AU2005316200A1 - Determinants of sensitivity to chemotherapeutic agents - Google Patents

Determinants of sensitivity to chemotherapeutic agents Download PDF

Info

Publication number
AU2005316200A1
AU2005316200A1 AU2005316200A AU2005316200A AU2005316200A1 AU 2005316200 A1 AU2005316200 A1 AU 2005316200A1 AU 2005316200 A AU2005316200 A AU 2005316200A AU 2005316200 A AU2005316200 A AU 2005316200A AU 2005316200 A1 AU2005316200 A1 AU 2005316200A1
Authority
AU
Australia
Prior art keywords
ankrd1
expression
ankyrin repeat
tumour
repeat protein
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
AU2005316200A
Other versions
AU2005316200B2 (en
Inventor
Anna De Fazio
Alexander David Guminski
Paul Robert Harnett
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Sydney West Area Health Service SWAHS
Original Assignee
Sydney West Area Health Service SWAHS
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from AU2004907126A external-priority patent/AU2004907126A0/en
Application filed by Sydney West Area Health Service SWAHS filed Critical Sydney West Area Health Service SWAHS
Priority to AU2005316200A priority Critical patent/AU2005316200B2/en
Priority claimed from PCT/AU2005/001890 external-priority patent/WO2006063397A1/en
Publication of AU2005316200A1 publication Critical patent/AU2005316200A1/en
Application granted granted Critical
Publication of AU2005316200B2 publication Critical patent/AU2005316200B2/en
Ceased legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Description

