WO2009033941A1 - A method for predicting the response of a tumor in a patient suffering from or at risk of developing recurrent gynecologic cancer towards a chemotherapeutic agent - Google Patents

A method for predicting the response of a tumor in a patient suffering from or at risk of developing recurrent gynecologic cancer towards a chemotherapeutic agent Download PDF

Info

Publication number
WO2009033941A1
WO2009033941A1 PCT/EP2008/061088 EP2008061088W WO2009033941A1 WO 2009033941 A1 WO2009033941 A1 WO 2009033941A1 EP 2008061088 W EP2008061088 W EP 2008061088W WO 2009033941 A1 WO2009033941 A1 WO 2009033941A1
Authority
WO
WIPO (PCT)
Prior art keywords
nucleic acid
expression
cancer
regulated
stromal
Prior art date
Application number
PCT/EP2008/061088
Other languages
French (fr)
Inventor
Mathias Gehrmann
Jan Christoph Brase
Marcus Schmidt
Original Assignee
Siemens Healthcare Diagnostics Gmbh
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
Application filed by Siemens Healthcare Diagnostics Gmbh filed Critical Siemens Healthcare Diagnostics Gmbh
Priority to EP08787449A priority Critical patent/EP2191015A1/en
Priority to US12/674,782 priority patent/US20110143946A1/en
Publication of WO2009033941A1 publication Critical patent/WO2009033941A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6883Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
    • C12Q1/6886Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material for cancer
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/106Pharmacogenomics, i.e. genetic variability in individual responses to drugs and drug metabolism
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/112Disease subtyping, staging or classification
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/158Expression markers

