WO2016198807A1 - Method for predicting the sensitivity of a gastrointestinal tumor to an inhibitor treatment - Google Patents

Method for predicting the sensitivity of a gastrointestinal tumor to an inhibitor treatment Download PDF

Info

Publication number
WO2016198807A1
WO2016198807A1 PCT/FR2016/051419 FR2016051419W WO2016198807A1 WO 2016198807 A1 WO2016198807 A1 WO 2016198807A1 FR 2016051419 W FR2016051419 W FR 2016051419W WO 2016198807 A1 WO2016198807 A1 WO 2016198807A1
Authority
WO
WIPO (PCT)
Prior art keywords
qprt
tumor
expression
treatment
protein
Prior art date
Application number
PCT/FR2016/051419
Other languages
French (fr)
Inventor
Stéphane ANSIEAU
Alain Puisieux
Guillaume Collin
Pierre SAINTIGNY
Original Assignee
Centre Leon Berard
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 Centre Leon Berard filed Critical Centre Leon Berard
Publication of WO2016198807A1 publication Critical patent/WO2016198807A1/en

Links

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/57407Specifically defined cancers
    • G01N33/57446Specifically defined cancers of stomach or intestine
    • 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
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/573Immunoassay; Biospecific binding assay; Materials therefor for enzymes or isoenzymes
    • 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
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/90Enzymes; Proenzymes
    • G01N2333/91Transferases (2.)
    • G01N2333/91091Glycosyltransferases (2.4)
    • G01N2333/91142Pentosyltransferases (2.4.2)
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/52Predicting or monitoring the response to treatment, e.g. for selection of therapy based on assay results in personalised medicine; Prognosis

