(19)
(11)EP 3 359 508 B1

(12)EUROPEAN PATENT SPECIFICATION

(45)Mention of the grant of the patent:
09.09.2020 Bulletin 2020/37

(21)Application number: 16852924.6

(22)Date of filing:  04.10.2016
(51)International Patent Classification (IPC): 
C12Q 1/6886(2018.01)
(86)International application number:
PCT/CA2016/000247
(87)International publication number:
WO 2017/059521 (13.04.2017 Gazette  2017/15)

(54)

TARGETING THE HISTONE PATHWAY TO DETECT AND OVERCOME ANTHRACYCLIN RESISTANCE

ABZIELUNG AUF DEN HISTONSIGNALWEG ZUR DETEKTION UND ÜBERWINDUNG VON ANTHRACYCLINRESISTENZ

CIBLAGE DE LA VOIE DE L'HISTONE POUR DÉTECTER ET VAINCRE LA RÉSISTANCE À L'ANTHRACYCLINE


(84)Designated Contracting States:
AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

(30)Priority: 06.10.2015 US 201562237928 P

(43)Date of publication of application:
15.08.2018 Bulletin 2018/33

(73)Proprietor: Ontario Institute for Cancer Research
Toronto, Ontario M5G 1L7 (CA)

(72)Inventors:
  • SPEARS, Melanie
    Ontario M8Z 3V5 (CA)
  • BARTLETT, John, M. S.
    Toronto, Ontario M4G 1S9 (CA)
  • BRAUNSTEIN, Marsela
    Waterloo, Ontario N2T 2V2 (CA)
  • KRZYZANOWKSI, Paul, M.
    Toronto, Ontario M6C 3X8 (CA)
  • KALATSKAYA, Irina
    Toronto, Ontario M5T 2Y5 (CA)
  • STEIN, Lincoln
    Toronto, Ontario M4E 2R3 (CA)

(74)Representative: Elsy, David 
Withers & Rogers LLP 4 More London Riverside
London SE1 2AU
London SE1 2AU (GB)


(56)References cited: : 
WO-A1-2014/047723
WO-A2-2007/085497
  
  • BRAUNSTEIN ET AL: "Abstract: P 5-08 -14: Deregulated histone and cell cycle pathways are associated with anthracycline resistance in breast cancer''", CANCER RESEARCH, PROCEEDINGS: AACR ANNUAL MEETING 2014; APRIL 5-9, 2014; SAN DIEGO, CA , vol. 73, no. 24 1 December 2013 (2013-12-01), XP009509146, ISSN: 0008-5472, DOI: 10.1158/0008-5472.SABCS13-P5-08-14 Retrieved from the Internet: URL:http://cancerres.aacrjournals.org/cont ent/73/24_Supplement/P5-08-14
  • DAVID J VILLENEUVE ET AL: "cDNA microarray analysis of isogenic paclitaxel and doxorubicin-resistant breast tumor cell lines reveals distinct drug-specific genetic signatures of resistance", BREAST CANCER RESEARCH AND TREATMENT, KLUWER ACADEMIC PUBLISHERS, BO, vol. 96, no. 1, 1 March 2006 (2006-03-01), pages 17-39, XP019274976, ISSN: 1573-7217
  • NAYAK SHWETA R ET AL: "A Role for Histone H2B Variants in Endocrine-Resistant Breast Cancer", HORMONES AND CANCER, SPRINGER NEW YORK LLC, US, vol. 6, no. 5, 26 June 2015 (2015-06-26), pages 214-224, XP035964490, ISSN: 1868-8497, DOI: 10.1007/S12672-015-0230-5 [retrieved on 2015-06-26]
  • PAMELA MÜNSTER ET AL: "Phase I Trial of Histone Deacetylase Inhibition by Valproic Acid Followed by the Topoisomerase II Inhibitor Epirubicin in Advanced Solid Tumors: A Clinical and Translational Study", JOURNAL OF CLINICAL ONCOLOGY, vol. 25, no. 15, 20 May 2007 (2007-05-20), pages 1979-1985, XP055576868, US ISSN: 0732-183X, DOI: 10.1200/JCO.2006.08.6165
  • BRAUNSTEIN ET AL.: 'Abstract: P 5-08 -14: Deregulated histone and cell cycle pathways are associated with anthracycline resistance in breast cancer''.' CANCER RESEARCH vol. 73, no. 24, 01 December 2013, XP009509146 DOI: 10.1158/0008-5472.SABCS13-P5-08-14 ISSN: 1538-7445 Retrieved from the Internet: <URL:http://cancerres.aacrjournals.org/cont ent/73/24_Supplement/P5-08-14> [retrieved on 2016-12-07]
  • SANCHEZ-GONZALEZ ET AL.: 'Antileukemia activity of the combination of an anthracycline with a histone deacetylase inhibitor''.' BLOOD vol. 108, no. 4, 15 August 2006, ISSN 0006-4971 pages 1174 - 1182, XP055372703
  • DATABASE GEO 17 June 2010 NCBI: 'Illumina HumanHT-12 V4.0 expression beadchip''.', XP055372713 Database accession no. GPL10558
  • CHERNOVA ET AL.: 'Histone modifications and cancer: biomarkers of prognosis?''.' AMERICAN JOURNAL OF CANCER RESEARCH vol. 2, no. 5, 15 September 2012, ISSN 2156-6976 pages 589 - 597, XP055116681
  • BRAUNSTEIN ET AL.: 'Downregulation of Histone H2A and H2B pathways is associated with anthracycline sensitivity in breast cancer''.' BREAST CANCER RESEARCH vol. 18, no. 16, 01 January 2016, pages 1 - 16, XP055520565 DOI: 10.1186/S13058-016-0676-6
  
Note: Within nine months from the publication of the mention of the grant of the European patent, any person may give notice to the European Patent Office of opposition to the European patent granted. Notice of opposition shall be filed in a written reasoned statement. It shall not be deemed to have been filed until the opposition fee has been paid. (Art. 99(1) European Patent Convention).


Description

FIELD OF THE INVENTION



[0001] The invention relates to the targeting of the histone pathway in order to assess and overcome anthracycline resistance.

BACKGROUND OF THE INVENTION



[0002] Breast cancer is the second leading cause of cancer death for women. Most patients present with early disease and are treated with surgery often followed by adjuvant radiotherapy and chemotherapy +/- endocrine therapy or trastuzumab given with curative intent; nevertheless, 40-50% of high-risk patients treated with adjuvant chemotherapy ultimately relapse as a result of them having resistant disease (EBCTCG 2005). Despite the advent of targeted therapies, chemotherapy is also central to the treatment of women with metastatic disease, who often respond to palliative chemotherapy but in due course relapse due to drug resistance, including cross-resistance to structurally unrelated anti-cancer drugs (Guo et al. 2004).

[0003] The taxanes and anthracyclines are widely used as adjuvant therapy, but also in the metastatic setting. Both target rapidly proliferating cancer cells. The taxanes interfere with microtubule depolymerisation, causing cell-cycle arrest (Ringel and Horwitz 1991; Chazard et al. 1994), whereas anthracyclines introduce DNA breaks, form free radicals and covalently bind topoisomerase II-DNA complexes (Minotti et al. 2004; Minotti et al. 2004). The taxanes and anthracyclines are both natural products and susceptible to resistance mediated by over-expression of the multidrug transporter P-glycoprotein. A well-established in vitro mechanism of resistance involves activity of MDR1 and MDR2/3, which bind non-specifically to multiple drugs and actively export them across the cellular membrane (Schinkel et al. 1991; van der Bliek et al. 1988). Although this results in decreased intracellular drug concentrations and cytotoxicity, the clinical relevance of MDR genes remains to be determined. Other mechanisms include reduced topoisomerase activity (Giaccone et al. 1992; de Jong et al. 1990), reduced Fas ligand expression (Friesen et al. 1997) and downregulation of TP53 expression (Lowe et al. 1993). However, the molecular drivers of clinical anthracycline resistance remain largely unknown. Applicant previously identified duplication of centromeric region on chromosome 17 (CEP17), a surrogate marker of chromosomal instability, as a predictive marker of clinical anthracycline sensitivity (Munro et al. 2012; Pritchard et al. 2012; Bartlett et al. 2015). However, identifying pathways that could be targeted in the clinic to eliminate anthracycline-resistant breast cancer remains a major challenge.

[0004] WO 2007/085497 A2 describes methods comprising the measurement of transcription of at least one of a group of genes for determining the prognosis or outcome of treatment of anthracycline in a subject with cell proliferative disorder.

[0005] Braunstein et al (Abstract P5-08-14: Dereguated histone and cell cycle pathways are associated with anthracycline resistance in breast cancer; AACR Annual meeting 2014) described deregulation of histone H2A and H2B genes in epirubicin-resistant breast cancer cell lines.

[0006] Villeneuve, DJ et al (cDNA microarray analysis of isogenic paclitaxel- and doxorubicin-resistant breast tumour cell lines reveals distinct drug-specific genetic signatures of resistance, Breast Cancer Research and Treatment, (2006) 96: 17-39) described drug-specific changes in gene expression in doxorubicin resistance including HIST1-H2BC.

[0007] Nayak, SR et al (A role for histone H2B variants in endocrine-resistant breast cancer, Hormones and Cancer, (2015) 6: 214-224) showed that overexpression of histone variants may be important in resistance to aromatase inhibitors and endocrine response in estrogen receptor-positive breast cancer.

[0008] WO 2014/047723 A1 describe a method of prognosing cancer recurrence in non-small cell lung cancer based on gene expression data from 22 genes, including HIST3H2A.

SUMMARY OF THE INVENTION



[0009] The invention is defined in the appended claims. In one embodiment, there is provided a method for determining a likelihood of resistance to anthracycline in a patient with breast cancer comprising: detecting a level of expression in a sample from the patient, of HIST1H2BK, HIST1H2BD, NEDD9, SYTL2, NHP2, ARPP19, TXNRD1, CENPF, STMN1, CCT5, APRT, UBEC2C, BAX, HDAC1, E2F1, E2F2, E2F4 and CDKN2A; comparing the level of the genes detected to a level of expression of the genes in a control sample; and wherein there is a likelihood of anthracycline resistance if there is a relatively higher level of expression of the genes in the subject sample compared to the control sample.

[0010] In another embodiment, there is provided a method for prognosticating survival in cancer patient comprising: detecting a level of expression in the sample of HIST1H2BK, HIST1H2BD, NEDD9, SYTL2, NHP2, ARPP19, TXNRD1, CENPF, STMN1, CCT5, APRT, UBEC2C, BAX, HDAC1, E2F1, E2F2, E2F4 and CDKN2A; comparing the level of the genes detected to a level of expression of the genes in a control sample; and wherein there is a likelihood of poor survival if there is a relatively higher level of expression of the genes in the subject sample compared to the control sample.

