AU2012200431B2 - Gene expression markers for predicting response to chemotherapy - Google Patents

Gene expression markers for predicting response to chemotherapy Download PDF

Info

Publication number
AU2012200431B2
AU2012200431B2 AU2012200431A AU2012200431A AU2012200431B2 AU 2012200431 B2 AU2012200431 B2 AU 2012200431B2 AU 2012200431 A AU2012200431 A AU 2012200431A AU 2012200431 A AU2012200431 A AU 2012200431A AU 2012200431 B2 AU2012200431 B2 AU 2012200431B2
Authority
AU
Australia
Prior art keywords
chemotherapy
expression
cancer
rna
gene
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.)
Active
Application number
AU2012200431A
Other versions
AU2012200431A1 (en
Inventor
Joffre B. Baker
Luca Gianni
Steven Shak
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.)
Fondazione IRCCS Istituto Nazionale dei Tumori
Genomic Health Inc
Original Assignee
Fondazione IRCCS Istituto Nazionale dei Tumori
Genomic Health Inc
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 AU2011204944A external-priority patent/AU2011204944B2/en
Application filed by Fondazione IRCCS Istituto Nazionale dei Tumori, Genomic Health Inc filed Critical Fondazione IRCCS Istituto Nazionale dei Tumori
Priority to AU2012200431A priority Critical patent/AU2012200431B2/en
Publication of AU2012200431A1 publication Critical patent/AU2012200431A1/en
Priority to AU2013206082A priority patent/AU2013206082B2/en
Application granted granted Critical
Publication of AU2012200431B2 publication Critical patent/AU2012200431B2/en
Priority to AU2016210735A priority patent/AU2016210735B2/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Landscapes

  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)

Abstract

The present invention provides sets of genes the expression of which is important in the prognosis of cancer. In particular, the invention provides gene expression information 5 useful for predicting whether cancer patients are likely to have a beneficial treatment response to chemotherapy. 3098652_1 (GHMatters) P82417.AU.3

