simultaneously or sequentially, dependent upon the condition to be treated.

The formulations may conveniently be presented in unit dosage form and may be prepared by any methods well known in the art of pharmacy. Such methods include the step of bringing the active compound into association with a carrier, which may constitute one or more accessory ingredients. In general, the formulations are prepared by uniformly and intimately bringing into

association the active compound with liquid carriers or finely divided solid carriers or both, and then if necessary shaping the product .

The agents disclosed herein for the treatment of estrogen receptor a ( ERa ) positive breast cancer may be administered to a subject by any convenient route of administration, whether systemically/ peripherally or at the site of desired action, including but not limited to, oral (e.g. by ingestion); topical (including e.g. transdermal, intranasal, ocular, buccal, and sublingual); pulmonary (e.g. by inhalation or insufflation therapy using, e.g. an aerosol, e.g. through mouth or nose);

rectal; vaginal; parenteral, for example, by injection, including subcutaneous, intradermal, intramuscular, intravenous,

intraarterial, intracardiac, intrathecal, intraspinal,

intracapsular, subcapsular, intraorbital, intraperitoneal, intratracheal, subcuticular, intraarticular, subarachnoid, and intrasternal ; by implant of a depot, for example, subcutaneously or intramuscularly.

Formulations suitable for oral administration (e.g., by

ingestion) may be presented as discrete units such as capsules, cachets or tablets, each containing a predetermined amount of the active compound; as a powder or granules; as a solution or suspension in an aqueous or non-aqueous liquid; or as an oil-in- water liquid emulsion or a water-in-oil liquid emulsion; as a bolus; as an electuary; or as a paste.

Formulations suitable for parenteral administration (e.g., by injection, including cutaneous, subcutaneous, intramuscular, intravenous and intradermal) , include aqueous and non-aqueous isotonic, pyrogen-free, sterile injection solutions which may contain anti-oxidants , buffers, preservatives, stabilisers, bacteriostats , and solutes which render the formulation isotonic with the blood of the intended recipient; and aqueous and nonaqueous sterile suspensions which may include suspending agents and thickening agents, and liposomes or other microparticulate systems which are designed to target the compound to blood components or one or more organs. Examples of suitable isotonic vehicles for use in such formulations include Sodium Chloride Injection, Ringer's Solution, or Lactated Ringer's Injection. Typically, the concentration of the active compound in the solution is from about 1 ng/ml to about 10 μg/ml, for example from about 10 ng/ml to about 1 μg/ml. The formulations may be presented in unit-dose or multi-dose sealed containers, for example, ampoules and vials, and may be stored in a freeze-dried (lyophilised) condition requiring only the addition of the sterile liquid carrier, for example water for injections, immediately prior to use. Extemporaneous injection solutions and suspensions may be prepared from sterile powders, granules, and tablets. Formulations may be in the form of liposomes or other microparticulate systems which are designed to target the active compound to blood components or one or more organs . Compositions comprising agents disclosed herein for the treatment estrogen receptor (ER) positive breast cancer may be used in the methods described herein in combination with standard

chemotherapeutic regimes or in conjunction with radiotherapy. Examples of other chemotherapeutic agents include Amsacrine (Amsidine) , Bleomycin, Busulfan, Capecitabine (Xeloda) ,

Carboplatin, Carmustine (BCNU) , Chlorambucil (Leukeran) ,

Cisplatin, Cladribine (Leustat) , Clofarabine (Evoltra) , Crisantaspase (Er inase) , Cyclophosphamide, Cytarabine (ARA-C) , Dacarbazine (DTIC) , Dactinomycin (Actinomycin D) , Daunorubicin, Docetaxel (Taxotere) , Doxorubicin, Epirubicin, Etoposide

(Vepesid, VP-16), Fludarabine (Fludara) , Fluorouracil (5-FU) , Gemcitabine (Gemzar) , Hydroxyurea (Hydroxycarbamide, Hydrea)

, Idarubicin (Zavedos) . Ifosfamide (Mitoxana) , Irinotecan (CPT-11, Campto) , Leucovorin (folinic acid) , Liposomal doxorubicin

