WO2015078906A1 - A method for predicting responsiveness to a treatment with an egfr inhibitor - Google Patents

A method for predicting responsiveness to a treatment with an egfr inhibitor Download PDF

Info

Publication number
WO2015078906A1
WO2015078906A1 PCT/EP2014/075651 EP2014075651W WO2015078906A1 WO 2015078906 A1 WO2015078906 A1 WO 2015078906A1 EP 2014075651 W EP2014075651 W EP 2014075651W WO 2015078906 A1 WO2015078906 A1 WO 2015078906A1
Authority
WO
WIPO (PCT)
Prior art keywords
patient
egfr
cancer
hsa
egfr inhibitor
Prior art date
Application number
PCT/EP2014/075651
Other languages
French (fr)
Inventor
Raphaële THIEBAUT
Original Assignee
Integragen
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 Integragen filed Critical Integragen
Priority to AU2014356506A priority Critical patent/AU2014356506A1/en
Priority to CA2931176A priority patent/CA2931176A1/en
Priority to JP2016534255A priority patent/JP2017503478A/en
Priority to KR1020167016884A priority patent/KR20160089488A/en
Priority to CN201480064646.4A priority patent/CN105765081A/en
Priority to MX2016006782A priority patent/MX2016006782A/en
Priority to EP14805833.2A priority patent/EP3074530A1/en
Priority to US15/038,826 priority patent/US20160376661A1/en
Priority to BR112016012001A priority patent/BR112016012001A2/en
Publication of WO2015078906A1 publication Critical patent/WO2015078906A1/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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • 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
    • C12Q2600/158Expression markers
    • 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
    • C12Q2600/178Oligonucleotides characterized by their use miRNA, siRNA or ncRNA