[0011] In one example, there is provided a use of a histone deacetylase inhibitor in the treatment of a cancer patient receiving anthracycline and exhibiting upregulation of at least one histone gene.

[0012] In another example, there is provided a use of a histone deacetylase inhibitor in the treatment of a breast cancer patient receiving anthracycline.

[0013] In yet another example, there is provided a method of sensitizing, or re-sensitizing, a patient with breast cancer to anthracycline, comprising administering to the patient a histone deacetylase inhibitor.

[0014] In yet another example, there is provided a composition comprising a plurality of reagents, preferably nucleic acid sequences, wherein each of the reagents is for detecting a level of expression in the sample of a gene in the regulatory pathway of at least one histone gene from the H1, H2A, H2B, H3 and H4 gene families.

[0015] In one example, there is provided an array comprising, for a plurality of genes in the regulatory pathway of at least one histone gene from the H1, H2A, H2B, H3 and H4 gene families, one or more polynucleotide probes complementary and hybridizable to an expression product of the gene.

[0016] In another example, there is provided a kit for determining a likelihood of resistance to anthracycline in a patient, comprising detection agents for detecting a level of expression in the sample of a gene in the regulatory pathway of at least one histone gene from the H1, H2A, H2B, H3 and H4 gene families, and instructions for use.

[0017] In yet another example, there is provided a kit for prognosticating survival in cancer patient, comprising detection agents for detecting a level of expression in the sample of a gene in the regulatory pathway of at least one histone gene from the H1, H2A, H2B, H3 and H4 gene families, and instructions for use.

[0018] In another example, there is provided a computer program product for use in conjunction with a computer having a processor and a memory connected to the processor, the computer program product comprising a computer readable storage medium having a computer mechanism encoded thereon, wherein the computer program mechanism may be loaded into the memory of the computer and cause the computer to carry out the method as defined in the claims.

[0019] In one embodiment, there is provided a computer implemented product for determining a likelihood of resistance to anthracycline in a breast cancer patient comprising: a means for receiving values corresponding to a subject expression profile in a subject sample; a database comprising a control expression profile associated with HIST1H2BK, HIST1H2BD, NEDD9, SYTL2, NHP2, ARPP19, TXNRD1, CENPF, STMN1, CCT5, APRT, UBEC2C, BAX, HDAC1, E2F1, E2F2, E2F4 and CDKN2A; and a processor disposed to compare the subject expression profile to the control expression profile and determine a likelihood of anthracycline resistance if there is a relatively higher level of expression of the genes in the subject sample compared to the control sample.

[0020] In another embodiment, there is provided a computer implemented product for prognosticating survival in a breast cancer patient comprising: a means for receiving values corresponding to a subject expression profile in a subject sample; and a database comprising a control expression profile associated HIST1H2BK, HIST1H2BD, NEDD9, SYTL2, NHP2, ARPP19, TXNRD1, CENPF, STMN1, CCT5, APRT, UBEC2C, BAX, HDAC1, E2F1, E2F2, E2F4 and CDKN2A; and a processor disposed to compare the subject expression profile to the control expression profile and determine there is a likelihood of poor survival if there is a relatively higher level of expression of the at least one gene in the subject sample compared to the control sample.

BRIEF DESCRIPTION OF FIGURES



[0021] These and other features of the preferred embodiments of the invention will become more apparent in the following detailed description in which reference is made to the appended drawings wherein:

Figure 1 shows characterization of epirubicin-resistant cell lines. Native and resistant cells were exposed to drug concentrations ranging from 0.3nM to 3000nM. Cell viability was determined 72h later by CCK-8 assay. A) Percent of live cells relative to DMSO control was plotted against epirubicin concentration. Black = native cells, magenta = resistant cells. B) IC50 values in nM concentration ± standard deviation. Resistance factor is shown in parenthesis and represents resistant IC50/native IC50.

Figure 2 shows expression of conventional breast cancer biomarkers and select multidrug resistance genes. Cell lysates were prepared in RIPA buffer supplemented with Complete Mini protease inhibitor and PhosSTOP phosphatase inhibitor. 10-50µg of total protein was run on a 10% gel (MDR1), 4-20% precast gels (EGFR, ER, PgR, TOPOIIα) and Any kD precast gels (HER2, HER3), transferred onto PVDF membrane and developed using chemiluminescence substrate. Nat = native; Epi-R = epirubicin resistant.

Figure 3 shows resistant cell lines overcome epirubicin-induced G2/M arrest. (A-D) Cells were synchronized by a double-thymidine block and treated with DMSO or epirubicin at selection doses established for each resistant cell line: 25nM epirubicin to MDA-MB-231, 30nM epirubicin to MCF7, 15nM epirubicin to SKBR3 and 15nM epirubicin to ZR-75-1. Epirubicin concentration was increased to 100nM for MCF7 and SKBR3 cells since G2/M block was not observed at the lower doses of epirubicin. Cells were collected at 48h, stained by PI and analyzed by flow cytometry. Debris was gated out.

Figure 4 shows network-based analysis of epirubicin-resistant cell lines. A) Venn diagram of genes with significant changes in expression in breast cancer cell lines. B) Histone module identified from functional interaction network analysis. Coloured rings denote genes demonstrating consistent changes across all 4 lines. Red rings (darker) = upregulated genes, green rings (lighter) = downregulated genes, diamonds = linker genes. C) qRT-PCR performed on RNA isolated from native and epirubicin-resistant cell lines. Bar graphs indicate average quantitative means, while error bars represent SEM. p-values were calculated using unpaired t-test; ns = non-significant. D) Immunoblotting of total H2A and H2B histone proteins in native and epirubicin-resistant cell lines. GAPDH was used as a housekeeping control. E) Reactome pathways significantly enriched within the module shown in panel B.

Figure 5 shows histone gene knockdown is not sufficient to resensitize breast cancer cells to epirubicin. A total of 7 x 104 ZR75-1 EpiR cells and MDA-MB-231 EpiR cells were transfected with 30nM of each siRNAs (Dharmacon, Waltman, USA) targeting HIST1H2AC and HIST1H2BK (individual knockdowns not shown for simplicity). Negative controls included media only, lipofectamine only or mock transfection with non-targeting siRNA. Percent gene expression knockdown is shown in Table 4. B) IC50 values were generated using non-linear regression analysis and average values of two independent experiments were graphed. Error bars represent standard deviation.

Figure 6 shows histone module is a biological marker for anthracycline therapy. High expression and low expression of histone module were tested for association with distant recurrence free survival (DRFS) and overall survival (OS) in BR9601 trial in which patients were treated with standard chemotherapy (CMF) or anthracycline-containing chemotherapy (E-CMF). A) DRFS and OS for patients treated with E-CMF versus CMF split into high or low histone gene expression groups. B) Multivariate, treatment by marker analysis after adjustment for HER2 status, ER status, nodal status, grade and age. HR=hazard ratio, CI=confidence interval.

Figure 7 shows HDAC inhibitors induce cytotoxicity in epirubicin-resistant cells lines. A) Examples of inhibitors that were more cytotoxic for resistant-cell lines (pracinostat for MDA-MB-231, ST-2-92 for MCF7, oxamflatin for SKBR3) or had no selective differences between the native and epirubicin-resistant cells (ZR-75-1). IC50 values are shown in Table 5. B) Working models of molecular mechanisms involved in epirubicin resistance. There are three proposed mechanisms by which HDACi sensitize cells to epirubicin: 1) by transcriptional activation of repressors and pro-apoptotic genes, 2) by repression of resistance genes and 3) due to increased accessibility to DNA.

Figure 8 shows clinical trial BR9601 information. A) Schematic representation of the patient samples available for analysis. B) Patient information available for the histone analysis.

Figures 9A-9D together show the entire Functional Interaction network from 61 consistently changing genes. Red (darker) circles = upregulated genes ; green (lighter) circles = downregulated genes; diamonds = linker genes. Figures 9A-9D form one figure when arranged by quadrants as follows: upper left, lower left, upper right, lower right respectively.

Figure 10 shows heatmaps of probes for the 61 consistently changing genes in four breast cancer cell lines. Rows labeled with gene symbol and microarray probe IDs. A) Raw expression values. B) Row scaled expression values.

Figure 11 shows combination of pre-processing methods. The most optimal method selected was at the top, indicated by the black colour (high-rank).

Figure 12 shows sample by gene heatmap. Row represent patients and columns represent genes. Patients and genes are clustered using ward clustering algorithm.

Figure 13 shows functional Interaction network generated from the histone module. Circles = genes within the module, diamonds = linker genes.

Figure 14 shows multiplot showing scaled mRNA abundance levels for each histone gene. A treatment-by-marker interaction Cox proportional hazards model was fit for each gene and results were visualized on the right with the squares representing the hazard ratios (HR) and the ends of the segments representing the 95% confidence intervals in log2 scale. Patients were sorted by DRFS events on the x-axis and genes by decreasing log2 HR on the y-axis.


DETAILED DESCRIPTION



[0022] In the following description, numerous specific details are set forth to provide a thorough understanding of the invention. However, it is understood that the invention may be practiced without these specific details.

[0023] Drug resistance in breast cancer is the major obstacle to effective treatment with chemotherapy. While upregulation of multidrug resistance (MDR) genes is a key component of drug resistance in multiple cancers, the complexity and hierarchy of non-MDR driven drug resistance pathways are still largely unknown. The present study aimed to establish anthracycline-resistant breast cancer cell lines to elucidate mechanisms driving resistance, which could be tested in clinical trial cohorts. Cell lines were chosen to reflect four major breast cancer subtypes (Perou et al. 2000; Sorlie et al. 2001): MCF7 (ER+HER2-, luminal A), ZR-75-1 (ER+HER2+, luminal B), SKBR3 (ER-HER2+, HER2-amplified) and MDA-MB-231 (ER-/PR-/HER2-, triple negative), and exposed to increasing concentrations of epirubicin until resistant cells were generated. To identify mechanisms driving epirubicin resistance, the investigators used complementary approaches including gene expression analyses to identify signaling pathways involved in resistance, and small-molecule inhibitors to reverse resistance. Applicant demonstrated that overexpression of histones H2A and H2B were associated with epirubicin resistance and that small-molecule inhibitors targeting histone pathways reversed resistance and induced cytotoxicity in all epirubicin-resistant cell lines. Most importantly, the identified mechanism of resistance was recapitulated in the BR9601 clinical trial as the patients with low expression of the histone module benefited from anthracycline treatment compared to patients with high expression of the same module (HR: 0.35, 95%CI 0.13-0.96, p=0.042). Thus, our study has identified that chromatin remodeling represents an important mechanism of anthracycline resistance in breast cancer and established a reliable in vitro model system for investigating anthracycline resistance in all four breast cancer subtypes; as the histone modification can be targeted with small-molecule inhibitors, it presents a possible means of reversing clinical anthracycline resistance.