Definitions

  • the present invention is in the medical field and more specifically in the field of anti-cancer therapy.
  • the main subject of the invention is an in vitro or ex vivo method of selecting patients with gastrointestinal tumors that may benefit from anti-tumor therapy comprising the administration of an inhibitor of the kynurenine pathway.
  • a method according to the invention comprises in particular a step of determining the expression of the QPRT protein in a test sample of said gastrointestinal tumor, the expression of QPRT in epithelial cells of the test sample being indicative of the sensitivity said patient to an inhibitory treatment of the kynurenine pathway.
  • the present invention also relates to the use of the protein QPRT as a biomarker for predicting the sensitivity of a patient suffering from a gastrointestinal tumor to an inhibitory treatment of the kynurenine pathway.
  • This method comprises determining the expression of the QPRT protein in the epithelial cells of a sample of the patient's tumor prior to any treatment, a higher expression of the QPRT protein in the test sample compared to a reference sample being indicative of the patient's sensitivity to an inhibitory treatment of the kynurenine pathway.
  • classifiers to monitor the evolution of Crohn's disease and colitis have been described (Montero-Melendez et al., 2013).
  • the publication by Watanabe (2013) describes a gene expression signature and a method for predicting the progression from ulcerative colitis to colorectal cancer.
  • the signature described in this document comprises 40 differentially expressed genes according to whether patients with neoplastic lesions, or not affected by such lesions.
  • telomeres Chronic inflammation is fertile ground for cancer development. In inflamed mucosa, however, oxidative stress induces erosion of telomeres and signs of replicative senescence in intestinal stem cells.
  • the inventors have shown that the comparison of the gene expression profile of human colon epithelial cells that has escaped the senescence induced by oxidative stress with the gene profile of the parental line highlights the induction of a specific gene program, characterized by the activation of the expression of about thirty genes and the repression of about thirty others.
  • This gene program is orchestrated by ZEB transcription factors and is differentiated from the epithelio-mesenchymal transition (EMT) program commonly induced by these factors.
  • EMT epithelio-mesenchymal transition
  • This new gene program is named ALEP for Alternative to EMT Program.
  • the inventors have shown in vitro that the expression of QPRT confers a survival advantage to cells having escaped senescence, under conditions of chronic inflammation or when they are subjected to an oxidizing agent.
  • the inventors have also shown that QPRT depletion of cells that have escaped senescence induces the death of these cells by apoptosis under conditions of chronic inflammation, and that the inhibition of QPRT expression prevents these cells from tolerating oncogenic activations. They also demonstrated that intestinal cancer cell lines with an active ALEP program (high signature score) are dependent on the QPRT protein for proliferation. Beyond the QPRT protein, the inhibition of another key enzyme of the kynurenine pathway located upstream of QPRT, the TD02 protein, very similarly inhibits the proliferation of these lines. The activity of the kynurenine pathway as a whole would therefore be necessary for the proliferation of colorectal lines in which the ALEP program is activated.
  • the inventors have shown that the QPRT protein is absent in the enterocytes of normal intestinal tissue, whereas an increase in the number of labeled epithelial cells during the detection of QPRT is observed in a significant proportion of dysplasias of low- or high-grade and cancers developed in an inflammatory context, as well as in epithelial cells with a significant proportion of adenomas and sporadic cancers.
  • a proportion of oesophageal adenocarcinomas express the QPRT protein whereas no labeling is observed in the squamous epithelium, used as internal control.
  • the QPRT protein, and beyond it the other enzymes of the kynurenine pathway, are therefore particularly interesting therapeutic targets and their inactivation therefore constitutes a promising anti-tumor therapeutic approach, if appropriate in combination with another anti-tumor treatment, in particular with a therapy capable of inducing oxidative stress.
  • the present invention relates to an ex vivo method of selecting a patient suffering from a gastrointestinal tumor, likely to benefit from an inhibitory treatment of the kynurenine pathway, from a test sample of said tumor before treatment of said patient, said method comprising the following steps:
  • step b) The comparison of the expression of the QPRT protein in the epithelial cells of the said test sample with the expression of QPRT in a reference sample
  • step c) The selection of a patient likely to benefit from an inhibitor treatment of the pathway of kynurenine from the comparison of step b), a higher expression level of QPRT protein in the test sample epithelial cells than that of the reference sample being indicative of the sensitivity of said test sample. tumor to an inhibitory treatment of the kynurenine pathway.
  • the present invention also relates to an ex vivo method for predicting the sensitivity of a patient suffering from a gastrointestinal tumor (before treatment) to an inhibitory treatment of the kynurenine pathway, said method comprising the following steps: i) obtaining a test sample of said tumor, ii) determining the expression of QPRT protein in the epithelial cells of said test sample, iii) comparing the expression of QPRT protein in said test sample with the expression of QPRT in a reference sample, iv) predicting the sensitivity of said patient to an inhibitory treatment of the kynurenine pathway from the comparison of step iii), a higher expression level of QPRT protein epithelial cells of the test sample relative to that of the reference sample being indicative of the sensitivity of said patient to an inhibitory treatment of the kynurenine pathway.
  • sensitivity of a patient to a treatment is meant the observation of a therapeutic effect such as improving the survival of a patient having received said therapeutic treatment.
  • the absence of a patient's response, or the patient's lack of sensitivity to a therapeutic treatment is instead defined by the term "resistance" of a patient to said therapeutic treatment, in which no therapeutic effect is observed.
  • before treatment or “untreated” means before or after a surgical procedure, such as an excision, performed on the tumor but before any therapeutic treatment associated but non-surgical, such as chemotherapy, radiotherapy, hormone therapy or immunotherapy.
  • Quinolinate phosphoribosyltransferase or QPRT (UniProtKB Q 15274 updated on May 18, 2010, SEQ ID NO: 1), also known as Nicotinate-nucleotide pyrophosphorylase catalyzes the conversion of quinolinic acid (QA) to nicotinamide mononucleotide (NAMN) by transfer.
  • Nicotinate-nucleotide pyrophosphorylase catalyzes the conversion of quinolinic acid (QA) to nicotinamide mononucleotide (NAMN) by transfer.
  • QA quinolinic acid
  • NAMN nicotinamide mononucleotide
  • PRPP 5-phospho-D-ribose-1-diphosphate
  • kynurenine pathway is meant the metabolic pathway leading to the de novo production of NAD + from tryptophan, an essential amino acid.
  • the enzymes involved in this pathway have been described in particular by Vécsei et al., 2013.
  • inhibitory treatment of the kynurenine pathway is meant the application of a treatment intended to inhibit the activity and / or the effects of at least one of the enzymes which contribute to this metabolic pathway, namely tryptophan 2 , 3-dioxygenase (TD02), kynurenine formylase, kynurenine 3-monooxygenase (KMO), kynureninase (KYNU), 3-hydroxyanthranilic acid oxygenase (3HAO) or QPRT.
  • tryptophan 2 3-dioxygenase (TD02), kynurenine formylase, kynurenine 3-monooxygenase (KMO), kynureninase (KYNU), 3-hydroxyanthranilic acid oxygenase (3HAO) or QPRT.
  • This treatment includes, for example, the use of inhibitors of the tryptophan protein 2,3-dioxygenase or TD02 such as the molecule 690C91 ((E) -6-fluoro-3- [2- (3-pyridyl) vinyl] -1H indole, Sigma), inhibitors of kynurenine 3-monooxygenase or KMO such as 2-3 teriflunomide and Ro-61-8048 (Selleckchem), PDE10A (Pfizer), JM6 (3,4-dimethoxy-N- (4) - (3-nitrophenyl) -5- (piperidin-1-ylmethyl) thiazol-2-yl) benzenesulfonamide), inhibitors of the enzyme 3HAO as described by Vallerini et al., 2013 or QPRT inhibitors.
  • a known example of an antagonist of the enzymatic activity of QPRT is phthalic acid (Malik et al., 2013).
  • inhibitor of the kynurenine pathway is meant any substance, molecule, compound or complex capable of specifically inhibiting, in particular by competition or conformation, the activity and / or the effects of at least one enzyme of this type. metabolic pathway.
  • these inhibitors may be of the allosteric type, substrate analog (s), product analogue (s) or enzymatic reaction intermediate (s), especially non-catabolizable, specific for the enzyme concerned.
  • the inhibitory treatment of the kynyrenine pathway is preferably an inhibitor treatment of QPRT or TD02, and more particularly QPRT.
  • the subject of the invention is an ex vivo method of selecting a patient suffering from a gastrointestinal tumor, likely to benefit from an inhibitory treatment of the kynurenine pathway, from a test sample of said tumor prior to treatment of said patient, said method comprising the following steps:
  • step b) comparing the expression of the QPRT protein in the epithelial cells of said test sample with the expression of QPRT in a reference sample, c) The selection of a patient likely to benefit from an inhibitory treatment of the kynurenine pathway from the comparison of step b), a higher expression level of QPRT protein in the epithelial cells of the sample test relative to that of the reference sample being indicative of the sensitivity of said tumor to an inhibitory treatment of the kynurenine pathway.
  • the subject of the invention is an ex vivo method of selecting a patient suffering from a gastrointestinal tumor, before treatment, likely to benefit from an inhibitory treatment of the kynurenine pathway, comprising the determining the level of expression of QPRT protein in the epithelial cells of a tumor test sample, wherein the tumor test sample is selected from: tumor tissue, an operative specimen and a biopsy.
  • the selection method according to the invention relates to an ex vivo method of a patient suffering from a gastrointestinal tumor likely to benefit from an inhibitory treatment of the kynurenine pathway, before said patient has been treated for said tumor.
  • the sample from the patient with a gastrointestinal tumor is obtained before treatment; said patient being considered untreated for said tumor.
  • the sample of the gastrointestinal tumor may be derived from the primary tumor or from metastasis (s). According to a particular aspect of the ex vivo selection process according to the invention, the test sample of said tumor is derived from the primary tumor.
  • the expression "higher expression level” indicates that the level of expression observed in said test sample is greater than the level of expression observed in the reference sample, in terms of the number of cells in which the Expression is detected and / or the intensity of the detected signal and / or the location of the cells for which the protein is detected.
  • the inventors have shown that the QPRT protein is absent in the enterocytes of normal intestinal tissue, whereas an increase in the number of labeled epithelial cells during the detection of QPRT is observed in a significant proportion of dysplasias of low- or high-grade and cancers developed in an inflammatory context, as well as in epithelial cells with a significant proportion of adenomas and sporadic cancers.
  • oesophageal adenocarcinomas express the QPRT protein whereas no labeling is observed in the squamous epithelium, used as internal control.
  • the expression of the QPRT protein is determined by comparison with the expression of the QPRT protein in a reference sample, the expression of the QPRT protein in a reference sample being especially chosen from: the expression of QPRT in a test sample of healthy tissue of the same nature as the tissue of the test sample, the expression of QPRT in a tissue sample of a patient reaches inflammatory gastrointestinal disease not associated with dysplastic (ie, non-tumor) lesions, and / or expression of QPRT in a tissue sample of the subject patient taken from the patient did not have a gastrointestinal tumor.
  • the expression of the QPRT protein in a reference sample being especially chosen from: the expression of QPRT in a test sample of healthy tissue of the same nature as the tissue of the test sample, the expression of QPRT in a tissue sample of a patient reaches inflammatory gastrointestinal disease not associated with dysplastic (ie, non-tumor) lesions, and / or expression of QPRT in a tissue sample of the subject patient taken from the patient did not have
  • the known non-tumorous inflammatory gastrointestinal diseases are: Crohn's disease, ulcerative colitis; chronic gastritis and chronic gastroesophageal reflux (as part of adenocarcinoma).
  • the determination of the level of expression of the QPRT protein is carried out quantitatively or semi-quantitatively by any method known to those skilled in the art allowing the specific detection and / or quantification of a particular protein in a sample.
  • a determination method is chosen in particular from methods using QPRT-binding antibodies, including in particular immunohistochemistry (IHC) and immunofluorescence (IF) detection.
  • QPRT can be defined by means of a histological score taking into account, on the one hand, the observed marking intensity and, on the other hand, the percentage of labeled cells, or by combining the percentage labeled cells and the intensity of labeling. According to this method, tumor sample sections are analyzed by microscopy after bringing said sections into contact with an antibody specifically binding QPRT.
  • labeled cells cells for which a signal is detected when looking for expression of the QPRT protein.
  • the subject of the invention is an ex vivo method of selecting a patient suffering from a gastrointestinal tumor likely to benefit from an inhibitory treatment of the kynurenine pathway, in which the Determination of QPRT protein expression comprises determining the presence of QPRT comprising at least one immunohistochemistry step.
  • immunohistochemistry is meant a method of locating proteins in the cells of a tissue section, by detecting a particular antigen by means of polyclonal or monoclonal antibodies binding that antigen.
  • the antibody-antigen pair can be visualized for example by conjugation to an enzyme which can catalyze a color production reaction, by the use of labeled antibodies, in particular by a fluorophore, or by the use of antibodies known as " secondary antibodies capable of binding antigen-binding antibodies.
  • the tissue is placed on a solid support.
  • a primary antibody bound to a detection system (or a primary antibody followed by a secondary antibody) is added.
  • the signal is observed by microscopy techniques.
  • the test sample of said tumor is contacted with a QPRT-binding antibody, said antibody being selected from polyclonal anti-QPRT antibodies and anti-QPRT monoclonal antibodies.
  • said antibody binds QPRT under native conditions, one skilled in the art can easily choose an antibody meeting this criterion, there may be mentioned in particular the Sigma HPA011887 antibody.
  • the subject of the invention is an in vitro method for selecting a patient suffering from a gastrointestinal tumor that may benefit from an inhibitory treatment of the kynurenine pathway, in which the determination of the presence of the QPRT protein in the test sample of said tumor is carried out by means of at least one immunohistochemistry step comprising the localization of labeled cells within the epithelial tissue of the test sample.
  • an ex vivo method for predicting the sensitivity of a patient suffering from a gastrointestinal tumor to an inhibitory treatment of the kynurenine pathway comprises the determination of the percentage of labeled epithelial cells within the epithelial tissue of the test sample.
  • the subject of the invention is an ex vivo method of selecting a patient suffering from a gastrointestinal tumor likely to benefit from an inhibitory treatment of the kynurenine pathway, in which the determination of the QPRT protein expression comprises determining the percentage of labeled tumor cells.
  • a level of expression of QPRT protein in the test sample corresponding to at least 10% of the labeled epithelial cells is indicative of the sensitivity of said tumor to a treatment inhibitor of the way of kynurenine.
  • a method according to the invention comprises determining the amount of an mRNA encoding the QPRT protein, particularly in the epithelial cells of the tumor.
  • the determination of the amount of the mRNA encoding the QPRT protein can be carried out by any method known to those skilled in the art allowing the specific detection of an mRNA, for example after microdissection of the epithelial cells of a tumor.
  • Such a method notably comprises the specific detection of an mRNA using nucleic acid probes complementary to the sequence of said mRNA, in particular by RT-PCR, by a Northern-Blot type method. It can also be obtained by hybridization on an immobilized probe (in situ hybridization).
  • Quantitative PCR is used to measure the initial amount of nucleic acid.
  • the amplified nucleic acids, and in particular the mRNAs can be detected and quantified by any method known to a person skilled in the art, such as in particular by 260 nm spectrometry or by Northern blotting using a hybridization method for radioactive or fluorescent probes.
  • amplified nucleic acid The nucleotide sequence of the mRNA corresponding to quinolinate phosphoribosyltransferase is known (NCBI reference NM 014298, updated on March 15, 2015, SEQ ID NO: 2).
  • primers capable of specifically hybridizing to the nucleic acid encoding QPRT, thus enabling its detection.
  • the primers described in the present patent application can be used in a process according to the invention.
  • the subject of the invention is an ex vivo method of selecting a patient suffering from a gastrointestinal tumor likely to benefit from an inhibitory treatment of the kynurenine pathway, starting from a sample of said tumor before any treatment, said method comprising the following steps:
  • test sample ii. a higher expression level of QPRT protein in the test sample epithelial cells and a lower level of expression of at least one ALEP program protein in the test sample epithelial cells relative to that of the test sample.
  • reference sample being indicative of the sensitivity of said tumor to an inhibitory treatment of the kynurenine pathway.
  • ALEP program proteins refer to the 59 proteins encoded by the genes shown in Figure 13, which the inventors have demonstrated that the expression is either higher or lower in epithelial cells resistant to oxidative stress.
  • the genes encoding the ALEP program proteins whose expression is greater are:
  • APOE SPARC, C6ORF160, SNHG5, SLC2A3, GALC, MT1G, SH3GL2, GNE, SUNC1, EXOSC8, LCP1, KCNJ8, NDRG1, SNCG, FGD3, LGALS1, TXNIP, SUM03, EMILIN2, TNNC1, TNNT1, TGFB1, LOC728031, KRT19 and GABARAPL1.
  • the genes coding for the ALEP program proteins whose expression is lower are: IL32, ETV5, S100A3, SGK1, PRSS1, IRAK2, S100A9, GPR56, TNFAIP3, PIM2, PRRX2, BIRC3, CXCL5, DIAPH1, AD AMI9, LRIG1, SLC7A11, EVL, WNT5A, TNFAIP2, EN02, TNFRSF1B, PROM1, TNF, TMPRSS2, LOC100134294, NFIX, RIN2, CXCL1, CXCL2, CARD9, CCL20.
  • step b) consists in comparing the expression of the QPRT protein and at least the expression of the APOE protein in the epithelial cells of said test sample, it being understood that a higher expression level of QPRT and of APOE is indicative of the sensitivity of said tumor to an inhibitory treatment of the kynurenine pathway, in particular to a QPRT or TD02 inhibitor, in particular to a QPRT inhibitor.
  • said gastrointestinal tumor is selected from: colorectal cancer, gastric cancer and adenocarcinoma of the esophagus.
  • tumor refers to a malignant tumor or cancer.
  • colonal cancer refers to a tumor resulting from the neoplastic transformation of the epithelial cells of the mucosa of the colon or rectum. Colorectal cancer can develop in an inflammatory context (ulcerative colitis or Crohn's disease) or not and may result from malignant progression of dysplasia or polyps. Depending on the size and depth of the tumor, ganglion infiltration and number, the tumors are classified as O to 4 (TNM classification).
  • Gastric adenocarcinoma refers to a tumor resulting from the neoplastic transformation of the stomach lining and results from the progression of chronic gastritis in atropic chronic gastritis, metaplasia, dysplasia before possible progression to adenocarcinoma. Localized, locally advanced and metastatic stages are distinguished.
  • Esophageal adenocarcinoma refers to a tumor derived from the epithelium of the
  • Tumors are classified according to the TNM classification).
  • said gastrointestinal tumor is intestinal is developed in a chronic inflammatory context.
  • chronic inflammatory context refers to the repetition of stages of inflammation and remission, this period can extend over years, as for patients with Crohn's disease or ulcerative colitis, leading progressively to an alteration of the epithelium, for example by inducing genetic instability in the epithelial cells.
  • This chronic inflammation increases the incidence of cancer in the long term.
  • the subject of the invention is an ex vivo method for predicting the sensitivity of a patient suffering from a gastrointestinal tumor developed on a chronic inflammatory context to an inhibitor treatment of the gastrointestinal tract. kynurenine, according to the invention, said gastrointestinal tumor being a colorectal cancer.
  • said gastrointestinal tumor intestinal is a sporadic colorectal cancer.
  • Tumors that do not develop in an inflammatory context are said to be sporadic; they generally result from the mutation of the APC gene.
  • said gastrointestinal tumor intestinal is a colorectal cancer that affects patients with germline mutations on the APC gene. These patients develop juvenile adenomatous polyposes (PAF), which can evolve into CRC.
  • PAF juvenile adenomatous polyposes
  • the subject of the invention is an ex vivo method for predicting the sensitivity of a patient suffering from a tumor to an inhibitory treatment of the kynurenine pathway, combined with another antitumor treatment. More particularly, in a method according to the invention, said anti-tumor treatment is chosen from: surgery, chemotherapy and radiotherapy.
  • the subject of the invention is an ex vivo method for predicting the sensitivity of a patient suffering from a gastrointestinal tumor to an inhibitory treatment of the kynurenine pathway, said patient having made the subject of a surgical step for removing said tumor (or excision).
  • the subject of the present invention is also an ex vivo method for identifying a tumor capable of responding to a treatment comprising the administration of a kynurenine inhibiting agent, from a test sample of said tumor before treatment of said tumor. patient, said method comprising the steps of:
  • the expression of QPRT can be determined by the protein expression of QPRT or the transcriptional expression of QPRT, in particular the expression of the QPRT mRNA. According to a particularly preferred embodiment of this subject of the invention, it is the expression of the QPRT protein which is determined.
  • the invention relates to the use of QPRT as a protein biomarker for predicting the sensitivity of a patient with a gastrointestinal tumor to an inhibitory treatment of the kynurenine pathway.
  • the invention also relates to the use of QPRT as a biomarker for predicting the sensitivity of a patient suffering from a gastrointestinal tumor to an inhibitory treatment of the kynurenine pathway, preferably chosen from a QPRT inhibitor and a TD02 inhibitor, even more preferably QPRT inhibitor treatment, said patient being untreated for said tumor and QPRT protein expression being measured in epithelial cells of a sample of said tumor.
  • the invention also relates to the use of QPRT as a biomarker for predicting the sensitivity of a patient suffering from a gastrointestinal tumor to an inhibitory treatment of the kynurenine pathway, preferably selected from a QPRT inhibitor and a TD02 inhibitor, combined with an anti-tumor treatment, such as oxaliplatin, 5-FU or irrinotecan, and even more preferably the sensitivity of a patient with a gastrointestinal tumor -intestinal to an inhibitory treatment of QPRT.
  • an inhibitory treatment such as oxaliplatin, 5-FU or irrinotecan
  • Figures 1A and 1B The escape to senescence induced in response to chronic inflammation allows the emergence of risk cells.
  • Fig.lA submission of colon epithelial cells to chronic inflammation forces them to adapt to oxidative stress.
  • Immortalized human colon epithelial cells (HCEC-hTERT) were subjected every two days to activated macrophage supernatants (AMS).
  • AMS macrophage supernatants
  • Cells rapidly stop proliferating (as demonstrated by H3-phosphorylated histone level, pS10-H3), enter senescence as demonstrated by detection of SA- ⁇ -galactosidase activity, accumulation of acetylated p53 protein ( Ac-p53) and the induction of its target gene CIP1 (p21). After a month, the cells begin to proliferate.
  • Fig. 1B Emergent cells are resistant to oxidative stress and tolerate mitogenic activations.
  • the cells that have escaped senescence (Ech-Inf A and Ech-Inf B from two separate experiments) and the Parental HCEC cells (Par) were either treated with tert-butyl-hydoxy-peroxide (TBHP) (panel left) or transduced by an activated version of the RAS protein (HRASG12V, right panel) and stained with 12-day crystal violet later.
  • the level of RAS protein is analyzed by Western blot.
  • Figures 2A and 2B ZEB transcription factors orchestrate the mechanism of adaptation to oxidative stress.
  • Fig. 2A Escaping to induced senescence in response to chronic inflammation or chemical peroxide results from induction of ZEB embryonic genes. Analysis of SNAI1, TWIST2, ZEB1 and ZEB2 gene expression by q-RT-PCR in cells escaping chronic inflammation (left panel) or oxidative stress induced by a chemical peroxide (right panel). Relative expression is defined in comparison to the HPRT1 household gene and parental HCEC cells. The expression of the SNAI2 and TWIST1 genes is below the detection threshold.
  • Fig. 2B The ectopic expression of one of the two ZEB transcription factors is sufficient to allow cells to adapt to oxidative stress.
  • HCEC-hTERT cells were transduced by Zebl or Zeb2, submitted to either activated macrophage supernatants (AMS) or TBHP. After 12 days of treatment, the cells were stained with crystal violet.
  • AMS activate
  • FIGS 3A and 3B The resistance to oxidative stress is related to the induction of a specific gene program.
  • Fig. 3A Comparison of the HCEC-hTERT gene profiles that escaped chronic inflammation (from two independent experiments, Ech-Inf A and B), chemical oxidative stress induced (Ech-TBHP) or transduced by Zebl (HCEC-ZeW) or Zeb2 (HCEC-Zeb2) highlights a set of 27 commonly induced genes and 32 commonly repressed genes (see Figure 13). Among this list, the two most induced genes are QPRT and APOE.
  • Fig. 3B The ectopic expression of either of these two proteins is sufficient to protect cells from oxidative stress.
  • HCEC cells were transduced by QPRT or APOE and subjected to either activated macrophage supernatants (AMS) or TBHP treatment. After 12 days of treatment, the cells were stained with crystal violet.
  • FIGS. 4A and 4B The activity of the QPRT protein contributes to resistance to oxidative stress.
  • Fig. 4A Survival of cells that have escaped senescence induced in response to chronic inflammation is dependent on QPRT expression.
  • HCEC cells that escaped senescence induced in response to chronic inflammation were infected with lentiviral vectors expressing two distinct shRNAs directed against QPRT RNA (shRNA QPRT A and B) and subjected activated macrophage supernatants (SMA). After 12 days of treatment, the cells are stained with crystal violet.
  • the parental HCEC (Par) and Ech-Inf cells infected with a control shRNA were used respectively as negative and positive controls.
  • Fig. 4B QPRT protein activity is essential to allow cells that have escaped senescence induced in response to chronic inflammation to tolerate activations oncogenic.
  • Panel at the top HCEC cells that escaped induced senescence in response to chronic inflammation (Ech-Inf) were disrupted in QPRT by RNA interference and infected with a retroviral vector expressing the activated version of the HRAS protein (HRASG12V). 12 days after infection, the cells were stained with crystal violet.
  • Bottom panel QPRT and RAS analysis by Western blot.
  • Figure 5 Classification of human lines of colorectal carcinoma based on the ALEP score.
  • the lines used in the following tests and in which the program is induced are OUMS-23, C2BBel, CL-14; the lines in which the program is not induced are SW116, HCT116, HT-29, SW403, COLO-320.
  • FIG. 6 The proliferation of colorectal cancer lines in which the ALEP program is induced is dependent on the expression of QPRT.
  • Top panels different colorectal lines in which the alternative program is induced (ALEP active) or not (ALEP inactive) were infected with lentiviral vectors expressing shRNAs directed against QPRT RNA (shRNA QPRT A and B) and stained with purple crystal twelve days after infection.
  • Bottom panel QPRT analysis in the different lines by Western blot.
  • Figure 7 Panels A and B.
  • the inhibition of the kynurenine pathway in ALEP -positive ORMS-23 (panel A) or C2BBel (panel B) colorectal lines is accompanied by a proliferation arrest, a induction of autophagy and DNA damage.
  • Panels from the top Inhibition of QPRT expression via shRNA (A or B) induces cell proliferation arrest as judged by proliferation curves (panels on the upper left) and histone H3 phospho accumulation Serine 10 (pS10-H3, panels top right).
  • Figure 8 QPRT analysis on human samples by immunohistochemistry (I). QPRT marking examples by immunohistochemistry on healthy colon samples (upper left) of inflamed mucous membranes (top right), dysplasias of low- or high-grade (2 nd and 3 rd row from the high) or carcinomas developed on an inflammatory context. Scale: 100 ⁇ .
  • Figure 9 QPRT analysis on human samples by immunohistochemistry (II). Examples of QPRT staining by immunohistochemistry in samples of weakly or strongly dysplastic adenomas and in sporadic cancers. Scale 100 ⁇ .
  • FIGS 10A and 10B The ALEP program is induced in the inflamed mucosa of patients with ulcerative colitis or Crohn's disease.
  • the comparison of the ALEP program signature with the gene profiles of patients with Crohn's disease (CD) or ulcerative colitis (UC) makes it possible to segregate a certain number of patients. Note in the series of patients with ulcerative colitis GSE37283 the ability of the signature to segregate those with or without cancerous colorectal lesions.
  • Figures 11A to 11C Analysis of the biological significance of the alternative program on a large cohort of patients with sporadic colorectal cancers (primary resected tumors of patients who have not yet undergone any non-surgical associated therapeutic treatment). The analysis is conducted on a cohort of 566 colorectal carcinomas (GSE39582) classified according to their gene profile into six molecular types (from C1 to C6, Marisa et al., 2013).
  • Fig. 11A Analysis of the signature score of the alternative program in the six molecular types.
  • Fig. 11B Analysis of the correlation between the scores of the alternative program signatures and intestinal stem cells Lgr5 and EphB2.
  • Fig. 11C Analysis of the biological significance of the alternative program on a large cohort of patients with sporadic colorectal cancers (primary resected tumors of patients who have not yet undergone any non-surgical associated therapeutic treatment). The analysis is conducted on a cohort of 566 colorectal carcinomas (GSE39582) classified according to
  • Kaplan-Meier representation of the recurrence-free survival of colorectal cancer patients by high, intermediate, or low ALEP score (with thresholds taken at 4000 and 6000 by combining the scores of each of the overexpressed or under-expressed genes even extinct).
  • Figure 12 QPRT analysis by immunohistochemistry in oesophageal adenocarcinoma.
  • the squamous epithelium of the lower third of the esophagus is used as an internal control (panel at the bottom right). Scale 100 ⁇ .
  • Figure 13 List of genes commonly induced or repressed in HCEC cells that have escaped senescence induced in response to chronic inflammation (Ech-Inf A and B) to a chemical peroxide (Ech-TBHP A and B) or transduced by Zebl or Zeb2.
  • the HRAS G12V , ZEB1 and ZEB2 pBabe Puro constructs have been previously described (Morel et al., 2012).
  • the expression vectors ⁇ 3 and QPRT in Pbabe Puro were generated from the vectors pLenti-GIII-hAPOE-GFP-2A-Puro constructs (abm) and QPRT pCMV6 (Origene).
  • the QPRT RK138 / 139 cDNA was synthesized by the Genscript company and subcloned into the Puro pBabe retro viral vector (Addgene).
  • shRNAs (5'-TCGCTCTGAAGGTGGAGT-3 ', SEQ ID NO: 5) and shRNA QPRT B (5'-AGTCCTAAACCGGAAGAGG-3', SEQ ID NO: 6) in pLKO.I were obtained from Sigma .
  • HT-29, HCT116, SW403, COLO320HSR and C2BBel were obtained from ATCC.
  • the lines OUMS23 and CL-14 were provided by the JCRB cellular bank (Japan) and the DSMZ GmbH (Germany), respectively.
  • the lines OUMS23 and c2BBel were cultured in DMEM medium (Life Technologies) supplemented with 10% FCS (Cambrex) and 100 U / ml Penicillin / Streptomycin (PS, Life Technologies), lines HT-29 and HCT116 in McCoy's 5A medium ( Life Technologies) supplemented with 10% FCS + 100 U / ml PS, lines SW116, SW403 and COLO320 in RPMI medium (Life Technologies) supplemented with 10% FBS + 100 U / ml PS, and the line CL-14 in DMEM / HAMF 12 1: 1 medium (Invitrogen) supplemented with 20% FBS and 100 U / ml PS.
  • the primary epithelial colon cells were obtained from the abm company (T4056), immortalized by transduction at HTERT and cultured in Prigrow III medium (TM003, abm) supplemented with 5% FBS and 100 U / ml PS on collagen plates. (BD BioCoat Collagen Type I, Dutscher).
  • the monocyte line THP-1 was obtained from ATCC and cultured in RPMI medium (Life Technologies) supplemented with 10% FBS, 100 U / ml PS, 1 mM Sodium pyruvate (Life Technologies) and non-essential amino acids (Life Technologies).
  • Cell differentiation (5.10 7 cells) was induced by a treatment with PMA (0.162 ⁇ , Sigma) for one day and the activation of the macrophages thus obtained then induced by LPS (ultra-pure LPS-EB of the E line. coli 0111: B4, InvivoGen) for one day.
  • the supernatant was collected, filtered over 0.45 ⁇ l, diluted half in the HCEC culture medium and placed on the HCEC cells (5 ⁇ 10 5 in a 6-well dish). The supernatant of activated macrophages was renewed every other day. Similarly, the treatment of HCEC cells with tert-Butyl hydroperoxide (30 ⁇ l, Sigma) was repeated every other day. The cells were stained with crystal violet after ten days of treatment. SA- ⁇ -galactosidase assays were performed as previously described (Dimri et al., 1995).
  • Retroviral and lentiviral infections Ectopic production of proteins in HCEC-hTERT cells was performed through retroviral infections as previously described (Gras et al., 2014). Briefly, the cells are "murine" by the ectopic expression of an ecotropic receptor (Baker et al., 1992; Boehm et al., 2005) before being infected with the retroviral expression vectors. Sequential infections are spaced 48 hours apart and selection is initiated 24 hours after the second infection with puromycin (1.5 ⁇ g / ml).
  • the lentiviral particles of the QPRT shRNAs were generated by transfection of the HEK293T line with the pLKO.l vectors, pCMV AR8.91 (gag-pop-Tat-Rev) (Zufferey et al., 1997) and phCMVG-VSVG (env). (Burns et al., 1993) by the calcium phosphate technique. Depletion in QPRT was obtained by three successive infections, each separated by 12h. We confirmed the infection of more than 90%> cells.
  • the markings were performed on sections of 40 ⁇ of formalin fixed tissues and incubated in paraffin.
  • the slides were successively deparaffinized by three successive baths of xylene and rehydrated with baths of pure ethanol, 90% and 80%.
  • the antigen was then unmasked by heating the slides in the microwave (5 and 10 min) in a PH6 (Dako) unmasking solution.
  • the slides were incubated in the presence of QPRT antibodies (Atlas antibody, HPA011887, Sigma) at room temperature for 1 h at a dilution at l / 100th.
  • QPRT antibodies Alphas antibody, HPA011887, Sigma
  • Gene Expression profiles and data analysis were performed through the ProfileXpert platform (Lyon, France). The expression profiles were analyzed on the basis of chips containing 47231 probes (HumanHT-12 v4 Expression BeadChip, Illumina Inc., USA). Total RNA (500 ng) was amplified and labeled with biotin using the Illumina TotalPrep TM RNA Amplification Kit kit (Ambion Inc., USA). Hybridization was performed with 750 ng of biotin labeled cRNA. The chips were scanned using Illumina's standard protocol and theiScan scanner (Illumina Inc., USA) and the data was standardized using Genome Studio 2010 software (Illumina Inc., USA).
  • Example 2 The escape of senescence in response to chronic inflammation leads to the emergence of risk cells and results from reprogramming of the cells, orchestrated by the transcription factor ZEB1.
  • the model established in vitro is based on the use of immortalized colon epithelial cells (HCEC-hTERT) cultured in the presence of activated macrophage supernatant (SMA, conditions mimicking chronic inflammation). These experimental conditions lead to the induction of single-stranded DNA damage that evolves into double-stranded damage (confirmed by ⁇ 2 ⁇ protein accumulation).
  • HCEC-hTERT immortalized colon epithelial cells
  • SMA activated macrophage supernatant
  • Emergent cells are not only able to proliferate under conditions of chronic inflammation but also to withstand oxidative stress that is induced in response to a peroxide chemical (such as tert-butyl-hydroxy-peroxide or TBHP) or in response to oncogenic activation (transduction of an activated version of the RAS protein, H-RAS G12V ) ( Figure 1B). Emerging cells have therefore acquired intrinsic resistance to oxidative stress and tolerant oncogenic activation is likely to evolve into a malignant phenotype.
  • TBHP chemical peroxide
  • TBHP chemical peroxide
  • the status of the TP53 gene is wild in the Ech-Inf cells and the signaling pathway remains inducible in response to genotoxic stress.
  • the analysis of the expression of the members of the three main families Snail, Twist and Zeb was analyzed. Invariably, on three independent experiments, the oxidative stress escape induced by inflammation or chemical peroxide is associated with an induction of the ZEB1 or ZEB2 gene ( Figure 2A), suggesting that ZEB transcription factors orchestrate the mechanism of adaptation to oxidative stress.
  • Example 3 The ZEB1 and ZEB2 proteins induce a specific gene program that gives the cells resistance to oxidative stress.
  • QPRT protein for quinolinate phosphoribosyl transferase is an enzyme originally described as being involved in the kynurenine pathway (Liu et al., 2007). Although its contribution to the de novo production of NAD + is effective in the liver and kidney, the QPRT protein is also present in other cell types, presumably providing other functions, including inhibition of the activation of NAD +. caspase 3 as recently described in HeLa cells (Ishidoh et al., 2010). Depletion of HCEC Esc cells into QPRT induces apoptotic cell death under conditions of chronic inflammation ( Figure 4A). Interestingly, inhibition of QPRT expression also prevents these cells from tolerating oncogenic activations (Figure 4B).
  • the ALEP signature score is parallel to the level of QPRT protein detected in Western blot ( Figure 6). However, although the program is not active in the COLO-320 line, the QPRT protein is detectable at a lower rate. In immunohistochemistry (IHC), the signal detected in this line is sufficiently high to be considered as weakly positive.
  • Inhibition of the QPRT protein induces proliferation arrest of the ALEP-positive colorectal lines as judged by the accumulation of phosphorylated H3 histone on the Ser10 residue. This arrest is accompanied by an increase in autophagy (visualized by the accumulation of LC3 protein) and the accumulation of damage to DNA (visualized by the accumulation of ⁇ 2 ⁇ ). Inactivation of the TD02 enzyme upstream of the QPRT protein in the kynurenine pathway has similar consequences ( Figure 7). ALEP-positive colorectal cells are therefore dependent on the activity of the kynurenine pathway to maintain their proliferation.
  • EXAMPLE 5 The expression of QPRT is induced in the early phases of the development of colorectal cancers, developed on an inflammatory context or not.
  • Example 6 The ALEP program is induced in intestinal stem cells and is a factor of poor prognosis.
  • the gene signature of the ALEP program was established on an epithelial cell model. To refine this signature so that it can be used on tumors (taking into account their cellular heterogeneity), it has been compared to two cohorts of patients suffering from ulcerative colitis (UC) or Crohn's disease (CD) since over seven years (datasets GSE36807 and GSE37283, (Montero-Melendez et al., 2013, Pekow et al., 2013)). These two series were selected because the gene profiles were established on macroscopically non-inflamed mucosa so as to limit the direct impact of inflammation on transcript levels. The analysis of the score of this signature allows in both cohorts of patients to clearly segregate two subgroups (Figure 10 A-B).
  • the ALEP signature score and that of the Lgr5 and EphB2 intestinal stem cell signatures correlate significantly, supporting the hypothesis that ALEP is induced in stem cells. intestinal precursors (Figure 11B).
  • the ALEP signature proves to be predictive of a risk of high recurrence (Figure 11 C).
  • Esophageal adenocarcinomas originate from Barrett's esophagus (located in the lower third of the esophagus) and chronic inflammation related to the rise of gastric juices plays a key role.

Abstract

The invention primarily relates to an in vitro method for selecting a gastrointestinal cancer patient who is likely to respond to anticancer treatment involving the administration of a quinolinate phosphoribosyltransferase (QPRT) inhibitor. A method of the invention involves in particular a step of determining QPRT expression in a test sample of the gastrointestinal tumor, QPRT expression in cells of the test sample being an indicator that the patient is sensitive to a QPRT inhibitor treatment. The invention also relates to the use of QPRT as a biomarker for predicting the sensitivity of a gastrointestinal tumor patient to a QPRT inhibitor treatment.