(Caelyx, Myocet) , Liposomal daunorubicin (DaunoXome®) Lomustine, Melphalan, Mercaptopurine, Mesna, Methotrexate, Mitomycin,

Mitoxantrone , Oxaliplatin (Eloxatin) , Paclitaxel (Taxol) ,

Pemetrexed (Alimta) , Pentostatin (Nipent) , Procarbazine,

Raltitrexed (Tomudex®) , Streptozocin (Zanosar®), Tegafur-uracil

(Uftoral), Temozolomide (Temodal), Teniposide (Vumon) , Thiotepa, Tioguanine (6-TG) (Lanvis) , Topotecan (Hycamtin) , Treosulfan, Vinblastine (Velbe) , Vincristine (Oncovin) , Vindesine (Eldisine) and Vinorelbine (Navelbine) .

Administration in vivo can be effected in one dose, continuously or intermittently (e.g., in divided doses at appropriate

intervals) throughout the course of treatment. Methods of determining the most effective means and dosage of administration are well known to those of skill in the art and will vary with the formulation used for therapy, the purpose of the therapy, the target cell being treated, and the subject being treated. Single or multiple administrations can be carried out with the dose level and pattern being selected by the treating physician.

In general, a suitable dose of the active compound is in the range of about 100 μg to about 250 mg per kilogram body weight of the subject per day. Where the active compound is a salt, an ester, prodrug, or the like, the amount administered is

calculated on the basis of the parent compound, and so the actual weight to be used is increased proportionately. Experimental Examples

METHODS

Cell lines, compounds and siRNA All cell lines were obtained from ATCC (USA) and maintained according to the supplier's instructions. Cell lines were grown and transfected with SMARTpool siRNAs using Dharmafect 3 (DF3) , Dharmafect 4 (DF4) (Dharmacon) , Oligofectamine , Lipofectamine 2000 or RNAiMAX (Invitrogen) transfection reagents. The siRNA library (siARRAY - targeting 714 known and putative human protein kinase genes) was obtained in nine 96 well plates from Dharmacon

(USA) . Each well in this library contained a SMARTpool of four distinct siRNA species targeting different sequences of the target transcript. Each plate was supplemented with siCONTROL

(ten wells, Dharmacon (USA)) .

Antibodies

Antibodies targeting the following were used as per manufacturers instructions: ACTIN and C-MYC (Santa Cruz Biotech), TTK, ER, PR, ERBB2, CYCLIN Dl , TFF1, FOXOl, C-JUN and ADCK2 (AbCam) . All secondary antibodies used for western blot analysis were HRP conj ugated .

RNA interference screening

We transfected cells with the SMARTpool library, where each well of the 96 wellplate contained a pool of four different siRNAs (a SMARTpool) targeting one gene. After five population doublings, cell viability in each well was estimated by use of a highly sensitive luminescent assay measuring cellular ATP levels (Cell Titre Glo, Promega) . To identify loss of viability/failure to proliferate effects in each cell line, luminescence readings from each well were log transformed and then centred by the plate median, to account for plate-to-plate variation common in high- throughput screens. Well position effects were identified and eliminated and the quality of data from each plate estimated by calculating Z' -factors (24) based on positive (siRNA targeting PLK1 ) and negative (non-targeting siRNA) controls in each plate. The dynamic range of each screen was determined by calculating Z prime values (24); we use a threshold of Z prime >0.3 to define acceptable screens, based upon previous screens where Z prime >0.3 is predictive of reproducible data (25, 26) . To allow data to be compared between different cell lines, plate- centred data from each screen were standardised by the use of a Z score statistic, where Z=0 represents no effect on viability and negative Z scores represent loss of viability. Here, Z scores were calculated using the Median Absolute Deviation (MAD) of all effects in each cell line (5) . In each screen the Z score data approximated a normal distribution allowing comparison of the individual siRNA effects across cell lines. siRNA screens were carried out in triplicate and comparison of Z score data from replica screens of each cell line showed the screening process to be highly robust, as demonstrated by Spearman's r2 values approaching 1 for all comparisons. Raw luminescence values and processed data are now deposited on the ROCK Breast Cancer

Functional Genomics Database (23) (rock.icr.ac.uk) as a community resource. This rigorous quality control method reduced our cell line panel to 20 cell lines with robust viability profiles.