Description

AUSTRALIA Patents Act 1990 COMPLETE SPECIFICATION Standard Patent Applicants: FONDAZIONE IRCCS ISTITUTO NAZIONALE DEI TUMORI and GENOMIC HEALTH, INC. Invention Title: GENE EXPRESSION MARKERS FOR PREDICTING RESPONSE TO CHEMOTHERAPY The following statement is a full description of this invention, including the best method for performing it known to us: Gene Expression Markers for Predicting Response to Chemotherapy The entire disclosure in the complete specification of our Australian Patent Application No. 2011204944 is by this cross-reference incorporated into the present 5 specification. Background of the Invention Field of the Invention The present invention provides sets of genes the expression of which is important in 10 the prognosis of cancer. In particular, the invention provides gene expression information useful for predicting whether cancer patients are likely to have a beneficial treatment response to chemotherapy. Description of the Related Art 15 Oncologists have a number of treatment options available to them, including different combinations of chemotherapeutic drugs that are characterized as "standard of care," and a number of drugs that do not carry a label claim for particular cancer, but for which there is evidence of efficacy in that cancer. Best likelihood of good treatment outcome requires that patients be assigned to optimal available cancer treatment, and that this assignment be made 20 as quickly as possible following diagnosis. In particular, it is important to determine the likelihood of patient response to "standard of care" chemotherapy because chemotherapeutic drugs such as anthracyclines and taxanes have limited efficacy and are toxic. The identification of patients who are most or least likely to respond thus could increase the net benefit these drugs have to offer, and decrease the net morbidity and toxicity, via more 25 intelligent patient selection. Currently, diagnostic tests used in clinical practice are single analyte, and therefore do not capture the potential value of knowing relationships between dozens of different markers. Moreover, diagnostic tests are frequently not quantitative, relying on immunohistochemistry. This method often yields different results in different laboratories, in part because the 30 reagents are not standardized, and in part because the interpretations are subjective and cannot be easily quantified. RNA-based tests have not often been used because of the problem of RNA degradation over time and the fact that it is difficult to obtain fresh tissue samples from patients for analysis. Fixed paraffin-embedded tissue is more readily available 2 3098552_1 (GHMatters) P52417.AU.3 and methods have been established to detect RNA in fixed tissue. However, these methods typically do not allow for the study of large numbers of genes (DNA or RNA) from small amounts of material. Thus, traditionally fixed tissue has been rarely used other than for immunohistochhemistry detection of proteins. 5 In the last few years, several groups have published studies concerning the classification of various cancer types by microarray gene expression analysis (see, e.g. Golub et al., Science 286:531-537 (1999); Bhattacharjae et al., Proc. Nail. Acad Sci. USA 98:13790-13795 (2001); Chen-Hsiang et al., Bioinformatics 17 (Suppl. 1):S316-S322 (2001); Ramaswamy et al., Proc. Nati. Acad Sci. USA 98:15149-15154 (2001)). Certain 10 classifications of human breast cancers based on gene expression patterns have also been reported (Martin et al., Cancer Res. 60:2232-2238 (2000); West et al., Proc. Nati. Acad. Sci. USA 98:11462-11467 (2001); Sorlie et al., Proc. Natl. Acad. Sci. USA 98:10869-10874 (2001); Yan el al., Cancer Res. 61:8375-8380 (2001)). However, these studies mostly focus on improving and refining the already established classification of various types of cancer, 15 including breast cancer, and generally do not provide new insights into the relationships of the differentially expressed genes, and do not link the findings to treatment strategies in order to improve the clinical outcome of cancer therapy. Although modern molecular biology and biochemistry have revealed hundreds of genes whose activities influence the behavior of tumor cells, state of their differentiation, and 20 their sensitivity or resistance to certain therapeutic drugs, with a few exceptions, the status of these genes has not been exploited for the purpose of routinely making clinical decisions about drug treatments. One notable exception is the use of estrogen receptor (ER) protein expression in breast carcinomas to select patients to treatment with anti-estrogen drugs, such as tamoxifen. Another exceptional example is the use of ErbB2 (Her2) protein expression in 25 breast carcinomas to select patients with the Her2 antagonist drug Herceptin* (Genentech, Inc., South San Francisco, CA). Despite recent advances, the challenge of cancer treatment remains to target specific treatment regimens to pathogenically distinct tumor types, and ultimately personalize tumor treatment in order to maximize outcome. Hence, a need exists for tests that simultaneously 30 provide predictive information about patient responses to the variety of treatment options. This is particularly true for breast cancer, the biology of which is poorly understood. It is clear that the classification of breast cancer into a few subgroups, such as the ErbB2 positive subgroup, and subgroups characterized by low to absent gene expression of the estrogen 3 3098652_1 (GHMatters) P62417 AU 3 receptor (ER) and a few additional transcriptional factors (Perou et al., Nature 406:747-752 (2000)), does not reflect the cellular and molecular heterogeneity of breast cancer, and does not allow the design of treatment strategies maximizing patient response. Breast cancer is the most common type of cancer among women in the United States and is 5 the leading cause of cancer deaths among women ages 40 - 59. Therefore, there is a particularly great need for a clinically validated breast cancer test predictive of patient response to chemotherapy. It is to be understood that, if any prior art publication is referred to herein, such reference does not constitute an admission that the publication forms a part of the common 10 general knowledge in the art, in Australia or any other country. Summary of the Invention The present invention provides gene sets useful in predicting the response of cancer, e.g. breast cancer patients to chemotherapy. In addition, the invention provides a clinically 15 validated cancer, e.g. breast cancer, test, predictive of patient response to chemotherapy, using multi-gene RNA analysis. The present invention accommodates the use of archived paraffin-embedded biopsy material for assay of all markers in the relevant gene sets, and therefore is compatible with the most widely available type of biopsy material. In one aspect, the present invention concerns a method for predicting the response of 20 a subject diagnosed with cancer to chemotherapy comprising determining the expression level of one or more prognostic RNA transcripts or their expression products in a biological sample comprising cancer cells obtained from said subject, wherein the predictive RNA transcript is the transcript of one or more genes selected from the group consisting of TBP; ILT.2; ABCC5; CDI8; GATA3; DICERl; MSH3; GBPl; IRSl; CD3z; fasl; TUBB; BAD; 25 ERCCl; MCM6; PR; APC; GGPSI; KRTI8; ESRRG; E2F1; AKT2; A.Catenin; CEGPI; NPDO09; MAPKl4; RUNX1; ID2; G.Catenin; FBXO5; FHIT; MTA1; ERBB4; FUS; BBC3; IGFlR; CD9; TP53BPl; MUCl; IGFBP5; rhoC; RALBPl; CDC20; STAT3; ERKI; HLA.DPB1; SGCB; CGA; DHPS; MGMT; CRIP2; MMP12; ErbB3; RAPlGDS1; CDC25B; IL6; CCNDI; CYBA; PRKCD; DR4; Hepsin; CRABPI; AK055699; Contig.51037; 30 VCAMl; FYN; GRB7; AKAP.2; RASSFl; MCPl; ZNF38; MCM2; GBP2; SEMA3F; CD31; COLIAl; ER2; BAGl; AKT1; COLlA2; STATIC; Wnt.5a; PTPDl; RAB6C; TKI, ErbB2, CCNB1, BIRC5, STK6, MK167, MYBL2, MMP1 1, CTSL2, CD68, GSTM1, BCL2, ESRI wherein 4 3098852.1 (GHMatters) P62417.AU.3 (a) for every unit of increased expression of one or more of ILT.2; CD18; GBP1; CD3z; fasl; MCM6; E2Fl; ID2; FBXO5; CDC20; HLA.DPBI; CGA; MMP12; CDC25B; IL6; CYBA; DR4; CRABPI; Contig.51037; VCAMI; FYN; GRB7; AKAP.2; RASSFl; MCP1; MCM2; GBP2; CD31; ER2; STAT1; TK1; ERBB2, CCNB1, BIRC5, STK6, MK167, 5 MYBL2, MMPl 1, CTSL2 and CD68; or the corresponding expression product, said subject is predicted to have an increased likelihood of response to chemotherapy; and (b) for every unit of increased expression of one or more of TBP; ABCC5; GATA3; DICER1; MSH3; IRS 1; TUBB; BAD; ERCC1; PR; APC; GGPS1; KRT18; ESRRG; AKT2; A.Catenin; CEGPI; NPDO09; MAPKl4; RUNXl; G.Catenin; FHIT; 10 MTAI; ErbB4; FUS; BBC3; IGFIR; CD9; TP53BPl; MUCI; IGFBP5; rhoC; RALBPl; STAT3; ERK1; SGCB; DHPS; MGMT; CRIP2; ErbB3; RAPlGDS1; CCND1; PRKCD; Hepsin; AK055699; ZNF38; SEMA3F; COLlAl; BAGI; AKTI; COLlA2; Wnt.5a; PTPDI; RAB6C; GSTM1, BCL2, ESRI; or the corresponding expression product, said subject is predicted to have a decreased likelihood of response to chemotherapy. 15 In a particular embodiment, in the above method the predictive RNA transcript is the transcript of one or more genes selected from the group consisting of TBP; ILT.2; ABCC5; CD18; GATA3; DICER1; MSH3; GBP1; IRSI; CD3z; fasl; TUBB; BAD; ERCCl; MCM6; PR; APC; GGPSl; KRT18; ESRRG; E2Fl; AKT2; A.Catenin; CEGP1; NPDO09; MAPK14; RUNXI; ID2; G.Catenin; FBX05; FHIT; MTAl; ERBB4; FUS; BBC3; IGFlR; CD9; 20 TP53BPI; MUCI; IGFBP5; rhoC; RALBPI; CDC20; STAT3; ERKI; HLA.DPBl; SGCB; CGA; DHPS; MGMT; CRIP2; MMP12; ErbB3; RAPlGDSl; CDC25B; IL6; CCND1; CYBA; PRKCD; DR4; Hepsin; CRABPl; AK055699; Contig.51037; VCAMI; FYN; GRB7; AKAP.2; RASSFl; MCPl; ZNF38; MCM2; GBP2; SEMA3F; CD31; COLlAl; ER2; BAGI; AKTI; COL1A2; STATl; Wnt.5a; PTPDI; RAB6C; and TKl. 25 In another embodiment, the response is a complete pathological response. In a preferred embodiment, the subject is a human patient. The cancer can be any types of cancer but preferably is a solid tumor, such as breast cancer, ovarian cancer, gastric cancer, colon cancer, pancreatic cancer, prostate cancer and lung cancer. 30 If the tumor is breast cancer, it can, for example, be invasive breast cancer, or stage II or stage III breast cancer. In a particular embodiment, the chemotherapy is adjuvant chemotherapy. In another embodiment, the chemotherapy is neoadjuvant chemotherapy. 5 309852_1 (GHMatters) P62417.AU.3 The neoadjuvant chemotherapy may, for example, comprise the administration of a taxane derivativ, such as docetaxel and/or paclitaxel, and/or other anti-cancer agents, such as, members of the anthracycline class of anti-cancer agents, doxorubicin, topoisomerase inhibitors, etc. 5 The method may involve determination of the expression levels of at least two, or at least five, or at least ten, or at least 15 of the prognostic transcripts listed above, or their expression products. The biological sample may be e.g. a tissue sample comprising cancer cells, where the tissue can be fixed, paraffin-embedded, or fresh, or frozen. 10 In a particular embodiment, the tissue is from fine needle, core, or other types of biopsy. In another embodiment, the tissue sample is obtained by fine needle aspiration, bronchial lavage, or transbronchial biopsy. The expression level of said prognostic RNA transcript or transcripts can be 15 determined, for example, by RT-PCR or an other PCR-based method, immunohistochemistry, proteomics techniques, or any other methods known in the art, or their combination. In an embodiment, the assay for the measurement of said prognostic RNA transcripts or their expression products is provided is provided in the form of a kit or kits. 20 In another aspect, the invention concerns an array comprising polynucleotides hybridizing to a plurality of the following genes: TBP; ILT.2; ABCC5; CD18; GATA3; DICERI; MSH3; GBPl; IRSI; CD3z; fasl; TUBB; BAD; ERCCl; MCM6; PR; APC; GGPS1; KRT18; ESRRG; E2F1; AKT2; A.Catenin; CEGP1; NPDO09; MAPK14; RUNX1; ID2; G.Catenin; FBXO5; FHIT; MTAl; ERBB4; FUS; BBC3; IGF1R; CD9; TP53BPl; 25 MUCl; IGFBP5; rhoC; RALBPl; CDC20; STAT3; ERKI; HLA.DPBl; SGCB; CGA; DHPS; MGMT; CRIP2; MMP12; ErbB3; RAP1GDS1; CDC25B; IL6; CCNDl; CYBA; PRKCD; DR4; Hepsin; CRABPl; AK055699; Contig.51037; VCAM1; FYN; GRB7; AKAP.2; RASSFl; MCP1; ZNF38; MCM2; GBP2; SEMA3F; CD31; COLIAI; ER2; BAGl; AKTl; COLIA2; STATIC; Wnt.5a; PTPD1; RAB6C; TK1, ErbB2, CCNB1, BIRC5, 30 STK6, MK167, MYBL2, MMPI 1, CTSL2, CD68, GSTM1, BCL2, ESR1. In an embodiment, the array compises polynucleotides hybridizing to a plurality of the following genes: TBP; ILT.2; ABCC5; CD18; GATA3; DICERI; MSH3; GBPl; IRS1; CD3z; fasl; TUBB; BAD; ERCCI; MCM6; PR; APC; GGPSI; KRTI8; ESRRG; E2F1; 6 3098652_1 (GHMatters) PS2417.AU.3 AKT2; A.Catenin; CEGPl; NPDO09; MAPK14; RUNXI; ID2; G.Catenin; FBX05; FHIT; MTAl; ERBB4; FUS; BBC3; IGFlR; CD9; TP53BPl; MUCI; IGFBP5; rhoC; RALBPl; CDC20; STAT3; ERKI; HLA.DPBl; SGCB; CGA; DHPS; MGMT; CRIP2; MMPl2; ErbB3; RAPlGDSl; CDC25B; IL6; CCNDl; CYBA; PRKCD; DR4; Hepsin; CRABPI; 5 AK055699; Contig.51037; VCAMI; FYN; GRB7; AKAP.2; RASSFI; MCP1; ZNF38; MCM2; GBP2; SEMA3F; CD31; COLlAI; ER2; BAGI; AKTl; COLlA2; STATIC; Wnt.5a; PTPDI; RAB6C; TKl. In another embodiment, the array comprises polynucleotides hybridizing to a plurality of the following genes: ILT.2; CD18; GBPl; CD3z; fasl; MCM6; E2F1; ID2; 10 FBX05; CDC20; HLA.DPBl; CGA; MMP12; CDC25B; IL6; CYBA; DR4; CRABPI; Contig.51037; VCAMl; FYN; GRB7; AKAP.2; RASSFl; MCPl; MCM2; GBP2; CD31; ER2; STAT1; TK1; ERBB2, CCNB1, BIRC5, STK6, MK167, MYBL2, MMP 11, CTSL2 and CD68. In yet another embodiment, the array comprises polynucleotides hybridizing to a 15 plurality of the following genes: ILT.2; CD18; GBPl; CD3z; fasl; MCM6; E2FI; ID2; FBX05; CDC20; HLA.DPBl; CGA; MMP12; CDC25B; IL6; CYBA; DR4; CRABPl; Contig.51037; VCAMI; FYN; GRB7; AKAP.2; RASSFl; MCPl; MCM2; GBP2; CD31; ER2; STATIC; TKI In a still further embodiment, the array comprises polynucleotides hybridizing to a 20 plurality of the following genes: TBP; ABCC5; GATA3; DICERI; MSH3; IRS]; TUBB; BAD; ERCCI; PR; APC; GGPSl; KRT18; ESRRG; AKT2; A.Catenin; CEGPl; NPDO09; MAPKI4; RUNXl; G.Catenin; FHIT; MTAl; ErbB4; FUS; BBC3; IGFlR; CD9; TP53BPl; MUCI; IGFBP5; rhoC; RALBPl; STAT3; ERKI; SGCB; DHPS; MGMT; CRIP2; ErbB3; RAPIGDSI; CCNDl; PRKCD; Hepsin; AK055699; ZNF38; SEMA3F; COLlAl; BAGI; 25 AKT1; COLl A2; Wnt.5a; PTPD1; RAB6C; GSTM1, BCL2, ESR1. In another embodiment, the array comprises polynucleotides hybridizing to a plurality of the following genes: TBP; ABCC5; GATA3; DICERI; MSH3; IRSl; TUBB; BAD; ERCCl; PR; APC; GGPSI; KRT18; ESRRG; AKT2; A.Catenin; CEGPl; NPDO09; MAPK14; RUNXI; G.Catenin; FHIT; MTAl; ErbB4; FUS; BBC3; IGFIR; CD9; TP53BPl; 30 MUCI; IGFBP5; rhoC; RALBPl; STAT3; ERKI; SGCB; DHPS; MGMT; CRIP2; ErbB3; RAPIGDS1; CCNDI; PRKCD; Hepsin; AK055699; ZNF38; SEMA3F; COLlAl; BAGI; AKTl; COLlA2; Wnt.5a; PTPDl; RAB6C. 7 3098652_1 (GHMatters) P62417.AU.3 In various embodiments, the array comprises at least five, or at least 10, or at least 15, or at least 10 of such polynucleotides. In a particular embodiment, the array comprises polynucleotides hybridizing to all of the genes listed above. 5 In another particular embodiment, the array comprises more than one polynucleotide hybridizing to the same gene. In another embodiment, at least one of the the polynucleotides comprises an intron based sequence the expression of which correlates with the expression of a corresponding exon sequence. 10 In various embodiments, the polynucleotides can be cDNAs or oligonucleotides. In another aspect, the invention concerns a method of preparing a personalized genomics profile for a patient comprising the steps of: (a) determining the normalized expression levels of the RNA transcripts or the expression products of a gene or gene set selected from the group consisting of TBP; ILT.2; 15 ABCC5; CD18; GATA3; DICER1; MSH3; GBP1; IRS1; CD3z; fasl; TUBB; BAD; ERCC1; MCM6; PR; APC; GGPS1; KRT18; ESRRG; E2F1; AKT2; A.Catenin; CEGP1; NPDO09; MAPKI4; RUNXI; ID2; G.Catenin; FBX05; FHIT; MTAI; ERBB4; FUS; BBC3; IGFIR; CD9; TP53BPl; MUCI; IGFBP5; rhoC; RALBPl; CDC20; STAT3; ERKI; HLA.DPBI; SGCB; CGA; DHPS; MGMT; CRIP2; MMP12; ErbB3; RAP1GDSl; CDC25B; IL6; 20 CCNDI; CYBA; PRKCD; DR4; Hepsin; CRABP1; AK055699; Contig.51037; VCAMl; FYN; GRB7; AKAP.2; RASSF1; MCP1; ZNF38; MCM2; GBP2; SEMA3F; CD31; COLIAl; ER2; BAGI; AKTI; COL1A2; STATIC; Wnt.5a; PTPDI; RAB6C; TKI, ErbB2, CCNB1, BIRC5, STK6, MK167, MYBL2, MMP1 1, CTSL2, CD68, GSTMI, BCL2, ESRI, in a cancer cell obtained from said patient; and 25 (b) creating a report summarizing the data obtained by the gene expression analysis. In a specific embodiment, if increased expression of one or more of ILT.2; CD18; GBP1; CD3z; fasl; MCM6; E2F1; ID2; FBX05; CDC20; HLA.DPBl; CGA; MMP12; CDC25B; IL6; CYBA; DR4; CRABPI; Contig.51037; VCAMI; FYN; GRB7; AKAP.2; 30 RASSFl; MCP1; MCM2; GBP2; CD31; ER2; STATIC; TKI; ERBB2, CCNBI, BIRC5, STK6, MK167, MYBL2, MMP1 1, CTSL2 and CD68; or the corresponding expression product, is determined, the report includes a prediction that said subject has an increased 8 309852.1 (GHMatters) P62417 AU.3 likelihood of response to chemotherapy. In this case, in a particular embodiment, the method includes the additional step of treating the patient with a chemotherapeutic agent. In the foregoing method, if increased expression of one or more of TBP; ABCC5; GATA3; DICERI; MSH3; IRSI; TUBB; BAD; ERCCI; PR; APC; GGPSI; KRT18; 5 ESRRG; AKT2; A.Catenin; CEGPI; NPDO09; MAPK14; RUNXl; G.Catenin; FHIT; MTAl; ErbB4; FUS; BBC3; IGFIR; CD9; TP53BPl; MUCI; IGFBP5; rhoC; RALBPl; STAT3; ERKI; SGCB; DHPS; MGMT; CRIP2; ErbB3; RAP1GDSI; CCNDI; PRKCD; Hepsin; AK055699; ZNF38; SEMA3F; COLIAl; BAGI; AKTI; COLIA2; Wnt.5a; PTPDI; RAB6C; GSTMI, BCL2, ESRI; or the corresponding expression product, is 10 determined, the report includes a prediction that said subject has a decreased likelihood of response to chemotherapy. In another aspect, the invention concerns a method for determining the likelihood of the response of a patient to chemotherapy, comprising: (a) determining the expression levels of the RNA transcripts of following genes 15 .ACTB, BAG 1, BCL2, CCNB1, CD68, SCUBE2, CTSL2, ESRI, GAPD, GRB7, GSTM1, GUSB, ERBB2, MK167, MYBL2, PGR, RPLPO, STK6, MMPl 1, BIRC5, TFRC , or their expression products, and (b) calculating the recurrence score (RS). In an embodiment, patients having an RS > 50 are in the upper 50 percentile of 20 patients who are likely to respond to chemotherapy. In enother embodiment, patients having an RS < 35 are in the lower 50 percentile of patients who are likely to response to chemotherapy. In a further embodiment, RS is determined by creating the following gene subsets: (i) growth factor subset: GRB7 and HER2; 25 (ii) estrogen receptor subset: ER, PR, Bcl2, and CEGPl; (iii) proliferation subset: SURV, Ki,67, MYBL2, CCNB1, and STKI5; and (iv) invasion subset: CTSL2, and STMY3; wherein a gene within any of subsets (i)-(iv) can be substituted by substitute gene which coexpresses with said gene in said tumor with a Pearson correlation coefficient of> 30 0.40; and (c) calculating the recurrence score (RS) for said subject by weighting the contributions of each of subsets (i) - (iv), to breast cancer recurrence. 