Description

PROCÉDÉ DE PRÉDICTION DE LA SENSIBILITÉ D'UNE TUMEUR  METHOD FOR PREDICTING THE SENSITIVITY OF A TUMOR
GASTRO-INTESTINALE À UN TRAITEMENT INHIBITEUR  GASTROINTESTINAL TO INHIBITORY TREATMENT
La présente invention se situe dans le domaine médical et plus précisément dans le domaine de la thérapie anti-cancéreuse. L'invention a principalement pour objet un procédé in vitro ou ex vivo de sélection de patients atteints de tumeurs gastro-intestinales susceptibles de bénéficier d'une thérapie anti-tumorale comprenant l'administration d'un inhibiteur de la voie de la kynurénine. Un procédé selon l'invention comprend notamment une étape de détermination de l'expression de la protéine QPRT dans un échantillon test de ladite tumeur gastro-intestinale, l'expression de QPRT dans des cellules épithéliales de l'échantillon test étant indicateur de la sensibilité dudit patient à un traitement inhibiteur de la voie de la kynurénine. The present invention is in the medical field and more specifically in the field of anti-cancer therapy. The main subject of the invention is an in vitro or ex vivo method of selecting patients with gastrointestinal tumors that may benefit from anti-tumor therapy comprising the administration of an inhibitor of the kynurenine pathway. A method according to the invention comprises in particular a step of determining the expression of the QPRT protein in a test sample of said gastrointestinal tumor, the expression of QPRT in epithelial cells of the test sample being indicative of the sensitivity said patient to an inhibitory treatment of the kynurenine pathway.
La présente invention a également pour objet l'utilisation de la protéine QPRT en tant que biomarqueur pour la prédiction de la sensibilité d'un patient atteint d'une tumeur gastro-intestinale à un traitement inhibiteur de la voie de la kynurénine.  The present invention also relates to the use of the protein QPRT as a biomarker for predicting the sensitivity of a patient suffering from a gastrointestinal tumor to an inhibitory treatment of the kynurenine pathway.
Afin d'optimiser les traitements thérapeutiques et la pertinence des essais cliniques dans le domaine de la thérapie anti-cancéreuse, l'identification de nouvelles cibles thérapeutiques et de biomarqueurs prédictifs de la sensibilité des patients à ce traitement, qu'il soit utilisé seul d'un ou en combinaison avec d'autres approches thérapeutiques, demeure un objectif important pour le développement futur de ces traitements. Il est donc nécessaire de disposer de nouveaux traitements et de procédés efficaces permettant de prédire la sensibilité d'un patient à ces traitements lourds et associés à des effets secondaires importants.  In order to optimize therapeutic treatments and the relevance of clinical trials in the field of anti-cancer therapy, the identification of new therapeutic targets and biomarkers predictive of the patients' sensitivity to this treatment, be it used alone. one or in combination with other therapeutic approaches, remains an important goal for the future development of these treatments. It is therefore necessary to have new treatments and effective methods to predict the sensitivity of a patient to these heavy treatments and associated with significant side effects.
Les inventeurs ont maintenant développé un nouveau procédé permettant la sélection de patients atteints d'une tumeur gastro-intestinale susceptibles de bénéficier d'un traitement inhibiteur de la voie de la kynurénine. Ce procédé comprend la détermination de l'expression de la protéine QPRT dans les cellules épithéliales d'un échantillon de la tumeur du patient avant tout traitement, une expression supérieure de la protéine QPRT dans l'échantillon test par rapport à un échantillon de référence étant indicatrice de la sensibilité du patient à un traitement inhibiteur de la voie de la kynurénine.  The inventors have now developed a new method for the selection of patients with a gastrointestinal tumor likely to benefit from an inhibitory treatment of the kynurenine pathway. This method comprises determining the expression of the QPRT protein in the epithelial cells of a sample of the patient's tumor prior to any treatment, a higher expression of the QPRT protein in the test sample compared to a reference sample being indicative of the patient's sensitivity to an inhibitory treatment of the kynurenine pathway.
Plusieurs signatures d'expression génique (« classifiers ») permettant de suivre l'évolution des maladies de Crohn et les colites ont été décrites (Montero-Melendez et al., 2013).  Several gene expression signatures ("classifiers") to monitor the evolution of Crohn's disease and colitis have been described (Montero-Melendez et al., 2013).
La publication de Watanabe (2013) décrit une signature d'expression génique et une méthode de prédiction de l'évolution d'une colite ulcérative vers un cancer colorectal. La signature décrite dans ce document comprend 40 gènes exprimés de façon différentielle selon que les patients atteints de lésions néoplasiques, ou non atteints par de telles lésions.  The publication by Watanabe (2013) describes a gene expression signature and a method for predicting the progression from ulcerative colitis to colorectal cancer. The signature described in this document comprises 40 differentially expressed genes according to whether patients with neoplastic lesions, or not affected by such lesions.
La publication de Malik et al. (2014) décrit la structure cristallographique de la QPRT humaine sous une forme complexée avec son inhibiteur, l'acide phtalique. Cet inhibiteur est cependant peu spécifique, la concentration à laquelle il est effectif in vitro est de l'ordre du millimolaire (10"3M). La publication de Sahm ei al. (2013) enseigne que l'acide quinolique, métabolite endogène du tryptophane et précurseur du NAD+, confère aux gliomes une résistance au stress oxydatif. The publication of Malik et al. (2014) describes the crystallographic structure of human QPRT in a form complexed with its inhibitor, phthalic acid. This inhibitor is however not very specific, the concentration at which it is effective in vitro is of the order of millimolar (10 "3 M). The publication of Sahm et al. (2013) teaches that quinolic acid, an endogenous metabolite of tryptophan and a precursor of NAD + , confers resistance to oxidative stress on gliomas.
L'inflammation chronique constitue un terrain fertile au développement de cancers. Dans les muqueuses enflammées, pour autant le stress oxydatif induit une érosion des télomères et des signes de sénescence réplicative au sein des cellules souches intestinales.  Chronic inflammation is fertile ground for cancer development. In inflamed mucosa, however, oxidative stress induces erosion of telomeres and signs of replicative senescence in intestinal stem cells.
Les inventeurs ont montré que la comparaison du profil d'expression génique de cellules épithéliales de colon humaines ayant échappé à la sénescence induite par un stress oxydatif avec le profil génique de la lignée parentale met en exergue l'induction d'un programme génique spécifique, caractérisé par l'activation de l'expression d'une trentaine de gènes et la répression d'une trentaine d'autres. Ce programme génique est orchestré par les facteurs de transcription ZEB et se différencie du programme de transition épithélio-mésenchymateuse (EMT selon l'acronyme anglais) communément induit par ces facteurs. Ce nouveau programme génique est nommé ALEP pour Alternative to EMT Program. Les inventeurs ont montré in vitro que l'expression de QPRT confère un avantage de survie aux cellules ayant échappé à la sénescence, dans des conditions d'inflammation chronique ou lorsqu'elles sont soumises à un agent oxydant. Les inventeurs ont aussi montré que la déplétion en QPRT des cellules ayant échappé à la sénescence induit la mort de ces cellules par apoptose dans des conditions d'inflammation chronique, et que l'inhibition de l'expression de QPRT empêche ces cellules de tolérer des activations oncogéniques. Ils ont également démontré que des cellules de lignées cancéreuses intestinales ayant un programme ALEP actif (score de la signature élevé) sont dépendantes la protéine QPRT pour leur prolifération. Au-delà de la protéine QPRT, l'inhibition d'une autre enzyme clef de la voie de la kynurénine située en amont de QPRT, la protéine TD02, inhibe de manière très similaire la prolifération de ces lignées. L'activité de la voie de la kynurénine dans sa globalité serait donc nécessaire à la prolifération des lignées colorectales dans lesquelles le programme ALEP est activé.  The inventors have shown that the comparison of the gene expression profile of human colon epithelial cells that has escaped the senescence induced by oxidative stress with the gene profile of the parental line highlights the induction of a specific gene program, characterized by the activation of the expression of about thirty genes and the repression of about thirty others. This gene program is orchestrated by ZEB transcription factors and is differentiated from the epithelio-mesenchymal transition (EMT) program commonly induced by these factors. This new gene program is named ALEP for Alternative to EMT Program. The inventors have shown in vitro that the expression of QPRT confers a survival advantage to cells having escaped senescence, under conditions of chronic inflammation or when they are subjected to an oxidizing agent. The inventors have also shown that QPRT depletion of cells that have escaped senescence induces the death of these cells by apoptosis under conditions of chronic inflammation, and that the inhibition of QPRT expression prevents these cells from tolerating oncogenic activations. They also demonstrated that intestinal cancer cell lines with an active ALEP program (high signature score) are dependent on the QPRT protein for proliferation. Beyond the QPRT protein, the inhibition of another key enzyme of the kynurenine pathway located upstream of QPRT, the TD02 protein, very similarly inhibits the proliferation of these lines. The activity of the kynurenine pathway as a whole would therefore be necessary for the proliferation of colorectal lines in which the ALEP program is activated.
Par ailleurs, les inventeurs ont montré que la protéine QPRT est absente dans les entérocytes de tissu normal de l'intestin, alors qu'une augmentation du nombre de cellules épithéliales marquées lors de la détection de QPRT est observée dans une proportion significative de dysplasies de bas- ou de haut-grade et de cancers développés dans un contexte inflammatoire, ainsi que dans les cellules épithéliales d'une proportion significative d'adénomes et de cancers sporadiques. De plus, une proportion des adénocarcinomes œsophagiens exprime la protéine QPRT alors qu'aucun marquage n'est observé dans l'épithélium malpighien, utilisé comme contrôle interne.  Moreover, the inventors have shown that the QPRT protein is absent in the enterocytes of normal intestinal tissue, whereas an increase in the number of labeled epithelial cells during the detection of QPRT is observed in a significant proportion of dysplasias of low- or high-grade and cancers developed in an inflammatory context, as well as in epithelial cells with a significant proportion of adenomas and sporadic cancers. In addition, a proportion of oesophageal adenocarcinomas express the QPRT protein whereas no labeling is observed in the squamous epithelium, used as internal control.
La protéine QPRT, et au-delà de celle-ci les autres enzymes de la voie de la kynurénine, sont donc des cibles thérapeutiques particulièrement intéressantes et leur inactivation constitue donc une approche thérapeutique anti-tumorale prometteuse, le cas échéant en association avec un autre traitement anti-tumoral, en particulier avec une thérapie capable d'induire un stress oxydant. L'invention sera maintenant décrite de manière détaillée à l'aide d'exemples destinés à l'illustrer sans la limiter, diverses modifications pouvant y être apportées sans sortir du cadre général de l'invention. The QPRT protein, and beyond it the other enzymes of the kynurenine pathway, are therefore particularly interesting therapeutic targets and their inactivation therefore constitutes a promising anti-tumor therapeutic approach, if appropriate in combination with another anti-tumor treatment, in particular with a therapy capable of inducing oxidative stress. The invention will now be described in detail with the aid of examples intended to illustrate it without limiting it, various modifications that may be made without departing from the general scope of the invention.
La présente invention a pour objet un procédé ex vivo de sélection d'un patient atteint d'une tumeur gastro-intestinale, susceptible de bénéficier d'un traitement inhibiteur de la voie de la kynurénine, à partir d'un échantillon test de ladite tumeur avant traitement dudit patient, ledit procédé comprenant les étapes suivantes :  The present invention relates to an ex vivo method of selecting a patient suffering from a gastrointestinal tumor, likely to benefit from an inhibitory treatment of the kynurenine pathway, from a test sample of said tumor before treatment of said patient, said method comprising the following steps:
a) La détermination de l'expression de la protéine QPRT dans les cellules épithéliales dudit échantillon test,  a) determination of the expression of the QPRT protein in the epithelial cells of said test sample,
b) La comparaison de l'expression de la protéine QPRT dans les cellules épithéliales dudit échantillon test avec l'expression de QPRT dans un échantillon de référence, c) La sélection d'un patient susceptible de bénéficier d'un traitement inhibiteur de la voie de la kynurénine à partir de la comparaison de l'étape b), un niveau d'expression supérieur de protéine QPRT dans les cellules épithéliales de l'échantillon test par rapport à celui de l'échantillon de référence étant indicateur de la sensibilité de ladite tumeur à un traitement inhibiteur de la voie de la kynurénine.  b) The comparison of the expression of the QPRT protein in the epithelial cells of the said test sample with the expression of QPRT in a reference sample, c) The selection of a patient likely to benefit from an inhibitor treatment of the pathway of kynurenine from the comparison of step b), a higher expression level of QPRT protein in the test sample epithelial cells than that of the reference sample being indicative of the sensitivity of said test sample. tumor to an inhibitory treatment of the kynurenine pathway.
La présente invention a également pour objet un procédé ex vivo de prédiction de la sensibilité d'un patient atteint d'une tumeur gastro-intestinale (avant traitement) à un traitement inhibiteur de la voie de la kynurénine, ledit procédé comprenant les étapes suivantes : i) l'obtention d'un échantillon test de ladite tumeur, ii) la détermination de l'expression de protéine QPRT dans les cellules épithéliales dudit échantillon test, iii) la comparaison de l'expression de protéine QPRT dans ledit échantillon test avec l'expression de QPRT dans un échantillon de référence, iv) la prédiction de la sensibilité dudit patient à un traitement inhibiteur de la voie de la kynurénine à partir de la comparaison de l'étape iii), un niveau d'expression supérieur de protéine QPRT dans les cellules épithéliales de l'échantillon test par rapport à celui de l'échantillon de référence étant indicateur de la sensibilité dudit patient à un traitement inhibiteur de la voie de la kynurénine.  The present invention also relates to an ex vivo method for predicting the sensitivity of a patient suffering from a gastrointestinal tumor (before treatment) to an inhibitory treatment of the kynurenine pathway, said method comprising the following steps: i) obtaining a test sample of said tumor, ii) determining the expression of QPRT protein in the epithelial cells of said test sample, iii) comparing the expression of QPRT protein in said test sample with the expression of QPRT in a reference sample, iv) predicting the sensitivity of said patient to an inhibitory treatment of the kynurenine pathway from the comparison of step iii), a higher expression level of QPRT protein epithelial cells of the test sample relative to that of the reference sample being indicative of the sensitivity of said patient to an inhibitory treatment of the kynurenine pathway.
Par « sensibilité d'un patient à un traitement », on entend l'observation d'un effet thérapeutique tel que l'amélioration de la survie d'un patient ayant reçu ledit traitement thérapeutique. L'absence de réponse d'un patient, ou l'absence de sensibilité du patient à un traitement thérapeutique, est au contraire définie par le terme de « résistance » d'un patient audit traitement thérapeutique, dans laquelle aucun effet thérapeutique n'est observé. Un procédé ex vivo de sélection d'un patient susceptible de bénéficier d'un traitement inhibiteur de la voie de la kynurénine, permet de prédire la sensibilité d'un patient atteint d'une tumeur gastro-intestinale à un traitement inhibiteur de la voie de la kynurénine.  By "sensitivity of a patient to a treatment" is meant the observation of a therapeutic effect such as improving the survival of a patient having received said therapeutic treatment. The absence of a patient's response, or the patient's lack of sensitivity to a therapeutic treatment, is instead defined by the term "resistance" of a patient to said therapeutic treatment, in which no therapeutic effect is observed. An ex vivo method of selecting a patient likely to benefit from an inhibitory treatment of the kynurenine pathway makes it possible to predict the sensitivity of a patient suffering from a gastrointestinal tumor to an inhibitor treatment of the gastrointestinal tract. kynurenine.
Par l'expression « avant traitement » ou « non-traité », on entend avant ou après un acte chirurgical, tel qu'une exérèse, pratiqué sur la tumeur mais avant tout traitement thérapeutique associé mais non chirurgical, tel que la chimiothérapie, la radiothérapie, l'hormonothérapie ou 1 ' immunothérapie . The expression "before treatment" or "untreated" means before or after a surgical procedure, such as an excision, performed on the tumor but before any therapeutic treatment associated but non-surgical, such as chemotherapy, radiotherapy, hormone therapy or immunotherapy.
La quinolinate phosphoribosyltransférase ou QPRT (UniProtKB Q 15274 mise à jour le 18 mai 2010 ; SEQ ID NO :1), aussi dénommée Nicotinate-nucleotide pyrophosphorylase catalyse la conversion de l'acide quinolinique (QA) en nicotinamide mononucleotide (NAMN) par le transfert du groupe phosphoribosyl du 5-phospho-D-ribose-l-diphosphate (PRPP), lors de la première étape de biosynthèse du NAD+.  Quinolinate phosphoribosyltransferase or QPRT (UniProtKB Q 15274 updated on May 18, 2010, SEQ ID NO: 1), also known as Nicotinate-nucleotide pyrophosphorylase catalyzes the conversion of quinolinic acid (QA) to nicotinamide mononucleotide (NAMN) by transfer. phosphoribosyl group of 5-phospho-D-ribose-1-diphosphate (PRPP), during the first stage of NAD + biosynthesis.
Par « voie de la kynurénine », on entend la voie métabolique conduisant à la production de novo de NAD+ à partir de tryptophane, un acide aminé essentiel. Les enzymes impliquées dans cette voie ont été décrites notamment par Vécsei et al., 2013. By "kynurenine pathway" is meant the metabolic pathway leading to the de novo production of NAD + from tryptophan, an essential amino acid. The enzymes involved in this pathway have been described in particular by Vécsei et al., 2013.
Par « traitement inhibiteur de la voie de la kynurénine » on entend l'application d'un traitement destiné à inhiber l'activité et/ou les effets d'au moins une des enzymes qui contribuent à cette voie métabolique, à savoir le tryptophane 2,3-dioxygénase (TD02), la kynurénine formylase, la kynurénine 3-monooxygénase (KMO), la kynuréninase (KYNU), la 3-hydroxyanthranilic acid oxygénase (3HAO) ou QPRT. Ce traitement inclut par exemple l'utilisation d'inhibiteurs de la protéine tryptophane 2,3-dioxygénase ou TD02 tel que la molécule 690C91 ((E)-6-fluoro-3-[2-(3- pyridyl)vinyl]-lH-indole, Sigma), d'inhibiteurs de la kynurénine 3-monooxygénase ou KMO tel que 2-3 teriflunomide et Ro-61-8048 (Selleckchem), PDE10A (Pfizer), JM6 (3,4-dimethoxy-N-(4- (3-nitrophenyl)-5-(piperidin-l -ylmethyl)thiazol-2-yl)benzenesulfonamide), d'inhibiteurs de l'enzyme 3HAO tels que décrits par Vallerini et al., 2013 ou d'inhibiteurs de QPRT. Un exemple connu d'un antagoniste de l'activité enzymatique de QPRT est l'acide phtalique (Malik et al. 2013).  By "inhibitory treatment of the kynurenine pathway" is meant the application of a treatment intended to inhibit the activity and / or the effects of at least one of the enzymes which contribute to this metabolic pathway, namely tryptophan 2 , 3-dioxygenase (TD02), kynurenine formylase, kynurenine 3-monooxygenase (KMO), kynureninase (KYNU), 3-hydroxyanthranilic acid oxygenase (3HAO) or QPRT. This treatment includes, for example, the use of inhibitors of the tryptophan protein 2,3-dioxygenase or TD02 such as the molecule 690C91 ((E) -6-fluoro-3- [2- (3-pyridyl) vinyl] -1H indole, Sigma), inhibitors of kynurenine 3-monooxygenase or KMO such as 2-3 teriflunomide and Ro-61-8048 (Selleckchem), PDE10A (Pfizer), JM6 (3,4-dimethoxy-N- (4) - (3-nitrophenyl) -5- (piperidin-1-ylmethyl) thiazol-2-yl) benzenesulfonamide), inhibitors of the enzyme 3HAO as described by Vallerini et al., 2013 or QPRT inhibitors. A known example of an antagonist of the enzymatic activity of QPRT is phthalic acid (Malik et al., 2013).
Par « inhibiteur de la voie de la kynurénine » on entend toute substance, molécule, composé ou complexe capable d'inhiber de manière spécifique, notamment par compétition ou conformation, l'activité et/ou les effets d'au moins une enzyme de cette voie métabolique. Par exemple, ces inhibiteurs peuvent être de type allostérique, analogue de substrat(s), analogue de produit(s) ou intermédiaire(s) de réaction enzymatique notamment non catabolisable, spécifiques de l'enzyme concernée.  By "inhibitor of the kynurenine pathway" is meant any substance, molecule, compound or complex capable of specifically inhibiting, in particular by competition or conformation, the activity and / or the effects of at least one enzyme of this type. metabolic pathway. For example, these inhibitors may be of the allosteric type, substrate analog (s), product analogue (s) or enzymatic reaction intermediate (s), especially non-catabolizable, specific for the enzyme concerned.
Dans tous les aspects de l'invention, le traitement inhibiteur de la voie de la kynyrénine est de préférence un traitement inhibiteur de QPRT ou de TD02, et plus particulièrement de QPRT.  In all aspects of the invention, the inhibitory treatment of the kynyrenine pathway is preferably an inhibitor treatment of QPRT or TD02, and more particularly QPRT.
Par conséquent, de manière préférée, l'invention a pour objet un procédé ex vivo de sélection d'un patient atteint d'une tumeur gastro-intestinale, susceptible de bénéficier d'un traitement inhibiteur de la voie de la kynurénine, à partir d'un échantillon test de ladite tumeur avant traitement dudit patient, ledit procédé comprenant les étapes suivantes :  Therefore, preferably, the subject of the invention is an ex vivo method of selecting a patient suffering from a gastrointestinal tumor, likely to benefit from an inhibitory treatment of the kynurenine pathway, from a test sample of said tumor prior to treatment of said patient, said method comprising the following steps:
a) La détermination de l'expression de la protéine QPRT dans les cellules épithéliales dudit échantillon test,  a) determination of the expression of the QPRT protein in the epithelial cells of said test sample,