WO 2006/063397 PCT/AU2005/001890 Determinants of Sensitivity to Chemotherapeutic Agents Technical Field The present invention relates to methods of determining the level of sensitivity or resistance of tumour cells to chemotherapeutic agents, such as platinum-based agents, and thus the 5 responsiveness of patients with tumours to such agents. The invention further relates to methods of reducing resistance (increasing sensitivity) of tumour cells to chemotherapeutic agents and to constructs and compositions for achieving the same. Background of the Invention Ovarian cancer is a major contributor to cancer mortalities worldwide and is the fifth most 1o common cause of cancer death in Australian women, with approximately 1200 new cases diagnosed and 750 deaths each year (Australian Institute of Health and Welfare, 2002). It is an insidious disease with no specific symptoms and currently no accurate screening test. Consequently, ovarian cancer is usually not diagnosed until extensive spread has occurred. Ovarian cancers show a high response rate to chemotherapy, but unfortunately this does not 15 translate to high cure rates. Indeed, although chemotherapy has increased survival times, there has been little improvement in cure rate, with only 20% of patients with advanced stage ovarian cancer surviving more than 5 years (Kricker, 2002). There is a need for improved therapies for the treatment of advanced ovarian cancer. Platinum-based drugs including cisplatin and its analogues carboplatin and oxaliplatin, are 20 commonly used chemotherapeutic agents that are effective as single agents or in combination with other drugs in the treatment of a wide variety of malignant solid tumrnours including germ cell tumours, and cancers arising in lung, upper aero-digestive tract, urothelium as well as the ovarian epithelium. However not all patients respond equally to these drugs (for example approximately 30% of ovarian cancers do not respond) and some patients that do respond to initial treatment 25 subsequently develop resistance to the drugs and do not respond when their disease relapses. Accordingly, there is an urgent need for the development of new therapeutic and management strategies for cancers resistant to chemotherapeutic-drugs Pr-ogress in this regard requires improved understanding of the molecular and genetic changes that result in resistance to chemotherapy. 30 A number of genes have been implicated in resistance to platinum-based chemotherapy. For example demethylation of one of the key genes in the Fanconi anaemia-BRCA1 pathway, FANCF, has been shown to result in increased cisplatin resistance (Taniguchi et al., 2003). At the level of transmembrane transport, there is also evidence that the transmembrane transport protein MRP2, is a platinum export pump and that lack of expression increases platinum sensitivity WO 2006/063397 PCT/AU2005/001890 -2 (Guminski et al., 2005). In addition, the induction of TP53 expression following DNA strand breaks leads to apoptosis and also contributes to cisplatin cytotoxicity (Niedner et al., 2001). However it is not yet possible to predict sensitivity to platinum-based drugs in patients, nor to design rational strategies to overcome resistance. The ability to predict drug response would assist in identifying 5 those patients that fail to respond to therapy without significant benefit, thereby enabling the early selection of a potentially more effective chemotherapy regime. Similarly, a detailed understanding of the pathways involved in the determination of drug sensitivity will lead to novel, targeted strategies for overcoming resistance. Despite the lack of detailed understanding of the mechanisms that underlie clinical drug o10 tumour resistance, targets have been implicated and clinical trials initiated. These include co administration of inhibitors of p-glycoprotein and MRP1 drug efflux pumps; however no increase in response rates have been seen in trials conducted thus far. Detailed understanding of the pathways and underlying mechanisms involved in chemotherapeutic response is clearly required to maximize the likelihood of success of these types of approaches. 15 Further, despite the number of drug-resistance mechanisms that have been described in vitro, to date none of these mechanisms has been shown unequivocally to be important in the clinical setting. This can be attributed to some extent to the models used to study drug resistance, In many studies, sensitive cell lines have been exposed to increasing doses of cytotoxic drugs to generate resistant cell lines and as a consequence cell lines with several-hundred-fold greater 20 resistance to the drug have been generated. However, the clinical relevance of resistance mechanisms generated by such methods has been questioned (Agarwal and Kaye, 2003). In clinical practice, tumours are exposed to repeated fixed drug concentrations and this could select for resistance via different mechanisms. These observations support the need for further searches for genes and pathways that might be clinically useful determinants of tumour response to 25 , chemotherapy. The present invention is predicated on the inventors' finding that the muscle ankyrin repeat protein ANKRD1 (ankyrin repeat domain 1 (cardiac muscle) protein) also known as CARP, is expressed in human ovarian and breast tumours and that alterations in the level of expression of ANKRD1 modulates the sensitivity of the tumour cells to cisplatin. 30 Summary of the Invention According to a first aspect of the present invention there is provided a method for determining the level of resistance of a tumour cell to one or more chemotherapeutic agents, the method comprising measuring the level-of expression of a muscle ankyrin repeat protein in the tumour cell.
WO 2006/063397 PCT/AU2005/001890 -3 The muscle ankyrin repeat protein may be selected from the group consisting of ANKRD1, ANKRD2 and ANKRD23. The muscle ankyrin repeat protein may be ANKRD1 and the ANKRD1 protein may comprise an amino acid sequence as set forth in SEQ ID NO: 1 or SEQ ID NO: 3. 5 The ANKRD1 protein may be encoded by polynucleotide comprising a nucleic acid sequence as set forth in SEQ ID NO: 2 or SEQ ID NO: 4. The expression of the muscle ankyrin repeat protein may be determined by measuring protein expression or mRNA expression levels. The tumour cell may be an ovarian, breast, lung, bladder, testicular, cervical, endometrial or o10 bowel tumour cell or a cell from a head or neck tumour. The chemotherapeutic agent may be a platinum-based agent, an anthracycline or a taxane. The platinum-based chemotherapeutic agent may be cisplatin or a metabolite, derivative or analogue thereof. The analogue may be carboplatin or oxaliplatin. The anthracycline may be adriamycin or a metabolite, derivative or analogue' thereof. The taxane may be paclitaxel, 15is docataxel, or metabolite, derivative or analogue thereof. According to a second aspect of the present invention there is provided a method of assessing the responsiveness of a patient to one or more chemotherapeutic agents, the method comprising: (a) obtaining a biological sample containing at least one tumour cell from the patient, and 20 (b) analyzing the level of expression of a muscle ankyrin repeat protein in the at least one tumour cell, wherein the detected level of expression of the muscle ankyrin repeat protein correlates with the sensitivity level of the at least one tumour cell to the chemotherapeutic agent. The muscle ankyrin repeat protein may be selected from the group consisting of ANKRDI, 25 ANKRD2 and ANKRD23. The ANKRD1 polypeptide may comprise an amino acid sequence as set forth in SEQ ID NO: 1 or SEQ ID NO: 3. The ANKRD1 polypeptide may be encoded by polynucleotide comprising a nucleic acid sequence as set forth in SEQ ID NO: 2 or SEQ ID NO: 4. 30 The expression of the muscle ankyrin repeat protein may be determined by measuring protein expression or mRNA expression levels. The tumour cell may be an ovarian, breast, lung, bladder, testicular, cervical, endometrial or bowel tumour cell or a cell from a head or neck tumour. The chemotherapeutic agent may be a platinum-based agent, an anthracycline or a taxane. 35 The platinum-based chemotherapeutic agent may be cisplatin or a metabolite, derivative or WO 2006/063397 PCT/AU2005/001890 -4 analogue thereof. The analogue may be carboplatin or oxaliplatin. The anthracycline may be adriamycin or a metabolite, derivative or analogue thereof. The taxane may be paclitaxel, docataxel, or metabolite, derivative or analogue thereof. According to a third aspect of the present invention there is provided a method for increasing 5 sensitivity of a tumour cell to one or more chemotherapeutic agents, the method comprising administering to the cell an effective amount of an antagonist of a muscle ankyrin repeat protein. According to a fourth aspect of the present invention there is provided a method for substantially inhibiting expression of a muscle ankyrin repeat protein in tumour cells, the method comprising introducing into the tumour cells an effective amount of an antagonist of the muscle o10 ankyrin repeat protein. According to a fifth aspect of the present invention there is provided a composition for increasing sensitivity of a tumour cell to one or more chemotherapeutic agents, the composition comprising an antagonist of a muscle ankyrin repeat protein together with one or more pharmaceutically acceptable carriers, diluents or adjuvants. 15 According to embodiments of the third, fourth and fifth aspects, the antagonist may be a nucleic acid-based inhibitor, a peptide-based inhibitor or a small molecule inhibitor of the muscle ankyrin repeat protein or polynucleotide encoding the same. The nucleic acid-based inhibitor may be an siRNA molecule or an antisense construct. The muscle ankyrin repeat protein may be ANKRD1 and the nucleic acid-based inhibitor may 20 be an siRNA molecule. The siRNA molecule may comprise a nucleotide sequence as set forth in any one of SEQ ID NOs:5, 6 or 7, or a fragment thereof. According to a sixth aspect of the present invention there is provided an isolated inhibitory nucleic acid construct comprising a nucleotide sequence specific to at least a portion of a polynucleotide encoding a muscle ankyrin repeat protein, wherein the nucleic acid construct 25 substantially inhibits expression of the muscle ankyrin repeat protein in tumour cells. The portion of the polynucleotide may include the coding region of the gene encoding the muscle ankyrin repeat protein and/or one or more regulatory regions of the gene. The inhibitory nucleic acid construct may be located in a vector. The vector may direct transcription of the construct. 30 The inhibitory nucleic acid construct may be an siRNA molecule. In one embodiment, the muscle ankyrin repeat protein is ANKRD1 and the nucleotide sequence of the siRNA molecule comprises: (a) the nucleotide sequence set forth in any one of SEQ ID NOs:5, 6 or 7, or a fragment thereof; or WO 2006/063397 PCT/AU2005/001890 -5 (b) a nucleotide sequence having at least 85% identity to the nucleotide sequence set forth Sin any one of SEQ ID NOs: 5, 6 or 7, or fragment thereof. In other embodiments the nucleotide sequence may have at least 85%, at least 90% identity, or at least 95% identity, to the nucleotide sequence set forth in any one of SEQ ID NOs: 5, 6 or 7, s or fragment thereof. According to a seventh aspect of the present invention there is provided a method for substantially inhibiting expression of a muscle ankyrin repeat protein in tumour cells, the method comprising introducing into the tumour cells an effective amount of an inhibitory nucleic acid construct according to the sixth aspect. 10 According to an eighth aspect of the present invention there is provided a method for increasing sensitivity of tumour cells to one or more chemotherapeutic agents, the method comprising introducing into the tumour cells an effective amount of an inhibitory nucleic acid construct according to the sixth aspect. According to a ninth aspect of the present invention there is provided a composition for use in 15 the method of the seventh or eighth aspects, the composition comprising an inhibitory nucleic acid construct according to the sixth aspect. The composition may further comprise one or more chemotherapeutic agents. The composition may further comprise one or more pharmaceutically acceptable carriers, diluents or adjuvants. 20 Definitions In the context of this specification, the term "comprising" means "including principally, but not necessarily solely". Furthermore, variations of the word "comprising", such as "comprise" and "comprises", have correspondingly varied meanings. The term "polynucleotide" as used herein refers to a single- or double- stranded polymer of 25 deoxyribonucleotide, ribonucleotide bases or known analogues of natural nucleotides, or mixtures thereof. The term includes reference to the specified sequence as well as to the sequence complimentary thereto, unless otherwise indicated. The term "polypeptide" means a polymer made up of amino acids linked together by peptide bonds. The terms "polypeptide" and "protein" are used interchangeably herein, although for the 30 purposes of the present invention a "polypeptide" may constitute a portion of a full length protein. As used herein the term "effective amount" includes within its meaning a non-toxic but sufficient amount of an antisense construct to provide the desired effect. The exact amount required will vary depending on factors such as the level of expression of muscle ankyrin repeat protein in the absence of the construct, the type of tumour to be treated, the severity of the tumour, WO 2006/063397 PCT/AU2005/001890 -6 the drug resistancei1e4el of the tumour, the species being treated, the age and general condition of the subject, the particular construct being administered, the mode of administration and so forth. Thus, it is not possible to specify an exact "effective amount". However, for any given case, an appropriate "effective amount" may be determined by one of ordinary skill in the art using only 5 routine experimentation. In the context of this specification, the term "antagonist" refers to any agent capable of inhibiting the expression or activity of a protein, polypeptide portion thereof, or polynucleotide. Thus, the antagonist may operate to prevent transcription, translation, post-transcriptional or post translational processing or otherwise inhibit the activity of the protein, polypeptide or polynucleotide o10 in any way, via either direct or indirect action. The antagonist may for example be nucleic acid, peptide, any other suitable chemical compound or molecule or any combination of these. Additionally, it will be understood that in indirectly impairing the activity of the protein, polypeptide or polynucleotide, the antagonist may affect the activity of other cellular molecules which may in turn act as regulators of the protein, polypeptide or polynucleotide itself. Similarly, the antagonist may 15 affect the activity of molecules which are themselves subject to regulation or modulation by the protein, polypeptide or polynucleotide. The term "inhibiting" as used herein as it pertains to expression or activity of a muscle ankyrin repeat protein or polynucleotide encoding the same does not necessarily mean completely inhibiting expression. Rather, expression of the protein, polypeptide or polynucleotide is inhibited to 20 an extent, and/or for a time, sufficient to produce the desired effect. The term "expression" as used herein refers interchangeably to expression of a polypeptide or protein and expression of a polynucleotide or gene. Expression of a polynucleotide may be determined, for example, by measuring the production of messenger RNA (mRNA) transcript levels. Expression of a protein or polypeptide may be determined, for example, by immunoassay using an 25 antibody(ies) that bind with the polypeptide. In the context of this specification, the term "activity" as it pertains to a protein, polypeptide or polynucleotide means any cellular function, action, effect or influence exerted by the protein, polypeptide or polynucleotide, either by a nucleic acid sequence or fragment thereof, or by the protein or polypeptide itself or any fragment thereof. 30 In the context of this specification, the term "specific" when used in relation to the nucleotide sequence of an inhibitory nucleic acid construct of the invention means substantially specific, but not necessarily exclusively so. For example, the nucleotide sequence of an inhibitory nucleic acid construct according to the present invention may display less than 100% sequence identity with one particular muscle ankyrin repeat protein-encoding polynucleotide and retain specificity thereto.