9 3098852_1 (GHMatters) P62417.AU 3 The foregoing method may further comprise determining the RNA transcripts of CD68, GSTMI and BAGI or their expression products, or corresponding substitute genes or their expression products, and including the contribution of said genes or substitute genes to breast cancer recurrence in calculating the RS 5 RS may, for example, be determined by using the following equation: RS = (0.23 to 0.70) x GRB7axisthresh - (0.17 to 0.55) x ERaxis + (0.52 to 1.56) x prolifaxisthresh + (0.07 to 0.21) x invasionaxis + (0.03 to 0.15) x CD68 - (0.04 to 0.25) x GSTM1 - (0.05 to 0.22) x BAGI wherein 10 (i) GRB7 axis = (0.45 to 1.35) x GRB7 + (0.05 to 0.15) x HER2; (ii) if GRB7 axis < -2, then GRB7 axis thresh = -2, and if GRB7 axis -2, then GRB7 axis thresh = GRB7 axis; (iii) ER axis = (EstI + PR + Bcl2 + CEGP1)/4; (iv) prolifaxis = (SURV + Ki.67 + MYBL2 + CCNBI + STKI5)/5; 15 (v) if prolifaxis < -3.5, then prolifaxisthresh = -3.5, if prolifaxis> -3.5, then prolifaxishresh = prolifaxis; and (vi) invasionaxis = (CTSL2 + STMY3)/2, wherein the individual contributions of the genes in (iii), (iv) and (vi) are weighted by a factor of 0.5 to 1.5, and wherein a higher RS represents an increased likelihood of breast 20 cancer recurrence. In another embodiment, RS is determined by using the following equation: RS (range, 0 - 100) = + 0.47 x HER2 Group Score - 0.34 x ER Group Score + 1.04 x Proliferation Group Score + 0.10 x Invasion Group Score + 0.05 x CD68 - 0.08 x GSTM1 - 0.07 x BAG1 Brief Description of the Drawings Figure 1 shows the relationship between recurrence score (RS) and likelihood of 25 patient response to chemotherapy, based on results from a clinical trial with pathologic complete response endpoint. 10 309852_1 (GHMaters) P52417.AU.3 Table 1 shows a list of genes, the expression of which correlates, positively or negatively, with breast cancer response to adriamycin and taxane neoadjuvant chemotherapy. Results from a clinical trial with pathologic complete response endpoint. Statistical analysis utilized univarite generalized linear models with a probit link function. 5 Table 2 presents a list of genes, the expression of which predicts breast cancer response to chemotherapy. Results from a retrospective clinical trial. The table includes accession numbers for the genes, sequences for the forward and reverse primers (designated by "f' and "r", respectively) and probes (designated by "p") used for PCR amplification. Table 3 shows the amplicon sequences used in PCR amplification of the indicated 10 genes. Detailed Description A. Definitions Unless defined otherwise, technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention 15 belongs. Singleton et al., Dictionary of Microbiology and Molecular Biology 2nd ed., J. Wiley & Sons (New York, NY 1994), and March, Advanced Organic Chemistry Reactions, Mechanisms and Structure 4th ed., John Wiley & Sons (New York, NY 1992), provide one skilled in the art with a general guide to many of the terms used in the present application. One skilled in the art will recognize many methods and materials similar or 20 equivalent to those described herein, which could be used in the practice of the present invention. Indeed, the present invention is in no way limited to the methods and materials described. For purposes of the present invention, the following terms are defined below. In the claims which follow and in the description of the invention, except where the context requires otherwise due to express language or necessary implication, the word 25 "comprise" or variations such as "comprises" or "comprising" is used in an inclusive sense, i.e. to specify the presence of the stated features but not to preclude the presence or addition of further features in various embodiments of the invention. The term "microarray" refers to an ordered arrangement of hybridizable array elements, preferably polynucleotide probes, on a substrate. 30 The term "polynucleotide," when used in singular or plural, generally refers to any polyribonucleotide or polydeoxribonucleotide, which may be unmodified RNA or DNA or modified RNA or DNA. Thus, for instance, polynucleotides as defined herein include, without limitation, single- and double-stranded DNA, DNA including single- and double 11 30986521 (GHMatters) P62417 AU.3 stranded regions, single- and double-stranded RNA, and RNA including single- and double stranded regions, hybrid molecules comprising DNA and RNA that may be single-stranded or, more typically, double-stranded or include single- and double-stranded regions. In addition, the term "polynucleotide" as used herein refers to triple-stranded regions 5 comprising RNA or DNA or both RNA and DNA. The strands in such regions may be from the same molecule or from different molecules. The regions may include all of one or more of the molecules, but more typically involve only a region of some of the molecules. One of the molecules of a triple-helical region often is an oligonucleotide. The term "polynucleotide" specifically includes cDNAs. The term includes DNAs (including cDNAs) 10 and RNAs that contain one or more modified bases. Thus, DNAs or RNAs with backbones modified for stability or for other reasons are "polynucleotides" as that term is intended herein. Moreover, DNAs or RNAs comprising unusual bases, such as inosine, or modified bases, such as tritiated bases, are included within the term "polynucleotides" as defined herein. In general, the term "polynucleotide" embraces all chemically, enzymatically and/or 15 metabolically modified forms of unmodified polynucleotides, as well as the chemical forms of DNA and RNA characteristic of viruses and cells, including simple and complex cells. The term "oligonucleotide" refers to a relatively short polynucleotide, including, without limitation, single-stranded deoxyribonucleotides, single- or double-stranded ribonucleotides, RNA:DNA hybrids and double-stranded DNAs. Oligonucleotides, such as 20 single-stranded DNA probe oligonucleotides, are often synthesized by chemical methods, for example using automated oligonucleotide synthesizers that are commercially available. However, oligonucleotides can be made by a variety of other methods, including in vitro recombinant DNA-mediated techniques and by expression of DNAs in cells and organisms. The terms "differentially expressed gene," "differential gene expression" and their 25 synonyms, which are used interchangeably, refer to a gene whose expression is activated to a higher or lower level in a subject suffering from a disease, specifically cancer, such as breast cancer, relative to its expression in a normal or control subject. The terms also include genes whose expression is activated to a higher or lower level at different stages of the same disease. It is also understood that a differentially expressed gene may be either activated or 30 inhibited at the nucleic acid level or protein level, or may be subject to alternative splicing to result in a different polypeptide product. Such differences may be evidenced by a change in mRNA levels, surface expression, secretion or other partitioning of a polypeptide, for example. Differential gene expression may include a comparison of expression between two 12 3098652_1 (GHMatlers) P2417.AU.3 or more genes or their gene products, or a comparison of the ratios of the expression between two or more genes or their gene products, or even a comparison of two differently processed products of the same gene, which differ between normal subjects and subjects suffering from a disease, specifically cancer, or between various stages of the same disease. Differential 5 expression includes both quantitative, as well as qualitative, differences in the temporal or cellular expression pattern in a gene or its expression products among, for example, normal and diseased cells, or among cells which have undergone different disease events or disease stages. For the purpose of this invention, "differential gene expression" is considered to be present when there is at least an about two-fold, preferably at least about four-fold, more 10 preferably at least about six-fold, most preferably at least about ten-fold difference between the expression of a given gene in normal and diseased subjects, or in various stages of disease development in a diseased subject. The term "normalized" with regard to a gene transcript or a gene expression product refers to the level of the transcript or gene expression product relative to the mean levels of 15 transcripts/products of a set of reference genes, wherein the reference genes are either selected based on their minimal variation across, patients, tissues or treatments ("housekeeping genes"), or the reference genes are the totality of tested genes. In the latter case, which is commonly referred to as "global normalization", it is important that the total number of tested genes be relatively large, preferably greater than 50. Specifically, the term 20 'normalized' with respect to an RNA transcript refers to the transcript level relative to the mean of transcript levels of a set of reference genes. More specifically, the mean level of an RNA transcript as measured by TaqMan@ RT-PCR refers to the Ct value minus the mean Ct values of a set of reference gene transcripts. The terms "expression threshold," and "defined expression threshold" are used 25 interchangeably and refer to the level of a gene or gene product in question above which the gene or gene product serves as a predictive marker for patient response or resistance to a drug. The threshold typically is defined experimentally from clinical studies. The expression threshold can be selected either for maximum sensitivity (for example, to detect all responders to a drug), or for maximum selectivity (for example to detect only responders to a 30 drug), or for minimum error. The phrase "gene amplification" refers to a process by which multiple copies of a gene or gene fragment are formed in a particular cell or cell line. The duplicated region (a stretch of amplified DNA) is often referred to as "amplicon." Often, the amount of the 13 3098652_1 (GHMatters) P62417.AU.3 messenger RNA (mRNA) produced, i.e., the level of gene expression, also increases in the proportion to the number of copies made of the particular gene. The term "prognosis" is used herein to refer to the prediction of the likelihood of cancer-attributable death or progression, including recurrence, metastatic spread, and drug 5 resistance, of a neoplastic disease, such as breast cancer. The term "prediction" is used herein to refer to the likelihood that a patient will respond either favorably or unfavorably to a drug or set of drugs, and also the extent of those responses, or that a patient will survive, following surgical removal or the primary tumor and/or chemotherapy for a certain period of time without cancer recurrence. The predictive methods of the present invention can be used 10 clinically to make treatment decisions by choosing the most appropriate treatment modalities for any particular patient. The predictive methods of the present invention are valuable tools in predicting if a patient is likely to respond favorably to a treatment regimen, such as surgical intervention, chemotherapy with a given drug or drug combination, and/or radiation therapy, or whether long-term survival of the patient, following surgery and/or termination 15 of chemotherapy or other treatment modalities is likely. The term "long-term" survival is used herein to refer to survival for at least 3 years, more preferably for at least 8 years, most preferably for at least 10 years following surgery or other treatment. The term "tumor," as used herein, refers to all neoplastic cell growth and 20 proliferation, whether malignant or benign, and all pre-cancerous and cancerous cells and tissues. The terms "cancer" and "cancerous" refer to or describe the physiological condition in mammals that is typically characterized by unregulated cell growth. Examples of cancer include but are not limited to, breast cancer, colon cancer, lung cancer, prostate cancer, 25 hepatocellular cancer, gastric cancer, pancreatic cancer, cervical cancer, ovarian cancer, liver cancer, bladder cancer, cancer of the urinary tract, thyroid cancer, renal cancer, carcinoma, melanoma, and brain cancer. The "pathology" of cancer includes all phenomena that compromise the well-being of the patient. This includes, without limitation, abnormal or uncontrollable cell growth, 30 metastasis, interference with the normal functioning of neighboring cells, release of cytokines or other secretory products at abnormal levels, suppression or aggravation of inflammatory or immunological response, neoplasia, premalignancy, malignancy, invasion of surrounding or distant tissues or organs, such as lymph nodes, etc. 14 3098052_1 (GHMauers) P02417.AU 3 "Patient response" can be assessed using any endpoint indicating a benefit to the patient, including, without limitation, (1) inhibition, to some extent, of tumor growth, including slowing down and complete growth arrest; (2) reduction in the number of tumor cells; (3) reduction in tumor size; (4) inhibition (i.e., reduction, slowing down or complete 5 stopping) of tumor cell infiltration into adjacent peripheral organs and/or tissues; (5) inhibition (i.e. reduction, slowing down or complete stopping) of metastasis; (6) enhancement of anti-tumor immune response, which may, but does not have to, result in the regression or rejection of the tumor; (7) relief, to some extent, of one or more symptoms associated with the tumor; (8) increase in the length of survival following treatment; and/or (9) decreased 10 mortality at a given point of time following treatment. "Neoadjuvant therapy" is adjunctive or adjuvant therapy given prior to the primary (main) therapy. Neoadjuvant therapy includes, for example, chemotherapy, radiation therapy, and hormone therapy. Thus, chemotherapy may be administered prior to surgery to shrink the tumor, so that surgery can be more effective, or, in the case of previously 15 unoperable tumors, possible. "Stringency" of hybridization reactions is readily determinable by one of ordinary skill in the art, and generally is an empirical calculation dependent upon probe length, washing temperature, and salt concentration. In general, longer probes require higher temperatures for proper annealing, while shorter probes need lower temperatures. 20 Hybridization generally depends on the ability of denatured DNA to reanneal when complementary strands are present in an environment below their melting temperature. The higher the degree of desired homology between the probe and hybridizable sequence, the higher the relative temperature which can be used. As a result, it follows that higher relative temperatures would tend to make the reaction conditions more stringent, while lower 25 temperatures less so. For additional details and explanation of stringency of hybridization reactions, see Ausubel et al., Current Protocols in Molecular Biology, Wiley Interscience Publishers, (1995). "Stringent conditions" or "high stringency conditions", as defined herein, typically: (1) employ low ionic strength and high temperature for washing, for example 0.015 M 30 sodium chloride/0.0015 M sodium citrate/0.1% sodium dodecyl sulfate at 50*C; (2) employ during hybridization a denaturing agent, such as formamide, for example, 50% (v/v) formamide with 0.1% bovine serum albumin/0.1% Ficoll/0.1% polyvinylpyrrolidone/50mM sodium phosphate buffer at pH 6.5 with 750 mM sodium chloride, 75 mM sodium citrate at 15 3098852_ (GHMatler) P62417 AU.3 42*C; or (3) employ 50% formamide, 5 x SSC (0.75 M NaCl, 0.075 M sodium citrate), 50 mM sodium phosphate (pH 6.8), 0.1% sodium pyrophosphate, 5 x Denhardt's solution, sonicated salmon sperm DNA (50 pg/ml), 0.1% SDS, and 10% dextran sulfate at 42 0 C, with washes at 42*C in 0.2 x SSC (sodium chloride/sodium citrate) and 50% formamide at 55*C, 5 followed by a high-stringency wash consisting of 0.1 x SSC containing EDTA at 55 0 C. "Moderately stringent conditions" may be identified as described by Sambrook et al., Molecular Cloning: A Laboratory Manual, New York: Cold Spring Harbor Press, 1989, and include the use of washing solution and hybridization conditions (e.g., temperature, ionic strength and %SDS) less stringent that those described above. An example of moderately 10 stringent conditions is overnight incubation at 37 0 C in a solution comprising: 20% formamide, 5 x SSC (150 mM NaCl, 15 mM trisodium citrate), 50 mM sodium phosphate (pH 7.6), 5 x Denhardt's solution, 10% dextran sulfate, and 20 mg/ml denatured sheared salmon sperm DNA, followed by washing the filters in I x SSC at about 37-50*C. The skilled artisan will recognize how to adjust the temperature, ionic strength, etc. as necessary 15 to accommodate factors such as probe length and the like. In the context of the present invention, reference to "at least one," "at least two," "at least five," etc. of the genes listed in any particular gene set means any one or any and all combinations of the genes listed. B. Detailed Description 20 The practice of the present invention will employ, unless otherwise indicated, conventional techniques of molecular biology (including recombinant techniques), microbiology, cell biology, and biochemistry, which are within the skill of the art. Such techniques are explained fully in the literature, such as, "Molecular Cloning: A Laboratory Manual", 2 "d edition (Sambrook et al., 1989); "Oligonucleotide Synthesis" (M.J. Gait, ed., 25 1984); "Animal Cell Culture" (R.I. Freshney, ed., 1987); "Methods in Enzymology" (Academic Press, Inc.); "Handbook of Experimental Immunology", 4 th edition (D.M. Weir & C.C. Blackwell, eds., Blackwell Science Inc., 1987); "Gene Transfer Vectors for Mammalian Cells" (J.M. Miller & M.P. Calos, eds., 1987); "Current Protocols in Molecular Biology" (F.M. Ausubel et al., eds., 1987); and "PCR: The Polymerase Chain Reaction", (Mullis et al., 30 eds., 1994). 