b) La comparaison de l'expression de la protéine QPRT dans les cellules épithéliales dudit échantillon test avec l'expression de QPRT dans un échantillon de référence, c) La sélection d'un patient susceptible de bénéficier d'un traitement inhibiteur de la voie de la kynurénine à partir de la comparaison de l'étape b), un niveau d'expression supérieur de protéine QPRT dans les cellules épithéliales de l'échantillon test par rapport à celui de l'échantillon de référence étant indicateur de la sensibilité de ladite tumeur à un traitement inhibiteur de la voie de la kynurénine. b) comparing the expression of the QPRT protein in the epithelial cells of said test sample with the expression of QPRT in a reference sample, c) The selection of a patient likely to benefit from an inhibitory treatment of the kynurenine pathway from the comparison of step b), a higher expression level of QPRT protein in the epithelial cells of the sample test relative to that of the reference sample being indicative of the sensitivity of said tumor to an inhibitory treatment of the kynurenine pathway.
Selon un aspect particulier, l'invention a pour objet un procédé ex vivo de sélection d'un patient atteint d'une tumeur gastro-intestinale, avant traitement, susceptible de bénéficier d'un traitement inhibiteur de la voie de la kynurénine, comprenant la détermination du niveau d'expression de protéine QPRT dans les cellules épithéliales d'un échantillon test de tumeur, dans lequel l'échantillon test de tumeur est choisi parmi : un tissu tumoral, une pièce opératoire et une biopsie.  According to a particular aspect, the subject of the invention is an ex vivo method of selecting a patient suffering from a gastrointestinal tumor, before treatment, likely to benefit from an inhibitory treatment of the kynurenine pathway, comprising the determining the level of expression of QPRT protein in the epithelial cells of a tumor test sample, wherein the tumor test sample is selected from: tumor tissue, an operative specimen and a biopsy.
Le procédé de sélection selon l'invention concerne un procédé ex vivo d'un patient atteint d'une tumeur gastro-intestinale susceptible de bénéficier d'un traitement inhibiteur de la voie de la kynurénine, avant que ledit patient n'ait été traité pour ladite tumeur.  The selection method according to the invention relates to an ex vivo method of a patient suffering from a gastrointestinal tumor likely to benefit from an inhibitory treatment of the kynurenine pathway, before said patient has been treated for said tumor.
D'une manière générale, dans le cadre de la présente invention, l'échantillon issu du patient atteint d'une tumeur gastro-intestinale est obtenu avant traitement ; ledit patient étant considéré comme non-traité pour ladite tumeur.  In general, in the context of the present invention, the sample from the patient with a gastrointestinal tumor is obtained before treatment; said patient being considered untreated for said tumor.
L'échantillon de la tumeur gastro-intestinale peut être issu de la tumeur primaire ou bien de métastase(s). Selon un aspect particulier du procédé de sélection ex vivo selon l'invention, l'échantillon test de ladite tumeur est issu de la tumeur primaire.  The sample of the gastrointestinal tumor may be derived from the primary tumor or from metastasis (s). According to a particular aspect of the ex vivo selection process according to the invention, the test sample of said tumor is derived from the primary tumor.
L'expression «niveau d'expression supérieur » indique que le niveau d'expression observé dans ledit échantillon test est plus important que le niveau d'expression observé dans l'échantillon de référence, du point de vue du nombre de cellules dans lesquelles l'expression est détectée et/ou de l'intensité du signal détecté et/ou de la localisation des cellules pour lesquelles la protéine est détectée.  The expression "higher expression level" indicates that the level of expression observed in said test sample is greater than the level of expression observed in the reference sample, in terms of the number of cells in which the Expression is detected and / or the intensity of the detected signal and / or the location of the cells for which the protein is detected.
En effet, les inventeurs ont montré que la protéine QPRT est absente dans les entérocytes de tissu normal de l'intestin, alors qu'une augmentation du nombre de cellules épithéliales marquées lors de la détection de QPRT est observée dans une proportion significative de dysplasies de bas- ou de haut- grade et de cancers développés dans un contexte inflammatoire, ainsi que dans les cellules épithéliales d'une proportion significative d'adénomes et des cancers sporadiques.  Indeed, the inventors have shown that the QPRT protein is absent in the enterocytes of normal intestinal tissue, whereas an increase in the number of labeled epithelial cells during the detection of QPRT is observed in a significant proportion of dysplasias of low- or high-grade and cancers developed in an inflammatory context, as well as in epithelial cells with a significant proportion of adenomas and sporadic cancers.
De plus, une proportion des adénocarcinomes œsophagiens exprime la protéine QPRT alors qu'aucun marquage n'est observé dans l'épithélium malpighien, utilisé comme contrôle interne.  In addition, a proportion of oesophageal adenocarcinomas express the QPRT protein whereas no labeling is observed in the squamous epithelium, used as internal control.
De façon avantageuse, dans un procédé selon l'invention, l'expression de la protéine QPRT est déterminée par comparaison avec l'expression de la protéine QPRT dans un échantillon de référence, l'expression de la protéine QPRT dans un échantillon de référence étant notamment choisie parmi : l'expression de QPRT dans un échantillon test de tissu sain de même nature que le tissu de l'échantillon test, l'expression de QPRT dans un échantillon de tissu d'un patient atteint d'une maladie gastro-intestinale inflammatoire non associée à des lésions dysplasiques (c'est-à-dire de nature non tumorale), et/ou l'expression de QPRT dans un échantillon de tissu du patient testé, prélevé alors que le patient n'était pas atteint d'une tumeur gastro-intestinale. Advantageously, in a method according to the invention, the expression of the QPRT protein is determined by comparison with the expression of the QPRT protein in a reference sample, the expression of the QPRT protein in a reference sample being especially chosen from: the expression of QPRT in a test sample of healthy tissue of the same nature as the tissue of the test sample, the expression of QPRT in a tissue sample of a patient reaches inflammatory gastrointestinal disease not associated with dysplastic (ie, non-tumor) lesions, and / or expression of QPRT in a tissue sample of the subject patient taken from the patient did not have a gastrointestinal tumor.
Les maladies gastro-intestinales inflammatoires non tumorales connues sont : la maladie de Crohn, la colite ulcérative ; la gastrite chronique et les reflux gastro-œsophagiens chroniques (dans le cadre de l'adénocarcinome).  The known non-tumorous inflammatory gastrointestinal diseases are: Crohn's disease, ulcerative colitis; chronic gastritis and chronic gastroesophageal reflux (as part of adenocarcinoma).
La détermination du niveau d'expression de la protéine QPRT est réalisée quantitativement ou semi-quantitativement par tout procédé connu par l'homme du métier permettant la détection spécifique et/ou la quantification d'une protéine particulière dans un échantillon. Un tel procédé de détermination est notamment choisi parmi les procédés utilisant des anticorps liant QPRT, incluant en particulier l'immunohistochimie (IHC), une détection par immunofluorescence (IF).  The determination of the level of expression of the QPRT protein is carried out quantitatively or semi-quantitatively by any method known to those skilled in the art allowing the specific detection and / or quantification of a particular protein in a sample. Such a determination method is chosen in particular from methods using QPRT-binding antibodies, including in particular immunohistochemistry (IHC) and immunofluorescence (IF) detection.
L'expression de QPRT peut être définie à l'aide d'un score histologique prenant en compte, d'une part, l'intensité de marquage observée et, d'autre part, le pourcentage de cellules marquées, ou en combinant le pourcentage de cellules marquées et l'intensité de marquage. Selon ce procédé, des sections d'échantillon tumoral sont analysées par microscopie après avoir mis en présence lesdites sections avec un anticorps liant spécifiquement QPRT.  The expression of QPRT can be defined by means of a histological score taking into account, on the one hand, the observed marking intensity and, on the other hand, the percentage of labeled cells, or by combining the percentage labeled cells and the intensity of labeling. According to this method, tumor sample sections are analyzed by microscopy after bringing said sections into contact with an antibody specifically binding QPRT.
Par « cellules marquées » on entend des cellules pour lesquelles un signal est détecté lors de la recherche de l'expression de la protéine QPRT.  By "labeled cells" is meant cells for which a signal is detected when looking for expression of the QPRT protein.
Selon un mode plus particulier de réalisation, l'invention a pour objet un procédé ex vivo de sélection d'un patient atteint d'une tumeur gastro-intestinale susceptible de bénéficier d'un traitement inhibiteur de la voie de la kynurénine, dans lequel la détermination de l'expression de la protéine QPRT comprend la détermination de la présence de QPRT comprenant au moins une étape d'immunohistochimie.  According to a more particular embodiment, the subject of the invention is an ex vivo method of selecting a patient suffering from a gastrointestinal tumor likely to benefit from an inhibitory treatment of the kynurenine pathway, in which the Determination of QPRT protein expression comprises determining the presence of QPRT comprising at least one immunohistochemistry step.
Par « immunohistochimie », on entend d'une méthode de localisation de protéines dans les cellules d'une coupe de tissu, par la détection d'un antigène particulier au moyen d'anticorps polyclonaux ou monoclonaux liant cet antigène. Le couple anticorps-antigène peut être visualisé par exemple par la conjugaison à une enzyme qui peut catalyser une réaction de production de couleur, par l'utilisation d'anticorps marqués, notamment par un fluorophore, ou par l'utilisation d'anticorps dits « secondaires » capables de lier les anticorps liant l'antigène.  By "immunohistochemistry" is meant a method of locating proteins in the cells of a tissue section, by detecting a particular antigen by means of polyclonal or monoclonal antibodies binding that antigen. The antibody-antigen pair can be visualized for example by conjugation to an enzyme which can catalyze a color production reaction, by the use of labeled antibodies, in particular by a fluorophore, or by the use of antibodies known as " secondary antibodies capable of binding antigen-binding antibodies.
Dans une méthode d'immunohistochimie, le tissu est placé sur un support solide. Un anticorps primaire lié à un système de détection (ou un anticorps primaire suivi d'un anticorps secondaire) est ajouté. Le signal est observé par des techniques de microscopie.  In an immunohistochemical method, the tissue is placed on a solid support. A primary antibody bound to a detection system (or a primary antibody followed by a secondary antibody) is added. The signal is observed by microscopy techniques.
Dans un mode particulier de réalisation d'un procédé selon l'invention de sélection d'un patient atteint d'une tumeur gastro-intestinale susceptible de bénéficier d'un traitement inhibiteur de la voie de la kynurénine, l'échantillon test de ladite tumeur est mis en contact avec un anticorps liant QPRT, ledit anticorps étant choisi parmi les anticorps polyclonaux anti-QPRT et les anticorps monoclonaux anti-QPRT. Selon un mode particulier de réalisation d'un procédé selon l'invention, ledit anticorps lie QPRT en conditions natives, l'homme de l'art peut aisément choisir un anticorps répondant à ce critère, on peut citer notamment l'anticorps Sigma HPA011887. In a particular embodiment of a method according to the invention for selecting a patient suffering from a gastrointestinal tumor likely to benefit from an inhibitory treatment of the kynurenine pathway, the test sample of said tumor is contacted with a QPRT-binding antibody, said antibody being selected from polyclonal anti-QPRT antibodies and anti-QPRT monoclonal antibodies. According to a particular embodiment of a method according to the invention, said antibody binds QPRT under native conditions, one skilled in the art can easily choose an antibody meeting this criterion, there may be mentioned in particular the Sigma HPA011887 antibody.
Selon un mode encore plus particulier de réalisation, l'invention a pour objet un procédé in vitro de sélection d'un patient atteint d'une tumeur gastro-intestinale susceptible de bénéficier d'un traitement inhibiteur de la voie de la kynurénine, dans lequel la détermination de la présence de la protéine QPRT dans l'échantillon test de ladite tumeur est réalisée au moyen d'au moins une étape d'immunohistochimie comprenant la localisation de cellules marquées au sein du tissu épithélial de l'échantillon test.  According to an even more particular embodiment, the subject of the invention is an in vitro method for selecting a patient suffering from a gastrointestinal tumor that may benefit from an inhibitory treatment of the kynurenine pathway, in which the determination of the presence of the QPRT protein in the test sample of said tumor is carried out by means of at least one immunohistochemistry step comprising the localization of labeled cells within the epithelial tissue of the test sample.
Selon un mode encore plus particulier, un procédé ex vivo de prédiction de la sensibilité d'un patient atteint d'une tumeur gastro-intestinale à un traitement inhibiteur de la voie de la kynurénine, selon l'invention, comprend la détermination du pourcentage de cellules épithéliales marquées au sein du tissu épithélial de l'échantillon test.  According to an even more particular embodiment, an ex vivo method for predicting the sensitivity of a patient suffering from a gastrointestinal tumor to an inhibitory treatment of the kynurenine pathway, according to the invention, comprises the determination of the percentage of labeled epithelial cells within the epithelial tissue of the test sample.
Selon un aspect particulier, l'invention a pour objet un procédé ex vivo de sélection d'un patient atteint d'une tumeur gastro-intestinale susceptible de bénéficier d'un traitement inhibiteur de la voie de la kynurénine, dans lequel la détermination de l'expression de protéine QPRT comprend la détermination du pourcentage de cellules tumorales marquées.  According to a particular aspect, the subject of the invention is an ex vivo method of selecting a patient suffering from a gastrointestinal tumor likely to benefit from an inhibitory treatment of the kynurenine pathway, in which the determination of the QPRT protein expression comprises determining the percentage of labeled tumor cells.
Selon un aspect plus particulier, dans un procédé selon l'invention, un niveau d'expression de protéine QPRT dans l'échantillon test correspondant à au moins 10% des cellules épithéliales marquées est indicateur de la sensibilité de ladite tumeur à un traitement inhibiteur de la voie de la kynurénine.  According to a more particular aspect, in a method according to the invention, a level of expression of QPRT protein in the test sample corresponding to at least 10% of the labeled epithelial cells is indicative of the sensitivity of said tumor to a treatment inhibitor of the way of kynurenine.
Dans le cadre de la présente invention, il est également décrit un procédé ex vivo de prédiction de la sensibilité d'un patient atteint d'une tumeur à un traitement inhibiteur de la voie la voie de la kynurénine, dans lequel la détermination du niveau d'expression de QPRT comprend la détermination du niveau d'expression d'un ARNm codant pour la protéine QPRT. Plus particulièrement, un procédé selon l'invention comprend la détermination de la quantité d'un ARNm codant pour la protéine QPRT, en particulier dans les cellules épithéliales de la tumeur. La détermination de la quantité de l'ARNm codant pour la protéine QPRT peut être réalisée par tout procédé connu par l'homme du métier permettant la détection spécifique d'un ARNm, par exemple après microdissection des cellules épithéliales d'une tumeur. Un tel procédé comprend notamment la détection spécifique d'un ARNm à l'aide de sondes d'acide nucléique complémentaires de la séquence dudit ARNm, notamment par RT-PCR, par un procédé de type Northern-Blot. Elle peut être également obtenue par hybridation sur une sonde immobilisée (Hybridation in situ).  In the context of the present invention, there is also disclosed an ex vivo method for predicting the sensitivity of a tumor patient to an inhibitory treatment of the kynurenine pathway, wherein the determination of the level of QPRT expression includes the determination of the level of expression of an mRNA encoding the QPRT protein. More particularly, a method according to the invention comprises determining the amount of an mRNA encoding the QPRT protein, particularly in the epithelial cells of the tumor. The determination of the amount of the mRNA encoding the QPRT protein can be carried out by any method known to those skilled in the art allowing the specific detection of an mRNA, for example after microdissection of the epithelial cells of a tumor. Such a method notably comprises the specific detection of an mRNA using nucleic acid probes complementary to the sequence of said mRNA, in particular by RT-PCR, by a Northern-Blot type method. It can also be obtained by hybridization on an immobilized probe (in situ hybridization).
La PCR quantitative permet de mesurer la quantité initiale d'acide nucléique. Les acides nucléiques amplifiés, et notamment les ARNm, peuvent être détectés et quantifiés par tout procédé connu par un homme du métier, tel que notamment par spectrométrie à 260 nm ou par Northern blot en utilisant un procédé d'hybridation de sondes radioactives ou fluorescentes sur l'acide nucléique amplifié. La séquence nucléotidique de l'ARNm correspondant à la quinolinate phosphoribosyltransférase est connue (NCBI référence NM 014298, mise à jour le 15 mars 2015, SEQ ID NO :2). Un homme du métier spécialiste du domaine technique concevra aisément, à partir de cette séquence nucléotidique, des amorces capables de s'hybrider de manière spécifique à l'acide nucléique codant pour QPRT, permettant ainsi sa détection. A titre d'exemple, les amorces décrites dans la présente demande de brevet (SEQ ID NO : 3 et SEQ ID NO : 4) sont utilisables dans un procédé selon l'invention. Quantitative PCR is used to measure the initial amount of nucleic acid. The amplified nucleic acids, and in particular the mRNAs, can be detected and quantified by any method known to a person skilled in the art, such as in particular by 260 nm spectrometry or by Northern blotting using a hybridization method for radioactive or fluorescent probes. amplified nucleic acid. The nucleotide sequence of the mRNA corresponding to quinolinate phosphoribosyltransferase is known (NCBI reference NM 014298, updated on March 15, 2015, SEQ ID NO: 2). A person skilled in the technical field will readily conceive, from this nucleotide sequence, primers capable of specifically hybridizing to the nucleic acid encoding QPRT, thus enabling its detection. By way of example, the primers described in the present patent application (SEQ ID NO: 3 and SEQ ID NO: 4) can be used in a process according to the invention.
Selon un autre aspect particulier, l'invention a pour objet un procédé ex vivo de sélection d'un patient atteint d'une tumeur gastro-intestinale susceptible de bénéficier d'un traitement inhibiteur de la voie de la kynurénine, à partir d'un échantillon test de ladite tumeur avant tout traitement, ledit procédé comprenant les étapes suivantes :  According to another particular aspect, the subject of the invention is an ex vivo method of selecting a patient suffering from a gastrointestinal tumor likely to benefit from an inhibitory treatment of the kynurenine pathway, starting from a sample of said tumor before any treatment, said method comprising the following steps:
a) La détermination, dans ledit échantillon test, du niveau de l'expression de la protéine QPRT et d'au moins une autre protéine du programme ALEP, dans les cellules épithéliales de la tumeur  a) Determining, in said test sample, the level of expression of the QPRT protein and at least one other protein of the ALEP program, in the epithelial cells of the tumor
b) La comparaison de l'expression protéique, dans ledit échantillon test, de QPRT et d'au moins une autre protéine du programme ALEP, avec l'expression protéique de QPRT et d'au moins l'autre protéine du programme ALEP, dans les cellules épithéliales d'un échantillon de référence,  b) comparing the protein expression, in said test sample, of QPRT and at least one other protein of the ALEP program, with the protein expression of QPRT and at least the other protein of the ALEP program, in the epithelial cells of a reference sample,
c) La sélection d'un patient susceptible de bénéficier d'un traitement inhibiteur de la voie de la kynurénine à partir de la comparaison de l'étape b),  c) The selection of a patient likely to benefit from an inhibitory treatment of the kynurenine pathway from the comparison of step b),
i. un niveau d'expression supérieur de protéine QPRT dans les cellules épithéliales de l'échantillon test et un niveau d'expression supérieur d'au moins une autre protéine du programme ALEP dans les cellules épithéliales de l'échantillon test par rapport à celui de l'échantillon de référence, et/ou  i. a higher expression level of QPRT protein in the test sample epithelial cells and a higher level of expression of at least one other ALEP program protein in the test sample epithelial cells than in the test sample reference sample, and / or
ii. un niveau d'expression supérieur de protéine QPRT dans les cellules épithéliales de l'échantillon test et un niveau d'expression inférieure d'au moins une protéine du programme ALEP dans les cellules épithéliales de l'échantillon test par rapport à celui de l'échantillon de référence étant indicateur de la sensibilité de ladite tumeur à un traitement inhibiteur de la voie de la kynurénine.  ii. a higher expression level of QPRT protein in the test sample epithelial cells and a lower level of expression of at least one ALEP program protein in the test sample epithelial cells relative to that of the test sample. reference sample being indicative of the sensitivity of said tumor to an inhibitory treatment of the kynurenine pathway.
Les protéines du programme ALEP, désigne les 59 protéines codées par les gènes indiqués dans la Figure 13, dont les inventeurs ont démontré que l'expression est soit supérieure soit inférieure dans des cellules épithéliales résistantes au stress oxydatif.  ALEP program proteins refer to the 59 proteins encoded by the genes shown in Figure 13, which the inventors have demonstrated that the expression is either higher or lower in epithelial cells resistant to oxidative stress.
Les gènes codant les protéines du programme ALEP dont l'expression est supérieure sont : The genes encoding the ALEP program proteins whose expression is greater are:
APOE, SPARC, C6ORF160, SNHG5, SLC2A3, GALC, MT1G, SH3GL2, GNE, SUNC1, EXOSC8, LCPl, KCNJ8, NDRGl, SNCG, FGD3, LGALSl, TXNIP, SUM03, EMILIN2, TNNCl, TNNT1, TGFB1, LOC728031, KRT19 et GABARAPL1. Les gènes codant les protéines du programme ALEP dont l'expression est inférieure sont : IL32, ETV5, S100A3, SGK1, PRSS1, IRAK2, S100A9, GPR56, TNFAIP3, PIM2, PRRX2, BIRC3, CXCL5, DIAPH1, AD AMI 9, LRIG1, SLC7A11, EVL, WNT5A, TNFAIP2, EN02, TNFRSF1B, PROM1, TNF, TMPRSS2, LOC100134294, NFIX, RIN2, CXCL1, CXCL2, CARD9, CCL20. APOE, SPARC, C6ORF160, SNHG5, SLC2A3, GALC, MT1G, SH3GL2, GNE, SUNC1, EXOSC8, LCP1, KCNJ8, NDRG1, SNCG, FGD3, LGALS1, TXNIP, SUM03, EMILIN2, TNNC1, TNNT1, TGFB1, LOC728031, KRT19 and GABARAPL1. The genes coding for the ALEP program proteins whose expression is lower are: IL32, ETV5, S100A3, SGK1, PRSS1, IRAK2, S100A9, GPR56, TNFAIP3, PIM2, PRRX2, BIRC3, CXCL5, DIAPH1, AD AMI9, LRIG1, SLC7A11, EVL, WNT5A, TNFAIP2, EN02, TNFRSF1B, PROM1, TNF, TMPRSS2, LOC100134294, NFIX, RIN2, CXCL1, CXCL2, CARD9, CCL20.