Definitions

  • the present invention relates to methods for predicting the response of a tumor in a patient suffering from or at risk of developing recurrent gynecologic cancer towards a chemotherapeutic agent.
  • breast cancer Every fourth cancer finding in women is breast cancer. Therewith, breast cancer is the most common cancer and the second most common cause of death among women in western in- dustrial nations (Jemal et al . , 2007) 1 . It is estimated that every eighth to tenth woman will develop breast cancer during her lifetime. With a total share of 10 % it is the third most common cancer worldwide (Veronesi et al . , 2005) 2 . With an incidence of 130 cases per 100000 women there are about 55000 new cases in Germany annually from which 18000 cases will cause death (GEKID, 2006) 3 .
  • the mamma carcinoma is a very heterogenous disease with many subtypes. Therefore, even pathologically similar tumors show a different clinical development towards the same therapy. For this reason, the current histopathological markers can not predict the clinical response adequate. Therefore, it is very difficult to perform an optimized therapy. Hence, a therapy often will be chosen due to empirical experiences, and most of the women will be treated systemically as a precaution (Bast et al., 2001; Goldhirsch et al . , 2005) 4 ' 5 .
  • CMF Cyclophosphamid, Meth- otrexat and 5-Fluorouracil
  • 5- Fluorouracil inhibits for instance the Thymidylate Synthetase irreversible and therewith the DNA synthesis (Longley et al . , 2003) 6 .
  • anthracyclines are intercalators, which can incorporate into the DNA, dissolve their structure and inhibit the topoisom- erase II (Capranico et al . , 1989) 7 .
  • the administration of an anthracycline leads to a reduction of recurrent incidences about 12 % and to a reduction of the death rate about 11 % in comparison to a CMF therapy (Misset et al . , 1996) 8 .
  • Paclitaxel was the first taxane, which was used for breast cancer therapy. Paclitaxel binds to the beta- tubuli-unities of the mikrotubuli and stabilizes them (Par- ness and Horwitz, 1981) 12 .
  • platin derivatives Carboplatin, Cisplatin
  • the cytotoxic effect of the platin derivatives is caused by a cross-linking of DNA single strands and double strands, which are disabled thereby.
  • Another problem of chemotherapy is occurrence of adverse effects that might be life threatening or severely impairing the quality of life.
  • RNA from primary tumor tissues for gene expression analysis before neoadjuvant chemotherapy.
  • the chemotherapeutic success can be directly evaluated via tumor reduction and correlated with the gene expres- sion data.
  • predictive gene signatures could be identified (Ayers et al . , 2004; Hess et al., 2006; Gianni et al . , 2005; Thuerigen et al . , 2006) 17 ' 18 ' 19 ' 20 .
  • monotherapies instead of monotherapies have been analyzed.
  • prediction relates to an individual assessment of the malignancy of a tumor, or to the expected survival rate (DFS, disease free survival) of a pa- tient, if the tumor is treated with a given therapy.
  • DFS expected survival rate
  • Prediction of the response to chemotherapy shall be understood to be the act of determining a likely outcome of a chemotherapy in a patient inflicted with cancer.
  • the prediction of a response is preferably made with reference to probability values for reaching a desired or non-desired outcome of the chemotherapy.
  • the predictive methods of the present invention can be used clinically to make treatment decisions by choosing the most appropriate treatment modalities for any particular patient .
  • the "response of a tumor to chemotherapy” relates to any response of the tumor to chemotherapy, preferably to a change in tumor mass and/or volume after initiation of neoadjuvant chemotherapy.
  • Tumor response may be assessed in a neoadjuvant situation where the size of a tumor after systemic intervention can be compared to the initial size and dimensions as measured by CT, PET, mammogram, ultrasound or palpation, usually recorded as "clinical response" of a patient.
  • Response may also be assessed by caliper measurement or pathological examination of the tumor after biopsy or surgical resection.
  • neoadjuvant therapy may be recorded in a quantitative fashion like percentage change in tumor volume or in a qualitative fashion like "no change” (NC) , "partial remission” (PR) , "complete remission” (CR) or other qualitative criteria.
  • Assessment of tumor response may be done early after the onset of neoadjuvant therapy e.g. af- ter a few hours, days, weeks or preferably after a few months.
  • a typical endpoint for response assessment is upon termination of neoadjuvant chemotherapy or upon surgical removal of residual tumor cells and/or the tumor bed. This is typically three month after initiation of neoadjuvant ther- apy.
  • the term "response marker” relates to a marker which can be used to predict the clinical response of a patient towards a given treatment.
  • carcinomas e.g., carcinoma in situ, invasive carcinoma, metastatic carcinoma
  • pre-malignant con- ditions neomorphic changes independent of their histological origin.
  • carcinomas e.g., carcinoma in situ, invasive carcinoma, metastatic carcinoma
  • pre-malignant con- ditions neomorphic changes independent of their histological origin.
  • cancer is not limited to any stage, grade, histomorphological feature, invasiveness, aggressiveness or malignancy of an affected tissue or cell aggregation. In particular stage 0 cancer, stage I cancer, stage II cancer, stage III cancer, stage IV cancer, grade I cancer, grade II cancer, grade III cancer, malignant cancer and primary carcinomas are included.
  • gynecologic cancers refers to can- cer which are diagnosed in female breast and reproductive organs that include the uterus, ovaries, cervix, fallopian tubes, vulva, and vagina.
  • examples of gynecologic cancers include, but are not limited to breast cancer, ovarian cancer, vulvar cancer, vaginal cancer, tubal cancer, endometrian can- cer and/or cervical cancer.
  • endometrian cancer also called endometrial cancer or uterine cancer, includes malignant growth of cells in the endometrium, the lining of the uterus.
  • tumor refers to all neoplastic cell growth and proliferation, whether malignant or benign, and all pre-cancerous and cancerous cells and tissues.
  • determining the status refers to a measurable property of a gene and its products, especially on the nucleotide level and the gene level including mutation status and gene expression status.
  • a number of parameters to determine the status of a gene and its products can be used including, but not limited to, determining the level of protein expression, the amplification or expression status on RNA level or DNA level, of polynucleotides and of polypeptides, and the analysis of haplotype or the mutation status of the gene.
  • An exemplary determinable property correlated with the status of estrogen receptor or progesterone receptor is the amount of the estrogen receptor or progesterone receptor RNA, DNA or other polypeptide in the sample or the presence of nucleotide polymorphisms.
  • biological sample refers to a sample obtained from a patient.
  • the sample may be of any biological tissue or fluid.
  • samples include, but are not limited to, sputum, blood, serum, plasma, blood cells (e.g., white cells) , tissue, core or fine needle biopsy samples, cell-containing body fluids, free floating nucleic acids, urine, peritoneal fluid, and pleural fluid, or cells there from.
  • Biological samples may also include sections of tissues such as frozen or fixed sections taken for histological pur- poses or microdissected cells or extracellular parts thereof.
  • a biological sample to be analyzed is tissue material from neoplastic lesion taken by aspiration or punctuation, excision or by any other surgical method leading to biopsy or resected cellular material.
  • tissue material from neoplastic lesion taken by aspiration or punctuation, excision or by any other surgical method leading to biopsy or resected cellular material.
  • Such biological sample may comprise cells obtained from a patient. The cells may be found in a cell "smear" collected, for example, by a nipple aspiration, ductal lavarge, fine needle biopsy or from provoked or spontaneous nipple discharge.
  • the sample is a body fluid.
  • Such fluids include, for example, blood flu- ids, serum, plasma, lymph, ascitic fluids, gynecological fluids, or urine but not limited to these fluids.
  • array or “matrix” is meant an arrangement of addressable locations or “addresses” on a device.
  • the locations can be arranged in two dimensional arrays, three dimensional arrays, or other matrix formats.
  • the number of locations can range from several to at least hundreds of thousands. Most importantly, each location represents a totally independent reac- tion site.
  • Arrays include but are not limited to nucleic acid arrays, protein arrays and antibody arrays.
  • a “nucleic acid array” refers to an array containing nucleic acid probes, such as oligonucleotides, polynucleotides or larger portions of genes.
  • the nucleic acid on the array is preferably single stranded.
  • oligonucleotide arrays wherein the probes are oligonucleotides are referred to as "oligonucleotide arrays" or “oligonucleotide chips.”
  • a “protein array” refers to an array containing polypeptide probes or protein probes which can be in native form or denatured.
  • An “antibody array” refers to an array containing antibodies which include but are not limited to monoclonal antibodies (e.g. from a mouse), chimeric antibodies, humanized antibodies or phage antibodies and single chain antibodies as well as fragment
  • regulated refers to both upregulation [i.e., activation or stimulation (e.g., by agonizing or potentiating] and down regulation [i.e., inhibition or suppression (e.g., by antagonizing, decreasing or inhibiting) ] .
  • transcriptome relates to the set of all messenger RNA (mRNA) molecules, or "transcripts", produced in one or a population of cells .
  • the term can be applied to the total set of transcripts in a given organism, or to the specific subset of transcripts present in a particular cell type.
  • the transcriptome can vary with external environmental conditions. Because it includes all mRNA transcripts in the cell, the transcriptome reflects the genes that are being actively expressed at any given time, with the exception of mRNA degradation phenomena such as transcriptional attenuation.
  • the discipline of transcriptom- ics examines the expression level of mRNAs in a given cell population, often using high-throughput techniques based on DNA microarray technology.
  • expression levels refers, e.g., to a determined level of gene expression.
  • pattern of expression levels refers to a determined level of gene expression com- pared either to a reference gene (e.g. housekeeper or inversely regulated genes) or to a computed average expression value (e.g. in DNA-chip analyses) .
  • a pattern is not limited to the comparison of two genes but is more related to multiple comparisons of genes to reference genes or samples.
  • a certain “pattern of expression levels” may also result and be determined by comparison and measurement of several genes disclosed hereafter and display the relative abundance of these transcripts to each other.
  • a differentially expressed gene disclosed herein may be used in methods for identifying reagents and compounds and uses of these reagents and compounds for the treatment of cancer as well as methods of treatment.
  • the differential regulation of the gene is not limited to a specific cancer cell type or clone, but rather displays the interplay of cancer cells, muscle cells, stromal cells, connective tissue cells, other epithelial cells, endothelial cells of blood vessels as well as cells of the immune system (e.g. lymphocytes, macrophages, killer cells) .
  • a “reference pattern of expression levels”, within the meaning of the invention shall be understood as being any pattern of expression levels that can be used for the comparison to another pattern of expression levels.
  • a reference pattern of expression levels is, e.g., an average pattern of expression levels observed in a group of healthy or diseased individuals, serving as a reference group.
  • Probe pairs and “probes”, within the meaning of the invention shall have the ordinary meaning of this term which is well known to the person skilled in the art of molecular bi- ology.
  • primer pairs and “probes” shall be understood as being polynucleotide molecules having a sequence identical, complementary, homologous, or homologous to the complement of regions of a target polynucleotide which is to be detected or quantified.
  • nucleotide analogues are also comprised for usage as primers and/or probes.
  • marker refers to a biological molecule, e.g., a nucleic acid, peptide, protein, hormone, etc., whose presence or concentration can be detected and correlated with a known condition, such as a disease state.
  • marker gene refers to a differentially expressed gene whose expression pattern may be util- ized as part of a predictive, prognostic or diagnostic process in malignant neoplasia or cancer evaluation, or which, alternatively, may be used in methods for identifying compounds useful for the treatment or prevention of malignant neoplasia and head and neck, colon or breast cancer in par- ticular.
  • a marker gene may also have the characteristics of a target gene.
  • expression level relates to the process by which a gene's DNA sequence is converted into functional protein (i.e. ligands) and particularly to the amount of said conversion.
  • substantially homologous refers to any probe that can hybridize (i.e., it is the complement of) the single-stranded nucleic acid sequence under conditions of low stringency as described above.
  • hybridization is used in reference to the pairing of complementary nucleic acids.
  • hybridization based method refers to methods imparting a process of combining complementary, single-stranded nucleic acids or nucleotide analogues into a single double stranded molecule. Nucleotides or nucleotide analogues will bind to their complement under normal conditions, so two perfectly complementary strands will bind to each other readily. In bioanalytics, very often labeled, single stranded probes are in order to find complementary target sequences. If such sequences exist in the sample, the probes will hybridize to said sequences which can then be detected due to the label. Other hybridization based methods comprise microarray and/or biochip methods.
  • probes are immobilized on a solid phase, which is then exposed to a sample. If complementary nucleic acids exist in the sample, these will hybridize to the probes and can thus be detected.
  • array based methods are also known as “array based methods”.
  • PCR an- other hybridization based method is PCR, which is described below. When it comes to the determination of expression levels, hybridization based methods may for example be used to determine the amount of mRNA for a given gene.
  • a PCR based method refers to methods comprising a polymerase chain reaction (PCR) .
  • PCR polymerase chain reaction
  • This is an approach for exponentially amplifying nucleic acids, like DNA or RNA, via enzymatic replication, without using a living organism.
  • PCR is an in vitro technique, it can be performed without restrictions on the form of DNA, and it can be extensively modified to perform a wide array of genetic manipulations.
  • a PCR based method may for example be used to detect the presence of a given mRNA by (1) reverse transcription of the complete mRNA pool (the so called transcriptome) into cDNA with help of a reverse transcriptase enzyme, and (2) detecting the presence of a given cDNA with help of respective primers.
  • This approach is commonly known as reverse tran- scriptase PCR (rtPCR)
  • determining the protein level refers to methods which allow the quantitative and/or qualita- tive determination of one or more proteins in a sample. These methods include, among others, protein purification, including ultracentrifugation, precipitation and chromatography, as well as protein analysis and determination, including the use protein microarrays, two-hybrid screening, blotting methods including western blot, one- and two dimensional gelelectrophoresis, isoelectric focusing and the like.
  • anamnesis relates to patient data gained by a physician or other healthcare professional by asking specific questions, either of the patient or of other people who know the person and can give suitable information (in this case, it is sometimes called heteroanamnesis) , with the aim of obtaining information useful in formulating a diagnosis and providing medical care to the patient.
  • This kind of informa- tion is called the symptoms, in contrast with clinical signs, which are ascertained by direct examination.
  • the term "etiopathology” relates to the course of a disease, that is its duration, its clinical symptoms, and its outcome.
  • the present invention provides new diagnostic criteria for the treatment of gynecologic cancer and an optimal predictive gene signature for different chemotherapeutic agents.
  • a method for predicting a response of a tumor in a patient suffering from or at risk of developing recurrent gynecologic cancer, preferably breast cancer, towards a chemotherapeutic agent comprises the steps of:
  • molecular subtype is selected from the group com ⁇ prising the subtypes basal, stromal-high, stromal-low, luminal A, immune system-high, immune system-low, proliferation-high and/or proliferation-low.
  • the molecular subtypes are divided into said groups based on the gene expression of the tumor.
  • one or more genes coregulated with at least one of these genes may be used in addition, or in replacement, of the said genes.
  • Replacement of the said genes with one or more genes coregulated therewith is advan ⁇ tageous in cases, where determining the expression level of the genes enumerated under item b) is critical. Further, an additional use of coregulated genes increases the specificity of said method.
  • Such genes may be selected from the following table.
  • said method comprises the further steps of
  • chemotherapeutic agent is selected from the group comprising Epirubicin, Paclitaxel, 5-Fluorouracil and/or Car- boplatin .
  • the chemotherapeutics may be selected from the group consist- ing of Cyclophosphamid (Endoxan®, Cyclostin®) . Adriamycin
  • Doxorubicin (Doxorubicin) (Adriblastin®) , BCNU (Carmustin) (Carmubris®) , Busulfan (Myleran®) , Bleomycin (Bleomycin®), Carboplatin (Car- boplat®) , Chlorambucil (Leukeran®) , Cis-Platin (Cisplatin®) , Platinex (Platiblastin®) , dacarbazin (DTIC®; Detimedac®) , Do- cetaxel (Taxotere®) , Epirubicin (Farmorubicin®) , Etoposid (Vepesid®) , 5-Fluorouracil (Fluroblastin®, Fluorouracil®) , Gemcitabin (Gemzar®) , Ifosfamid (Holoxan®) , Interferon alpha (Roferon®) , Irinote
  • a first step for the identification of an optimal predictive gene signature for single chemotherapeutic agents four different molecular subtypes (basal, luminal A, stromal-high and stromal-low) whose tumors differ in their response to- wards chemotherapy have been identified via the identified marker genes MLPH, ESRl, PGR and COMP.
  • the basal subtype is
  • hormone receptor negative progesterone receptor negative
  • - Her2 receptor negative sensitive towards most of the chemotherapeutic agents, and exhibits the gene MLPH as differentially expressed gene (down-regulated) and marker gene.
  • the luminal A subtype is a subtype of the luminal A subtype.
  • the stromal-high subtype is
  • the stromal-low subtype is
  • in vitro chemosensitivity assays of pri- mary tumors are performed to determine the response of a tumor towards a single chemotherapeutic agent.
  • the primary tumors were cultivated in different assays with increasing concentrations of the agents. After 6 days of incubation the vitality of the tumor cells were determined with an ATP-measurement .
  • the growing inhibition for the different agent concentrations could be determined and a dose-response curve could be pro- vided.
  • AUC Area under the dose-response curve
  • the AUC is used to evaluate the response of a tumor towards a chemo- therapeutic agent.
  • the tumor samples were classi- fied according to their sensitivity towards the agents into three classes (resistant, intermediate, sensitive) via the tertiles of the AUC arrangement.
  • RNA from the tumor tis- sues was used for molecular profiling with microarrays .
  • Unsu- pervised hierarchical clustering and principal component analysis identified the molecular subtypes.
  • cut off values relate to gene expression values determined by HG-U133a arrays of Affymetrix using MAS5.0 software with target intensity settings of 500.
  • Tumor tissues with an expression MLPH ⁇ 2000 have been characterized as the basal molecular subtype.
  • the expression of the genes ESRl and PGR were > 6000 and 160, respectively, the tumor tissues have been characterized as the subtype luminal A.
  • the remaining tumor tissues have been divided into two different subtypes via the stromal gene COMP (cut-off score 300) .
  • Table 2 Overview regarding the different molecular subtypes and their predictive gene expression signatures
  • the basal molecular subtype predictive for Epirubicin sensitivity is characterized by down-regulated MLPH- expression
  • the luminal A molecular subtype predictive for Epirubicin resistance is characterized by up- regulated ESRl and PGR expression
  • the immune system- high molecular subtype predictive for Epirubicin sensitivity is characterized by up-regulated IGKC and CCL5 expression
  • the immune system-low molecular subtype predictive for Epirubicin resistance is characterized by down-regulated IGKC and CCL5 expression
  • the basal molecular subtype predictive for Paclitaxel sensitivity is characterized by down-regulated MLPH expression
  • the luminal A molecular subtype predictive for Paclitaxel resistance is characterized by up- regulated ESRl and PGR expression
  • the stromal-low molecular subtype predictive for Paclitaxel sensitivity is characterized by down-regulated DCN expression
  • the stromal-high molecular subtype predictive for Paclitaxel resistance is characterized by up- regulated DCN expression
  • the stromal-low molecular subtype predictive for _5 ⁇ Fluorouracil sensitivity is characterized by down- regulated FBNl expression
  • the stromal-high mo- lecular subtype predictive for 5-Fluorouracil resistance is characterized by up-regulated FBNl expression
  • the stromal/proliferation-low molecular subtype pre- dictive for Carboplatin sensitivity is characterized by down-regulated FBN1/UBE2C expression ratio
  • the stromal/proliferation-high molecular subtype predictive for Carboplatin resistance is characterized by up-regulated FBN1/UBE2C expression ratio.
  • the marker genes MLPH and ESRl and PGR, respectively, are used for the prediction towards said agents.
  • IGKC normalized B-cell
  • CCL5 normalized T-cell
  • the defined cut-off score for MLPH predictive for Epirubicin is 2000, the defined score for ESRl predictive for Epirubicin is 6000, the defined score for PGR predictive for Epirubicin is 160 and the defined score for the immune system score (average of IGKC and CCL5) predictive for Epirubicin is 1.5.
  • An overview about the predictive gene (expression) signature for Epirubicin in breast cancer is demonstrated in Table 3.
  • Table 3 Predictive gene (expression) signature for Epirubicin in breast cancer
  • the defined cut-off score for MLPH predictive for Paclitaxel is 2000, the defined score for ESRl predictive for Paclitaxel is 6000, the defined score for PGR predictive for Paclitaxel is 160 and the defined score for DCN predictive for Paclitaxel is 1500.
  • An overview about the predictive gene (expression) signature for Paclitaxel in breast cancer is demonstrated in Table 4.
  • Table 4 Predictive gene (expression) signature for Paclitaxel in breast cancer
  • the defined cut-off score for FBNl predictive for 5- Fluorouracil is 3500.
  • An overview about the predictive gene (expression) signature for 5-Fluorouracil in breast cancer is demonstrated in Table 5.
  • Table 5 Predictive gene (expression) signature for 5- Fluorouracil in breast cancer
  • the defined cut-off score for the ratio between FBNl and UBE2C predictive for Carboplatin is 1.
  • An overview about the predictive gene (expression) signature for Carboplatin in breast cancer is demonstrated in Table 6.
  • Table 6 Predictive gene (expression) signature for Car- boplatin in breast cancer
  • Pa- clitaxel resistance in the basal molecular subtype is charac ⁇ terized by up-regulated AKRlCl expression.
  • the said expression level is determined by
  • the expression level of at least one of the said genes is determined with rtPCR (reverse transcrip ⁇ tase polymerase chain reaction) of the gene related mRNA.
  • the expression level of at least one of the said genes is determined in formalin and/or paraffin fixed tissue samples.
  • tissue samples are taken as bi ⁇ opsies from a patient and undergo diagnostic procedures.
  • the samples are fixed in formaline and/or par- rafine and are then examined with immunohistochemistry methods.
  • the formaline treatment leads to the inactivation of en ⁇ zymes, as for example the ubiquitous RNA-digesting enzymes (RNAses) .
  • RNAses ubiquitous RNA-digesting enzymes
  • the formaline treatment leads to partial depolymeri- zation of the individual mRNA molecules.
  • the current doctrine is that formaline fixed tissue samples can not be used for the analysis of the transcriptome of said tissue .
  • the samples are treated with silica-coated magnetic particles and a cha- otropic salt, in order to purify the nucleic acids contained in said sample for further determination.
  • Collaborators of the inventors of the present invention have developed an approach which however allows successful purification of mRNA out of tissue samples fixed in such manner, and which is disclosed, among others, in WO03058649,
  • Said method comprises the use of magnetic particles coated with silica (Si ⁇ 2) •
  • the silica layer is closed and tight and is characterized by having an extremely small thickness on the scale of a few nanometers.
  • These particles are produced by an improved method that leads to a product having a closed silica layer and thus entail a highly improved purity.
  • the said method prevents an uncontrolled formation of aggregates and clusters of silicates on the magnetite surface whereby positively influencing the additional cited properties and biological applications.
  • the said magnetic particles exhibit an optimized magnetization and suspension behavior as well as a very advantageous run-off behavior from plastic surfaces.
  • These highly pure magnetic particles coated with silicon dioxide are used for isolating nucleic acids, including DNA and RNA, from cell and tissue samples, the separating out from a sample matrix ensuing by means of magnetic fields.
  • These par- tides are particularly well-suited for the automatic purification of nucleic acids, mostly from biological body samples for the purpose of detecting them with different amplification methods.
  • the said approach is particularly useful for the purification of mRNA out of formaline and/or paraffine fixed tissue samples.
  • the said approach creates mRNA fragments which are large enough to al- low specific primer hybridzation and/or specific probe hybridization.
  • a minimal size of at least 100 bp, more preferably 200 base pairs is needed for specific and robust detection of target gene expression.
  • Other issues of perturbance of expression data by sample preparation problems relate to the contamination level with DNA, which is lower compared to other bead based technologies. This of particular importance, as the inventors have observed, that DNAse treatment is not efficient in approximately 10% of FFPE samples generated by standard procedures and stored at room temperature for some years before cutting and RNA extraction.
  • the said approach thus allows a highly specific determination of candidate gene expression levels with one of the above introduced methods, particularly with hybridization based methods, PCR based methods and/or array based methods, even in formaline and/or paraffine fixed tissue samples, and is thus extremely beneficial in the context of the present invention, as it allows the use of tissue samples fixid with formaline and/or paraffine, which are available in tissue banks and connected to clinical databases of sufficient follow-up to allow retrospective analysis.
  • said gynecologic cancer is selected from the group comprising breast cancer, ovarian cancer, vulvar cancer, vaginal cancer, tubal cancer, endometrian cancer and/or cervical cancer.
  • said gynecologic cancer is breast cancer.
  • the method according to the invention may be used for the analysis of a wide variety of neoplastic cell growth and proliferation of the breast tissues including, but not limited to ductal carcinoma in situ, lobular carcinoma, colloid carcinoma, tubular carcinoma, medullary carcinoma, metaplastic carcinoma, intraductal carcinoma in situ, lobular carcinoma in situ and pap- illary carcinoma in situ.
  • kit useful for carrying out one of the said methods comprising at least
  • a primer pair and/or a probe each having a sequence sufficiently complementary to a marker gene according to the present invention; and/or b) at least an antibody directed against a marker according to the present invention
  • a method for correlating the clinical outcome of a patient suffering from or at risk of developing recurrent gynecologic cancer, preferably breast cancer, with the presence or non-presence of a defect in marker gene expression comprising the steps of:
  • nucleic acid molecule selected from the group comprising
  • nucleic acid molecule presented as SEQ ID NO: 1 - 30, b) a nucleic acid molecule having a length of 4 - 80 nucleotides, preferably 18 - 30 nucleotides, the sequence of which corresponds to the sequence of a single stranded fragment of a gene encoding for a marker from the group comprising MLPH, ESRl, PGR, COMP, DCN, IGKC, CCL5, FBNl, UBE2C and/or AKRlCl, c) a nucleic acid molecule that is a fraction, variant, homologue, derivative, or fragment of the nucleic acid molecule presented as SEQ ID NO: 1 - 30, d) a nucleic acid molecule that is capable of hybridiz- ing to any of the nucleic acid molecules of a) - c) under stringent conditions, e) a nucleic acid molecule that is capable of hybridizing to the complement of any of the nucleic acid molecules of a
  • nucleic acid is se- lected from the group comprising DNA, RNA, PNA, LNA and/or
  • the nucleic acid may, in a preferred embodiment, be labelled with at least one detectable marker. This feature is applicable particularly for those nucleic acids which serve as detectable probes for monitoring the polymerase chain reaction process
  • Such detectable markers may for example comprise at least one label selected from the group consisting of fluorescent molecules, luminescent molecules, radioactive molecules, enzy- matic molecules and/or quenching molecules.
  • the said detectable probes are labeled with a fluorescent marker at one end and a quencher of fluorescence at the opposite end of the probe.
  • the close proximity of the reporter to the quencher prevents detection of its fluorescence; breakdown of the probe by the 5 ' to 3 ' exonuclease activity of the taq polymerase breaks the reporter-quencher proximity and thus allows unquenched emission of fluorescence, which can be detected.
  • An increase in the product targeted by the reporter probe at each PCR cycle therefore causes a proportional increase in fluorescence due to the breakdown of the probe and release of the reporter.
  • kits of primers and/or detection probes comprising at least one of the nucleic acids according to the above enumeration and/or their fractions, variants, homologues, derivatives, fragments, complements, hybridizing counterparts, or molecules sharing a sequence identity of at least 70%, preferably 95 %.
  • Said kit may, in another preferred embodiment, comprise at least one of the nucleic acid molecules presented as SEQ ID NO: 1 - 30, and/or their fractions, variants, homologues, derivatives, fragments, complements, hybridizing counterparts, or molecules sharing a sequence identity of at least 70%, preferably 95 %, for the detection of at least one marker gene according to the present invention.
  • nucleic acid according as recited above, or of a kit as recited above for the prediction of a clinical response of a patient suffering from or at risk of developing recurrent gynecologic cancer, preferably breast cancer, towards a chemotherapeutic agent is provided.
  • the predictive gene signatures for the chemotherapeutic agents Epirubicin, 5-Fluorouracil and Paclitaxel have been validated in neoadjuvant studies via the defined cut-off scores.
  • the Epirubicin prediction markers have been tested, to which extent they can predict the relative tumor reduction towards a neoadjuvant EC (Epirubin, Cyclophosphamid) combination therapy in 86 patients.
  • the predictive genes for Epirubicin and 5-Fluorouracil were tested in a study, in which 39 patients received a neoadjuvant FEC (5- Fluorouracil, Epirubicin and Cyclophosphamid) therapy.
  • the inventors of the present invention described for the first time methods for predicting the response of a tumor in a patient suffering from or at risk of developing recurrent breast cancer towards single chemotherapeutic agents.
  • the expression values of the individual genes might be combined by first normalising each individual value and then combining the normalised values into a single meta-value.
  • the ESRl expression value might be divided by e.g. 5000
  • the PGR expression value by e.g. 150
  • the normalised values are added to a combined ESRl-PGR score.
  • the expression value of IGKC (214669_s_at) might be divided by e.g.
  • the value of CCL5 might be divided by e.g. 399 and both normalised values might then be added and subsequently divided by two in order to obtain an immune system metagen score.
  • the ratio between stromal and proliferation might be deduced from normalised expression values of FBNl and UBE2C by e.g. dividing the FBNl value by e.g. 3366 and dividing the value of UBE2C by 973 and subsequently calculating a ratio between the normalised values.
  • the constant used for normalisation are typically derived as the median expression value of that gene found in samples collected from a representative patient cohort.
  • a representative patient cohort is a population of breast cancer patient of a) sufficient size, e.g. preferentially more than 30 breast cancer patients and b) preferen- tially containing a proportion of grade 1, 2, 3, as well as estrogen receptor positive and estrogen receptor negative tumors of at least 5 %.
  • the TAC study and the T-FAC study differ too much compared to the in vitro study. Therefore, the Paclitaxel prediction markers should be analyzed in further studies.
  • AKRlCl has as predictive gene marker for Paclitaxel in the different subtypes defined in the in vitro study. Since in the in vitro study not enough tumor samples exist in the single subtypes to enable an appropriate analysis, the largest available validation dataset was used.
  • the tumors of the neoadjuvant T-FAC study (Hess et al . , 2006) have been classified via the defined predictive gene signatures for Paclitaxel into four subtypes. Subsequently, it could be analyzed to what extent the AKRlCl gene expression in the subtypes differs in the patients, who received a complete remission due to the neoadjuvant therapy, compared to the rest.
  • Figures 1 - 4 demonstrate the chemosensitivity of the tumors towards the chemotherapeutic agents Epirubicin (Fig. 1), Pa- clitaxel (Fig. 2), 5-Fluorouracil (Fig. 3) and Carboplatin (Fig. 4) after classification with predictive gene signatures.
  • the results are depicted via a Box-Whisker-Plot of the AUC (Area under the dose-response curve) of the prediction classes for Epirubicin (basal, immune system-high, immune system-low, luminal A) , Paclitaxel (basal, stromal-low, stro- mal-high, luminal A) , 5-Fluorouracil (stromal-low, stromal- high) and Carboplatin (ratio stromal/proliferation-low, ratio stromal/proliferation-high) (43 tumors for the agents Pacli- taxel/Epirubicin and 34 tumors for 5- Fluorouracil/Carboplatin) .
  • AUC Average under the dose-response curve
  • Figure 5 demonstrates the relative reduction of 86 tumors towards a neoadjuvant EC combination therapy.
  • the results are depicted via a Box-Whisker-Plot of the relative tumor reduction of the Epirubicin prediction classes (basal, immune system-high, immune system-low and luminal A) .
  • Significant re- suits of a nonparametric test are demonstrated for the comparison of all subtypes adjacent the Figure and for the pair- wise comparison of the subtypes within the Figure (* : p- score ⁇ 0.05, **: p-score ⁇ 0.01 , ***: p-score ⁇ 0.001) (Tab. 9) .
  • Figures 6 and 7 demonstrate the relative reduction of 39 tumors towards a neoadjuvant FEC combination therapy. The results are depicted via a Box-Whisker-Plot of the relative tumor reduction of the prediction classes, which are defined via the Epirubicin (Fig. 6) and 5-Fluorouracil (Fig. 7) prediction markers. Significant results of a nonparametric test are demonstrated for the comparison of all subtypes adjacent the Figures and for the pairwise comparison of the subtypes within the Figures (* : p-score ⁇ 0.05) (Tab. 9) .
  • Figures 8 and 9 demonstrate prediction classes of the Pacli- taxel prediction markers in neoadjuvant studies.
  • the percentage of the tumors with "(pathological) complete remission” (pCR/CR) (in each case the bar on the left side) and with incomplete remission (in each case the bar on the right side) in a neoadjuvant TAC (Fig. 8) and T-FAC (Fig. 9) study is demonstrated.
  • Figures 10 to 13 demonstrate the general classification schema using preferred marker genes for the different chemo- therapeutic agents.
  • Figure 14 demonstrates the chemosensitivity of different cell lines towards Paclitaxel.
  • NCI 60 cell lines were classified with the marker gene AKRlCl (cut-off score 5000) .
  • AKRlCl cut-off score 5000
  • concentration of Palitaxel is depicted, which is needed for the inhibition of half of the cells (GI-50) .
  • GI-50 values for the cell lines were provided.
  • Figure 15 demonstrates the expression of the gene AKRlCl in a neoadjuvant T-FAC study for the molecular subtypes basal, stromal-low, stromal-high and luminal A. For each group there occurred a classification of the tumors, which received a pathological complete remission (depicted in each case on the right side) or an incomplete remission (depicted in each case on the left side) towards the neoadjuvant therapy.
  • Paclitaxel predicin vitro tor study p-score
  • Carboplatin prein vitro dictor study p-score stromal (low) vs , 0.0029* stromal (high)