Definitions

  • the present invention provides methods for individualizing chemotherapy for cancer treatment, and particularly for evaluating a patient's responsiveness to one or more epidermal growth factor receptor (EGFR) inhibitors prior to treatment with such agents, based on the determination of the expression level of at least one target gene of hsa- mi ' R-31 -3p (SEQ ID NO: 1 ) miRNA, wherein said target gene of hsa-mi ' R-31 -3p is selected from DBNDD2 and EPB41 L4B.
  • EGFR epidermal growth factor receptor
  • the epidermal growth factor receptor (EGFR) pathway is crucial in the development and progression of human epithelial cancers.
  • the combined treatment with EGFR inhibitors has a synergistic growth inhibitory and pro-apoptotic activity in different human cancer cells which possess a functional EGFR-dependent autocrine growth pathway through to a more efficient and sustained inhibition of Akt.
  • EGFR inhibitors have been approved or tested for treatment of a variety of cancers, including non-small cell lung cancer (NSCLC) , head and neck cancer, colorectal carcinoma, and Her2-positive breast cancer, and are increasingly being added to standard therapy.
  • NSCLC non-small cell lung cancer
  • EGFR inhibitors which may target either the intracellular tyrosine kinase domain or the extracellular domain of the EGFR target, are generally plagued by low population response rates, leading to ineffective or non-optimal chemotherapy in many instances, as well as unnecessary drug toxicity and expense.
  • a reported clinical response rate for treatment of colorectal carcinoma with cetuximab is about 1 1 % (Cunningham et al, N Engl Med 2004;351 : 337-45), and a reported clinical response rate for treatment of NSCLC with erlotinib is about 8.9% (Shepherd F A, et al, N Engl J Med 2005; 353 : 123-132) .
  • PCT/EP2012/073535 describes an in vitro method for predicting whether a patient with a cancer is likely to respond to an epidermal growth factor receptor (EGFR)inhibitor, which comprises determining the expression level of hsa-mi ' R-31 -3p (previously named hsa-miR-31 * , SEQ ID NO:1 ) miRNA in a sample of said patient. More particularly, the lower the expression of hsa-miR-31 -3p is, the more likely the patient is to respond to the EGFR inhibitor treatment.
  • EGFR epidermal growth factor receptor
  • DBNDD2 (dysbindin (dystrobrevin binding protein 1 ) domain containing 2) has been disclosed to be a binding partner of human casein kinase-1 (Yin H et al. Biochemistry. 2006 Apr 25;45(16):5297-308).
  • DBNDD2 (dysbindin (dystrobrevin binding protein 1 ) domain containing 2) has been disclosed to be a binding partner of human casein kinase-1 (Yin H et al. Biochemistry. 2006 Apr 25;45(16):5297-308).
  • WO2010065940 WO2010059742 ; WO2009131710 ; WO2007112097
  • cancer cells sensitive or resistant torapamycin WO2011017106
  • tamoxifen (WO2010127338).
  • this gene does not seem to have been specifically associated to cancer, and no involvement of this gene in prediction of response to EGFR inhibitors has been disclosed.
  • EPB41 L4B erythrocyte membrane protein band 4.1 like 4B is a protein of the FERM family proteins, which can link transmembrane proteins to the cytoskeleton or link kinase and/or phosphatase enzymatic activity to the plasma membrane, and have been described to be involved in carcinogenesis and metastasis.
  • EPB41 L4B also known as EHM2
  • EHM2 has been associated to increased aggressiveness of prostate cancer
  • breast cancer Yama H et al.
  • Mol Cancer Res 2010;8: 1 501 - 1512 This gene has thus been associated to aggressiveness and poor prognosis of at least two types of cancer. Moreover, it has been found to be differentially expressed between cancer cell lines sensitive and resistant to taxotere (docetaxel, see WO2007072225 and WO2008138578). However, there has been no disclosure of its association to the ability of a cancer patient to respond or not to EGFR inhibitors.
  • the inventors implemented a new database incorporating information from the 6 databases, which may be interrogated either based on the name of a miRNA, or based on a gene name.
  • the database returns genes names considered as candidate targets of the queried miRNA, based on published or structural information, candidate target genes being ranked from the most probable to the less probable based on available information.
  • the database returns candidates miRNAs, for which the queried gene might (or not) be a target.
  • hsa-mi ' R-31 -3p With the aim to understand why increased expression of hsa-mi ' R-31 -3p is associated to lower response to EGFR inhibitor treatment, the inventors tried to identify target genes of this miRNA. For this purpose, they transfected three colorectal adenocarcinoma (CRC) cell lines that naturally weakly express hsa-miR-31 -3p with a mimic of hsa-miR- 31 -3p or a negative control mimic and analyzed genes differentially expressed between cell lines overexpressing or expressing weakly hsa-miR-31 -3p. A total of 74genes significantly down- or up-regulated was identified.
  • CRC colorectal adenocarcinoma
  • miRNAs function mainly by decreasing expression of their target genes, the inventors focused on the 47 down- regulated genes. To limit the number of candidate targets and avoid the false direct target genes, the inventors further performed in silico analyses based on information available in 6 databases relating to miRNAs and candidate targets. It is important to note that, most miRNA target genes provided in public databases are not validated, but only more or less probable candidates, based on structural or fragmental experimental data. 25 candidate target genes of hsa-miR-31 -3p were selected for further analysis on this basis. The inventors further analyzed the expression of these candidate target genes of hsa-mi ' R-31 -3p in tumor samples of patients treated with EGFR inhibitors, whose treatment response status based on RECIST criteria were known.
  • DBNDD2 and EPB41 L4B are both hsa-miR-31 -3p target genes, since their expression is significantly down- regulated by overexpression of hsa-miR-31 -3p in cancer cell lines, and that each of these genes is independently significantly associated to the ability of cancer patients to respond to EGFR inhibitor treatment. They further confirmed that each of these genes may alone be used for reliably predicting response to EGFR inhibitors in cancer patients.
  • the two genes found to be significantly down-regulated in patients not responding to EGFR inhibitor treatment are a gene not specifically known to be associated to cancer (DBNDD2) and a gene known to be associated to cancer (EPB41 L4B), but for which high expression level was associated to poor prognosis.
  • DBNDD2 a gene not specifically known to be associated to cancer
  • EPB41 L4B a gene known to be associated to cancer
  • the present invention provides an in vitro method for predicting whether a patient with a cancer is likely to respond to an epidermal growth factor receptor (EGFR) inhibitor, which comprises determining the expression level of at least one target gene of hsa-miR-31 -3p (SEQ ID NO:1 ) miRNA in a sample of said patient, wherein said target gene of hsa-miR-31 -3p is selected from DBNDD2 and EPB41 L4B.
  • EGFR epidermal growth factor receptor
  • the patient has a KRAS wild-type cancer.
  • the cancer preferably is a colorectal cancer, preferably a metastatic colorectal cancer.
  • the invention provides an in vitro method for predicting whether a patient with a metastatic colorectal carcinoma is likely to respond to an epidermal growth factor receptor (EGFR) inhibitor, such as cetuximab or panitumumab, which method comprises determining the expression level of at least one target gene of hsa-miR-31 -3p (SEQ ID N0:1 ) miRNA in a tumor sample of said patient, wherein said target gene of hsa-miR-31 -3p is selected from DBNDD2 and EPB41 L4B.
  • EGFR epidermal growth factor receptor
  • the invention also provides a kit for determining whether a patient with a cancer is likely to respond to an epidermal growth factor receptor (EGFR) inhibitor, comprising or consisting of: reagents for determining the expression level of at least one target gene of hsa-miR-31 -3p (SEQ ID NO:1 ) miRNA in a sample of said patient, wherein said target gene of hsa-miR-31 -3p is selected from DBNDD2 and EPB41 L4B, and reagents for determining at least one other parameter positively or negatively correlated to response to EGFR inhibitors.
  • EGFR epidermal growth factor receptor
  • the invention further relates to an EGFR inhibitor for use in treating a patient affected with a cancer, wherein the patient has been classified as being likely to respond, by the method according to the invention.
  • the invention also relates to the use of an EGFR inhibitor for the preparation of a drug intended for use in the treatment of cancer in patients that have been classified as "responder" by the method of the invention.
  • the invention also relates to a method for treating a patient affected with a cancer, which method comprises (i) determining whether the patient is likely to respond to an EGFR inhibitor, by the method of the invention, and (ii) administering an EGFR inhibitor to said patient if the patient has been determined to be likely to respond to the EGFR inhibitor.
  • FIGURES Figure 1 Correlation between log 2 expression levels of DBNDD2 (in Figure 1A) and EPB41 L4B (in Figure 1B) and hsa-miR-31 -3p in the 20 mCRC patients of Example 1 .
  • Figure 2 Correlation between log 2 expression levels of DBNDD2 and hsa-miR-31 -3p in the 20 mCRC patients of Example 2.
  • Figure 3 In A: Nomogram tool established based on log 2 expression of DBNDD2 in the 20 mCRC patients of Example 2, in order to predict risk of progression (i.e. risk of non- response) of mCRC patients treated with anti-EGFR based chemotherapy.
  • Figure 4 Multivariate Cox proportional hazards models with DBNDD2 expression as covariate in the 20 mCRC patients of Example 2.
  • Figure 5 Correlation between log 2 expression levels of DBNDD2 (in Figure 5A) and EPB41 L4B (in Figure 5B) and hsa-miR-31 -3p in the 42 mCRC patients of Example 3.
  • Figure 6 Expression of DBNDD2 (in Figure 6A) and EPB41 L4B (in Figure 6B) in patients of Example 3 according to their risk of progression (low or high), as predicted based on hsa-miR-31 -3p expression level.
  • the "patient” may be any mammal, preferably a human being, whatever its age or sex.
  • the patient is afflicted with a cancer.
  • the patient may be already subjected to a treatment, by any chemotherapeutic agent, or may be untreated yet.
  • the cancer is preferably a cancer in which the signaling pathway through EGFR is involved.
  • it may be e.g. colorectal, lung, breast, ovarian, endometrial, thyroid, nasopharynx, prostate, head and neck, kidney, pancreas, bladder, or brain cancer (Ciardello F et al. N Engl J Med. 2008 Mar 13;358(11 ):1160-74 ; Wheeler DL et al. Nat RevClinOncol. 2010 September ; 7(9): 493-507 ; Zeineldin R et al. J Oncol. 2010;2010:414676;Albitar L et al. Mol Cancer 2010;9:166; Leslie KK et al.
  • the tumor is a solid tissue tumor and/or is epithelial in nature.
  • the patient may be a colorectal carcinoma patient, a Her2-positive or Her2- negative (in particular triple negative, i.e. Her2-negative, estrogen receptor negative and progesterone receptor negative) breast cancer patient, a non-small cell lung cancer (NSCLC) patient, a head and neck cancer patient (in particular a squamous-cell carcinoma of the head and neck patient), a pancreatic cancer patient, or an endometrial cancer patient.
  • NSCLC non-small cell lung cancer
  • the patient may be a colorectal carcinoma patient, a Her2-positive or Her2- negative (in particular triple negative) breast cancer patient, a lung cancer (in particular a NSCLC) patient, a head and neck cancer patient (in particular a squamous-cell carcinoma of the head and neck patient), or a pancreatic cancer patient.
  • a Her2-positive or Her2- negative (in particular triple negative) breast cancer patient a lung cancer (in particular a NSCLC) patient, a head and neck cancer patient (in particular a squamous-cell carcinoma of the head and neck patient), or a pancreatic cancer patient.
  • the cancer is a colorectal cancer, still preferably the cancer is a metastatic colorectal cancer.
  • DBNDD2 or EPB41 L4B expression level may be used as a predictor of response to EGFR inhibitors (and in particular to anti-EGFR monoclonal antibodies such as cetuximab and panitumumab) treatment in colorectal cancer.
  • the cancer is a Her2-positive or Her2- negative (in particular triple negative) breast cancer, preferably a Her2- negative (in particular triple negative) breast cancer.
  • the cancer is a lung cancer, in particular a non- small cell lung cancer (NSCLC).
  • NSCLC non- small cell lung cancer
  • the cancer is a pancreatic cancer.
  • the patient's tumor is preferably EGFR positive.
  • the patient has a KRAS wild-type tumor, i.e. , the KRAS gene in the tumor of the patient is not mutated in codon 12, 13 (exon 1 ), or 61 (exon 3) .
  • the KRAS gene is wild-type on codons 12, 13 and 61 .
  • Wild type, i.e. non mutated, codons 12, 13 (exon 1 ),and 61 (exon 3) respectively correspond to glycine (Gly, codon 12), glycine (Gly, codon 13), and glutamine (Gin, codon 61 ).
  • the wild-type reference KRAS amino acid sequence may be found in Genbank accession number NP_004976.2 (SEQ ID NO: 24).
  • Gly12Cys (GGT>TGT)
  • Gly12Asp (GGT>GAT)
  • the KRAS gene of the patient's tumor does also not show any of the following mutations (Demiralay et al. Surgical Science, 2012, 3, 111 -115):
  • the KRAS gene of the patient's tumor does also not show any of the following mutations (Bos. Cancer Res 1989;49:4682-4689; Tarn et al. Clin Cancer Res2006;12:1647-1653 ; Edkins et al. Cancer BiolTher. 2006 August ; 5(8): 928-932;
  • a tumor tissue is microdissected and DNA extracted from paraffin- embedded tissue blocks. Regions covering codons 12, 13, and 61 of the KRAS gene are amplified using polymerase chain reaction (PCR). Mutation status is determined by allelic discrimination using PCR probes (Laurent-Puig P, et al, J ClinOncol. 2009, 27(35) :5924-30) or by any other methods such as pyrosequencing (Ogino S, et al. J MolDiagn 2008;7:413-21 ).
  • PCR polymerase chain reaction
  • the “sample” may be any biological sample derived from a patient, which contains nucleic acids. Examples of such samples include fluids (including blood, plasma, saliva, urine, seminal fluid), tissues, cell samples, organs, biopsies, etc.
  • the sample is a tumor sample, preferably a tumor tissue biopsy or whole or part of a tumor surgical resection.
  • the sample may be collected according to conventional techniques and used directly for diagnosis or stored.
  • a tumor sample may be fresh, frozen or paraffin- embedded. Usually, available tumor samples are frozen or paraffin-embedded, most of the time paraffin-embedded. The inventors have shown that both frozen and paraffin- embedded tumor samples may be used.
  • a tumor sample notably a tumor biopsy or whole or part of a tumor surgical resection
  • a pool of reference samples comprises at least one (preferably several, more preferably at least 5, more preferably at least 6, at least 7, at least 8, at least 9, at least 10) responder patient(s) and at least one (preferably several, more preferably at least 6, at least 7, at least 8, at least 9, at least 10) non responder patient(s).
  • a patient who is "likely to respond” or is “responder” refers to a patient who may respond to a treatment with an EGFR inhibitor, i.e. at least one of his symptoms is expected to be alleviated, or the development of the disease is stopped, or slowed down.
  • Complete responders, partial responders, or stable patients according to the RECIST criteria are considered as "likely to respond” or "responder” in the context of the present invention.
  • the RECIST criteria are an international standard based on the presence of at least one measurable lesion.
  • “Complete response” means disappearance of all target lesions;
  • Partial response means 30% decrease in the sum of the longest diameter of target lesions,
  • progressive disease means 20% increase in the sum of the longest diameter of target lesions,
  • stable disease means changes that do not meet above criteria.
  • a "responder" patient is predicted to show a good progression free survival (PFS), i.e. the patient is likely to survive at least 25 weeks without aggravation of the symptoms of the disease, and/or such patient shows a good overall survival (OS), i.e. the patient is likely to survive at least 14 months.
  • PFS progression free survival
  • OS overall survival
  • predicting refers to a probability or likelihood for a patient to respond to the treatment with an EGFR inhibitor.
  • the sensitivity of tumor cell growth to inhibition by an EGFR inhibitor is predicted by whether and to which level such tumor cells express hsa-miR- 31 -3p target genes DBNDD2 and EPB41 L4B.
  • treating means stabilizing, alleviating, curing, or reducing the progression of the cancer.
  • a “miRNA” or “microRNA” is a single-stranded molecule of about 21 -24 nucleotides, preferably 21 -23 in length, encoded by genes that are transcribed from DNA but not translated into protein (non-coding RNA); instead they are processed from primary transcripts known as pri-miRNA to short stem-loop structures called pre-miRNA and finally to functional miRNA. During maturation, each pre-miRNA gives rise to two distinct fragments with high complementarity, one originating from the 5' arm the other originating from the 3' arm of the gene encoding the pri-miRNA. Mature miRNA molecules are partially complementary to one or more messenger RNA (mRNA) molecules, and their main function is to downregulate gene expression.
  • mRNA messenger RNA
  • miRNAs There is an international nomenclature of miRNAs (see Ambros V et al, RNA 2003 9(3):277-279 ; Griffiths-Jones S. NAR 2004 32(Database lssue):D109-D111 ; Griffiths- Jones S et al. NAR 2006 34(Database lssue):D140-D144; Griffiths- Jones S et al. NAR 2008 36(Database lssue) :D154-D158; and Kozomara A et al. NAR 2011 39(Database lssue):D152-D157), which is available from miRBase at http://www.mirbase.org/.
  • Each miRNA is assigned a unique name with a predefined format, as follows: • For a mature miRNA: sss-miR-X-Y, wherein "
  • o sss is a three letters code indicating the species of the miRNA, "hsa" standing for human,
  • o X is the unique arbitrary number assigned to the sequence of the miRNA in the particular species, which may be followed by a letter if several highly homologous miRNAs are known. For instance, "20a" and “20b” refer to highly homologous miRNAs.
  • o Y indicates whether the mature miRNA, which has been obtained by cutting of the pre-miRNA, corresponds to the 5' arm (Y is then "5p") or 3' arm (Y is then "3p") of the gene encoding the pri-mRNA.
  • "-Y" was not present.
  • the two mature miRNAs obtained either from the 5' or the 3' arm of the gene encoding the pri-miRNA were then distinguished by the presence or absence of a "*" sign just after n. The presence of the "*" sign indicated that the sequence corresponded to the less often detected miRNA. Since such classification was subject to changes, a new nomenclature using the
  • o sss is a three letters code indicating the species of the miRNA, "hsa" standing for human,
  • o n is the unique arbitrary number assigned to the sequence of the miRNA in the particular species, which may be followed by a letter if several highly homologous miRNAs are known.
  • Each miRNA is also assigned an accession number for its sequence.
  • hsa-miR-31 -3p The miRNA targeted by the two genes detected in the present invention (DBNDD2 and EPB41 L4B) is hsa-miR-31 -3p (previously named hsa-miR-31*).
  • hsa means that it relates to a human miRNA
  • miR refers to a mature miRNA
  • 31 refers to the arbitrary number assigned to this particular miRNA
  • 3p means that the mature miRNAs has been obtained from the 3' arm of the gene encoding the pri-miRNA.
  • hsa-miR-31 -3p is UGCUAUGCCAACAUAUUGCCAU(SEQ ID NO : 1 )
  • DNDD2 is the official symbol of NCBI Entrez Gene database for "dysbindin (dystrobrevin binding protein 1 ) domain containing 2" gene (official name and symbol approved by the HUGO Gene Nomenclature Committee (HGNC)), located in humans in chromosome 20 (20q13.12). It corresponds to UniGene database accession number Hs.730643. Further symbols used for this gene include CK1 BP (for "casein kinase-1 binding protein”), HSMNP1 , RP3-453C12.9, and C20orf35. It is also known as "SCF apoptosis response protein 1 ". Five isoforms (a to e) of this protein are known, encoded by several mRNA variants, as detailed in Table 1 below.
  • Table 1 isoforms of DBNDD2 and corresponding mRNA and protein reference sequences provided by NCBI EntrezGene database, on July 1 , 2013.
  • EPB41 L4B is the official symbol of NCBI Entrez Gene database for "erythrocyte membrane protein band 4.1 like 4B" gene (official name and symbol approved by the HGNC), located in humans in chromosome 9 (9q31 -q32). It corresponds to UniGene database accession number Hs.591901 . Further symbols used for this gene include CG1 and EHM2 (for “Expressed in Highly Metastatic cells 2"). It is also known as “FERM- containing protein CG1 ". Two isoforms (1 and 2) of this protein are known, encoded by two mRNA variants, as detailed in Table 2 below.
  • Table 2 isoforms of EPB41 L4Band corresponding mRNA and protein reference sequences provided by NCBI EntrezGene database, as updated on July 1 , 2013.
  • the expression level of hsa-miR-31 -3p target gene(s) DBNDD2 and/or EPB41 L4B may be determined by any technology known by a person skilled in the art.
  • each gene expression level may be measured in vitro, starting from the patient's sample, at the genomic and/or nucleic acid and/or proteic level.
  • the expression profile is determined by measuring in vitro the amount of nucleic acid transcripts of each gene.
  • the expression profile is determined by measuring in vitro the amount of protein produced by each of the genes.
  • Such measures are made in vitro, starting from a patient's sample, in particular a tumor sample, and necessary involve transformation of the sample. Indeed, no measure of a specific gene expression level can be made without some type of transformation of the sample.
  • the claimed method may thus also comprise a preliminary step of extracting DNA, mRNA or proteins from the patient's sample.
  • mRNAs when extracted, they are generally retrotranscribed into cDNA, which is more stable than mRNA.
  • the claimed methods may thus also comprise a step of retrotranscribing mRNA extracted from the patient's sample into cDNA.
  • Detection by mass spectrometry does not necessary involve preliminary binding to specific reagents. However, it is most of the time performed on extracted DNA, mRNA or proteins. Even when preformed directly on the sample, without preliminary extraction steps, it involves some extraction of molecules from the sample by the laser beam, which extracted molecules are then analysed by the spectrometer.
  • the amount of nucleic acid transcripts can be measured by any technology known by a person skilled in the art.
  • the measure may be carried out directly on an extracted messenger RNA (mRNA) sample, or on retrotranscribed complementary DNA (cDNA) prepared from extracted mRNA by technologies well-known in the art.
  • mRNA messenger RNA
  • cDNA retrotranscribed complementary DNA
  • the amount of nucleic acid transcripts may be measured using any technology known by a person skilled in the art, including nucleic microarrays, quantitative PCR, next generation sequencing and hybridization with a labelled probe.
  • qRT-PCR real time quantitative RT-PCR
  • qRT-PCR can be used for both the detection and quantification of RNA targets (Bustin et al., 2005, Clin. Sci., 109:365-379). Quantitative results obtained by qRT-PCR can sometimes be more informative than qualitative data, and can simplify assay standardization and quality management. Thus, in some embodiments, qRT-PCR- based assays can be useful to measure hsa-miR-31 -3p target gene(s) DBNDD2 and/or EPB41 L4B expression levels during cell-based assays. The qRT-PCR method may be also useful in monitoring patient therapy. qRT-PCR is a well-known and easily available technology for those skilled in the art and does not need a precise description.
  • qRT-PCR-based methods can be found, for example, in U.S. Pat. No. 7,101 ,663.
  • Commercially available qRT-PCR based methods e.g. ,Taqman® Array
  • the design of primers and/or probe being easily made based on the sequences of DBNDD2 and/or EPB41 L4B disclosed in Tables 1 and 2 above.
  • Nucleic acid assays or arrays can also be used to assess in vitro the expression level of the gene in a sample, by measuring in vitro the amount of gene transcripts in a patient's sample.
  • a nucleic acid microarray can be prepared or purchased.
  • An array typically contains a solid support and at least one nucleic acid (cDNA or oligonucleotide) contacting the support, where the oligonucleotide corresponds to at least a portion of a gene.
  • Any suitable assay platform can be used to determine the presence of hsa-mi ' R-31 -3p target gene(s) DBNDD2 and/or EPB41 L4B in a sample.
  • an assay may be in the form of a membrane, a chip, a disk, a test strip, a filter, a microsphere, a multiwell plate, and the like.
  • An assay system may have a solid support on which a nucleic acid (cDNA or oligonucleotide) corresponding to the gene is attached.
  • the solid support may comprise, for example, a plastic, silicon, a metal, a resin, or a glass.
  • the assay components can be prepared and packaged together as a kit for detecting a gene.
  • a target nucleic sample is labelled, contacted with the microarray in hybridization conditions, leading to the formation of complexes between target nucleic acids that are complementary to probe sequences attached to the microarray surface. The presence of labelled hybridized complexes is then detected.
  • Many variants of the microarray hybridization technology are available to the person skilled in the art.
  • the measure in vitro of hsa-miR-31 -3p target gene(s) DBNDD2 and/or EPB41 L4B expression level(s) may be performed by sequencing of transcripts (mRNA or cDNA) of the gene extracted from the patient's sample.
  • the measure in vitro of hsa-miR-31 -3p target gene(s) DBNDD2 and/or EPB41 L4B expression level(s) may be performed by the use of a protein microarray, for measuring the amount of the gene encoded protein in total proteins extracted from the patient's sample.
  • Such a control value may be determined based on a pool of reference samples, as defined above.
  • Figure 6 clearly shows that, based on a pool of reference samples, a control value for DBNDD2 and EPB41 L4B level of expression (the logged DBNDD2:EPB41 L4B level of expression) may be defined that permits to predict response or non-response to EGFR inhibitor treatment.
  • the method further comprises determining a prognostic score or index based on the expression level of at least one o fhsa-mi ' R-31 -3p target gene(s) DBNDD2 and EPB41 L4B, wherein the prognostic score indicates whether the patient is likely to respond to the EGFR inhibitor.
  • said prognosis score may indicate whether the patient is likely to respond to the EGFR inhibitor depending if it is higher or lower than a predetermined threshold value (dichotomized result).
  • a discrete probability of response or non-response to the EGFR inhibitor may be derived from the prognosis score.
  • the probability that a patient responds to an EGFR inhibitor treatment is linked to the probability that this patient survives, with or without disease progression, if the EGFR inhibitor treatment is administered to said patient.
  • a prognosis score may be determined based on the analysis of the correlation between the expression level of at least one of hsa-miR-31 -3p target gene(s) DBNDD2 and EPB41 L4B and progression free survival (PFS) or overall survival (OS) of a pool of reference samples, as defined above.