De préférence, l'étape b) consiste à comparer l'expression de la protéine QPRT et au moins l'expression de la protéine APOE dans les cellules épithéliales dudit échantillon test, étant entendu qu'un niveau d'expression supérieur de QPRT et de APOE est indicateur de la sensibilité de ladite tumeur à un traitement inhibiteur de la voie de la kynurénine, en particulier à un inhibiteur de QPRT ou TD02, en tout particulièrement à un inhibiteur de QPRT.  Preferably, step b) consists in comparing the expression of the QPRT protein and at least the expression of the APOE protein in the epithelial cells of said test sample, it being understood that a higher expression level of QPRT and of APOE is indicative of the sensitivity of said tumor to an inhibitory treatment of the kynurenine pathway, in particular to a QPRT or TD02 inhibitor, in particular to a QPRT inhibitor.
Selon un mode particulier de réalisation, dans un procédé ex vivo de prédiction de la sensibilité d'un patient atteint d'une tumeur gastro-intestinale à un traitement inhibiteur de la voie de la kynurénine, selon l'invention, ladite tumeur gastro-intestinale est choisie parmi : le cancer colorectal, le cancer gastrique et l'adénocarcinome de l'œsophage.  According to a particular embodiment, in an ex vivo method for predicting the sensitivity of a patient suffering from a gastrointestinal tumor to an inhibitory treatment of the kynurenine pathway, according to the invention, said gastrointestinal tumor is selected from: colorectal cancer, gastric cancer and adenocarcinoma of the esophagus.
L'expression « tumeur » désigne une tumeur maligne ou cancer.  The term "tumor" refers to a malignant tumor or cancer.
L'expression « cancer colorectal » désigne une tumeur issue de la transformation néoplasique des cellules épithéliales de la muqueuse du colon ou du rectum. Le cancer colorectal peut se développer sur un contexte inflammatoire (colites ulcératives ou maladie de Crohn) ou non et selon résulter de la progression maligne de dysplasie ou de polypes. Selon la taille et la profondeur de la tumeur, l'infiltration des ganglions et leur nombre, les tumeurs sont classées des stades de O à 4 (classification TNM).  The term "colorectal cancer" refers to a tumor resulting from the neoplastic transformation of the epithelial cells of the mucosa of the colon or rectum. Colorectal cancer can develop in an inflammatory context (ulcerative colitis or Crohn's disease) or not and may result from malignant progression of dysplasia or polyps. Depending on the size and depth of the tumor, ganglion infiltration and number, the tumors are classified as O to 4 (TNM classification).
L'adénocarcinome gastrique désigne une tumeur issue de la transformation néoplasique de la muqueuse de l'estomac et résulte de la progression d'une gastrite chronique en gastrite chronique atropique, métaplasie, dysplasie avant l'éventuelle évolution en adénocarcinome. On distingue les stades localisés, localement avancés et métastatiques.  Gastric adenocarcinoma refers to a tumor resulting from the neoplastic transformation of the stomach lining and results from the progression of chronic gastritis in atropic chronic gastritis, metaplasia, dysplasia before possible progression to adenocarcinoma. Localized, locally advanced and metastatic stages are distinguished.
L'adénocarcinome œsophagien désigne une tumeur issue de l'épithélium de la glande de Esophageal adenocarcinoma refers to a tumor derived from the epithelium of the
Barrett et résulte d'une cicatrisation anormale de lésions liées à l'agression chronique de la muqueuse du bas de l'œsophage par un reflux gastro-œsophagiens chronique. Les tumeurs sont classées selon la classification TNM). Barrett and results from an abnormal healing of lesions related to chronic aggression of the lower esophageal lining by chronic gastroesophageal reflux. Tumors are classified according to the TNM classification).
Selon un mode plus particulier de réalisation, dans un procédé ex vivo de prédiction de la sensibilité d'un patient atteint d'une tumeur gastro-intestinale à un traitement inhibiteur de la voie de la kynurénine, selon l'invention, ladite tumeur gastro-intestinale est développée dans un contexte inflammatoire chronique.  According to a more particular embodiment, in an ex vivo method for predicting the sensitivity of a patient suffering from a gastrointestinal tumor to an inhibitory treatment of the kynurenine pathway, according to the invention, said gastrointestinal tumor is intestinal is developed in a chronic inflammatory context.
L'expression « contexte inflammatoire chronique » désigne la répétition de phases d'inflammation et de rémission, cette période peut s'étendre sur des années, comme pour les patients atteints de maladie de Crohn ou de colites ulcératives, conduisant progressivement à une altération de l'épithélium, en induisant par exemple une instabilité génétique dans les cellules épithéliales. Cette inflammation chronique augmente sur du long terme l'incidence de cancers. Selon un mode encore plus particulier de réalisation, l'invention a pour objet un procédé ex vivo de prédiction de la sensibilité d'un patient atteint d'une tumeur gastro-intestinale développée sur un contexte inflammatoire chronique à un traitement inhibiteur de la voie de la kynurénine, selon l'invention, ladite tumeur gastro-intestinale étant un cancer colorectal. The term "chronic inflammatory context" refers to the repetition of stages of inflammation and remission, this period can extend over years, as for patients with Crohn's disease or ulcerative colitis, leading progressively to an alteration of the epithelium, for example by inducing genetic instability in the epithelial cells. This chronic inflammation increases the incidence of cancer in the long term. According to an even more specific embodiment, the subject of the invention is an ex vivo method for predicting the sensitivity of a patient suffering from a gastrointestinal tumor developed on a chronic inflammatory context to an inhibitor treatment of the gastrointestinal tract. kynurenine, according to the invention, said gastrointestinal tumor being a colorectal cancer.
Selon un autre mode particulier de réalisation, dans un procédé ex vivo de prédiction de la sensibilité d'un patient atteint d'une tumeur gastro-intestinale à un traitement inhibiteur de la voie de la kynurénine, selon l'invention, ladite tumeur gastro-intestinale est un cancer colorectal sporadique.  According to another particular embodiment, in an ex vivo method for predicting the sensitivity of a patient suffering from a gastrointestinal tumor to an inhibitory treatment of the kynurenine pathway, according to the invention, said gastrointestinal tumor intestinal is a sporadic colorectal cancer.
Les tumeurs qui ne se développent pas sur un contexte inflammatoire sont dites sporadiques, elles résultent en général de la mutation du gène APC.  Tumors that do not develop in an inflammatory context are said to be sporadic; they generally result from the mutation of the APC gene.
Selon un autre mode particulier de réalisation, dans un procédé ex vivo de prédiction de la sensibilité d'un patient atteint d'une tumeur gastro-intestinale à un traitement inhibiteur de la voie de la kynurénine, selon l'invention, ladite tumeur gastro-intestinale est un cancer colorectal qui atteint des patients ayant des mutations germinales sur le gène APC. Ces patients développent des polyposes adénomateuses juvéniles (FAP), qui peuvent évoluer en CRC.  According to another particular embodiment, in an ex vivo method for predicting the sensitivity of a patient suffering from a gastrointestinal tumor to an inhibitory treatment of the kynurenine pathway, according to the invention, said gastrointestinal tumor intestinal is a colorectal cancer that affects patients with germline mutations on the APC gene. These patients develop juvenile adenomatous polyposes (PAF), which can evolve into CRC.
Selon un autre aspect particulier, l'invention a pour objet un procédé ex vivo de prédiction de la sensibilité d'un patient atteint d'une tumeur à un traitement inhibiteur de la voie de la kynurénine, combiné à un autre traitement anti-tumoral. Plus particulièrement, dans un procédé selon l'invention, ledit traitement anti-tumoral est choisi parmi : la chirurgie, la chimiothérapie et la radiothérapie.  According to another particular aspect, the subject of the invention is an ex vivo method for predicting the sensitivity of a patient suffering from a tumor to an inhibitory treatment of the kynurenine pathway, combined with another antitumor treatment. More particularly, in a method according to the invention, said anti-tumor treatment is chosen from: surgery, chemotherapy and radiotherapy.
Selon un aspect encore plus particulier, l'invention a pour objet un procédé ex vivo de prédiction de la sensibilité d'un patient atteint d'une tumeur gastro-intestinale à un traitement inhibiteur de la voie de la kynurénine, ledit patient ayant fait l'objet d'une étape de chirurgie destinée à retirer ladite tumeur (ou exérèse).  According to a still more particular aspect, the subject of the invention is an ex vivo method for predicting the sensitivity of a patient suffering from a gastrointestinal tumor to an inhibitory treatment of the kynurenine pathway, said patient having made the subject of a surgical step for removing said tumor (or excision).
La présente invention a également pour objet un procédé ex vivo d'identification d'une tumeur susceptible de répondre à un traitement comprenant l'administration d'un agent inhibiteur de la kynurénine, à partir d'un échantillon test de ladite tumeur avant traitement dudit patient, ledit procédé comprenant les étapes suivantes :  The subject of the present invention is also an ex vivo method for identifying a tumor capable of responding to a treatment comprising the administration of a kynurenine inhibiting agent, from a test sample of said tumor before treatment of said tumor. patient, said method comprising the steps of:
a. La détermination de l'expression de QPRT dans les cellules épithéliales dudit échantillon test,  at. Determination of QPRT expression in the epithelial cells of said test sample,
b. La comparaison de l'expression de QPRT dans les cellules épithéliales dudit échantillon test avec l'expression de QPRT dans les cellules épithéliales d'un échantillon de référence,  b. Comparing QPRT expression in epithelial cells of said test sample with expression of QPRT in epithelial cells of a reference sample,
c. L'identification de la tumeur susceptible de répondre à un traitement comprenant l'administration d'un agent inhibiteur de la voie de la kynurénine, à partir de la comparaison de l'étape b), un niveau d'expression supérieur de QPRT dans les cellules épithéliales de l'échantillon test par rapport à celui des cellules épithéliales d'un échantillon de référence étant indicateur de la sensibilité de ladite tumeur à un traitement inhibiteur de la voie de la kynurénine. vs. The identification of the tumor likely to respond to a treatment comprising the administration of an inhibitory agent of the kynurenine pathway, from the comparison of step b), a higher expression level of QPRT in the epithelial cells of the test sample compared to that of the epithelial cells of a reference sample being indicative of the sensitivity of said tumor to an inhibitory treatment of the kynurenine pathway.
Dans le cadre de cet objet de l'invention, l'expression de QPRT peut être déterminée par l'expression protéique de QPRT ou l'expression transcriptionnelle de QPRT, en particulier l'expression de l'ARNm de QPRT. Selon un mode de réalisation particulièrement préférée de cet objet de l'invention, c'est l'expression de la protéine QPRT qui est déterminée.  In the context of this subject of the invention, the expression of QPRT can be determined by the protein expression of QPRT or the transcriptional expression of QPRT, in particular the expression of the QPRT mRNA. According to a particularly preferred embodiment of this subject of the invention, it is the expression of the QPRT protein which is determined.
Selon un autre aspect, l'invention a pour objet l'utilisation de QPRT en tant que biomarqueur protéique pour la prédiction de la sensibilité d'un patient atteint d'une tumeur gastrointestinale à un traitement inhibiteur de la voie de la kynurénine.  In another aspect, the invention relates to the use of QPRT as a protein biomarker for predicting the sensitivity of a patient with a gastrointestinal tumor to an inhibitory treatment of the kynurenine pathway.
L'invention a également pour objet l'utilisation de QPRT en tant que biomarqueur pour la prédiction de la sensibilité d'un patient atteint d'une tumeur gastro-intestinale à un traitement inhibiteur de la voie de la kynurénine, de préférence choisi parmi un inhibiteur de QPRT et un inhibiteur de TD02, de manière encore plus préférée à un traitement inhibiteur de QPRT, ledit patient étant non-traité pour ladite tumeur et l'expression de la protéine QPRT étant mesurée dans les cellules épithéliales d'un échantillon de ladite tumeur.  The invention also relates to the use of QPRT as a biomarker for predicting the sensitivity of a patient suffering from a gastrointestinal tumor to an inhibitory treatment of the kynurenine pathway, preferably chosen from a QPRT inhibitor and a TD02 inhibitor, even more preferably QPRT inhibitor treatment, said patient being untreated for said tumor and QPRT protein expression being measured in epithelial cells of a sample of said tumor.
Selon un aspect particulier, l'invention a également pour objet l'utilisation de QPRT en tant que biomarqueur pour la prédiction de la sensibilité d'un patient atteint d'une tumeur gastrointestinale à un traitement inhibiteur de la voie de la kynurénine, de préférence choisi parmi un inhibiteur de QPRT et un inhibiteur de TD02, combiné à un traitement anti-tumoral, tel l'oxaliplatine, 5-FU ou irrinotecan, et de manière encore plus préférence de la sensibilité d'un patient atteint d'une tumeur gastro-intestinale à un traitement inhibiteur de QPRT.  In a particular aspect, the invention also relates to the use of QPRT as a biomarker for predicting the sensitivity of a patient suffering from a gastrointestinal tumor to an inhibitory treatment of the kynurenine pathway, preferably selected from a QPRT inhibitor and a TD02 inhibitor, combined with an anti-tumor treatment, such as oxaliplatin, 5-FU or irrinotecan, and even more preferably the sensitivity of a patient with a gastrointestinal tumor -intestinal to an inhibitory treatment of QPRT.
D'autres caractéristiques et avantages de l'invention apparaissent dans les exemples et figures suivants. Other features and advantages of the invention appear in the following examples and figures.
BREVE DESCRIPTION DES FIGURES BRIEF DESCRIPTION OF THE FIGURES
Figures 1A et 1B : L'échappement à la sénescence induite en réponse à une inflammation chronique permet l'émergence de cellules à risque. Figures 1A and 1B: The escape to senescence induced in response to chronic inflammation allows the emergence of risk cells.
Fig.lA : La soumission de cellules épithéliales de colon à une inflammation chronique les force à s'adapter au stress oxy datif. Des cellules épithéliales de colon humaines immortalisées (HCEC- hTERT) ont été soumises tous les deux jours à des surnageants de macrophages activés (SMA). Les cellules arrêtent rapidement de proliférer (comme démontré par le taux d'histone H3-phosphorylé, pS10-H3), entrent en sénescence comme démontré par la détection d'une activité SA-β- galactosidase, l'accumulation de protéine p53 acétylé (Ac-p53) et l'induction de son gène cible CIP1 (p21). Après un délai d'un mois, les cellules se remettent à proliférer. Fig. 1B : Les cellules émergentes sont résistantes au stress oxydatif et tolèrent des activations mitogéniques. Les cellules ayant échappé à la sénescence (Ech-Inf A et Ech-Inf B issues de deux expériences distinctes) et les cellules HCEC parentales (Par) ont été soit traitées au tert-butyl-hydoxy-peroxyde (TBHP) (panel de gauche) soit transduites par une version activée de la protéine RAS (HRASG12V, panel de droite) et colorées au cristal violet 12 jours plus tard. Le taux de protéine RAS est analysé par Western-blot. Fig.lA: Submission of colon epithelial cells to chronic inflammation forces them to adapt to oxidative stress. Immortalized human colon epithelial cells (HCEC-hTERT) were subjected every two days to activated macrophage supernatants (AMS). Cells rapidly stop proliferating (as demonstrated by H3-phosphorylated histone level, pS10-H3), enter senescence as demonstrated by detection of SA-β-galactosidase activity, accumulation of acetylated p53 protein ( Ac-p53) and the induction of its target gene CIP1 (p21). After a month, the cells begin to proliferate. Fig. 1B: Emergent cells are resistant to oxidative stress and tolerate mitogenic activations. The cells that have escaped senescence (Ech-Inf A and Ech-Inf B from two separate experiments) and the Parental HCEC cells (Par) were either treated with tert-butyl-hydoxy-peroxide (TBHP) (panel left) or transduced by an activated version of the RAS protein (HRASG12V, right panel) and stained with 12-day crystal violet later. The level of RAS protein is analyzed by Western blot.
Figures 2 A et 2B : Les facteurs de transcription ZEB orchestrent le mécanisme d'adaptation au stress oxydatif. Fig. 2A : L'échappement à la sénescence induite en réponse à une inflammation chronique ou un peroxyde chimique résulte de l'induction des gènes embryonnaires ZEB. Analyse de l'expression des gènes SNAI1, TWIST2, ZEB1 et ZEB2 par q-RT-PCR dans les cellules ayant échappé à l'inflammation chronique (panel de gauche) ou au stress oxydatif induit par un peroxyde chimique (panel de droite). L'expression relative est définie en comparaison au gène de ménage HPRTl et aux cellules HCEC parentales. L'expression des gènes SNAI2 et TWIST1 est en dessous du seuil de détection. Fig. 2B : L'expression ectopique de l'un des deux facteurs de transcription ZEB est suffisante pour permettre aux cellules de s'adapter au stress oxydatif. Les cellules HCEC- hTERT ont été transduites par Zebl ou Zeb2, soumises soit à des surnageants de macrophages activés (SMA) ou à du TBHP. Après 12 jours de traitement, les cellules ont été colorées au cristal violet. Figures 2A and 2B: ZEB transcription factors orchestrate the mechanism of adaptation to oxidative stress. Fig. 2A: Escaping to induced senescence in response to chronic inflammation or chemical peroxide results from induction of ZEB embryonic genes. Analysis of SNAI1, TWIST2, ZEB1 and ZEB2 gene expression by q-RT-PCR in cells escaping chronic inflammation (left panel) or oxidative stress induced by a chemical peroxide (right panel). Relative expression is defined in comparison to the HPRT1 household gene and parental HCEC cells. The expression of the SNAI2 and TWIST1 genes is below the detection threshold. Fig. 2B: The ectopic expression of one of the two ZEB transcription factors is sufficient to allow cells to adapt to oxidative stress. HCEC-hTERT cells were transduced by Zebl or Zeb2, submitted to either activated macrophage supernatants (AMS) or TBHP. After 12 days of treatment, the cells were stained with crystal violet.
Figures 3A et 3B : La résistance au stress oxydatif est liée à l'induction d'un programme génique spécifique. Fig. 3A : La comparaison des profils géniques de cellules HCEC-hTERT ayant échappé à l'inflammation chronique (issues de deux expériences indépendantes, Ech-Inf A et B), au stress oxydatif induit par un peroxyde chimique (Ech-TBHP) ou transduites par Zebl (HCEC-ZeW) ou Zeb2 (HCEC-Zeb2) met en exergue un ensemble de 27 gènes communément induits et de 32 gènes communément réprimés (voir Figure 13). Parmi cette liste, les deux gènes les plus induits sont QPRT et APOE. Fig. 3B : L'expression ectopique de l'une ou de l'autre de ces deux protéines est suffisante pour protéger les cellules du stress oxydatif. Les cellules HCEC ont été transduites par QPRT ou APOE et soumises soit à des surnageants de macrophages activés (SMA) soit un traitement au TBHP. Après 12 jours de traitement, les cellules ont été colorées au cristal violet. Figures 4A et 4B : L'activité de la protéine QPRT contribue à la résistance au stress oxydatif. Fig. 4A : La survie des cellules ayant échappé à la sénescence induite en réponse à une inflammation chronique est dépendante de l'expression de QPRT. Panel du haut : les cellules HCEC ayant échappé à la sénescence induite en réponse à une inflammation chronique (Ech-Inf) ont été infectées par des vecteurs lentiviraux exprimant deux shRNA distincts dirigés contre l'ARN QPRT (shRNA QPRT A et B) et soumises à des surnageants de macrophage activés (SMA). Après 12 jours de traitement, les cellules sont colorées au cristal violet. Les cellules HCEC parentales (Par) et Ech-Inf infectées par un shRNA contrôle ont été utilisées respectivement comme contrôles négatif et positif. Panel du bas : analyse de QPRT et de la caspase 3 clivée (cl. casp. 3) par Western blot.  Figures 3A and 3B: The resistance to oxidative stress is related to the induction of a specific gene program. Fig. 3A: Comparison of the HCEC-hTERT gene profiles that escaped chronic inflammation (from two independent experiments, Ech-Inf A and B), chemical oxidative stress induced (Ech-TBHP) or transduced by Zebl (HCEC-ZeW) or Zeb2 (HCEC-Zeb2) highlights a set of 27 commonly induced genes and 32 commonly repressed genes (see Figure 13). Among this list, the two most induced genes are QPRT and APOE. Fig. 3B: The ectopic expression of either of these two proteins is sufficient to protect cells from oxidative stress. HCEC cells were transduced by QPRT or APOE and subjected to either activated macrophage supernatants (AMS) or TBHP treatment. After 12 days of treatment, the cells were stained with crystal violet. FIGS. 4A and 4B: The activity of the QPRT protein contributes to resistance to oxidative stress. Fig. 4A: Survival of cells that have escaped senescence induced in response to chronic inflammation is dependent on QPRT expression. Panel at the top: HCEC cells that escaped senescence induced in response to chronic inflammation (Ech-Inf) were infected with lentiviral vectors expressing two distinct shRNAs directed against QPRT RNA (shRNA QPRT A and B) and subjected activated macrophage supernatants (SMA). After 12 days of treatment, the cells are stained with crystal violet. The parental HCEC (Par) and Ech-Inf cells infected with a control shRNA were used respectively as negative and positive controls. Bottom panel: analysis of QPRT and cleaved caspase 3 (Cl, case 3) by Western blot.
Fig. 4B : L'activité de la protéine QPRT est essentielle pour permettre aux cellules ayant échappé à la sénescence induite en réponse à une inflammation chronique de tolérer des activations oncogéniques. Panel du haut : les cellules HCEC ayant échappé à la sénescence induite en réponse à une inflammation chronique (Ech-Inf) ont été déplétées en QPRT par ARN interférence et infectées par un vecteur rétroviral exprimant la version activée de la protéine HRAS (HRASG12V). 12 jours après infection, les cellules ont été colorées au cristal violet. Panel du bas : analyse de QPRT et RAS par Western blot. Fig. 4B: QPRT protein activity is essential to allow cells that have escaped senescence induced in response to chronic inflammation to tolerate activations oncogenic. Panel at the top: HCEC cells that escaped induced senescence in response to chronic inflammation (Ech-Inf) were disrupted in QPRT by RNA interference and infected with a retroviral vector expressing the activated version of the HRAS protein (HRASG12V). 12 days after infection, the cells were stained with crystal violet. Bottom panel: QPRT and RAS analysis by Western blot.
Figure 5 : Classification des lignées humaines de carcinomes colorectaux en fonction du score du programme ALEP. Les lignées utilisées dans les tests suivants et dans lesquelles le programme est induit sont OUMS-23, C2BBel, CL-14 ; les lignées dans lesquelles le programme n'est pas induit sont SW116, HCT116, HT-29, SW403, COLO-320.  Figure 5: Classification of human lines of colorectal carcinoma based on the ALEP score. The lines used in the following tests and in which the program is induced are OUMS-23, C2BBel, CL-14; the lines in which the program is not induced are SW116, HCT116, HT-29, SW403, COLO-320.