Landscapes

  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Engineering & Computer Science (AREA)
  • Immunology (AREA)
  • Pathology (AREA)
  • Analytical Chemistry (AREA)
  • Zoology (AREA)
  • Genetics & Genomics (AREA)
  • Wood Science & Technology (AREA)
  • Physics & Mathematics (AREA)
  • Biotechnology (AREA)
  • Microbiology (AREA)
  • Molecular Biology (AREA)
  • Hospice & Palliative Care (AREA)
  • Biophysics (AREA)
  • Oncology (AREA)
  • Biochemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)

Abstract

The present invention relates to a method for predicting a response of a tumor in a patient suffering from or at risk of developing recurrent gynecologic cancer towards a chemotherapeutic agent, said method comprising the steps of : a) obtaining a biological sample from said patient; b) determining the pattern of expression level of at least one gene of the group comprising AKR1C1, MLPH, ESR1, PGR, COMP, DCN, IGKC, CCL5, FBNl and/or UBE2C, or of genes coregulated therewith, in said sample; c) comparing the pattern of expression levels determined in (b) with one or several reference pattern (s) of expression levels; d) identifying at least one marker gene; e) determining a molecular subtype for said sample on the basis of (d); and f) predicting from said molecular subtype response of a tumor for a chemotherapeutic agent, wherein the molecular subtype is selected from the group comprising the subtypes basal, stromal-high, stromal-low, luminal A, immune system-high, immune system-low, proliferation-high and/or proliferation-low.