  • PFS and/or OS score which is a function correlating PFS or OS to the expression level of at least one of hsa-miR-31 -3p target gene(s) DBNDD2 and EPB41 L4B, may thus be used as prognosis score for prediction of response to an EGFR inhibitor.
  • a PFS score is used, since absence of disease progression is a clear indicator of response to the EGFR inhibitor treatment.
  • Prognosis score a * x + b, wherein x is the logged expression level ofDBNDD2 (preferably log in base 2, referred to as "log 2 ") and/or EPB41 L4Bmeasured in the patient's sample, and a and b are parameters that have been previously determined based on a pool of reference samples, as defined above.
  • the patient may then be predicted as responding to the EGFR inhibitor if his/her prognosis score is greater than or equal to/lower than or equal to a threshold value c, and not responding to the EGFR inhibitor if his/her prognosis score is lower than/greater than threshold value c, wherein the value of c has also been determined based on the same pool of reference samples:
  • the patient may then be predicted as responding to the EGFR inhibitor if his/her prognosis score is greater than or equal to threshold value c, and not responding to the EGFR inhibitor if his/her prognosis score is lower than threshold value c.
  • the patient may be predicted as responding to the EGFR inhibitor if his/her prognosis score is lower than or equal to threshold value c, and not responding to the EGFR inhibitor if his/her prognosis score is greater than threshold value c.
  • a discrete probability of response or non-response to the EGFR inhibitor may be derived from the above a * x + b prognosis score.
  • a precise correlation between the prognosis score and the probability of response to the EGFR inhibitor treatment may be determined based on the same set of reference samples. Depending if a is positive/negative, a higher/lower prognosis score indicates a higher probability of response to the EGFR inhibitor treatment: • If a is positive, the higher the prognosis score, the higher is the probability of response to the EGFR inhibitor treatment (i.e. the lower is the probability of disease progression in the case of a PFS score).
  • This prediction of whether a patient with a cancer is likely to respond to an EGFR inhibitor may also be made using a nomogram.
  • a nomogram points scales are established for each variable of a score of interest. For a given patient, points are allocated to each of the variables by selecting the corresponding points from the points scale of each variable. For a discrete variable (such as a gene expression level), the number of points attributed to a variable is linearly correlated to the value of the variable. For a dichotomized variable (only two values possible), two distinct values are attributed to each of the two possible values or the variable. The score of interest is then calculated by adding the points allocated for each variable (total points).
  • the patient may then be given either a good or bad response prognosis depending on whether the composite score is inferior or superior to a threshold value (dichotomized score), or a probability of response or non-response to the treatment.
  • nomograms are mainly useful when several distinct variables are combined in a composite score (see below the possibility to use composite scores combining DBNDD2 and EPB41 L4B expression levels; DBNDD2 and/or EPB41 L4B expression levels and hsa-mi ' R-31 -3p expression level; or DBNDD2 and/or EPB41 L4B expression level(s) and BRAF status).
  • a nomogram may also be used to represent a prognosis score based on only one variable, such as DBNDD2 or EPB41 L4B expression level. In this case, total points correspond to points allocated to the single variable.
  • the method further comprises determining a risk of non-response based on a nomogram calibrated based on a pool of reference samples.
  • the nomogram may be calibrated based on OS or PFS data. If calibrated based on OS, the risk of non-response corresponds to a risk of death. If calibrated based on PFS, the risk of non-response corresponds to a risk of disease progression (see Figure 3).
  • each of DBNDD2 and EPB41 L4B has been found to be a target gene of hsa-miR-31 -3p and to be independently significantly associated to response to EGFR inhibitors, so that the expression level of only one of DBNDD2 and EPB41 L4B may be measured and used for prediction in a method according to the invention.
  • the method according to the invention may also comprise determining the expression levels of both DBNDD2 and EPB41 L4B in the patient's sample, and predicting response or non-response based on the combined expression of DBNDD2 and EPB41 L4B.
  • a composite score combining the expression levels of DBNDD2 and EPB41 L4B may notably be created based on a pool of reference samples.
  • a nomogram may also be used to combine the expression levels of DBNDD2 and EPB41 L4B and obtain the composite score, which may then be correlated to the risk of non-response (i.e. the risk of disease progression for a PFS score) .
  • response to EGFR inhibitors can be predicted based only on the expression level of at least one of hsa-miR-31 -3p target genes DBNDD2 and EPB41 L4B (see Examples 1 , 2 and 3)
  • the method according to the invention may also comprise determining at least one other parameter positively or negatively correlated to response to EGFR inhibitors.
  • a composite score combining the expression level(s) of DBNDD2 and/or EPB41 L4B and the other parameter(s) may notably be created based on a pool of reference samples.
  • a nomogram, in which points scales are established for each variable of the composite score may also be used to combine the expression level(s) of DBNDD2 and/or EPB41 L4B and the other parameter(s), and obtain the composite score, which may then be correlated to the risk of non-response (i.e. the risk of disease progression for a PFS score).
  • points are allocated to each of the variables by selecting the corresponding points from the points scale of each variable.
  • the number of points attributed to a variable is linearly correlated to the value of the variable.
  • a dichotomized variable only two values possible, such as BRAF mutation status or gender
  • two distinct values are attributed to each of the two possible values or the variable.
  • a composite score is then calculated by adding the points allocated for each variable (total points) . Based on the value of the composite score, the patient may then be given either a good or bad response prognosis depending on whether the composite score is inferior or superior to a threshold value (dichotomized score), or a probability of response or non-response to the treatment.
  • the points scale of each variable, as well the threshold value over/under which the response prognosis is good or bad or the correlation between the composite score and the probability of response or non-response may be determined based on the same pool of reference samples.
  • Such other parameters positively or negatively correlated to response to EGFR inhibitors may notably be selected from:
  • the expression level of hsa-miR-31 -3p which may be measured at the genomic and/or nucleic (in particular by measuring the amount of nucleic acid transcripts of each gene)and/or proteic level, by any method disclosed above for measuring the expression level of DBNDD2 and EPB41 L4B; and/or
  • Such mutations may be detected by any method known to those skilled in the art and notably include those mentioned in Table 3 below:
  • EGFR Inhibitors are defined by mention of the codon number in the protein, preceded by the one letter code for the wild-type amino acid, and optionally followed by the replacement amino acid. When no replacement amino acid is mentioned, the replacement amino acid may be any amino acid different from the wild-type amino acid.
  • EGFR Inhibitors are defined by mention of the codon number in the protein, preceded by the one letter code for the wild-type amino acid, and optionally followed by the replacement amino acid. When no replacement amino acid is mentioned, the replacement amino acid may be any amino acid different from the wild-type amino acid.
  • the present invention makes it possible to predict a patient's responsiveness to one or more epidermal growth factor receptor (EGFR) inhibitors prior to treatment with such agents.
  • EGFR epidermal growth factor receptor
  • the EGRF inhibitor may be an EGFR tyrosine kinase inhibitor, or may alternatively target the extracellular domain of the EGFR target.
  • the EGFR inhibitor is a tyrosine kinase inhibitor such as Erlotinib, Gefitinib, or Lapatinib, or a molecule that targets the EGFR extracellular domain such as Cetuximab or Panitumumab.
  • the EGFR inhibitor is an anti-EGFR antibody, preferably a monoclonal antibody, in particular Cetuximab or Panitumumab.
  • Molecules that target the EGFR extracellular domain including anti-EGFR monoclonal antibodies such as Cetuximab or Panitumumab, are mainly used in the treatment of colorectal cancer or breast cancer treatment.
  • the method according to the invention may preferably be used to predict response to molecules that target the EGFR extracellular domain, and in particular to anti-EGFR monoclonal antibodies, such as Cetuximab or Panitumumab.
  • tyrosine kinase EGFR inhibitors are mainly used in the treatment of lung cancer (in particular non-small cell lung cancer, NSCLC), so that if the patient's cancer is lung cancer (in particular non-small cell lung cancer, NSCLC), then the method according to the invention may preferably be used to predict response to tyrosine kinase EGFR inhibitors, such as Erlotinib, Gefitinib, or Lapatinib.
  • pancreatic cancer or head and neck cancer in particular squamous cell carcinoma of the head and neck (SCCHN)
  • both tyrosine kinase EGFR inhibitors and anti-EGFR monoclonal antibodies are being tested as therapy, so that if the patient's cancer is pancreatic cancer or head and neck cancer (in particular squamous cell carcinoma of the head and neck (SCCHN)), then the method according to the invention may be used to predict response either to tyrosine kinase EGFR inhibitors (such as Erlotinib, Gefitinib, or Lapatinib) or to anti-EGFR monoclonal antibodies (such as Cetuximab or Panitumumab).
  • tyrosine kinase EGFR inhibitors such as Erlotinib, Gefitinib, or Lapatinib
  • anti-EGFR monoclonal antibodies such as Cetuximab or Panitumumab
  • Cetuximab and Panitumumab are currently the clinically mostly used anti-EGFR monoclonal antibodies.
  • further anti-EGFR monoclonal antibodies are in development, such as Nimotuzumab (TheraCIM-h-R3), Matuzumab (EMD 72000), and Zalutumumab (HuMax-EGFr).
  • the method according to the invention may also be used to predict response to these anti-EGFR monoclonal antibodies or any other anti-EGFR monoclonal antibodies (including fragments) that might be further developed, in particular if the patient is suffering from colorectal cancer (in particular metastatic colorectal cancer), breast cancer, pancreatic cancer or head and neck cancer (in particular squamous cell carcinoma of the head and neck (SCCHN)).
  • colorectal cancer in particular metastatic colorectal cancer
  • breast cancer in particular pancreatic cancer or head and neck cancer
  • SCHN squamous cell carcinoma of the head and neck
  • Erlotinib, Gefitinib, and Lapatinib are currently the clinically mostly used tyrosine kinase EGFR inhibitors.
  • tyrosine kinase EGFR inhibitors are in development, such as Canertinib (CI-1033),Neratinib (HKI-272), Afatinib (BIBW2992), Dacomitinib (PF299804,PF-00299804), TAK-285, AST-1306, ARRY334543, AG-1478 (Tyrphostin AG-1478), AV-412, OSI-420 (DesmethylErlotinib), AZD8931 , AEE788 (NVP- AEE788), Pelitinib (EKB-569), CUDC-101 , AG 490, PD153035 HCl, XL647, and BMS-599626 (AC480).
  • the method according to the invention may also be used to predict response to these tyrosine kinase EGFR inhibitors or any other tyrosine kinase EGFR inhibitors that might be further developed, in particular if the patient
  • the present invention also relates to a kit for determining whether a patient with a cancer is likely to respond to an epidermal growth factor receptor (EGFR) inhibitor, comprising or consisting of:
  • Such reagents may notably include reagents for:
  • determining the expression level of at least one miRNA positively or negatively correlated to response to EGFR inhibitors in particular hsa- miR-31 -3p (SEQ ID N0:1 ) miRNA or particular hsa-miR-31 -5p (SEQ ID NO:34) in a sample (preferably a tumor sample, such as a tumor biopsy or whole or part of a tumor surgical resection) of said patient, and /or, ii) detecting at least one mutation positively or negatively correlated to response to EGFR inhibitors, such as those mentioned in Table 3 above.
  • Reagents for determining the expression level of at least one of hsa-miR-31 -3p target gene(s) DBNDD2 and EPB41 L4B or of at least one miRNA positively or negatively correlated to response to EGFR inhibitors, in particular hsa-miR-31 -3p itself or hsa-miR- 31 -5p, in a sample of said patient may notably comprise or consist of primers pairs (forward and reverse primers) and/or probes specific for at least one of hsa-miR-31 -3p target gene(s) DBNDD2 and EPB41 L4B or a microarray comprising a sequence specific for at least one of hsa-miR-31 -3p target gene(s) DBNDD2 and EPB41 L4B.
  • the design of primers and/or probe can be easily made by those skilled in the art based on the sequences of DBNDD2 and/or EPB41 L4B disclosed in Tables 1 and
  • Reagents for detecting at least one mutation positively or negatively correlated to response to EGFR inhibitors may include at least one primer pair for amplifying whole or part of the gene of interest before sequencing or a set of specific probes labeled with reporter dyes at their 5' end, for use in an allelic discrimination assay, for instance on an ABI 7900HT Sequence Detection System (Applied Biosystems, Foster City, CA) (see Laurent-Puig P, et al, J ClinOncol. 2009, 27(35):5924-30 and Lievre et al. J ClinOncol. 2008 Jan 20;26(3):374-9 for detection of BRAF mutation V600).
  • the kit of the invention may further comprise instructions for determining whether the patient is likely to respond to the EGFR inhibitor based on the expression level of at least one of hsa-miR-31 -3p target gene(s) DBNDD2 and EPB41 L4B and the other tested parameter.
  • a nomogram including points scales of all variables included in the composite score and correlation between the composite score (total number of points) and the prediction (response/non-response or probability of response or non- response) may be included.
  • the method of the invention predicts patient responsiveness to EGFR inhibitors at rates that match reported clinical response rates for the EGFR inhibitors.
  • a method for treating a patient with a cancer comprises administering to the patient at least one EGFR inhibitor, wherein the patient has been predicted (or classified) as "responder” or “likely to respond” by the method as described above.
  • the invention concerns a method for treating a patient affected with a cancer, which method comprises (i) determining whether the patient is likely to respond to an EGFR inhibitor, by the method according to the invention, and (ii) administering an EGFR inhibitor to said patient if the patient has been determined to be likely to respond to the EGFR inhibitor.
  • the method may further comprise, if the patient has been determined to be unlikely to respond to the EGFR inhibitor a step (in) of administering an alternative anticancer treatment to the patient.
  • an alternative anticancer treatment depends on the specific cancer and on previously tested treatments, but may notably be selected from radiotherapy, other chemotherapeutic molecules, or other biologies such as monoclonal antibodies directed to other antigens (anti-Her2, anti-VEGF, anti-EPCAM, anti-CTLA4).
  • the alternative anticancer treatment administered in step (iii) may be selected from:
  • a VEGF inhibitor in particular an anti-VEGF monoclonal antibodies (such as bevacizumab), advantageously in combination with FOLFOX (a combination of leucovorin (folinic acid), 5-fluorouracil (5-FU), and oxaliplatin) or FOLFIRI (a combination of leucovorin (folinic acid), 5-fluorouracil (5-FU), and irinotecan) chemotherapy.
  • FOLFOX a combination of leucovorin (folinic acid), 5-fluorouracil (5-FU), and oxaliplatin
  • FOLFIRI a combination of leucovorin (folinic acid), 5-fluorouracil (5-FU), and irinotecan
  • VEGF inhibitor optionally in combination with FOLFOX or FOLFIRI chemotherapy
  • 5-FU optionally in combination with Mitomycin B
  • Best supportive care defined as a treatment administered with the intent to maximize quality of life without a specific antineoplastic regimen (i.e. not an anticancer treatment) may further be administered to the patient.
  • Another subject of the invention is an EGFR inhibitor, for use in treating a patient affected with a cancer, wherein the patient has been classified as being likely to respond by the method as defined above.
  • the invention also relates to an EGFR inhibitor for use in treating a patient affected with a cancer, wherein said treatment comprises a preliminary step of predicting if said patient is or not likely to respond to the EGFR inhibitor by the method as defined above, and said EGFR inhibitor is administered to the patient only is said patient has been predicted as likely to respond to the EGFR inhibitor by the method as defined above.
  • Said patient may be affected with a colorectal cancer, more particularly a metastatic colorectal cancer.
  • said patient may be affected with a breast cancer, in particular a triple negative breast cancer.
  • said patient may be affected with a lung cancer, in particular a non-small cell lung cancer (NSCLC).
  • NSCLC non-small cell lung cancer
  • said patient may be affected with a head and neck cancer, in particular a squamous-cell carcinoma of the head and neck.
  • said patient may be affected with a pancreatic cancer.
  • the invention also relates to the use of an EGFR inhibitor for the preparation of a medicament intended for use in the treatment of cancer in patients that have been classified as "responder" by the method of the invention as described above.
  • the EGFR inhibitor is an anti-EGFR antibody, preferably cetuximab or panitumumab.
  • the EGFR inhibitor may be a tyrosine kinase EGFR inhibitor, in particular Erlotinib, Gefitinib, or Lapatinib.
  • the patient is afflicted with a colorectal cancer, in particular a metastatic colorectal cancer, and the EGFR inhibitor is an anti-EGFR antibody, preferably cetuximab or panitumumab;
  • the patient is afflicted with a breast cancer, in particular a triple negative breast cancer, and the EGFR inhibitor is an anti-EGFR antibody, preferably cetuximab or panitumumab;
  • the patient is afflicted with a lung cancer, in particular a non-small cell lung cancer (NSCLC), and the EGFR inhibitor is a tyrosine kinase EGFR inhibitor, in particular Erlotinib, Gefitinib, or Lapatinib;
  • NSCLC non-small cell lung cancer
  • the EGFR inhibitor is a tyrosine kinase EGFR inhibitor, in particular Erlotinib, Gefitinib, or Lapatinib;
  • the patient is afflicted with a head and neck cancer, in particular a squamous- cell carcinoma of the head and neck, or a pancreatic cancer
  • the EGFR inhibitor is an anti-EGFR antibody (preferably cetuximab or panitumumab) or a tyrosine kinase EGFR inhibitor (in particular Erlotinib, Gefitinib, or Lapatinib).
  • Example 1 DBNDD2 and EPB41 L4B are targets of hsa-miR-31 -3p and independently predict response to EGFR inhibitors
  • the set of patients was made of 20mCRC(metastatic colorectal cancer) patients, 14 males, 6 females. The median of age was 66.49 ⁇ 11.9 years. All patients received a combination of irinotecan and cetuximab. The number of chemotherapy lines before the introduction of Cetuximab was recorded. The median of follow-up until progression was 20 weeks and the median overall survival was 10 months. All tumor sample came from resections and were fixed in formalin and paraffin embedded (FFPE).
  • FFPE formalin and paraffin embedded
  • HTB-37 3 colorectal adenocarcinoma cell lines from the American Type Culture Collection (ATCC, Manassas, CA) that express weaklyhsa-miR-31 -3p: HTB-37, CCL-222 and CCL-220-1.
  • HTB-37 cells were maintained in a Dulbecco's Modified Eagle Medium (DMEM) culture medium with stable glutamine with 20% Fetal Bovine serum and 1% Penicillin/Streptomycin.
  • DMEM Dulbecco's Modified Eagle Medium
  • CCL-222 and CCL-220-1 cells were maintained in a RPMI 1640 culture media with stable glutamine with 10% fetal bovine serum. The cells were incubated at a temperature of 37 °C with 5% C02.
  • Gene expression microarray was performed using the AffymetrixHuman Gene 1 .0. Fifty ng of total RNA was reverse transcribed following the Ovation PicoSL WTA System V2 (Nugen, San Carlos, CA). Then, amplification was done based on SPIA technology. After purification according to Nugen protocol, 2.5 pg of single strand DNA was used for fragmentation and biotin labelling using Encore Biotin Module (Nugen). After control of fragmentation using Bioanalyzer 2100, cDNA was then hybridized to GeneChip® human Gene 1.0 ST (Affymetrix) at 45 ° C for 17 hours.
  • Chips were washed on the fluidic station FS450 following specific protocols (Affymetrix) and scanned using the GCS3000 7G. The image was then analyzed with Expression Console software (Affymetrix) to obtain raw data (CELfiles) and metrics for Quality Controls.
  • Affymetrix Expression Console software
  • qRT-PCR validation of the target expression on cell lines and FFPE patients samples were performed on 20ng of total RNA for FFPE samples or 50ng of total RNA cell culture samples using ABI7900HT Real-Time PCR System (Applied Biosystem). All reactions were performed in triplicate. Expression levels were normalized to the RNA18S and GAPDH levels through the AACt method.
  • False-discovery rate (FDR)-adjusted p-values were calculated using the Benjamini and Hochberg procedure for multiple testing correction. The cor. test function was used to calculate Pearson correlations between expression values together with matching p- values. Statistical significance was set at p ⁇ 0.05 for all analyses.
  • CRC cell lines that weakly express hsa-miR-31 -3p were transfected with hsa-miR- 31 -3p mimic or with a mimic control.
  • the transfection efficacy was attested by an average rise of hsa-miR-31 -3p level of 1500 times without mortality or growth defect.
  • Expression profile analysis of the transfected cells allowed us to identify 47 genes significantly down-regulated (fc ⁇ 0.77, p ⁇ 0.05), and 27 genes significantly up-regulated by hsa-miR-31 -3p (fc ⁇ 1 .3, p ⁇ 0.05), as described in Table 4 below.
  • Table 4 List of the genes with a fc ⁇ 0.77 or fc>1 .3 and a pvalue>0.05 identified in the expression array made on the 3 cell lines (fc: fold change in expression between cell lines transfected with hsa-miR-31 -3p mimic and cell lines transfected with a mimic control)
  • the database including information from 6 web-available predicts the 54 down -regulated genes as hsa-miR-31 -3p putative target.
  • the database may be queried either by miRNA name, or by gene name.
  • miRNA name When a miRNA name is queried, the database returns a list of candidate target genes, ranked by order of probability (from the most probable to the less probable) that the genes are true targets of the queried miRNA, based on structural and potential experimental data included in the database.
  • the database returns a list of miRNA candidates, ranked by order of probability (from the most probable to the less probable) that the mi ' RNAs truly target the queried gene, based on structural and potential experimental data included in the database.
  • the database was queried with hsa-miR-31 -3p name and with the names of genes found to be down-regulated in CRC cell lines overexpressing hsa-miR-31 -3p (47 genes, cf Table 4) .
  • Table 5below shows down-regulated genes of Table 4, including DBNDD2 and EPB41 L4B, which were identified as a putative direct target of has-miR-31 -3p. lt also indicates the rank of hsa-miR-31 -3p if the database was queried using the gene name, and the rank of the gene if the database was queried using hsa-miR-31 -3p name.
  • Table 5 Target predictions from in silico database are indicated for the down- regulated genes depending on the request: Column 2: database was interrogated with a gene of interest, and reported all candidate microRNAs potentially targeting this gene, ranked from the most likely to the less likely. The rank of hsa-miR-31 -3p and the total number of microRNA candidates are indicated; Column 3:database was interrogated with hsa-miR-31 -3p, and reported all putative targets, ranked from the most likely to the less likely for a total of 1620 putative targeted genes. Then rank of the queried gene is indicated. Only down-regulated genes listed in hsa-miR-31 -3p 1620 putative targeted genes are presented in Table 5. Data relating to DBNDD2 and EPB41 L4B are in bold.
  • the 25 putative direct target genes and the 27 indirect target genes were validated on qRT-PCR, out of these47genes, 45 displayed an expression level comparable to the level obtained in the array.
  • DBNDD2 and EPB41 L4B showed a significant negative correlation with hsa-miR-31 -3p expression levels: DBNDD2 and EPB41 L4B (see Figure 1A and 1 B) .
  • these results suggest that expression of DBNDD2 and EPB41 L4B could distinguish between mCRC patients with poor or good prognosis, i.e. between non-responders and responders mCRC patients.
  • Example 2 Creation of a tool with DBNDD2 and EPB41L4B expression to predict response to EGFR inhibitors
  • the set of patients was made of 20mCRC patients, 13 males and 7 females. The median of age was 67 ⁇ 11 .2 years. All had a metastatic disease at the time of the inclusion. All these patients developed a KRAS wild type metastatic colon cancer. All patients were considered refractory to a 5-fluorouracil-based regimen combined with irinotecan and oxaliplatin. They received an anti-EGFR-based chemotherapy, 8 patients with panitumumab, 10 patients with cetuximab and 2 patients received a combination of panitumumab and cetuximab. The number of chemotherapy lines before the introduction of Cetuximab and panitumumab was recorded. The median of follow-up until progression was 21 weeks and the median overall survival was 8.9 months. Measurement of gene expression
  • Example 3 Replication of the predictive value of DBNDD2 and EPB41 L4B to EGFR inhibitors in a new and independent cohort PA TIENTS AND METHODS
  • the set of patients was made of 42 mCRC(metastatic colorectal cancer) patients, 27 males and 15 females. The median of age was 59 ⁇ 12.1years. All had a metastatic disease at the time of the inclusion. All patients were treated with 3rd line therapy by a combination of irinotecan and panitumumab after progression with oxaliplatin and irinotecan chemotherapy based regimens. The median of follow-up until progression was 23 weeks and the median overall survival was 9.6 months. 26 samples were available in FFPE and 16 in frozen tissue.
  • qRT-PCR validation of the target expression on frozen or FFPE patients samples were performed on 20ng of total RNA using ABI7900HT Real-Time PCR System (Applied Biosystem). All reactions were performed in triplicate. Expression levels were normalized to the RNA18S or GAPDH levels through the AACt method.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Zoology (AREA)
  • Immunology (AREA)
  • Wood Science & Technology (AREA)
  • Engineering & Computer Science (AREA)
  • Analytical Chemistry (AREA)
  • Genetics & Genomics (AREA)
  • Pathology (AREA)
  • General Health & Medical Sciences (AREA)
  • Physics & Mathematics (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Oncology (AREA)
  • General Engineering & Computer Science (AREA)
  • Hospice & Palliative Care (AREA)
  • Biochemistry (AREA)
  • Molecular Biology (AREA)
  • Microbiology (AREA)
  • Biophysics (AREA)
  • Biotechnology (AREA)
  • Public Health (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Animal Behavior & Ethology (AREA)
  • Medicinal Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
  • Peptides Or Proteins (AREA)