WO 2006/063397 PCT/AU2005/001890 -7 Brief Description of the Drawings The present invetion will now be described, by way of example only, with reference to the following drawings: Figure 1 is a schematic illustration of the derivation of cell line models according to the 5 present invention. Mutations were induced in the parental CHO-K1 cell line by treatment with the chemical mutagen, ethyl methane sulfonate (EMS). Mutant clones with increased sensitivity to cisplatin were identified. One of these mutants (MMS-2) was chosen for further characterization and was sub-cloned to ensure a pure, clonal population (CHO-K1-MMS-2-SC2 referred to as SC2 cells). o10 Figure 2. Cisplatin sensitivity of CHO cell lines. Parental CHO-K1 and the platinum sensitive clone SC2 were exposed to a range of cisplatin (CDDP) concentrations for 2 hrs. The drug was then removed and the surviving cell fraction was determined by colony formation over 6 days. Colonies were fixed, stained with 0.1% crystal violet and counted. The results are expressed as a percentage of vehicle-treated control (mean and standard error from duplicate plates in at 15 least three separate experiments) at differing cisplatin (CDDP) concentrations. Figure 3. Expression of ANKRD1 in CHO-K1 cells, cisplatih sensitive (SC2) CHO cells and 10C5 cells with intermediate cisplatin sensitivity by Northern blot. The blot was also probed for 18S RNA as a control for RNA loading and transfer. Figure 4. Amino acid sequence alignment of the Hamster (CHO-K1) ANKRD1 gene product 20 with chicken, human, pig, rabbit and rat ANKRDI sequences. Identical amino acids are shaded black, similar amino acids are shaded grey and unique amino acids are unshaded. The consensus amino acid sequence is also shown. Sequence analysis conducted using GCG Eclustalw and Prettybox programs via the Australian National Genomic Information Service (ANGIS). Figure 5. Increased resistance of SC2 cells to cisplatin following transfection with ANKRDI, 25 Rat ANKRD1 cDNA was transfected into SC2 cells and transfected clones selected. ANKRDI transfected -clones were exposed to 6 [M cisplatin and the surviving fraction was measured by clonogenic assay. Survival in 4 transfected clones (SC2+ANKRD1) was compared with untransfected SC2 cells and empty vector-control transfected SC2 cells (SC2 + vector). Figure 6. Increased sensitivity to cisplatin in CHO-K1 cells following reduction of ANKRD1 30 expression by siRNA. CHO-K1 were transfected with one of three inserts designed to knock-down ANKRD1 expression by targeting regions of the open reading frame encoding the N-terminus (siRNA41; SEQ ID NO: 5), the central portion (siRNA58; SEQ ID NO: 6) or the C-terminus (siRNA77; SEQ ID NO: 7). Pools (P1) of siRNA transfected cells were selected and maintained in hygromycin B. A. ANKRD1 mRNA was measured by real-time PCR. Results are shown as a s35 percentage of the parent CHO-K1 ANKRD1 mRNA levels and corrected for rodent GAPDH WO 2006/063397 PCT/AU2005/001890 -8 (glyceraldehyde-3-phosphate dehydrogenase) and 18S rRNA. B. Cisplatin (CDDP) sensitivity in CHO-K1 and siRNA transfected cells was measured by clonogenic assay. The results are expressed as the mean and standard error of duplicate plates from at least three separate experiments. 5 Figure 7. Expression of ANKRD1 in ovarian and breast cell lines. RNA was extracted from a range of normal and malignant ovarian and breast cell lines. ANKRD1 expression was measured by real-time PCR. A. Results are shown as relative mRNA levels, corrected for human P0 (acidic phosphoprotein P0; also known as 36B4, part of the 60S ribosomal RNA complex) and 18S rRNA. All cell lines are human. HOSE 17.1 and BRE-80 are normal ovarian and breast cell lines o10 respectively; the remaining cell lines are malignant. B. Cisplatin (CDDP) sensitivity was determined for 13 of the cell lines. Survival was determined by MTS assay after exposing the cells to drug for 72 hr. The IC37 is shown for each cell line, results are the mean determined from between two and seven independent experiments. Asterix indicates IC37 not determined. C. Association between growth inhibition by cisplatin (IC37) and expression of ANKRD1 mRNA in 13 cell lines. Regression 15is analysis was performed on cell lines expressing detectable levels of ANKRD1 (triangles; R 2 = 0.9827, Linear Regression Analysis) and did not include cell lines with ANKRD1 levels at or below the level of detection (closed squares). Figure 8. Expression of ANKRD1 in human ovarian tumours and association with response to chemotherapy. Two subgroups of ovarian carcinoma patients were selected from the extreme 20 ends of the spectrum of clinical outcome. Cases were designated 'chemo-sensitive' if the patient had a sustained response to therapy (>4 years) and 'chemo-resistant' if the tumour progressed while the patient was on primary platinum-based treatment. The two cohorts were not different with respect to other clinical features known to be associated with patient outcome such as surgical stage and extent of residual disease following debulking surgery (Table 2). RNA was extracted 25 from frozen ovarian tumour specimens. In one patient (number 146) RNA was extracted from tissue collected from two anatomical sites, the primary ovarian tumour and a metastasis to the omentum. ANKRD1 expression was measured by real-time PCR and results are shown as relative mRNA levels, corrected for human P0 and 18S rRNA. The ANKRD1 mRNA levels for case 146 are represented by triangles, where the filled triangle represents the primary tumour and the open 30 triangle represents the metastasis. Samples from the other two patients in the chemo-sensitive cohort are represented by filled squares, and filled circles represent the samples from the chemo resistant patients. Figure 9. Effect of carboplatin (A) and oxaliplatin (B) on cell survival in CHO-K1 cells (diamonds) and SC2 cells (squares). Cell survival was measured by modified MTS assay. Results 35 are expressed as a percentage of the vehicle-treated control, as a mean and standard error of WO 2006/063397 PCT/AU2005/001890 -9 triplicate wells from six independent experiments. Results were subjected to a paired t-test where * is p<0.05 and ** is p<0.005. The amino acid sequence set forth in SEQ ID NO:1 is the amino acid sequence of the human ANKRD1 protein, deposited under GenBank database accession number NM_014391. s The nucleotide sequence set forth in SEQ ID NO:2 is the nucleotide sequence of the gene encoding the human ANKRD1 protein, deposited under GenBank database accession number NM_014391. The amino acid sequence set forth in SEQ ID NO:3 is the amino acid sequence of the ANKRD1 protein from the Chinese Hamster Ovary cell line CHO-K1. to The nucleotide sequence set forth in SEQ ID NO:4 is the nucleotide sequence of the gene encoding the ANKRD1 protein from the Chinese Hamster Ovary cell line CHO-KI. The nucleotide sequences set forth in SEQ ID NOs:5 to 7 are nucleotide sequences of the siRNA molecules, siRNA41, siRNA58 and siRNA77 respectively, as disclosed herein. Best Mode of Performing the Invention is The present inventors have used a unique cell line model and stringent differential display strategy to identify gene determinants of platinum-based drug sensitivity. The inventors' utilized a cell line model where random mutagenesis was used to create clones that are exquisitely sensitive to platinum-based therapy. In this model, neither the parent cell line (which is intrinsically platinum resistant) nor the sensitive clones have ever been exposed to chemotherapeutic drugs. Thus, 20 rather than seeking to identify genes associated with resistance, the approach has been to identify gene determinants of drug sensitivity - as the ultimate clinical goal is to increase the sensitivity of tumors to chemotherapeutic agents. One of the genes identified by this strategy encodes ankyrin repeat domain 1 (cardiac muscle) protein (ANKRDI), also known as cardiac ankyrin repeat protein (CARP) and cardiac adriamycin-responsive protein. ANKRD1 is also homologous to the human 25 protein C-193 (cytokine inducible nuclear protein). ANKRD1 belongs to a family of structurally related proteins with conserved biological function known as muscle ankyrin repeat proteins (MARPs), the known members of which currently include ANKRD1, ANKRD2 (also known as Arpp) and ANKRD23 (also known as DARP (diabetes-related ankyrin repeat protein)) (see Miller et al., 2003; Kojic et al., 2004). Each of these three proteins contains an N-terminally-located nuclear 30 localization signal and four C-terminal ankyrin repeat motifs of 33 amino acid residues, the second of which represents a distinct subclass (Miller et al., 2003). The term MARP is used herein to refer, collectively, to the family of muscle ankyrin repeat proteins exemplified by ANKRD1 (CARP), ANKRD2/Arpp and ANKRD23 (DARP).
WO 2006/063397 PCT/AU2005/001890 -10 The ANKRD1 gene encodes a nuclear co-repressor for cardiac gene expression; it is downstream of the cardiac homeobox gene, Nkx 2.5 and is an early marker of the cardiac muscle lineage. ANKRD1 represses muscle-specific promoters in cardiomyocytes, interacting with the transcription factor YB1 and repressing expression of the ventricular isoform of myosin light chain s 2, a trial natriuretic factor, and cardiac troponin C genes. It is developmentally downregulated in cardiac tissue and is re-expressed in several cardiac pathologies including cardiac hypertrophy (Baudet, 2003). ANKRDI is also induced in regenerating myofibers of patients with Duchenne muscular dystrophy (Nakada et al., 2003). ANKRD1 is most abundant in the heart but it is also found in skeletal muscle, lung, liver, kidney, prostate and endothelial cells (Nakada et al., 2003; 10 Zou et al., 1997; Chu et al., 1995). As disclosed herein the present inventors have now found that ANKRDI is expressed in normal human ovarian epithelial cells, in ovarian and breast cancer cell lines and ovarian tumour specimens. ANKRD1 was however found to be barely detectable in a mutated ovarian cell line (SC2) with unusual sensitivity to cisplatin. In addition gene transfection and gene knock-down 5is experiments (siRNA) revealed that cisplatin cytotoxicity could be altered by changing ANKRD1 expression levels. These results indicate that ANKRDI, or a molecular pathway involving ANKRD1, has a direct pro-survival effect following exposure to cisplatin. Neither ANKRDI nor its homologues have previously been associated with ovarian cancer or response to platinum therapy. Accordingly, one aspect of the present invention provides a method for determining the level 20 of sensitivity/resistance of a tumour cell to one or more chemotherapeutic agents, the method comprising measuring the level or expression of a muscle ankyrin repeat protein in the tumour cell. The invention also provides a method for assessing the responsiveness of a patient to one or more chemotherapeutic agents. Further, another aspect of the invention provides a method for increasing sensitivity of a tumour cell to one or more chemotherapeutic agents, the method 25 comprising administering to the cell an effective amount of an antagonist of a muscle ankyrin repeat protein. Although exemplified herein in relation to the muscle ankyrin repeat protein ANKRD1, those skilled in the art will readily appreciate that the present invention is not so limited and extends to other members, both known and yet to be elucidated, of the family of related muscle ankyrin repeat 30 proteins, including but not limited to ANKRD2/Arpp and ANKRD23 (DARP). Muscle ankyrin repeat proteins and expression thereof Typically the polypeptides of and polynucleotides encoding muscle ankyrin repeat proteins (MARPs) to which the methods and compositions of the present invention relate are the human protein and gene. The amino acid sequence of the human ANKRD1 protein is shown in SEQ ID 35 NO:1 (GenBank Accession No. NM_014391), and the nucleotide sequence of the human ANKRD1 WO 2006/063397 PCT/AU2005/001890 -11 gene is shown in SEQ ID NO:2 (GenBank Accession No. NM_014391). Also disclosed herein are the CHO ANKRD1 protein and nucleotide sequences, set forth in SEQ ID NOs: 3 and 4, respectively. The nucleotide and polypeptide sequences of human ANKRD2/Arpp are found under GenBank Accession No. NM_020349, and the nucleotide and polypeptide sequences of human s ANKRD23 (DARP) are found under GenBank Accession No. NM_144994. According to embodiments of the invention, the ANKRD1 polypeptide may have the amino sequence as set forth in SEQ ID NO:1 or 3 or display sufficient sequence identity thereto to hybridise to the sequence of SEQ ID NO:1 or 3. In alternative embodiments, the nucleotide sequence of the polynucleotide may share at least 50%, 60%, 70%, 80%, 85%, 90%, 96%, 97%, o10 98% or 99% identity with the sequence set forth in SEQ ID NO:3. According to embodiments of the invention, the ANKRD1 polynucleotide may have the nucleotide sequence as set forth in SEQ ID NO:2 or 4 or display sufficient sequence identity thereto to hybridise to the sequence of SEQ ID NO:2 or 4. In alternative embodiments, the nucleotide sequence of the polynucleotide may share at least 30%, 40%, 50%, 60%, 70%, 80%, 85%, 90%, 15 96%, 97%, 98% or 99% identity with the sequence set forth in SEQ ID NO:2 or 4. Within the scope of the terms "protein", "polypeptide" and "polynucleotide" as used herein are fragments and variants thereof. The term "fragment" refers to a nucleic acid or polypeptide sequence that encodes a constituent or is a constituent of a full-length MARP protein. In terms of the polypeptide the 20 fragment possesses qualitative biological activity in common with the full-length protein. The term "variant" as used herein refers to substantially similar sequences. Generally, nucleic acid sequence variants encode polypeptides which possess qualitative biological activity in common. Generally, polypeptide sequence variants also possess qualitative biological activity in common. Further, these polypeptide sequence variants may share at least 50%, 55%, 60%, 65%, 25 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98% or 99% sequence identity. Further, a variant polypeptide may include analogues, wherein the term "analogue" means a polypeptide which is a derivative of a MARP, which derivative comprises addition, deletion, substitution of one or more amino acids, such that the polypeptide retains substantially the same function as the native MARP from which it is derived. The term "conservative amino acid 30 substitution" refers to a substitution or replacement of one amino acid for another amino acid with similar properties within a polypeptide chain (primary sequence of a protein). For example, the substitution of the charged amino acid glutamic acid (Glu) for the similarly charged amino acid aspartic acid (Asp) would be a conservative amino acid substitution. Expression. of polynucleotides, proteins or polypeptides may be determined by any one of a 35 number of techniques well known to those skilled in the art. For example, expression may be WO 2006/063397 PCT/AU2005/001890 -12 determined by assaying mRNA transcript abundance in a sample. mRNA abundance may be measured, for example, by reverse transcriptase-PCR. Alternatively expression of a protein or polypeptide may be determined using an antibody that binds to the protein or polypeptide or a fragment thereof, using a technique such as enzyme-linked immunosorbent assay (ELISA). 