Heatmaps of Z scores that separated different phenotypes as defined by a median difference permutation test were created to show the significant genes ranked row-wise according to their median difference and ordered column-wise by phenotype.

Western blots

Protein lysates were prepared using RIPA lysis buffer (50 nM Tris pH 8.0, 150 mM NaCl, 0.1% SDS, 0.1% DOC, 1% TritonX-100, 50 mM NaF, 1 mM Na 3 V0 4 and protease inhibitors) . lOOmg of total cell lysate was loaded onto prefabricated 4-12% Bis-Tris gels

(Invitrogen) , with full range rainbow molecular weight marker (GE Healthcare, UK) as a size reference, and resolved by SDS-PAGE electrophoresis. Proteins were transferred to nitrocellulose membrane (Bio-rad, USA) , blocked and probed with primary antibody diluted 1 in 1000 in 5% milk overnight at 4°C. Secondary antibodies were diluted 1 in 5000 in 5% milk and incubated for one hour at room temperature. Protein bands were visualised using ECL (GE Healthcare, UK) and MR or XAR film (Kodak) .

Validation of gene silencing by siRNA Validation of RNAi gene silencing was determined by western blotting and by viability assays of silencing effects with individual oligos. Cells were transfected with individual ERBB2 , ADCK2, ESR1, PIK3CA or TTK siGenome oligos ( Dharmacon) . Protein lysates were collected 48 hours following transfection for western blot analysis. Cell viability was measured using

CellTiter Glo (Promega, USA) after 5 populations doublings.

Survival assays

For measurement of sensitivity to TTK inhibitor treatment, cells were plated in 96 well plates and exposed to the drug at the indicated concentrations. Cells were dosed at 24 and 96 hours. After 7 days, cell viability was measured using CellTiter Glo Luminescent Cell Viability Assay (Promega, USA) . Surviving fractions were calculated and drug sensitivity curves plotted as previously described (27) .

Metaphase Spreads

After 24 hours of treatment with 2uM AZ3146, HCT116 PTEN wt or PTEN null cells were treated with colcemide (lOng/ml, Sigma) and MG132 (20uM, Sigma) for 1 hour. Cells were then lysed in hypotonic solution (0.03M Sodium Citrate) for 20 minutes at 37°C and fixed in methanol/acetic acid (3:1) . Two or three drops of suspended cells were applied to glass slides and chromosomes were stained with DAPI .

FISH

Cell pellets of CA151 and Hec-lb were washed twice with PBS and fixated with methanol/acetic acid solution (3:1) onto slides. LSI PTEN (10q23, red/orange) /chromosome 10 centromere (CEP 10, green) Dual Colour Probe (Abbott Molecular, IL, US) was

hybridised to representative slides of the cell lines according to the manufacturer's instructions. Signals were counted in 100 non-overlapping nuclei using the Leica TCS SP2 confocal

microscope (Leica, Milton Keynes, UK) .

Transcript profiling RNA was extracted from cell lines with Trizol and

phenol/chloroform extraction followed by isopropanol

precipitation. For each cell line, triplicate extractions and profiles were performed. Biotin-labelled cRNA was produced by means of a linear amplification kit (IL1791; Ambion, Austin, TX, http://www. ambion.com) using 250 ng of quality-checked total RNA as input. Chip hybridisations, washing, Cy3-streptavidin

(Amersham Bio- sciences) staining, and scanning were performed on an Illumina BeadStation 500 (San Diego, http://www.illumina.com) platform using reagents and following protocols supplied by the manufacturer as previously described (28) . cRNA

samples were hybridised on Illumina human-6 v2 BeadChips, covering approximately 47,000 RefSeq transcripts. The random distribution of large populations of oligonucleotide-coated beads across the available positions within the human-6 v2 chip enables, on average, 30 intensity measurements per RefSeq, yielding quantitative assessments of gene expression. All basic expression data analysis was carried out using the manufacturer' s software BeadStudio 3.1. Illumina expression profiles were performed in triplicate, the raw data were then variance- stabilizing transformed and robust spine normalised using the lumi package in the Bioconductor software. Expression values for each sample were median scaled and the mean expression value was established over the three replicates. Genes with significant difference in expression between cell lines were identified by one-way analysis of variance (ANOVA) .