[0024] In one embodiment, there is provided a method for determining a likelihood of resistance to anthracycline in a patient with cancer comprising: detecting a level of expression in the sample of HIST1H2BK, HIST1H2BD, NEDD9, SYTL2, NHP2, ARPP19, TXNRD1, CENPF, STMN1, CCT5, APRT, UBEC2C, BAX, HDAC1, E2F1, E2F2, E2F4 and CDKN2A; comparing the level of the at least one gene detected to a level of expression of the at least one gene in a control sample; and wherein there is a likelihood of anthracycline resistance if there is a relatively higher level of expression of the at least one gene in the subject sample compared to the control sample.

[0025] In another embodiment, there is provided a method for prognosticating survival in cancer patient comprising: detecting a level of expression in the sample of HIST1H2BK, HIST1H2BD, NEDD9, SYTL2, NHP2, ARPP19, TXNRD1, CENPF, STMN1, CCT5, APRT, UBEC2C, BAX, HDAC1, E2F1, E2F2, E2F4 and CDKN2A; comparing the level of the genes detected to a level of expression of the genes in a control sample; and wherein there is a likelihood of poor survival if there is a relatively higher level of expression of the genes in the subject sample compared to the control sample.

[0026] Five major families of histones exist: H1/H5, H2A, H2B, H3 and H4.[2][4][5] Histones H2A, H2B, H3 and H4 are known as the core histones, while histones H1 and H5 are known as the linker histones.

[0027] The H1 family comprises the H1F subfamily comprising H1F0, H1FNT, H1FOO, and H1FX; and the H1H1 subfamily comprising HIST1H1A, IST1H1B, HIST1H1C, HIST1H1D, HIST1H1E and HIST1H1T.

[0028] The H2A family comprises the H2AF subfamily comprising H2AFB1 , H2AFB2, H2AFB3, H2AFJ, H2AFV, H2AFX, H2AFY, H2AFY2 and H2AFZ; the H2A1 subfamily comprising HIST1H2AA, HIST1H2AB, HIST1H2AC, HIST1H2AD, HIST1H2AE, HIST1H2AG, HIST1H2AI, HIST1H2AJ, HIST1H2AK, HIST1H2AL, and HIST1H2AM; the H2A2 subfamily comprising HIST2H2AA3, HIST2H2AC.

[0029] The H2B family comprises the H2BF subfamily comprising H2BFM, H2BFS, and H2BFWT; the H2B1 subfamily comprising HIST1H2BA, HIST1H2BB, HIST1H2BC, HIST1H2BD, HIST1H2BE, HIST1H2BF, HIST1H2BG, HIST1H2BH, HIST1H2BI, HIST1H2BJ, HIST1H2BK, HIST1H2BL, HIST1H2BM, HIST1H2BN, and HIST1H2BO; and the H2B2 subfamily comprising HIST2H2BE.

[0030] The H3 family comprises the H3A1 subfamily comprisingHIST1H3A, HIST1H3B, HIST1 H3C, HIST1 H3D, HIST1 H3E, HIST1H3F, HIST1 H3G, HIST1 H3H, HIST1 H3I, and HIST1 H3J; the H3A2 subfamily comprising HIST2H3C; and the H3A3 subfamily comprising HIST3H3.

[0031] The H4 family comprises the H41 subfamily comprising HIST1H4A, HIST1H4B, HIST1H4C, HIST1H4D, HIST1H4E, HIST1H4F, HIST1H4G, HIST1H4H, HIST1H4I, HIST1 H4J, HIST1 H4K, and HIST1 H4L; and the H44 subfamily comprising HIST4H4.

[0032] The aspects described herein may be practiced with any number of cancers. In some embodiments, the cancer is a multidrug resistant cancer. Cancers could include Adrenal Cancer, Anal Cancer, Bile Duct Cancer, Bladder Cancer, Bone Cancer, Brain/CNS Tumors, Breast Cancer, Castleman Disease, Cervical Cancer, Colon/Rectum Cancer, Endometrial Cancer, Esophagus Cancer, Ewing Family Of Tumors, Eye Cancer, Gallbladder Cancer, Gastrointestinal Carcinoid Tumors, Gastrointestinal Stromal Tumor (GIST), Gestational Trophoblastic Disease, Hodgkin Disease, Kaposi Sarcoma, Kidney Cancer, Laryngeal and Hypopharyngeal Cancer, Leukemia, Liver Cancer, Lung Cancer, Lung Carcinoid Tumor, Lymphoma, Malignant Mesothelioma, Multiple Myeloma, Myelodysplastic Syndrome, Nasal Cavity and Paranasal Sinus Cancer, Nasopharyngeal Cancer, Neuroblastoma, Non-Hodgkin Lymphoma, Oral Cavity and Oropharyngeal Cancer, Osteosarcoma, Ovarian Cancer, Pancreatic Cancer, Penile Cancer, Pituitary Tumors, Prostate Cancer, Retinoblastoma, Rhabdomyosarcoma, Salivary Gland Cancer, Sarcoma, Skin Cancer, Small Intestine Cancer, Stomach Cancer, Testicular Cancer, Thymus Cancer, Thyroid Cancer, Uterine Sarcoma, Vaginal Cancer, Vulvar Cancer, Waldenstrom Macroglobulinemia, and Wilms Tumor.

[0033] The term "level of expression" or "expression level" as used herein refers to a measurable level of expression of the products of biomarkers, such as, without limitation, the level of messenger RNA transcript expressed or of a specific exon or other portion of a transcript, the level of proteins or portions thereof expressed of the biomarkers, the number or presence of DNA polymorphisms of the biomarkers, the enzymatic or other activities of the biomarkers, and the level of specific metabolites.

[0034] In addition, a person skilled in the art will appreciate that a number of methods can be used to determine the amount of a protein product of the biomarker of the invention, including immunoassays such as Western blots, ELISA, and immunoprecipitation followed by SDS-PAGE and immunocytochemistry.

[0035] As used herein, the term "control" refers to a specific value or dataset that can be used to prognose or classify the value e.g. expression level or reference expression profile obtained from the test sample associated with an outcome class. A person skilled in the art will appreciate that the comparison between the expression of the biomarkers in the test sample and the expression of the biomarkers in the control will depend on the control used.

[0036] The term "differentially expressed" or "differential expression" as used herein refers to a difference in the level of expression of the biomarkers that can be assayed by measuring the level of expression of the products of the biomarkers, such as the difference in level of messenger RNA transcript or a portion thereof expressed or of proteins expressed of the biomarkers. In a preferred embodiment, the difference is statistically significant. The term "difference in the level of expression" refers to an increase or decrease in the measurable expression level of a given biomarker, for example as measured by the amount of messenger RNA transcript and/or the amount of protein in a sample as compared with the measurable expression level of a given biomarker in a control.

[0037] The term "sample" as used herein refers to any fluid, cell or tissue sample from a subject that can be assayed for biomarker expression products and/or a reference expression profile, e.g. genes differentially expressed in subjects.

[0038] Important histone genes may be from the H2A or H2B families, preferably selected from the group consisting of H2AFB1, H2AFB2, H2AFB3, H2AFJ, H2AFV, H2AFX, H2AFY, H2AFY2, H2AFZ, HIST1H2AA, HIST1H2AB, HIST1H2AC, HIST1H2AD, HIST1H2AE, HIST1H2AG, HIST1H2AI, HIST1H2AJ, HIST1H2AK, HIST1H2AL, HIST1H2AM, HIST2H2AA3, HIST2H2AC, H2BFM, H2BFS, H2BFWT, HIST1H2BA, HIST1H2BB, HIST1H2BC, HIST1H2BD, HIST1H2BE, HIST1H2BF, HIST1H2BG, HIST1H2BH, HIST1H2BI, HIST1H2BJ, HIST1H2BK, HIST1H2BL, HIST1H2BM, HIST1H2BN, HIST1H2BO, and HIST2H2BE; or combinations thereof.

[0039] The histone genes of the invention comprise all of the genes in Table 7.

[0040] The method may further comprise treating the patient with adjuvant therapy that does not comprise anthracycline if there is a relatively higher level of expression of the genes in the subject sample compared to the control sample.

[0041] The method may also further comprise administering to the patient anthracycline along with an inhibitor of at least one gene in the regulatory pathway of at least one histone gene, if there is a relatively higher level of expression of the at least one gene in the subject sample compared to the control sample.

[0042] In some examples, the inhibitor is a histone deacetylase inhibitor, preferably panobinostat, quisinostat, givinostat, abexinostat, pracinostat, belinostat mocetinostat, Apicidin A, CAY10603, Oxamflatin, Trichostatin A, Sciptaid, CBHA or Dacinostat.

[0043] In an embodiment, the breast cancer is early breast cancer, preferably selected from the following subtype: ER+HER2-, luminal A, ER+HER2+, luminal B, ER-HER2+, HER2-amplified and ER-/PR-/HER2-, triple negative.

[0044] In some embodiments, the anthracycline is Daunorubicin Doxorubicin, Epirubicin, Idarubicin, Valrubicin, or Mitoxantrone, preferably Epirubicin.

[0045] Also described is a use of a histone deacetylase inhibitor in the treatment of a cancer patient receiving anthracycline and exhibiting upregulation of at least one histone gene.

[0046] In another example is described a use of a histone deacetylase inhibitor in the treatment of a breast cancer patient receiving anthracycline.

[0047] In another example, there is provided a method of sensitizing, or re-sensitizing, a patient with breast cancer to anthracycline, comprising administering to the patient a histone deacetylase inhibitor.

[0048] In an example, there is provided a composition comprising a plurality of reagents, preferably nucleic acid sequences, wherein each of the reagents is for detecting a level of expression in the sample of a gene in the regulatory pathway of at least one histone gene from the H1, H2A, H2B, H3 and H4 gene families.

[0049] In another example, there is provided an array comprising, for a plurality of genes in the regulatory pathway of at least one histone gene from the H1, H2A, H2B, H3 and H4 gene families, one or more polynucleotide probes complementary and hybridizable to an expression product of the gene.

[0050] Also described is a kit for determining a likelihood of resistance to anthracycline in a patient, comprising detection agents for detecting a level of expression in the sample of a gene in the regulatory pathway of at least one histone gene from the H1, H2A, H2B, H3 and H4 gene families, and instructions for use.

[0051] In another example, there is provided a kit for prognosticating survival in cancer patient, comprising detection agents for detecting a level of expression in the sample of a gene in the regulatory pathway of at least one histone gene from the H1, H2A, H2B, H3 and H4 gene families, and instructions for use.

[0052] In an example, there is provided a computer program product for use in conjunction with a computer having a processor and a memory connected to the processor, the computer program product comprising a computer readable storage medium having a computer mechanism encoded thereon, wherein the computer program mechanism may be loaded into the memory of the computer and cause the computer to carry out the method of the invention.