Abstract

The present invention relates to a method for predicting whether a patient with a cancer is likely to respond to an epidermal growth factor receptor (EGFR) inhibitor, which method comprises determining the expression level of at least one target gene of hsa-miR-31 -3p (SEQ ID NO:1 ) miRNA in a sample of said patient, wherein said target gene of hsa-miR-31 -3p is selected from DBNDD2 and EPB41 L4B. The invention also relates to kits for measuring the expression of DBNDD2 and/or EPB41 L4B and at least one other parameter positively or negatively correlated to response to EGFR inhibitors. The invention also relates to therapeutic uses of an EGFR inhibitor in a patient predicted to respond to said EGFR inhibitor.

Description

A METHOD FOR PREDICTING RESPONSIVENESS TO A TREATMENT WITH AN EGFR INHIBITOR
TECHNICAL FIELD OF THE INVENTION The present invention provides methods for individualizing chemotherapy for cancer treatment, and particularly for evaluating a patient's responsiveness to one or more epidermal growth factor receptor (EGFR) inhibitors prior to treatment with such agents, based on the determination of the expression level of at least one target gene of hsa- mi'R-31 -3p (SEQ ID NO: 1 ) miRNA, wherein said target gene of hsa-mi'R-31 -3p is selected from DBNDD2 and EPB41 L4B.
BACKGROUND OF THE INVENTION
The epidermal growth factor receptor (EGFR) pathway is crucial in the development and progression of human epithelial cancers. The combined treatment with EGFR inhibitors has a synergistic growth inhibitory and pro-apoptotic activity in different human cancer cells which possess a functional EGFR-dependent autocrine growth pathway through to a more efficient and sustained inhibition of Akt.
EGFR inhibitors have been approved or tested for treatment of a variety of cancers, including non-small cell lung cancer (NSCLC) , head and neck cancer, colorectal carcinoma, and Her2-positive breast cancer, and are increasingly being added to standard therapy. EGFR inhibitors, which may target either the intracellular tyrosine kinase domain or the extracellular domain of the EGFR target, are generally plagued by low population response rates, leading to ineffective or non-optimal chemotherapy in many instances, as well as unnecessary drug toxicity and expense. For example, a reported clinical response rate for treatment of colorectal carcinoma with cetuximab (a chimeric monoclonal antibody targeting the extracellular domain of EGFR) is about 1 1 % (Cunningham et al, N Engl Med 2004;351 : 337-45), and a reported clinical response rate for treatment of NSCLC with erlotinib is about 8.9% (Shepherd F A, et al, N Engl J Med 2005; 353 : 123-132) .
In particular resistance has been observed in case of KRAS mutation .
In colorectal cancer, as KRAS mutations are clearly associated with resistance to anti- EGFR antibodies (Lievre et al, Cancer Res. 2006 66(8) :3992-5) , one of the major challenges is to identify, in non-mutated KRAS patients, other markers that can predict lack of response to this therapy. Among them, amplification or activating mutations of oncogenes and inactivating mutations of tumor suppressor genes described above are relevant candidates, such as the level of activation of EGFR downstream signaling pathway evaluated by the measurement of EGFR downstream phosphoprotein expression. In lung cancer, three groups of patients are emerging: one counts the patients with EGFR mutated tumors for which the use of EGFR tyrosine kinase inhibitors (EGFR TKI) was proven to improve outcome, the second counts the patients with KRAS mutated tumors for which anti-EGFR therapies are probably not the good alternatives, and the third group counts the non-EGFR and non-KRAS mutated tumors for which response cannot be predicted. No marker linked to drug response in the non-mutated tumor group has proved valuable so far.
Thus, there is a need for predicting patient responsiveness to EGFR inhibitors prior to treatment with such agents, so as to better individualize patent therapy.
There are many documents in the prior art concerning the involvement of micro RNAs (miRNAs) in sensitivity or resistance to various anticancer treatments. In particular, PCT/EP2012/073535 describes an in vitro method for predicting whether a patient with a cancer is likely to respond to an epidermal growth factor receptor (EGFR)inhibitor, which comprises determining the expression level of hsa-mi'R-31 -3p (previously named hsa-miR-31*, SEQ ID NO:1 ) miRNA in a sample of said patient. More particularly, the lower the expression of hsa-miR-31 -3p is, the more likely the patient is to respond to the EGFR inhibitor treatment.
Similarly, there are many documents in the prior art concerning the involvement of various genes in sensitivity or resistance to various anticancer treatments. However, in most cases, studies are partial, incomplete, and actually do not permit a true prediction of clinical response or non-response to treatment. Indeed, in many cases, studies are limited to the analysis of the expression of genes in vitro, in cell lines sensitive or resistant to a particular treatment, or in tumor cells isolated from a patient tumor. In addition, in many studies, while differences in expression level between two populations of cells or patients are shown, no threshold value or score actually permitting to predict response or non-response in a new patient are provided. This is partly linked to the first shortage that many studies lack data obtained in a clinical setting. Moreover, even when some data obtained in a clinical setting is presented, these data are most of the time only retrospective, and data validating a prediction method in an independent cohort are often lacking.
In view of various shortcomings of prior art studies, there is still a need for true and validated methods for predicting response to EGFR inhibitors in patients for which such therapy is one of several options. The present invention provides a response to this need.
DBNDD2 (dysbindin (dystrobrevin binding protein 1 ) domain containing 2) has been disclosed to be a binding partner of human casein kinase-1 (Yin H et al. Biochemistry. 2006 Apr 25;45(16):5297-308). In addition, using microarray global profiling, it has been found, among many other genes, to be differentially expressed in various tumor cells (WO2010065940 ; WO2010059742 ; WO2009131710 ; WO2007112097), or between cancer cells sensitive or resistant torapamycin (WO2011017106) or tamoxifen (WO2010127338). However, this gene does not seem to have been specifically associated to cancer, and no involvement of this gene in prediction of response to EGFR inhibitors has been disclosed.
EPB41 L4B (erythrocyte membrane protein band 4.1 like 4B) is a protein of the FERM family proteins, which can link transmembrane proteins to the cytoskeleton or link kinase and/or phosphatase enzymatic activity to the plasma membrane, and have been described to be involved in carcinogenesis and metastasis. In particular, EPB41 L4B (also known as EHM2) has been associated to increased aggressiveness of prostate cancer (Wang J, et al. Prostate. 2006 Nov 1 ;66(15):1641 -52 ; Schulz WA, et al. BMC Cancer.2010 Sep 22;10:505) and breast cancer (Yu H et al. Mol Cancer Res 2010;8: 1 501 - 1512) . This gene has thus been associated to aggressiveness and poor prognosis of at least two types of cancer. Moreover, it has been found to be differentially expressed between cancer cell lines sensitive and resistant to taxotere (docetaxel, see WO2007072225 and WO2008138578). However, there has been no disclosure of its association to the ability of a cancer patient to respond or not to EGFR inhibitors.
The inventors implemented a new database incorporating information from the 6 databases, which may be interrogated either based on the name of a miRNA, or based on a gene name. In the first case (query based on miRNA name), the database returns genes names considered as candidate targets of the queried miRNA, based on published or structural information, candidate target genes being ranked from the most probable to the less probable based on available information. When the query is based on a gene name, the database returns candidates miRNAs, for which the queried gene might (or not) be a target.
SUMMARY OF THE INVENTION
With the aim to understand why increased expression of hsa-mi'R-31 -3p is associated to lower response to EGFR inhibitor treatment, the inventors tried to identify target genes of this miRNA. For this purpose, they transfected three colorectal adenocarcinoma (CRC) cell lines that naturally weakly express hsa-miR-31 -3p with a mimic of hsa-miR- 31 -3p or a negative control mimic and analyzed genes differentially expressed between cell lines overexpressing or expressing weakly hsa-miR-31 -3p. A total of 74genes significantly down- or up-regulated was identified. Since miRNAs function mainly by decreasing expression of their target genes, the inventors focused on the 47 down- regulated genes. To limit the number of candidate targets and avoid the false direct target genes, the inventors further performed in silico analyses based on information available in 6 databases relating to miRNAs and candidate targets. It is important to note that, most miRNA target genes provided in public databases are not validated, but only more or less probable candidates, based on structural or fragmental experimental data. 25 candidate target genes of hsa-miR-31 -3p were selected for further analysis on this basis. The inventors further analyzed the expression of these candidate target genes of hsa-mi'R-31 -3p in tumor samples of patients treated with EGFR inhibitors, whose treatment response status based on RECIST criteria were known.
Based on these analyses, the inventors surprisingly found that DBNDD2 and EPB41 L4B are both hsa-miR-31 -3p target genes, since their expression is significantly down- regulated by overexpression of hsa-miR-31 -3p in cancer cell lines, and that each of these genes is independently significantly associated to the ability of cancer patients to respond to EGFR inhibitor treatment. They further confirmed that each of these genes may alone be used for reliably predicting response to EGFR inhibitors in cancer patients. None of the other 23 candidate target genes of hsa-miR-31 -3p was found to be significantly associated to the ability of cancer patients to respond to EGFR inhibitor treatment, although some of these genes were considered in databases as a candidate target gene of hsa-miR-31 -3p with higher probability, such as HAUS4, and known to be associated to cancer, such as STAT3, FEM1A, EHBP1 and SEC31A. This clearly indicates that mere association of a gene to cancer is not sufficient to reasonably expect that the gene may be used as a biomarker of response to a particular cancer treatment. This also illustrates that only a few of the numerous candidate target genes of a particular miRNA disclosed in public databases are true targets of this miRNA, and that the true targets are not necessarily the best ranked candidates.
Surprisingly, the two genes found to be significantly down-regulated in patients not responding to EGFR inhibitor treatment are a gene not specifically known to be associated to cancer (DBNDD2) and a gene known to be associated to cancer (EPB41 L4B), but for which high expression level was associated to poor prognosis. In contrast, in the present invention, it is a low expression of EPB41 L4B that is associated to absence of response to EGFR inhibitors, and thus to poor prognosis. These results further confirm that biomarkers of prognosis (in general) may not be reasonably expected to be also biomarkers of response to a particular treatment.
Based on the results obtained by the inventors (see Example 1 ), the present invention provides an in vitro method for predicting whether a patient with a cancer is likely to respond to an epidermal growth factor receptor (EGFR) inhibitor, which comprises determining the expression level of at least one target gene of hsa-miR-31 -3p (SEQ ID NO:1 ) miRNA in a sample of said patient, wherein said target gene of hsa-miR-31 -3p is selected from DBNDD2 and EPB41 L4B.
Preferably the patient has a KRAS wild-type cancer.
The cancer preferably is a colorectal cancer, preferably a metastatic colorectal cancer. In a most preferred embodiment, the invention provides an in vitro method for predicting whether a patient with a metastatic colorectal carcinoma is likely to respond to an epidermal growth factor receptor (EGFR) inhibitor, such as cetuximab or panitumumab, which method comprises determining the expression level of at least one target gene of hsa-miR-31 -3p (SEQ ID N0:1 ) miRNA in a tumor sample of said patient, wherein said target gene of hsa-miR-31 -3p is selected from DBNDD2 and EPB41 L4B.
The invention also provides a kit for determining whether a patient with a cancer is likely to respond to an epidermal growth factor receptor (EGFR) inhibitor, comprising or consisting of: reagents for determining the expression level of at least one target gene of hsa-miR-31 -3p (SEQ ID NO:1 ) miRNA in a sample of said patient, wherein said target gene of hsa-miR-31 -3p is selected from DBNDD2 and EPB41 L4B, and reagents for determining at least one other parameter positively or negatively correlated to response to EGFR inhibitors.
The invention further relates to an EGFR inhibitor for use in treating a patient affected with a cancer, wherein the patient has been classified as being likely to respond, by the method according to the invention. The invention also relates to the use of an EGFR inhibitor for the preparation of a drug intended for use in the treatment of cancer in patients that have been classified as "responder" by the method of the invention.
The invention also relates to a method for treating a patient affected with a cancer, which method comprises (i) determining whether the patient is likely to respond to an EGFR inhibitor, by the method of the invention, and (ii) administering an EGFR inhibitor to said patient if the patient has been determined to be likely to respond to the EGFR inhibitor.
BRIEF DESCRIPTION OF THE FIGURES Figure 1 : Correlation between log2 expression levels of DBNDD2 (in Figure 1A) and EPB41 L4B (in Figure 1B) and hsa-miR-31 -3p in the 20 mCRC patients of Example 1 .
Figure 2: Correlation between log2 expression levels of DBNDD2 and hsa-miR-31 -3p in the 20 mCRC patients of Example 2.
Figure 3: In A: Nomogram tool established based on log2 expression of DBNDD2 in the 20 mCRC patients of Example 2, in order to predict risk of progression (i.e. risk of non- response) of mCRC patients treated with anti-EGFR based chemotherapy.
Figure 4: Multivariate Cox proportional hazards models with DBNDD2 expression as covariate in the 20 mCRC patients of Example 2.
Figure 5: Correlation between log2 expression levels of DBNDD2 (in Figure 5A) and EPB41 L4B (in Figure 5B) and hsa-miR-31 -3p in the 42 mCRC patients of Example 3.
Figure 6: Expression of DBNDD2 (in Figure 6A) and EPB41 L4B (in Figure 6B) in patients of Example 3 according to their risk of progression (low or high), as predicted based on hsa-miR-31 -3p expression level.
DETAILED DESCRIPTION OF THE INVENTION
Definitions
The "patient" may be any mammal, preferably a human being, whatever its age or sex. The patient is afflicted with a cancer. The patient may be already subjected to a treatment, by any chemotherapeutic agent, or may be untreated yet.
The cancer is preferably a cancer in which the signaling pathway through EGFR is involved. In particular, it may be e.g. colorectal, lung, breast, ovarian, endometrial, thyroid, nasopharynx, prostate, head and neck, kidney, pancreas, bladder, or brain cancer (Ciardello F et al. N Engl J Med. 2008 Mar 13;358(11 ):1160-74 ; Wheeler DL et al. Nat RevClinOncol. 2010 September ; 7(9): 493-507 ; Zeineldin R et al. J Oncol. 2010;2010:414676;Albitar L et al. Mol Cancer 2010;9:166; Leslie KK et al. GynecolOncol. 2012 Nov;127(2):345-50; Mimeault M et al. PLoS One.2012;7(2):e31919; Liebner DA et al. TherAdvEndocrinolMetab. 2011 Oct;2(5):173-95; Leboulleux S et al. Lancet Oncol. 2012 Sep;13(9):897-905; Pan J et al. Head Neck. 2012 Sep 13; Chan SL et al. Expert OpinTher Targets. 2012 Mar;16 Suppl 1 :S63-8; Chu H et al. Mutagenesis.2012 Oct 15; Li Y et al. Oncol Rep. 2010 Oct;24(4):1019-28; Thomasson M et al. Br J Cancer 2003, 89:1285-1289; Thomasson M et al. BMC Res Notes.2012 May 3;5:216). In certain embodiments, the tumor is a solid tissue tumor and/or is epithelial in nature. For example, the patient may be a colorectal carcinoma patient, a Her2-positive or Her2- negative (in particular triple negative, i.e. Her2-negative, estrogen receptor negative and progesterone receptor negative) breast cancer patient, a non-small cell lung cancer (NSCLC) patient, a head and neck cancer patient (in particular a squamous-cell carcinoma of the head and neck patient), a pancreatic cancer patient, or an endometrial cancer patient. More particularly, the patient may be a colorectal carcinoma patient, a Her2-positive or Her2- negative (in particular triple negative) breast cancer patient, a lung cancer (in particular a NSCLC) patient, a head and neck cancer patient (in particular a squamous-cell carcinoma of the head and neck patient), or a pancreatic cancer patient.
In a preferred embodiment, the cancer is a colorectal cancer, still preferably the cancer is a metastatic colorectal cancer. Indeed, data presented in Example 1 clearly indicate that DBNDD2 or EPB41 L4B expression level may be used as a predictor of response to EGFR inhibitors (and in particular to anti-EGFR monoclonal antibodies such as cetuximab and panitumumab) treatment in colorectal cancer.
These results, obtained in a cancer in which the EGFR signaling pathway is known to be involved, clearly suggest that DBNDD2 and/or EPB41 L4B expression level might be used as a predictor of response to EGFR inhibitors (and in particular to anti-EGFR monoclonal antibodies such as cetuximab and panitumumab) in any other cancer in which the EGFR signaling pathway is known to be involved, such as lung, ovarian, endometrial, thyroid, nasopharynx, prostate, head and neck, kidney, pancreas, bladder, or brain cancer. Therefore, in another preferred embodiment, the cancer is a Her2-positive or Her2- negative (in particular triple negative) breast cancer, preferably a Her2- negative (in particular triple negative) breast cancer.
In still another preferred embodiment, the cancer is a lung cancer, in particular a non- small cell lung cancer (NSCLC).
In still another preferred embodiment, the cancer is a pancreatic cancer.
Since the prediction relates to EGFR inhibitors treatment, the patient's tumor is preferably EGFR positive.