Array CGH (aCGH) analysis method

Genomic DNA was extracted from cell lines using the QIAamp DNA Blood Mini Kit (51104, Qiagen) , according to manufacturer's instructions. Microarray-based CGH analysis was performed on an in-house 32K tiling path BAC array platform as previously described (29) . For copy number correlations, the circular binary segmentation (CBS) ratios of BACs containing the gene of interest were used for copy number correlations, and copy number assigned as previously described (29) . Briefly, AWS smoothed log2 ratio values <-0.12 were categorised as losses, those >0.12 as gains, and those in between as unchanged. Amplifications were defined as smoothed log2 ratio values >0.4 (29) . Data processing and analysis were carried out in R 2.9.0 (http://www.r-project.org/) and BioConductor 1.5 (http://www.bioconductor.org/), making extensive use of modified versions of the packages aCGH, marray and aws in particular.

Correlation of gene expression and aCGH data

aCGH and gene expression were compared by direct Pearson

correlation of gene expression log intensity values with smoothed log ratio values for every probe in the gene expression data. Correlation p values were adjusted for multiple comparison testing using the local False Discovery Rate (FDR) method of Benjamini and Hochberg (30) as previously described (28) . Mann Whitney U tests were preformed for each gene to compare gene expression values in groups defined as assigned as amplified or not, gained or not, lost or not and deleted or not using the thresholded aCGH calls. Wilcox test p-values were corrected for multiple comparison testing within contiguously altered regions. For each gene, cases in which genes were amplified, gained, lost or deleted were recorded along with the fold changes between samples carrying a copy number change and those which did not. Total counts of gains, losses, amplifications and deletions were also recorded.

Correlation of siRNA Z score with gene expression and aCGH data The correlation between siRNA Z score and normalised gene expression was examined for genes where siRNA caused significant loss of viability (Z<-2) . Z score was compared to normalized gene expression using Pearson correlation coefficient. A gene was taken as being significantly correlated if the Pearson

correlation coefficient was significantly different to the null hypothesis, the correlation was inverse, and the variation in gene expression between cells lines were significantly different as assessed by one-way AN0VA. For aCGH overlay, Z scores for the common 714 genes in the gene expression and the kinase library were correlated directly using Pearson correlation and p-values adjusted for multiple comparison testing using the FDR (28) . Gene expression values were

subsequently compared in those cell lines that displayed Z score hits for each gene and those that did not. Fold changes between these groups for each gene were also calculated. Array CGH copy number changes for each gene were used to divide the cell lines into those carrying amplifications, gains, losses or deletions, from those which did not and Mann Whitney U tests were performed to compare gene expression between these groups for each gene. The numbers of cases harbouring amplifications, gains losses and deletions which also responded with Z score hits in the same cell line was also calculated.

Clustering Methods

Supervised analysis was performed by calculating the absolute difference in the medians of the two groups, followed by

estimating a p-value by permuting the labels to create a

distribution of median differences against which to compare the actual effects. We used a significance cut-off of p=0.05 with no corrections for multiple comparisons. The combined gene

expression, Z scores and aCGH matrices were subjected to

hierarchichal clustering using a Pearson correlation distance measure and Wards clustering algorithm. Equivalent heatmaps were drawn which clustered the samples and the genes in the combined overlay using the corresponding Z scores. Heatmaps of gene expression and copy number changes were then displayed under the same dendrograms derived by clustering by Z score. Stability of the clustering was established by pvclust giving an approximately unbiased (AU) p-value greater than 0.9. All analysis was performed in R 2.9.0 using in-house scripts.

Cell Line Verification

STR (short tandem repeat) was used for profiling to verify authenticity of the panel of cell lines. We simultaneously amplified 8 STR loci, in a multiplex PCR reaction (Promega

PowerPlex 1.2 System) and used the ATCC database for comparison, where possible. In addition, gain and loss of expression of particular subtype markers, described by Hollestelle et al . (14), were confirmed by western blot as well as microarray analysis.