Description

Description
A method for predicting the response of a tumor in a patient suffering from or at risk of developing recurrent gynecologic cancer towards a chemotherapeutic agent
Field of the invention
The present invention relates to methods for predicting the response of a tumor in a patient suffering from or at risk of developing recurrent gynecologic cancer towards a chemotherapeutic agent.
Background of the invention
Every fourth cancer finding in women is breast cancer. Therewith, breast cancer is the most common cancer and the second most common cause of death among women in western in- dustrial nations (Jemal et al . , 2007) 1. It is estimated that every eighth to tenth woman will develop breast cancer during her lifetime. With a total share of 10 % it is the third most common cancer worldwide (Veronesi et al . , 2005) 2. With an incidence of 130 cases per 100000 women there are about 55000 new cases in Germany annually from which 18000 cases will cause death (GEKID, 2006) 3.
The mamma carcinoma is a very heterogenous disease with many subtypes. Therefore, even pathologically similar tumors show a different clinical development towards the same therapy. For this reason, the current histopathological markers can not predict the clinical response adequate. Therefore, it is very difficult to perform an optimized therapy. Hence, a therapy often will be chosen due to empirical experiences, and most of the women will be treated systemically as a precaution (Bast et al., 2001; Goldhirsch et al . , 2005) 4'5.
Usually, a combination of several agents will be used since due to the different therapeutic mechanisms of the single chemotherapeutic agents the development of crossresistances is unlikely. A very common combination therapy used already many years as standard therapy is CMF (Cyclophosphamid, Meth- otrexat and 5-Fluorouracil) . These three chemotherapeutic agents have different molecular mechanisms of action. 5- Fluorouracil inhibits for instance the Thymidylate Synthetase irreversible and therewith the DNA synthesis (Longley et al . , 2003) 6.
Over the years new chemotherapeutic agents were identified, which might have an advantage for the treatment of patients. In clinical practice there exists the problem that a new substance can not be tested independently from other agents since they are added to an already established combination therapy. With the results from this therapy it can be determined, whether there could arise an improvement over the standard therapy.
Thus, after the discovery of anthracyclines it could be demonstrated in clinical studies that for instance the combination therapy EC/AC (Epirubicin and Doxorubicin, respectively, and Cyclophosphamid) has an advantage over the CMF therapy. Anthracyclines are intercalators, which can incorporate into the DNA, dissolve their structure and inhibit the topoisom- erase II (Capranico et al . , 1989) 7. The administration of an anthracycline leads to a reduction of recurrent incidences about 12 % and to a reduction of the death rate about 11 % in comparison to a CMF therapy (Misset et al . , 1996) 8.
In current clinical studies it could be demonstrated that the completion of an anthacycline based chemotherapy with a tax- ane leads to an additional advantage of survival (Nabholtz et al., 1999; Henderson et al . , 2003; Bishop et al . , 1999) 9, io, ii _ Tne agent Paclitaxel was the first taxane, which was used for breast cancer therapy. Paclitaxel binds to the beta- tubuli-unities of the mikrotubuli and stabilizes them (Par- ness and Horwitz, 1981) 12. Newly, platin derivatives (Carboplatin, Cisplatin) are used for the treatment of the mamma carcinoma. The cytotoxic effect of the platin derivatives is caused by a cross-linking of DNA single strands and double strands, which are disabled thereby.
One problem of chemotherapy is development of resistances of some tumors towards single or several agents which can in- hibit the success of a chemotherapy. To date, only a few resistance mechanisms are known. Furthermore, the order of the application seems to be important regarding crossresistances (Paridaens et al . , 2000) 13. Thus, the identification at an early stage of those resistances is very important to change a therapy in good time. However, it would be optimal to evaluate possible resistances already before a treatment.
Another problem of chemotherapy is occurrence of adverse effects that might be life threatening or severely impairing the quality of life.
In the last years the heterogeneity and complexity of mamma carcinoma have been analyzed on molecular level to analyze which genes are expressed in these tumors.
An already known approach for the identification of predictive gene signatures is the cultivation of cell lines with the purpose to compare the gene expression of resistant and sensitive breast cancer tumor cells. Different genes could be identified which could be appropriate for the prediction of a tumor response towards single chemotherapeutic agents (Ville- neuve et al . , 2006; Gyorffy et al . , 2006) 14'15. Since the expression profile of cell lines in the in vitro situation is much different from the in vivo situation of primary tumor tissues (Ross and Perou, 2001) 16, it has to be evaluated, to which extent the identified genes could be appropriate for prediction in clinical practice. In research neoadjuvant chemotherapy is very important as well since breast tumor response towards chemotherapeutic agents can be directly analyzed via the tumor reduction status .
A common approach is to isolate RNA from primary tumor tissues for gene expression analysis before neoadjuvant chemotherapy. The chemotherapeutic success can be directly evaluated via tumor reduction and correlated with the gene expres- sion data. In several neoadjuvant studies predictive gene signatures could be identified (Ayers et al . , 2004; Hess et al., 2006; Gianni et al . , 2005; Thuerigen et al . , 2006) 17'18'19'20. In most of the studies neoadjuvant combination therapies instead of monotherapies have been analyzed. Thus, it is difficult to identify the cause of resistances as well as to transfer the identified gene signatures upon other combination therapies.
To date, there exists no reliable predictive marker which can predict the response towards a chemotherapeutic agent in breast cancer. To date, no gene signature could be validated in an independent dataset.
Therefore, a need exists to identify those patients who are likely to respond to a chemotherapeutic agent by providing predictive information. There is a great need for predictive methods that can assist the practicing physician to make treatment choices based on reliable analysis.
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 belongs.
The term "prediction", as used herein, relates to an individual assessment of the malignancy of a tumor, or to the expected survival rate (DFS, disease free survival) of a pa- tient, if the tumor is treated with a given therapy.
"Prediction of the response to chemotherapy", within the meaning of the invention, shall be understood to be the act of determining a likely outcome of a chemotherapy in a patient inflicted with cancer. The prediction of a response is preferably made with reference to probability values for reaching a desired or non-desired outcome of the chemotherapy. The predictive methods of the present invention can be used clinically to make treatment decisions by choosing the most appropriate treatment modalities for any particular patient .
The "response of a tumor to chemotherapy", within the meaning of the invention, relates to any response of the tumor to chemotherapy, preferably to a change in tumor mass and/or volume after initiation of neoadjuvant chemotherapy. Tumor response may be assessed in a neoadjuvant situation where the size of a tumor after systemic intervention can be compared to the initial size and dimensions as measured by CT, PET, mammogram, ultrasound or palpation, usually recorded as "clinical response" of a patient. Response may also be assessed by caliper measurement or pathological examination of the tumor after biopsy or surgical resection. Response may be recorded in a quantitative fashion like percentage change in tumor volume or in a qualitative fashion like "no change" (NC) , "partial remission" (PR) , "complete remission" (CR) or other qualitative criteria. Assessment of tumor response may be done early after the onset of neoadjuvant therapy e.g. af- ter a few hours, days, weeks or preferably after a few months. A typical endpoint for response assessment is upon termination of neoadjuvant chemotherapy or upon surgical removal of residual tumor cells and/or the tumor bed. This is typically three month after initiation of neoadjuvant ther- apy. The term "response marker" relates to a marker which can be used to predict the clinical response of a patient towards a given treatment.
The terms "cancer" and "cancerous" refer to or describe the physiological condition in mammals that is typically characterized by unregulated cell growth. The term "cancer" as used herein includes carcinomas, (e.g., carcinoma in situ, invasive carcinoma, metastatic carcinoma) and pre-malignant con- ditions, neomorphic changes independent of their histological origin. The term "cancer" is not limited to any stage, grade, histomorphological feature, invasiveness, aggressiveness or malignancy of an affected tissue or cell aggregation. In particular stage 0 cancer, stage I cancer, stage II cancer, stage III cancer, stage IV cancer, grade I cancer, grade II cancer, grade III cancer, malignant cancer and primary carcinomas are included.
As used herein, the term "gynecologic cancers" refers to can- cer which are diagnosed in female breast and reproductive organs that include the uterus, ovaries, cervix, fallopian tubes, vulva, and vagina. Examples of gynecologic cancers include, but are not limited to breast cancer, ovarian cancer, vulvar cancer, vaginal cancer, tubal cancer, endometrian can- cer and/or cervical cancer. As used herein, the term "endometrian cancer", also called endometrial cancer or uterine cancer, includes malignant growth of cells in the endometrium, the lining of the uterus.
The term "tumor" as used herein, refers to all neoplastic cell growth and proliferation, whether malignant or benign, and all pre-cancerous and cancerous cells and tissues.
The term "determining the status" as used herein, refers to a measurable property of a gene and its products, especially on the nucleotide level and the gene level including mutation status and gene expression status. A number of parameters to determine the status of a gene and its products can be used including, but not limited to, determining the level of protein expression, the amplification or expression status on RNA level or DNA level, of polynucleotides and of polypeptides, and the analysis of haplotype or the mutation status of the gene. An exemplary determinable property correlated with the status of estrogen receptor or progesterone receptor is the amount of the estrogen receptor or progesterone receptor RNA, DNA or other polypeptide in the sample or the presence of nucleotide polymorphisms.
The term "biological sample", as used herein, refers to a sample obtained from a patient. The sample may be of any biological tissue or fluid. Such samples include, but are not limited to, sputum, blood, serum, plasma, blood cells (e.g., white cells) , tissue, core or fine needle biopsy samples, cell-containing body fluids, free floating nucleic acids, urine, peritoneal fluid, and pleural fluid, or cells there from. Biological samples may also include sections of tissues such as frozen or fixed sections taken for histological pur- poses or microdissected cells or extracellular parts thereof. A biological sample to be analyzed is tissue material from neoplastic lesion taken by aspiration or punctuation, excision or by any other surgical method leading to biopsy or resected cellular material. Such biological sample may comprise cells obtained from a patient. The cells may be found in a cell "smear" collected, for example, by a nipple aspiration, ductal lavarge, fine needle biopsy or from provoked or spontaneous nipple discharge. In another embodiment, the sample is a body fluid. Such fluids include, for example, blood flu- ids, serum, plasma, lymph, ascitic fluids, gynecological fluids, or urine but not limited to these fluids.
By "array" or "matrix" is meant an arrangement of addressable locations or "addresses" on a device. The locations can be arranged in two dimensional arrays, three dimensional arrays, or other matrix formats. The number of locations can range from several to at least hundreds of thousands. Most importantly, each location represents a totally independent reac- tion site. Arrays include but are not limited to nucleic acid arrays, protein arrays and antibody arrays. A "nucleic acid array" refers to an array containing nucleic acid probes, such as oligonucleotides, polynucleotides or larger portions of genes. The nucleic acid on the array is preferably single stranded. Arrays wherein the probes are oligonucleotides are referred to as "oligonucleotide arrays" or "oligonucleotide chips." A "microarray, " herein also refers to a "biochip" or "biological chip", an array of regions having a density of discrete regions of at least about 100/cm2, and preferably at least about 1000/cm2. The regions in a microarray have typical dimensions, e.g., diameters, in the range of between about 10-250 μm, and are separated from other regions in the array by about the same distance. A "protein array" refers to an array containing polypeptide probes or protein probes which can be in native form or denatured. An "antibody array" refers to an array containing antibodies which include but are not limited to monoclonal antibodies (e.g. from a mouse), chimeric antibodies, humanized antibodies or phage antibodies and single chain antibodies as well as fragments from antibodies .
The terms "regulated" or "regulation" as used herein refer to both upregulation [i.e., activation or stimulation (e.g., by agonizing or potentiating] and down regulation [i.e., inhibition or suppression (e.g., by antagonizing, decreasing or inhibiting) ] .
The term "transcriptome" relates to the set of all messenger RNA (mRNA) molecules, or "transcripts", produced in one or a population of cells . The term can be applied to the total set of transcripts in a given organism, or to the specific subset of transcripts present in a particular cell type. Unlike the genome, which is roughly fixed for a given cell line (exclud- ing mutations) , the transcriptome can vary with external environmental conditions. Because it includes all mRNA transcripts in the cell, the transcriptome reflects the genes that are being actively expressed at any given time, with the exception of mRNA degradation phenomena such as transcriptional attenuation. The discipline of transcriptom- ics examines the expression level of mRNAs in a given cell population, often using high-throughput techniques based on DNA microarray technology.
The term "expression levels" refers, e.g., to a determined level of gene expression. The term "pattern of expression levels" refers to a determined level of gene expression com- pared either to a reference gene (e.g. housekeeper or inversely regulated genes) or to a computed average expression value (e.g. in DNA-chip analyses) . A pattern is not limited to the comparison of two genes but is more related to multiple comparisons of genes to reference genes or samples. A certain "pattern of expression levels" may also result and be determined by comparison and measurement of several genes disclosed hereafter and display the relative abundance of these transcripts to each other.
Alternatively, a differentially expressed gene disclosed herein may be used in methods for identifying reagents and compounds and uses of these reagents and compounds for the treatment of cancer as well as methods of treatment. The differential regulation of the gene is not limited to a specific cancer cell type or clone, but rather displays the interplay of cancer cells, muscle cells, stromal cells, connective tissue cells, other epithelial cells, endothelial cells of blood vessels as well as cells of the immune system (e.g. lymphocytes, macrophages, killer cells) .
A "reference pattern of expression levels", within the meaning of the invention shall be understood as being any pattern of expression levels that can be used for the comparison to another pattern of expression levels. In a preferred embodi- ment of the invention, a reference pattern of expression levels is, e.g., an average pattern of expression levels observed in a group of healthy or diseased individuals, serving as a reference group. "Primer pairs" and "probes", within the meaning of the invention, shall have the ordinary meaning of this term which is well known to the person skilled in the art of molecular bi- ology. In a preferred embodiment of the invention "primer pairs" and "probes" shall be understood as being polynucleotide molecules having a sequence identical, complementary, homologous, or homologous to the complement of regions of a target polynucleotide which is to be detected or quantified. In yet another embodiment, nucleotide analogues are also comprised for usage as primers and/or probes.
The term "marker" refers to a biological molecule, e.g., a nucleic acid, peptide, protein, hormone, etc., whose presence or concentration can be detected and correlated with a known condition, such as a disease state.
The term "marker gene, " as used herein, refers to a differentially expressed gene whose expression pattern may be util- ized as part of a predictive, prognostic or diagnostic process in malignant neoplasia or cancer evaluation, or which, alternatively, may be used in methods for identifying compounds useful for the treatment or prevention of malignant neoplasia and head and neck, colon or breast cancer in par- ticular. A marker gene may also have the characteristics of a target gene.
The term "expression level", as used herein, relates to the process by which a gene's DNA sequence is converted into functional protein (i.e. ligands) and particularly to the amount of said conversion.
When used in reference to a single-stranded nucleic acid sequence, the term "substantially homologous" refers to any probe that can hybridize (i.e., it is the complement of) the single-stranded nucleic acid sequence under conditions of low stringency as described above. As used herein, the term "hybridization" is used in reference to the pairing of complementary nucleic acids.
The term "hybridization based method", as used herein, refers to methods imparting a process of combining complementary, single-stranded nucleic acids or nucleotide analogues into a single double stranded molecule. Nucleotides or nucleotide analogues will bind to their complement under normal conditions, so two perfectly complementary strands will bind to each other readily. In bioanalytics, very often labeled, single stranded probes are in order to find complementary target sequences. If such sequences exist in the sample, the probes will hybridize to said sequences which can then be detected due to the label. Other hybridization based methods comprise microarray and/or biochip methods. Therein, probes are immobilized on a solid phase, which is then exposed to a sample. If complementary nucleic acids exist in the sample, these will hybridize to the probes and can thus be detected. These approaches are also known as "array based methods". Yet an- other hybridization based method is PCR, which is described below. When it comes to the determination of expression levels, hybridization based methods may for example be used to determine the amount of mRNA for a given gene.
The term "a PCR based method" as used herein refers to methods comprising a polymerase chain reaction (PCR) . This is an approach for exponentially amplifying nucleic acids, like DNA or RNA, via enzymatic replication, without using a living organism. As PCR is an in vitro technique, it can be performed without restrictions on the form of DNA, and it can be extensively modified to perform a wide array of genetic manipulations. When it comes to the determination of expression levels, a PCR based method may for example be used to detect the presence of a given mRNA by (1) reverse transcription of the complete mRNA pool (the so called transcriptome) into cDNA with help of a reverse transcriptase enzyme, and (2) detecting the presence of a given cDNA with help of respective primers. This approach is commonly known as reverse tran- scriptase PCR (rtPCR)
The term "determining the protein level", as used herein, refers to methods which allow the quantitative and/or qualita- tive determination of one or more proteins in a sample. These methods include, among others, protein purification, including ultracentrifugation, precipitation and chromatography, as well as protein analysis and determination, including the use protein microarrays, two-hybrid screening, blotting methods including western blot, one- and two dimensional gelelectrophoresis, isoelectric focusing and the like.
The term "anamnesis" relates to patient data gained by a physician or other healthcare professional by asking specific questions, either of the patient or of other people who know the person and can give suitable information (in this case, it is sometimes called heteroanamnesis) , with the aim of obtaining information useful in formulating a diagnosis and providing medical care to the patient. This kind of informa- tion is called the symptoms, in contrast with clinical signs, which are ascertained by direct examination.
The term "etiopathology" relates to the course of a disease, that is its duration, its clinical symptoms, and its outcome.
Object of the invention
It is one object of the present invention to provide an improved method for the prediction of a response of a tumor in a patient suffering from gynecologic cancer towards a chemo- therapeutic agent to current status tests.