5 Antagonists Embodiments of the present invention provide methods and compositions for inhibiting the expression of a MARP using an antagonist thereof. Typically the antagonist may be nucleic-acid based, peptide-based or other suitable chemical compound. In particular embodiments the antagonist is a nucleic-acid based inhibitor of expression of o10 polynucleotide encoding a MARP or a fragment thereof. Suitable molecules include small interfering RNA (siRNA) species, antisense constructs, such as antisense oligonucleotides, and catalytic antisense nucleic acid constructs. Suitable molecules can be manufactured by chemical synthesis, recombinant DNA procedures or, in the case of antisense RNA, by transcription in vitro or in vivo when linked to a promoter, by methods known to those skilled in the art. s15 One suitable technology for inhibiting gene expression, known as RNA interference (RNAi), (see, eg. Chuang et al. (2000) PNAS USA 97: 4985) may be used for the purposes of the present invention, according to known methods in the art (for example Fire et al. (1998) Nature 391: 806 811; Hammond, et al. (2001) Nature Rev, Genet. 2:110-1119; Hammond et al. (2000) Nature 404: 293-296; Bernstein et aL. (2001) Nature 409: 363-366; Elbashir et al (2001) Nature 411: 494-498; 20 WO 99/49029 and WO 01/70949, the disclosures of which are incorporated herein by reference), to inhibit the expression of MARPs. RNAi refers to a means of selective post-transcriptional gene silencing by destruction of specific mRNA by small interfering RNA molecules (siRNA). The siRNA is typically generated by cleavage of double stranded RNA, where one strand is identical to the message to be inactivated. Double-stranded RNA molecules may be synthesised in which one 25 strand is identical to a specific region of the mRNA transcript of the MARP of interest and introduced directly. Alternatively corresponding dsDNA can be employed, which, once presented intracellularly is converted into dsRNA. Methods for the synthesis of suitable siRNA molecules for use in RNAi and for achieving post-transcriptional gene silencing are known to those of skill in the art. The skilled addressee will appreciate that a range of suitable siRNA constructs capable of 30 inhibiting the expression of a MARP can be identified and generated based on knowledge of the sequence of the gene in question using routine procedures known to those skilled in the art without undue experimentation. Those skilled in the art will appreciate that there need not necessarily be 100% nucleotide sequence match between the target sequence and the siRNA sequence. The capacity for 35 mismatch therebetween is dependent largely on the location of the mismatch within the sequences.
WO 2006/063397 PCT/AU2005/001890 -13 In some instances, mismatches of 2 or 3 nucleotide may be acceptable but in other instances a single nucleotide mismatch is enough to negate the effectiveness of the siRNA. The suitability of a particular siRNA molecule may be determined using routine procedures known to those skilled in the art without undue experimentation. By way of example, the nucleotide sequences of exemplary 5 siRNA molecules for the inhibition of ANKRD1 are set forth in any one of SEQ ID NOs:5, 6 or 7. Suitable siRNA molecules may display at least 80% sequence identity to the sequences set forth in SEQ ID NOs: 5, 6 or 7. In alternative embodiments, the nucleotide sequence of the siRNA molecule may share at least 85%, at least 90%, or at least 95% identity with the sequence set forth in any one of SEQ ID NOs:5, 6 or 7. Those skilled in the art will appreciate that one or base o10 substitutions, additions or deletions of these sequences may be made in generating a sequence of at least 80% nucleotide sequence identity, at least 85% nucleotide sequence identity, at least 90% nucleotide sequence identity or at least 95% nucleotide sequence identity. Further, the skilled addressee will appreciate that a number of other suitable siRNA constructs capable of inhibiting the expression of ANKRD1 can be identified and generated using routine procedures known to those 15 skilled in the art without undue experimentation. Sequences of antisense constructs of the invention may be derived from various regions of a MARP gene. Antisense constructs may be designed to target and bind to regulatory regions of the nucleotide sequence or to coding (exon) or non-coding (intron) sequences. Antisense constructs of the invention may be generated which are at least substantially complementary across their length 20 to a region of the gene in question. Binding of an antisense construct to its complementary cellular sequence may interfere with transcription, RNA processing, transport, translation and/or mRNA stability. Antisense constructs of the present invention may be derived from the human MARP gene, or non-human animal variants thereof. For example, antisense constructs derived from non-human 25 genes having at least 50% sequence identity with the human gene and can be used in the methods of the invention for administration to tumours of humans and/or non-human animals. Non-human MARP genes may have at least 60%, at least 70%, at least 80% or at least 90% sequence identity with their human homologue. Suitable antisense oligonucleotides may be prepared by methods well known to those of skill 30 in the art. Typically antisense oligonucleotides will be synthesized on automated synthesizers. Suitable antisense oligonucleotides may include modifications designed to improve their delivery into cells, their stability once inside a cell, and/or their binding to the appropriate target. For example, the antisense oligonucleotide may be modified by the addition of one or more phosphorothioate linkages, or the inclusion of one or morpholine rings into the backbone.
WO 2006/063397 PCT/AU2005/001890 -14 In particular embodiments of the invention suitable inhibitory nucleic acid molecules may be administered to the tumour cells in a vector. The vector may be a plasmid vector, a viral vector, or any other suitable vehicle adapted for the insertion and foreign sequences and introduction into eukaryotic cells. Preferably the vector is an expression vector capable of directing the transcription 5 of the DNA sequence of an inhibitory nucleic acid molecule of the invention into RNA. Viral expression vectors include, for example, epstein-barr virus-, bovine papilloma virus-, adenovirus and adeno-associated virus-based vectors. In one embodiment, the vector is episomal. The use of a suitable episomal vector provides a means of maintaining the inhibitory nucleic acid molecule in the tumour cells in high copy number extra-chromosomally thereby eliminating potential effects of 10 chromosomal integration. A further means of substantially inhibiting MARP gene expression may be achieved by introducing catalytic antisense nucleic acid constructs, such as ribozymes, which are capable of cleaving RNA transcripts and thereby preventing the production of wildtype protein. Ribozymes are targeted to and anneal with a particular sequence by virtue of two regions of sequence 15 complementarity to the target flanking the ribozyme catalytic site. After binding the ribozyme cleaves the target in a site-specific manner. The design and testing of ribozymes which specifically recognize and cleave MARP sequences can be achieved by techniques well known to those in the art (for example Lieber and Strauss, (1995) Mo. Cell. Biol. 15:540-551, the disclosure of which is incorporated herein by reference). 20 Alternative antagonists of MARPs may include antibodies. Suitable antibodies include, but are not limited to polyclonal antibodies, monoclonal antibodies, chimeric antibodies, humanised antibodies, single chain antibodies and Fab fragments. Antibodies may be prepared from discrete regions or fragments of the polypeptide of interest. An antigenic polypeptide contains at least about 5, and preferably at least about 10, amino acids. 25 Methods for the generation of suitable antibodies will be readily appreciated by those skilled in the art. For example, a suitable monoclonal antibody, typically containing Fab portions, may be prepared using. the hybridoma technology described in Antibodies-A Laboratory Manual, Harlow and Lane, eds. Cold Spring Harbor Laboratory, N.Y. (1988), the disclosure of which is incorporated herein by reference. 30 Similarly, there are various procedures known in the art which may be used for the production of polyclonal antibodies to polypeptides of interest as disclosed herein. For the production of polyclonal antibodies, various host animals, including but not limited to rabbits, mice, rats, sheep, goats, etc, can be immunized by injection with a polypeptide, or fragment or analogue thereof. Further, the polypeptide or fragment or analogue thereof can be. conjugated to an 35 immunogenic carrier, e.g., bovine serum albumin (BSA) or keyhole limpet hemocyanin (KLH). Also, WO 2006/063397 PCT/AU2005/001890 -15 various adjuvants may be used to increase the immunological response, including but not limited to Freund's (complete and incomplete), mineral gels such as aluminium hydroxide, surface active substances such as lysolecithin, pluronic polyols, polyanions, peptides, oil emulsions, keyhole limpet hemocyanins, dinitrophenol, and potentially useful human adjuvants such as BCG (Bacillus 5 Calmette-Guerin) and Corynebacterium parvum. Screening for the desired antibody can also be accomplished by a variety of techniques known in the art. Assays for immunospecific binding of antibodies may include, but are not limited to, radioimmunoassays, ELISAs (enzyme-linked immunosorbent assay), sandwich immunoassays, immunoradiometric assays, gel diffusion precipitation reactions, immunodiffusion assays, in situ o10 immunoassays, Western blots, precipitation reactions, agglutination assays, complement fixation assays, immunofluorescence assays, protein A assays, and immunoelectrophoresis assays, and the like (see, for example, Ausubel et al., eds, 1994, Current Protocols in Molecular Biology, Vol. 1, John Wiley & Sons, Inc., New York). Antibody binding may be detected by virtue of a detectable label on the primary antibody. Alternatively, the primary antibody may be detected by virtue of its 15 binding with a secondary antibody or reagent which is appropriately labelled. A variety of methods are known in the art for detecting binding in an immunoassay and are within the scope of the present invention. Also included within the scope of the present invention are alternative forms of inhibition of MARP expression, including, for example, small molecule or other non-nucleic acid or non 20 proteinaceous inhibitors. Such inhibitors may be identified by those skilled in the art by screening using routine techniques. Chemotherapeutic Agents According to methods of the invention, antisense constructs of the invention and other antagonists may be employed to increase the sensitivity (decrease the resistance) of tumour cells 25 to chemotherapeutic agents, such as platinum-based agents, anthracyclines and taxanes. Platinum-based agents include those agents which inhibit tumour cell growth by binding to DNA and inducing DNA damage. Several numbers of platinum-based drugs have been used as chemotherapeutic agents. For the purposes of the present invention, examples of such drugs include, but are not limited to, cisplatin and metabolites, derivatives or analogues thereof. Such 30 derivatives and analogues include carboplatin and oxaliplatin. Exemplary anthracyclines include adriamycin and metabolites, derivatives and analogues thereof. Exemplary taxanes include paclitaxel, docataxel, and metabolites, derivatives and analogues thereof. Those skilled in the art will appreciate that the chemotherapeutic agents to which the present invention refers are not limited to the above-mentioned specific agents but include any compound 35 suitable for chemotherapeutic use.
WO 2006/063397 PCT/AU2005/001890 -16 Tumours Embodiments of the present invention provide methods for the determination and the alteration of levels of resistance/sensitivity of tumour cells to chemotherapeutic agents, predicated on the inventors' finding of ANKRDI expression in human ovarian and breast tumours and of s modulation in the sensitivity of tumour cells to cisplatin in response to modulation of the expression of ANKRDI. Accordingly the methods and compositions of the present invention relate to the analysis and treatment of a variety of tumour cell types, depending on the nature of the chemotherapeutic agent the resistance to which assessment or alteration is desired. For example in the case of platinum-based chemotherapeutic agents, the tumour cells may be ovarian, breast, 1o lung, bladder, testicular, cervical, endometrial or bowel tumour cells or cells from a head or neck tumour. Methods of Treatment Embodiments of the present invention relate to the use of antagonists of the invention in methods and compositions for treating individuals having cancer, the individual either being in need 15is of or undergoing platinum-based chemotherapy. Accordingly, the present invention contemplates the administration of antagonists of the invention to such individuals having cancer in order to inhibit ANKRD1 expression thereby increasing sensitivity of the cancer cells to one or more platinum-based chemotherapeutic agents. Accordingly, antagonists of the invention may be administered in combination therapy with such platinum-based chemotherapeutic agents. For such 20 combination therapies, each component of the combination therapy may be administered at the same time, or sequentially in any order, or at different times, so as to provide the desired therapeutic effect. One or more suitable antagonist may be combined with one or more suitable platinum-based chemotherapeutic agent in a single composition, optionally also comprising one or more pharmaceutically acceptable carriers, diluents or adjuvants. 25 It will be understood that the specific dose level of a composition of the invention for any particular individual will depend upon a variety of factors including, for example, the activity of the specific antagonists and/or platinum-based chemotherapeutic agents employed, the age, body weight, general health and diet of the individual to be treated, the time of administration, rate of excretion, and combination with any other treatment or therapy. Single or multiple administrations 30 can be carried out with dose levels and pattern being selected by the treating physician. Examples of pharmaceutically acceptable carriers or diluents are demineralised or distilled water; saline solution; vegetable based oils such as peanut oil, safflower oil, olive oil, cottonseed oil, maize oil, sesame oils such as peanut oil, safflower oil, olive oil, cottonseed oil, maize oil, sesame oil, arachis oil or coconut oil; silicone oils, including polysiloxanes, such as methyl 35 polysiloxane, phenyl polysiloxane and methylphenyl polysolpoxane; volatile silicones; mineral oils WO 2006/063397 PCT/AU2005/001890 -17 such as liquid paraffin, soft paraffin or squalane; cellulose derivatives such as methyl cellulose, ethyl cellulose, carboxymethylcellulose, sodium carboxymethylcellulose or hydroxypropylmethylcellulose; lower alkanols, for example ethanol or iso-propanol; lower aralkanols; lower polyalkylene glycols or lower alkylene glycols, for example polyethylene glycol, 5 polypropylene glycol, ethylene glycol, propylene glycol, 1,3-butylene glycol or glycerin; fatty acid esters such as isopropyl palmitate, isopropyl myristate or ethyl oleate; polyvinylpyrridone; agar; carrageenan; gum tragacanth or gum acacia, and petroleum jelly. Typically, the carrier or carriers will form from 10% to 99.9% by weight of the compositions. The compositions of the invention may be in a form suitable for parenteral administration, or o10 in the form of a formulation suitable for oral ingestion (such as capsules, tablets, caplets, elixirs, for example). For administration as an injectable solution or suspension, non-toxic parenterally acceptable diluents or carriers can include, Ringer's solution, isotonic saline, phosphate buffered saline, ethanol and 1,2 propylene glycol. 