Preferably, the patient has a KRAS wild-type tumor, i.e. , the KRAS gene in the tumor of the patient is not mutated in codon 12, 13 (exon 1 ), or 61 (exon 3) . In other words, the KRAS gene is wild-type on codons 12, 13 and 61 .
Wild type, i.e. non mutated, codons 12, 13 (exon 1 ),and 61 (exon 3) respectively correspond to glycine (Gly, codon 12), glycine (Gly, codon 13), and glutamine (Gin, codon 61 ). The wild-type reference KRAS amino acid sequence may be found in Genbank accession number NP_004976.2 (SEQ ID NO: 24).
Especially the KRAS gene of the patient's tumor does not show any of the following mutations (Bos. Cancer Res 1989;49:4682-4689; Edkins et al. Cancer BiolTher. 2006 August ; 5(8): 928-932; Demiralay et al. Surgical Science, 2012, 3, 111 -115):
Gly12Ser (GGT>AGT)
Gly12Arg (GGT>CGT)
Gly12Cys (GGT>TGT) Gly12Asp (GGT>GAT)
Gly12Ala (GGT>GCT)
Gly12Val (GGT>GTT)
Gly13Arg (GGC>CGC)
Gly13Cys (GGC>TGC)
Gly13Asp (GGC>GAC)
Gly13Ala (GGC>GCC)
Gly13Val (GGOGTC)
Preferably, the KRAS gene of the patient's tumor does also not show any of the following mutations (Demiralay et al. Surgical Science, 2012, 3, 111 -115):
Gly12Phe (GGT>TTT)
Gly13Ser (GGOAGC)
Preferably, the KRAS gene of the patient's tumor does also not show any of the following mutations (Bos. Cancer Res 1989;49:4682-4689; Tarn et al. Clin Cancer Res2006;12:1647-1653 ; Edkins et al. Cancer BiolTher. 2006 August ; 5(8): 928-932;
Demiralay et al. Surgical Science, 2012, 3, 111 -115):
Gln61 His (CAA>CAC)
Gln61 His (CAA>CAT)
Gln61Arg (CAA>CGA)
Gln61 Leu (CAA>CTA)
Gln61 Glu (CAA>GAA)
Gln61 Lys (CAA>AAA)
Gln61 Pro (CAA>CCA)
Any method known in the art may be used to know the KRAS status of the patient.
For example, a tumor tissue is microdissected and DNA extracted from paraffin- embedded tissue blocks. Regions covering codons 12, 13, and 61 of the KRAS gene are amplified using polymerase chain reaction (PCR). Mutation status is determined by allelic discrimination using PCR probes (Laurent-Puig P, et al, J ClinOncol. 2009, 27(35) :5924-30) or by any other methods such as pyrosequencing (Ogino S, et al. J MolDiagn 2008;7:413-21 ).
The "sample" may be any biological sample derived from a patient, which contains nucleic acids. Examples of such samples include fluids (including blood, plasma, saliva, urine, seminal fluid), tissues, cell samples, organs, biopsies, etc. Preferably the sample is a tumor sample, preferably a tumor tissue biopsy or whole or part of a tumor surgical resection. The sample may be collected according to conventional techniques and used directly for diagnosis or stored. A tumor sample may be fresh, frozen or paraffin- embedded. Usually, available tumor samples are frozen or paraffin-embedded, most of the time paraffin-embedded. The inventors have shown that both frozen and paraffin- embedded tumor samples may be used.
By a "reference sample", it is meant a tumor sample (notably a tumor biopsy or whole or part of a tumor surgical resection) of a patient whose positive or negative response to an EGFR inhibitor treatment is known. Preferably, a pool of reference samples comprises at least one (preferably several, more preferably at least 5, more preferably at least 6, at least 7, at least 8, at least 9, at least 10) responder patient(s) and at least one (preferably several, more preferably at least 6, at least 7, at least 8, at least 9, at least 10) non responder patient(s). The highest the number of responders (also referred to as "positive") and non-responders (also referred to as "negative") reference samples, the better for the reliability of the method of prediction according to the invention.
Within the context of this invention, a patient who is "likely to respond" or is "responder" refers to a patient who may respond to a treatment with an EGFR inhibitor, i.e. at least one of his symptoms is expected to be alleviated, or the development of the disease is stopped, or slowed down. Complete responders, partial responders, or stable patients according to the RECIST criteria (Eisenhauer et al, European Journal of Cancer, 2009, 45:228-247) are considered as "likely to respond" or "responder" in the context of the present invention.
In solid tumors, the RECIST criteria are an international standard based on the presence of at least one measurable lesion. "Complete response" means disappearance of all target lesions; "partial response" means 30% decrease in the sum of the longest diameter of target lesions, "progressive disease" means 20% increase in the sum of the longest diameter of target lesions, "stable disease" means changes that do not meet above criteria.
More preferably, a "responder" patient is predicted to show a good progression free survival (PFS), i.e. the patient is likely to survive at least 25 weeks without aggravation of the symptoms of the disease, and/or such patient shows a good overall survival (OS), i.e. the patient is likely to survive at least 14 months.
The term "predicting" or "prognosis" refers to a probability or likelihood for a patient to respond to the treatment with an EGFR inhibitor.
According to the invention, the sensitivity of tumor cell growth to inhibition by an EGFR inhibitor is predicted by whether and to which level such tumor cells express hsa-miR- 31 -3p target genes DBNDD2 and EPB41 L4B.
The term "treating" or "treatment" means stabilizing, alleviating, curing, or reducing the progression of the cancer.
A "miRNA" or "microRNA" is a single-stranded molecule of about 21 -24 nucleotides, preferably 21 -23 in length, encoded by genes that are transcribed from DNA but not translated into protein (non-coding RNA); instead they are processed from primary transcripts known as pri-miRNA to short stem-loop structures called pre-miRNA and finally to functional miRNA. During maturation, each pre-miRNA gives rise to two distinct fragments with high complementarity, one originating from the 5' arm the other originating from the 3' arm of the gene encoding the pri-miRNA. Mature miRNA molecules are partially complementary to one or more messenger RNA (mRNA) molecules, and their main function is to downregulate gene expression.
There is an international nomenclature of miRNAs (see Ambros V et al, RNA 2003 9(3):277-279 ; Griffiths-Jones S. NAR 2004 32(Database lssue):D109-D111 ; Griffiths- Jones S et al. NAR 2006 34(Database lssue):D140-D144; Griffiths- Jones S et al. NAR 2008 36(Database lssue) :D154-D158; and Kozomara A et al. NAR 2011 39(Database lssue):D152-D157), which is available from miRBase at http://www.mirbase.org/. Each miRNA is assigned a unique name with a predefined format, as follows: • For a mature miRNA: sss-miR-X-Y, wherein "
o sss is a three letters code indicating the species of the miRNA, "hsa" standing for human,
o the upper case "R" in mi'R indicates that it is referred to a mature miRNA. However, some authors in the literature abusively use "mir" also for mature miRNA. In this case, it may be recognized that it is referred to a mature miRNA by the presence of "-Y",
o X is the unique arbitrary number assigned to the sequence of the miRNA in the particular species, which may be followed by a letter if several highly homologous miRNAs are known. For instance, "20a" and "20b" refer to highly homologous miRNAs.
o Y indicates whether the mature miRNA, which has been obtained by cutting of the pre-miRNA, corresponds to the 5' arm (Y is then "5p") or 3' arm (Y is then "3p") of the gene encoding the pri-mRNA. In previous international nomenclature of miRNAs, "-Y" was not present. The two mature miRNAs obtained either from the 5' or the 3' arm of the gene encoding the pri-miRNA were then distinguished by the presence or absence of a "*" sign just after n. The presence of the "*" sign indicated that the sequence corresponded to the less often detected miRNA. Since such classification was subject to changes, a new nomenclature using the
"3p" and "5p" code has been implemented.
• For a pri-miRNA:sss-mir-X, wherein
o sss is a three letters code indicating the species of the miRNA, "hsa" standing for human,
o the lower case "r" in mir indicates that it is referred to a pri-miRNA and not to a mature miRNA, which is confirmed by the absence of "-Y", o n is the unique arbitrary number assigned to the sequence of the miRNA in the particular species, which may be followed by a letter if several highly homologous miRNAs are known.
Each miRNA is also assigned an accession number for its sequence.
The miRNA targeted by the two genes detected in the present invention (DBNDD2 and EPB41 L4B) is hsa-miR-31 -3p (previously named hsa-miR-31*). In this name, "hsa" means that it relates to a human miRNA, "miR" refers to a mature miRNA, "31 " refers to the arbitrary number assigned to this particular miRNA, and "3p" means that the mature miRNAs has been obtained from the 3' arm of the gene encoding the pri-miRNA. hsa-miR-31 -3p is UGCUAUGCCAACAUAUUGCCAU(SEQ ID NO : 1 )
(Accession number MIMAT0004504 on http://www.mirbase.org)
"DBNDD2" is the official symbol of NCBI Entrez Gene database for "dysbindin (dystrobrevin binding protein 1 ) domain containing 2" gene (official name and symbol approved by the HUGO Gene Nomenclature Committee (HGNC)), located in humans in chromosome 20 (20q13.12). It corresponds to UniGene database accession number Hs.730643. Further symbols used for this gene include CK1 BP (for "casein kinase-1 binding protein"), HSMNP1 , RP3-453C12.9, and C20orf35. It is also known as "SCF apoptosis response protein 1 ". Five isoforms (a to e) of this protein are known, encoded by several mRNA variants, as detailed in Table 1 below.
Figure imgf000011_0001
Table 1 : isoforms of DBNDD2 and corresponding mRNA and protein reference sequences provided by NCBI EntrezGene database, on July 1 , 2013.
"EPB41 L4B" is the official symbol of NCBI Entrez Gene database for "erythrocyte membrane protein band 4.1 like 4B" gene (official name and symbol approved by the HGNC), located in humans in chromosome 9 (9q31 -q32). It corresponds to UniGene database accession number Hs.591901 . Further symbols used for this gene include CG1 and EHM2 (for "Expressed in Highly Metastatic cells 2"). It is also known as "FERM- containing protein CG1 ". Two isoforms (1 and 2) of this protein are known, encoded by two mRNA variants, as detailed in Table 2 below.
Figure imgf000011_0002
Table 2: isoforms of EPB41 L4Band corresponding mRNA and protein reference sequences provided by NCBI EntrezGene database, as updated on July 1 , 2013.
Methods of Detecting DBNDD2 and /or EPB41 L4BExpression Levels in a Sample The expression level of hsa-miR-31 -3p target gene(s) DBNDD2 and/or EPB41 L4B may be determined by any technology known by a person skilled in the art. In particular, each gene expression level may be measured in vitro, starting from the patient's sample, at the genomic and/or nucleic acid and/or proteic level. In a preferred embodiment, the expression profile is determined by measuring in vitro the amount of nucleic acid transcripts of each gene. In another embodiment, the expression profile is determined by measuring in vitro the amount of protein produced by each of the genes.
Such measures are made in vitro, starting from a patient's sample, in particular a tumor sample, and necessary involve transformation of the sample. Indeed, no measure of a specific gene expression level can be made without some type of transformation of the sample.
Most technologies rely on the use of reagents specifically binding to the gene DNA, transcripts or proteins, thus resulting in a modified sample further including the detection reagent.
In addition, most technologies also involve some preliminary extraction of DNA, mRNA or proteins from the patient's sample before binding to a specific reagent. The claimed method may thus also comprise a preliminary step of extracting DNA, mRNA or proteins from the patient's sample. In addition, when mRNAs are extracted, they are generally retrotranscribed into cDNA, which is more stable than mRNA. The claimed methods may thus also comprise a step of retrotranscribing mRNA extracted from the patient's sample into cDNA.
Detection by mass spectrometry does not necessary involve preliminary binding to specific reagents. However, it is most of the time performed on extracted DNA, mRNA or proteins. Even when preformed directly on the sample, without preliminary extraction steps, it involves some extraction of molecules from the sample by the laser beam, which extracted molecules are then analysed by the spectrometer.
In any case, no matter which technology is used, the state of the sample after measure of a gene expression level has been transformed compared to the initial sample taken from the patient.
The amount of nucleic acid transcripts can be measured by any technology known by a person skilled in the art. In particular, the measure may be carried out directly on an extracted messenger RNA (mRNA) sample, or on retrotranscribed complementary DNA (cDNA) prepared from extracted mRNA by technologies well-known in the art. From the mRNA or cDNA sample, the amount of nucleic acid transcripts may be measured using any technology known by a person skilled in the art, including nucleic microarrays, quantitative PCR, next generation sequencing and hybridization with a labelled probe. In particular, real time quantitative RT-PCR (qRT-PCR) may be useful. In some embodiments, qRT-PCR can be used for both the detection and quantification of RNA targets (Bustin et al., 2005, Clin. Sci., 109:365-379). Quantitative results obtained by qRT-PCR can sometimes be more informative than qualitative data, and can simplify assay standardization and quality management. Thus, in some embodiments, qRT-PCR- based assays can be useful to measure hsa-miR-31 -3p target gene(s) DBNDD2 and/or EPB41 L4B expression levels during cell-based assays. The qRT-PCR method may be also useful in monitoring patient therapy. qRT-PCR is a well-known and easily available technology for those skilled in the art and does not need a precise description. Examples of qRT-PCR-based methods can be found, for example, in U.S. Pat. No. 7,101 ,663. Commercially available qRT-PCR based methods (e.g. ,Taqman® Array) may for instance be employed, the design of primers and/or probe being easily made based on the sequences of DBNDD2 and/or EPB41 L4B disclosed in Tables 1 and 2 above.
Nucleic acid assays or arrays can also be used to assess in vitro the expression level of the gene in a sample, by measuring in vitro the amount of gene transcripts in a patient's sample. In some embodiments, a nucleic acid microarray can be prepared or purchased. An array typically contains a solid support and at least one nucleic acid (cDNA or oligonucleotide) contacting the support, where the oligonucleotide corresponds to at least a portion of a gene. Any suitable assay platform can be used to determine the presence of hsa-mi'R-31 -3p target gene(s) DBNDD2 and/or EPB41 L4B in a sample. For example, an assay may be in the form of a membrane, a chip, a disk, a test strip, a filter, a microsphere, a multiwell plate, and the like. An assay system may have a solid support on which a nucleic acid (cDNA or oligonucleotide) corresponding to the gene is attached. The solid support may comprise, for example, a plastic, silicon, a metal, a resin, or a glass. The assay components can be prepared and packaged together as a kit for detecting a gene. To determine the expression profile of a target nucleic sample, said sample is labelled, contacted with the microarray in hybridization conditions, leading to the formation of complexes between target nucleic acids that are complementary to probe sequences attached to the microarray surface. The presence of labelled hybridized complexes is then detected. Many variants of the microarray hybridization technology are available to the person skilled in the art.
In another embodiment, the measure in vitro of hsa-miR-31 -3p target gene(s) DBNDD2 and/or EPB41 L4B expression level(s) may be performed by sequencing of transcripts (mRNA or cDNA) of the gene extracted from the patient's sample.
In still another embodiment, the measure in vitro of hsa-miR-31 -3p target gene(s) DBNDD2 and/or EPB41 L4B expression level(s) may be performed by the use of a protein microarray, for measuring the amount of the gene encoded protein in total proteins extracted from the patient's sample.
Classifying the patient Classification based on DBNDD2 and/or EPB41L4B expression level(s)
The higher the expression of hsa-miR-31 -3p target gene(s) DBNDD2 and/or EPB41 L4B is, the better for the patient. Therefore, the higher the level of expression of hsa-miR-31 - 3p target gene(s) DBNDD2 and/or EPB41 L4B is, the more likely the patient is to respond to the EGFR inhibitor treatment. In an embodiment, the patient is considered as "responder", or likely to respond to a treatment with an EGFR inhibitor, when the expression of hsa-miR-31 -3p target gene(s) DBNDD2 and/or EPB41 L4B is higher than a control value.
Such a control value may be determined based on a pool of reference samples, as defined above. In particular, Figure 6 clearly shows that, based on a pool of reference samples, a control value for DBNDD2 and EPB41 L4B level of expression (the logged DBNDD2:EPB41 L4B level of expression) may be defined that permits to predict response or non-response to EGFR inhibitor treatment.
However, in a preferred embodiment, the method further comprises determining a prognostic score or index based on the expression level of at least one o fhsa-mi'R-31 -3p target gene(s) DBNDD2 and EPB41 L4B, wherein the prognostic score indicates whether the patient is likely to respond to the EGFR inhibitor. In particular, said prognosis score may indicate whether the patient is likely to respond to the EGFR inhibitor depending if it is higher or lower than a predetermined threshold value (dichotomized result). In another embodiment, a discrete probability of response or non-response to the EGFR inhibitor may be derived from the prognosis score.
The probability that a patient responds to an EGFR inhibitor treatment is linked to the probability that this patient survives, with or without disease progression, if the EGFR inhibitor treatment is administered to said patient.
As a result, a prognosis score may be determined based on the analysis of the correlation between the expression level of at least one of hsa-miR-31 -3p target gene(s) DBNDD2 and EPB41 L4B and progression free survival (PFS) or overall survival (OS) of a pool of reference samples, as defined above. A PFS and/or OS score, which is a function correlating PFS or OS to the expression level of at least one of hsa-miR-31 -3p target gene(s) DBNDD2 and EPB41 L4B, may thus be used as prognosis score for prediction of response to an EGFR inhibitor. Preferably, a PFS score is used, since absence of disease progression is a clear indicator of response to the EGFR inhibitor treatment.