[0053] In one embodiment, there is provided a computer implemented product for determining a likelihood of resistance to anthracycline in a patient comprising: a means for receiving values corresponding to a subject expression profile in a subject sample; a database comprising a control expression profile associated with HIST1H2BK, HIST1H2BD, NEDD9, SYTL2, NHP2, ARPP19, TXNRD1, CENPF, STMN1, CCT5, APRT, UBEC2C, BAX, HDAC1, E2F1, E2F2, E2F4 and CDKN2A; and processor disposed to compare the subject expression profile to the control expression profile and determine a likelihood of anthracycline resistance if there is a relatively higher level of expression of the genes in the subject sample compared to the control sample.

[0054] In another embodiment, there is provided a computer implemented product for prognosticating survival in cancer patient comprising: a means for receiving values corresponding to a subject expression profile in a subject sample; and a database comprising a control expression profile associated with HIST1H2BK, HIST1H2BD, NEDD9, SYTL2, NHP2, ARPP19, TXNRD1, CENPF, STMN1, CCT5, APRT, UBEC2C, BAX, HDAC1, E2F1, E2F2, E2F4 and CDKN2A; and a processor disposed to compare the subject expression profile to the control expression profile and determine there is a likelihood of poor survival if there is a relatively higher level of expression of the genes in the subject sample compared to the control sample.

[0055] As used herein, "pharmaceutically acceptable carrier" means any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like that are physiologically compatible. Examples of pharmaceutically acceptable carriers include one or more of water, saline, phosphate buffered saline, dextrose, glycerol, ethanol and the like, as well as combinations thereof. In many cases, it will be preferable to include isotonic agents, for example, sugars, polyalcohols such as mannitol, sorbitol, or sodium chloride in the composition. Pharmaceutically acceptable carriers may further comprise minor amounts of auxiliary substances such as wetting or emulsifying agents, preservatives or buffers, which enhance the shelf life or effectiveness of the pharmacological agent.

[0056] As used herein, "therapeutically effective amount' refers to an amount effective, at dosages and for a particular period of time necessary, to achieve the desired therapeutic result. A therapeutically effective amount of the pharmacological agent may vary according to factors such as the disease state, age, sex, and weight of the individual, and the ability of the pharmacological agent to elicit a desired response in the individual. A therapeutically effective amount is also one in which any toxic or detrimental effects of the pharmacological agent are outweighed by the therapeutically beneficial effects.

[0057] The advantages of the present invention are further illustrated by the following examples. The examples and their particular details set forth herein are presented for illustration only and should not be construed as a limitation on the claims of the present invention.

EXAMPLES


Methods and Materials


BR9601 trial



[0058] The BR9601 trial recruited 374 pre- and post-menopausal women with completely excised, histologically confirmed breast cancer and a clear indication for adjuvant chemotherapy. Patients were randomized between 8 cycles of CMF (i.v. cyclophosphamide 750 mg/m2, methotrexate 50 mg/m2 and 5-fluorouracil 600 mg/m2) every 21 days, and E-CMF (4 cycles of epirubicin 100 mg/m2 every 21 days followed by 4 cycles of the same CMF regimen) (Poole et al. 2006)(Figure 8). The protocol was approved by central and local ethics committees, and each patient provided written informed consent prior to randomization. For the current analysis, tissue blocks were retrieved and RNA was extracted. The primary outcomes of the BR9601 study were RFS and OS, although distant relapse-free survival was also reported (Poole et al. 2006).

Cell culture



[0059] Breast cancer cell lines (MDA-MB-231, MCF7, ZR-75-1, SKBR3) were purchased from ATCC and cultured in DMEM (except SKBR3, cultured in RPMI) supplemented with 10% heat-inactivated fetal bovine serum and 1% L-glutamine (Gibco, Burlington, Canada). Epirubicin-resistant cell lines were generated by exposing native cells to increasing concentrations of epirubicin with an initial concentration set at 0.5nM. Resistance was defined when IC50 value superseded the IC50 value of the corresponding native cell line, and resistant cells could not tolerate further increase in drug concentration. Drug resistance and cross resistance were determined by exposing cells to drug concentrations ranging from 0.3-3000nM for 72h. Cell viability was determined by Cell Counting Kit-8 (CCK-8, Dojindo, Cedarlane, Burlington, Canada). IC50 were calculated in GraphPad Prism5.

Flow cytometry



[0060] For cell cycle, cells were synchronized by the double-thymidine block (Whitfield et al. 2000) and incubated with DMSO or epirubicin doses established for each cell line: 25nM for MDA-MB-231, 30nM for MCF7, 15nM for SKBR3, 10nM for ZR-75-1. Cells were collected at 48h, fixed with 80% ethanol and incubated with 2mg/ml RNase A and 0.1mg/ml propidium iodide (both from Sigma, Oakville, Canada) prior to analysis. For apoptosis experiments, cells were treated with DMSO or epirubicin at concentrations described above, and collected at 72h for staining with Annexin V apoptosis-detection eFluor450 (eBioscience, San Diego, USA). Data were collected by FACSCanto II and FACSDiva (BD Biosciences, Mississauga, Canada) and analyzed by FlowJo (Treestar, Ashland, USA).

Proliferation



[0061] Cells were cultured in the presence or absence of epirubicin for up to 96h (see Flow Cytometry for epirubicin concentrations). Cells were collected at 24, 48, 72 and 96 hours and counted by ViCell (Beckman Coulter, Mississauga, Canada). Data were analyzed in GraphPad Prism5 software.

Microarray



[0062] Illumina Human HT-12-V4 Bead Chips were used for the whole genome microarray analysis by the UHN Microarray Centre, Toronto, Canada. Total RNA was extracted with the RNeasy Mini kit (Qiagen, Toronto, Canada) and used for profiling gene expression changes. Raw data were normalized with the R3.0.0 lumi package using Simple Scaling Normalization; the 10% most variable probes were retained for differential analysis using the genefilter package. Differentially expressed probes were identified using limma with a Benjamini-Hochberg corrected P-value cutoff of 0.05.

Network-based analysis



[0063] To identify functionally relevant modules, genes demonstrating consistent directionality of significant expression changes were analyzed using the Cytoscape Reactome Functional Interaction (FI) plugin in Cytoscape 2.8.3. Symbols were loaded as a gene set and interactions from the FI network 2012 version, including FI annotations and linker genes. Network modules were identified using spectral clustering and Pathway Enrichment computed for each module using the Reactome FI plugin functions. Reactome pathways exhibiting FDR values<0.01 were considered enriched.

Pharmaceutical inhibitors



[0064] All inhibitors were provided by the Drug Discovery group at the Ontario Institute for Cancer Research (OICR, Toronto, Canada). Cells were seeded at 1000-1500 cells/well into 384-well plates (Greiner, Mississauga, Canada). After 24h, resistant cells were exposed to epirubicin at the selection doses established (see Flow Cytometry), then exposed to HDACi dissolved in DMSO in 12 concentrations ranging from 0.0026-10µM using D300 digital compound dispenser (HP/Tecan, San Jose, USA); DMSO concentration did not exceed 0.5% in the final drug solution. After 72h, the effects of inhibitors were determined using CellTiter-Glo Luminescent Cell Viability Assay (Promega, Madison, USA) and the Wallac EnVision 2104 Multilabel Reader (Perkin-Elmer, Woodbridge, Canada). Raw data were normalized to negative (media) and positive (20µM staurosporine) controls and analyzed in GraphPad Prism5.

Quantitative RT-PCR



[0065] RNA was isolated from cultured cell lines using RNeasy Mini Kit (Qiagen, Toronto, Canada). A total of 20ng of RNA was analysed using TaqMan Gene Expression Assays (HIST1H2BD - Hs00371070_m1; HIST1H2BK - Hs00955067_g1; HIST1H2AC - Hs00185909_m1) and EXPRESS One-Step Superscript qRT-PCR universal kit according to manufacturer's protocol (Life Technologies, Burlington, Canada). Reactions were run using Applied Biosystems Viia 7 real-time PCR instrument and software (Life Technologies, Burlington, Canada); transcript levels were quantified from the standard curve generated from the control, Universal Human Reference RNA samples (Agilent, Mississauga, Canada). Statistical significance was determined using unpaired t-test.

Immunoblotting



[0066] Whole cell lysates (WCL) were prepared in RIPA buffer supplemented with Complete Mini protease and PhosSTOP phosphatase inhibitors (Roche, Laval, Canada). For cell line characterization, 10-50µg of total protein was run on 4-20% Mini-Protean TGX precast gels (Bio-Rad, Mississauga, Canada). For histones, cells were collected in 0.1 % NP40-PBS to release nuclei. WCL were prepared by adding equal volume of 2x RIPA buffer, supplemented with 25 units of benzonase nuclease (Sigma-Aldrich, Oakville, Canada) and Complete Mini protease inhibitor cocktail (Roche, Laval, Canada), incubating on ice for 30 minutes and sonicating for 15 minutes with 30-second on-off intervals. Twenty µg of WCL were run on a 12% gel. A list of primary antibodies used in immunoblotting is provided in Table 6. Signals were developed with the BM Chemiluminescence Blotting Substrate POD (Roche, Laval, Canada) and ChemiDoc Imaging System (Bio-Rad, Mississauga, Canada).

RNAi transfection of ZR75-1 and MDA-MB-231 resistant cells



[0067] A total of 7 x 104 ZR75-1 EpiR cells and MDA-MB-231 EpiR cells were transfected with Lipofectamine RNAiMAX (Invitrogen, Canada) and 30nM siRNAs (Dharmacon, Waltman, USA) targeting HIST1H2AC, HIST1H2BK, or both according to manufacturer's instructions. Negative controls included media only, lipofectamine only, or mock transfection with non-targeting siRNA. RNA was collected at 48h and analyzed by qRT-PCR as described above; IC50 values were generated in GraphPad Prism5.

nCounter codeset and data pre-processing



[0068] nCounter gene expression codeset included 7 genes within the histone module and 11 additional genes that were identified in Kegg pathways (Kanehisa et al. 2014) as being important for histone function (Table 7); HIST1H2AC was excluded from the codeset since probes cross-hybridized to other genes. All 18 genes were functionally related (Figure 13). mRNA codesets were processed on nCounter according to manufacturer's instructions (NanoString Technologies, Seattle, USA). Raw mRNA abundance data were pre-processed using the NanoStringNorm R package. A range of pre-processing schemes was assessed to optimize normalization parameters as previously described (Sabine et al., submitted).