16 309852_1 (GHMatters) P6241?AU.3 1 . Gene Ex pression Profiling Methods of gene expression profiling include methods based on hybridization analysis of polynucleotides, methods based on sequencing of polynucleotides, and proteomics-based methods. The most commonly used methods known in the art for the 5 quantification of mRNA expression in a sample include northern blotting and in situ hybridization (Parker & Barnes, Methods in Molecular Biology 106:247-283 (1999)); RNAse protection assays (Hod, Biotechniques 13:852-854 (1992)); and PCR-based methods, such as reverse transcription polymerase chain reaction (RT-PCR) (Weis et al., Trends in Genetics 8:263-264 (1992)). Alternatively, antibodies may be employed that can recognize specific 10 duplexes, including DNA duplexes, RNA duplexes, and DNA-RNA hybrid duplexes or DNA-protein duplexes. Representative methods for sequencing-based gene expression analysis include Serial Analysis of Gene Expression (SAGE), and gene expression analysis by massively parallel signature sequencing (MPSS). 15 2. PCR-based Gene Expression Profiling Methods a. Reverse Transcriptase PCR (RT-PCR) One of the most sensitive and most flexible quantitative PCR-based gene expression profiling methods is RT-PCR, which can be used to compare mRNA levels in different sample populations, in normal and tumor tissues, with or without drug treatment, to 20 characterize patterns of gene expression, to discriminate between closely related mRNAs, and to analyze RNA structure. The first step is the isolation of mRNA from a target sample. The starting material is typically total RNA isolated from human tumors or tumor cell lines, and corresponding normal tissues or cell lines, respectively. Thus RNA can be isolated from a variety of primary 25 tumors, including breast, lung, colon, prostate, brain, liver, kidney, pancreas, spleen, thymus, testis, ovary, uterus, etc., tumor, or tumor cell lines, with pooled DNA from healthy donors. If the source of mRNA is a primary tumor, mRNA can be extracted, for example, from frozen or archived paraffin-embedded and fixed (e.g. formalin-fixed) tissue samples. General methods for mRNA extraction are well known in the art and are disclosed in 30 standard textbooks of molecular biology, including Ausubel et al., Current Protocols of Molecular Biology, John Wiley and Sons (1997). Methods for RNA extraction from paraffin embedded tissues are disclosed, for example, in Rupp and Locker, Lab Invest. 56:A67 (1987), and De Andrds et al., BioTechniques 18:42044 (1995). In particular, RNA isolation 17 3098852_1 (GHMatters) P82417.AU.3 can be performed using purification kit, buffer set and protease from commercial manufacturers, such as Qiagen, according to the manufacturer's instructions. For example, total RNA from cells in culture can be isolated using Qiagen RNeasy mini-columns. Other commercially available RNA isolation kits include MasterPureTM Complete DNA and RNA 5 Purification Kit (EPICENTRE@, Madison, WI), and Paraffin Block RNA Isolation Kit (Ambion, Inc.). Total RNA from tissue samples can be isolated using RNA Stat-60 (Tel Test). RNA prepared from tumor can be isolated, for example, by cesium chloride density gradient centrifugation. As RNA cannot serve as a template for PCR, the first step in gene expression 10 profiling by RT-PCR is the reverse transcription of the RNA template into cDNA, followed by its exponential amplification in a PCR reaction. The two most commonly used reverse transcriptases are avilo myeloblastosis virus reverse transcriptase (AMV-RT) and Moloney murine leukemia virus reverse transcriptase (MMLV-RT). The reverse transcription step is typically primed using specific primers, random hexamers, or oligo-dT primers, depending 15 on the circumstances and the goal of expression profiling. For example, extracted RNA can be reverse-transcribed using a GeneAmp RNA PCR kit (Perkin Elmer, CA, USA), following the manufacturer's instructions. The derived cDNA can then be used as a template in the subsequent PCR reaction. Although the PCR step can use a variety of thermostable DNA-dependent DNA 20 polymerases, it typically employs the Taq DNA polymerase, which has a 5'-3' nuclease activity but lacks a 3'-5' proofreading endonuclease activity. Thus, TaqMan@ PCR typically utilizes the 5'-nuclease activity of Taq or Tth polymerase to hydrolyze a hybridization probe bound to its target amplicon, but any enzyme with equivalent 5' nuclease activity can be used. Two oligonucleotide primers are used to generate an amplicon typical of a PCR 25 reaction. A third oligonucleotide, or probe, is designed to detect nucleotide sequence located between the two PCR primers. The probe is non-extendible by Taq DNA polymerase enzyme, and is labeled with a reporter fluorescent dye and a quencher fluorescent dye. Any laser-induced emission from the reporter dye is quenched by the quenching dye when the two dyes are located close together as they are on the probe. During the amplification reaction, 30 the Taq DNA polymerase enzyme cleaves the probe in a template-dependent manner. The resultant probe fragments disassociate in solution, and signal from the released reporter dye is free from the quenching effect of the second fluorophore. One molecule of reporter dye is 18 30988521 (GHMtters) P82417.AU.3 liberated for each new molecule synthesized, and detection of the unquenched reporter dye provides the basis for quantitative interpretation of the data. TaqMan@ RT-PCR can be performed using commercially available equipment, such as, for example, ABI PRISM 7 7 0 0 TM Sequence Detection System TM (Perkin-Elmer-Applied 5 Biosystems, Foster City, CA, USA), or Lightcycler (Roche Molecular Biochemicals, Mannheim, Germany). In a preferred embodiment, the 5' nuclease procedure is run on a real time quantitative PCR device such as the ABI PRISM 7 7 00 TM Sequence Detection SystemT. The system consists of a thermocycler, laser, charge-coupled device (CCD), camera and computer. The system amplifies samples in a 96-well format on a thermocycler. During 10 amplification, laser-induced fluorescent signal is collected in real-time through fiber optics cables for all 96 wells, and detected at the CCD. The system includes software for running the instrument and for analyzing the data. 5'-Nuclease assay data are initially expressed as Ct, or the threshold cycle. As discussed above, fluorescence values are recorded during every cycle and represent the 15 amount of product amplified to that point in the amplification reaction. The point when the fluorescent signal is first recorded as statistically significant is the threshold cycle (C). To minimize errors and the effect of sample-to-sample variation, RT-PCR is usually performed using a reference RNA which ideally is expressed at a constant level among different tissues, and is unaffected by the experimental treatment. RNAs most frequently 20 used to normalize patterns of gene expression are mRNAs for the housekeeping genes glyceraldehyde-3-phosphate-dehydrogenase (GAPD) and p-actin (ACTB). A more recent variation of the RT-PCR technique is the real time quantitative PCR, which measures PCR product accumulation through a dual-labeled fluorigenic probe (i.e., TaqMan@ probe). Real time PCR is compatible both with quantitative competitive PCR, 25 where internal competitor for each target sequence is used for normalization, and with quantitative comparative PCR using a normalization gene contained within the sample, or a housekeeping gene for RT-PCR. For further details see, e.g. Held et al., Genome Research 6:986-994 (1996). b. MassARRA Y System 30 In the MassARRAY-based gene expression profiling method, developed by Sequenom, Inc. (San Diego, CA) following the isolation of RNA and reverse transcription, the obtained cDNA is spiked with a synthetic DNA molecule (competitor), which matches the targeted cDNA region in all positions, except a single base, and serves as an internal 19 3098852.1 (GHMaters) P62417.AU.3 standard. The cDNA/competitor mixture is PCR amplified and is subjected to a post-PCR shrimp alkaline phosphatase (SAP) enzyme treatment, which results in the dephosphorylation of the remaining nucleotides. After inactivation of the alkaline phosphatase, the PCR products from the competitor and cDNA are subjected to primer extension, which generates 5 distinct mass signals for the competitor- and cDNA-derives PCR products. After purification, these products are dispensed on a chip array, which is pre-loaded with components needed for analysis with matrix-assisted laser desorption ionization time-of flight mass spectrometry (MALDI-TOF MS) analysis. The cDNA present in the reaction is then quantified by analyzing the ratios of the peak areas in the mass spectrum generated. For 10 further details see, e.g. Ding and Cantor, Proc. Natl. Acad Sci. USA 100:3059-3064 (2003). c. Other PCR-based Methods Further PCR-based techniques include, for example, differential display (Liang and Pardee, Science 257:967-971 (1992)); amplified fragment length polymorphism (iAFLP) (Kawamoto et al., Genome Res. 12:1305-1312 (1999)); BeadArray T M technology (Illumina, San 15 Diego, CA; Oliphant et al., Discovery of Markers for Disease (Supplement to Biotechniques), June 2002; Ferguson et al., Analytical Chemistry 72:5618 (2000)); BeadsArray for Detection of Gene Expression (BADGE), using the commercially available Luminex 100 LabMAP system and multiple color-coded microspheres (Luminex Corp., Austin, TX) in a rapid assay for gene expression (Yang et al., Genome Res. 11:1888-1898 (2001)); 20 and high coverage expression profiling (HiCEP) analysis (Fukumura et al., NucL. Acids. Res. 31(16) e94 (2003)). 3. Microarrays Differential gene expression can also be identified, or confirmed using the microarray technique. Thus, the expression profile of breast cancer-associated genes can be measured in 25 either fresh or paraffin-embedded tumor tissue, using microarray technology. In this method, polynucleotide sequences of interest (including cDNAs and oligonucleotides) are plated, or arrayed, on a microchip substrate. The arrayed sequences are then hybridized with specific DNA probes from cells or tissues of interest. Just as in the RT-PCR method, the source of mRNA typically is total RNA isolated from human tumors or tumor cell lines, and 30 corresponding normal tissues or cell lines. Thus RNA can be isolated from a variety of primary tumors or tumor cell lines. If the source of mRNA is a primary tumor, mRNA can be extracted, for example, from frozen or archived paraffin-embedded and fixed (e.g. 20 3098652_1 (GHMatters) P62417.AU.3 formalin-fixed) tissue samples, which are routinely prepared and preserved in everyday clinical practice. In a specific embodiment of the microarray technique, PCR amplified inserts of cDNA clones are applied to a substrate in a dense array. Preferably at least 10,000 nucleotide 5 sequences are applied to the substrate. The microarrayed genes, immobilized on the microchip at 10,000 elements each, are suitable for hybridization under stringent conditions. Fluorescently labeled cDNA probes may be generated through incorporation of fluorescent nucleotides by reverse transcription of RNA extracted from tissues of interest. Labeled cDNA probes applied to the chip hybridize with specificity to each spot of DNA on the array. 10 After stringent washing to remove non-specifically bound probes, the chip is scanned by confocal laser microscopy or by another detection method, such as a CCD camera. Quantitation of hybridization of each arrayed element allows for assessment of corresponding mRNA abundance. With dual color fluorescence, separately labeled cDNA probes generated from two sources of RNA are hybridized pairwise to the array. The relative abundance of the 15 transcripts from the two sources corresponding to each specified gene is thus determined simultaneously. The miniaturized scale of the hybridization affords a convenient and rapid evaluation of the expression pattern for large numbers of genes. Such methods have been shown to have the sensitivity required to detect rare transcripts, which are expressed at a few copies per cell, and to reproducibly detect at least approximately two-fold differences in the 20 expression levels (Schena et al., Proc. Nati. Acad. Sci. USA 93(2):106-149 (1996)). Microarray analysis can be performed by commercially available equipment, following manufacturer's protocols, such as by using the Affymetrix GenChip technology, or Incyte's microarray technology. The development of microarray methods for large-scale analysis of gene expression 25 makes it possible to search systematically for molecular markers of cancer classification and outcome prediction in a variety of tumor types. 4. Serial Analysis of Gene Expression (SAGE) Serial analysis of gene expression (SAGE) is a method that allows the simultaneous and quantitative analysis of a large number of gene transcripts, without the need of providing 30 an individual hybridization probe for each transcript. First, a short sequence tag (about 10-14 bp) is generated that contains sufficient information to uniquely identify a transcript, provided that the tag is obtained from a unique position within each transcript. Then, many transcripts are linked together to form long serial molecules, that can be sequenced, revealing 21 3098652_1 (GHMOter) P62417.AU.3 the identity of the multiple tags simultaneously. The expression pattern of any population of transcripts can be quantitatively evaluated by determining the abundance of individual tags, and identifying the gene corresponding to each tag. For more details see, e.g. Vclculescu et al., Science 270:484-487 (1995); and Velculescu et al., Cell 88:243-51 (1997). 5 5. Gene Expression Analysis by Massively Parallel Signature Sequencing (MPSS) This method, described by Brenner et al., Nature Biotechnology 18:630-634 (2000), is a sequencing approach that combines non-gel-based signature sequencing with in vitro cloning of millions of templates on separate 5 pim diameter microbeads. First, a microbead 10 library of DNA templates is constructed by in vitro cloning. This is followed by the assembly of a planar array of the template-containing microbeads in a flow cell at a high density (typically greater than 3 x 106 microbeads/cm 2 ). The free ends of the cloned templates on each microbead are analyzed simultaneously, using a fluorescence-based signature sequencing method that does not require DNA fragment separation. This method 15 has been shown to simultaneously and accurately provide, in a single operation, hundreds of thousands of gene signature sequences from a yeast cDNA library. 6. Immunohistochemistry Immunohistochemistry methods are also suitable for detecting the expression levels of the prognostic markers of the present invention. Thus, antibodies or antisera, preferably 20 polyclonal antisera, and most preferably monoclonal antibodies specific for each marker are used to detect expression. The antibodies can be detected by direct labeling of the antibodies themselves, for example, with radioactive labels, fluorescent labels, hapten labels such as, biotin, or an enzyme such as horse radish peroxidase or alkaline phosphatase. Alternatively, unlabeled primary antibody is used in conjunction with a labeled secondary antibody, 25 comprising antisera, polyclonal antisera or a monoclonal antibody specific for the primary antibody. Immunohistochemistry protocols and kits are well known in the art and are commercially available. 7. Proteomics The term "proteome" is defined as the totality of the proteins present in a sample (e.g. 30 tissue, organism, or cell culture) at a certain point of time. Proteomics includes, among other things, study of the global changes of protein expression in a sample (also referred to as "expression proteomics"). Proteomics typically includes the following steps: (1) separation of individual proteins in a sample by 2-D gel electrophoresis (2-D PAGE); (2) identification 22 3098852_1 (GHMatIers) Pe2417.AU.3 of the individual proteins recovered from the gel, e.g. my mass spectrometry or N-terminal sequencing, and (3) analysis of the data using bioinformatics. Proteomics methods are valuable supplements to other methods of gene expression profiling, and can be used, alone or in combination with other methods, to detect the products of the prognostic markers of the 5 present invention. 8. General Description of mRNA Isolation, Purification and Amplification The steps of a representative protocol for profiling gene expression using fixed, paraffin-embedded tissues as the RNA source, including mRNA isolation, purification, primer extension and amplification are given in various published journal articles (for 10 example: T.E. Godfrey et al. J. Molec. Diagnostics 2: 84-91 [2000]; K. Specht et al., Am. J. Pathol. 