The present invention provides new diagnostic criteria for the treatment of gynecologic cancer and an optimal predictive gene signature for different chemotherapeutic agents.
These objects are met with methods and means according to the independent claims of the present invention. The dependent claims are related to preferred embodiments. It is yet to be understood that value ranges delimited by numerical values are to be understood to include the said delimiting values.
Summary of the invention
Before the invention is described in detail, it is to be understood that this invention is not limited to the particular component parts of the devices described or process steps of the methods described as such devices and methods may vary. It is also to be understood that the terminology used herein is for purposes of describing particular embodiments only, and is not intended to be limiting. It must be noted that, as used in the specification and the appended claims, the singu- lar forms "a, " "an" and "the" include singular and/or plural referents unless the context clearly dictates otherwise. It is moreover to be understood that, in case parameter ranges are given which are delimited by numeric values, the ranges are deemed to include these limitation values.
According to the invention, a method is provided for predicting a response of a tumor in a patient suffering from or at risk of developing recurrent gynecologic cancer, preferably breast cancer, towards a chemotherapeutic agent. Said method comprises the steps of:
a) obtaining a biological sample from said patient; b) determining the pattern of expression level of at least one gene of the group comprising AKRlCl, MLPH, ESRl, PGR, COMP, DCN, IGKC, CCL5, FBNl and/or UBE2C, or of genes coregulated therewith, in said sample; c) comparing the pattern of expression levels determined in (b) with one or several reference pattern (s) of expression levels; d) identifying at least one marker gene; e) determining a molecular subtype for said sample on the basis of (d) ; and f) predicting from said molecular subtype response of a tumor for a chemotherapeutic agent,
wherein the molecular subtype is selected from the group com¬ prising the subtypes basal, stromal-high, stromal-low, luminal A, immune system-high, immune system-low, proliferation-high and/or proliferation-low.
The molecular subtypes are divided into said groups based on the gene expression of the tumor.
It is vital to the present invention that, in addition to the genes enumerated under item b) , one or more genes coregulated with at least one of these genes may be used in addition, or in replacement, of the said genes. Replacement of the said genes with one or more genes coregulated therewith is advan¬ tageous in cases, where determining the expression level of the genes enumerated under item b) is critical. Further, an additional use of coregulated genes increases the specificity of said method.
Such genes may be selected from the following table.
Table 1 : Coregulated genes of Interest
Figure imgf000016_0001
Figure imgf000017_0001
Figure imgf000018_0001
Figure imgf000019_0001
Figure imgf000020_0001
Figure imgf000021_0001
Figure imgf000022_0001
Figure imgf000023_0001
Figure imgf000024_0001
at
217148 X at 0. 891 IGLJ3 immunoglobulin lambda joining 3
214677 X at 0. 888 IGLJ3 immunoglobulin lambda joining 3 similar to bK24 6H3.1 (lmmu- noglobulm lambda-like
215946 X polypeptide 1, pre-B-cell spe- at 0. 887 LOC91316 cific)
IGKV1OR2-
108; IGOl;
IGKV1OR210
O ;
217378 X IGKV1/OR2- at 0. 887 108
214768 X at 0. 885 IGKC immunoglobulin kappa constant
214777 at 0. 883 IGKC immunoglobulin kappa constant
IGKV1OR15-
118;
IGKVP2;
IGKV1OR118
IGKV1/OR-
118;
217480 X IGKV1/OR15 at 0. 882 -118
216401 X at 0. 879 IGKV immunoglobulin heavy constant
211430 S gamma 3 at 0. 877 IGHG3 (G3m marker) similar to bK24 6H3.1 (lmmu- noglobulm lambda-like
213502 X IGLL3; polypeptide 1, pre-B-cell spe- at 0. 872 16.1 cific)
216984 X 0. 867 IGLJ3 immunoglobulin lambda joining 3 at
211798 X at 0.852 IGLJ3 immunoglobulin lambda joining 3
209374 S at 0.845 IGHM immunoglobulin heavy constant mu
215214 at 0.834 IGLg immunoglobulin lambda locus
211634 X immunoglobulin heavy constant at 0.83 IGHM gamma 3 (G3m marker)
217281 X immunoglobulin heavy constant at 0.828 IGHG3 gamma 3 (G3m marker)
POU domain, class 2, associating
205267 at 0.819 POU2AF1 factor 1
211881 X at 0.819 IGLJ3 immunoglobulin lambda joining 3
217258 X at 0.819
217179 X at 0.817 IGL@ immunoglobulin lambda locus
217235 X at 0.808 IGLJ3 immunoglobulin lambda joining 3
216365 X at 0.796 immunoglobulin lambda joining 3
217227 X at 0.792 IGL@ immunoglobulin lambda locus immunoglobulin J polypeptide, linker protein for immunoglobulin alpha and mu po -
212592 at 0.791 IGJ lypeptides
216560 X at 0.786 immunoglobulin lambda locus tumor necrosis factor receptor
206641 at 0.785 TNFRSF17 superfamily, member 17
211868 X at 0.784
211637 X at 0.78 IGHM immunoglobulin heavy constant mu
214973 X 0.78 IGVH3 immunoglobulin heavy constant
Figure imgf000027_0001
Figure imgf000028_0001
Figure imgf000029_0001
Figure imgf000030_0001
Figure imgf000031_0001
204620 S chondroitin sulfate proteoglycan at 0 .848 CSPG2 2 (versican)
201438 at 0 .845 COL6A3 collagen, type VI, alpha 3
211161 S at 0 .845
207173 X cadherin 11, type 2, OB-cadherin at 0 .84 CDHIl (osteoblast)
204619 S chondroitin sulfate proteoglycan at 0 .836 CSPG2 2 (versican)
215446 S at 0 .836 LOX lysyl oxidase secreted protein, acidic, cys-
212667 at 0 .828 SPARC teine-rich (osteonectin) collagen, type III, alpha 1
215076 S (Ehlers-Danlos syndrome at 0 .827 COL3A1 type IV, autosomal dominant)
202404 S at 0 .823 COL1A2 collagen, type I, alpha 2
202450 S at 0 .823 CTSK cathepsin K (pycnodysostosis)
212488 at 0 .82 COL5A1 collagen, type V, alpha 1 transcription factor 8 (represses interleukin 2 expres¬
212764 at 0 .819 TCF8 sion)
DKFZP434B0 hypothetical protein
221541 at 0 .816 44 DKFZp434B044
Homo sapiens cDNA FLJ31066 fis,
213790 at 0 .815 clone HSYRA2001153.
221730 at 0 .815 COL5A2 collagen, type V, alpha 2
202403 S at 0 .812 COL1A2 collagen, type I, alpha 2
201792 at 0 .805 AEBPl AE binding protein 1
209335 at 0 .805 DCN decorin
203083 at 0 .803 THBS2 thrombospondin 2
219087 at 0 .8 ASPN asporin (LRR class 1) protease, serine, 11 (IGF bin¬
201185 at 0 .798 PRSSIl ding)
Figure imgf000033_0001
Figure imgf000034_0001
Figure imgf000035_0001
Figure imgf000036_0001
Figure imgf000037_0001
Figure imgf000038_0001
Figure imgf000039_0001
However, other genes not mentioned in the above table, which are yet coregulated with at least one of the genes enumerated under item b) , do also fall under the scope of the present invention. The teaching presented herein will make it obvious to the person skilled in the art to find such coregulated genes in literature, in databases or the like, without the need of inventive step.
In a preferred embodiment, said method comprises the further steps of
g) selecting at least one chemotherapeutic agent which is predicted to be successful,
wherein the chemotherapeutic agent is selected from the group comprising Epirubicin, Paclitaxel, 5-Fluorouracil and/or Car- boplatin .
The chemotherapeutics may be selected from the group consist- ing of Cyclophosphamid (Endoxan®, Cyclostin®) . Adriamycin
(Doxorubicin) (Adriblastin®) , BCNU (Carmustin) (Carmubris®) , Busulfan (Myleran®) , Bleomycin (Bleomycin®), Carboplatin (Car- boplat®) , Chlorambucil (Leukeran®) , Cis-Platin (Cisplatin®) , Platinex (Platiblastin®) , Dacarbazin (DTIC®; Detimedac®) , Do- cetaxel (Taxotere®) , Epirubicin (Farmorubicin®) , Etoposid (Vepesid®) , 5-Fluorouracil (Fluroblastin®, Fluorouracil®) , Gemcitabin (Gemzar®) , Ifosfamid (Holoxan®) , Interferon alpha (Roferon®) , Irinotecan (CPT 11, Campto®) , Melphalan (AIk- eran®) , Methotrexat (Methotrexat®, Farmitrexat®) , Mitomycin C (Mitomycin®) , Mitoxantron (Novantron®) , Oxaliplatin
(Eloxatine®) , Paclitaxel (Taxol®) , Prednimustin (Sterecyt®) , Procarbazin (Natulan®) , Ralitrexed (Tomudex®) , Trofosfamid (Ixoten®), Vinblastin (Velbe®) , Vincristin (Vincristin®) , Vindesin (Eldisine®) , Vinorelbin (Navelbine®) .
In a first step for the identification of an optimal predictive gene signature for single chemotherapeutic agents four different molecular subtypes (basal, luminal A, stromal-high and stromal-low) whose tumors differ in their response to- wards chemotherapy have been identified via the identified marker genes MLPH, ESRl, PGR and COMP. The basal subtype is
hormone receptor negative, progesterone receptor negative, - Her2 receptor negative, sensitive towards most of the chemotherapeutic agents, and exhibits the gene MLPH as differentially expressed gene (down-regulated) and marker gene.
The luminal A subtype is
resistant towards most of the chemotherapeutic agents, and - exhibits the genes ESRl and PGR as differentially expressed genes (up-regulated) and marker gene.
The stromal-high subtype is
- resistant towards most of the chemotherapeutic agents, and exhibits the gene COMP as differentially expressed gene (up-regulated) and marker gene.
The stromal-low subtype is
sensitive towards most of the chemotherapeutic agents, and exhibits the gene COMP as differentially expressed gene (down-regulated) and marker gene.
In a second step for the identification of an optimal predictive gene signature in vitro chemosensitivity assays of pri- mary tumors are performed to determine the response of a tumor towards a single chemotherapeutic agent. In said in vitro chemosensitivity assays the primary tumors were cultivated in different assays with increasing concentrations of the agents. After 6 days of incubation the vitality of the tumor cells were determined with an ATP-measurement . Hereby, the growing inhibition for the different agent concentrations could be determined and a dose-response curve could be pro- vided. For each tumor the Area under the dose-response curve (AUC) could be determined for the different agents. The AUC is used to evaluate the response of a tumor towards a chemo- therapeutic agent. The bigger the AUC, the more sensitive is the tumor towards the agent. The tumor samples were classi- fied according to their sensitivity towards the agents into three classes (resistant, intermediate, sensitive) via the tertiles of the AUC arrangement.
For the expression analyses isolated RNA from the tumor tis- sues was used for molecular profiling with microarrays . Unsu- pervised hierarchical clustering and principal component analysis identified the molecular subtypes.
In a quantitative diagnostic assay it is a common practice to use a cut-off score which distinguishes between two different test results (up-regulated expression and down-regulated expression) . The following cut off values relate to gene expression values determined by HG-U133a arrays of Affymetrix using MAS5.0 software with target intensity settings of 500.
Tumor tissues with an expression MLPH < 2000 (cut-off score) have been characterized as the basal molecular subtype. In case the expression of the genes ESRl and PGR were > 6000 and 160, respectively, the tumor tissues have been characterized as the subtype luminal A. The remaining tumor tissues have been divided into two different subtypes via the stromal gene COMP (cut-off score 300) .
The response of these molecular subtypes differs signifi- cantly towards single agents.
To improve separation between resistant and sensitive tumors for Epirubicin, Paclitaxel, 5-Fluorouracil and Carboplatin, additional biological motives were included in the analysis. Predictive gene signatures were defined for the single agents (Fig. 1 - 4) . See Table 2 for an overview regarding the different molecular subtypes and their predictive gene expression signatures.
Table 2: Overview regarding the different molecular subtypes and their predictive gene expression signatures
Figure imgf000043_0001
+ positive negative t up-regulated i down-regulated
In a preferred embodiment
a) the basal molecular subtype predictive for Epirubicin sensitivity is characterized by down-regulated MLPH- expression, the luminal A molecular subtype predictive for Epirubicin resistance is characterized by up- regulated ESRl and PGR expression, the immune system- high molecular subtype predictive for Epirubicin sensitivity is characterized by up-regulated IGKC and CCL5 expression, and/or the immune system-low molecular subtype predictive for Epirubicin resistance is characterized by down-regulated IGKC and CCL5 expression;
b) the basal molecular subtype predictive for Paclitaxel sensitivity is characterized by down-regulated MLPH expression, the luminal A molecular subtype predictive for Paclitaxel resistance is characterized by up- regulated ESRl and PGR expression, the stromal-low molecular subtype predictive for Paclitaxel sensitivity is characterized by down-regulated DCN expression, and/or the stromal-high molecular subtype predictive for Paclitaxel resistance is characterized by up- regulated DCN expression;
c) the stromal-low molecular subtype predictive for _5^ Fluorouracil sensitivity is characterized by down- regulated FBNl expression, and/or the stromal-high mo- lecular subtype predictive for 5-Fluorouracil resistance is characterized by up-regulated FBNl expression; and/or
d) the stromal/proliferation-low molecular subtype pre- dictive for Carboplatin sensitivity is characterized by down-regulated FBN1/UBE2C expression ratio, and/or the stromal/proliferation-high molecular subtype predictive for Carboplatin resistance is characterized by up-regulated FBN1/UBE2C expression ratio.
Since the tumors of the basal and luminal A subtype are particularly sensitive and resistant, respectively, towards the chemotherapeutic agents Epirubicin and Paclitaxel, the marker genes MLPH and ESRl and PGR, respectively, are used for the prediction towards said agents.
In favor of the stromal subtypes the marker gene DCN is used for the prediction towards Paclitaxel (p=0.0188) (Fig. 2) . For the agent Epirubicin the tumors, which can not be classified as luminal A and basal subtypes, are divided via the expression average of the immune system genes IGKC (normalized B-cell) and CCL5 (normalized T-cell) into a resistant and sensitive group (p=0.0341) (Fig. 1) . In case of signal intensities obtained by MAS5.0 software from HG-U133a arrays using scaling to a target intensity of 500, IGKC values are divided by 2338 and CCL5 values are divided by 399.5, corresponding to their respective median expression values in a reference cohort. In order to get an immune system score, both normalized values are added and divided by two.
For 5-Fluorouracil all tumors are divided into two groups via the stromal gene FBNl (p=0.0005) (Fig. 3) . And regarding Car- boplatin the proliferation gene UBE2C and the stromal gene FBNl are used for classification. Due to the inverse correlation of said motives regarding chemosensitivity the ratio between UBE2C and FBNl is calculated (Fig. 4) . The tumors with high expressed proliferation genes and with low expressed stromal genes differ significantly (p=0.0029) to the tumors with an inverse ratio.
The defined cut-off score for MLPH predictive for Epirubicin is 2000, the defined score for ESRl predictive for Epirubicin is 6000, the defined score for PGR predictive for Epirubicin is 160 and the defined score for the immune system score (average of IGKC and CCL5) predictive for Epirubicin is 1.5. An overview about the predictive gene (expression) signature for Epirubicin in breast cancer is demonstrated in Table 3.
Table 3: Predictive gene (expression) signature for Epirubicin in breast cancer
Figure imgf000045_0001
Figure imgf000046_0001
+ pos itive negative t up-regulated i down-regulated
The defined cut-off score for MLPH predictive for Paclitaxel is 2000, the defined score for ESRl predictive for Paclitaxel is 6000, the defined score for PGR predictive for Paclitaxel is 160 and the defined score for DCN predictive for Paclitaxel is 1500. An overview about the predictive gene (expression) signature for Paclitaxel in breast cancer is demonstrated in Table 4.
Table 4: Predictive gene (expression) signature for Paclitaxel in breast cancer
Figure imgf000046_0002
+ positive - negative t up-regulated i down-regulated
The defined cut-off score for FBNl predictive for 5- Fluorouracil is 3500. An overview about the predictive gene (expression) signature for 5-Fluorouracil in breast cancer is demonstrated in Table 5.
Table 5: Predictive gene (expression) signature for 5- Fluorouracil in breast cancer
Figure imgf000047_0001
+ positive negative t up-regulated I down-regulated
The defined cut-off score for the ratio between FBNl and UBE2C predictive for Carboplatin is 1. An overview about the predictive gene (expression) signature for Carboplatin in breast cancer is demonstrated in Table 6.
Table 6: Predictive gene (expression) signature for Car- boplatin in breast cancer
Figure imgf000047_0002
+ positive negative t up-regulated I down-regulated In another preferred embodiment of the present invention, Pa- clitaxel resistance in the basal molecular subtype is charac¬ terized by up-regulated AKRlCl expression.
It is particularly preferred that, in the method according to the invention, the said expression level is determined by
a) a hybridization based method; b) a PCR based method; c) determining the protein level, and/or by d) an array based method.
In yet another preferred embodiment of the present invention, it is provided that the expression level of at least one of the said genes is determined with rtPCR (reverse transcrip¬ tase polymerase chain reaction) of the gene related mRNA.
In another preferred embodiment of the present invention, it is provided that the expression level of at least one of the said genes is determined in formalin and/or paraffin fixed tissue samples.
Routinely, in tumor diagnosis tissue samples are taken as bi¬ opsies from a patient and undergo diagnostic procedures. For this purpose, the samples are fixed in formaline and/or par- rafine and are then examined with immunohistochemistry methods. The formaline treatment leads to the inactivation of en¬ zymes, as for example the ubiquitous RNA-digesting enzymes (RNAses) . For this reason, the mRNA status of the tissue (the so called transcriptome) , remains undigested.
However, the formaline treatment leads to partial depolymeri- zation of the individual mRNA molecules. For this reason, the current doctrine is that formaline fixed tissue samples can not be used for the analysis of the transcriptome of said tissue . For this reason, it is provided in a preferred embodiment of the present invention that after lysis, the samples are treated with silica-coated magnetic particles and a cha- otropic salt, in order to purify the nucleic acids contained in said sample for further determination.
Collaborators of the inventors of the present invention have developed an approach which however allows successful purification of mRNA out of tissue samples fixed in such manner, and which is disclosed, among others, in WO03058649,
WO2006136314A1 and DE10201084A1, the content of which is incorporated herein by reference.
Said method comprises the use of magnetic particles coated with silica (Siθ2) • The silica layer is closed and tight and is characterized by having an extremely small thickness on the scale of a few nanometers. These particles are produced by an improved method that leads to a product having a closed silica layer and thus entail a highly improved purity. The said method prevents an uncontrolled formation of aggregates and clusters of silicates on the magnetite surface whereby positively influencing the additional cited properties and biological applications. The said magnetic particles exhibit an optimized magnetization and suspension behavior as well as a very advantageous run-off behavior from plastic surfaces. These highly pure magnetic particles coated with silicon dioxide are used for isolating nucleic acids, including DNA and RNA, from cell and tissue samples, the separating out from a sample matrix ensuing by means of magnetic fields. These par- tides are particularly well-suited for the automatic purification of nucleic acids, mostly from biological body samples for the purpose of detecting them with different amplification methods.
The selective binding of these nucleic acids to the surface of said particles is due to the affinity of negatively charged nucleic acids to silica containing media in the presence of chaotropic salts like guanidinisothiocyanate . Said binding properties are known as the so called "boom principle". They are described in the European patent EP819696.
The said approach is particularly useful for the purification of mRNA out of formaline and/or paraffine fixed tissue samples. In contrast to most other approaches, which leave very small fragments behind that are not suitable for later determination by PCR and/or hybridization technologies, the said approach creates mRNA fragments which are large enough to al- low specific primer hybridzation and/or specific probe hybridization. A minimal size of at least 100 bp, more preferably 200 base pairs is needed for specific and robust detection of target gene expression. Moreover it is also necessary to not have too many inter-sample variations with regard to the size of the RNA fragments to guarantee comparability of gene expression results. Other issues of perturbance of expression data by sample preparation problems relate to the contamination level with DNA, which is lower compared to other bead based technologies. This of particular importance, as the inventors have observed, that DNAse treatment is not efficient in approximately 10% of FFPE samples generated by standard procedures and stored at room temperature for some years before cutting and RNA extraction.