RESULTS

Generation of functional viability profiles of breast cancer To generate functional profiles of breast cancer, we used an approach that involves high-throughput RNA interference/short interfering RNA (siRNA) viability screening of multiple cell lines and the integration of viability profiles with gene expression, genetic, genomic and histological analysis (3) . We reasoned that siRNAs causing significant loss of cell viability in all of the cell lines assayed likely targeted genes that had an essential ubiquitous function in both normal and tumor cells. Similarly, siRNAs that had no significant effect on viability in any of the cell lines were either not functional or targeted non-essential genes. Finally, siRNAs that caused significant lethality in only some but not all cell lines likely identified genes that represented tumour-specific dependencies and candidate therapeutic targets.

To generate functional viability profiles for breast cancer we selected a panel of 34 breast cancer-derived cell lines and optimised these for high-throughput siRNA screening.

Subsequently, each cell line was transfected with a 96 well-plate arrayed siRNA library targeting 714 kinases and kinase-related genes (see Methods) . After five population doublings, cell viability in each well was estimated by use of a highly sensitive luminescent assay measuring cellular ATP levels. To identify loss of viability /inhibition/ failure to proliferate effects in each cell line, luminescence readings from each well were log transformed and then centred by the plate median, to account for plate-to-plate variation. To allow data to be compared between different cell lines, plate-centred data from each screen was standardised by the use of a Z score statistic, where Z=0 represents no effect on viability and negative Z scores

represent loss of viability. RNA interference screens were carried out in triplicate and comparison of Z score data from replica screens of each cell line showed the screening process to be highly robust.

Having screened and processed data from 34 breast cancer lines, we selected 20 for further analysis, based on stringent QC thresholds (such as Z prime-factors) . This panel encapsulated each of the major breast cancer subtypes (4) and included hormone receptor (Estrogen Receptor (ER) , Progesterone Receptor (PR) ) , ERBB2 positive as well as ER, PR and ERBB2 negative models

(triple negative) , as characterised by parallel

immunohistochemical, fluorescent in situ hybridisation (FISH) and western blot analysis. To complement this functional viability profiling, we also characterised the breast tumor panel using transcript microarrays, array-based comparative genomic

hybridisation (aCGH) , drug sensitivity for a small number of targeted agents, as well as gene mutation data. In the siRNA screens we used a stringent threshold of Z<-2 for defining loss of viability effects (approximately equal to Q=0.02275 in one screen replica, and Q= 1.21 xl0-5 in triplicate screens (5)) and identified 330 genes whose depletion by siRNA caused loss of viability in at least one cell line, and 180 genes that caused loss of viability in two or more cell lines. This analysis enabled us to identify the predominant dependencies in each cell.

Identification of de facto genetic dependencies using functional viability profiling

The delineation of functional viability profiles for the breast cell line panel provided a framework for identifying dependencies in particular genetically-defined subgroups of the disease. As proof of this principle, we used supervised hierarchical

clustering of the viability data and integration with DNA sequence, aCGH, FISH and western blot data to identify additional gene dependencies in PIK3CA mutant or ERBB2 amplified cell lines. Somatic mutations of PIK3CA, which encodes the pllO catalytic subunit of Pl3-kinase, have been shown to induce oncogenic transformation in vitro and in vivo (6). PIK3CA mutations are found in 8% - 35% of human breast cancers making them one of the most common genetic aberrations in this disease (7) . Supervised hierarchical clustering of functional viability data according to PIK3CA mutation status identified 30 siRNAs that caused loss of viability preferentially in the PIK3CA mutant. Of 714 genes profiled in the cell line panel, the highest ranked PIK3CA mutant selective effect was targeting of PIK3CA itself (p=0.014; Fig. lb) . In six of seven PIK3CA mutant lines, PIK3CA siRNA gives effects of Z<-2, suggesting that PIK3CA addiction is one of the most significant effects in PIK3CA mutant tumor cells.