Figure 6 : La prolifération des lignées cancéreuses colorectales dans lesquelles le programme ALEP est induit est dépendante de l'expression de QPRT. Panels du haut: différentes lignées colorectales dans lesquelles le programme alternatif est induit (ALEP actif) ou non (ALEP inactif) ont été infectées par des vecteurs lentiviraux exprimant des shRNA dirigés contre l'ARN QPRT (shRNA QPRT A et B) et colorées au cristal violet douze jours après infection. Panel du bas: analyse de QPRT dans les différentes lignées par Western blot. Figure 6: The proliferation of colorectal cancer lines in which the ALEP program is induced is dependent on the expression of QPRT. Top panels: different colorectal lines in which the alternative program is induced (ALEP active) or not (ALEP inactive) were infected with lentiviral vectors expressing shRNAs directed against QPRT RNA (shRNA QPRT A and B) and stained with purple crystal twelve days after infection. Bottom panel: QPRT analysis in the different lines by Western blot.
Figure 7 : Panels A et B. L'inhibition de la voie de la kynurénine dans des lignées colorectales ALEP -positives OUMS-23 (panel A) ou C2BBel (panel B) s'accompagne d'un arrêt de prolifération, d'une induction d'une autophagie et de dommages à l'ADN. Panels du haut. L'inhibition de l'expression de QPRT par le biais de shRNA (A ou B) induit un arrêt de prolifération des cellules à en juger par des courbes de prolifération (panels en haut à gauche) et l'accumulation d'histone H3 phospho-serine 10 (pS10-H3, panels en haut à droite). L'inhibition de QPRTçw les shRNA ou l'inactivation de l'enzyme TD02 par le composé chimique 680C91 induit de l'autophagie, à en juger par l'accumulation de vacuoles (panels en bas à gauche) et de la protéine LC3 (panels en bas centraux et à droite) ainsi que l'accumulation de dommages à l'ADN à en juger par l'accumulation de la protéine γ-Η2ΑΧ (panels en bas centraux et à droite).  Figure 7: Panels A and B. The inhibition of the kynurenine pathway in ALEP -positive ORMS-23 (panel A) or C2BBel (panel B) colorectal lines is accompanied by a proliferation arrest, a induction of autophagy and DNA damage. Panels from the top. Inhibition of QPRT expression via shRNA (A or B) induces cell proliferation arrest as judged by proliferation curves (panels on the upper left) and histone H3 phospho accumulation Serine 10 (pS10-H3, panels top right). Inhibition of QPRTcw shRNA or inactivation of the enzyme TD02 by chemical compound 680C91 induces autophagy, judging by the accumulation of vacuoles (panels on the lower left) and the LC3 protein (panels central and right) and the accumulation of DNA damage, judged by the accumulation of the γ-Η2ΑΧ protein (central bottom and right panels).
Figure 8 : Analyse de QPRT sur des échantillons humains par immunohistochimie (I). Exemples de marquage de QPRT par immunohistochimie sur des échantillons de colon sain (en haut à gauche), de muqueuses enflammées (en haut à droite), de dysplasies de bas- ou de haut-grade (2nde et 3ieme rangée en partant du haut) ou des carcinomes développés sur un contexte inflammatoires. Echelle : 100 μιη. Figure 8: QPRT analysis on human samples by immunohistochemistry (I). QPRT marking examples by immunohistochemistry on healthy colon samples (upper left) of inflamed mucous membranes (top right), dysplasias of low- or high-grade (2 nd and 3 rd row from the high) or carcinomas developed on an inflammatory context. Scale: 100 μιη.
Figure 9 : Analyse de QPRT sur des échantillons humains par immunohistochimie (II). Exemples de marquage de QPRT par immunohistochimie sur des échantillons d'adénomes faiblement ou fortement dysplasiques et dans des cancers sporadiques. Echelle 100 μιη.  Figure 9: QPRT analysis on human samples by immunohistochemistry (II). Examples of QPRT staining by immunohistochemistry in samples of weakly or strongly dysplastic adenomas and in sporadic cancers. Scale 100 μιη.
Figures 10A et 10B : Le programme ALEP est induit dans les muqueuses enflammées de patients atteints de colites ulcératives ou de maladie de Crohn. La comparaison de la signature du programme ALEP aux profils géniques de patients atteints de maladie de Crohn (CD) ou de colite ulcérative (UC) (séries GSE36807 et GSE37283) permet de ségréger un certain nombre de patients. Notons dans la série de patients atteints de colites ulcératives GSE37283 la capacité de la signature à ségréger ceux ayant ou non développé des lésions colorectales cancéreuses. Figures 10A and 10B: The ALEP program is induced in the inflamed mucosa of patients with ulcerative colitis or Crohn's disease. The comparison of the ALEP program signature with the gene profiles of patients with Crohn's disease (CD) or ulcerative colitis (UC) (series GSE36807 and GSE37283) makes it possible to segregate a certain number of patients. Note in the series of patients with ulcerative colitis GSE37283 the ability of the signature to segregate those with or without cancerous colorectal lesions.
Figures 11A à 11C : Analyse de la signification biologique du programme alternatif sur une large cohorte de patients atteints de cancers colorectaux sporadiques (tumeurs primaires réséquées de patients n'ayant encore subi aucun traitement thérapeutique associé non chirurgical). L'analyse est menée sur une cohorte de 566 carcinomes colorectaux (GSE39582) classifiés selon leur profil génique en six types moléculaires (de Cl à C6, Marisa et al., 2013). Fig. 11A. Analyse du score de la signature du programme alternatif dans les six types moléculaires. Fig. 11B. Analyse de la corrélation entre les scores des signatures du programme alternatif et des cellules souches intestinales Lgr5 et EphB2. Fig. 11C. Représentation de Kaplan-Meier de la survie sans récidive de patients atteints de cancers colorectaux selon le score du programme ALEP, élevé, intermédiaire ou faible (avec des seuils pris à 4000 et 6000 en combinant les scores de chacun des gènes surexprimés ou sous-exprimés voire éteints). Figures 11A to 11C: Analysis of the biological significance of the alternative program on a large cohort of patients with sporadic colorectal cancers (primary resected tumors of patients who have not yet undergone any non-surgical associated therapeutic treatment). The analysis is conducted on a cohort of 566 colorectal carcinomas (GSE39582) classified according to their gene profile into six molecular types (from C1 to C6, Marisa et al., 2013). Fig. 11A. Analysis of the signature score of the alternative program in the six molecular types. Fig. 11B. Analysis of the correlation between the scores of the alternative program signatures and intestinal stem cells Lgr5 and EphB2. Fig. 11C. Kaplan-Meier representation of the recurrence-free survival of colorectal cancer patients by high, intermediate, or low ALEP score (with thresholds taken at 4000 and 6000 by combining the scores of each of the overexpressed or under-expressed genes even extinct).
Figure 12 : Analyse de QPRT par immunohistochimie dans les adénocarcinomes œsophagiens. Exemples d' adénocarcinomes œsophagiens exprimant (QPRT+) ou non (QPRT-) la protéine QPRT. L'épithélium malpighien du tiers inférieur de l'œsophage est utilisé comme contrôle interne (panel en bas à droite). Echelle 100 μιη.  Figure 12: QPRT analysis by immunohistochemistry in oesophageal adenocarcinoma. Examples of oesophageal adenocarcinoma expressing (QPRT +) or non - QPRT - protein QPRT. The squamous epithelium of the lower third of the esophagus is used as an internal control (panel at the bottom right). Scale 100 μιη.
Figure 13 : Liste des gènes communément induits ou réprimés dans les cellules HCEC ayant échappé à la sénescence induite en réponse à une inflammation chronique (Ech-Inf A et B) à un peroxyde chimique (Ech-TBHP A et B) ou transduites par Zebl ou Zeb2.  Figure 13: List of genes commonly induced or repressed in HCEC cells that have escaped senescence induced in response to chronic inflammation (Ech-Inf A and B) to a chemical peroxide (Ech-TBHP A and B) or transduced by Zebl or Zeb2.
EXEMPLES EXAMPLES
Exemple 1 : Matériel et méthodes  Example 1: Material and methods
Plasmides et lignées : Les constructions HRASG12V, ZEB1 et ZEB2 pBabe Puro ont été décrites antérieurement (Morel et al., 2012). Les vecteurs d'expression ΑΡΟΕε3 et QPRT en pbabe Puro ont été générés à partir des vecteurs pLenti-GIII-hAPOE-GFP-2A-Puro constructs (abm) et QPRT pCMV6 (Origene). Le cDNA QPRT RK138/139 a été synthétisé par la société Genscript et sous- cloné dans le vecteur rétro viral pBabe Puro (Addgene). Les shRNA QPRT A (5'- TCGCTCTGAAGGTGGAGT-3' ; SEQ ID NO : 5) and shRNA QPRT B (5'- AGTCCTAAACCGGAAGAGG-3'; SEQ ID NO : 6) en pLKO.l ont été obtenus auprès de la société Sigma. Les lignées HT-29, HCT116, SW403, COLO320HSR and C2BBel ont été obtenues auprès de l'ATCC. Les lignées OUMS23 et CL- 14 ont été fournies par la banque cellulaire JCRB (Japon) et le DSMZ GmbH (Allemagne), respectivement. Les lignées OUMS23 and c2BBel ont été cultivées en milieu DMEM (Life Technologies) complémenté par 10% SVF (Cambrex) et 100 U/ml Penicillin/Streptomycin (PS, Life Technologies), les lignées HT-29 et HCT116 en milieu McCoy's 5A (Life Technologies) complémenté par 10% SVF + 100 U/ml PS, les lignées SW116, SW403 and COLO320 en milieu RPMI (Life Technologies) complémenté avec 10% FBS + 100 U/ml PS, et la lignée CL- 14 en milieu DMEM/HAMF 12 1:1 (Invitrogen) complémenté avec 20% FBS et 100 U/ml PS. Les cellules primaires épithéliales de colon ont été obtenues auprès de la société abm (T4056), immortalisées par transduction à'hTERT et cultivées in milieu Prigrow III (TM003, abm) complémenté 5% FBS and 100 U/ml PS sur des boites de collagène (BD BioCoat Collagen type I, Dutscher). Plasmids and Lineages: The HRAS G12V , ZEB1 and ZEB2 pBabe Puro constructs have been previously described (Morel et al., 2012). The expression vectors ΑΡΟΕε3 and QPRT in Pbabe Puro were generated from the vectors pLenti-GIII-hAPOE-GFP-2A-Puro constructs (abm) and QPRT pCMV6 (Origene). The QPRT RK138 / 139 cDNA was synthesized by the Genscript company and subcloned into the Puro pBabe retro viral vector (Addgene). QPRT A shRNAs (5'-TCGCTCTGAAGGTGGAGT-3 ', SEQ ID NO: 5) and shRNA QPRT B (5'-AGTCCTAAACCGGAAGAGG-3', SEQ ID NO: 6) in pLKO.I were obtained from Sigma . HT-29, HCT116, SW403, COLO320HSR and C2BBel were obtained from ATCC. The lines OUMS23 and CL-14 were provided by the JCRB cellular bank (Japan) and the DSMZ GmbH (Germany), respectively. The lines OUMS23 and c2BBel were cultured in DMEM medium (Life Technologies) supplemented with 10% FCS (Cambrex) and 100 U / ml Penicillin / Streptomycin (PS, Life Technologies), lines HT-29 and HCT116 in McCoy's 5A medium ( Life Technologies) supplemented with 10% FCS + 100 U / ml PS, lines SW116, SW403 and COLO320 in RPMI medium (Life Technologies) supplemented with 10% FBS + 100 U / ml PS, and the line CL-14 in DMEM / HAMF 12 1: 1 medium (Invitrogen) supplemented with 20% FBS and 100 U / ml PS. The primary epithelial colon cells were obtained from the abm company (T4056), immortalized by transduction at HTERT and cultured in Prigrow III medium (TM003, abm) supplemented with 5% FBS and 100 U / ml PS on collagen plates. (BD BioCoat Collagen Type I, Dutscher).
Induction d'un stress oxydatif dans les cellules HCEC : La lignée monocytaire THP-1 a été obtenue auprès de l'ATCC et cultivée en milieu RPMI (Life Technologies) complémenté avec 10% FBS, 100 U/ml PS, 1 mM Sodium pyruvate (Life Technologies) and des acides aminés non- essentiels (Life Technologies). La différentiation cellulaire (5.107 cellules) a été induite par un traitement au PMA (0.162 μΜ, Sigma) pendant une journée et l'activation des macrophages ainsi obtenus ensuite induite par du LPS (LPS-EB ultra-pure de la lignée E.coli 0111:B4, InvivoGen) pendant une journée. Le surnageant a été collecté, filtré sur 0.45 μΜ, dilué au demi dans le milieu de culture des HCEC et placé sur les cellules HCEC (5 xlO5 en boite 6 puits). Le surnageant des macrophages activés a été renouvelé tous les deux jours. De manière similaire, le traitement des cellules HCEC avec du tert-Butyl-hydroperoxide (30 μΜ, Sigma) a été renouvelé tous les deux jours. Les cellules ont été colorées au cristal violet après dix jours de traitement. Les tests SA-β- galactosidase ont été effectués comme préalablement décrits (Dimri et al., 1995). Induction of oxidative stress in HCEC cells: The monocyte line THP-1 was obtained from ATCC and cultured in RPMI medium (Life Technologies) supplemented with 10% FBS, 100 U / ml PS, 1 mM Sodium pyruvate (Life Technologies) and non-essential amino acids (Life Technologies). Cell differentiation (5.10 7 cells) was induced by a treatment with PMA (0.162 μΜ, Sigma) for one day and the activation of the macrophages thus obtained then induced by LPS (ultra-pure LPS-EB of the E line. coli 0111: B4, InvivoGen) for one day. The supernatant was collected, filtered over 0.45 μl, diluted half in the HCEC culture medium and placed on the HCEC cells (5 × 10 5 in a 6-well dish). The supernatant of activated macrophages was renewed every other day. Similarly, the treatment of HCEC cells with tert-Butyl hydroperoxide (30 μl, Sigma) was repeated every other day. The cells were stained with crystal violet after ten days of treatment. SA-β-galactosidase assays were performed as previously described (Dimri et al., 1995).
Infections rétrovirales and lentivirales : La production ectopique de protéines dans les cellules HCEC-hTERT a été effectuée par le biais d'infections rétrovirales comme préalablement décrit (Gras et al., 2014). Brièvement, les cellules sont « murinisées » par l'expression ectopique d'un récepteur écotrope (Baker et al., 1992; Boehm et al., 2005) avant d'être infectées par les vecteurs d'expression rétroviraux. Les infections séquentielles sont espacées de 48h et la sélection est initiée 24h après la seconde infection avec de la puromycine (1.5 μg/ml). Les particules lentivirales des shRNA QPRT ont été générées par transfection de la lignée HEK293T avec les vecteurs pLKO.l, pCMV AR8.91 (gag-pop-Tat-Rev) (Zufferey et al., 1997) et phCMVG-VSVG (env) (Burns et al., 1993) par la technique de calcium phosphate. La déplétion en QPRT a été obtenue par trois infections successives, chacune séparée de 12h. Nous avons confirmé l'infection de plus de 90%> des cellules. Retroviral and lentiviral infections: Ectopic production of proteins in HCEC-hTERT cells was performed through retroviral infections as previously described (Gras et al., 2014). Briefly, the cells are "murine" by the ectopic expression of an ecotropic receptor (Baker et al., 1992; Boehm et al., 2005) before being infected with the retroviral expression vectors. Sequential infections are spaced 48 hours apart and selection is initiated 24 hours after the second infection with puromycin (1.5 μg / ml). The lentiviral particles of the QPRT shRNAs were generated by transfection of the HEK293T line with the pLKO.l vectors, pCMV AR8.91 (gag-pop-Tat-Rev) (Zufferey et al., 1997) and phCMVG-VSVG (env). (Burns et al., 1993) by the calcium phosphate technique. Depletion in QPRT was obtained by three successive infections, each separated by 12h. We confirmed the infection of more than 90%> cells.
Analyse des protéines par Western-bot : Les extraits cellulaires ont été effectués en milieu RIPA (100 mM NaCl, 1% NP40, 0.1% SDS, 50 mM Tris pH8) complémenté par des cocktails d'inhibiteurs de protéase (Roche) et de phosphatases (Sigma) et les débris cellulaires éliminés par centrifugation. Les protéines ont été analysées par Western-blot en utilisant les anticorps monoclonaux anti-TWISTl 2Cla (Abcam), anti-p21CIPl/WAFl clone SX118 (Dako) et anti- QPRT (ab57125, Abcam), et les anticorps polyclonaux anti-H-RAS N-18 (Santa-Cruz), anti-ZEBl H102 (Santa-Cruz), anti-ZEB2 (Sayan et al., 2009), p-histone H3 (SerlO)-R (Santa-Cruz), and phospho-histone H2A.X Serl39 (Cell Signaling) et des anticorps secondaires couplés à la peroxidase (Dako). Les complexes antigène-anticorps ont été révélés par le réactif Luminol (Santa Cruz). Immunohistochimie : Les échantillons de patients ont été obtenus auprès du Centre de Ressources Biologiques de l'Hôpital Lyon Est et Saint-Antoine (Paris) avec l'accord des comités éthiques locaux. Les échantillons ont été utilisés avec l'accord écrit informé des patients. Cette étude a été approuvée par les comités éthiques des deux institutions. Les marquages ont été opérés sur des sections de 40 μιη de tissus fixés en formol et incubés dans de la paraffine. Les lames ont été successivement déparaffinées par trois bains successifs de xylène et réhydratées par des bains en éthanol pur, à 90% et 80%. L'antigène a ensuite été démasqué en chauffant les lames au microonde (5 et 10 min) dans une solution de démasquage à PH6 (Dako). Après avoir bloqué l'activité peroxydase endogène par un bain d'eau oxygénée à 0.3% pendant 10 min, les lames ont été incubées en présence d'anticorps QPRT (anticorps Atlas, HPA011887, Sigma) à température ambiante pendant lh à une dilution au l/100ème. Le marquage est ensuite révélé en utilisant le Polymer Kit Novolink (Leica). Western-bot protein analysis: The cell extracts were carried out in RIPA medium (100 mM NaCl, 1% NP40, 0.1% SDS, 50 mM Tris pH8) supplemented with protease inhibitor (Roche) and phosphatase cocktails. (Sigma) and cell debris removed by centrifugation. The proteins were analyzed by Western blot using anti-TWIST1 2Cla monoclonal antibodies (Abcam), anti-p21CIP1 / WAF1 clone SX118 (Dako) and anti-QPRT (ab57125, Abcam), and polyclonal anti-H-antibodies. RAS N-18 (Santa Cruz), anti-ZEB1 H102 (Santa Cruz), anti-ZEB2 (Sayan et al., 2009), p-histone H3 (Ser10) -R (Santa Cruz), and histone H2A.X Ser139 (Cell Signaling) and secondary antibodies coupled to peroxidase (Dako). The antigen-antibody complexes were revealed by the Luminol reagent (Santa Cruz). Immunohistochemistry: Patient samples were obtained from the Biological Resources Center of Lyon Est and Saint-Antoine Hospital (Paris) with the agreement of local ethics committees. The samples were used with the informed written agreement of the patients. This study has been approved by the ethical committees of both institutions. The markings were performed on sections of 40 μιη of formalin fixed tissues and incubated in paraffin. The slides were successively deparaffinized by three successive baths of xylene and rehydrated with baths of pure ethanol, 90% and 80%. The antigen was then unmasked by heating the slides in the microwave (5 and 10 min) in a PH6 (Dako) unmasking solution. After blocking the endogenous peroxidase activity by a 0.3% hydrogen peroxide bath for 10 min, the slides were incubated in the presence of QPRT antibodies (Atlas antibody, HPA011887, Sigma) at room temperature for 1 h at a dilution at l / 100th. The labeling is then revealed using the Novolink Polymer Kit (Leica).
Analyse de l'expression transcriptionnelle des gènes par q-RT-PCR : La préparation des ADN et la transcription reverse ont été effectuées comme précédemment décrit (Gras et al., 2014). Les amorces de PCR ont été sélectionnées par le biais du logiciel primer3. Le gène de ménage HPRT1 a été utilisé pour la normalisation. Les couples d'amorces suivants ont été sélectionnés:  Analysis of the transcriptional expression of the genes by q-RT-PCR: The preparation of the DNAs and the reverse transcription were performed as previously described (Gras et al., 2014). The PCR primers were selected using the primer3 software. The household gene HPRT1 was used for normalization. The following primer pairs have been selected:
Figure imgf000017_0001
Figure imgf000017_0001
Tableau 1 Profils d'expression génique : Les profils d'expression génique et l'analyse des données ont été effectués par le biais de la plateforme ProfileXpert (Lyon, France). Les profils d'expression ont été analysés sur la base de puces contenant 47231 sondes (HumanHT-12 v4 Expression BeadChip;Illumina Inc., USA). L'ARN total (500 ng) a été amplifié et marqué à la biotine en utilisant le kit Illumina TotalPrepTM RNA Amplification Kit (Ambion Inc., USA). L'hybridation a été réalisée avec 750 ng de cRNA marqué à la biotine. Les puces ont été scannées en utilisant le protocole standard d'Illumina et le scanner theiScan (Illumina Inc., USA) et les données ont été normalisées en utilisant le logiciel Génome Studio 2010 (Illumina Inc.,USA). Table 1 Gene Expression Profiles: Gene expression profiles and data analysis were performed through the ProfileXpert platform (Lyon, France). The expression profiles were analyzed on the basis of chips containing 47231 probes (HumanHT-12 v4 Expression BeadChip, Illumina Inc., USA). Total RNA (500 ng) was amplified and labeled with biotin using the Illumina TotalPrep ™ RNA Amplification Kit kit (Ambion Inc., USA). Hybridization was performed with 750 ng of biotin labeled cRNA. The chips were scanned using Illumina's standard protocol and theiScan scanner (Illumina Inc., USA) and the data was standardized using Genome Studio 2010 software (Illumina Inc., USA).
Analyses in silico de la pertinence du programme alternatif : L'analyse des données a été effectuée par le biais du logiciel Array Studio (Omicsoft Corporation). Les données brutes des séries analysées (CEL) ont été traitées en utilisant la normalisation quantile et l'algorithme Robust multi-array average (PMA et transformées en Log2 (Irizarry et al., 2003). Différents outils informatiques dont le « Hierarchical clustering analysis », le « Principal Component Analysis (PCA) et la méthodologie de projection ssGSEA ont été utilisés. Le score de la signature a été déterminé en utilisant les « Empirical cumulative Distribution Functions » (ECDF) des gènes et les corrélations entre les signatures déterminées par une corrélation de Pearson. In silico analyzes of the relevance of the alternative program: The analysis of the data was carried out through the software Array Studio (Omicsoft Corporation). The raw data from the analyzed series (CEL) were processed using quantile normalization and the Robust multi-array average algorithm (PMA and transformed into Log2 (Irizarry et al., 2003). Various computer tools including the "Hierarchical clustering analysis The Principal Component Analysis (PCA) and ssGSEA projection methodology were used.The signature score was determined using the "Empirical cumulative Distribution Functions" (ECDF) genes and correlations between signatures determined by a Pearson correlation.
Exemple 2 : L'échappement à îa sénescence ndui e en réponse à une inflammation chronique conduit à l'émergence de cellules à risque et résulte d'une reprogrammation des cellules, orchestrée par le facteur de transcription ZEB1. Example 2: The escape of senescence in response to chronic inflammation leads to the emergence of risk cells and results from reprogramming of the cells, orchestrated by the transcription factor ZEB1.