Experimental data obtained by the inventors shows that the probability for a patient to respond to an EGFR inhibitor treatment is linearly and negatively correlated to the logged expression level of each of DBNDD2 and EPB41 L4B (see Figures 1 , 2 and 5). In a preferred embodiment, said prognosis score is thus represented by the following formula:
Prognosis score = a * x + b, wherein x is the logged expression level ofDBNDD2 (preferably log in base 2, referred to as "log2") and/or EPB41 L4Bmeasured in the patient's sample, and a and b are parameters that have been previously determined based on a pool of reference samples, as defined above.
Depending if a is positive/negative, the patient may then be predicted as responding to the EGFR inhibitor if his/her prognosis score is greater than or equal to/lower than or equal to a threshold value c, and not responding to the EGFR inhibitor if his/her prognosis score is lower than/greater than threshold value c, wherein the value of c has also been determined based on the same pool of reference samples:
· If a is positive, the patient may then be predicted as responding to the EGFR inhibitor if his/her prognosis score is greater than or equal to threshold value c, and not responding to the EGFR inhibitor if his/her prognosis score is lower than threshold value c.
• Alternatively, if a is negative, then the patient may be predicted as responding to the EGFR inhibitor if his/her prognosis score is lower than or equal to threshold value c, and not responding to the EGFR inhibitor if his/her prognosis score is greater than threshold value c.
In another embodiment, a discrete probability of response or non-response to the EGFR inhibitor may be derived from the above a * x + b prognosis score. A precise correlation between the prognosis score and the probability of response to the EGFR inhibitor treatment may be determined based on the same set of reference samples. Depending if a is positive/negative, a higher/lower prognosis score indicates a higher probability of response to the EGFR inhibitor treatment: • If a is positive, the higher the prognosis score, the higher is the probability of response to the EGFR inhibitor treatment (i.e. the lower is the probability of disease progression in the case of a PFS score).
• Alternatively, if a is negative, then the lower the prognosis score, the higher is the probability of response to the EGFR inhibitor treatment (i.e. the lower is the probability of disease progression in the case of a PFS score).
This prediction of whether a patient with a cancer is likely to respond to an EGFR inhibitor may also be made using a nomogram. In a nomogram, points scales are established for each variable of a score of interest. For a given patient, points are allocated to each of the variables by selecting the corresponding points from the points scale of each variable. For a discrete variable (such as a gene expression level), the number of points attributed to a variable is linearly correlated to the value of the variable. For a dichotomized variable (only two values possible), two distinct values are attributed to each of the two possible values or the variable. The score of interest is then calculated by adding the points allocated for each variable (total points). Based on the value of the score, the patient may then be given either a good or bad response prognosis depending on whether the composite score is inferior or superior to a threshold value (dichotomized score), or a probability of response or non-response to the treatment.
It is clear that nomograms are mainly useful when several distinct variables are combined in a composite score (see below the possibility to use composite scores combining DBNDD2 and EPB41 L4B expression levels; DBNDD2 and/or EPB41 L4B expression levels and hsa-mi'R-31 -3p expression level; or DBNDD2 and/or EPB41 L4B expression level(s) and BRAF status). However, a nomogram may also be used to represent a prognosis score based on only one variable, such as DBNDD2 or EPB41 L4B expression level. In this case, total points correspond to points allocated to the single variable.
An example of a nomogram permitting determination of a risk of progression (i.e. of a risk of non-response to EGFR inhibitors) in colorectal cancer patients based on DBNDD2 logged (log2) expression level is displayed in Figure 3 (see also Example 2 below).
Therefore, in an embodiment of the method for predicting whether a patient with a cancer is likely to respond to an EGFR inhibitor according to the invention, the method further comprises determining a risk of non-response based on a nomogram calibrated based on a pool of reference samples. The nomogram may be calibrated based on OS or PFS data. If calibrated based on OS, the risk of non-response corresponds to a risk of death. If calibrated based on PFS, the risk of non-response corresponds to a risk of disease progression (see Figure 3). As explained above, each of DBNDD2 and EPB41 L4B has been found to be a target gene of hsa-miR-31 -3p and to be independently significantly associated to response to EGFR inhibitors, so that the expression level of only one of DBNDD2 and EPB41 L4B may be measured and used for prediction in a method according to the invention.
However, the method according to the invention may also comprise determining the expression levels of both DBNDD2 and EPB41 L4B in the patient's sample, and predicting response or non-response based on the combined expression of DBNDD2 and EPB41 L4B. A composite score combining the expression levels of DBNDD2 and EPB41 L4B may notably be created based on a pool of reference samples. A nomogram may also be used to combine the expression levels of DBNDD2 and EPB41 L4B and obtain the composite score, which may then be correlated to the risk of non-response (i.e. the risk of disease progression for a PFS score) .
Classification based on DBNDD2 and/or EPB41L4B expression level(s) and further parameters positively or negatively correlated to response to EGFR inhibitors
While response to EGFR inhibitors can be predicted based only on the expression level of at least one of hsa-miR-31 -3p target genes DBNDD2 and EPB41 L4B (see Examples 1 , 2 and 3) , the method according to the invention may also comprise determining at least one other parameter positively or negatively correlated to response to EGFR inhibitors.
In this case, a composite score combining the expression level(s) of DBNDD2 and/or EPB41 L4B and the other parameter(s) may notably be created based on a pool of reference samples.
A nomogram, in which points scales are established for each variable of the composite score, may also be used to combine the expression level(s) of DBNDD2 and/or EPB41 L4B and the other parameter(s), and obtain the composite score, which may then be correlated to the risk of non-response (i.e. the risk of disease progression for a PFS score). For a given patient, points are allocated to each of the variables by selecting the corresponding points from the points scale of each variable. For a discrete variable (such as DBNDD2 or EPB41 L4B expression level or age), the number of points attributed to a variable is linearly correlated to the value of the variable. For a dichotomized variable (only two values possible, such as BRAF mutation status or gender), two distinct values are attributed to each of the two possible values or the variable.
A composite score is then calculated by adding the points allocated for each variable (total points) . Based on the value of the composite score, the patient may then be given either a good or bad response prognosis depending on whether the composite score is inferior or superior to a threshold value (dichotomized score), or a probability of response or non-response to the treatment.
The points scale of each variable, as well the threshold value over/under which the response prognosis is good or bad or the correlation between the composite score and the probability of response or non-response may be determined based on the same pool of reference samples.
Such other parameters positively or negatively correlated to response to EGFR inhibitors may notably be selected from:
· age;
• gender;
• the expression level of hsa-miR-31 -3p, which may be measured at the genomic and/or nucleic (in particular by measuring the amount of nucleic acid transcripts of each gene)and/or proteic level, by any method disclosed above for measuring the expression level of DBNDD2 and EPB41 L4B; and/or
• the presence or absence of at least one mutation positively or negatively correlated to response to EGFR inhibitors.
Such mutations may be detected by any method known to those skilled in the art and notably include those mentioned in Table 3 below:
Figure imgf000017_0001
* Mutations are defined by mention of the codon number in the protein, preceded by the one letter code for the wild-type amino acid, and optionally followed by the replacement amino acid. When no replacement amino acid is mentioned, the replacement amino acid may be any amino acid different from the wild-type amino acid. EGFR Inhibitors
The present invention makes it possible to predict a patient's responsiveness to one or more epidermal growth factor receptor (EGFR) inhibitors prior to treatment with such agents.
The EGRF inhibitor may be an EGFR tyrosine kinase inhibitor, or may alternatively target the extracellular domain of the EGFR target. In certain embodiments, the EGFR inhibitor is a tyrosine kinase inhibitor such as Erlotinib, Gefitinib, or Lapatinib, or a molecule that targets the EGFR extracellular domain such as Cetuximab or Panitumumab.
Preferably the EGFR inhibitor is an anti-EGFR antibody, preferably a monoclonal antibody, in particular Cetuximab or Panitumumab.
Molecules that target the EGFR extracellular domain, including anti-EGFR monoclonal antibodies such as Cetuximab or Panitumumab, are mainly used in the treatment of colorectal cancer or breast cancer treatment. As a result, if the patient's cancer is colorectal cancer (in particular metastatic colorectal cancer) or breast cancer, then the method according to the invention may preferably be used to predict response to molecules that target the EGFR extracellular domain, and in particular to anti-EGFR monoclonal antibodies, such as Cetuximab or Panitumumab.
Conversely, tyrosine kinase EGFR inhibitors are mainly used in the treatment of lung cancer (in particular non-small cell lung cancer, NSCLC), so that if the patient's cancer is lung cancer (in particular non-small cell lung cancer, NSCLC), then the method according to the invention may preferably be used to predict response to tyrosine kinase EGFR inhibitors, such as Erlotinib, Gefitinib, or Lapatinib.
In pancreatic cancer or head and neck cancer (in particular squamous cell carcinoma of the head and neck (SCCHN)), both tyrosine kinase EGFR inhibitors and anti-EGFR monoclonal antibodies are being tested as therapy, so that if the patient's cancer is pancreatic cancer or head and neck cancer (in particular squamous cell carcinoma of the head and neck (SCCHN)), then the method according to the invention may be used to predict response either to tyrosine kinase EGFR inhibitors (such as Erlotinib, Gefitinib, or Lapatinib) or to anti-EGFR monoclonal antibodies (such as Cetuximab or Panitumumab).
Cetuximab and Panitumumab are currently the clinically mostly used anti-EGFR monoclonal antibodies. However, further anti-EGFR monoclonal antibodies are in development, such as Nimotuzumab (TheraCIM-h-R3), Matuzumab (EMD 72000), and Zalutumumab (HuMax-EGFr). The method according to the invention may also be used to predict response to these anti-EGFR monoclonal antibodies or any other anti-EGFR monoclonal antibodies (including fragments) that might be further developed, in particular if the patient is suffering from colorectal cancer (in particular metastatic colorectal cancer), breast cancer, pancreatic cancer or head and neck cancer (in particular squamous cell carcinoma of the head and neck (SCCHN)). Similarly, Erlotinib, Gefitinib, and Lapatinib are currently the clinically mostly used tyrosine kinase EGFR inhibitors. However, further tyrosine kinase EGFR inhibitors are in development, such as Canertinib (CI-1033),Neratinib (HKI-272), Afatinib (BIBW2992), Dacomitinib (PF299804,PF-00299804), TAK-285, AST-1306, ARRY334543, AG-1478 (Tyrphostin AG-1478), AV-412, OSI-420 (DesmethylErlotinib), AZD8931 , AEE788 (NVP- AEE788), Pelitinib (EKB-569), CUDC-101 , AG 490, PD153035 HCl, XL647, and BMS-599626 (AC480).The method according to the invention may also be used to predict response to these tyrosine kinase EGFR inhibitors or any other tyrosine kinase EGFR inhibitors that might be further developed, in particular if the patient is suffering from of lung cancer (in particular non-small cell lung cancer, NSCLC), pancreatic cancer, or head and neck cancer (in particular squamous cell carcinoma of the head and neck (SCCHN)) .
Kits
The present invention also relates to a kit for determining whether a patient with a cancer is likely to respond to an epidermal growth factor receptor (EGFR) inhibitor, comprising or consisting of:
a) reagents for determining the expression level of at least one target gene of hsa- miR-31 -3p (SEQ ID NO:1 ) miRNA in a sample (preferably a tumor sample, such as a tumor biopsy or whole or part of a tumor surgical resection) of said patient, wherein said target gene of hsa-miR-31 -3p is selected from DBNDD2 and EPB41 L4B, and
b) reagents for determining at least one other parameter positively or negatively correlated to response to EGFR inhibitors.
Such reagents may notably include reagents for:
i) determining the expression level of at least one miRNA positively or negatively correlated to response to EGFR inhibitors, in particular hsa- miR-31 -3p (SEQ ID N0:1 ) miRNA or particular hsa-miR-31 -5p (SEQ ID NO:34) in a sample (preferably a tumor sample, such as a tumor biopsy or whole or part of a tumor surgical resection) of said patient, and /or, ii) detecting at least one mutation positively or negatively correlated to response to EGFR inhibitors, such as those mentioned in Table 3 above.
Reagents for determining the expression level of at least one of hsa-miR-31 -3p target gene(s) DBNDD2 and EPB41 L4B or of at least one miRNA positively or negatively correlated to response to EGFR inhibitors, in particular hsa-miR-31 -3p itself or hsa-miR- 31 -5p, in a sample of said patient, may notably comprise or consist of primers pairs (forward and reverse primers) and/or probes specific for at least one of hsa-miR-31 -3p target gene(s) DBNDD2 and EPB41 L4B or a microarray comprising a sequence specific for at least one of hsa-miR-31 -3p target gene(s) DBNDD2 and EPB41 L4B. The design of primers and/or probe can be easily made by those skilled in the art based on the sequences of DBNDD2 and/or EPB41 L4B disclosed in Tables 1 and 2 above.
Reagents for detecting at least one mutation positively or negatively correlated to response to EGFR inhibitors may include at least one primer pair for amplifying whole or part of the gene of interest before sequencing or a set of specific probes labeled with reporter dyes at their 5' end, for use in an allelic discrimination assay, for instance on an ABI 7900HT Sequence Detection System (Applied Biosystems, Foster City, CA) (see Laurent-Puig P, et al, J ClinOncol. 2009, 27(35):5924-30 and Lievre et al. J ClinOncol. 2008 Jan 20;26(3):374-9 for detection of BRAF mutation V600).
The kit of the invention may further comprise instructions for determining whether the patient is likely to respond to the EGFR inhibitor based on the expression level of at least one of hsa-miR-31 -3p target gene(s) DBNDD2 and EPB41 L4B and the other tested parameter. In particular, a nomogram including points scales of all variables included in the composite score and correlation between the composite score (total number of points) and the prediction (response/non-response or probability of response or non- response) may be included.
Drugs, therapeutic uses and methods of treating
The method of the invention predicts patient responsiveness to EGFR inhibitors at rates that match reported clinical response rates for the EGFR inhibitors.
It is thus further provided a method for treating a patient with a cancer, which method comprises administering to the patient at least one EGFR inhibitor, wherein the patient has been predicted (or classified) as "responder" or "likely to respond" by the method as described above.
In particular, the invention concerns a method for treating a patient affected with a cancer, which method comprises (i) determining whether the patient is likely to respond to an EGFR inhibitor, by the method according to the invention, and (ii) administering an EGFR inhibitor to said patient if the patient has been determined to be likely to respond to the EGFR inhibitor.
The method may further comprise, if the patient has been determined to be unlikely to respond to the EGFR inhibitor a step (in) of administering an alternative anticancer treatment to the patient. Such alternative anticancer treatment depends on the specific cancer and on previously tested treatments, but may notably be selected from radiotherapy, other chemotherapeutic molecules, or other biologies such as monoclonal antibodies directed to other antigens (anti-Her2, anti-VEGF, anti-EPCAM, anti-CTLA4...). In particular, in the case of colorectal cancer, if the patient has been determined to be unlikely to respond to the EGFR inhibitor, the alternative anticancer treatment administered in step (iii) may be selected from:
• a VEGF inhibitor, in particular an anti-VEGF monoclonal antibodies (such as bevacizumab), advantageously in combination with FOLFOX (a combination of leucovorin (folinic acid), 5-fluorouracil (5-FU), and oxaliplatin) or FOLFIRI (a combination of leucovorin (folinic acid), 5-fluorouracil (5-FU), and irinotecan) chemotherapy.
• Alternatively, if the patient has already been treated unsuccessfully with a VEGF inhibitor, optionally in combination with FOLFOX or FOLFIRI chemotherapy, it may be administered with 5-FU, optionally in combination with Mitomycin B. Best supportive care, defined as a treatment administered with the intent to maximize quality of life without a specific antineoplastic regimen (i.e. not an anticancer treatment) may further be administered to the patient. Another subject of the invention is an EGFR inhibitor, for use in treating a patient affected with a cancer, wherein the patient has been classified as being likely to respond by the method as defined above. The invention also relates to an EGFR inhibitor for use in treating a patient affected with a cancer, wherein said treatment comprises a preliminary step of predicting if said patient is or not likely to respond to the EGFR inhibitor by the method as defined above, and said EGFR inhibitor is administered to the patient only is said patient has been predicted as likely to respond to the EGFR inhibitor by the method as defined above. Said patient may be affected with a colorectal cancer, more particularly a metastatic colorectal cancer. Alternatively, said patient may be affected with a breast cancer, in particular a triple negative breast cancer. Alternatively, said patient may be affected with a lung cancer, in particular a non-small cell lung cancer (NSCLC). Alternatively, said patient may be affected with a head and neck cancer, in particular a squamous-cell carcinoma of the head and neck. Alternatively, said patient may be affected with a pancreatic cancer. The invention also relates to the use of an EGFR inhibitor for the preparation of a medicament intended for use in the treatment of cancer in patients that have been classified as "responder" by the method of the invention as described above.