Statistically, the chance probability, Q, of a PIK3CA siRNA effect of <-2 in six independently-derived PIK3CA mutant lines is approximately Q=1.39xl0 ~10 (where Z -2 in one cell line is approximately equal to, a Q = 0.02275 effect, or 1 in 44; in six cell lines this is a Q = (0.02275) 6 chance effect), demonstrating a strong dependency on PIK3CA in breast cancers with PIK3CA mutations despite their diverse genetic backgrounds. We also noted that PIK3CA mutant breast cancer lines were preferentially sensitive to AKT2 and AKT3 siRNAs (Fig. lc) . Although the mechanisms by which mutant PIK3CA mediates cellular

transformation are not completely understood, it is likely that part of this effect is mediated by signalling through AKT (8) . The preferential sensitivity of PIK3CA mutant cells to AKT targeting supported the hypothesis that the viability profiles had the ability to illuminate true addiction pathways. Our analysis of the functional viability profiles based on PIK3CA mutation status also suggested that cell lines with mutations in the kinase domain of PIK3CA tend to be more sensitive to PIK3CA siRNA targeting than cell lines harbouring non-kinase domain PIK3CA mutations. Whilst a wide variety of tumour specific PIK3CA mutations have been identified, the vast majority occur in two hotspots, either in the kinase domain (for example p.H1047R) or in the helical domain (e.g. p.E542K or p.E545K) . It has been suggested that helical and kinase domain mutants have distinct physiologic phenotypes in human cells (9, 10), and the

differential effects of PIK3CA targeting in helical vs. kinase domain mutants could also suggest differences in PIK3CA

addiction.

Interrogation of the functional profiles also identified other well-established oncogene addiction effects. For example, approximately 15% of breast cancers exhibit amplification and overexpression of the ERBB2 gene and the addiction of some tumor types to ERBB2 signalling is the basis for the clinical success of ERBB2-targeting agents such as trastuzumab and lapatinib.

Supervised clustering of viability data based on ERBB2

amplification status (based upon an integration with aCGH and FISH data identified a number of genes that when silenced were selectively lethal in ERBB2 amplified breast cancer lines (Fig. Id) . One of the most dominant effects was targeting of ERBB2 itself (p=0.003), supporting the hypothesis that ERBB2

amplification can lead to an oncogene addiction effect (Fig.

ld,e) . This reaffirms that our approach may have merit in identifying genes functionally relevant to tumor cell viability in clinically defined subgroups. Moreover many ERBB2 amplified cell lines are highly sensitive to PIK3CA siRNA (Fig. Id) supporting the clinical development of PI3K inhibitors in ERBB2- dependent breast cancer regardless of PIK3CA mutation status.

Genetic dependency on ADCK2 in ER+ve breast tumor models

As a second example of the potential of functional viability profiling we interrogated our datasets to identify candidate genes whose targeting was selective for ER+ve versus ER-ve subtypes. The most significant effect we identified was the sensitivity of ER+ve breast tumor cell line models to ADCK2 siRNA (p=0.004; Fig. 2a, b), an effect validated in an additional four ER+ve breast tumor cell line models not examined in the original screen. This suggested that the genetic dependency of ER+ve breast tumor cells to ADCK2 is a relatively common phenomenon. Integration of viability data with transcript and protein profiling also identified a correlation between sensitivity to ADCK2 silencing and ADCK2 mRNA/protein perhaps suggesting that elevated expression of this protein could be essential

for the survival of particular tumor cells.

ADCK2 (Uncharacterized aarF domaincontaining protein kinase 2) is a member of a family of aarF-domain containing proteins. Other members of this protein family are localised to the mitochondria and mitochondrial membranes and have been implicated in

ubiquinone biosynthesis (20) . Given that the most dominant driver in ER+ve breast cancers is estrogen signalling itself, we hypothesised that ADCK2 could target ER+ve breast tumor cells by modulating estrogen signalling. Supporting this hypothesis we noted that sensitivity to siRNA targeting of ESR1 , the gene encoding ER , closely correlated with the inhibitory effect of ADCK2 siRNA (see the Table below) suggesting that ADCK2 is an important dependency in breast tumor cells with a strong

addiction to estrogen signalling. More specifically we noted that ADCK2 silencing reduced estrogen signalling, as measured by the expression of well established ER target genes, including ESR1 itself (Fig. 2c), estrogen stimulation stimulated ADCK2 expression, suppression of estrogen signalling (with