15 Some examples of suitable carriers, diluents, excipients and adjuvants for oral use include peanut oil, liquid paraffin, sodium carboxymethylcellulose, methylcellulose, sodium alginate, gum acacia, gum tragacanth, dextrose, sucrose, sorbitol, mannitol, gelatine and lecithin. In addition these oral formulations may contain suitable flavouring and colourings agents. When used in capsule form the capsules may be coated with compounds such as glyceryl monostearate or 20 glyceryl distearate which delay disintegration. Adjuvants typically include emollients, emulsifiers, thickening agents, preservatives, bactericides and buffering agents. Solid forms for oral administration may contain binders acceptable in human and veterinary pharmaceutical practice, sweeteners, disintegrating agents, diluents, flavourings, coating agents, 25 preservatives, lubricants and/or time delay agents. Suitable binders include gum acacia, gelatine, corn starch, gum tragacanth, sodium alginate,. carboxymethylcellulose or polyethylene glycol. Suitable sweeteners include sucrose, lactose, glucose, aspartame or saccharine. Suitable disintegrating agents include corn starch, methylcellulose, polyvinylpyrrolidone, guar gum, xanthan gum, bentonite, alginic acid or agar. Suitable diluents include lactose, sorbitol, mannitol, dextrose, 30 kaolin, cellulose, calcium carbonate, calcium silicate or dicalcium phosphate. Suitable flavouring agents include peppermint oil, oil of wintergreen, cherry, orange or raspberry flavouring. Suitable coating agents include polymers or copolymers of acrylic acid and/or methacrylic acid and/or their esters, waxes, fatty alcohols, zein, shellac or gluten. Suitable preservatives include sodium benzoate, vitamin E, alpha-tocopherol, ascorbic acid, methyl paraben, propyl paraben or sodium WO 2006/063397 PCT/AU2005/001890 -18 bisulphite. Suitable lubricants include magnesium stearate, stearic acid, sodium oleate, sodium chloride or talc. Suitable time delay agents include glyceryl monostearate or glyceryl distearate. Liquid forms for oral administration may contain, in addition to the above agents, a liquid carrier. Suitable liquid carriers include water, oils such as olive oil, peanut oil, sesame oil, 5 sunflower oil, safflower oil, arachis oil, coconut oil, liquid paraffin, ethylene glycol, propylene glycol, polyethylene glycol, ethanol, propanol, isopropanol, glycerol, fatty alcohols, triglycerides or mixtures thereof. Suspensions for oral administration may further comprise dispersing agents and/or suspending agents. Suitable suspending agents include sodium carboxymethylcellulose, 10 methylcellulose, hydroxypropylmethyl-cellulose, poly-vinyl-pyrrolidone, sodium alginate or acetyl alcohol. Suitable dispersing agents include lecithin, polyoxyethylene esters of fatty acids such as stearic acid, polyoxyethylene sorbitol mono- or di-oleate, -stearate or -laurate, polyoxyethylene sorbitan mono- or di-oleate, -stearate or -laurate and the like. The emulsions for oral administration may further comprise one or more emulsifying agents. s15 Suitable emulsifying agents include dispersing agents as exemplified above or natural gums such as guar gum, gum acacia or gum tragacanth. Methods for preparing parenterally administrable compositions are apparent to those skilled in the art, and are described in more detail in, for example, Remington's Pharmaceutical Science, 15th ed., Mack Publishing Company, Easton, Pa., hereby incorporated by reference herein. 20 The composition may incorporate any suitable surfactant such as an anionic, cationic or non-ionic surfactant such as sorbitan esters or polyoxyethylene derivatives thereof. Suspending agents such as natural gums, cellulose derivatives or inorganic materials such as silicaceous silicas; and other ingredients such as lanolin, may also be included. The compositions may also be administered in the form of liposomes. Liposomes are 25 generally derived from phospholipids or other lipid substances, and are formed by mono- or multi lamellar hydrated liquid crystals that are dispersed in an aqueous medium. Any non-toxic, physiologically acceptable and metabolisable lipid capable of forming liposomes can be used. The compositions in liposome form may contain stabilisers, preservatives, excipients and the like. The preferred lipids are the phospholipids and the phosphatidyl cholines (lecithins), both natural and 30 synthetic. Methods to form liposomes are known in the art, and in relation to this specific reference is made to: Prescott, Ed., Methods in Cell Biology, Volume XIV, Academic Press, New York, N.Y. (1976), p. 33 et seq., the contents of which are incorporated herein by reference.
WO 2006/063397 PCT/AU2005/001890 -19 The present invention will now be further described in greater detail by reference to the following specific examples, which should not be construed as in any way limiting the scope of the invention. Examples 5 Example 1 - Cell line model of platinum sensitivity The present inventors have used a novel approach to identify genes that determine sensitivity to platinum-based chemotherapeutic agents. This strategy utilized the mutant Chinese hamster ovary (CHO) cell line SC2 which is more sensitive to platinum than the parent CHO-K1 cell line. Parental CHO-K1 and mutant CHO cell lines were provided as a gift from Dr I Hickson and Dr 1o C Robson (Imperial Cancer Research Fund, Oxford UK) (Robson, C, et al, 1985). The strategy adopted is outlined in Figure 1. CHO-K1 cells were mutagenised with ethyl methane sulphonate (EMS). Mutants with altered sensitivity to cisplatin were isolated using replica plating. Colonies were grown in agar then transferred onto replica plates - untreated or drug selection plates. Clones growing on untreated but not drug treated plates were isolated and characterized. One 5is exquisitely cisplatin-sensitive mutant, MMS-2, was utilized for further analysis and was subcloned to ensure a pure clonal population of cells (CHO-K1-MMS-2-SC2, referred to herein as SC2 cells). Parental CHO-K1 and the platinum-sensitive clone SC2 were exposed to a range of cisplatin concentrations for 2 hrs. The drug was then removed and the surviving cell fraction was determined by colony formation over 6 days. Colonies were fixed, stained with 0.1% crystal violet 20 and counted. Exposure to 10 uM cisplatin for 2 hr results in less than 2% cell survival of SC2 cells, compared with greater than 80% survival in CHO-K1 cells (see Figure 2). The mechanisms underlying cisplatin sensitivity in this mutant were then investigated and at least two defects related to cisplatin response, increased drug influx and defective DNA repair, were identified (Segelov, E. et al, 1998). 25 Example 2- Identification of a new regulator of platinum sensitivity A stringent differential display PCR strategy was used to isolate differentially expressed genes in the pair of CHO lines with differing platinum sensitivity, CHO-K1 and SC2. RNA was extracted from CHO-K1 cells, SC2 cells and a partial revertant (10C5) for comparison. 30 Differential display-PCR (DD-PCR) was used to examine differential gene expression between CHO K1 and SC2 cells. RNA was extracted from exponentially growing early passage cells. Primer sets for differential display were obtained commercially (Genomyx, Foster City, CA, USA). Reverse transcription (RT) was performed using a DD anchor primer (final concentration 2 pM) and 2 pl of RNA from each clone freshly diluted to a concentration of 100 ng/il. The reaction WO 2006/063397 PCT/AU2005/001890 - 20 mix included dNTP mix (final concentration 25 jiM each), DTT (final concentration 10 mM), RNase inhibitor (final concentration 1 U/Il, RNasin, Promega, Madison, WI, USA), RT enzyme (final concentration 2 U/p l, Superscript II Life Technologies, Gibco BRL, Grand Island, NY, USA), RT enzyme buffer (lx) and sterile water to 20 gl. Negative controls lacking either RT enzyme or RNA 5 template were performed as was a positive control using HeLa RNA supplied by the manufacturer (Genomyx). The RT reaction was performed in a hot lid thermal cycler (Hybaid, Integrated Sciences, Willoughby, NSW, Australia) and conditions were 250 for 10 min, 420C for 60 min, 7000 for 15 min then 40C. RT products were either used immediately or stored at -200C. PCR mix consisted of 3' 10 anchor primer and 5' arbitrary primer (final concentration 0.2 pM each, Genomyx), dNTP mix (final concentration 20 gM each, Genomyx), PCR buffer (final concentration lx), a[ 3 3 P]dATP (final concentration 0.125 pCi/pl), Taq polymerase (final concentration 0.05 units/pl, AmpliTaq, Perkin Elmer, NJ, USA), 2 Il RT product and DNase free water to 20 pl. PCR cycling parameters were: 95 0 C for 2 min; 92 0 C for 15 sec, 460C for 30 sec, 720C for 2 min (four cycles); 920C for 15 sec, 15 600C for 30 sec, 720C for 2 min (25 cycles); and extension at 720C for 7 min. DD-PCR products were resolved on a 6% denaturing polyacrylamide gel. To reduce the risk of false negatives independent, duplicate harvests of RNA were performed from the CHO clones to be compared and the RT-PCR reaction was performed in duplicate on each of these harvests. Thus for each anchor primer/arbitrary primer combination, a total of 4 lanes for each clone were run 20 on a gel. Bands were only selected if the difference between clones was consistent across at least 3 out of the 4 replicates. Differential display analysis revealed 28 differentially expressed bands confirmed on duplicate RNA samples. The selected bands were cut from the dried polyacrylamide gel, amplified by PCR, isolated by cloning into a bacterial vector (pGEMT-Easy Cloning Kit; Promega) and 25 sequenced using an ABI automated sequencer (PE Applied Biosystems, Foster City, CA, USA). All sequence searches were performed using data and tools available at ANGIS (Australian National Genomic Information Service, (http://www.angis.org.au) and the NCBI website (http://www.ncbi.nlm.nih.pov/). Four genes were confirmed to be consistently altered in either the sensitive or resistant cell lines and their identity revealed by sequence analysis. These genes and 30 their expression relative to cisplatin resistance are indicated in Table 1 below.
WO 2006/063397 PCT/AU2005/001890 -21 Table 1 GENE DIRECTION OF FOLD CHANGEa CHANGE Ribosomal protein L19 Increased in resistant cells 2x Cardiac adriamycin response protein (ANKRD1) increased in resistant cells 9.3x Ribosomal protein L39 Decreased in resistant cells 2x EST 25111 lb Decreased in resistant cells 1.7x a. Determined by Northern Blot Analysis or Real-time quantitative PCR b. Bcr/Abl regulated protein 5 One of the differentially expressed genes was found to be homologous to ANKRD1 (cardiac ankyrin repeat protein (Zou et al. 1997); known also as cardiac adriamycin-responsive protein (Jeyaseelan et al., 1997) and homologous to the human gene, C-193 (Chu et al., 1995). Expression of ANKRD1 in CHO-K1 and SC2 cells was determined by Northern blot analysis. Total RNA was isolated by lysis in guanidinium isothiocyanate solution followed by CsCI o10 centrifugation using standard methods (Sambrook et al., 1989) or by using Tri-reagent according to manufacturer's instructions (Sigma Aldrich, Steinheim, Germany) and Northern blot analysis was performed by standard techniques. An oligonucleotide probe to ANKRD1 was designed from common sequence of the published human, rat and mouse ANKRD1 cDNA (5'-GTC CAG GGG TTC AGC CAC AA-3'; SEQ ID NO:8). An oligonucleotide designed to the 18S rRNA sequence (5' 15is ACG GTA TCT GAT CGT CTT CGA ACC-3'; SEQ ID NO:9) was used to control for equal loading and transfer on Northern blots. Probes were end-labeled with (y- 3 2 P)ATP (5,000 Ci/mmol) using T4 polynucleotide enzyme (Promega) and unincorporated label was removed using G25 Sephadex spin column. Following hybridisation the filters were washed then exposed to either radiographic film (BioMax, Eastman 20 Kodak, Rochester, NY, USA) or a phosphoimager screen (Molecular Dynamics, Sunnyvale, CA, USA). Lane intensity was compared using the Image Quant program (Molecular Dynamics). Filters were then stripped and reprobed for 18S. By Northern blotting ANKRD1 was confirmed to be expressed in CHO-K1 and to be undetectable in SC2 cells (Figure 3). By real-time PCR (see Example 6) ANKRD1 expression was 25 found to be 9.3 fold higher in the parental cell line (0.185 ng in CHO-K1 compared to 0.02 ng in SC2) (Table 1). The full length ANKRD1 gene from CHO-K1 was sequenced. The full-length open reading frame was obtained using the SMART T M 5'-; 3'-RACE cDNA Amplification kit (BD Biosciences Clontech, Palo Alto, CA, USA). RACE was carried out according to the manufacturer's protocol. 30 The two initial primers, forward 5'-AAC ATG ATG GTG CTG AGA GTA GAG GAG CCG-3' (SEQ ID WO 2006/063397 PCT/AU2005/001890 - 22 NO:10) and reverse 5'-GCA CCA TCA TGT TGG CAG CAG TGA GTC T-3' (SEQ ID NO:11), were designed from the sequence isolated from the DD-PCR. An additional internal primer was required to obtain the full-length sequence, 5'-TGC GCT GGA GAA CAA ACT GCC AGT TG-3' (SEQ ID NO:12) at position 383 bp (primers synthesized by Sigma Genosys, Castle Hill, NSW, Australia). 5 The full-length sequence was then compared to all known ANKRD1 genes using multisequence analysis programs available at ANGIS. This analysis revealed a 957 bp open reading frame with high homology to ANKRD1 in other species; amino acid identities of 91.2%, 93.4%, 91.8% to human, mouse, and rat sequences respectively (see Figure 4). Example 3 - Increased expression of ANKRD1 decreases cisplatin sensitivity o10 To determine whether ANKRD1 has the ability to decrease sensitivity to cisplatin, rat ANKRD1 cDNA in the pcDNA3.1 expression vector, under the control of a CMV promoter, (a gift from the laboratory of Prof. L. Kedes, University of Southern California, USA) was transfected into the ANKRDI-deficient, platinum-sensitive SC2 mutant using Lipofectamine 2000 (Invitrogen Life Technologies, Carlsbad, CA, USA). Transfected clones were selected and maintained in 400 15 pg/ml G-418 (Gibco-BRL). ANKRD1 transfection resulted in increased resistance to cisplatin (Figure 5), providing evidence that ANKRD1 may play a role in modulating sensitivity to cisplatin. Example 4- Decreased expression of ANKRD1 increases cisplatin sensitivity To confirm that ANKRD1 was able to modulate platinum sensitivity, the converse hypothesis was tested, that decreasing ANKRD1 expression in the resistant, CHO-K1 cell line would increase 20 sensitivity. Three 19 bp siRNA oligonucleotides were designed to regions encoding the N- and C terminus as well as to a central portion of the coding region of the CHO ANKRD1 sequence. The sequences of the sense strands of these siRNA oligonucleotides are shown in in SEQ ID NOs: 5 (siRNA41), 6 (siRNA58) and 7 (siRNA77). These siRNA oligonucleotides were designed so as to also allow targeting to the human ANKRD1 sequence Hairpin siRNA encoding DNA oligonucleotide 25 insert sequences were cloned into the pSilencer T M 2.1-U6 hygro vector (Ambion, Austin, TX, USA) and transfected into CHO-K1 using Lipofectamine 2000. Transfected cells were selected and maintained in 500 pg/mL hygromycin B (Invitrogen). Additionally, a silencing negative control pSilencer vector supplied by the manufacturer expressing an siRNA (siN9) with low homology to any known sequences in the human, mouse, and rat genomes, was also stably expressed in CHO 30 K1 cells. As shown in Figure 6A, ANKRD1 expression was virtually ablated in the cells transfected with ANKRD1 siRNA41 (SEQ ID NO:5), targeting the region encoding the N-terminus. ANKRD1 expression was similarly reduced in cells expressing siRNA58 (SEQ ID NO:6) and siRNA77 (SEQ ID NO:7). The negative control (siN9) had minimal effect on ANKRD1 expression.