158: 419-29 [2001]). Briefly, a representative process starts with cutting about 10 [im thick sections of paraffin-embedded tumor tissue samples. The RNA is then extracted, and protein and DNA are removed. After analysis of the RNA concentration, RNA repair and/or amplification steps may be included, if necessary, and RNA is reverse transcribed using gene 15 specific promoters followed by RT-PCR. Finally, the data are analyzed to identify the best treatment option(s) available to the patient on the basis of the characteristic gene expression pattern identified in the tumor sample examined. 9. Cancer Chemotherapy Chemotherapeutic agents used in cancer treatment can be divided into several groups, 20 depending on their mechanism of action. Some chemotherapeutic agents directly damage DNA and RNA. By disrupting replication of the DNA such chemotherapeutics either completely halt replication, or result in the production of nonsense DNA or RNA. This category includes, for example, cisplatin (Platinol@), daunorubicin (Cerubidine@), doxorubicin (Adriamycin@), and etoposide (VePesid@). Another group of cancer 25 chemotherapeutic agents interfere with the formation of nucleotides or deoxyribonucleotides, so that RNA synthesis and cell replication is blocked. Examples of drugs in this class include methotrexate (Abitrexate@), mercaptopurine (Purinethol@), fluorouracil (Adrucil@), and hydroxyurea (Hydrea@). A third class of chemotherapeutic agents effects the synthesis or breakdown of mitotic spindles, and, as a result, interrupt cell division. Examples of drugs in 30 this class include Vinblastine (Velban®), Vincristine (Oncovin®) and taxenes, such as, Pacitaxel (Taxol®), and Tocetaxel (Taxotere@) Tocetaxel is currently approved in the United States to treat patients with locally advanced or metastatic breast cancer after failure of prior 23 3098652_1 (GHMatters) P62417.AU.3 chemotherapy, and patients with locally advanced or metastatic non-small cell lung cancer after failure of prior platinum-based chemotherapy. A common problem with chemotherapy is the high toxicity of chemotherapeutic agents, such as anthracyclines and taxenes, which limits the clinical benefits of this treatment 5 approach. Most patients receive chemotherapy immediately following surgical removal of tumor. This approach is commonly referred to as adjuvant therapy. However, chemotherapy can be administered also before surgery, as so called neoadjuvant treatment. Although the use of neo-adjuvant chemotherapy originates from the treatment of advanced and inoperable 10 breast cancer, it has gained acceptance in the treatment of other types of cancers as well. The efficacy of neoadjuvant chemotherapy has been tested in several clinical trials. In the multi center National Surgical Adjuvant Breast and Bowel Project B-18 (NSAB B-18) trial (Fisher et al., J. Clin. Oncology 15:2002-2004 (1997); Fisher et al., J. Clin. Oncology 16:2672-2685 (1998)) neoadjuvant therapy was performed with a combination of adriamycin and 15 cyclophosphamide ("AC regimen"). In another clinical trial, neoadjuvant therapy was administered using a combination of 5-fluorouracil, epirubicin and cyclophosphamide ("FEC regimen") (van Der Hage et al., J. Clin. Oncol. 19:4224-4237 (2001)). Newer clinical trials have also used taxane-containing neoadjuvant treatment regiments. See, e.g. Holmes et al., J. Natl. Cancer Inst. 83:1797-1805 (1991) and Moliterni et al., Seminars in Oncology, 24:S17 20 10-S-17-14 (1999). For further information about neoadjuvant chemotherapy for breast cancer see, Cleator et al., Endocrine-Related Cancer 9:183-195 (2002). 10. Cancer Gene Set, Assayed Gene Subsequences, and Clinical Application of Gene Expression Data An important aspect of the present invention is to use the measured expression of 25 certain genes by breast cancer tissue to provide prognostic information. For this purpose it is necessary to correct for (normalize away) differences in the amount of RNA assayed, variability in the quality of the RNA used, and other factors, such as machine and operator differences. Therefore, the assay typically measures and incorporates the use of reference RNAs, including those transcribed from well-known housekeeping genes, such as GAPD and 30 ACTB. A precise method for normalizing gene expression data is given in "User Bulletin #2" for the ABI PRISM 7700 Sequence Detection System (Applied Biosystems; 1997). Alternatively, normalization can be based on the mean or median signal (Ct) of all of the assayed genes or a large subset thereof (global normalization approach). In the study 24 3098852_1 (GHMatters) P62417.AU.3 described in the following Example, a so called central normalization strategy was used, which utilized a subset of the screened genes, selected based on lack of correlation with clinical outcome, for normalization. 11. Recurrence and Response to Therapy Scores and their Applications 5 Copending application Serial No. 60/486,302, filed on July 10, 2003, describes an algorithm-based prognostic test for determining the likelihood of cancer recurrence and/or the likelihood that a patient responds well to a treatment modality. Features of the algorithm that distinguish it from other cancer prognostic methods include: 1) a unique set of test mRNAs (or the corresponding gene expression products) used to determine recurrence likelihood, 2) 10 certain weights used to combine the expression data into a formula, and 3) thresholds used to divide patients into groups of different levels of risk, such as low, medium, and high risk groups. The algorithm yields a numerical recurrence score (RS) or, if patient response to treatment is assessed, response to therapy score (RTS). The test requires a laboratory assay to measure the levels of the specified mRNAs or 15 their expression products, but can utilize very small amounts of either fresh tissue, or frozen tissue or fixed, paraffin-embedded tumor biopsy specimens that have already been necessarily collected from patients and archived. Thus, the test can be noninvasive. It is also compatible with several different methods of tumor tissue harvest, for example, via core biopsy or fine needle aspiration. 20 According to the method, cancer recurrence score (RS) is determined by: (a) subjecting a biological sample comprising cancer cells obtained from said subject to gene or protein expression profiling; (b) quantifying the expression level of multiple individual genes [i.e., levels of mRNAs or proteins] so as to determine an expression value for each gene; 25 (c) creating subsets of the gene expression values, each subset comprising expression values for genes linked by a cancer-related biological function and/or by co expression; (d) multiplying the expression level of each gene within a subset by a coefficient reflecting its relative contribution to cancer recurrence or response to therapy within said 30 subset and adding the products of multiplication to yield a term for said subset; (e) multiplying the term of each subset by a factor reflecting its contribution to cancer recurrence or response to therapy; and 25 3098852.1 (GHMatters) P62417.AU.3 (f) producing the sum of terms for each subset multiplied by said factor to produce a recurrence score (RS) or a response to therapy (RTS) score, wherein the contribution of each subset which does not show a linear correlation with cancer recurrence or response to therapy is included only above a predetermined threshold 5 level, and wherein the subsets in which increased expression of the specified genes reduce risk of cancer recurrence are assigned a negative value, and the subsets in which expression of the specified genes increase risk of cancer recurrence are assigned a positive value. In a particular embodiment, RS is determined by: 10 (a) determining the expression levels of GRB7, HER2, EstRI, PR, Bcl2, CEGP1, SURV, Ki.67, MYBL2, CCNBI, STK15, CTSL2, STMY3, CD68, GSTM1, and BAGI, or their expression products, in a biological sample containing tumor cells obtained from said subject; and (b) calculating the recurrence score (RS) by the following equation: 15 RS = (0.23 to 0.70) x GRB7axisthresh - (0.17 to 0.51) x ERaxis + (0.53 to 1.56) x prolifaxisthresh + (0.07 to 0.21) x invasionaxis + (0.03 to 0.15) x CD68 - (0.04 to 0.25) x GSTMI - (0.05 to 0.22) x BAGI wherein (i) GRB7 axis = (0.45 to 1.35) x GRB7 + (0.05 to 0.15) x HER2; 20 (ii) if GRB7 axis < -2, then GRB7 axis thresh = -2, and if GRB7 axis > -2, then GRB7 axis thresh = GRB7 axis; (iii) ER axis = (EstI + PR + Bcl2 + CEGP])/4; (iv) prolifaxis = (SURV + Ki.67 + MYBL2 + CCNBI + STK15)/5; (v) if prolifaxis < -3.5, then prolifaxisthresh = -3.5, 25 if prolifaxis -3.5, then prolifaxishresh = prolifaxis; and (vi) invasionaxis = (CTSL2 + STMY3)/2, wherein the terms for all individual genes for which ranges are not specifically shown can vary between about 0.5 and 1.5, and wherein a higher RS represents an increased likelihood of cancer recurrence. 30 Further details of the invention will be described in the following non-limiting Example. 26 3098852_1 (GHMatters) P02417.AU.3 Example A Retrospective Study of Neoadjuvant Chemotherapy in Invasive Breast Cancer: Gene Expression Profiling of Paraffin-Embedded Core Biopsy Tissue This was a collaborative study involving Genomic Health, Inc., (Redwood City 5 California), and Institute Tumori, Milan, Italy. The primary objective of the study was to explore the correlation between pre-treatment molecular profiles and pathologic complete response (pCR) to neoadjuvant chemotherapy in locally advanced breast cancer. Patient inclusion criteria: Histologic diagnosis of invasive breast cancer (date of surgery 1998-2002); diagnosis 10 of locally advanced breast cancer defined by skin infiltration and or N2 axillary status and or homolateral supraclavicular positive nodes; core biopsy, neoadjuvant chemotherapy and surgical resection performed at Istituto Nazionale Tumori, Milan; signed informed consent that the biological material obtained for histological diagnosis or diagnostic procedures would be used for research; and histopathologic assessment indicating adequate amounts of 15 tumor tissue for inclusion in this research study. Exclusion criteria: Distant metastases; no tumor block available from initial core biopsy or from the surgical resection; or no tumor or very little tumor (<5% of the overall tissue on the slide) in block as assessed by examination of the H&E slide by the Pathologist. 20 Studv design Eighty-nine evaluable patients (from a set of 96 clinically evaluable patients) were identified and studied. The levels of 384 mRNA species were measured by RT-PCR, representing products of candidate cancer-related genes that were selected from the biomedical research literature. Only one gene was lost due to inadequate signal. 25 Patient characteristics were as follows: Mean age: 50 years; Tumor grades: 24% Well, 55% Moderate, and 21% Poor; Sixty-three % of patients were ER positive {by immunohistochemistry}; Seventy % of patients had positive lymph nodes. All patients were given primary neoadjuvant chemotherapy: Doxorubicin plus Taxol 3weeks/3 cycles followed by Taxol* (paclitaxel) Iweek/12 cycles. Surgical removal of the 30 tumor followed completion of chemotherapy. Core tumor biopsy specimens were taken prior to start of chemotherapy, and served as the source of RNA for the RT-PCR assay. 27 3098852 1 (GHMatter) P62417 AU.3 Materials and Methods Fixed paraffin-embedded (FPE) tumor tissue from biopsy was obtained prior to and after chemotherapy. Core biopsies were taken prior to chemotherapy. In that case, the pathologist selected the most representative primary tumor block, and submitted nine 10 5 micron sections for RNA analysis. Specifically, a total of 9 sections (10 microns in thickness each) were prepared and placed in three Costar Brand Microcentrifuge Tubes (Polypropylene, 1.7 mL tubes, clear; 3 sections in each tube) and pooled. Messenger RNA was extracted using the MasterPure TM RNA Purification Kit (Epicentre Technologies) and quantified by the RiboGreen@ fluorescence method 10 (Molecular probes). Molecular assays of quantitative gene expression were performed by RT PCR, using the ABI PRISM 7900TM Sequence Detection SystemTM (Perkin-Elmer-Applied Biosystems, Foster City, CA, USA). ABI PRISM 7900TM consists of a thermocycler, laser, charge-coupled device (CCD), camera and computer. The system amplifies samples in a 384-well format on a thermocycler. During amplification, laser-induced fluorescent signal is 15 collected in real-time for all 384 wells, and detected at the CCD. The system includes software for running the instrument and for analyzing the data. Analysis and Results Tumor tissue was analyzed for 384 genes. The threshold cycle (CT) values for each patient were normalized based on the median of a subset of the screened genes for that 20 particular patient, selected based on lack of correlation with clinical outcome (central normalization strategy). Patient beneficial response to chemotherapy was defined as pathologic complete response (pCR). Patients were formally assessed for response at the completion of all chemotherapy. A clinical complete response (cCR) requires complete disappearance of all clinically 25 detectable disease, either by physical examination or diagnostic breast imaging. A pathologic complete response (pCR) requires absence of residual breast cancer on histologic examination of biopsied breast tissue, lumpectomy or mastectomy specimens following primary chemotherapy. Residual ductal carcinoma in situ (DCIS) may be present. Residual cancer in regional nodes may not be present. Of the 89 evaluable patients I1 (12%) 30 had a pathologic complete response (pCR). Seven of these patients were ER negative. A partial clinical response was defined as a > 50% decrease in tumor area (sum of the products of the longest perpendicular diameters) or a > 50% decrease in the sum of the 28 3098652_1 (GHMatters) P62417 AU.3 products of the longest perpendicular diameters of multiple lesions in the breast and axilla. No area of disease may increase by > 25% and no new lesions may appear. Analysis was performed by comparing the relationship between normalized gene expression and the binary outcomes of pCR or no pCR. Univariate generalized models were 5 used with probit or logit link functions. See, e.g. Van K. Borooah, LOGIT and PROBIT, Ordered Multinominal Models, Sage University Paper, 2002. Table I presents pathologic response correlations with gene expression, and lists the 86 genes for which the p-value for the differences between the groups was <0.1. The second column (with the heading "Direction") denotes whether increased expression correlates with 10 decreasing or increasing likelihood of response to chemotherapy. The statistical significance of the predictive value for each gene is given by P-value (right hand column) 29 3098852.1 (GHMatters) P82417.AU.3 Probit Link Gene Direction Intercept Slope P-value TBP Decreasing 0.0575 2.4354 0.0000 ILT.2 Increasing 0.5273 -0.9489 0.0003 ABCC5 Decreasing 0.9872 0.8181 0.0003 CD18 Increasing 3.4735 -1.0787 0.0007 GATA3 Decreasing 0.6175 0.2975 0.0008 DICER1 Decreasing -0.9149 1.4875 0.0013 MSH3 Decreasing 2.6875 0.9270 0.0013 GBP1 Increasing 1.7649 -0.5410 0.0014 IRS1 Decreasing 1.3576 0.5214 0.0016 CD3z Increasing 0.1567 -0.5162 0.0018 Fasl Increasing -0.6351 -0.4050 0.0019 TUBB Decreasing 1.2745 0.8267 0.0025 BAD Decreasing 0.9993 1.1325 0.0033 ERCC1 Decreasing 0.0327 1.0784 0.0039 MCM6 Increasing 0.1371 -0.8008 0.0052 PR Decreasing 1.6079 0.1764 0.0054 APC Decreasing 0.7264 1.0972 0.0061 GGPS1 Decreasing 1.0906 0.8124 0.0062 KRT18 Decreasing -0.8029 0.4506 0.0063 ESRRG Decreasing 2.0198 0.2262 0.0063 E2F1 Increasing 0.2188 -0.5277 0.0068 AKT2 Decreasing -1.3566 1.1902 0.0074 A.Catenin Decreasing -0.6859 0.9279 0.0079 CEGP1 Decreasing 1.3355 0.1875 0.0091 NPDO09 Decreasing 1.3996 0.2971 0.0092 MAPK14 Decreasing 2.6253 1.6007 0.0093 RUNX1 Decreasing -0.4138 0.7214 0.0103 ID2 Increasing 1.7326 -0.7032 0.0104 G.Catenin Decreasing -0.1221 0.5954 0.0110 FBXO5 Increasing 0.3421 -0.4935 0.0110 FHIT Decreasing 1.9966 0.4989 0.0113 MTA1 Decreasing 0.3127 0.6069 0.0133 ERBB4 Decreasing 1.4591 0.1436 0.0135 FUS Decreasing -0.6150 0.9415 0.0137 BBC3 Decreasing 2.4796 0.6495 0.0138 IGF1R Decreasing 1.1998 0.3116 0.0147 CD9 Decreasing -0.9292 0.5747 0.0156 TPS3BP1 Decreasing 1.4325 0.8122 0.0169 MUC1 Decreasing 0.8881 0.2140 0.0175 IGFBP5 Decreasing -0.6180 0.4880 0.0181 rhoC Decreasing -0.1726 0.6860 0.0184 RALBP1 Decreasing 0.2383 0.9509 0.0185 CDC20 Increasing 1.3204 -0.4390 0.0186 STAT3 Decreasing -0.9763 0.7023 0.0194 ERK1 Decreasing 0.8577 0.6496 0.0198 HLA.DPB1 Increasing 3.6300 -0.6035 0.0202 SGCB Decreasing 0.6171 0.7823 0.0208 30 3098852.1 (GHMaters) P62417.AU.3 Probit Link Gene Direction Intercept Slope P-value CGA Increasing 0.0168 -0.1450 0.0209 DHPS Decreasing 0.2957 0.7840 0.0216 MGMT Decreasing 0.9238 0.6876 0.0226 CRIP2 Decreasing 0.5524 0.4394 0.0230 MMP12 Increasing 0.4208 -0.2419 0.0231 ErbB3 Decreasing 0.9438 0.2798 0.0233 RAP1GDS1 Decreasing 0.2617 0.7672 0.0235 CDC25B Increasing 1.6965 -0.5356 0.0264 IL6 Increasing 0.0592 -0.2388 0.0272 CCND1 Decreasing 0.2260 0.2992 0.0272 CYBA Increasing 2.6493 -0.5175 0.0287 PRKCD Decreasing 0.2125 0.6745 0.0291 DR4 Increasing 0.3039 -0.5321 0.0316 Hepsin Decreasing 1.9211 0.1873 0.0318 CRABP1 Increasing 1.0309 -0.1287 0.0320 AK055699 Decreasing 2.0442 0.1765 0.0343 Contig.51037 Increasing 0.7857 -0.1131 0.0346 VCAM1 Increasing 1.1866 -0.3560 0.0346 FYN Increasing 1.5502 -0.5624 0.0359 GRB7 Increasing 1.3592 -0.1646 0.0375 AKAP.2 Increasing 1.7946 -0.7008 0.0382 RASSF1 Increasing 1.1972 -0.0390 0.0384 MCP1 Increasing 1.3700 -0.3805 0.0388 ZNF38 Decreasing 1.7957 0.4993 0.0395 MCM2 Increasing 1.0574 -0.4695 0.0426 GBP2 Increasing 1.4095 -0.4559 0.0439 SEMA3F Decreasing 1.2706 0.3725 0.0455 CD31 Increasing 1.9913 -0.5955 0.0459 COL1A1 Decreasing -1.9861 0.3812 0.0466 ER2 Increasing -0.5204 -0.2617 0.0471 BAG1 Decreasing 0.6731 0.5070 0.0472 AKT1 Decreasing -0.4467 0.5768 0.0480 COL1A2 Decreasing -1.0233 0.3804 0.0490 STAT1 Increasing 1.9447 -0.4062 0.0498 Wnt.5a