faslodex/ICI182780) inhibited ADCK2 expression (Fig. 2d), and finally that ADCK2 co-immunoprecipitated with ERa (Fig. 2e) , indicating that the estrogen receptor and ADCK2 interact, either directly or indirectly. Taken together, these observations suggest that the genetic dependency of ER+ve breast tumor cells for ADCK2 is explained by ADCK2 playing a role in estrogen signalling itself. To extend this observation we examined ADCK2 expression in publically available breast cancer gene expression datasets . ER+ve breast tumours divide into two broad and overlapping subgroups termed luminal A and B, based predominantly upon proliferative rate and strength of estrogen signalling (luminal A being low proliferative with strong estrogen

signalling (21)) . We noted in two independent studies that ADCK2 expression was elevated in the luminal A subgroup when compared to luminal B (Fig. 2f; p=0.0135 and 0.0054 in the two studies, respectively (11, 22)), in concordance with our in vitro

observations .

Table : ADCK2 and estrogen receptor (ESR1) information in the breast tumour cell line panel. The ADCK2 and ESR1 copy number, expression (as assessed by microarray, immunohistochemistry (IHC; for ESR1 only) and western blot) and cell line dependency for ADCK2 and ESR1 (as defined by Z score) is summarized here.

Highlighted are six ER positive lines. ND = not determined. NPI = normalized percent inhibition, which is an estimation of survival where 1 = no cell inhibition and 0 = complete cell inhibition. Expression by western blot analysis was graded none, low, medium and high as determined by the intensity of the ADCK2 and ESR1 bands. IHC positive refers to the presence of ESR1 staining and negative refers to the absence of staining.

References

The documents disclosed herein are all expressly incorporated by reference in their entirety.

1. Ash orth A, Lord CJ, Reis-Filho JS . Genetic interactions in cancer progression and treatment. Cell 2011;145:30-8.

2. Baselga J, Swain SM. Novel anticancer targets: revisiting ERBB2 and discovering ERBB3. Nat Rev Cancer 2009;9:463-75.

3. Iorns E, Lord CJ, Grigoriadis A, McDonald S, Fenwick K, Mackay A, et al . Integrated functional, gene expression and genomic analysis for the identification of cancer targets. PLoS One 2009; 4 :e5120.

4. Neve RM, Chin K, Fridlyand J, Yeh J, Baehner FL, Fevr T, et al . A collection of breast cancer cell lines for the study of functionally distinct cancer subtypes. Cancer Cell 2006; 10:515- 27.

5. Boutros M, Bras LP, Huber W. Analysis of cell-based RNAi screens. Genome Biol 2006;7:R66.

6. Zhao L, Vogt PK. Class I PI3K in oncogenic cellular

transformation. Oncogene 2008;27:5486-96.

7. Stemke-Hale K, Gonzalez-Angulo AM, Lluch A, Neve RM, Kuo WL,

Davies M, et al . An integrative genomic and proteomic analysis of PIK3CA, PTEN, and AKT mutations in breast cancer. Cancer Res 2008;68:6084-91.

8. Manning BD, Cantley LC . AKT/PKB signaling: navigating

downstream. Cell 2007;129:1261-74.

9. Zhao L, Vogt PK. Helical domain and kinase domain mutations in pllOalpha of phosphatidylinositol 3-kinase induce gain of function by different mechanisms. Proc Natl Acad Sci U S A

2008;105:2652-7.

10. Pang H, Flinn R, Patsialou A, Wyckoff J, Roussos ET, Wu H, et al . Differential enhancement of breast cancer cell motility and metastasis by helical and kinase domain mutations of class IA phosphoinositide 3-kinase. Cancer Res 2009;69:8868-76.

11. Natrajan R, Weigelt B, Mackay A, Geyer FC, Grigoriadis A, Tan DS, et al . An integrative genomic and transcriptomic analysis reveals molecular pathways and networks regulated by copy number aberrations in basal-like, HER2 and luminal cancers. Breast Cancer Res Treat 2010;121:575-89.

12. Santarius T, Shipley J, Brewer D, Stratton MR, Cooper CS . A census of amplified and overexpressed human cancer genes. Nat Rev Cancer 2010;10:59-64.

13. Sorlie T, Perou CM, Tibshirani R, Aas T, Geisler S, Johnsen H, et al . Gene expression patterns of breast carcinomas

distinguish tumor subclasses with clinical implications. Proc Natl Acad Sci U S A 2001;98:10869-74.