WO 2006/063397 PCT/AU2005/001890 - 23 Cisplatin sensitivity was determined in the pools of ANKRD1 siRNA transfectants. siRNA mediated reduction in ANKRD1 expression resulted in a significant decrease in resistance to cisplatin (Figure 6B), with the 2-fold reduction in IC5o compared with vector-transfected controls. No significant difference in increased sensitivity to cisplatin was observed between the siRNAs s targeting different regions of the ANKRDI gene. This data confirmed that alteration in ANKRD1 expression was able to alter sensitivity to cisplatin in CHO cells. Example 5- Detection of ANKRD1 in human ovarian tumours The inventors measured ANKRD1 mRNA levels by real-time PCR (see Example 6) in a cohort of 64 serous ovarian adenocarcinomas (approved by the Institutional Human Research 10to Ethics Committee). The majority were of moderate or high grade (low n=1, moderate n=28, high n=34, unknown n=1), and from patients with advanced stage disease (stage I n=2, stage II n=1, stage Ill n=56, stage IV n=5, determined in accordance with FIGO criteria). These patients reflect the most common epithelial ovarian cases treated with platinum-based chemotherapy. The tissue specimens examined came from patients treated between 1988 and 2002 in the Department of 15 Gynaecological Oncology at Westmead Hospital. Tissue examined was excised at the time of primary surgery, prior to the administration of chemotherapy in all but one case. Tissues were snap-frozen and stored in liquid nitrogen in the Department of Gynaecological Oncology tissue bank. Cases were selected according the tumor histological type indicated by original pathology reports, and only cases of serous adenocarcinoma were included. 20 For each case, RNA was extracted from cryosections or pieces of snap frozen tumor specimens and an adjacent haematoxylin and eosin stained section was taken to verify tissue content. Total RNA was isolated from frozen sections using the Absolutely RNA Microprep kit (Stratagene, LaJolla, CA, USA) following homogenisation with a hand operated pestle grinder in the lysis buffer provided; or from pulverized frozen tissue pieces by lysis in guanidinium 25 isothiocyanate solution followed by CsCI centrifugation using standard methods. RNA quality was assessed using an Agilent 2100 Bioanalyser and RNA Integrity Number software (Agilent Technologies, Palo Alto, CA, USA). ANKRD1 mRNA levels were measured by real-time PCR and analysis was performed on coded samples with the operator blinded to all clinical data. ANKRD1 mRNA was relatively low, or absent, in 72% of cases (46/64) and moderate or relatively high 30 expression was seen in 28% of cases (18/64). The level of ANKRD1 expression was significantly associated with overall survival (p = 0.03, Cox Regression Analysis), patients with a high tumor level of ANKRD1 having a significantly poorer outcome compared with patients whose tumors had low ANKRD1 expression. Multivariate analysis indicated that the relationship between ANKRD1 level and overall survival was independent of the extent of residual disease following debulking 35 laparotomy and patient age at diagnosis.
WO 2006/063397 PCT/AU2005/001890 - 24 Example 6 - ANKRD1 expression in human ovarian and breast cell lines ANKRDI expression has not previously been reported in ovarian epithelial cells. In the present study ANKRD1 mRNA levels were measured in 16 human ovarian and breast cell lines by real-time PCR. Total RNA was isolated by lysis in guanidinium isothiocyanate solution followed by 5 CsCI centrifugation using standard methods (see for example Sambrook, J., et al, 1989, and Wilson in Current Protocols in Molecular Biology (Ausubel, F.M. et al. Eds.) Greene Publishing Associated and Wiley-lnterscience, NY 1994) or by Stratagene Absolutely RNA Microprep Kit used according to manufacturer's protocol. RNA was reverse transcribed into cDNA using Superscript Ill First Strand Synthesis System (Invitrogen) according to manufacturer's instructions, in a final o10 volume of 20 pL. To examine ANKRD1 mRNA expression, 100 ng of cDNA used as templates in real-time PCR using the ABI PRISM 7700 Sequence Detection System (PE Applied Biosystems). Samples were made up in a final volume of 25 pL with Platinum Quantitative PCR Supermix-UDG (Invitrogen) and 250 nM of each primer set. The amplification conditions were 50°C for 2 min, 15 95 0 C for 10 min then 50 cycles of 950C for 15 sec and 600C for 60 sec. Relative concentrations of ANKRD1 and housekeeping genes were determined by comparison of Ct values to a standard curve produced using a sample containing cDNA produced from RNA of known concentration. Primers for ANKRD1 were designed using the LUX T M primer design software (Invitrogen), labelled with FAM and were: 5'-GAA CCT TCG GCA CAT CCA CAG GTC-FAM (sense) (SEQ ID NO:13) 20 and 5'-AGA AAC AGC GAG AGG CAG AGC (antisense) (SEQ ID NO:14). Equal cDNA concentrations were confirmed using commercially available primer and probe sets for two housekeeping genes, the VIC-labelled 18S and human P0 PDARs (Applied Biosystems). As shown in Figure 7A ANKRD1 expression was detected at moderate to high levels in 5/9 ovarian cancer cell lines as well as in the normal ovarian cell line HOSE 17.1 and in 1/5 breast 25 cancer cell lines, with little detectable expression in the normal breast cell line BRE-80. Moreover a greater than 1000-fold difference in the level of expression in the ovarian cancer cell lines was observed, with very high expression in COLO 316 and Caov-3, moderate expression in PE01 and low expression in JAM, SK-OV-3 and A2780. Relative cisplatin sensitivity was measured in 13 cell lines using a colorimetric proliferation 30 assay. Cells were seeded into 96-well plates in a volume of 100 Id at 500 to 2000 cells per well. The following day 50 pl. of complete medium containing concentrations of cisplatin ranging from 625 nM to 20 pM were added to duplicate wells and MTS assays (based on color conversion by viable cells MTS, 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulphenyl)-2H terazolium, inner salt) were carried out as described by the manufacturer (Promega) on the day of s35 seeding and subsequently at days 1, 2, 3, 4 and 6 post-drug exposure. IC037 values were calculated WO 2006/063397 PCT/AU2005/001890 - 25 for each cell line from survival curves, using data generated from between two to seven independent experiments. As depicted in Figure 7B, a range of response was observed, with COLO 316, T-47D and MCF-7 being among the most resistant cell lines and HOSE 17.1, A2780 and PEO1, amongst the 5 most sensitive. When the relative levels of ANKRD1 mRNA were compared with the C1037 for cisplatin, the cell lines clearly separated into two groups. In one group of six cell lines (COLO 316, MDA-MB-231, PEO14, HOSE 17.1, NIH:OVCAR3 and Caov3) a linear relationship between ANKRD1 levels and platinum response was observed (R 2 =0.9827; Linear Regression Analysis). The second group of cell lines (HCC1937, A2780, PEO1, T-47D, SK-OV-3, JAM, MCF-7) did not 1o express ANKRD1, and consequently there was no relationship with the ICa37 for cisplatin. No highly platinum-sensitive cell lines were found to express appreciable levels of ANKRD1 (Figure 70) which may indicate that high levels of ANKRD1 are not compatible with a high level of sensitivity to cisplatin, further supporting our hypothesis that ANKRD1 plays an important role in determining response to cisplatin. s15 Example 7- Association between clinical chemo-sensitivity and ANKRD1 expression in human ovarian tumours To determine whether ANKRD1 expression is associated with sensitivity to platinum-based chemotherapy in ovarian cancer, a rare subgroup of ovarian carcinoma patients (n=3) with extreme sensitivity to chemotherapy were selected. These patients were diagnosed with Stage Ill, serous 20 ovarian carcinoma and had macroscopic disease remaining following debulking surgery i.e. these cases were similar to the majority of cases of epithelial ovarian cancer, however the patients were classified as chemo-sensitive as the tumour did not recur following primary treatment and overall survival was greater than 4 years in all cases. This included 2 patients who remained disease free after minimum follow-up of 10 years (Table 2). These cases were compared with a group that were 25 chemo-resistant (n=5) ie patients with evidence of disease progression during primary platinum based therapy (persistent elevation of the serum tumour marker CA125, and/or increasing disease measured by imaging or palpation) with a correspondingly short median survival (approx 1 year) (Table 2). The two subgroups were from the extreme ends of the spectrum of clinical outcome but were not significantly different in respect of features that have been associated with clinical 30 outcome in ovarian cancer, including surgical stage, extent of residual disease following debulking surgery, patient age or tumour grade (Table 2). Patients were identified retrospectively from hospital records and were women diagnosed with primary carcinoma of the ovary or peritoneum. Clinical and pathological features are summarised in Table 2. In all cases the histological tumour type was serous or papillary serous 35 adenocarcinoma.
WO 2006/063397 PCT/AU2005/001890 - 26 Table 2. Clinical and histopathological characteristics of ovarian cancer cohort. (Cases are grouped according to clinical response to platinum-based chemotherapy) Clinical Patient Patient Tumour FIGO Residual Progression- Overall Response Number Age Grade Stage disease free survival survival (weeks) (weeks) 83A 67 3 III < 1 cm 674a 674a Chemo sensitive 351A 52 3 III > 2 cm 243a 243a 146B, Cd 55 4 III > 2 cm 553a 553a 117A 31 1 11I > 2 cm nlab 60 224B c 48 3 III > 2 cm n/ab 53 Chemo resistant 261A 55 3 III > 2 cm n/ab 58 317A 62 2 III > 2 cm n/ab 54 380A 65 2 IV < 1 cm n/ab 53 5 a Censored (ie. the patient had not relapsed (progression-free survival) or died (overall survival) before completion of the study) b Not applicable as disease progression was evident during primary treatment 0 Patient received 1 cycle of carboplatin/cyclophosphamide prior to surgery. d RNA was extracted from tissue collected from the primary tumour site (B) and from the metastasis to o10 the omentum (C). Patients were treated by surgery, and adjuvant chemotherapy according to standard protocols specifying cisplatin or carboplatin, usually in combination with cyclophosphamide or paclitaxel. In the majority of cases, tumour examined was excised at the time of primary surgery, is prior to the administration of chemotherapy. One patient who had tumour excised post-platinum based chemotherapy were also examined. Details of tumour histological type and grade were derived from original pathology reports and the research protocol was approved by the Human Research Ethics Committee, Westmead Hospital, NSW, Australia. Tumour specimens were snap frozen and stored cryogenically in the 20 Westmead Hospital Gynaecological Oncology Tumour Bank. Total RNA was isolated from pulverized frozen sections / pieces by lysis in guanidinium isothiocyanate solution followed by CsCI centrifugation using standard methods. ANKRD1 mRNA levels were measured by real-time PCR and analysis was performed on coded samples with the operator blinded to all clinical data. ANKRD1 mRNA was detected in all tumours examined (8/8, 100%) with a greater than 50 25 fold difference in the level of expression found between the tumours. Expression of ANKRD1 mRNA was low in all cases (3/3, 100%) that were classified as chemo-sensitive (Figure 8) and did not differ between the primary tumour and a metastasis in the one case examined. ANKRD1 expression was higher in 3/5 chemo-resistant cases, compared with the chemo-sensitive cases, and a wide range of ANKRD1 expression was found in this group. This data is highly consistent WO 2006/063397 PCT/AU2005/001890 - 27 with the data from earlier examples that low ANKRD1 expression is indicative of sensitivity to platinum-based chemotherapy. Example 8-ANKRDI expression and sensitivity to cisplatin analogues Carboplatin is a cisplatin analogue with a similar pattern of activity to cisplatin, with reduced 5 neuro- and nephrotoxicity. Oxaliplatin is an analogue with activity against colorectal carcinomas (unlike cisplatin) and appears to have greater activity in vitro against tumour cells that have acquired DNA mismatch repair deficiencies. In the present study the inventors compared the response to carboplatin and oxaliplatin in the 2 CHO cell lines with differing ANKRD1 expression: the CHO-K1 parental line, with high ANKRD1 gene expression, and the SC2 cells, with very low 10 ANKRD1 gene expression. CHO-K1 and SC2 cells were each seeded into 24-well plates, incubated overnight then exposed to a range of concentrations of carboplatin or oxaliplatin for 2 hr. The drugs were removed, wells washed and the growth media replaced. Cells were incubated for 6 days and cell proliferation measured using a modified colorimetric MTS (3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxy 15 methoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium, inner salt) proliferation assay. MTS solution was added to each well and colour development measured on a plate reader at 490 nm. Figure 9 demonstrates that CHO-K1 cells are significantly more resistant to both carboplatin and oxaliplatin compared to the SC2 cells. This differential response to carboplatin and oxaliplatin is very similar to that observed with the parent compound, cisplatin, tested in the same CHO cell 20 lines with differing levels of ANKRD1 expression. These results suggest that ANKRDI expression may be associated with response to platinum-based agents in general. References Agarwal, R and Kaye, SB, Ovarian cancer: Strategies for overcoming resistance to 25 chemotherapy. Nature Rev Cancer 3: 502-16, 2003. Australian Institute of Health and Welfare and Australian Association of Cancer Registries. Cancer in Australia 1999. AIHW, Canberra, 2002. Baudet, S, Another activity for the cardiac biologist: ANKRD1 fishing. Cardiovasc Res 59: 529-31, 2003. 30 Chu, W, et al., Identification and characterization of a novel cytokine-inducible nuclear protein from human endothelial cells. J Biol Chem 270:10236-45,1995. Guminski, AD, Balleine, RL, Chiew, Y-E, Webster, LR, Tapner, M, Farrell, G, Harnett, PR, and deFazio, A, MRP2 (ABCC2) expression and cisplatin sensitivity in hepatocytes and human ovarian carcinoma. Gynecol. Oncol. 2005 [Epub ahead of print].
WO 2006/063397 PCT/AU2005/001890 - 28 Jeyaseelan, R. et al. A novel cardiac-restricted target for doxorubicin. CARP, a nuclear modulator of gene expression in cardiac progenitor cells and cardiomyocytes. J Biol Chem 272(369): 22800-08. 1997 Kojic, S. et al., The Ankrd2 protein, a link between the sarcomere and the nucleus in skeletal 5 muscle. J Mol Biot 339: 313-325, 2004. Kricker, A, Ovarian cancer in Australian women. National Ovarian Cancer Program. National Breast Cancer Centre 2002. Miller, M.K. et al., The muscle ankyrin repeat proteins: CARP, ankrd2/Arpp and DARP as a family of titin filament-based stress response modulators. J Mol Blol 333: 951-964, 2003. 10 Nakada, C, et al., Caridac-restricted ankyrin-repeated protein is differentially induced in duchenne and congenital muscular dystrophy. Lab Invest 83:711-9, 2003. Niedner, H, et al., Identification of genes that mediate sensitivity to cisplatin. Mol Pharmacol 60:1153-60, 2001. Robson, C, et al Isolation and characterization of Chinese hamster ovary cell lines sensitive 15 to mitomycin C and bleomycin. Cancer Res 45: 5304-9, 1985. Sambrook, J., et al. Molecular Cloning - A Laboratory Manual, 2nd Edition. Cold Spring Habour Laboratory Press, New York, USA. 1989. Segelov, E., Mann, G., deFazio, A. and Harnett, P.R. Mechanisms determining sensitivity to cisplatin in three mutant Chinese hamster ovary cell lines. Mut. Res. 407(3): 243-52, 1998. 20 Taniguchi, T, et al., Disruption of the Fanconi anemia-BRCA pathway in cisplatin-sensitive ovarian tumors. Nature Med 9: 568-74, 2003. Zou, Y, et al., ANKRD1, a cardiac ankyrin repeat protein, is downstream in the Nkx2-5 homeobox gene pathway. Development 124: 793-804, 1997.