Survival modelling



[0069] To assess whether individual genes are prognostic of survival, each gene was median dichotomized into low- and high-expression groups and univariate Cox proportional hazards models were fit (Figure 14). Survival analysis of clinical variables modelled age as binary variable (dichotomized at age >50), while nodal status, pathological grade, ER status and HER2 status were modelled as ordinal variables (Figure 8B). Tumor size was treated as a continuous variable.

mRNA network analysis



[0070] The investigators hypothesized that integrating molecular modules could improve residual risk prediction relative to DRFS) and OS. For each module the investigators calculated a 'module-dysregulation score' (MDS; Methods), which were used in a univariate Cox proportional hazards model. A stratified 5-fold cross validation approach was applied; models were trained in the training cohort and validated in the k-th testing cohort using 10-year DRFS as an end-point. All survival modelling was performed on DRFS and OS, in the R statistical environment with the survival package (v2.37-7). Treatment by marker interaction term was calculated using Cox proportional hazards model.

mRNA Abundance Data Processing



[0071] Raw mRNA abundance counts data were preprocessed using R package NanoStringNorm (v1.1.19). In total, 252 preprocessing schemes were assessed, including the use of six positive controls, eight negative controls and six housekeeping genes (TRFC, TBP, GUSB, TMED10, SF3A1, and PUM1) followed by global normalization (Figure 11). The investigators used two criteria to help identify the optimal preprocessing parameters as previously described (Sabine et al., submitted). First, each of the 252 combinations of preprocessing schemes was ranked based on their ability to maximize Euclidean distance of ERBB2 mRNA abundance levels between HER2-positive and HER2-negative patients. For robustness, the entire process was repeated for 1 million random subsets of HER2-positive and HER2-negative samples for each of the preprocessing schemes. Second, the investigators included 5 replicates of an RNA pool extracted from randomly selected anonymized FFPE breast tumour samples; the rationale here was to assess each of the different preprocessing schemes for their inter-batch variation and rank them as previously described (Sabine et al. submitted). For this evaluation, a mixed effects linear model was used and residual estimate was used as a metric for inter-batch variation (R package: nlme v3.1-120). Lastly, the investigators estimated the cumulative ranks using RankProduct (Breitling et al. 2004) based on the two criteria and identified the optimal pre-processing scheme as using geometric mean derived from the top 75 expressing genes for sample content followed by quantile normalisation (Figure 12). No samples were removed after QAQC. Six samples were run in duplicates, and their raw counts were averaged and subsequently treated as a single sample.

Module Dysregulation Score (MDS)



[0072] As previously described (Sabine et al. submitted, Haider et al., submitted), predefined functional modules were scored using a two-step process. First, weights (β) of all the genes were estimated by fitting a multivariate Cox proportional hazards model and were obtained from the treatment by marker interaction term (Training cohort only). Second, these weights were applied to scaled mRNA abundance profiles to estimate per-patient module dysregulation score using the following equation 1:


(1)
where n represents the number of genes in a given module and Xi is the scaled (z-score) abundance of gene i. MDS was subsequently used in the multivariate Cox proportional hazards model alongside clinical covariates.

Survival Modelling



[0073] Using a stratified 5-fold cross validation approach, MDS profiles (equation 1) of patients within each training set were used to fit a univariate Cox proportional hazards model. The parameters estimated by the univariate model were applied to patient-wise MDS in the testing set of each fold to generate per-patient risk scores. These continuous risk scores were dichotomized based on the median threshold derived from each training set, and the resulting dichotomized groups were evaluated through Kaplan-Meier analysis. Models were trained and validated using DRFS truncated to 10 years as an end-point.

Results and Discussion


Generation and characterization of epirubicin-resistant breast cancer cell lines



[0074] Resistant cell lines generated from epirubicin-sensitive native cell lines MDA-MB-231, MCF7, SKBR3 and ZR-75-1, exhibited 7- to 67-fold increased resistance to epirubicin (Figure 1). The investigators tested whether epirubicin-resistant cell lines are cross-resistant to doxorubicin, paclitaxel, docetaxel, SN-38 and carboplatin, drugs used in breast cancer clinical trials. All four epirubicin-resistant cell lines were resistant to doxorubicin (Figure 1B). While MDA-MB-231, MCF7 and ZR-75-1 epirubicin-resistant cells were not taxane-resistant, SKBR3 epirubicin-resistant cells were cross-resistant to both, paclitaxel and docetaxel (Figure 1B). MDA-MB-231 and SKBR3 cells were cross-resistant to SN-38, whereas MCF7 and ZR-75-1 tolerated only small increases in SN-38 concentrations. None of the cell lines were cross-resistant to carboplatin (Figure 1B).

[0075] Epirubicin-resistant cells showed no marked alterations in EGFR, HER2 and HER3 expression levels (Figure 2); ER and PR expression decreased slightly in epirubicin-resistant ZR-75-1 cells compared to native cells. MDR1 was only upregulated in resistant SKBR3 cells, which may explain their cross resistance to taxanes (Figure 1B). TOPOIIα expression was downregulated in epirubicin-resistant ZR-75-1 cells (Figure 2); no changes in MDR or TOPOIIα were observed in epirubicin-resistant MDA-MB-231 and MCF7 cell lines. These results suggest that anthracycline resistance is not MDR-driven for three of four cell lines and that epirubicin-resistant cell lines remained unaltered with respect to the expression of conventional breast cancer biomarkers.

[0076] To determine cell-doubling time, the investigators cultured cells with or without epirubicin for up to 96h. In the absence of epirubicin, the native MDA-MB-231 and MCF7 cell populations doubled every 25h and 29h, respectively (Table 2), whereas native SKBR3 and ZR-75-1 cells grew more slowly, doubling every 45h and 50h, respectively. In the presence of epirubicin, doubling time increased 2.8-fold for the MDA-MB-231 (p=0.0371), 2.5-fold for MCF7 (ns), 1.3-fold for SKBR3 (p=0.0494) and 1.9-fold for ZR-75-1 (p=0.0258) for native cells. In contrast to the native cell lines, there were no marked changes in the doubling time of the resistant cells, regardless of whether epirubicin was added (Table 2). Interestingly, in the absence of epirubicin, none of the resistant cells proliferated as rapidly as native cells indicating that epirubicin selection induced permanent changes in resistant cells.

Impaired apoptosis in anthracycline-resistant cells



[0077] To assess the effects of epirubicin on apoptosis, apoptotic cells were scored by flow cytometry after 72h of exposure to epirubicin. The apoptotic index was consistently lower for resistant cells compared to native controls (Table 1). In particular, MDA-MB-231 and SKBR3 resistant cells required a substantially higher concentration of epirubicin (1000nM) to induce apoptosis; even at this concentration of epirubicin, the apoptotic index was still nearly 50% lower compared to the native cells (Table 1).

Resistant cell lines overcome epirubicin-induced G2/M arrest



[0078] Cells were synchronized prior to exposure to DMSO or epirubicin. All DMSO-treated cell lines progressed through the cell cycle (Figure 3). When 25nM and 10nM epirubicin were added to the MDA-MB-231 and ZR-75-1 cell lines respectively, native cells arrested in G2/M phase whereas resistant cells progressed through (Figure 3A, C). When 30nM and 15nM epirubicin were added to the MCF7 and SKBR3 cell lines respectively, the investigators observed only a modest effect on the cell cycle (Figure 3B, D); this necessitated increasing epirubicin concentrations to 100nM at which native cells arrested in G2/M phase, but with minimal effect on the epirubicin-resistant cells (Figure 3B, D). Therefore, overcoming a G2/M block may be part of the process leading to epirubicin resistance.

Gene expression analyses identify histone H2A and H2B containing pathways as potential functional drivers of epirubicin resistance



[0079] Whole genome expression analysis revealed 209 genes in common, differentially expressed between all four pairs of native and epirubicin-resistant cell lines (Figure 4A). Of these, 61 genes were regulated in the same direction in all four cell lines: 26 genes were consistently upregulated and 35 were consistently downregulated (Table 3, Figure 4). These 61 genes were used to generate a gene interaction network and identify candidate pathways involved in epirubicin resistance. A minimal set of linker genes was used to connect the network. Identifying clustered genes within the network revealed four modules (Figure 9); however, only modules I and II contained significantly enriched pathway annotations with a False Discovery Rate (FDR) <0.01. Module I contained three histone genes (HIST1 H2AC, HIST1H2BK, HIST1H2BD) and several genes involved in RNA processing and mitosis (Figure 4B). Importantly, all three histone genes were upregulated in all four cell lines and directly interconnected without linker genes. Within module I, significantly enriched pathways included cell-cycle regulation (Figure 4E), consistent with our results in Figure 3. Module II contained three directly connected genes (TACC3, AURKA, NFKBIA) involved in Aurora A kinase signaling; while NFKBIA was upregulated, TACC3 and AURKA were downregulated.

[0080] The investigators focused on the histone-containing module 1 since all three histones were upregulated, tightly interconnected without linker genes and implicated in several molecular pathways. Elevated levels of all three histone transcripts were validated by qRT-PCR (Figure 4C). Since antibodies specific to individual histone variants are not commercially available, the investigators assessed protein expression using pan H2A and H2B antibodies; the investigators observed no difference in the total H2A and H2B levels between resistant and native cell lines (Figure 4D). Overall, our findings suggest that histone upregulation is a common event associated with epirubicin resistance in breast cancer cells and that histone-related pathways might be functional drivers of epirubicin resistance.

Histone gene knockdown is not sufficient to resensitize breast cancer cells to epirubicin



[0081] The investigators performed a series of gene knockdown experiments in MDA-MB-231 and ZR-75-1 resistant cells in which HIST1H2AC, HIST1H2BK, or both were silenced prior to exposing cells to epirubicin. HIST1 H2BK, rather than HIST1 H2BD, was selected because high transcript levels of this variant were associated with poor survival of breast cancer patients in our in silico analysis (data not shown; for online tool see reference (Gyorffy et al. 2010)). Following gene knockdown, a proliferation assay was performed to assess whether resistant cells were resensitized to epirubicin. A decrease in histone transcripts was confirmed by qRT-PCR and summarized in Table 4. Interestingly, transient knockdown of either histone alone, or both, did not re-sensitise cell lines to epirubicin (Figure 5 and data not shown). The results suggest that downregulation of one or two histone genes is insufficient to reverse epirubicin resistance and that future approaches may have to target multiple molecules within the histone module.

Histone module is a clinical marker of anthracycline sensitivity



[0082] The prognostic significance of the 18-gene histone module was tested on the entire BR9601 clinical cohort, irrespective of allocated adjuvant chemotherapy. High histone module expression was associated with reduced distant relapse free survival (DRFS; HR: 2.64, 95%CI 1.7- 4.09, p=1.44 x 10-5), indicating that elevated histone module is prognostic for poor survival.