The said approach thus allows a highly specific determination of candidate gene expression levels with one of the above introduced methods, particularly with hybridization based methods, PCR based methods and/or array based methods, even in formaline and/or paraffine fixed tissue samples, and is thus extremely beneficial in the context of the present invention, as it allows the use of tissue samples fixid with formaline and/or paraffine, which are available in tissue banks and connected to clinical databases of sufficient follow-up to allow retrospective analysis.
However, other methods are appropriate for the purification of mRNA out of formaline and/or paraffine fixed tissue samples as well. In a yet preferred embodiment of the present invention said gynecologic cancer is selected from the group comprising breast cancer, ovarian cancer, vulvar cancer, vaginal cancer, tubal cancer, endometrian cancer and/or cervical cancer.
In an especially preferred embodiment of the present invention said gynecologic cancer is breast cancer. The method according to the invention may be used for the analysis of a wide variety of neoplastic cell growth and proliferation of the breast tissues including, but not limited to ductal carcinoma in situ, lobular carcinoma, colloid carcinoma, tubular carcinoma, medullary carcinoma, metaplastic carcinoma, intraductal carcinoma in situ, lobular carcinoma in situ and pap- illary carcinoma in situ.
Furthermore, a kit useful for carrying out one of the said methods, comprising at least
a) a primer pair and/or a probe each having a sequence sufficiently complementary to a marker gene according to the present invention; and/or b) at least an antibody directed against a marker according to the present invention
is provided.
In yet another embodiment of the invention a method for correlating the clinical outcome of a patient suffering from or at risk of developing recurrent gynecologic cancer, preferably breast cancer, with the presence or non-presence of a defect in marker gene expression is provided, said method comprising the steps of:
a) obtaining a biological sample from said patient; b) determining the expression level of at least one marker gene according to the present invention in said patient, and c) correlating the pattern of expression levels determined in (b) with said patient's data, said data being selected from the group consisting of etiopathology data, clinical symptoms, anamnesis data and/or data concerning the therapeutic regimen.
In yet another embodiment of the invention a nucleic acid molecule is provided, said nucleic acid molecule selected from the group comprising
a) the nucleic acid molecule presented as SEQ ID NO: 1 - 30, b) a nucleic acid molecule having a length of 4 - 80 nucleotides, preferably 18 - 30 nucleotides, the sequence of which corresponds to the sequence of a single stranded fragment of a gene encoding for a marker from the group comprising MLPH, ESRl, PGR, COMP, DCN, IGKC, CCL5, FBNl, UBE2C and/or AKRlCl, c) a nucleic acid molecule that is a fraction, variant, homologue, derivative, or fragment of the nucleic acid molecule presented as SEQ ID NO: 1 - 30, d) a nucleic acid molecule that is capable of hybridiz- ing to any of the nucleic acid molecules of a) - c) under stringent conditions, e) a nucleic acid molecule that is capable of hybridizing to the complement of any of the nucleic acid molecules of a) - d) under stringent conditions, f) a nucleic acid molecule that is capable of hybridizing to the complement of a nucleic acid molecule of e), g) a nucleic acid molecule having a sequence identity of at least 95 % with any of the nucleic acid molecules of a) - f) , h) a nucleic acid molecule having a sequence identity of at least 70 % with any of the nucleic acid molecules of a) - f), i) a complement of any of the nucleic acid molecules of a) - h) , and/or j) a nucleic acid molecule that comprises any nucleic acid molecule of a) - i) .
Genes of interest are listed in Table 7, and the sequence listing is depicted in Table 8. These nucleic acids are being used either as primers for a polymerase chain reaction protocol, or as detectable probes for monitoring the said process.
Furthermore it is provided that the said nucleic acid is se- lected from the group comprising DNA, RNA, PNA, LNA and/or
Morpholino. The nucleic acid may, in a preferred embodiment, be labelled with at least one detectable marker. This feature is applicable particularly for those nucleic acids which serve as detectable probes for monitoring the polymerase chain reaction process
Such detectable markers may for example comprise at least one label selected from the group consisting of fluorescent molecules, luminescent molecules, radioactive molecules, enzy- matic molecules and/or quenching molecules.
In a particularly preferred embodiment, the said detectable probes are labeled with a fluorescent marker at one end and a quencher of fluorescence at the opposite end of the probe. The close proximity of the reporter to the quencher prevents detection of its fluorescence; breakdown of the probe by the 5 ' to 3 ' exonuclease activity of the taq polymerase breaks the reporter-quencher proximity and thus allows unquenched emission of fluorescence, which can be detected. An increase in the product targeted by the reporter probe at each PCR cycle therefore causes a proportional increase in fluorescence due to the breakdown of the probe and release of the reporter. In another preferred embodiment of the present invention, a kit of primers and/or detection probes is provided, comprising at least one of the nucleic acids according to the above enumeration and/or their fractions, variants, homologues, derivatives, fragments, complements, hybridizing counterparts, or molecules sharing a sequence identity of at least 70%, preferably 95 %.
Said kit may, in another preferred embodiment, comprise at least one of the nucleic acid molecules presented as SEQ ID NO: 1 - 30, and/or their fractions, variants, homologues, derivatives, fragments, complements, hybridizing counterparts, or molecules sharing a sequence identity of at least 70%, preferably 95 %, for the detection of at least one marker gene according to the present invention.
Furthermore, the use of a nucleic acid according as recited above, or of a kit as recited above for the prediction of a clinical response of a patient suffering from or at risk of developing recurrent gynecologic cancer, preferably breast cancer, towards a chemotherapeutic agent is provided.
Brief description of the examples and drawings
Additional details, features, characteristics and advantages of the object of the invention are disclosed in the sub- claims, and the following description of the respective fig- ures and examples, which, in an exemplary fashion, show preferred embodiments of the present invention. However, these drawings should by no means be understood as to limit the scope of the invention.
Example 1
The predictive gene signatures for the chemotherapeutic agents Epirubicin, 5-Fluorouracil and Paclitaxel have been validated in neoadjuvant studies via the defined cut-off scores. The Epirubicin prediction markers have been tested, to which extent they can predict the relative tumor reduction towards a neoadjuvant EC (Epirubin, Cyclophosphamid) combination therapy in 86 patients. Furthermore, the predictive genes for Epirubicin and 5-Fluorouracil were tested in a study, in which 39 patients received a neoadjuvant FEC (5- Fluorouracil, Epirubicin and Cyclophosphamid) therapy.
Via the predictive gene signature for Epirubicin the tumors of the neoadjuvant studies were divided into four prediction classes. Most of the tumors being sensitive towards the che- motherapeutic agents were classified as basal subtype and the class with a high expression of the immune system genes. The resistant tumors can be mainly found in the luminal A subtype and in the group with a low expressed immune system. In the EC study the four molecular prediction classes differ among each other significantly (p=0.0008) . In particular noticeable is the difference, which occurs for the classification with the immune system markers (p=0.0038) (Fig. 5) . In the classification of the FEC study with the Epirubicin prediction markers only the basal subtype and the group with the low expression of the immune system markers diverged significantly (p=0.0207) . In contrast, between the two immune system subtypes there is a trend to statistical significance (p=0.0848) (Fig. 6) . The 5-Fluorouracil prediction markers divide the tumors into two prediction classes, which differ in regard to the tumor reduction with a trend to statistical significance (Fig. 7) .
For the validation of the predictive Paclitaxel gene signatures two different neoadjuvant studies could be used, wherein the clinical response towards TAC (Rody et al . , 2006) and T-FAC combination (Hess et al . , 2006) was analyzed. Since no data have been provided for the percental tumor reduction, it could be only performed a classification via the provided clinical information. For each prediction class of the Paclitaxel prediction markers the percentage of the (pathological) complete and incomplete remission (pCR/CR) , respectively, has been determined.
In the neoadjuvant TAC study with the Paclitaxel marker genes all patients with a complete remission (in each case the bar on the left side) were classified into the basal subtype and the subtype with low expression of the stromal motive, respectively (Fig. 8) . In the second validation study most of the tumors showing a pathological complete remission (in each case the bar on the left side) towards a T-FAC combination therapy have been classified into these two subtypes as well. A little percentage has been classified into the subtype with a high expression of the stromal genes (Fig. 9) .
Discussion
The inventors of the present invention described for the first time methods for predicting the response of a tumor in a patient suffering from or at risk of developing recurrent breast cancer towards single chemotherapeutic agents.
Figures 10 to 13 present an overview via a general classification schema using the preferred marker genes. Cut off values are derived after expression analysis using HG-U133a ar- rays and MAS5.0 software using global scaling and target intensity settings of TGT = 500. In case two or more genes are used for a classification step, the expression values of the individual genes might be combined by first normalising each individual value and then combining the normalised values into a single meta-value. For example, the ESRl expression value might be divided by e.g. 5000, the PGR expression value by e.g. 150, and the normalised values are added to a combined ESRl-PGR score. The expression value of IGKC (214669_s_at) might be divided by e.g. 2338, the value of CCL5 might be divided by e.g. 399 and both normalised values might then be added and subsequently divided by two in order to obtain an immune system metagen score. In addition, the ratio between stromal and proliferation might be deduced from normalised expression values of FBNl and UBE2C by e.g. dividing the FBNl value by e.g. 3366 and dividing the value of UBE2C by 973 and subsequently calculating a ratio between the normalised values. The constant used for normalisation are typically derived as the median expression value of that gene found in samples collected from a representative patient cohort. A representative patient cohort is a population of breast cancer patient of a) sufficient size, e.g. preferentially more than 30 breast cancer patients and b) preferen- tially containing a proportion of grade 1, 2, 3, as well as estrogen receptor positive and estrogen receptor negative tumors of at least 5 %.
However, the TAC study and the T-FAC study differ too much compared to the in vitro study. Therefore, the Paclitaxel prediction markers should be analyzed in further studies.
The results of the validations demonstrate that, although the validation studies included complex agent combinations and the effect of the other agents could not be analyzed, the predictive gene signatures for the single agents can classify sensitive and resistant tumors in a neoadjuvant combination therapy as well.
The question, which meaning the identified signatures might have for the clinical practice, has to be analyzed first in larger studies.
Example 2 To prove the effect of the expression of AKRlCl in cell lines, the study NCI 60 (Staunton et al . , 2001) 22 was used. In this study several tumor cell lines of different cancer types were tested for their sensitivity towards several che- motherapeutic agents. The GI-50 value (concentration, where half of the tumor cells are inhibited regarding growing) for Paclitaxel was used to test, whether AKRlCl is appropriate for a prediction of a Paclitaxel sensitivity in cell lines. The analysis demonstrates that in case of a high expression of AKRlCl the cell lines mainly are resistant towards the agent Paclitaxel. The defined cut-off score (5000) allowed a significant classification (p=0.0049) of the resistant and sensitive tumor cells of the NCI 60 cell lines (Fig. 14) .
In primary breast cancer tissues it was analyzed, which effect AKRlCl has as predictive gene marker for Paclitaxel in the different subtypes defined in the in vitro study. Since in the in vitro study not enough tumor samples exist in the single subtypes to enable an appropriate analysis, the largest available validation dataset was used. The tumors of the neoadjuvant T-FAC study (Hess et al . , 2006) have been classified via the defined predictive gene signatures for Paclitaxel into four subtypes. Subsequently, it could be analyzed to what extent the AKRlCl gene expression in the subtypes differs in the patients, who received a complete remission due to the neoadjuvant therapy, compared to the rest.
Within the basal subtype tumors with higher expression of AKRlC tended to receive an incomplete remission (trend for significance p=0.09; Fig. 15) .
Discussion
Only for the basal subtype it was possible regarding the ex- pression of AKRlCl to differ between resistant and sensitive tumors with a significant trend. This observation should be verified in subsequent studies.
In case it is possible to identify with this gene for the basal subtype tumors, which do not respond to Paclitaxel, this would be very important for a therapy. Patients, whose tumors belong to the basal subtype, have very often a poor clinical prediction since this tumor entity is very aggressive. At the same time, this tumor subtype responds very strong towards chemotherapeutic agents. Therefore, especially for this subtype it is important to elucidate resistance mechanisms to improve the life span via an optimal treatment. Figures
Figures 1 - 4 demonstrate the chemosensitivity of the tumors towards the chemotherapeutic agents Epirubicin (Fig. 1), Pa- clitaxel (Fig. 2), 5-Fluorouracil (Fig. 3) and Carboplatin (Fig. 4) after classification with predictive gene signatures. The results are depicted via a Box-Whisker-Plot of the AUC (Area under the dose-response curve) of the prediction classes for Epirubicin (basal, immune system-high, immune system-low, luminal A) , Paclitaxel (basal, stromal-low, stro- mal-high, luminal A) , 5-Fluorouracil (stromal-low, stromal- high) and Carboplatin (ratio stromal/proliferation-low, ratio stromal/proliferation-high) (43 tumors for the agents Pacli- taxel/Epirubicin and 34 tumors for 5- Fluorouracil/Carboplatin) . Significant results of a nonpara- metric test are demonstrated for the comparison of all subtypes adjacent the Figures and for the pairwise comparison of the subtypes within the Figures (* : p-score < 0.05, **: p- score < 0.01 , ***: p-score < 0.001) (Tab. 9) .
Figure 5 demonstrates the relative reduction of 86 tumors towards a neoadjuvant EC combination therapy. The results are depicted via a Box-Whisker-Plot of the relative tumor reduction of the Epirubicin prediction classes (basal, immune system-high, immune system-low and luminal A) . Significant re- suits of a nonparametric test are demonstrated for the comparison of all subtypes adjacent the Figure and for the pair- wise comparison of the subtypes within the Figure (* : p- score < 0.05, **: p-score < 0.01 , ***: p-score < 0.001) (Tab. 9) .
Figures 6 and 7 demonstrate the relative reduction of 39 tumors towards a neoadjuvant FEC combination therapy. The results are depicted via a Box-Whisker-Plot of the relative tumor reduction of the prediction classes, which are defined via the Epirubicin (Fig. 6) and 5-Fluorouracil (Fig. 7) prediction markers. Significant results of a nonparametric test are demonstrated for the comparison of all subtypes adjacent the Figures and for the pairwise comparison of the subtypes within the Figures (* : p-score < 0.05) (Tab. 9) .
Figures 8 and 9 demonstrate prediction classes of the Pacli- taxel prediction markers in neoadjuvant studies. The percentage of the tumors with "(pathological) complete remission" (pCR/CR) (in each case the bar on the left side) and with incomplete remission (in each case the bar on the right side) in a neoadjuvant TAC (Fig. 8) and T-FAC (Fig. 9) study is demonstrated.
Figures 10 to 13 demonstrate the general classification schema using preferred marker genes for the different chemo- therapeutic agents.
Figure 14 demonstrates the chemosensitivity of different cell lines towards Paclitaxel. NCI 60 cell lines were classified with the marker gene AKRlCl (cut-off score 5000) . Onto the ordinate the concentration of Palitaxel is depicted, which is needed for the inhibition of half of the cells (GI-50) . Altogether for Paclitaxel 54 GI-50 values for the cell lines were provided.
Figure 15 demonstrates the expression of the gene AKRlCl in a neoadjuvant T-FAC study for the molecular subtypes basal, stromal-low, stromal-high and luminal A. For each group there occurred a classification of the tumors, which received a pathological complete remission (depicted in each case on the right side) or an incomplete remission (depicted in each case on the left side) towards the neoadjuvant therapy.
The figures are described in the context of the respective examples . Disclaimer
To provide a comprehensive disclosure without unduly lengthening the specification, the applicant hereby incorporates by reference each of the patents and patent applications referenced above .
The particular combinations of elements and features in the above detailed embodiments are exemplary only; the inter- changing and substitution of these teachings with other teachings in this and the patents/applications incorporated by reference are also expressly contemplated. As those skilled in the art will recognize, variations, modifications, and other implementations of what is described herein can oc- cur to those of ordinary skill in the art without departing from the spirit and the scope of the invention as claimed. Accordingly, the foregoing description is by way of example only and is not intended as limiting. The invention's scope is defined in the following claims and the equivalents thereto. Furthermore, reference signs used in the description and claims do not limit the scope of the invention as claimed.
Table 7 : Genes of Interest
Figure imgf000061_0001
Figure imgf000062_0001
Table 8 : Primer sequences and probe sequences used in accor¬ dance with the present invention
Figure imgf000062_0002
Figure imgf000063_0001
Table 9: Molecular subtypes
Epirubi- Pacli- 5^ Carbopla- cin taxel Fluoroura- tin cil p-score p-score p-score p-score
Kruskal-Wallis test 0.002* 0.002* 0.039* 0.025* basal vs . stromal 0.023* 0.149 0.054 0.281
(low) stromal (high) vs . 0.011* 0.054 0.004* 0.076 stromal (low) stromal (high) vs . 0.005* 0.075 0.511 0.313 luminal . A basal vs . luminal A 0, 002* 0.004* 1.000 0.101 basal vs . stromal 0.705 0.006* 0.342 0.007*
(high) stromal (low) vs 0 . 365 0 . 024 * 0 . 282 0 . 345 luminal A
Epirubicin predicin vitro EC- FEC- tor : study validation validation p-score p-score p-score
Kruskal-Wallis test 0.0028* 0.0008* 0.0343* basal vs . immune 0.7715 0.4025 0.1672
(high) immune (high) vs. 0.0341* 0.0038* 0.0848 immune (low) immune (low) vs. 0.4758 0.1368 0.5657 luminal A basal vs . luminal A 0.0022* 0.0390* 0.1333 basal vs . immune 0.0327* 0.0029* 0.0207*
(low) immune (high) vs. 0.0015* 0.0328^ 0.1455 luminal A
Paclitaxel predicin vitro tor: study p-score
Kruskal-Wallis 0.0014* test basal vs . stromal 0.0759
(low) stromal (high) vs 0.0188* stromal (low) stromal (high) vs 0.0962 luminal A basal vs . luminal 0.0037*
A basal vs . stromal 0.0066*
(high) stromal (low) vs. 0.0241* luminal A 5-Fluorouracil in vitro FEC- predictor : study validation p-score p-score stromal (low) vs 0.0005* 0.0837 stromal (high)
Carboplatin prein vitro dictor : study p-score stromal (low) vs , 0.0029* stromal (high)
cell culture CaI 51 HCC 1954 MCF 7 study: p-score p-score p-score
AKRlCl resistant 0.004* 0.001* 0.016* sensitive :
significant results (p<0.05) are marked with an asterisk*
References
1. Jemal A et al . , CA Cancer J Clin 2007; 57:43-66.
2. Veronesi U et al . , Lancet 2005; 365:1727-1741. 3. Gesellschaft der epidemiologischen Krebsregister in Deutschland e.V. (GEKID) und RKI, Krebs in Deutschland 2006; 5.Auflage, Saarbrucken 2006.
4. Bast RC JR et al . , J Clin Oncol 2001; 19: 1865-1878.
5. Goldhirsch A et al . , Ann Oncol 2005; 16: 1569-83. 6. Longley DB et al . , Nat Rev Cancer 2003; 3: 330-338.
7. Capranico G et al . , Cancer Res 1989; 49: 2022-2027.
8. Misset JL et al . , J Clin Oncol 1996; 14: 1136-1145.
9. Nabholtz JM et al . , J Clin Oncol 1999; 17: 1413-1424.
10. Henderson IC et al . , J Clin Oncol 2003; 21: 976-83. 11. Bishop JF et al . , J Clin Oncol 1999; 17: 2355-64.
12. Parness J and Horwitz SB J Cell Biol 1981; 191: 479-87
13. Paridaens R et al . , J Clin Oncol 2000; 18: 724-733.
14. Villeneuve DJ et al . , Breast Cancer Res Treat 2006; 96:17-39. 15. Gyorffy B et al . , Int J Cancer 2006; 118:1699-712.
16. Ross DT and Perou CM, Dis Markers 2001; 17:99-109.
17. Ayers M et al . , J Clin Oncol 2004; 22:2284-2293.
18. Hess KR et al . , J Clin Oncol 2006; 24:4236-44.
19. Gianni L et al., J Clin Oncol 2005; 23:7265-77. 20. Thuerigen O et al . , J Clin Oncol 2006; 24:1839-45.
21. Rody A et al . , Zentralbl Gynakol 2006; 128: 76-81.
22. Staunton JE et al . , Proc Natl Acad Sci USA 2001; 98: 10787-92.