Claims (37)

1. A method for determining the level of resistance of a tumour cell to one or more chemotherapeutic agents, the method comprising measuring the level of expression of a muscle ankyrin repeat protein in the tumour cell. 5
2. The method of claim 1 wherein the muscle ankyrin repeat protein is selected from the group consisting of ANKRD1, ANKRD2 and ANKRD23.
3. The method of claim 2 wherein the muscle ankyrin repeat protein is ANKRD1.
4. The method of claim 3 wherein ANKRD1 comprises an amino acid sequence as set forth in SEQ ID NO: I or SEQ ID NO: 3. o10
5. The method of claim 3 wherein the ANKRD1 protein is encoded by polynucleotide comprising a nucleic acid sequence as set forth in SEQ ID NO: 2 or SEQ ID NO: 4.
6. The method of any one of the preceding claims wherein the tumour cell is an ovarian, breast, lung, bladder, testicular, cervical, endometrial or bowel tumour cell or a cell from a head or neck tumour. s15
7. The method of any one of the preceding claims wherein the chemotherapeutic agent is a platinum-based agent, an anthracycline or a taxane.
8. The method of claim 7 wherein the platinum-based chemotherapeutic agent is cisplatin or a metabolite, derivative or analogue thereof.
9. The method of claim 8 wherein the cisplatin analogue is carboplatin or oxaliplatin. 20
10. The method of claim 7 wherein the anthracycline is adriamycin or a metabolite, derivative or analogue thereof.
11. The method of claim 7 wherein the taxane is paclitaxel, docataxel, or metabolite, derivative or analogue thereof.
12. A method for determining the level of resistance of a tumour cell to one or more 25 platinum-based chemotherapeutic agents, the method comprising measuring the level of expression of ANKRDI in the tumour cell.
13. A method of assessing the responsiveness of a patient to one or more chemotherapeutic agents, the method comprising: (a) obtaining a biological sample containing at least one tumour cell from the patient, and 30 (b) analyzing the level of expression of a muscle ankyrin repeat protein in the at least one tumour cell, wherein the detected level of expression of the muscle ankyrin repeat protein correlates with the sensitivity level of the at least one tumour cell to the chemotherapeutic agent.
14. The method of claim 13 wherein the muscle ankyrin repeat protein is selected from the 35 group consisting of ANKRD1, ANKRD2 and ANKRD23. WO 2006/063397 PCT/AU2005/001890 - 30
15. The method of claim 13 or 14 wherein the tumour cell is an ovarian, breast, lung, bladder, testicular, cervical, endometrial or bowel tumour cell or a cell from a head or neck tumour.
16. The method of any one of claims 13 to 15 wherein the chemotherapeutic agent is a platinum-based agent, an anthracycline or a taxane. 5
17. A method of assessing the responsiveness of a patient to one or more platinum-based chemotherapeutic agents, the method comprising: (c) obtaining a biological sample containing at least one tumour cell from the patient, and (d) analyzing the level of expression of ANKRD1 in the at least one tumour cell, wherein the detected level of expression of ANKRDI correlates, with the sensitivity level of o10 the at least one tumour cell to the platinum-based chemotherapeutic agent.
18. A method for increasing sensitivity of a tumour cell to one or more chemotherapeutic agents, the method comprising administering to the cell an effective amount of an antagonist of a muscle ankyrin repeat protein.
19. The method of claim 18 wherein the antagonist is a nucleic acid-based inhibitor, a 15 peptide-based inhibitor or a small molecule inhibitor of the muscle ankyrin repeat protein or polynucleotide encoding the same.
20. The method of claim 19 wherein the nucleic acid-based inhibitor is an siRNA molecule or an antisense construct.
21. The method of claim 20 wherein the nucleic acid-based inhibitor is an siRNA molecule. 20
22. The method of claim 21 wherein the muscle ankyrin repeat protein is ANKRD1.
23. The method of claim 22 wherein the siRNA molecule comprises a nucleotide sequence as set forth in any one of SEQ ID NOs:5, 6 or 7, or a fragment thereof.
24. A method for increasing sensitivity of a tumour cell to one or more platinum-based chemotherapeutic agents, the method comprising administering to the cell an effective amount of 25 an antagonist of ANKRDI.
25. A method for substantially inhibiting expression of a muscle ankyrin repeat protein in tumour cells, the method comprising introducing into the tumour cells an effective amount of an antagonist of the muscle ankyrin repeat protein.
26. A method for substantially inhibiting expression of ANKRD1 in tumour cells, the 30 method comprising introducing into the tumour cells an effective amount of an antagonist of ANKRDI.
27. A composition for increasing sensitivity of a tumour cell to one or more chemotherapeutic agents, the composition comprising an antagonist of a muscle ankyrin repeat protein together with one or more pharmaceutically acceptable carriers, diluents or adjuvants. WO 2006/063397 PCT/AU2005/001890 -31
28. A composition for increasing sensitivity of a tumour cell to one or more platinum-based chemotherapeutic agents, the composition comprising an antagonist of ANKRD1 together with one or more pharmaceutically acceptable carriers, diluents or adjuvants.
29. An isolated inhibitory nucleic acid construct comprising a nucleotide sequence specific 5 to at least a portion of a polynucleotide encoding a muscle ankyrin repeat protein, wherein the nucleic acid construct substantially inhibits expression of the muscle ankyrin repeat protein in tumour cells.
30. The inhibitory nucleic acid construct of claim 29 wherein the portion of the polynucleotide comprises the coding region of the gene encoding the muscle ankyrin repeat protein .o and/or one or more regulatory regions of the gene.
31. The inhibitory nucleic acid construct of claim 29 or 30 wherein the muscle ankyrin repeat protein is ANKRD1.
32. The inhibitory nucleic acid construct of claim 31 wherein the construct is an siRNA molecule and the nucleotide sequence of the construct comprises: 15 (a) the nucleotide sequence set forth in any one of SEQ ID NOs:5, 6 or 7, or a fragment thereof; or (b) a nucleotide sequence having at least 85% identity to the nucleotide sequence set forth in any one of SEQ ID NOs: 5, 6 or 7, or fragment thereof.
33. The inhibitory nucleic acid construct of claim 32 wherein the nucleotide sequence 20 displays at least 85%, at least 90% identity, or at least 95% identity, to the nucleotide sequence set forth in any one of SEQ ID NOs: 5, 6 or 7, or fragment thereof.
34. A method for substantially inhibiting expression of a muscle ankyrin repeat protein in tumour cells, the method comprising introducing into the tumour cells an effective amount of an inhibitory nucleic acid construct according to any one of claims 29 to 33. 25
35. A method for increasing sensitivity of tumour cells to one or more chemotherapeutic agents, the method comprising introducing into the tumour cells an effective amount of an inhibitory nucleic acid construct according to any one of claims 29 to 33.
36. -A composition for use in the method of claim 34 or 35, the composition comprising an inhibitory nucleic acid construct according to any one of claims 29 to 33. 30
37. The composition of claim 36 further comprising one or more chemotherapeutic agents.
AU2005316200A 2004-12-14 2005-12-14 Determinants of sensitivity to chemotherapeutic agents Ceased AU2005316200B2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU2005316200A AU2005316200B2 (en) 2004-12-14 2005-12-14 Determinants of sensitivity to chemotherapeutic agents