[0083] Next, the investigators analysed the differential effects of the histone module on breast cancer-specific overall survival (OS) and DRFS between patients in the BR9601 trial receiving an anthracycline (E-CMF) and those given CMF alone by assessing hazard ratios and treatment by marker interactions. Patients whose tumours had low gene expression had an increased OS (HR: 0.38, 95%CI 0.19-0.76, p=0.005) when treated with E-CMF compared with patients treated with CMF alone; conversely, there was no apparent differential benefit of E-CMF vs CMF in patients with high histone module expression for OS (HR: 0.97, 95%CI 0.57-1.64, p=0.91) (Figure 6A). Similarly, patients whose tumour had low histone module expression had an increased DRFS (HR: 0.35, 95%Cl 0.17-0.73, p=0.0048) when treated with E-CMF compared with patients treated with CMF alone (Figure 6A); there was no apparent differential benefit of E-CMF vs CMF in patients with high histone module expression for DRFS (HR: 0.96, 95%Cl 0.58-1.59, p=0.87). In a multivariate analysis, after adjustment for HER2 status, nodal status, age, grade and ER status, treatment by marker interaction showed no statistical difference for OS (HR:0.50, 95% Cl 0.19-1.31, p=0.159); the likelihood of DRFS remained, however, low among patients with low histone module gene expression than in patients with high expression (HR:0.35, 95%CI 0.13-0.96, p=0.042) (Figure 6B).

HDAC inhibitors induce cytotoxicity in epirubicin-resistant cells lines



[0084] Gene expression analysis identified the histone module as significantly altered and possibly functionally required for epirubicin resistance. Consequently, the investigators tested whether alteration of histone activity may sensitize cells to epirubicin using histone deacetylase (HDAC) inhibitors, which reverse histone hypo-acetylation and permit transcriptional activation. Twenty four HDAC inhibitors (HDACi) were tested against the native and epirubicin-resistant cell lines; for resistant cell lines, all inhibitors were tested in the presence of selection doses of epirubicin. Positive hits were defined as compounds that exhibited cytotoxicity in at least 50% of population and had an IC50<5µM in all eight cell lines. As a result, 14 HDACi were cytotoxic to all native and epirubicin-resistant cells lines (Table 5). Importantly, three of four resistant cell lines were more sensitive to epirubicin than native cells when several HDACi were supplied. For instance, pracinostat was more cytotoxic for MDA-MB-231, ST-2-92 for MCF7 and oxamflatin for SKBR3 epirubicin-resistant cells compared to native cell lines (Figure 7A); no differences were observed between native and epirubicin-resistant ZR-75-1 cell for any cytotoxic HDACi tested (Figure 7A). Since inhibitors target different HDAC's and none of the inhibitors ubiquitously resensitized all four resistant cell lines (Table 5), it appears that different classes of HDAC's are involved in anthracycline resistance, possibly in breast cancer-subtype specific manner. Collectively, our data reveal a previously unrecognized role of histones and suggests that H2A and H2B histones are involved in clinical anthracycline resistance.

[0085] Anthracycline resistance represents a major obstacle to the effective treatment of women with breast cancer. Although various mechanisms may contribute to anthracycline resistance, including activation of drug transporters, reduced activity of TOPOIIα and inhibition of apoptosis, the majority of the molecular mechanisms involved in clinical drug resistance remain unknown. Using a panel of four paired cell lines representative of the major molecular subtypes of breast cancer the investigators have shown that deregulation of histones involved in chromosome maintenance, epigenetic pathways, cell division and gene regulation are observed consistently in epirubicin resistant cell lines. This observation was then validated clinically in the BR9601 adjuvant clinical trial cohort.

[0086] The dysregulation of histones is associated to increased cell cycle progression, specifically the release of a G2/M cell cycle block in the presence of epirubicin, and a reduction in apoptotic cell death. Interestingly, transcriptional knockdown of the two histone variants contributing to the dysregulation signature failed to resensitize cells to anthracycline, possibly due to two reasons. First, although the transcript levels were reduced by 6-53%, it is possible that the protein levels remained unchanged during our experimental window. Second, even if the protein levels were sufficiently diminished, it is still possible that other histone variants functionally substituted for the HIST1H2AC and HIST1H2BK since there are nine H2A and eleven H2B non-allelic histone variants (Bonenfant et al. 2006). Importantly, using small-molecule inhibitor screen the investigators have shown that drugs directly targeting HDAC function do reverse epirubicin resistance.

[0087] Epirubicin-resistant cell lines were generated by exposing native, non-resistant cell lines to increasing concentrations of epirubicin. Interestingly, only a single cell line, SKBR3, upregulated drug transporters and this was associated with cross resistance to taxanes. Previously, Hembruff et al. (Hembruff et al. 2008) developed epirubicin-resistant MCF-7 cells and established that a specific selection dose must be surpassed in order to activate drug transporters; for MCF-7, this critical threshold concentration was around 30nM (19). Although this concentration is identical to the selection dose of our resistant MCF-7 cells, MDR was not upregulated, suggesting a stochastic nature of molecular events that take place en route to drug resistance. Importantly, it indicates that there exist previously unappreciated MDR-independent mechanisms of resistance that should be evaluated for clinical relevance.

[0088] Our study revealed that one of those mechanisms involves upregulation of H2A and H2B genes and several pathways, including epigenetic and cell cycle pathways. H2A and H2B histones form octamers with H3 and H4 histones, which participate in packaging of DNA into nucleosomes (Wyrick and Parra 2009). These histones are replication-dependent and cell-cycle regulated, increasing 35-fold in S-phase during DNA replication (Harris et al. 1991). Thus, elevated histone transcript levels may be a consequence of a stalled cell cycle as cells struggle to repair epirubicin-induced DNA damage. However, since resistant cells did not stall, the investigators eliminated the possibility that upregulated histone transcripts were a mere reflection of accumulated mRNA.

[0089] An alternative explanation, supported by the ability of HDACi to sensitize resistant cells to epirubicin, is that upregulation of histones contributed to 1) the activation of resistance pathways, 2) the silencing of molecular pathways that sensitize cells to anthracyclines, and/or 3) a decreased accessibility of epirubicin to DNA. H3 and H4 histones modification patterns strongly associate with either active or repressed gene transcriptional status. Current understanding of H2A and H2B histone modifications is based on studies in yeast and few tumour cell lines; nonetheless, two important features of H2A and H2B histone modifications have been revealed. First, modified sites are acetylated, phosphorylated and ubiquitinated, but not methylated (Parra and Wyrick 2007; Parra et al. 2006; Beck et al. 2006), a modification most commonly observed with H3 and H4 histones. This highlights the appropriate use of HDACi in our study and their potency due to numerous acetylation sites, although this does not eliminate the possibility that the inhibitors were acting on H3 and H4 histones as well. Since acetylated sites on H2A and H2B are associated with transcriptional activation (Parra and Wyrick 2007; Parra et al. 2006), modifying the acetylation pattern may have activated transcriptional repressors and pro-apoptotic genes outlined in our model (Figure 7B, point 1). Second, the N-terminal end of H2A and H2B histones possesses a repression domain that inactivates gene transcription in approximately 10% of the yeast genome (Parra and Wyrick 2007; Parra et al. 2006), suggesting that these domains could have collaborated with acetylation patterns induced by HDACi to repress genes involved in resistance, such as those involved in cell cycle or apoptosis (Figure 7B, point 2). Lastly, our model also recognizes that resistance might have been reversed by an increased accessibility of epirubicin to DNA (Figure 7B, point 3).

[0090] Regel et al. (Regel et al. 2012) showed that HDACi panobinostat sensitizes gastric cancer cells to anthracyclines. Our findings are consistent with their study and show that multiple HDACi reverse anthracycline resistance in breast cancer cells. This is an important finding since many of the pharmacological inhibitors tested in our study are in use either as single-agents or as combination therapies in phase II/III clinical trials (Groselj et al. 2013; Wagner et al. 2010; Lee et al. 2012); HDAC inhibitors currently in clinical trials include panobinostat, quisinostat, givinostat, abexinostat, pracinostat, belinostat and mocetinostat (Table 5). Since anthracycline resistance may lead to cross-resistance to taxanes (Guo et al. 2004; Gosland et al. 1996) as it did in one of our resistant cell lines, it may be that taxanes, not anthracyclines, should be used in a first-line treatment (Paridaens et al. 2000). Furthermore, as cancer cells could acquire resistance to HDACi (Lee et al. 2012), sequential therapy involving HDACi, taxanes and anthracyclines will be an important aspect of clinical trial design and medical practice.

[0091] The investigators have identified novel pathways containing histone H2A and H2B genes as a mechanism of drug resistance across a spectrum of breast cancer cell lines and validated this finding in the BR9601 adjuvant clinical trial cohort. Furthermore, the investigators have developed a relevant model for studying clinical resistance as low histone expression correlated with better patient outcome. The model system opens avenues to its use for developing and testing novel single or combination, breast cancer therapies

[0092] In summary, the investigators generated paired native and epirubicin-resistant MDA-MB-231, MCF7, SKBR3 and ZR-75-1 epirubicin-resistant breast cancer cell lines to identify pathways contributing to anthracycline resistance. Native cell lines were exposed to increasing concentrations of epirubicin until resistant cells were generated; characterization of these cells revealed that they were cross-resistant to doxorubicin and SN-38, and had alterations in apoptosis and cell cycle profiles. To identify mechanisms driving epirubicin resistance, the investigators used a complementary approach including gene expression analyses to identify molecular pathways involved in resistance, and small-molecule inhibitors to reverse resistance. Gene expression analysis identified deregulation of histone H2A and H2B genes in all four cell lines. Histone deacetylase small-molecule inhibitors reversed resistance and were cytotoxic for epirubicin-resistant cell lines confirming that histone pathways are associated with epirubicin resistance. Gene expression analysis of the BR9601 adjuvant clinical trial revealed that patients with low expression of the histone module benefited from anthracycline treatment more than those with high expression (HR: 0.35, 95%CI 0.13-0.96, p=0.042). The present study has revealed a key pathway that contributes to anthracycline resistance and established model systems for investigating drug resistance in all four major breast cancer subtypes. As this process can be targeted with small-molecule inhibitors, it presents a possible means of reversing clinical anthracycline resistance.
Table 1: Percentages of apoptotic* cells following a 72h epirubicin treatment
MDA-MB-231DMSO1nM25nM1000nM
Native 18 17 41 94
25nM-R 10 10 8 50
MCF7DMSO1nM30nM1000nM
Native 32 29 49 77
30nM-R 20 24 23 78
SKBR3DMSO1nM15nM1000nM
Native 22 26 24 59
15nM-R 18 17 17 34
ZR-75-1DMSO1nM10nM1000nM
Native 36 44 47 71
10nM-R 29 28 29 62
*Apoptotic cells = Annexin V+. Debris and necrotic cells (Annexin V-, 7-AAD+) were gated out. Percentages reported here are from a single experiment; at least two independent experiments were done for each cell line.
Table 2 Doubling times (hours) of breast cancer cell lines
MDA-MB-231- epirubicin+ 25nM epirubicin
Native 25(1.2) 70(17.8)
25nM-Resistant 40 (4.2) 43 (3.0)
MCF7- epirubicin+ 30nM epirubicin
Native 29 (1.9) 74 (17.2)
30nM-Resistant 43(4.1) 37 (4.7)
SKBR3- epirubicin+ 15nM epirubicin
Native 45 (3.2) 57 (6.6)
15nM-Resistant 63 (2.0) 66 (9.2)*
ZR-75-1- epirubicin+ 10nM epirubicin
Native 50 (8.1) 95 (14.2)
10nM-Resistant 72 (15.9) 67 (4.0)
Data is based on three independent experiments and shows standard deviation in parentheses. * Indicates data based on two experiments.