Decreasing 2.2244 0.2983 0.0518 PTPD1 Decreasing 1.2950 0.4834 0.0552 RAB6C Decreasing 0.4841 0.5635 0.0717 TK1 Increasing 0.6127 -0.3625 0.0886 BcI2 Decreasing 1.1459 0.2509 0.0959 Based on the data set forth in Table 1, increased expression of the following genes correlates with increased likelihood of complete pathologic response to treatment: ILT.2; CD18; GBPl; CD3z; fasi; MCM6; E2FI; 1D2; FBX05; CDC20; HLA.DPB1; CGA; 5 MMP12; CDC25B; IL6; CYBA; DR4; CRABP1; Contig.51037; VCAMI; FYN; GRB7; AKAP.2; RASSF1; MCPl; MCM2; GBP2; CD31; ER2; STATI; TKI; while increased 31 3098652_ (GHMatters) P62417.AU.3 expression of the following genes correlates with decreased likelihood of complete pathologic response to treatment: TBP; ABCC5; GATA3; DICERl; MSH3; IRS 1; TUBB; BAD; ERCCI; PR; APC; GGPSI; KRT18; ESRRG; AKT2; A.Catenin; CEGP1; NPDO09; MAPK14; RUNXl; G.Catenin; FHIT; MTAI; ErbB4; FUS; BBC3; IGFIR; CD9; TP53BPl; 5 MUCI; IGFBP5; rhoC; RALBPl; STAT3; ERKI; SGCB; DHPS; MGMT; CRIP2; ErbB3; RAPIGDSI; CCND1; PRKCD; Hepsin; AK055699; ZNF38; SEMA3F; COLlAl; BAGI; AKTI; COLlA2; Wnt.5a; PTPDI; RAB6C; Bcl2. The relationship between the recurrence risk algorithm (described in copending U.S. application Serial No. 60/486,302) and pCR was also investigated. The algorithm 10 incorporates the measured levels of 21 mRNA species. Sixteen mRNAs (named below) were candidate clinical markers and the remaining 5 (ACTB, GAPD, GUSB, RPLPO, and TFRC) were reference genes. Reference-normalized expression measurements range from 0 to 15, where a one unit increase reflects a 2-fold increase in RNA. The Recurrence Score (RS) is calculated from the quantitative expression of four sets 15 of marker genes (an estrogen receptor group of 4 genes-ESRI, PGR, BCL2, and SCUBE2; a proliferation set of 5 genes-MK167, MYBL2, BIRC5, CCNB1, and STK6; a HER2 set of 2 genes-ERBB2 and GRB7, an invasion group of 2 genes-MMPI I and CTSL2) and 3 other individual genes-GSTMI, BAG 1, and CD68. Although the genes and the multiplication factors used in the equation may vary, in a 20 typical embodiment, the following equation may be used to calculate RS: RS (range, 0 - 100) = + 0.47 x HER2 Group Score - 0.34 x ER Group Score + 1.04 x Proliferation Group Score + 0.10 x Invasion Group Score + 0.05 x CD68 - 0.08 x GSTM1 - 0.07 x BAGI Application of this algorithm to study clinical and gene expression data sets yields a continuous curve relating RS to pCR values, as shown in Figure 1. Examination of these data 25 shows that patients with RS > 50 are in the upper 50 percentile of patients in terms of 32 30852_1 (GHMats3) P82417 AU.3 likelihood of response to chemotherapy, and that patients with RS < 35 are in the lower 50 percentile of patients in terms of likelihood of response to chemotherapy. All references cited throughout the disclosure are hereby expressly incorporated by reference. 5 While the invention has been described with emphasis upon certain specific embodiments, it is be apparent to those skilled in the art that variations and modification in the specific methods and techniques are possible. Accordingly, this invention includes all modifications encompassed within the spirit and scope of the invention as defined by the following claims. I0 33 309M521 (GHMetters) P62417.AU.3 TABLE 2 A-Catenin NM 00190 S2138/A 7 Cate.f2 CGTTCCGATCCTCTATACTGCAT 23 A-Catenin NM001 90 S21391A-Cate.r2 AGGTCCCTGTTGGOCTTATAGG 22 A-Catenin NM_00190 S4725/A'-Cate.p2 ATGCCTACAGCACCCTGATGTCGCA 25 ABCC5 NM_00568S5605/ABCC5.fl TGCAGACTGTACCATGCTGA 20 ABCC5 NM 00568 S5606/ABCC5.rl GGCCAGCACCATAATCCTAT 20 ABCC5 NM00568 S5607/ABCC5.pl CTGCACACGGTTCTAGGCTCCG 22 AK055699 AK055699 S2097/AK0556.fl CTGCATGTGATTGAATAAGAAACAAGA 27 AK055699 AK055699 S2098/AK0556.rl TGTGGACCTGATCCCTGTACAC 22 AK055699 AK055699 S5057/AK0556.pl TGACCACACCAAAGCCTCCCTGG 23 AKAP-2 NM 00720SI374/AKAP-2.fl ACGAATTGTCGGTGAGGTCT 20 AKAP-2 NM_00720S1375/AKAP-2.r1 GTCCATGCTGAAATCATTGG 20 AKAP-2 NM_00720 S4934/AKAP-2.p1 CAGGATACCACAGTCCTGGAGACCC 25 AKTI NM 00516 SOO1O/AKTI.f3 CGCTTCTATQGCGCTGAGAT 20 AKT1 NM_00516 S012/AKTt.r3 TCCCGGTACACCACGTTCTT 20 AKT1 NM_00516 S4776/AKT1.p3 CAGCCCTGGACTACCTGCACTCGG 24 AKT2 NM_00162S0828/AKT2.f3 TCCTGCCACCCTTCAAACC 19 AKT2 NM..00162 S0829/AKT2.r3. GGCGGTAAATTCATCATCGAA 21 AKT2 NM_00162S4727/AKT2.p3 CAGGTCACGTCCGAGGTCGACACA 24 APC NM_00003 S0022/APC.f4 GGACAGCAGGAATGTGTTTC 20 APC NM_00003 S0024/APC.r4 ACCCACTCGATTTGTTTCTG 20 APC NM_00003 S4888/APC.p4 CATTGGCTCCCCGTGACCTGTA 22 BAD. NM_03298 S201 1/BAD.fl GGGTCAGGTGCCTCGAGAT 19 BAD NM_03298 S2012/BAD.rl CTGCTCACTCGGCTCAAACTC 21 BAD NM'03298 S5058/BAD.pl TGGGCCCAGAGCATGTTCCAGATC 24 BAG1 NM_00432 SI386/BAG1,f2 CGTTGTCAGCACTTGGAATACAA 23 BAG1 NM_00432 S1387/BAG1 .r2 . GTTCAACCTPTTCCTGTGGACTGT 24 BAG1 NM_00432 S4731/BAG 1.p2 CCCAATTAACATGACCCGGCAACCAT . 26 BBC3 NM 01441 S1584/BBC3.f2 CCTGGAGGGTCCTGTACAAT 20 BBC3 NM_01441 S1585/BBC3.r2 CTAATTGGGCTCCATCTCG 19 BBC3 NM_01441 S4890/BBC3.p2 .CATCATGGGACTCCTGCCCTTACC . 24 Bcl2 NM_ 00063 S0043/Bcl2.f2 CAGATGGACCTAGTACCCACTGAGA 25 Bcl2 NM 00063 S0045/Bcl2.r2 CCTATGATTTAAGGGCATTTTTCC 24 Bc12 NM_00063 S4732/Bcl2.p2 TTCCACGCCGAAGGACAGCGAT 22 CCND1 NM_00175 S0058/CCND1..f3 GCATGTTCGTGGCCTCTAAGA . 21 CCNDI NM_00175 S0060/CCND1.r3 CGGTGTAGATGCACAGCTTCTC . 22 CCNDI NM_00175 S4986/CCND1.p3 AAGGAGACCATCCCCCTGACGGC 23 CD18 NM_00021 S0061/CD18.f2 CGTCAGGACCCACCATGTCT 20 CD18 NM_00021 S0063/CD18.r2 GGTTAATTGGTGACATCCTCAAGA 24 CD18 NM_00021 S 4987 /CD1 8 .p 2 CGCGGCCGAGACATGGCTTG 20 CD31 NM 00044 S1407/CD31.f3 TGTATTTCAAGACCTCTGTGCACTT . 25 CD31 NM_00044 S1408/CD31.r3 TTAGCCTGAGGAATTGCTGTGTT 23 CD31 NM 00044 S4939/CD31 .p3 TTTATGAACCTGCCCTGCTCCCACA 25 CD3z NM 00073 S0064/CD3z.f I AGATGAAGTGGAAGGCGCTT 20 CD3z NM_00073 S0066/CD3z.rl TGCCTCTGTAATCGGCAACTG 21 C03z NM_00073 S4988/CD3z.pl CACCGCGGCCATCCTGCA 18 CD9 NM 00176 S0686/CD9.f1 .GGGCGTGGAACAGTTTATCT 20 CD9 NM_00176 S0687/CD9.rl CACGGTGAAGGTTTCGAGT 19 CD9 NM 00176 S4792/CD9.pl AGACATCTGCCCCAAGAAGGACGT 24 CDC20 NM_00125 S4447/CDC20.fI TGGATTGGAGTTCTGGGAATG .21 CDC20 NM00 125 S4448/CDC20.rl GCTTGCACTCCACAGGTACACA 22 CDC20 NM_00125 S4449/CDC20.pl ACTGGCCGTGGCACTGGACAACA 23 CDC25B NM_02187 S1160/CDC25B.fl AAACGAGCAGTTTGCCATCAG . 21 CDC25B NM_02187 Si161/CDC25B.rl GTTGGTGATGTTCCGAAGCA 20 CDC25B NM_02187 S4842/CDC25Bpf CCTCACCGGCATAGACTGGAAGCG 24 CEGP1 NM_02097S1494/CEGP1.f2 TGACAATCAGCACACCTGCAT - 21 CEGPI NM_02097 SI495/CEGP1.r2 TGTGACTACAGCCGTGATCCTTA 23 CEGPI NM_02097 S4735/CEGP1.p2 CAGGCCCTCTTCCGAGCGGT 20 CGA (CHG NM_00127 S3221/CGA (C.f3 CTGAAGGAGCTCCAAGACCT 20 CGA (CHG NM-001 27 S3222/CGA (C.r3 CAAAACCGCTGTGTTTCTTC 20 CGA (CHG NM_00127 S3254/CGA (C.p3 TGCTGATGTGCCCTCTCCTTGG 22 COL1Al NM_00008 S4531 ICOLI A.fi GTGGCCATCCAGCTGACC 18 CO'lAl NM_00008 S45321COL1 Al .rl CAGTGGTAGGTGATGTTCTGGGA 23 COLIAl NM 00008 S45331COL1Al.pl TCCTGCGCCTGATGTCCACCG 21 COLIA2 NM_00008 S4534/COL1.A2.f1 CAGCCAAGAACTGGTATAGGAGCT 24 COL1A2 NM_00008 S4535/COL1A2.rl AAACTGGCTGCCAGCATTG 19 COL1A2 NM_0Q008 S4536/COL1A2.pl TCTCCTAGCCAGACGTGTTTCTTGTCCTTG 30 Contig 510 XM_05894 S2070/Contig.f1 CGACAGTTGCGATGAAAGTTCTAA 24 Contig 510 XM05894 S2071/Contig.rl. GGCTGCTAGAGACCATGGACAT 22 Contig 510 XMI0594 S5059/Contig.pi CCTCCTCCTGTTGCTGCCACTAATGCT 27 CRABPI NM_00437 S5441/CRABPI.f3 AACTTCAAGGTCGGAGAAGG 20 CRABP1 NM 00437 Sb442/RA P1.r 'I GU I AAACTCCTGCACTTG 20 CRABP1 NM_.00437 S5443/CRABP1.p3 CCGTCCACGGTCTCCTCCTCA 21 CRIP2 NM_00131 S5676/CRIP2.f3 GTGCTACGCCACCCTGTT 18. CRIP2 NM_00131 S5677/CRIP2.r3 CAGGGGCTTCTCGTAGATGT 20 CRIP2 NM -00131 S5678/CRIP2.p3 CCGATGTTCACGCCTTTGGGTC 22 .CYBA NM_00010 S5300/CYBA.fl GGTGCCTACTCCATTGTGG 19 CYBA NM_0001OS5301/CYBAr1 GTGGAGCCCTTCTTCCTCTT 20 CYBA NM_00010 S5302/CYBA.pl TACTCCAGCAGGCACACAAACACG 24 DHPS ., NM 01340 S4519/DHPS.f3 GGGAGAACGGGATCAATAGGAT 22 DHPS NM_01 340 S4520/DHPS.r3 GCATCAGCCAGTCCTCAAACT 21 DHPS NM_- 01340S45211DHPS.p3 CTCATTGGGCACCAGCAGGTTTCC 24 DICERI NM_17743 S5294/DICER1.f2 TCCAATTCCAGCATCACTGT 20 DICER1 NM 17743 S5295/DICER1.r2- GGCAGTGAAGGCGATAAAGT 20 DICERI NM_17743 S5296/DICERl.p2 AGAAAAGCTGTTTGTCTCCCCAGCA 25 DR4 NM_00384 S2532/DR4.f2 TGCACAGAGGGTGTGGGTTAC. 21 DR4 NM_00384 S2533/DR4.r2 TCTTCATCTGATTTACAAGCTGTACATG 28 DR4 NM_00384 S4981/DR4.p2 CAATGCTTCCAACAATTTGTTTGCTTGCC 29 E2F1 NM_.00522 S3063/E2F1.f3 ACTCCCTCTACCCTTGAGCA 20 * E2F1 NM_- 00522 S3064/E2F1.r3' CAGGCCTCAGTTOCTTCAGT 20 E2Fl NM_00522 S4821/E2F1.p3 CAGAAGAACAGCTCAGGGACCCCT 24 ER2 NM 00143 S0109/ER2.f2 TGGTCCATCGCCAGTTATCA 20 ER2 NM_00143 S011 1/ER2.r2 TGTTCTAGCGATCTTGCTTCACA 23 ER2 NM 00143 S5001/ER2.p2 ATCTGTATGCGGAACCTCAAAAGAGTCCCT 30 ErbB3 NM00198 S01 12/ErbB3.fl CGGTTATGTCATGCCAGATACAC 23 ErbB3 NM_00198 S01 14/ErbB3.rl GAACTGAGACCCACTGAAGAAAGG 24 ErbB3 NM_00198 S5002/ErbB3.pl CCTCAAAGGTACTCCCTCCTCCCGG 25 ERBB4 NM_00523 Si231/ERBB4.f3 TGGCTCTTAATCAGTTTCGTTACCT 25 ERBB4 NM_00523 Si 232/ERBB4.r3 CAAGGCATATCGATCCTCATAAAGT 25 ERBB4 NM_00523 S4891/ERBB4.p3 TGTCCCACGAATAATGCGTAAATTCTCCAG 30 ERCC1 NM 00198 S2437/ERCCI.f2 GTCCAGGTGGATGTGAAAGA 20 ERCC1 NMf00198 S2438/ERCC1 .r2 CGGCCAGGATACACATCTTA 20 ERCCI NM_00198S4920/ERCC1.p2 CAGCAGGCCCTCAAGGAGCTG 21 ERKi Z11696 S1560/ERK1.f3 ACGGATCACAGTGGAGGAAG -20 ERK1 Z11696 S1561/ERK1.r3 CTCATCCGTCGGGTCATAGT 20 ERKI Z11696 S4882/ERK1.p3 CGCTGGCTCACCCCTACCTG 20 ESRRG NM_.00143 S6130/ESRRG.f3 CCAGCACCATTGTTGAAGAT 20 ESRRG NM_00143S6131/ESRRG.r3 AGTCTCTTGGGCATCGAGTT . 20 ESRRG NM001 43 S6132/ESRRG.p3 CCCCAGACCAAGTGTGAATACATGCT 26 fasi . NM_00063 S0121/fasf2 GCACTTTGGGATTCTTTCOATTAT 24 fasl NM_00063 So1 23/fasI.r2 GCATGTAAGAAGACCCTCACTGAA 24 fasi NM_00063 S5004/fasi.p2 ACAACATTCTCGGTGCCTGTAACAAAGA 29 FBXO5 NM_01217 S2017/FBXO5.rl GGATTGTAGACTGTCACCGAAATTC 25 FBXO5 NM_01217 S2018/FBXO5.f 1 GGCTATTCCTCATTTTCTCTACAAAGTG 28 FBXO5 NM01217 S5061/FBXO5.p1 CCTCCAGGAGGCTACCTTCTTCATGTTCAC 30 FHIT NM_00201 S2443/FHIT.fl . CCAGTGGAGCGCTTCCAT 18 FHIT NM_00201 S2444/FHIT.r1 CTCTCTGGGTCGTCTGAAACAA 22 FHIT NM00;201 S4921/FHIT.pl TCGGCCACTTCATCAGGACGCAG 23 FUS NM_00496S2936/FUS.f1 GGATAATTCAGACAACAACACCATCT 26 FUS NM_00496 S2937/FUS.rl TGAAGTAATCAGCCACAGACTCAAT FUS NM_00496 S48D1/FUS.pl TCAATTGTAACATTCTCACCCAGGCCTTG 29 FYN NM-00203 S5695/FYN.f3 GAAGCGCAGATCATGAAGAA 20 FYN NM_00203 S5696/FYN.r3 CTCCTCAGACACCACTGCAT 20 FYN NM_00203 S5697/FYN.p3 CTGAAGCACGACAAGCTGGTCCAG 24 G-Catenin NM_00223 S2153/G-Cate.fl. TCAGCAGCAAGGGCATCAT 19 G-Catenin NM_00223 S2154/G-Cate.rl GGTGGTTTTCTTGAGCGTGTACT . 23 G-Catenin NM_00223 S5044/G-Cate.pl CGCCCGCAGGCCTCATCCT 19 GATA3 NM00205 SOI 27/GATA3.f3 CAAAGGAGCTCACTGTGGTGTCT 23 GATA3 NM00205 S01 29/GATA3.r3 GAGTCAGAATGGCTTATTCACAGATG 26 GATA3 NM00205 S5005/GATA3.p3 TGTTCCAACCACTGAATCTGGACC 24 GBP1 NM00205 S5698/GBP1.fl TTGGGAAATATTTGGGCATT 20 GBP1 NM_00205 S5699/GBP1.ri AGAAGCTAGGGTGGTTGTCC 20 GBP1 NM_.00205S5700/GBP1.pl TTGGGACATTGTAGACTTGGCCAGAC 26 GBP2 NM00412 S5707/GBP2.f2 GCATGGGAACCATCAACCA 19 GBP2 NM_00412 S5708/GBP2.r2 TGAGGAGTTTGCCTTGATTCG 21 GBP2 NM_00412 S5709/GBP2.p2 CCATGGACCAACTTCACTATGTGACAGAGC 30 GGPSI NM00483 SI 590/GGPSI.f1 CTCCGACGTGGCTTTCCA 18 GGPS1 NM_00483 Si 591 /GGPS1.rl CGTAATTGGCAGAATTGATGACA 23 GGPS1 NM_00483 S4896/GGPS1.pl TGGCCCACAGCATCTATGGAATCCC 25 GRB7 NM_00531 SO130/GRB7.f2 CCATCTGCATCCATCTTGTT 20 GRB7 NM_005,31 SO132/GRB7.r2 GGCCACCAGGGTATTATCTG 20 GRB7 NM_00531 S4726/GRB7.p2 CTCCCCACCCTTGAGAAGTGCCT 23 Hepsin NM_00215 S2269/Hepsin.f 1 AGGCTGCTGGAGGTCATCTC 20 Hepsin NM00215 S2270/Hepsin.r1 CTTCCTGCGGCCACAGTCT 19 Hepsin NM00215 S2271 /Hepsin.pl CCAGAGGCCGTTTCTTGGCCG 21 HLA-DPBI NM_00212 S4573/HLA-DP.fl TCCATGATGGTTCTGCAGGTT 21 HLA-DPB1 NM_00212 S4574/HLA-DP.r1 TGAGCAGCACCATCAGTAACG 21 HLA-DPB1 NM_00212 S4575/HLA-DP.p1 CCCCGGACAGTGGCTCTGACG 21 ID2 NM_00216 S01 51/D2.f4 AACGACTGCTACTCCAAGCTCAA 23 ID2 NM_00216 S0153/1ID2.r4 GGATTTCCATCTTGCTCACCTT 22 ID2 NM_00216 S5009/ID2.p4 TGCCCAGCATCCCCCAGAACAA 22 IGFIR NM 00087 S1249/IGF1R.f3 GCATGGTAGCCGAAGATTTCA 21 IGF1R NM_00087 S1250/IGF1R.r3 TTTCCGGTAATAGTCTGTCTCATAGATATC 30 IGF1R NM00087 S4895/lGF1 R.p3 CGCGTCATACCAAAATCTCCGATTTTGA 28 IL6 NM_00060 S0760/L6.f3 CCTGAACCTTCCAAAGATGG 20 IL6 NM_00060 S0761/IL6.r3 ACCAGGCAAGTCTCCTCATT- 20 IL6 NM00060 S4800/lL6.p3 CCAGATTGGAAGCATCCATC I I I I I CA 27 37 ILT-2 NM_00666 S1611/ILT-2.f2 AGCCATCACTCTCAGTGCAG 20 ILT-2 NM00666 Sl612/ILT-2.r2 ACTGCAGAGTCAGGGTCTCC 20 ILT-2 NM_00666 S4904/ILT-2.p2 CAGGTCCTATCGTGGCCCCTGA 22 IRSi NM_00554 S1943/IRS1.f3 CCACAGCTCACCTTCTGTCA 20 IRS1 NM00554 S1944/IRSI.r3 CCTCAGTGCCAGTCTCTTCC 20 IRSI NM_00554 S5050/IRSI.p3 TCCATCCCAGCTCCAGCCAG 20 KRT18 NM 00022 S1710/KRT18.f2 AGAGATCGAGGCTCTCAAGG 20 KRT18 NM_00022 S1711/KRT18.r2 GGCCTTTTACTTCCTCTTCG 20 KRT18 NM_00022 S4762/KRT18.p2 TGGTTCTTCTTCATGAAGAGCAGCTCC 27 MAPK14 NM_13901 S5557/MAPK14.f2 TGAGTGGAAAAGCCTGACCTATG 23 MAPK14 NM13901 S5558/MAPK14.r2 GGACTCCATCTCTTCTTGGTCAA 23 MAPK14 NM_13901 S5559/MAPKi4.p2 TGAAGTCATCAGCTTTGTGCCACCACC 27 MCM2 NM_00452S1602/MCM212 GACTTTTGCCCGCTACCTTTC 21 MCM2 NM00452 SI 603/MCM2.r2 GCCACTAACTGCTTCAGTATGAAGAG 26 MCM2 NM00452 S4900/MCM2.p2 ACAGCTCATTGTTGTCACGCCGGA 24 MCM6 NM00591 S1704/MCM6.f3 TGATGGTCCTATGTGTCACATTCA 24 MCM6 NM 00591 S1705/MCM6.r3 TGGGACAGGAAACACACCAA 20 MCM6 NM_00591 S4919/MCM6.p3 CrAGGTTTCATACCAACACAGGCTTCA CAC 30 MCPl NM 00298 S1955/MCP1.f1 CGCTCAGCCAGATGCAATC 19 MCP1 NM_00298 S1956/MCP1.rl GCACTGAGATCTTCCTATTGGTGAA 25 MCP1 NM00298 S5052/MCP1.pl TGCCCCAGTCACCTGCTGTTA 21 MGMT NM_00241 S1922/MGMT.fl GTGAAATGAAACGCACCACA 20 MGMT NM 00241 S1923/MGMT.rl GACCCTGCTCACAACCAGAC 20 MGMT NM_00241 S5045/MGMT.pl CAGCCCTTTGGGGAAGCTGG 20 MMP12 NM_00242 S4381 /MMP1 2.f2 CCAACGCTTGCCAAATCCT 19 MMP12 NM_00242 S4382/MMP12.r2 ACGGTAGTGACAGCATCAAAACTC 24 MMP12 NM_00242 S4383/MMP12.p2 AACCAGCTCTCTGTGACCCCAATT 24 MSH3 NM_00243 S5940/MSH3.f2 TGATTACCATCATGGCTCAGA 21 MSH3 NM_00243 S5941/MSH3.r2 CTTGTGAAAATGCCATCCAC 20 MSH3 NM_00243 S5942/MSH3.p2 TCCCAATTGTCGCTTCTTCTGCAG 24 MTAI NM_00468 S2369/MTA1.f I CCGCCCTCACCTGAAGAGA 19 MTA1 NM 00468 S2370/MTA1.rl GGAATAAGTTAGCCGCGCTTCT 22 MTA1 NM_00468 S4855/MTA1.pl CCCAGTGTCCGCCAAGGAGCG 21 MUCI NM_00245 S0782/MUC1.f2 GGCCAGGATCTGTGGTGGTA 20 MUC1 NM_00245 SO783/MUCI.r2 CTCCACGTCGTGGACATTGA 20 MUCi NM_00245'S4807/MUC1.p2 CTCTGGCCTTCCGAGAAGGTACC 23 NPDO09 (A NM_02068 S4474/NPDO09.f3 GGCTGTGGCTGAGGCTGTAG 20 NPDO09 (A NM 02068 S4475/NPDO09.r3 GGAGCATTCGAGGTCAAATCA 21 NPD009 (A NM_02068 S4476/NPDO09.p3. TTCCCAGAGTGTCTCACCTCCAGCAGAG 28 PR NM_00092 S1336/PR.f6 GCATCAaGCTGTCATTATGG 20 PR NM_00092 S1 337/PR.r6 AGTAGTTGTGCTGCCCTTCC 20 PR NM_00092 S4743/PR.p6 TGTCCTTACCTGTGGGAGCTGTAAGGTC 28 PRKCD NM00625 S1738/PRKCD.f2 CTGACACTTGCCGCAGAGAA 20 PRKCD NM_00625 S1739/PRKCD.r2 AGGTGGTCCTTGGTCTGGAA 20 PRKCD NM00625 S4923/PRKCD.p2 CCCTTTCTCACCCACCTCATCTGCAC 26 PTPDI NM_00703 S3069/PTPDi.f2 CGCTTGCCTAACTCATACTTTCC 23 PTPOI NM_00703 S3070/PTPD1.r2 CCATTCAGACTGCGCCACTT 20 PTPDI NM_00703 S4822/PTPD1.p2 TCCACGCAGCGTGGCACTG 19 RA86C NM_03214 S5535/RAB6C.fi - GCGACAGCTCCTCTAGTTCCA 21 RAB6C NM_03214 S5537/RAB6C.pl TTCCCGAAGTCTCCGCCCG 19 RAB6C NM_03214 S5538/RAB6C.rl GGAACACCAGCTTGAATTTCCT 22 RALBPI NM_00678 S5853/RALBP1.f I GGTGTCAGATATAAATGTGCAAATGC 26 RALBP1 NM_00678 S5854/RALBP1.r1 TftGATATTGCCAGCAGCTATAAA 24 RALBPI NM_00678 S5855/RALBPI.p1 TGCTGTCCTGTCGGTCTCAGTACGTTCA 28 RAPIGDS NM_02115 S5306/RAP1GD.f2 TGTGGATGCTGGATTGATTT 20 RAP1GDS NM 02115.S5307/RAPIGD.r2 AAGCAGCACTTCCTGGTCTT- 20 RAP1GDS NM_02115 S5308/RAP1GD.p2 CCACTGGTGCAGCTGCTAAATAGCA 25 RASSFI NM_00718 S2393/RASSF1.f3 AGTGGGAGACACCTGACCTT 20 RASSF1 NM 00718 S2394/RASSF1.r3 TGATCTGGGCATTGTACTCC 20 RASSFI NM 00718 S4909/RASSF1..p3 TTGATCTTCTGCTCAATCTCAGCTTGAGA 29 rhoC NM 00516 S2162/rhoC.fl CCCGTTCGGTCTGAGGAA 18 rhoC NM 00516 S2163/rhoC.rl GAGCACTCAAGGTAGCCAAAGG 22 rhoC NM 00516 S5042/rhoC.pl TCCGGTTCGCCATGTCCCG 19 RUNX1 NM_09175 S4588/RUNX1.f2 AACAGAGACATTGCCAACCA 20 RUNX1 -NM_00175 S4589/RUNX1.r2 GTGATTGCCCAGGAAGTTT 20 RUNXI NM_00175 S4590/RUNXI.p2 TTGGATCTGCVTGCTGTCCAAACC .24 SEMA3F NM 00418 S2857/SEMA3F.f3 CGCGAGCCCCTCATTATACA 20 SEMA3F NM 00418 S2858/SEMA3F.r3 CACTCGCCGTTGACATCCT 19 SEMA3F NM _00418 S4972/SEMA3F.p3 CTCCCCACAGCGCATCGAGGAA 22 SGCB NM_00023 S5752/SGCB.fi CAGTGGAGACCAGTTGGGTAGTG 23 SGCB NM_00023 S5753/SGCB.r1 CCTTGAAGAGCGTCCCATCA 20 SGCB NM_00023 S5754/SGCB.pi CACACATGCAGAGCTTGTAGCGTACCCA 28 STAT1 . NM. 00731 S1542/STAT1.f3 GGGCTCAGCTTTCAGAAGTG 20 STAT1 NM 00731 S1543/STAT1.r3 ACATGTTCAGCTGGTCCACA 20 STATIC NM 00731 S4878/STAT1.p3 TGGCAGTTTTCTTCTGTCACCAAAA 25 STAT3 NM_003151 S545/STAT3.f1 TCA.CATGCCACTTTGGTGTT 20 STAT3 NM_00315 S1546/STAT3.rl CTTGCAGGAAGCGGCTATAC 20 STAT3 NM00315 S4881/STAT3.pI TCCTGGGAGAGATTGACCAGCA 22 TBP NM 00319 SO262JTBP.fl GCCCGAAACbCCGAATATA 19 TBP NM_00319 S0264/TBP.rl CGTGGCTCTCTTATCCTCATGAT 23 TBP NM_00319 S4751/TBP.pl TACCGCAGCAAACCGCTTGGG 21 TK1 NM_00325 S0866/TK1.f2 GCCGGGAAGACCGTAATTGT 20 TKI NM_00325 S0927/TK1.r2 CAGCGGCACCAGGTTCAG 18 TK1 NM 00325 S4798/TKI.p2 CAAATGGCTTCCTCTOGAAGGTCCCA 26 TP53BP1 NM_00565 S1 747/TP53BP.f2 TGCTGTTGCTGAGTCTGTTG 20 TP53BP1 NM_00565-SI 748/TP53BP.r2 CTTGCCTGGCTTCACAGATA 20 TP53BPI NM_00 65 S4924/TP53BP.p2 CCAGTCCCCAGAAGACCATGTCTG 24 TUBB NM 00106 S5826/TUBB.f3 TGTGGTGAGGAAGGAGTCAG 20 TUBB NM_00106 S5827/TUBB.r3 CCCAGAGAGTGGGTCAGC 18 TUBB* NM 00106 S5828/TUBB.p3 CTGTGACTGTCTCCAGGGCTTCCA 24 VCAMI NM_00107 S3505NCAM1.f 1 TGGCTTCAGGAGCTGAATACC 21 VCAM1 NM 00107 S3506/VCAMI.rl TGCTGTOGTGATGAGAAAATAGTG 24 VCAM1 NM_00107 S3507NCAM1.pl CAGGCACACACAGGTGGGACACAAAT 26 Wnt-5a NM_00339 S6183/Wnt-5a.fI GTATCAGGACCACATGCAGTACATC 25 Wnt-5a NM 00339 S6184/Wnt-5a.rl TGTCGGAATTGATACTGGCATT 22 Wnt-5a NM_00339 S6185/Wnt-5a.pl TTGATGCCTGTCTTCGCGCCTTCT 24 ZNF38 NM_14591 S5593/ZNF38.f-3 TTTCCAAACATCAGCGAGTC 20 ZNF38 NM_14591 S5594/ZNF38.r3 AACAGGAGCGCTTGAAAGTT 20 ZNF38 NM14591 S5595/ZNF38.p3 ACGGTGCTTCTCCCTCTCCAGTG 23 TABLE 3 <0 0I-P 'U 00 O8<* <0 6 0 I< -I < 0 M unM I- m0 < 04 0D) 10o-< 00 0 u o7 0 <D< 8 001 0 L)0 <069O i.- 0 << 8oo884:I.~~ oo o8-1' CDD 8~C8o 0 u
«~~
0 0 O 60 C0~O OD *0~ 0.< < 00 Uo~ 6 u pD -C0 1-C 0 . (DC3 0(OD L-D- ou(~0 0 100 l R 0 cc0 a QD (C D CD P (DO cL c C. 0 1- 1 (DI. ~88Orto Z. ~~~m caccCD CD0~5 0 40 < CD 00 10D C <j 0 I- -C D CO i-D~ aC.) D 0 1. C < 0.0<0 8 0 CD ~OID OuCoIoL<C U0 uou .-aD .< 8 <0~~ 0 <~~CD 00o<Co0 V 'u 00D~ ~~00. DC3 CDOSD VCo 0D < DC c 0 C .) 0 00 ~C o ~D 4 oooD0 - a 9 LD ;.: Q- CD-D--C C.,q <CD<CD~ 8 g FwCD OfOC<D oD ~o-'69.. .) t- - o CD 0 UFO'gl ' R~ 08 L~ IL)o I- -- 8-< c L) 8 0 CD0