In a preferred embodiment the EGFR inhibitor is an anti-EGFR antibody, preferably cetuximab or panitumumab. Alternatively, the EGFR inhibitor may be a tyrosine kinase EGFR inhibitor, in particular Erlotinib, Gefitinib, or Lapatinib.
In preferred embodiments:
• the patient is afflicted with a colorectal cancer, in particular a metastatic colorectal cancer, and the EGFR inhibitor is an anti-EGFR antibody, preferably cetuximab or panitumumab;
· the patient is afflicted with a breast cancer, in particular a triple negative breast cancer, and the EGFR inhibitor is an anti-EGFR antibody, preferably cetuximab or panitumumab;
• the patient is afflicted with a lung cancer, in particular a non-small cell lung cancer (NSCLC), and the EGFR inhibitor is a tyrosine kinase EGFR inhibitor, in particular Erlotinib, Gefitinib, or Lapatinib;
• the patient is afflicted with a head and neck cancer, in particular a squamous- cell carcinoma of the head and neck, or a pancreatic cancer, and the EGFR inhibitor is an anti-EGFR antibody (preferably cetuximab or panitumumab) or a tyrosine kinase EGFR inhibitor (in particular Erlotinib, Gefitinib, or Lapatinib).
The examples and figures illustrate the invention without limiting its scope. EXAMPLES
Example 1 : DBNDD2 and EPB41 L4B are targets of hsa-miR-31 -3p and independently predict response to EGFR inhibitors
PATIENTS AND METHODS
Patients
The set of patients was made of 20mCRC(metastatic colorectal cancer) patients, 14 males, 6 females. The median of age was 66.49±11.9 years. All patients received a combination of irinotecan and cetuximab. The number of chemotherapy lines before the introduction of Cetuximab was recorded. The median of follow-up until progression was 20 weeks and the median overall survival was 10 months. All tumor sample came from resections and were fixed in formalin and paraffin embedded (FFPE).
Cell culture and transfection
We selected 3 colorectal adenocarcinoma cell lines from the American Type Culture Collection (ATCC, Manassas, CA) that express weaklyhsa-miR-31 -3p: HTB-37, CCL-222 and CCL-220-1. HTB-37 cells were maintained in a Dulbecco's Modified Eagle Medium (DMEM) culture medium with stable glutamine with 20% Fetal Bovine serum and 1% Penicillin/Streptomycin. CCL-222 and CCL-220-1 cells were maintained in a RPMI 1640 culture media with stable glutamine with 10% fetal bovine serum. The cells were incubated at a temperature of 37 °C with 5% C02.
All the cells were transfected with miRVana mi'RNA mimic negative control or hsa-mi'R- 31 -3p miRVana mi'RNA mimic (Ambion). For CCL-222, transfections were done with 2μΙ of lipofectamine RNAiMax with reverse transfection protocol according to the manufacturer's protocol using 25pmol of MiRNA mimic and 60 000 cells in a 12 wells plate. For CCL-220-1 and HBT27, transfections were done using 4μΙ of RiboCellin (BioCellChallenge, Toulon, France) according to the manufacturer's protocol using 12.5pmol of mi'RNA mimic and 100 000 cells in a 12 wells plate. For all the cell lines, cells were harvested 24h hours after transfection and Qiazol was used to protect RNA until extraction of total RNA with mi'RNeasy extraction kit (Qiagen). To assess for the efficacy of the transfection, specific quantification of mi'RNA hsa-miR-31 -3p expression level was done as described below.
Measurement of gene expression
Gene expression microarray was performed using the AffymetrixHuman Gene 1 .0. Fifty ng of total RNA was reverse transcribed following the Ovation PicoSL WTA System V2 (Nugen, San Carlos, CA). Then, amplification was done based on SPIA technology. After purification according to Nugen protocol, 2.5 pg of single strand DNA was used for fragmentation and biotin labelling using Encore Biotin Module (Nugen). After control of fragmentation using Bioanalyzer 2100, cDNA was then hybridized to GeneChip® human Gene 1.0 ST (Affymetrix) at 45 ° C for 17 hours. After hybridization, chips were washed on the fluidic station FS450 following specific protocols (Affymetrix) and scanned using the GCS3000 7G. The image was then analyzed with Expression Console software (Affymetrix) to obtain raw data (CELfiles) and metrics for Quality Controls.
qRT-PCR validation of the target expression on cell lines and FFPE patients samples were performed on 20ng of total RNA for FFPE samples or 50ng of total RNA cell culture samples using ABI7900HT Real-Time PCR System (Applied Biosystem). All reactions were performed in triplicate. Expression levels were normalized to the RNA18S and GAPDH levels through the AACt method.
In silico analysis
We developed a data portal integrating up-to-date microRNA target predictions from six individual prediction databases (PITA, picTar 5-way, Targetscan, microRNA.org, MicroCosm and mi'RDB). This portal allows to determine microRNAs potentially co- targeted by a list of candidate genes, taking into account the number of microRNA prediction databases predicting each microRNA/target relationship and the rank of prediction of each miRNA from individual prediction databases. This database has been updated in November 2012 to perform the reported analysis.
Statistical analyses
Survival statistical analysis was performed using the R packages 'survival' and 'rms' . Univariate and multivariate analyses used a Cox proportional regression hazard model and generated a hazard ratio (HR). Nomograms were developed based on Cox proportional regression hazard models, which predict the probability of free -progression survival.
False-discovery rate (FDR)-adjusted p-values were calculated using the Benjamini and Hochberg procedure for multiple testing correction. The cor. test function was used to calculate Pearson correlations between expression values together with matching p- values. Statistical significance was set at p<0.05 for all analyses.
RESULTS
Three CRC cell lines that weakly express hsa-miR-31 -3p were transfected with hsa-miR- 31 -3p mimic or with a mimic control. The transfection efficacy was attested by an average rise of hsa-miR-31 -3p level of 1500 times without mortality or growth defect. Expression profile analysis of the transfected cells allowed us to identify 47 genes significantly down-regulated (fc<0.77, p<0.05), and 27 genes significantly up-regulated by hsa-miR-31 -3p (fc<1 .3, p<0.05), as described in Table 4 below. Table 4: List of the genes with a fc<0.77 or fc>1 .3 and a pvalue>0.05 identified in the expression array made on the 3 cell lines (fc: fold change in expression between cell lines transfected with hsa-miR-31 -3p mimic and cell lines transfected with a mimic control)
Figure imgf000024_0001
As the role of a microRNA includes degradation of its transcript target, we studied if the database including information from 6 web-available predicts the 54 down -regulated genes as hsa-miR-31 -3p putative target. The database may be queried either by miRNA name, or by gene name. When a miRNA name is queried, the database returns a list of candidate target genes, ranked by order of probability (from the most probable to the less probable) that the genes are true targets of the queried miRNA, based on structural and potential experimental data included in the database. Conversely, when a gene name is queried, the database returns a list of miRNA candidates, ranked by order of probability (from the most probable to the less probable) that the mi'RNAs truly target the queried gene, based on structural and potential experimental data included in the database. The database was queried with hsa-miR-31 -3p name and with the names of genes found to be down-regulated in CRC cell lines overexpressing hsa-miR-31 -3p (47 genes, cf Table 4) .
Table 5below shows down-regulated genes of Table 4, including DBNDD2 and EPB41 L4B, which were identified as a putative direct target of has-miR-31 -3p. lt also indicates the rank of hsa-miR-31 -3p if the database was queried using the gene name, and the rank of the gene if the database was queried using hsa-miR-31 -3p name.
Hsa-miR-31 -3p ranking by the
Gene ranking by hsa-miR-31 -
Genes ID gene/ Number of predicted
3p(on 1620 putative targets) microRNA
AMFR 72/216 293
B4GALT1 94/223 293
CA12 48/182 293
CSGALNACT2 89/242 293
DBNDD2 41 /139 293 EHBP1 13/361 10
EPB41 L4B 101/425 86
FEM1A 21 /125 293
GMFB 211 348 293
HAUS4 1 /110 16
HSPB11 37/279 101
LSM14B 52/288 101
OSGIN2 119/289 293
OSTM1 86/305 67
PCP4 18/109 115
PLEKHB2 93/257 293
PNP 9/216 31
P0LR2K 5/162 2
POTEM 47/210 293
SEC31 A 9/238 78
STAT3 37/240 166
UBE2H 120/303 293
VDAC1 29/213 173
WDR45L 39/154 293
XPNPEP3 145/583 293
Table 5: Target predictions from in silico database are indicated for the down- regulated genes depending on the request: Column 2: database was interrogated with a gene of interest, and reported all candidate microRNAs potentially targeting this gene, ranked from the most likely to the less likely. The rank of hsa-miR-31 -3p and the total number of microRNA candidates are indicated; Column 3:database was interrogated with hsa-miR-31 -3p, and reported all putative targets, ranked from the most likely to the less likely for a total of 1620 putative targeted genes. Then rank of the queried gene is indicated. Only down-regulated genes listed in hsa-miR-31 -3p 1620 putative targeted genes are presented in Table 5. Data relating to DBNDD2 and EPB41 L4B are in bold.
Among the 47 down-regulated genes, 25 were predicted to be putative direct target of hsa-miR-31 -3p and displayed a good rank in the prediction database. This number and the ranking of the genes are significant (P<0.0001 for both test by permutation test) . As expected, none but one of the 27 up-regulated genes in the cells transfected with miR- 31 -3p was predicted to be a target of hsa-miR-31 -3p, and the only predicted one was the last target ranked.
The 25 putative direct target genes and the 27 indirect target genes were validated on qRT-PCR, out of these47genes, 45 displayed an expression level comparable to the level obtained in the array.
Finally, expression of these genes was analyzed in patient FFPE tumor samples and 2 of them showed a significant negative correlation with hsa-miR-31 -3p expression levels: DBNDD2 and EPB41 L4B (see Figure 1A and 1 B) . In addition, using non-parametric differential analysis, these 2 genes were found to be associated to the progression free survival (p=0.004, for DBNDD2 and p=0.025 for EPB41 L4B) . Together, these results suggest that expression of DBNDD2 and EPB41 L4B could distinguish between mCRC patients with poor or good prognosis, i.e. between non-responders and responders mCRC patients.
Example 2: Creation of a tool with DBNDD2 and EPB41L4B expression to predict response to EGFR inhibitors
PATIENTS AND METHODS Patients
The set of patients was made of 20mCRC patients, 13 males and 7 females. The median of age was 67±11 .2 years. All had a metastatic disease at the time of the inclusion. All these patients developed a KRAS wild type metastatic colon cancer. All patients were considered refractory to a 5-fluorouracil-based regimen combined with irinotecan and oxaliplatin. They received an anti-EGFR-based chemotherapy, 8 patients with panitumumab, 10 patients with cetuximab and 2 patients received a combination of panitumumab and cetuximab. The number of chemotherapy lines before the introduction of Cetuximab and panitumumab was recorded. The median of follow-up until progression was 21 weeks and the median overall survival was 8.9 months. Measurement of gene expression
qRT-PCR of DBNDD2 and EPB41 L4B expression on FFPE patients samples were performed on 20ng of total RNA using ABI7900HT Real-Time PCR System (Applied Biosystem) . All reactions were performed in triplicate. Expression levels were normalized to the GAPDH levels through the A Ct method. Statistical analyses
Survival statistical analysis was performed using the R packages 'survival' and 'rms' . Univariate and multivariate analyses used a Cox proportional regression hazard model and generated a hazard ratio (HR). Nomograms were developed based on Cox proportional regression hazard models, which predict the probability of free -progression survival.
Gene and miRNA expression value comparison analyses were done using non-parametric test (Kruskal-Wallis tests) with the pairwise Wilcox test function in R.
The cor.test function was used to calculate Pearson correlations between expression values together with matching p-values. Statistical significance was set at p<0.05 for all analyses. RESULTS
Expression of DBNDD2 and EPB41 L4B was analyzed in the tumor samples. Statistical analyses showed a significant negative correlation with hsa-mi'R-31 -3p expression levels: (see Figure 2for DBNDD2). ln addition, using non-parametric differential analysis, these 2 genes were found to be associated to the progression free survival (p=0.025, for DBNDD2) . Based on this results, to obtain a tool for predicting response of mCRC patient treated with anti-EGFR, multivariate Cox proportional hazards models status and log2 of the gene expression as covariate were used to construct a nomogram based on PFS, thus permitting to predict the risk of progression (i.e. the risk of non-response, see Figures 3 and 4).
Example 3: Replication of the predictive value of DBNDD2 and EPB41 L4B to EGFR inhibitors in a new and independent cohort PA TIENTS AND METHODS
Patients
The set of patients was made of 42 mCRC(metastatic colorectal cancer) patients, 27 males and 15 females. The median of age was 59±12.1years. All had a metastatic disease at the time of the inclusion. All patients were treated with 3rd line therapy by a combination of irinotecan and panitumumab after progression with oxaliplatin and irinotecan chemotherapy based regimens. The median of follow-up until progression was 23 weeks and the median overall survival was 9.6 months. 26 samples were available in FFPE and 16 in frozen tissue.
Measurement of gene expression
qRT-PCR validation of the target expression on frozen or FFPE patients samples were performed on 20ng of total RNA using ABI7900HT Real-Time PCR System (Applied Biosystem). All reactions were performed in triplicate. Expression levels were normalized to the RNA18S or GAPDH levels through the AACt method.
Statistical analyses
Survival statistical analysis was performed using the R packages 'survival' and 'rms' . Univariate and multivariate analyses used a Cox proportional regression hazard model. Gene and miRNA expression value comparison analyses were done using non-parametric test (Kruskal-Wallis tests) with the pairwise Wilcox test function in R.
Statistical significance was set at p<0.05 for all analyses.
RESULTS
Expression of DBNDD2 and EPB41 L4B was analyzed in the patient tumor FFPE samples. They showed a significant negative correlation with hsa-miR-31 -3p expression levels: (see Figure 5A and 5B).A correlation between the expression of these two genes and prediction of response/non-response calculated based on the expression level of hsa- mi'R-31 -3p as described in patent application PCT/EP2012/073535 was found (see Figure 6).
Using a cox model, these 2 genes were found to be associated to the progression free survival (p=0.004 for DBNDD2 with GAPDH normalization and p=0.027 for EPB41 L4B with RNA 18S normalization).
These results confirm that expression of DBNDD2 and EPB41 L4B could discriminate mCRC patients with poor or good prognosis, i.e. between non-responders and responders mCRC patients.
BIBLIOGRAPHIC REFERENCES
Albitar Let al. Mol Cancer 2010;9: 166;
Ambros V et al, RNA 2003 9(3):277-279 ;
Bair E. & R Tibshirani, PLOS Biology 2:51 1 -522, 2004;
Bos. Cancer Res 1989;49:4682-4689;
Bustin et al. , 2005, Clin. Sci. , 109:365-379 ;
Chan SL et al. Expert OpinTherTargets. 2012 Mar; 16Suppl 1 :S63-8;
Chang KW et al. Oral Oncol.2012 Jul 30,
Chu H et al. Mutagenesis.2012 Oct 15;
Ciardello F et al. N Engl J Med. 2008 Mar 13;358(11 ) :1160-74 ;
Cox, D. R. (1972) . Journal of the Royal Statistical Society, Series B 34 (2), 187-220;
Cunningham et al, N Engl Med 2004;351 : 337-45;
Demiralay et al. Surgical Science, 2012, 3, 11 1 -115;
Edkins et al. Cancer BiolTher. 2006 August ; 5(8) : 928-932
Eisenhauer et al, European Journal of Cancer, 2009, 45:228-247 ;
Griffiths-Jones S. NAR 2004 32(Database lssue) :D109-D111 ;
Griffiths-Jones S et al. NAR 2006 34(Database lssue):D140-D144;
Griffiths-Jones S et al. NAR 2008 36(Database lssue):D154-D158;
HatakeyamaH.et al. PLoS One.2010 Sep 13;5(9):e12702;
Kozomara A et al. NAR 2011 39(Database lssue):D152-D157 ;
Laurent-Puig P, et al, J Clin Oncol. 2009, 27(35):5924-30;
Leboulleux S et al. Lancet Oncol.2012 Sep;13(9):897-905;
Leslie KK et al. GynecolOncol.2012 Nov;127(2) :345-50;
Li Y et al. OncolRep. 2010 Oct;24(4):1019-28;
Liebner DA et al. TherAdvEndocrinolMetab. 201 1 Oct;2(5):173-95;
Lievre et al, Cancer Res. 2006 66(8) :3992-5;
Lievre et al. J ClinOncol.2008 Jan 20;26(3) :374-9 ;
Mimeault M et al. PLoS One.2012;7(2) :e31919;
MosakhaniN .et al. Cancer Genet.2012 Oct 22.doi :pii: S2210-7762(12)00229-3. 10.1016/j .cancergen.2012.08.003;
Ogino S, et al. J Mol Diagn 2008;7:413-21 ;
Pan J et al. Head Neck. 2012 Sep 13;
Ragusa M. et al. Mol Cancer Ther. 2010 Dec;9(12):3396-409;
Schulz WA, et al. BMC Cancer.2010 Sep 22;10:505;
Shepherd F A, et al, N Engl J Med 2005; 353:123-132;
Tarn et al. Clin Cancer Res 2006; 12:1647-1653;
Thomasson M et al. Br J Cancer 2003, 89: 1285-1289; Thomasson M et al. 2012 May 3;5:216;
U.S. Pat. No. 7,101 ,663;
Wang J, et al. Prostate.2006 Nov 1 ;66(15):1641 -52;
Wheeler DL et al. Nat RevClinOncol.2010 September ; 7(9): 493-507 ; WO2009/080437 ;
WO2010/121238;
WO2011 /135459;
WO2010065940 ;
WO2010059742 ;
WO2009131710 ;
WO2007112097;
WO2011017106;
WO2010127338;
WO2007072225;
WO2008138578;
Xiao W et al. 2012. PLoS ONE 7(6) : e38648;
Yin H et al. Biochemistry.2006 Apr 25;45(16) :5297-308;
Yu H et al. Mol Cancer Res 2010;8:1501 -1512;
Zeineldin R et al. J Oncol. 2010;2010:414676,
Zhao L. et al. Int J Biochem Cell Biol. 2012 Nov;44(1 1 ) :2051 -9.