Claims

What is claimed is:
1. A method for predicting a response of a tumor in a patient suffering from or at risk of developing recurrent gynecologic cancer towards a chemotherapeutic agent, said method comprising the steps of:
a) obtaining a biological sample from said patient; b) determining the pattern of expression level of at least one gene of the group comprising AKRlCl, MLPH, ESRl, PGR, COMP, DCN, IGKC, CCL5, FBNl and/or UBE2C, or of genes coregulated therewith, in said sample; c) comparing the pattern of expression levels determined in (b) with one or several reference pattern (s) of ex- pression levels; d) identifying at least one marker gene; e) determining a molecular subtype for said sample on the basis of (d) ; and f) predicting from said molecular subtype response of a tumor for a chemotherapeutic agent,
wherein the molecular subtype is selected from the group comprising the subtypes basal, stromal-high, stromal-low, luminal A, immune system-high, immune system-low, prolifera- tion-high and/or proliferation-low.
2. The method according to claim 1, said method comprising the further step of:
g) selecting at least one chemotherapeutic agent which is predicted to be successful,
wherein the chemotherapeutic agent is selected from the group comprising Epirubicin, Paclitaxel, 5-Fluorouracil and/or Car- boplatin.
3. The method according to any one of the preceding claims, wherein a) the basal molecular subtype predictive for Epirubicin sensitivity is characterized by down-regulated MLPH- expression, the luminal A molecular subtype predictive for Epirubicin resistance is characterized by up- regulated ESRl and PGR expression, the immune system- high molecular subtype predictive for Epirubicin sensitivity is characterized by up-regulated IGKC and CCL5 expression, and/or the immune system-low molecu- lar subtype predictive for Epirubicin resistance is characterized by down-regulated IGKC and CCL5 expression;
b) the basal molecular subtype predictive for Paclitaxel sensitivity is characterized by down-regulated MLPH expression, the luminal A molecular subtype predictive for Paclitaxel resistance is characterized by up- regulated ESRl and PGR expression, the stromal-low molecular subtype predictive for Paclitaxel sensitivity is characterized by down-regulated DCN expression, and/or the stromal-high molecular subtype predictive for Paclitaxel resistance is characterized by up- regulated DCN expression;
c) the stromal-low molecular subtype predictive for 5- Fluorouracil sensitivity is characterized by down- regulated FBNl expression, and/or the stromal-high molecular subtype predictive for 5-Fluorouracil resistance is characterized by up-regulated FBNl expres- sion; and/or
d) the stromal/proliferation-low molecular subtype predictive for Carboplatin sensitivity is characterized by down-regulated FBN1/UBE2C expression ratio, and/or the stromal/proliferation-high molecular subtype predictive for Carboplatin resistance is characterized by up-regulated FBN1/UBE2C expression ratio.
4. The method according to any one of the preceding claims, wherein Paclitaxel resistance in the basal molecular sub¬ type is characterized by up-regulated AKRlCl expression.
5. The method according to any one of the preceding claims, wherein the expression level is determined by
a) a hybridization based method; b) a PCR based method; c) determining the protein level, and/or by d) an array based method.
6. The method to any one of the preceding claims, character¬ ized in that the expression level of at least one of the said genes is determined with rtPCR (reverse transcrip¬ tase polymerase chain reaction) of the gene related mRNA.
7. The method according to any one of the preceding claims, characterized in that the expression level of at least one of the said genes is determined in formalin and/or paraffin fixed tissue samples.
8. The method according to any one of the preceding claims, wherein, after lysis, the samples are treated with sil- ica-coated magnetic particles and a chaotropic salt, in order to purify the nucleic acids contained in said sam¬ ple for further determination.
9. The method according to any one of the aforementioned claims characterized in that said gynecologic cancer is selected from the group comprising breast cancer, ovarian cancer, vulvar cancer, vaginal cancer, tubal cancer, en- dometrian cancer and/or cervical cancer.
10. The method according to any one of the aforementioned claims characterized in that said gynecologic cancer is breast cancer.
11. A kit useful for carrying out a method of any one of the preceding claims, comprising at least
a) a primer pair and/or a probe each having a sequence sufficiently complementary to a marker gene according to any one of the preceding claims; and/or b) at least an antibody directed against a marker according to any one of the preceding claims.
12. A method for correlating the clinical outcome of a patient suffering from or at risk of developing recurrent gynecologic cancer with the presence or non-presence of a defect in marker gene expression, said method comprising the steps of:
a. obtaining a biological sample from said patient; b. determining the expression level of at least one marker gene according to any one of the preceding claims in said patient, and c. correlating the pattern of expression levels determined in (b) with said patient's data, said data being selected from the group consisting of etiopathology data, clinical symptoms, anamnesis data and/or data concerning the therapeutic regimen .
13. A nucleic acid molecule, selected from the group comprising
a) the nucleic acid molecule presented as SEQ ID NO: 1 - 30, b) a nucleic acid molecule having a length of 4 - 80 nucleotides, preferably 18 - 30 nucleotides, the se- quence of which corresponds to the sequence of a single stranded fragment of a gene encoding for a marker from the group comprising MLPH, ESRl, PGR, COMP, DCN, IGKC, CCL5, FBNl, UBE2C and/or AKRlCl, c) a nucleic acid molecule that is a fraction, variant, homologue, derivative, or fragment of the nucleic acid molecule presented as SEQ ID NO: 1 - 30, d) a nucleic acid molecule that is capable of hybridizing to any of the nucleic acid molecules of a) - c) under stringent conditions, e) a nucleic acid molecule that is capable of hybridiz- ing to the complement of any of the nucleic acid molecules of a) - d) under stringent conditions, f) a nucleic acid molecule that is capable of hybridizing to the complement of a nucleic acid molecule of e), g) a nucleic acid molecule having a sequence identity of at least 95 % with any of the nucleic acid molecules of a) - f), h) a nucleic acid molecule having a sequence identity of at least 70 % with any of the nucleic acid molecules of a) - f) , i) a complement of any of the nucleic acid molecules of a) - h) , and/or j) a nucleic acid molecule that comprises any nucleic acid molecule of a) - i) .
14. The nucleic acid according to claim 13, characterized in that the said nucleic acid is selected from the group comprising DNA, RNA, PNA, LNA and/or Morpholino.
15. The nucleic acid according to any of claims 13 - 14, characterized in that it is labelled with at least one detectable marker.
16. A kit of primers and/or detection probes, comprising at least one of the nucleic acids according to any of claims 13 - 15 and/or their fractions, variants, homologues, derivatives, fragments, complements, hybridizing counter- parts, or molecules sharing a sequence identity of at least 70%, preferably 95 %.
17. The kit according to claim 16, comprising at least one of the nucleic acid molecules presented as SEQ ID NO: 1 - 30 and/or their fractions, variants, homologues, derivatives, fragments, complements, hybridizing counterparts, or molecules sharing a sequence identity of at least 70%, preferably 95 %, for the detection of at least one marker gene according to any one of the preceding claims.
18. Use of a nucleic acid according to any of claims 13 - 15 or of a kit according to any of claims 16 - 17 for predicting a clinical response of a patient suffering from or at risk of developing recurrent gynecologic cancer towards a chemotherapeutic agent.
PCT/EP2008/061088 2007-09-12 2008-08-25 A method for predicting the response of a tumor in a patient suffering from or at risk of developing recurrent gynecologic cancer towards a chemotherapeutic agent WO2009033941A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
EP08787449A EP2191015A1 (en) 2007-09-12 2008-08-25 A method for predicting the response of a tumor in a patient suffering from or at risk of developing recurrent gynecologic cancer towards a chemotherapeutic agent
US12/674,782 US20110143946A1 (en) 2007-09-12 2008-08-25 Method for predicting the response of a tumor in a patient suffering from or at risk of developing recurrent gynecologic cancer towards a chemotherapeutic agent