Claims (14)

1. A method for determining the likelihood of a beneficial response of a human patient with breast cancer to an anthracycline-based chemotherapy, comprising: 5 determining a normalized expression level of an RNA transcript, or its expression product, of ILT2 in a breast cancer sample obtained from said patient; and predicting the likelihood of a beneficial response of the patient to the anthracycline based chemotherapy using said normalized ILT2 expression level, wherein said normalized ILT2 expression level is positively correlated with the likelihood of a beneficial response of 10 the patient to the anthracycline-based chemotherapy.
2. The method of claim 1, wherein said breast cancer is invasive breast cancer.
3. The method of claim I or claim 2, wherein the anthracycline-based chemotherapy 15 comprises administering doxorubicin.
4. The method of any one of claims 1 to 3, wherein the anthracycline-based chemotherapy comprises administration of a taxane in addition to an anthracycline. 20
5. The method of claim 4, wherein the taxane is docetaxel.
6. The method of claim 4, wherein said taxane is paclitaxel.
7. The method of any one of claims I to 6, wherein said chemotherapy is adjuvant 25 chemotherapy.
8. The method of any one of claims 1 to 6, wherein said chemotherapy is neoadjuvant chemotherapy. 30
9. The method of any one of claims 1 to 8, wherein said breast cancer sample is fixed, paraffin-embedded, fresh, or frozen. 41 4111401 (GHMatters) P82417AU3 5-Mar-13
10. The method of any one of claims 1 to 9, wherein said RNA is isolated from a fixed, paraffin-embedded breast cancer sample of said patient.
11. The method of any one of claims I to 10, wherein the beneficial response is a clinical 5 complete response.
12. The method of any one of claims I to 10, wherein the beneficial response is a pathological complete response. 10
13. The method of any one of claims I to 12, wherein the expression level of said RNA transcript is determined by RT-PCR.
14. The method of claim 1, substantially as hereinbefore described with reference to the examples and figures. 42 41114601 (GHMatlers) P62417.AU.3 5-Mar-13
AU2012200431A 2004-04-09 2012-01-25 Gene expression markers for predicting response to chemotherapy Active AU2012200431B2 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
AU2012200431A AU2012200431B2 (en) 2004-04-09 2012-01-25 Gene expression markers for predicting response to chemotherapy
AU2013206082A AU2013206082B2 (en) 2004-04-09 2013-05-30 Gene expression markers for predicting response to chemotherapy
AU2016210735A AU2016210735B2 (en) 2004-04-09 2016-08-05 Gene expression markers for predicting response to chemotherapy