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
AU2004907126A AU2004907126A0 (en) 2004-12-14 Determinants of sensitivity to chemotherapeutic agents
AU2004907126 2004-12-14
AU2005316200A AU2005316200B2 (en) 2004-12-14 2005-12-14 Determinants of sensitivity to chemotherapeutic agents
PCT/AU2005/001890 WO2006063397A1 (en) 2004-12-14 2005-12-14 Determinants of sensitivity to chemotherapeutic agents

Publications (2)

Publication Number Publication Date
AU2005316200A1 true AU2005316200A1 (en) 2006-06-22
AU2005316200B2 AU2005316200B2 (en) 2010-08-26

Family

ID=38353456

Family Applications (1)

Application Number Title Priority Date Filing Date
AU2005316200A Ceased AU2005316200B2 (en) 2004-12-14 2005-12-14 Determinants of sensitivity to chemotherapeutic agents

Country Status (1)

Country Link
AU (1) AU2005316200B2 (en)

Also Published As

Publication number Publication date
AU2005316200B2 (en) 2010-08-26

Similar Documents

Publication Publication Date Title
DK2081586T4 (en) Rspondins as modulators of angiogenesis and vasculogenesis
US8722340B2 (en) JAB1 as a prognostic marker and a therapeutic target for human cancer
Khalyfa et al. Characterization of elongation factor-1A (eEF1A-1) and eEF1A-2/S1 protein expression in normal and wasted mice
US8486903B2 (en) TAZ/WWTR1 for diagnosis and treatment of cancer
CN108192972B (en) Methods for diagnosis, prognosis and treatment of breast cancer metastasis
WO2014017491A1 (en) Fusion gene of cep55 gene and ret gene
KR20150122731A (en) Method for the prognosis and treatment of cancer metastasis
KR20140057361A (en) Methods and compositions for the treatment and diagnosis of cancer
US7335475B2 (en) PR/SET-domain containing nucleic acids, polypeptides, antibodies and methods of use
US5654406A (en) Antibody to ERBB2 promoter binding factor
Fernandez et al. Mice lacking dystrophin or α sarcoglycan spontaneously develop embryonal rhabdomyosarcoma with cancer-associated p53 mutations and alternatively spliced or mutant Mdm2 transcripts
US8673869B2 (en) Determinants of sensitivity to chemotherapeutic agents
AU2005316200B2 (en) Determinants of sensitivity to chemotherapeutic agents
TW201204393A (en) Diagnostic agent and therapeutic agent of cancer
BRPI0707272A2 (en) use of a nucleic acid fragment of sequence id: 1 or 2, or a variant of sequence id: 1 or 2, nucleic acid fragment of sense and / or antisense of sequence id: 1 or 2, or a variant of sequence id: 1 or 2, composition, protein, method for diagnosing cancers of neuroectodermal origin, and diagnostic kit
US20190374506A1 (en) Compositions and methods for treating cancer
WO2004022750A1 (en) Novel diagnostic and therapeutic methods and reagents therefor
WO2008075713A1 (en) Marker for diagnosis of cancer, and target molecule for therapy
JP4617257B2 (en) Human cancer-related genes, products encoded by them and applications
CA2538442A1 (en) Eukaryotic genes for modulating cell cycle progression
AU2010355523B2 (en) MLK4 gene, a new diagnostic and prognostic marker in cancers
US8241846B1 (en) Hedgehog pathway modulation and uses thereof for treating, preventing and/or diagnosing cancer
KR101796091B1 (en) A biomarker composition for diagnosis of head and neck cancer comprising carboxyl-terminal modulator protein
WO2019037675A1 (en) Gene capable of promoting tumor occurrence and development and expression product and uses thereof
Huu Characterisation of the Expression and Function of an E3 Ubiquitin Ligase, WWP1 in Breast Cancer

Legal Events

Date Code Title Description
FGA Letters patent sealed or granted (standard patent)
MK14 Patent ceased section 143(a) (annual fees not paid) or expired