Le modèle établi in vitro repose sur l'utilisation de cellules épithéliales de colon immortalisées (HCEC-hTERT) cultivées en présence de surnageant de macrophages activés (SMA, conditions mimant une inflammation chronique). Ces conditions expérimentales conduisent à l'induction de dommages à l'ADN simple brin qui évoluent en dommages double brin (confirmée par l'accumulation de protéine γΗ2ΑΧ). Ces dommages induisent un arrêt de la prolifération cellulaire (démontré par la diminution du taux d'histone H3 phosphorylée sur le résidu SerlO, p-S10-H3) et l'engagement des cellules épithéliales dans un programme de sénescence (mis en évidence par l'acétylation de la protéine p53 (Ac-p53), l'induction de son gène cible CIP1 (p21) et la détection d'une activité SA- -galactosidase): une séquence d'événements récapitulant les observations faites in vivo. Pour autant, après un délai d'un mois, les cellules se remettent invariablement à proliférer (Figure 1A). Les cellules émergentes (notées ensuite « Ech-Inf » pour « échappement à l'inflammation ») sont non seulement capables de proliférer dans des conditions d'inflammation chronique mais également de supporter des stress oxydatifs qu'ils soient induits en réponse à un peroxyde chimique (tel le tert-butyl-hydroxy-peroxyde ou TBHP) ou en réponse à une activation oncogénique (transduction d'une version activée de la protéine RAS, H-RASG12V) (Figure 1B). Les cellules émergentes ont donc acquis une résistance intrinsèque aux stress oxydatifs et en tolérant des activations oncogéniques sont susceptibles d'évoluer vers un phénotype malin. La soumission des cellules à un peroxyde chimique (TBHP) engendre de la même manière un arrêt de prolifération des cellules. Après trois semaines, des cellules de nouveau se remettent à proliférer (notées ensuite Ech-TBHP). The model established in vitro is based on the use of immortalized colon epithelial cells (HCEC-hTERT) cultured in the presence of activated macrophage supernatant (SMA, conditions mimicking chronic inflammation). These experimental conditions lead to the induction of single-stranded DNA damage that evolves into double-stranded damage (confirmed by γΗ2ΑΧ protein accumulation). These damages induce a stop of the cellular proliferation (demonstrated by the reduction of the phosphorylated H3 histone level on the residue Ser10, p-S10-H3) and the engagement of the epithelial cells in a program of senescence (evidenced by the acetylation of the p53 protein (Ac-p53), induction of its CIP1 target gene (p21) and detection of an SA-galactosidase activity): a sequence of events summarizing the observations made in vivo. However, after a period of one month, the cells invariably begin to proliferate (Figure 1A). Emergent cells (later referred to as "Ech-Inf" for "inflammation escape") are not only able to proliferate under conditions of chronic inflammation but also to withstand oxidative stress that is induced in response to a peroxide chemical (such as tert-butyl-hydroxy-peroxide or TBHP) or in response to oncogenic activation (transduction of an activated version of the RAS protein, H-RAS G12V ) (Figure 1B). Emerging cells have therefore acquired intrinsic resistance to oxidative stress and tolerant oncogenic activation is likely to evolve into a malignant phenotype. Submission of cells to a chemical peroxide (TBHP) similarly generates a halt to proliferation of cells. After three weeks, cells again begin to proliferate (noted after Ech-TBHP).
Le statut du gène TP53 est sauvage dans les cellules 'Ech-Inf et la voie de signalisation reste inductible en réponse à un stress génotoxique. L'analyse de l'expression des membres des trois principales familles Snail, Twist et Zeb a été analysée. Invariablement, sur trois expériences indépendantes, l'échappement au stress oxydatif induit par l'inflammation ou par le peroxyde chimique est associé à une induction du gène ZEB1 ou ZEB2 (Figure 2A), suggérant que les facteurs de transcription ZEB orchestrent le mécanisme d'adaptation au stress oxydatif. En accord avec cette hypothèse, la production ectopique de la protéine murine ZEB 1 ou de la protéine murine apparentée ZEB2 (lignées HCEC-ZeW et HCEC-Zeb2) octroie aux cellules HCEC un avantage prolifératif dans des conditions de stress oxydatif (induit soit par une inflammation chronique ou par un traitement au TBHP) (Figure 2B).  The status of the TP53 gene is wild in the Ech-Inf cells and the signaling pathway remains inducible in response to genotoxic stress. The analysis of the expression of the members of the three main families Snail, Twist and Zeb was analyzed. Invariably, on three independent experiments, the oxidative stress escape induced by inflammation or chemical peroxide is associated with an induction of the ZEB1 or ZEB2 gene (Figure 2A), suggesting that ZEB transcription factors orchestrate the mechanism of adaptation to oxidative stress. Consistent with this hypothesis, ectopic production of murine ZEB 1 protein or the related murine ZEB2 protein (HCEC-ZeW and HCEC-Zeb2 lines) gives HCEC cells a proliferative advantage under oxidative stress conditions (induced either by chronic inflammation or TBHP treatment) (Figure 2B).
Exemple 3. Les protéines ZEB1 et ZEB2 induisent un programme génique spécifique accordant aux cellules une résistance au stress oxydatif. Example 3 The ZEB1 and ZEB2 proteins induce a specific gene program that gives the cells resistance to oxidative stress.
La comparaison des profils d'expression génique des lignées Ech-Inf, Ech-TBHP, HCEC-Zebl et HCEC-Zeb2 à la lignée parentale (HCEC-Par) a permis de mettre en évidence l'activation d'un programme génique commun constitué de l'activation d'une trentaine de gènes et de la répression d'une trentaine d'autres (Figure 3A, gènes activés listés dans la Figure 13). Ce programme appelé ALEP (pour Alternative to EMT Program, alternatif au programme d'EMT communément induit par ces facteurs de transcription) n'inclut aucun des gènes de détoxification connu. L'implication dans l'octroi de cet avantage de survie des deux gènes les plus induits de ce programme, à savoir QPRT et APOE, a été testée. L'expression ectopique de l'un ou de l'autre, avec une efficacité supérieure pour QPRT, confère effectivement un avantage de survie aux cellules dans des conditions d'inflammation chronique ou lorsqu'elles sont soumises à un peroxyde chimique (Figure 3B). Cette observation confirme que plusieurs composantes de ce programme contribuent à accorder un avantage de survie et/ou de prolifération à des cellules épithéliales de colon dans ces conditions de stress. Exemple 4 : L'activation du programme alternatif se traduit par une dépendance des cellules à la protéine QPRT pour leur prolifération ou survie.  The comparison of the gene expression profiles of the Ech-Inf, Ech-TBHP, HCEC-Zebl and HCEC-Zeb2 lines to the parental line (HCEC-Par) made it possible to highlight the activation of a common gene program constituted by the activation of about 30 genes and the repression of about thirty others (Figure 3A, activated genes listed in Figure 13). This program called ALEP (Alternative to EMT Program, an alternative to the EMT program commonly induced by these transcription factors) does not include any of the known detoxification genes. The implication in granting this survival advantage of the two most induced genes of this program, namely QPRT and APOE, was tested. The ectopic expression of either, with superior QPRT efficiency, effectively confers survival benefit to cells under conditions of chronic inflammation or when subjected to chemical peroxide (Figure 3B). . This observation confirms that several components of this program contribute to providing survival and / or proliferation benefit to colon epithelial cells under these stress conditions. Example 4 Activation of the alternative program results in a dependence of the cells on the QPRT protein for their proliferation or survival.
La protéine QPRT (pour quinolinate phospho-ribosyl-transférase) est une enzyme à l'origine décrite comme étant impliquée dans la voie de la kynurénine (Liu et al., 2007). Si sa contribution à la production de novo de NAD+ est effective dans le foie et le rein, la protéine QPRT est également présente dans d'autres types cellulaires, assurant vraisemblablement d'autres fonctions, dont l'inhibition de l'activation de la caspase 3 comme récemment décrit dans des cellules HeLa (Ishidoh et al., 2010). La déplétion des cellules HCEC Echap en QPRT induit la mort des cellules par apoptose dans des conditions d'inflammation chronique (Figure 4A). De manière intéressante, l'inhibition de l'expression de QPRT empêche également ces cellules de tolérer des activations oncogéniques (Figure 4B). La comparaison du programme alternatif aux profils d'expression des 55 lignées cancéreuses colorectales de la banque du Cancer Cell Line Encyclopedia (CCLE) (données GSE36133) (Barretina et al., 2012) met en évidence dans un certain nombre de lignées une expression conjointe de plusieurs des gènes induits du programme ALEP (pas d'expression conjointe des gènes réprimés). L'ensemble des gènes activés du programme ALEP a été utilisé comme signature, les lignées ont été classées selon leur score et leur addiction à QPRT a été évaluée (Figure 5). Sur l'ensemble des lignées analysées, seules les cellules ayant un programme alternatif activé (score de la signature élevé) semblent être dépendantes de la protéine QPRT pour leur prolifération, à en juger par une coloration au cristal violet (Figure 6) et par l'analyse de courbes de prolifération (résultats non montrés). L'ensemble de ces données démontre la pertinence biologique de la signature ALEP et souligne son intérêt en termes prédictifs d'une dépendance à QPRT. QPRT protein (for quinolinate phosphoribosyl transferase) is an enzyme originally described as being involved in the kynurenine pathway (Liu et al., 2007). Although its contribution to the de novo production of NAD + is effective in the liver and kidney, the QPRT protein is also present in other cell types, presumably providing other functions, including inhibition of the activation of NAD +. caspase 3 as recently described in HeLa cells (Ishidoh et al., 2010). Depletion of HCEC Esc cells into QPRT induces apoptotic cell death under conditions of chronic inflammation (Figure 4A). Interestingly, inhibition of QPRT expression also prevents these cells from tolerating oncogenic activations (Figure 4B). The comparison of the alternative program with the expression profiles of the 55 colorectal cancer lines of the Cancer Cell Line Encyclopedia (CCLE) database (data GSE36133) (Barretina et al., 2012) highlights in a certain number of lines a joint expression several of the genes induced by the ALEP program (no joint expression of the repressed genes). The set of activated genes from the ALEP program was used as a signature, the lines were ranked according to their score and their QPRT addiction was evaluated (Figure 5). Of all the lines analyzed, only the cells having an activated alternative program (high signature score) seem to be dependent on the QPRT protein for their proliferation, judging by a crystal violet coloration (FIG. proliferation curve analysis (results not shown). All of this data demonstrates the biological relevance of the ALEP signature and highlights its interest in predicting QPRT dependence.
Le score de la signature ALEP est parallèle au taux de protéine QPRT détectée en Western-blot (Figure 6). Pour autant, bien que le programme ne soit pas actif dans la lignée COLO-320, la protéine QPRT y est détectable, à un taux certes plus faible. En immunohistochimie (IHC), le signal détecté dans cette lignée est suffisamment élevé pour être considéré comme faiblement positif.  The ALEP signature score is parallel to the level of QPRT protein detected in Western blot (Figure 6). However, although the program is not active in the COLO-320 line, the QPRT protein is detectable at a lower rate. In immunohistochemistry (IHC), the signal detected in this line is sufficiently high to be considered as weakly positive.
L'inhibition de la protéine QPRT induit un arrêt de prolifération des lignées colorectales ALEP- positives à en juger par l'accumulation d'histone H3 phosphorylée sur le résidu SerlO. Cet arrêt s'accompagne d'une augmentation de l'autophagie (visualisée par l'accumulation de protéine LC3) et de l'accumulation de dommages à l'ADN (visualisée par l'accumulation de γΗ2ΑΧ). L'inactivation de l'enzyme TD02 située en amont de la protéine QPRT dans la voie de la kynurénine a des conséquences en tout point similaires (Figure 7). Les cellules colorectales ALEP- positives sont donc dépendantes de l'activité de la voie de la kynurénine pour maintenir leur prolifération.  Inhibition of the QPRT protein induces proliferation arrest of the ALEP-positive colorectal lines as judged by the accumulation of phosphorylated H3 histone on the Ser10 residue. This arrest is accompanied by an increase in autophagy (visualized by the accumulation of LC3 protein) and the accumulation of damage to DNA (visualized by the accumulation of γΗ2ΑΧ). Inactivation of the TD02 enzyme upstream of the QPRT protein in the kynurenine pathway has similar consequences (Figure 7). ALEP-positive colorectal cells are therefore dependent on the activity of the kynurenine pathway to maintain their proliferation.
Exemple 5 : L'expression de QPRT est induite dans les phases précoces du développement de cancers colorectaux, développés sur un contexte inflammatoire ou non. EXAMPLE 5 The expression of QPRT is induced in the early phases of the development of colorectal cancers, developed on an inflammatory context or not.
L'analyse de l'expression de QPRT par IHC sur une cohorte de patients atteints de maladies chroniques de l'intestin montre une absence de la protéine dans les entérocytes du tissu normal (à l'inverse des fibroblastes de la lamina propria et des cellules immunes infiltrées qui expriment fortement la protéine), une induction de son expression limitée à quelques cellules épithéliales situées au fond des cryptes dans les muqueuses enflammées (lieu où sont localisées les cellules souches intestinales) et une augmentation du nombre de cellules épithéliales marquées dans les dysplasies de bas-grade (45.4%, n=22), de haut-grade (18.2%, n=22) et de cancers (38.1%>, n=21) développés sur un contexte inflammatoire (Figure 8). De même, l'expression de QPRT est induite dans les cellules épithéliales des adénomes (faiblement ou fortement dysplasiques, 50%, n=20) et des cancers sporadiques (30%, n=10) (Figure 9). The analysis of IHC QPRT expression in a cohort of patients with chronic bowel disease shows an absence of the protein in the enterocytes of normal tissue (unlike lamina propria and cell fibroblasts). infiltrated immune cells that strongly express the protein), an induction of its expression limited to a few epithelial cells located at the bottom of crypts in inflamed mucosa (a place where intestinal stem cells are located) and an increase in the number of labeled epithelial cells in dysplasias low-grade (45.4%, n = 22), high-grade (18.2%, n = 22) and cancers (38.1%>, n = 21) developed in an inflammatory context (Figure 8). Similarly, the expression of QPRT is induced in epithelial cells adenomas (weakly or strongly dysplastic, 50%, n = 20) and sporadic cancers (30%, n = 10) (Figure 9).
Exemple 6 : Le programme ALEP est induit dans les cellules souches intestinales et constitue un facteur de mauvais pronostic. Example 6: The ALEP program is induced in intestinal stem cells and is a factor of poor prognosis.
La signature génique du programme ALEP a été établie sur un modèle de cellules épithéliales. Pour affiner cette signature de telle sorte à pouvoir l'utiliser sur des tumeurs (en tenant compte de leur hétérogénéité cellulaire), elle a été comparée à deux cohortes de patients atteints de colites ulcératives (UC) ou de maladie de Crohn (CD) depuis plus de sept ans (datasets GSE36807 et GSE37283, (Montero-Melendez et al., 2013; Pekow et al., 2013)). Ces deux séries ont été sélectionnées du fait que les profils géniques aient été établis sur des muqueuses macroscopiquement non- enflammées de telle sorte à limiter l'impact direct de l'inflammation sur les niveaux de transcrits. L'analyse du score de cette signature permet dans les deux cohortes de patients de ségréger clairement deux sous-groupes (Figure 10 A-B). Elle permet en particulier dans la seconde série de patients de distinguer à partir des lésions non-néoplasiques ceux ayant développé des dysplasies ou des carcinomes colorectaux (Figure 10 B). Le score de la signature du programme ALEP reflète donc l'avancée de la maladie. Les analyses de QPRT par IHC révèlent cependant un certain nombre de patients présentant des dysplasies de bas-grade positives et de haut-grade négatives. La détection de QPRT ne semble donc pas constituer un facteur de mauvais pronostic en soi.  The gene signature of the ALEP program was established on an epithelial cell model. To refine this signature so that it can be used on tumors (taking into account their cellular heterogeneity), it has been compared to two cohorts of patients suffering from ulcerative colitis (UC) or Crohn's disease (CD) since over seven years (datasets GSE36807 and GSE37283, (Montero-Melendez et al., 2013, Pekow et al., 2013)). These two series were selected because the gene profiles were established on macroscopically non-inflamed mucosa so as to limit the direct impact of inflammation on transcript levels. The analysis of the score of this signature allows in both cohorts of patients to clearly segregate two subgroups (Figure 10 A-B). In particular, in the second series of patients, it makes it possible to distinguish, from non-neoplastic lesions, those who have developed dysplasias or colorectal carcinomas (FIG. 10B). The signature score of the ALEP program thus reflects the progress of the disease. QPRT analyzes by IHC, however, reveal a number of patients with positive low-grade dysplasias and high-grade negative dysplasias. The detection of QPRT does not seem to be a factor of poor prognosis in itself.
La distribution de la signature ALEP a été analysée dans une troisième cohorte de patients (n=466) ayant développé exclusivement des carcinomes colorectaux sur un contexte non-inflammatoire (données GSE39582) (Marisa et al., 2013). Ces tumeurs ont récemment été classées sur la base de leur profil génique en 6 catégories (de Cl à C6). L'analyse de la signature ALEP met en exergue une activation du programme dans un certain nombre de tumeurs (18%>, n=566), indépendamment de leur sous-type moléculaire (Figure 11 A). On peut noter une proportion plus forte de tumeurs positives dans le sous-groupe C4, décrit pour être enrichi en cellules souches (Marisa et al., 2013). Dans cette cohorte, le score de la signature ALEP et celui des signatures de cellules souches intestinales Lgr5 et EphB2 (Merlos-Suarez et al., 2011) corrèlent significativement, confortant l'hypothèse selon laquelle le programme ALEP est induit dans les cellules souches/précurseurs intestinales (Figure 11 B). Comme préalablement démontré pour les signatures EphB2 et Lgr5 (Merlos-Suarez et al., 2011), et démontré pour le sous-groupe C4 (Marisa et al., 2013), la signature ALEP s'avère être prédictive d'un risque de récidive élevé (Figure 11 C).  The distribution of the ALEP signature was analyzed in a third cohort of patients (n = 466) who exclusively developed colorectal carcinomas in a non-inflammatory setting (data GSE39582) (Marisa et al., 2013). These tumors have recently been classified on the basis of their gene profile into 6 categories (from C1 to C6). The analysis of the ALEP signature highlights activation of the program in a number of tumors (18%>, n = 566), regardless of their molecular subtype (Figure 11A). There is a higher proportion of positive tumors in the C4 subgroup, described to be enriched in stem cells (Marisa et al., 2013). In this cohort, the ALEP signature score and that of the Lgr5 and EphB2 intestinal stem cell signatures (Merlos-Suarez et al., 2011) correlate significantly, supporting the hypothesis that ALEP is induced in stem cells. intestinal precursors (Figure 11B). As previously demonstrated for the signatures EphB2 and Lgr5 (Merlos-Suarez et al., 2011), and demonstrated for the C4 subgroup (Marisa et al., 2013), the ALEP signature proves to be predictive of a risk of high recurrence (Figure 11 C).
En conclusion cette analyse met en exergue un groupe de patients distincts, avec un risque de récidive élevé caractérisée par des scores de signatures souches intestinales et de programme ALEP élevé. Exemple 7 : Extension des observations à un deuxième modèle de tumeurs développées sur un contexte inflammatoire. In conclusion, this analysis highlights a group of distinct patients, with a high risk of recurrence characterized by intestinal strains signatures and high ALEP program scores. Example 7: Extension of the observations to a second model of tumors developed on an inflammatory context.
Les adénocarcinomes œsophagiens tirent leur origine de l'œsophage de Barrett (situé dans le tiers inférieur de l'œsophage) et l'inflammation chronique liée à la remontée de sucs gastriques y joue un rôle clef. L'analyse de QPRT par immunohistochimie montre qu'une proportion des adénocarcinomes testés exprime la protéine QPRT (20%, n=10) suggérant que l'induction du programme puisse dans ce contexte également jouer un rôle clef dans l'initiation tumorale. Aucun marquage n'est observé dans l'épithélium malpighien, utilisé comme contrôle interne (Figure 11). Conclusions : Ces résultats mettent en lumière l'intérêt potentiel que représente QPRT en tant que cible thérapeutique. Ces résultats mettent aussi en évidence un nouveau programme induit dans les cancers gastro-intestinaux, très vraisemblablement au sein des cellules souches. Au-delà de sa valeur prédictive en termes de risque de récidive, l'intérêt de cette signature est de constituer un nouvel outil pour identifier des tumeurs qui pourraient être ciblées par des inhibiteurs de QPRT.  Esophageal adenocarcinomas originate from Barrett's esophagus (located in the lower third of the esophagus) and chronic inflammation related to the rise of gastric juices plays a key role. QPRT analysis by immunohistochemistry shows that a proportion of the adenocarcinomas tested express the protein QPRT (20%, n = 10) suggesting that the induction of the program can in this context also play a key role in tumor initiation. No labeling is observed in the squamous epithelium, used as an internal control (Figure 11). Conclusions: These findings highlight the potential value of QPRT as a therapeutic target. These results also highlight a new program induced in gastrointestinal cancers, most likely within stem cells. Beyond its predictive value in terms of risk of recurrence, the interest of this signature is to constitute a new tool to identify tumors that could be targeted by QPRT inhibitors.
REFERENCES BIBLIOGRAPHIQUES BIBLIOGRAPHIC REFERENCES
Baker,B.W., et al. (1992). Nucleic Acids Res. 20, 5234. Baker, B.W., et al. (1992). Nucleic Acids Res. 20, 5234.
Barretina,J., et al. (2012). Nature 483, 603-607.  Barretina, J., Et al. (2012). Nature 483, 603-607.
Boehm,J.S., et al. (2005). Mol. Cell Biol. 25, 6464-6474. Boehm, J.S., et al. (2005). Mol. Cell Biol. 25, 6464-6474.
Burns,J.C, et al. (1993). Proc. Natl. Acad. Sci. U. S. A 90, 8033-8037.  Burns, J.C. et al. (1993). Proc. Natl. Acad. Sci. U.S.A. 90, 8033-8037.
Dimri,G.P., et al. (1995). Proc. Natl. Acad. Sci. U. S. A 92, 9363-9367.  Dimri, G. P., et al. (1995). Proc. Natl. Acad. Sci. U.S. 92, 9363-9367.
Gras,B., et al. (2014). PLoS. One. 9, e92254.  Fat, B, et al. (2014). PLoS. One. 9, e92254.
Irizarry,R.A., et al. (2003). Biostatistics. 4, 249-264.  Irizarry, R.A., et al. (2003). Biostatistics. 4, 249-264.
Ishidoh,K., et al. (2010). Biochim. Biophys. Acta 1803, 527-533. Ishidoh, K., Et al. (2010). Biochim. Biophys. Acta 1803, 527-533.
Liu,H., et al. (2007). J. Mol. Biol. 373, 755-763.  Liu, H., et al. (2007). J. Mol. Biol. 373, 755-763.
Malik,S.S., et al. (2014) Proteins 82, 405-14.  Malik, S.S., et al. (2014) Proteins 82, 405-14.
Marisa,L., et al. (2013). PLoS. Med. 10, el001453.  Marisa, L., et al. (2013). PLoS. Med. 10, el001453.
Merlos-Suarez,A., et al. (2011). Cell Stem Cell 8, 511-524.  Merlos-Suarez, A., et al. (2011). Cell Stem Cell 8, 511-524.
Montero-Melendez,T. et al. (2013). PLoS. One. 8, e76235. Montero-Melendez, T. et al. (2013). PLoS. One. 8, e76235.
MoreLA.P., et al. (2012). PLoS. Genêt. 8, e 1002723.  MoreLA.P., Et al. (2012). PLoS. Broom. 8, e 1002723.
Pekow,J. et al. (2013) Inflamm. Bowel. Dis. 19, 461-470.  Pekow, J. et al. (2013) Inflamm. Bowel. Dis. 19, 461-470.
Sahm,F., et al. (2013) Cancer Res. 3225-34.  Sahm, F., et al. (2013) Cancer Res. 3225-34.
Sayan,A.E., et al. (2009). Proc. Natl. Acad. Sci. U. S. A 106, 14884-14889.  Sayan, A.E., et al. (2009). Proc. Natl. Acad. Sci. U.S. A 106, 14884-14889.
Vallerini,G.P., et al. (2013). J. Med. Chem. 56, 9482-95. Vallerini, G.P., et al. (2013). J. Med. Chem. 56, 9482-95.
Vecsei, L., et al (2013). Nat. Rev. Drud Discov. 12, 64-82. Vecsei, L., et al (2013). Nat. Rev. Drud Discov. 12, 64-82.
Watanabe, T., et al (2013) Clin Can. Res. 13, 415-420. Watanabe, T., et al (2013) Clin Can. Res. 13, 415-420.
Zufferey,R., et al. (1997). Nat. Biotechnol. 15, 871-875. Zufferey, R., et al. (1997). Nat. Biotechnol. 15, 871-875.