14. Hollestelle A, Nagel JH, Smid M, Lam S, Elstrodt F,

Wasielewski M, et al . Distinct gene mutation profiles among luminal-type and basal-type breast cancer cell lines. Breast Cancer Res Treat; 121 : 53-64.

15. Hewitt L, Tighe A, Santaguida S, White AM, Jones CD,

Musacchio A, et al . Sustained Mpsl activity is required in mitosis to recruit 0-Mad2 to the Madl-C-Mad2 core complex. J Cell Biol 2010;190:25-34.

16. Lee C, Kim JS, Waldman T. PTEN gene targeting reveals a radiation-induced size checkpoint in human cancer cells. Cancer Res 2004;64:6906-14.

17. Puc J, Parsons R. PTEN loss inhibits CHK1 to cause double stranded-DNA breaks in cells. Cell Cycle 2005;4:927-9.

18. Shen WH, Balajee AS, Wang J, Wu H, Eng C, Pandolfi PP, et al . Essential role for nuclear PTEN in maintaining chromosomal integrity. Cell 2007;128:157-70.

19. Kwiatkowski N, Jelluma N, Filippakopoulos P, Soundararaj an M, Manak MS, Kwon M, et al . Small-molecule kinase inhibitors provide insight into Mpsl cell cycle function. Nat Chem Biol 2010; 6:359- 68.

20. Lagier-Tourenne C, Tazir M, Lopez LC, Quinzii CM, Assoum M,

Drouot N, et al . ADCK3, an ancestral kinase, is mutated in a form of recessive ataxia associated with coenzyme Q10 deficiency. Am J Hum Genet 2008;82:661-72.

21. Sotiriou C, Pusztai L. Gene-expression signatures in breast cancer. N Engl J Med 2009;360:790-800. 22. Wang Y, Klijn JG, Zhang Y, Sieu erts AM, Look MP, Yang F, et al . Geneexpression profiles to predict distant metastasis of lymph-node-negative primary breast cancer. Lancet 2005;365:671-9.

23. Sims D, Bursteinas B, Gao Q, Jain E, Mackay A, Mitsopoulos C, et al . ROCK: a breast cancer functional genomics resource. Breast

Cancer Res Treat 2010.

24. Zhang JH, Chung TD, Oldenburg KR. A Simple Statistical

Parameter for Use in Evaluation and Validation of High Throughput Screening Assays. J Biomol Screen 1999;4:67-73.

25. Iorns E, Turner NC, Elliott R, Syed N, Garrone 0, Gasco M, et al . Identification of CDK10 as an important determinant of resistance to endocrine therapy for breast cancer. Cancer Cell 2008;13:91-104.

26. Turner NC, Lord CJ, Iorns E, Brough R, Swift S, Elliott R, et al . A synthetic lethal siRNA screen identifying genes mediating sensitivity to a PARP inhibitor. EMBO J 2008;27:1368-77.

27. Farmer H, McCabe N, Lord CJ, Tutt AN, Johnson DA, Richardson TB, et al . Targeting the DNA repair defect in BRCA mutant cells as a therapeutic strategy. Nature 2005;434:917-21.

28. Natrajan R, Weigelt B, Mackay A, Geyer FC, Grigoriadis A, Tan DS, et al . An integrative genomic and transcriptomic analysis reveals molecular pathways and networks regulated by copy number aberrations in basal-like, HER2 and luminal cancers. Breast Cancer Res Treat; 121 : 575-89.

29. Geyer FC, Weigelt B, Natrajan R, Lambros MB, de Biase D,

Vatcheva R, et al. Molecular analysis reveals a genetic basis for the phenotypic diversity of metaplastic breast carcinomas. J Pathol; 220 : 562-73.

30. Hochberg Y, Benjamini Y. More powerful procedures for multiple significance testing. Stat Med 1990;9:811-8.

31. Natrajan R, Lambros MB, Rodriguez-Pinilla SM, Moreno-Bueno G, Tan DS, Marchio C, et al . Tiling path genomic profiling of grade 3 invasive ductal breast cancers. Clin Cancer Res 2009; 15 : 2711- 22.