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US60/561,035 2004-04-09
AU2011204944A AU2011204944B2 (en) 2004-04-09 2011-07-22 Gene expression markers for predicting response to chemotherapy
AU2012200431A AU2012200431B2 (en) 2004-04-09 2012-01-25 Gene expression markers for predicting response to chemotherapy

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
AU2011204944A Division AU2011204944B2 (en) 2004-04-09 2011-07-22 Gene expression markers for predicting response to chemotherapy

Related Child Applications (1)

Application Number Title Priority Date Filing Date
AU2013206082A Division AU2013206082B2 (en) 2004-04-09 2013-05-30 Gene expression markers for predicting response to chemotherapy

Publications (2)

Publication Number Publication Date
AU2012200431A1 AU2012200431A1 (en) 2012-02-16
AU2012200431B2 true AU2012200431B2 (en) 2013-06-27

Family

ID=45812245

Family Applications (2)

Application Number Title Priority Date Filing Date
AU2012200431A Active AU2012200431B2 (en) 2004-04-09 2012-01-25 Gene expression markers for predicting response to chemotherapy
AU2016210735A Active AU2016210735B2 (en) 2004-04-09 2016-08-05 Gene expression markers for predicting response to chemotherapy

Family Applications After (1)

Application Number Title Priority Date Filing Date
AU2016210735A Active AU2016210735B2 (en) 2004-04-09 2016-08-05 Gene expression markers for predicting response to chemotherapy

Country Status (1)

Country Link
AU (2) AU2012200431B2 (en)

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001074388A1 (en) * 2000-03-31 2001-10-11 Idec Pharmaceuticals Corporation Combined use of anti-cytokine antibodies or antagonists and anti-cd20 for the treatment of b cell lymphoma
WO2004074506A2 (en) * 2003-02-13 2004-09-02 Mergen Ltd Polynucleotide sequences and corresponding encoded polypeptides of particular secreted and membrane-bound proteins overexpressed in certain cancers
WO2004074518A1 (en) * 2003-02-20 2004-09-02 Genomic Health, Inc. Use of intronic rna to measure gene expression

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001074388A1 (en) * 2000-03-31 2001-10-11 Idec Pharmaceuticals Corporation Combined use of anti-cytokine antibodies or antagonists and anti-cd20 for the treatment of b cell lymphoma
WO2004074506A2 (en) * 2003-02-13 2004-09-02 Mergen Ltd Polynucleotide sequences and corresponding encoded polypeptides of particular secreted and membrane-bound proteins overexpressed in certain cancers
WO2004074518A1 (en) * 2003-02-20 2004-09-02 Genomic Health, Inc. Use of intronic rna to measure gene expression

Also Published As

Publication number Publication date
AU2012200431A1 (en) 2012-02-16
AU2016210735B2 (en) 2017-09-07
AU2013206082A1 (en) 2013-06-20
AU2016210735A1 (en) 2016-08-25

Similar Documents

Publication Publication Date Title
AU2010241525B2 (en) Gene expression markers for predicting response to chemotherapy
US10619215B2 (en) Prediction of likelihood of cancer recurrence
AU2004248120B2 (en) Gene expression markers for predicting response to chemotherapy
AU2011204944B2 (en) Gene expression markers for predicting response to chemotherapy
AU2016210735B2 (en) Gene expression markers for predicting response to chemotherapy
KR20070022694A (en) Gene Expression Markers for Predicting Response to Chemotherapy
AU2019261715A1 (en) Prediction of likelihood of cancer recurrence

Legal Events

Date Code Title Description
FGA Letters patent sealed or granted (standard patent)