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
DE102007043455 2007-09-12
DE102007043455.5 2007-09-12
EP07122919A EP2036988A1 (en) 2007-09-12 2007-12-11 A method for predicting the response of a tumor in a patient suffering from or at risk of developing recurrent gynecologic cancer towards a chemotherapeutic agent
EP07122919.9 2007-12-11

Publications (1)

Publication Number Publication Date
WO2009033941A1 true WO2009033941A1 (en) 2009-03-19

Family

ID=39146845

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2008/061088 WO2009033941A1 (en) 2007-09-12 2008-08-25 A method for predicting the response of a tumor in a patient suffering from or at risk of developing recurrent gynecologic cancer towards a chemotherapeutic agent

Country Status (3)

Country Link
US (1) US20110143946A1 (en)
EP (3) EP2036988A1 (en)
WO (1) WO2009033941A1 (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2013532489A (en) * 2010-08-02 2013-08-19 ザ ブロード インスティテュート, インコーポレイテッド Prediction and monitoring of response to cancer treatment based on gene expression profiling
JP2017055769A (en) * 2011-11-08 2017-03-23 ジェノミック ヘルス, インコーポレイテッド Method for predicting breast cancer prognosis
KR20180063004A (en) * 2018-05-28 2018-06-11 의료법인 성광의료재단 A composition, and marker for diagnosing ovarian cancer comprising FOXA2

Families Citing this family (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN105821125A (en) 2010-03-31 2016-08-03 斯维丹诊断有限责任公司 Method for breast cancer recurrence prediction under endocrine treatment
GB201018377D0 (en) 2010-10-29 2010-12-15 Niendorf Axel Method for the analysis of a biological sample of a patient
ES2654469T3 (en) 2013-02-01 2018-02-13 Sividon Diagnostics Gmbh Procedure for predicting the benefit of the inclusion of taxane in a chemotherapy regimen in patients with breast cancer
WO2015114146A1 (en) * 2014-02-03 2015-08-06 Sividon Diagnostics Gmbh Method for predicting the response to an anti-her2 containing therapy and/or chemotherapy in patients with breast cancer
WO2015134548A1 (en) * 2014-03-05 2015-09-11 Sorrento Therapeutics, Inc. Methods for determining the maximum achievable response for a cancer treatment agent
ES2693035T3 (en) * 2014-04-01 2018-12-07 Miltenyi Biotec Gmbh SSEA4 and ST3GAL2 as biomarkers of response to chemotherapeutic drugs
EP3313993A4 (en) 2015-06-29 2019-06-19 Regents of the University of Minnesota Anti-apobec3 antibodies and methods of making and using
WO2017165629A1 (en) * 2016-03-23 2017-09-28 Regents Of The University Of Minnesota Methods of detecting apobec3 expression and predicting clinical outcomes
EP3679160A4 (en) 2017-09-08 2021-05-19 Myriad Genetics, Inc. Method of using biomarkers and clinical variables for predicting chemotherapy benefit

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005033699A2 (en) * 2003-10-03 2005-04-14 Ncc Technology Ventures Pte Limited Materials and methods relating to breast cancer classification

Family Cites Families (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
NL8900725A (en) 1989-03-23 1990-10-16 Az Univ Amsterdam METHOD AND COMBINATION OF AGENTS FOR INSULATING NUCLEIC ACID.
AU2002322280A1 (en) * 2001-06-21 2003-01-21 Millennium Pharmaceuticals, Inc. Compositions, kits, and methods for identification, assessment, prevention, and therapy of breast cancer
US20030124579A1 (en) * 2001-09-05 2003-07-03 Eos Biotechnology, Inc. Methods of diagnosis of ovarian cancer, compositions and methods of screening for modulators of ovarian cancer
DE10201084A1 (en) 2002-01-14 2003-07-24 Bayer Ag Magnetic particles containing silicon, process for their production and use of the particles
US7927795B2 (en) * 2003-06-09 2011-04-19 Board Of Trustees Of The University Of Arkansas Gene expression profiling in primary ovarian serous papillary tumors and normal ovarian epithelium
SG155968A1 (en) * 2004-02-23 2009-10-29 Univ Erasmus Medical Ct Classification, diagnosis and prognosis of acute myeloid leukemia by gene expression profiling
WO2006133287A2 (en) * 2005-06-06 2006-12-14 Wyeth Expression profiles of peripheral blood mononuclear cells for inflammatory bowel diseases
GB0512299D0 (en) * 2005-06-16 2005-07-27 Bayer Healthcare Ag Diagnosis prognosis and prediction of recurrence of breast cancer
US20100063263A1 (en) 2005-06-23 2010-03-11 Siemens Medical Solutions Diagnostics Gmbh Magnetic particles with a closed ultrathin silica layer, method for the production thereof and their use
DE602006015966D1 (en) * 2005-07-08 2010-09-16 Siemens Healthcare Diagnostics METHOD FOR PREDICTING AND MONITORING AN IMMEDIATE RESPONSE TO CANCER THERAPY
US8445198B2 (en) * 2005-12-01 2013-05-21 Medical Prognosis Institute Methods, kits and devices for identifying biomarkers of treatment response and use thereof to predict treatment efficacy
ATE530668T1 (en) * 2005-12-03 2011-11-15 Sividon Diagnostics Gmbh BREAST CANCER PROGNOSIS PROCEDURES AND KITS

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005033699A2 (en) * 2003-10-03 2005-04-14 Ncc Technology Ventures Pte Limited Materials and methods relating to breast cancer classification

Non-Patent Citations (7)

* Cited by examiner, † Cited by third party
Title
BRENTON JAMES D ET AL: "Molecular classification and molecular forecasting of breast cancer: ready for clinical application?", JOURNAL OF CLINICAL ONCOLOGY, GRUNE AND STRATTON, NEW YORK, NY, US, vol. 23, no. 29, October 2005 (2005-10-01), pages 7350 - 7360, XP009096987, ISSN: 0732-183X *
DRESSMAN HOLLY K ET AL: "Gene expression profiles of multiple breast cancer phenotypes and response to neoadjuvant chemotherapy.", CLINICAL CANCER RESEARCH : AN OFFICIAL JOURNAL OF THE AMERICAN ASSOCIATION FOR CANCER RESEARCH 1 FEB 2006, vol. 12, no. 3 Pt 1, 1 February 2006 (2006-02-01), pages 819 - 826, XP002472390, ISSN: 1078-0432 *
LEWIS MICHAEL J ET AL: "Expression of progesterone metabolizing enzyme genes (AKR1C1, AKR1C2, AKR1C3, SRD5A1, SRD5A2) is altered in human breast carcinoma", BMC CANCER, BIOMED CENTRAL, LONDON, GB, vol. 4, no. 1, 22 June 2004 (2004-06-22), pages 27, XP021004625, ISSN: 1471-2407 *
ROUZIER ROMAN ET AL: "Breast cancer molecular subtypes respond differently to preoperative chemotherapy.", CLINICAL CANCER RESEARCH : AN OFFICIAL JOURNAL OF THE AMERICAN ASSOCIATION FOR CANCER RESEARCH 15 AUG 2005, vol. 11, no. 16, 15 August 2005 (2005-08-15), pages 5678 - 5685, XP002472391, ISSN: 1078-0432 *
SEEBACH J ET AL: "A Molecular Profiling of Breast Cancer", SEMINARS IN BREAST DISEASE, SAUNDERS, PHILADELPHIA, PA, US, vol. 9, no. 1, 2006, pages 19 - 24, XP009096977, ISSN: 1092-4450 *
SORLIE T: "Molecular portraits of breast cancer: tumour subtypes as distinct disease entities", EUROPEAN JOURNAL OF CANCER, PERGAMON PRESS, OXFORD, GB, vol. 40, no. 18, December 2004 (2004-12-01), pages 2667 - 2675, XP004661394, ISSN: 0959-8049 *
WIEBE JOHN P ET AL: "Activity and expression of progesterone metabolizing 5[alpha]-reductase, 20[alpha]-hydroxysteroid oxidoreductase and 3[alpha]([beta])-hydroxysteroid oxidoreductases in tumorigenic (MCF-7, MDA-MB-231, T-47D) and nontumorigenic (MCF-10A) human breast cancer cells", BMC CANCER, BIOMED CENTRAL, LONDON, GB, vol. 3, no. 1, 22 March 2003 (2003-03-22), pages 9, XP021016085, ISSN: 1471-2407 *

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2013532489A (en) * 2010-08-02 2013-08-19 ザ ブロード インスティテュート, インコーポレイテッド Prediction and monitoring of response to cancer treatment based on gene expression profiling
JP2017055769A (en) * 2011-11-08 2017-03-23 ジェノミック ヘルス, インコーポレイテッド Method for predicting breast cancer prognosis
KR20180063004A (en) * 2018-05-28 2018-06-11 의료법인 성광의료재단 A composition, and marker for diagnosing ovarian cancer comprising FOXA2
KR101890387B1 (en) 2018-05-28 2018-08-21 의료법인 성광의료재단 A composition, and marker for diagnosing ovarian cancer comprising FOXA2

Also Published As

Publication number Publication date
EP2390370A3 (en) 2012-04-25
EP2036988A1 (en) 2009-03-18
EP2390370A2 (en) 2011-11-30
US20110143946A1 (en) 2011-06-16
EP2390370B1 (en) 2017-03-29
EP2191015A1 (en) 2010-06-02

Similar Documents

Publication Publication Date Title
EP2390370B1 (en) A method for predicting the response of a tumor in a patient suffering from or at risk of developing recurrent gynecologic cancer towards a chemotherapeutic agent
US11913078B2 (en) Method for breast cancer recurrence prediction under endocrine treatment
US20080305473A1 (en) Propagation of primary cells
US20180251854A1 (en) Method For Predicting Therapy Responsiveness In Basal Like Tumors
CA2504403A1 (en) Prognostic for hematological malignancy
US20160355889A1 (en) Method to assess prognosis and to predict therapeutic success in cancer by determining hormone receptor expression levels
ZA200305097B (en) Lamin alpha 4 subunit as a diagnostic indicator of malignant tumors.
WO2011068841A1 (en) Biological pathways associated with chemotherapy treatment in breast cancer
WO2009068409A1 (en) A method to assess prognosis and to predict therapeutic response to endocrine treatment
EP2065473A1 (en) A method to assess prognosis and to predict therapeutic success in gynecologic cancer
WO2010003772A1 (en) Method for predicting adverse response to erythropoietin in breast cancer treatment
US20240229150A1 (en) Method for breast cancer recurrence prediction under endocrine treatment
EP3426797A1 (en) Method for determining the risk of recurrence of an estrogen receptor-positive and her2-negative primary mammary carcinoma under an endocrine therapy
JP2023142853A (en) Method for determining risk of recurrence or progression to muscle invasive bladder cancer of non-muscle invasive bladder cancer, and marker gene used for the same

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 08787449

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 2008787449

Country of ref document: EP

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 12674782

Country of ref document: US