Claims

1 . An in vitro method for predicting whether a patient with a cancer is likely to respond to an epidermal growth factor receptor (EGFR) inhibitor, which method comprises determining the expression level of at least one target gene of hsa- miR-31 -3p (SEQ ID NO:1 ) mi'RNA in a tumor sample of said patient, wherein said target gene of hsa-mi'R-31 -3p is selected from DBNDD2 and EPB41 L4B.
2. The method of claim 1 , wherein the patient has a KRAS wild-type cancer.
3. The method of any one of claims 1 to 2, wherein the patient is afflicted with a cancer selected from colorectal, lung, breast, ovarian, endometrial, thyroid, nasopharynx, prostate, head and neck, liver, kidney, pancreas, bladder, and brain.
4. The method of claim 3, wherein the cancer is a colorectal cancer, in particular a metastatic colorectal cancer.
5. The method of any one of claims 1 to 4, wherein the EGFR inhibitor is an anti- EGFR antibody, in particular cetuximab or panitumumab.
6. The method of any one of claims 1 to 5, wherein the sample is a tumor tissue biopsy or whole or part of a tumor surgical resection.
7. The method of any one of claims 1 to 6, wherein the level of expression of said at least one target gene of hsa-miR-31 -3p is determined at the nucleic acid level by measuring in vitro the amount of transcripts produced by said target gene(s) of hsa-miR-31 -3p, preferably by quantitative RT-PCR.
8. The method of any one of claims 1 to 7, wherein the higher the level of expression of said at least one target gene of hsa-mi'R-31 -3pis, the more likely the patient is to respond to the EGFR inhibitor treatment.
9. The method of any one of claims 1 to 8, further comprising determining a prognostic score based on the expression level of said at least one target gene of hsa-miR-31 -3p, wherein the prognostic score indicates whether the patient is likely to respond to the EGFR inhibitor.
10. The method of any one of claims 1 to 9, wherein the prognostic score is of formula:
Prognosis score = a * x + b,
wherein:
• x is the logged expression level of DBNDD2 measured in the patient's sample, • a and b are parameters that have been previously determined based on a pool of reference samples, and
• the patient is predicted as responding or non- responding to the EGFR inhibitor if his/her prognosis score is greater or lower than a threshold value c, wherein the value of c has been determined based on the same pool of reference samples :
o If a is positive, then the patient is predicted as responding to the EGFR inhibitor if his/her prognosis score is greater than or equal to threshold value c, and not responding to the EGFR inhibitor if its prognosis score is lower than threshold value c,
o If a is negative, then the patient may be predicted as responding to the EGFR inhibitor if his/her prognosis score is lower than or equal to threshold value c, and not responding to the EGFR inhibitor if his/her prognosis score is greater than threshold value c.
11. The method of any one of claims 1 to 9, wherein the prognostic score is of formula:
Prognosis score = a * x + b,
wherein:
• x is the logged expression level of DBNDD2 measured in the patient's sample,
• a and b are parameters that have been previously determined based on a pool of reference samples, and
• depending if a is positive or negative:
o If a is positive, the higher the prognosis score, the higher is the probability of response to the EGFR inhibitor treatment;
o if a is negative, then the lower the prognosis score, the higher is the probability of response to the EGFR inhibitor treatment.
12. The method of any one of claims 1 to 8, further comprising determining a risk of non-response based on a nomogram calibrated based on a pool of reference samples.
13. The method of any one of claims 1 to 12, further comprising determining at least one other parameter positively or negatively correlated to response to EGFR inhibitors, and calculating a composite score taking into account the expression level of said at least one target gene of hsa-miR-31 -3p and said other parameter(s), wherein the composite score indicates whether the patient is likely to respond to the EGFR inhibitor.
14. A kit for determining whether a patient with a cancer is likely to respond to an epidermal growth factor receptor (EGFR) inhibitor, comprising or consisting of: a) reagents for determining the expression level of at least one target gene of hsa-miR-31 -3p (SEQ ID NO:1 ) miRNA in a sample of said patient, wherein said target gene of hsa-miR-31 -3p is selected from DBNDD2 and EPB41 L4B, and
b) reagents for determining at least one other parameter positively or negatively correlated to response to EGFR inhibitors, wherein said reagents are selected from:
i) reagents for determining the expression level of at least one miRNA positively or negatively correlated to response to EGFR inhibitors, in particular hsa-miR-31 -3p (SEQ ID NO:1 ) miRNA or hsa-miR-31 -5p (SEQ ID NO:34) miRNA, and/or
ii) reagents for detecting at least one mutation positively or negatively correlated to response to EGFR inhibitors.
15. An EGFR inhibitor for use in treating a patient affected with a cancer, wherein the patient has been classified as being likely to respond to the EGFR inhibitor by the method according to any one of claims 1 to 13.
16. An EGFR inhibitor for use in treating a patient affected with a cancer, wherein said treatment comprises a preliminary step of predicting if said patient is or not likely to respond to the EGFR inhibitor by the method according to any one of claims 1 to 13, and said EGFR inhibitor is administered to the patient only is said patient has been predicted as likely to respond to the EGFR inhibitor by the method according to any one of claims 1 to 13.
17. A method for treating a patient affected with a cancer, which method comprises:
(i) determining whether the patient is likely to respond to an EGFR inhibitor, by the method according to the invention, and
(ii) administering an EGFR inhibitor to said patient if the patient has been determined to be likely to respond to the EGFR inhibitor.
18. The method according to claim 17, further comprise, if the patient has been determined to be unlikely to respond to the EGFR inhibitor a step (iii) of administering an alternative anticancer treatment to the patient.
19. The method according to claim 18, wherein said alternative anticancer treatment is selected from:
a) a VEGF inhibitor,
b) a VEGF inhibitor in combination with FOLFOX,
c) a VEGF inhibitor in combination with FOLFIRI,
d) 5-FU, and
e) 5-FU in combination with Mitomycin B.
PCT/EP2014/075651 2013-11-26 2014-11-26 A method for predicting responsiveness to a treatment with an egfr inhibitor WO2015078906A1 (en)

Priority Applications (9)

Application Number Priority Date Filing Date Title
AU2014356506A AU2014356506A1 (en) 2013-11-26 2014-11-26 A method for predicting responsiveness to a treatment with an EGFR inhibitor
CA2931176A CA2931176A1 (en) 2013-11-26 2014-11-26 A method for predicting responsiveness to a treatment with an egfr inhibitor
JP2016534255A JP2017503478A (en) 2013-11-26 2014-11-26 Method for predicting responsiveness to treatment with an EGFR inhibitor
KR1020167016884A KR20160089488A (en) 2013-11-26 2014-11-26 A method for predicting responsiveness to a treatment with an egfr inhibitor
CN201480064646.4A CN105765081A (en) 2013-11-26 2014-11-26 A method for predicting responsiveness to a treatment with an egfr inhibitor
MX2016006782A MX2016006782A (en) 2013-11-26 2014-11-26 A method for predicting responsiveness to a treatment with an egfr inhibitor.
EP14805833.2A EP3074530A1 (en) 2013-11-26 2014-11-26 A method for predicting responsiveness to a treatment with an egfr inhibitor
US15/038,826 US20160376661A1 (en) 2013-11-26 2014-11-26 A method for predicting responsiveness to a treatment with an egfr inhibitor
BR112016012001A BR112016012001A2 (en) 2013-11-26 2014-11-26 In vitro method of predicting whether a cancer patient is prone to react to an inhibitor, kit, egfr inhibitor, and treatment method of affected cancer patient

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP13306619 2013-11-26
EP13306619.1 2013-11-26

Publications (1)

Publication Number Publication Date
WO2015078906A1 true WO2015078906A1 (en) 2015-06-04

Family

ID=49713039

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2014/075651 WO2015078906A1 (en) 2013-11-26 2014-11-26 A method for predicting responsiveness to a treatment with an egfr inhibitor

Country Status (10)

Country Link
US (1) US20160376661A1 (en)
EP (1) EP3074530A1 (en)
JP (1) JP2017503478A (en)
KR (1) KR20160089488A (en)
CN (1) CN105765081A (en)
AU (1) AU2014356506A1 (en)
BR (1) BR112016012001A2 (en)
CA (1) CA2931176A1 (en)
MX (1) MX2016006782A (en)
WO (1) WO2015078906A1 (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN105701365A (en) * 2016-01-12 2016-06-22 西安电子科技大学 Cancer-related genes finding method by using miRNA expression data
WO2018034301A1 (en) * 2016-08-17 2018-02-22 中外製薬株式会社 Combination drug of anti-epiregulin antibody and anti-egfr antibody
JPWO2017187937A1 (en) * 2016-04-28 2019-02-28 デンカ株式会社 Method for determining resistance of cancer cells to epidermal growth factor receptor inhibitors

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1888095A4 (en) 2005-05-13 2009-08-26 Whitehead Biomedical Inst Modulators of alpha-synuclein toxicity
WO2009086306A1 (en) * 2007-12-21 2009-07-09 Whitehead Institute For Biomedical Research Modulators of alpha-synuclein toxicity
EP3722417A4 (en) * 2017-12-08 2021-09-22 Kyo Diagnostics K.K. Cancer spheroid production method and method for selecting colon cancer patients

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009021673A1 (en) * 2007-08-14 2009-02-19 F. Hoffmann-La Roche Ag Predictive markers for egfr inhibitors treatment
WO2010015538A2 (en) * 2008-08-05 2010-02-11 F. Hoffmann-La Roche Ag Predictive marker for egfr inhibitor treatment
WO2010145796A2 (en) * 2009-06-19 2010-12-23 Merck Patent Gmbh Biomarkers and methods for determining efficacy of anti-egfr antibodies in cancer therapy
WO2013076282A1 (en) * 2011-11-25 2013-05-30 Integragen A method for predicting responsiveness to a treatment with an egfr inhibitor

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20090181016A1 (en) * 2005-11-30 2009-07-16 University Of Southern California FCgamma POLYMORPHISMS FOR PREDICTING DISEASE AND TREATMENT OUTCOME
JP6348115B2 (en) * 2012-10-26 2018-06-27 ザ ユニバーシティー オブ クイーンズランド Use of endocytosis inhibitors and antibodies for cancer therapy

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009021673A1 (en) * 2007-08-14 2009-02-19 F. Hoffmann-La Roche Ag Predictive markers for egfr inhibitors treatment
WO2010015538A2 (en) * 2008-08-05 2010-02-11 F. Hoffmann-La Roche Ag Predictive marker for egfr inhibitor treatment
WO2010145796A2 (en) * 2009-06-19 2010-12-23 Merck Patent Gmbh Biomarkers and methods for determining efficacy of anti-egfr antibodies in cancer therapy
WO2013076282A1 (en) * 2011-11-25 2013-05-30 Integragen A method for predicting responsiveness to a treatment with an egfr inhibitor

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
ASTRID LIÈVRE ET AL: "KRAS mutation status is predictive of response to Cetuximab therapy in colorectal cancer", CANCER RESEARCH, AMERICAN ASSOCIATION FOR CANCER RESEARCH, US, vol. 66, no. 8, 15 April 2006 (2006-04-15), pages 3992 - 3995, XP008155311, ISSN: 0008-5472, DOI: 10.1158/0008-5472.CAN-06-0191 *
KAKIUCHI SOJI ET AL: "Prediction of sensitivity of advanced non-small cell lung cancers to gefitinib (Iressa, ZD1839)", HUMAN MOLECULAR GENETICS, OXFORD UNIVERSITY PRESS, SURREY, vol. 13, no. 24, 15 December 2004 (2004-12-15), pages 3029 - 3043, XP002440000, ISSN: 0964-6906, DOI: 10.1093/HMG/DDH331 *

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN105701365A (en) * 2016-01-12 2016-06-22 西安电子科技大学 Cancer-related genes finding method by using miRNA expression data
JPWO2017187937A1 (en) * 2016-04-28 2019-02-28 デンカ株式会社 Method for determining resistance of cancer cells to epidermal growth factor receptor inhibitors
WO2018034301A1 (en) * 2016-08-17 2018-02-22 中外製薬株式会社 Combination drug of anti-epiregulin antibody and anti-egfr antibody

Also Published As

Publication number Publication date
EP3074530A1 (en) 2016-10-05
JP2017503478A (en) 2017-02-02
BR112016012001A2 (en) 2017-09-26
AU2014356506A1 (en) 2016-06-09
KR20160089488A (en) 2016-07-27
MX2016006782A (en) 2016-08-19
US20160376661A1 (en) 2016-12-29
CA2931176A1 (en) 2015-06-04
CN105765081A (en) 2016-07-13

Similar Documents

Publication Publication Date Title
US20220049312A1 (en) microRNAs as Biomarkers for Endometriosis
AU2012342397B2 (en) A method for predicting responsiveness to a treatment with an EGFR inhibitor
US20120027753A1 (en) MicroRNAs in Never-Smokers and Related Materials and Methods
US20160376661A1 (en) A method for predicting responsiveness to a treatment with an egfr inhibitor
KR101785795B1 (en) Biomarker micro rna for predicting prognosis of head and neck squamous cell carcinoma
US20180142303A1 (en) Methods and compositions for diagnosing or detecting lung cancers
US20200248269A1 (en) Methods for predicting the outcome of a cancer in a patient by analysing gene expression
EP3198028B1 (en) A method for predicting responsiveness to a treatment with an egfr inhibitor
TW201827603A (en) Biomarker panel for prognosis of bladder cancer
EP2716767A1 (en) Method for determining the prognosis of pancreatic cancer
US20150344961A1 (en) Sera Based miRNAs as Non-Invasive Biomarkers in Melanoma
Wang et al. Increased expression of miR-221 and miR-222 in patients with thyroid carcinoma
EP3476951B1 (en) Microrna-based methods and compositions for the diagnosis and prognosis of vulvar carcinoma and vulvar intraepithelial lesions
KR101507656B1 (en) GNB2L1 gene polymorphisms marker for predicting survival in patients with lung cancer and method for predicting survival using the same
WO2023099350A1 (en) In vitro method for predicting the response of patients suffering from her2+ breast cancer to a treatment with anti-her2 neoadjuvant therapy

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: 14805833

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2931176

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 15038826

Country of ref document: US

ENP Entry into the national phase

Ref document number: 2016534255

Country of ref document: JP

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: MX/A/2016/006782

Country of ref document: MX

NENP Non-entry into the national phase

Ref country code: DE

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112016012001

Country of ref document: BR

ENP Entry into the national phase

Ref document number: 2014356506

Country of ref document: AU

Date of ref document: 20141126

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 20167016884

Country of ref document: KR

Kind code of ref document: A

REEP Request for entry into the european phase

Ref document number: 2014805833

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2014805833

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 112016012001

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20160525