Table 4 : Percent reduction in gene expression compared to non-targeting siRNA control
 MDA-MB-231 Epi-RZR-75-1 Epi-R
 H2AC expressionH2BK expressionH2AC expressionH2BK expression
siH2AC 24.4 (±3.2) - 27.5 (±0.16) -
siH2BK - 12.2 (±2.5) - 5.7 (±1.44)
siH2BA and siH2BK 40.7 (±10.9) 12.2 (13.7) 52.8 (±0.99) 7.9 (±2.25)
Table 5 Drugs targeting epirubicin-resistant breast cancer cells
Drug statusDrug nameIC50 values (µM)
  MCF7 NatMCF7 EpiR231 Nat231 EpiRSKBR3 NatSKBR3 EpiRZR75 NatZR75 EpiR
Phase III Panobinostat (LBH-589) 0.01 0.01 0.02 0.01 0.02 0.07 0.01 0.02
Phase II Quisinostat (JNJ-26481585) 0.01 0.01 0.01 0.01 0.01 0.22 0.01 0.01
Phase II Givinostat (ITF2357) 0.10 0.08 0.26 0.16 0.22 2.74 0.17 0.18
Phase II Abexinostat (PCI-24781) 0.11 0.09 0.27 0.12 0.21 2.25 0.14 0.16
Phase II Pracinostat (SB939) 0.16 0.12 0.54 0.18 0.26 0.92 0.15 0.23
Phase II Belinostat (PX-105684) 0.25 0.20 0.50 0.18 0.21 0.15 0.36 0.46
Phase II Mocetinostat (MGCD0103) 0.32 0.41 0.85 0.43 1.00 3.69 0.35 0.43
Preclinical Apicidin A (OSI-2040) 0.07 0.11 0.23 0.11 0.17 2.21 0.21 0.25
Preclinical CAY10603 (ST-2-92) 0.61 0.38 1.27 0.82 0.44 1.03 0.98 0.75
Preclinical Oxamflatin (107-0130) 0.62 0.25 0.59 0.29 1.28 0.69 0.68 1.20
Preclinical Trichostatin A 1.18 0.50 0.33 0.15 1.52 1.24 1.83 2.28
Preclinical Scriptaid 1.34 0.72 3.81 1.30 1.25 0.94 1.66 1.23
Tool compound CBHA 1.18 3.58 2.39 1.75 1.45 1.03 2.94 2.25
Discontinued - Phase I Dacinostat (LAQ824) 0.02 0.01 0.04 0.02 0.02 0.06 0.02 0.02
Table 6 : List of primary antibodies
AntibodyVendorClone
anti-EGFR Santa Cruz Biotech A-10
anti-PR Dako PgR 636
anti-HER2 Cell Signaling Technology Polyclonal (#2242)
anti-HER3 Dako DAK-H3-IC
anti-ERα Novocastra/Leica ER 6F11
anti-MDR1 Santa Cruz Biotech G-1
anti-TOPOIIα Cell Signaling Technology D10G9
anti-H2A Cell Signaling Technology Polyclonal (#2578)
anti-H2B Cell Signaling Technology 53H3
anti-actin Calbiochem JLA20
anti-GAPDH Cell Signaling Technology D16H11

Reference List



[0093] 

Bartlett JM, McConkey CC, Munro AF, Desmedt C, Dunn JA, Larsimont DP, O'Malley FP, Cameron DA, Earl HM, Poole CJ, Shepherd LE, Cardoso F, Jensen MB, Caldas C, Twelves CJ, Rea DW, Ejlertsen B, Di LA and Pritchard Kl. (2015). J Clin Oncol. 33(15),1680-7

Beck HC, Nielsen EC, Matthiesen R, Jensen LH, Sehested M, Finn P, Grauslund M, Hansen AM and Jensen ON. (2006). Mol Cell Proteomics, 5, 1314-1325.

Bonenfant D, Coulot M, Towbin H, Schindler P and van OJ. (2006). Mol Cell Proteomics, 5, 541-552.

Chazard M, Pellae-Cosset B, Garet F, Soares JA, Lucidi B, Lavail Y and Lenaz L. (1994). Bull Cancer, 81, 173-181.

de Jong S, Zijlstra JG, de Vries EG and Mulder NH. (1990). Cancer Res, 50, 304-309.

Early Breast Cancer Trialists' Collaborative Group (EBCTCG). (2005). Lancet, 365, 1687-1717.

Friesen C, Fulda S and Debatin KM. (1997). Leukemia, 11, 1833-1841.

Giaccone G, Gazdar AF, Beck H, Zunino F and Capranico G. (1992). Cancer Res, 52, 1666-1674.

Gosland MP, Gillespie MN, Tsuboi CP, Tofiq S, Olson JW, Crooks PA and Aziz SM. (1996). Cancer Chemother Pharmacol, 37, 593-600.

Groselj B, Sharma NL, Hamdy FC, Kerr M and Kiltie AE. (2013). Br J Cancer, 108, 748-754.

Guo B, Villeneuve DJ, Hembruff SL, Kirwan AF, Blais DE, Bonin M and Parissenti AM. (2004). Breast Cancer Res Treat, 85, 31-51.

Gyorffy B, Lanczky A, Eklund AC, Denkert C, Budczies J, Li Q and Szallasi Z. (2010). Breast Cancer Res Treat, 123, 725-731.

Harris ME, Bohni R, Schneiderman MH, Ramamurthy L, Schumperli D and Marzluff WF. (1991). Mol Cell Biol, 11, 2416-2424.

Hembruff SL, Laberge ML, Villeneuve DJ, Guo B, Veitch Z, Cecchetto M and Parissenti AM. (2008). BMC Cancer, 8, 318.

Kanehisa M, Goto S, Sato Y, Kawashima M, Furumichi M and Tanabe M. (2014). Nucleic Acids Res, 42, D199-D205.

Lee JH, Choy ML and Marks PA. (2012). Adv Cancer Res, 116, 39-86.

Lowe SW, Ruley HE, Jacks T and Housman DE. (1993). Cell, 74, 957-967.

Minotti G, Menna P, Salvatorelli E, Cairo G and Gianni L. (2004). Pharmacol Rev, 56, 185-229.

Munro AF, Twelves C, Thomas JS, Cameron DA and Bartlett JM. (2012). Br J Cancer, 107, 71-74.

Paridaens R, Biganzoli L, Bruning P, Klijn JG, Gamucci T, Houston S, Coleman R, Schachter J, Van VA, Sylvester R, Awada A, Wildiers J and Piccart M. (2000). J Clin Oncol, 18, 724-733.

Parra MA, Kerr D, Fahy D, Pouchnik DJ and Wyrick JJ. (2006). Mol Cell Biol, 26, 3842-3852.

Parra MA and Wyrick JJ. (2007). Mol Cell Biol, 27, 7641-7648.

Perou CM, Sorlie T, Eisen MB, van de Rijn M, Jeffrey SS, Rees CA, Pollack JR, Ross DT, Johnsen H, Akslen LA, Fluge O, Pergamenschikov A, Williams C, Zhu SX, Lonning PE, Borresen-Dale AL, Brown PO and Botstein D. (2000). Nature, 406, 747-752.

Poole CJ, Earl HM, Hiller L, Dunn JA, Bathers S, Grieve RJ, Spooner DA, Agrawal RK, Fernando IN, Brunt AM, O'Reilly SM, Crawford SM, Rea DW, Simmonds P, Mansi JL, Stanley A, Harvey P, McAdam K, Foster L, Leonard RC and Twelves CJ. (2006). 355, 1851-1862.

Pritchard KI, Munro A, O'Malley FP, Tu D, Li X, Levine MN, Shepherd L, Chia S and Bartlett JM. (2012). Breast Cancer Res Treat, 131, 541-551.

Regel I, Merkl L, Friedrich T, Burgermeister E, Zimmermann W, Einwachter H, Herrmann K, Langer R, Rocken C, Hofheinz R, Schmid R and Ebert MP. (2012). Gastroenterology, 143, 99-109.

Ringel I and Horwitz SB. (1991). J Natl Cancer Inst, 83, 288-291.

Schinkel AH, Roelofs EM and Borst P. (1991). Cancer Res, 51, 2628-2635.

Sorlie T, Perou CM, Tibshirani R, Aas T, Geisler S, Johnsen H, Hastie T, Eisen MB, van de Rijn M, Jeffrey SS, Thorsen T, Quist H, Matese JC, Brown PO, Botstein D, Lonning PE and Borresen-Dale AL. (2001). Proc Natl Acad Sci USA, 98, 10869-10874.

van der Bliek AM, Baas F, Van d, V, Biedler JL, Meyers MB, Ozols RF, Hamilton TC, Joenje H and Borst P. (1988). Cancer Res, 48, 5927-5932.

Wagner JM, Hackanson B, Lubbert M and Jung M. (2010). Clin Epigenetics, 1, 117-136.

Whitfield ML, Zheng LX, Baldwin A, Ohta T, Hurt MM and Marzluff WF. (2000). Mol Cell Biol, 20, 4188-4198.

Wyrick JJ and Parra MA. (2009). Biochim Biophys Acta, 1789, 37-44.

Breitling R, Armengaud P, Amtmann A and Herzyk P. (2004). FEBS Lett, 573, 83-92.




Claims

1. A method of determining a likelihood of resistance to anthracycline in a patient with breast cancer comprising:

a. detecting a level of expression in a sample from the patient, of HIST1H2BK, HIST1H2BD, NEDD9, SYTL2, NHP2, ARPP19, TXNRD1, CENPF, STMN1, CCT5, APRT, UBEC2C, BAX, HDAC1, E2F1, E2F2, E2F4 and CDKN2A;

b. comparing the level of the genes detected in a. to a level of expression of the genes in a control sample; and
wherein there is a likelihood of anthracycline resistance if there is a relatively higher level of expression of the genes in the subject sample compared to the control sample.


 
2. A method of prognosticating survival in a breast cancer patient comprising:

a. detecting a level of expression in a sample from the patient, of HIST1H2BK, HIST1H2BD, NEDD9, SYTL2, NHP2, ARPP19, TXNRD1, CENPF, STMN1, CCT5, APRT, UBEC2C, BAX, HDAC1, E2F1, E2F2, E2F4 and CDKN2A;

b. comparing the level of the genes detected in a. to a level of expression of the genes in a control sample; and
wherein there is a likelihood of poor survival if there is a relatively higher level of expression of the genes in the subject sample compared to the control sample.


 
3. The method of claim 1 or 2, wherein the breast cancer is early breast cancer, preferably selected from the following subtype: ER+HER2-, luminal A, ER+HER2+, luminal B, ER-HER2+, HER2-amplified and ER-/PR-/HER2-, triple negative.
 
4. The method of any one of claims 1-3, wherein the anthracycline is Daunorubicin Doxorubicin, Epirubicin, Idarubicin, Valrubicin, or Mitoxantrone, preferably wherein the anthracycline is Epirubicin.
 
5. A computer implemented product for determining a likelihood of resistance to anthracycline in a breast cancer patient comprising:

a. a means for receiving values corresponding to a subject expression profile in a subject sample;

b. a database comprising a control expression profile associated with HIST1H2BK, HIST1H2BD, NEDD9, SYTL2, NHP2, ARPP19, TXNRD1, CENPF, STMN1, CCT5, APRT, UBEC2C, BAX, HDAC1, E2F1, E2F2, E2F4 and CDKN2A; and

c. a processor disposed to compare the subject expression profile to the control expression profile and determine a likelihood of anthracycline resistance if there is a relatively higher level of expression of the genes in the subject sample compared to the control sample.


 
6. A computer implemented product for prognosticating survival in a breast cancer patient comprising:

a. a means for receiving values corresponding to a subject expression profile in a subject sample; and

b. a database comprising a control expression profile associated with HIST1H2BK, HIST1H2BD, NEDD9, SYTL2, NHP2, ARPP19, TXNRD1, CENPF, STMN1, CCT5, APRT, UBEC2C, BAX, HDAC1, E2F1, E2F2, E2F4 and CDKN2A; and

c. a processor disposed to compare the subject expression profile to the control expression profile and determine there is a likelihood of poor survival if there is a relatively higher level of expression of the genes in the subject sample compared to the control sample.


 


Ansprüche

1. Verfahren zum Bestimmen einer Wahrscheinlichkeit der Resistenz auf Anthracyclin in einem Patienten mit Brustkrebs, umfassend:

a. Erfassen eines Expressionspegels in einer Probe aus dem Patienten von HIST1H2BK, HIST1H2BD, NEDD9, SYTL2, NHP2, ARPP19, TXNRD1, CENPF, STMN1, CCT5, APRT, UBEC2C, BAX, HDAC1, E2F1, E2F2, E2F4 und CDKN2A,

b. Vergleichen des Pegels der in a erfassten Gene mit einem Expressionspegel
der Gene in einer Kontrollgruppe und

wobei eine Wahrscheinlichkeit der Resistenz auf Anthracyclin besteht, wenn ein vergleichsweise höherer Expressionspegel von Genen in der Patientenprobe im Vergleich zur Kontrollprobe vorhanden ist.
 
2. Verfahren zum Prognostizieren des Überlebens bei einem Brustkrebspatienten, umfassend:

a. Erfassen eines Expressionspegels in einer Probe aus dem Patienten von HIST1H2BK, HIST1H2BD, NEDD9, SYTL2, NHP2, ARPP19, TXNRD1, CENPF, STMN1, CCT5, APRT, UBEC2C, BAX, HDAC1, E2F1, E2F2, E2F4 und CDKN2A,

b. Vergleichen des Pegels der in a erfassten Gene mit einem Expressionspegel der Gene in einer Kontrollgruppe und

wobei eine geringe Überlebenswahrscheinlichkeit besteht, wenn ein vergleichsweise höherer Expressionspegel von Genen in der Patientenprobe im Vergleich zur Kontrollprobe vorhanden ist.
 
3. Verfahren nach Anspruch 1 oder 2, wobei der Brustkrebs ein früher Brustkrebs ist, vorzugsweise ausgewählt aus dem folgenden Subtyp: ER+HER2-, Luminal A, ER+HER2+, Luminal B, ER-HER2+, HER2-amplifiziert und ER-/PR-/HER2-, dreifach negativ.
 
4. Verfahren nach einem der Ansprüche 1 bis 3, wobei das Anthracyclin Daunorubicin, Doxorubicin, Epirubicin, Idarubicin, Valrubicin oder Mitoxathron is, wobei das Anthracyclin vorzugsweise Epirubicin ist.
 
5. Rechnerimplementiertes Produkt zum Bestimmen einer Wahrscheinlichkeit der Resistenz auf Anthracyclin in einem Patienten mit Brustkrebs, umfassend:

a. ein Mittel zur Aufnahme von Werten, die einem Patientenexpressionsprofils in einer Patientenprobe entspricht,

b. eine Datenbank, die ein Kontrollexpressionsprofil umfasst, das mit HIST1H2BK, HIST1H2BD, NEDD9, SYTL2, NHP2, ARPP19, TXNRD1, CENPF, STMN1, CCT5, APRT, UBEC2C, BAX, HDAC1, E2F1, E2F2, E2F4 und CDKN2A assoziiert ist, und

c. einen Prozessor, der dazu beschaffen ist, das Patientenexpressionsprofil mit dem Kontrollexpressionsprofil zu vergleichen und eine Wahrscheinlichkeit der Anthracyclinresistenz zu bestimmen, wenn ein vergleichsweise höherer Expressionspegel von Genen in der Patientenprobe im Vergleich zur Kontrollprobe vorhanden ist.


 
6. Rechnerimplementiertes Produkt zum Prognostizieren des Überlebens bei einem Brustkrebspatienten, umfassend:

a. ein Mittel zur Aufnahme von Werten, die einem Patientenexpressionsprofils in einer Patientenprobe entspricht, und

b. eine Datenbank, die ein Kontrollexpressionsprofil umfasst, das mit HIST1H2BK, HIST1H2BD, NEDD9, SYTL2, NHP2, ARPP19, TXNRD1, CENPF, STMN1, CCT5, APRT, UBEC2C, BAX, HDAC1, E2F1, E2F2, E2F4 und CDKN2A assoziiert ist, und

c. einen Prozessor, der dazu beschaffen ist, das Patientenexpressionsprofil mit dem Kontrollexpressionsprofil zu vergleichen und zu bestimmen, dass eine geringe Überlebenswahrscheinlichkeit besteht, wenn ein vergleichsweise höherer Expressionspegel von Genen in der Patientenprobe im Vergleich zur Kontrollprobe vorhanden ist.


 


Revendications

1. Méthode de détermination d'une probabilité de résistance à l'anthracycline dans un patient atteint d'un cancer du sein comprenant :

a. la détection dans un échantillon du patient d'un niveau d'expression de HIST1H2BK, HIST1H2BD, NEDD9, SYTL2, NHP2, ARPP19, TXNRD1, CENPF, STMN1, CCT5, APRT, UBEC2C, BAX, HDAC1, E2F1, E2F2, E2F4 et CDKN2A,

b. la comparaison du niveau de gènes détectés dans a. avec un niveau d'expression des gènes dans un échantillon témoin, et
dans laquelle il y a une probabilité de résistance à l'anthracycline s'il y a un niveau d'expression des gènes relativement plus élevé dans l'échantillon d'un sujet comparé avec l'échantillon témoin.


 
2. Méthode de pronostic de survie d'un patient atteint d'un cancer du sein comprenant :

a. la détection d'un niveau d'expression dans un échantillon du patient de HIST1H2BK, HIST1H2BD, NEDD9, SYTL2, NHP2, ARPP19, TXNRD1, CENPF, STMN1, CCT5, APRT, UBEC2C, BAX, HDAC1, E2F1, E2F2, E2F4 et CDKN2A,

b. la comparaison du niveau des gènes détectés dans a. avec un niveau d'expression des gènes dans un échantillon témoin, et
dans laquelle il y a une probabilité de faible taux de survie s'il y a un niveau relativement plus élevé d'expression des gènes dans l'échantillon d'un sujet comparé avec l'échantillon témoin.


 
3. Méthode suivant une des revendications 1 ou 2, dans laquelle le cancer du sein est un cancer du sein précoce, de préférence sélectionné parmi les sous-types suivants : ER+HER2-, luminal A, ER+HER2+, luminal b, ER-HER2+, HER2-amplifié et ER-/PR-/HER2-, triple négatif.
 
4. Méthode suivant une des revendications 1 à 3, dans laquelle l'anthracycline est Daunorubicine, Doxorubicine, Epirubicine, Idarubicine, Valrubicine, ou Mitoxantrone, de préférence l'anthracycline étant Epirubicine.
 
5. Produit informatique destiné à déterminer une probabilité de résistance à l'anthracycline dans un patient atteint du cancer du sein comprenant :

a. un moyen pour recevoir des valeurs correspondant à un profile d'expression d'un sujet dans un échantillon d'un sujet,

b. une base de données comprenant un profile d'expression témoin associé à HIST1H2BK, HIST1H2BD, NEDD9, SYTL2, NHP2, ARPP19, TXNRD1, CENPF, STMN1, CCT5, APRT, UBEC2C, BAX, HDAC1, E2F1, E2F2, E2F4 et CDKN2A, et

c. un processeur conçu pour comparer le profile d'expression d'un sujet au profile d'expression témoin et déterminer une probabilité de résistance à l'anthracycline s'il y a un niveau relativement plus élevé d'expression des gènes dans l'échantillon d'un sujet comparé avec l'échantillon témoin.


 
6. Produit informatique destiné à établir un prognostique de survie d'un patient atteint d'un cancer du sein comprenant :

a. des moyens pour recevoir des valeurs correspondant à un profile d'expression d'un sujet, et

b. une base de données comprenant un profile d'expression témoin associé à HIST1H2BK, HIST1H2BD, NEDD9, SYTL2, NHP2, ARPP19, TXNRD1, CENPF, STMN1, CCT5, APRT, UBEC2C, BAX, HDAC1, E2F1, E2F2, E2F4 et CDKN2A, et

c. un processeur conçu pour comparer le profile d'expression d'un sujet au profile d'expression témoin et déterminer qu'il y a une probabilité d'un taux faible de survie s'il y a un niveau relativement plus élevé d'expression des gènes dans l'échantillon d'un sujet comparé avec l'échantillon témoin.


 




Drawing
















































































Cited references

REFERENCES CITED IN THE DESCRIPTION



This list of references cited by the applicant is for the reader's convenience only. It does not form part of the European patent document. Even though great care has been taken in compiling the references, errors or omissions cannot be excluded and the EPO disclaims all liability in this regard.

Patent documents cited in the description




Non-patent literature cited in the description