Claims

REVENDICATIONS
Procédé ex vivo de sélection d'un patient atteint d'une tumeur gastro-intestinale susceptible de bénéficier d'un traitement inhibiteur de la voie de la kynurénine à partir d'un échantillon test de ladite tumeur avant traitement dudit patient, ledit procédé comprenant les étapes suivantes :  An ex vivo method for selecting a patient suffering from a gastrointestinal tumor that may benefit from an inhibitory treatment of the kynurenine pathway from a test sample of said tumor prior to treatment of said patient, said method comprising the following steps :
a) La détermination de l'expression de la protéine QPRT dans les cellules épithéliales dudit échantillon test,  a) determination of the expression of the QPRT protein in the epithelial cells of said test sample,
b) La comparaison de l'expression de la protéine QPRT dans les cellules épithéliales dudit échantillon test avec l'expression de QPRT dans un échantillon de référence, c) La sélection d'un patient susceptible de bénéficier d'un traitement inhibiteur de la voie de la kynurénine à partir de la comparaison de l'étape b), un niveau d'expression supérieur de protéine QPRT dans les cellules épithéliales de l'échantillon test par rapport à celui de l'échantillon de référence étant indicateur de la sensibilité de ladite tumeur à un traitement inhibiteur de la voie de la kynurénine.  b) The comparison of the expression of the QPRT protein in the epithelial cells of the said test sample with the expression of QPRT in a reference sample, c) The selection of a patient likely to benefit from an inhibitor treatment of the pathway of kynurenine from the comparison of step b), a higher expression level of QPRT protein in the test sample epithelial cells than that of the reference sample being indicative of the sensitivity of said test sample. tumor to an inhibitory treatment of the kynurenine pathway.
Procédé selon la revendication 1 , dans lequel ledit échantillon test de ladite tumeur est issu de la tumeur primaire. The method of claim 1, wherein said test sample of said tumor is derived from the primary tumor.
Procédé selon l'une des revendications 1 à 2, dans lequel la détermination de l'expression de QPRT comprend au moins une étape d'immunohistochimie. The method according to one of claims 1 to 2, wherein the determination of QPRT expression comprises at least one immunohistochemistry step.
Procédé selon l'une des revendications 2 à 3, dans lequel la détermination de l'expression de QPRT comprend la localisation de cellules marquées au sein du tissu épithélial de l'échantillon test. The method according to one of claims 2 to 3, wherein determining QPRT expression comprises locating labeled cells within the epithelial tissue of the test sample.
Procédé selon l'une des revendications 2 à 4 dans lequel la détermination de l'expression de QPRT comprend la détermination du pourcentage de cellules épithéliales marquées. The method of one of claims 2 to 4 wherein the determination of QPRT expression comprises determining the percentage of labeled epithelial cells.
Procédé selon l'une des revendications 1 à 5, dans lequel ladite tumeur gastro-intestinale est choisie parmi le cancer colorectal, le cancer gastrique et l'adénocarcinome de l'œsophage. The method according to one of claims 1 to 5, wherein said gastrointestinal tumor is selected from colorectal cancer, gastric cancer and adenocarcinoma of the esophagus.
Procédé selon l'une des revendications 1 à 6, dans lequel ladite tumeur gastro-intestinale est développée dans un contexte inflammatoire chronique. Method according to one of claims 1 to 6, wherein said gastrointestinal tumor is developed in a chronic inflammatory context.
Procédé selon la revendication 7, dans lequel ladite tumeur gastro-intestinale est un cancer colorectal développé sur un contexte inflammatoire chronique. The method of claim 7, wherein said gastrointestinal tumor is colorectal cancer developed in a chronic inflammatory context.
9. Procédé selon l'une des revendications 1 à 6, dans lequel ladite tumeur gastro-intestinale est un cancer colorectal sporadique. The method according to one of claims 1 to 6, wherein said gastrointestinal tumor is sporadic colorectal cancer.
10. Procédé selon l'une des revendications 1 à 9, dans lequel ledit traitement inhibiteur de la voie de la kynurénine est choisi parmi un traitement inhibiteur de QPRT et un traitement inhibiteur de TD02. The method according to one of claims 1 to 9, wherein said inhibitory treatment of the kynurenine pathway is selected from a QPRT inhibitor treatment and a TD02 inhibitor treatment.
11. Utilisation de QPRT en tant que biomarqueur pour la prédiction de la sensibilité d'un patient atteint d'une tumeur gastro-intestinale à un traitement inhibiteur de la voie de la kynurénine, ledit patient étant non-traité pour ladite tumeur et l'expression de la protéine QPRT étant mesurée dans les cellules épithéliales d'un échantillon de ladite tumeur. 11. Use of QPRT as a biomarker for predicting the sensitivity of a gastrointestinal tumor patient to an inhibitory treatment of the kynurenine pathway, said patient being untreated for said tumor and the expressing the QPRT protein being measured in the epithelial cells of a sample of said tumor.
12. Utilisation selon la revendication 11 , dans laquelle ledit traitement inhibiteur de la voie de la kynurénine est choisi parmi un traitement inhibiteur de QPRT et un traitement inhibiteur de TD02. The use of claim 11, wherein said kynurenine pathway inhibitory treatment is selected from a QPRT inhibitor treatment and a TD02 inhibitor treatment.
PCT/FR2016/051419 2015-06-12 2016-06-13 Method for predicting the sensitivity of a gastrointestinal tumor to an inhibitor treatment WO2016198807A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
FR1555357 2015-06-12
FR1555357A FR3037403A1 (en) 2015-06-12 2015-06-12 METHOD FOR PREDICTING SUSCEPTIBILITY OF GASTROINTESTINAL TUMOR TO INHIBITORY TREATMENT

Publications (1)

Publication Number Publication Date
WO2016198807A1 true WO2016198807A1 (en) 2016-12-15

Family

ID=54066039

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/FR2016/051419 WO2016198807A1 (en) 2015-06-12 2016-06-13 Method for predicting the sensitivity of a gastrointestinal tumor to an inhibitor treatment

Country Status (2)

Country Link
FR (1) FR3037403A1 (en)
WO (1) WO2016198807A1 (en)

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009140409A1 (en) * 2008-05-14 2009-11-19 Genomic Health Inc. Predictors of patient response to treatment with egf receptor inhibitors
US20140357693A1 (en) * 2013-02-25 2014-12-04 Whitehead Institute For Biomedical Research Metabolic gene mesenchymal signatures and uses thereof
US20150141273A1 (en) * 2012-04-26 2015-05-21 Stichting Vu-Vumc Biomarkers

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009140409A1 (en) * 2008-05-14 2009-11-19 Genomic Health Inc. Predictors of patient response to treatment with egf receptor inhibitors
US20150141273A1 (en) * 2012-04-26 2015-05-21 Stichting Vu-Vumc Biomarkers
US20140357693A1 (en) * 2013-02-25 2014-12-04 Whitehead Institute For Biomedical Research Metabolic gene mesenchymal signatures and uses thereof

Non-Patent Citations (23)

* Cited by examiner, † Cited by third party
Title
BAKER,B.W. ET AL., NUCLEIC ACIDS RES., vol. 20, 1992, pages 5234
BARRETINA,J. ET AL., NATURE, vol. 483, 2012, pages 603 - 607
BOEHM,J.S. ET AL., MOL. CELL BIOL., vol. 25, 2005, pages 6464 - 6474
BURNS,J.C. ET AL., PROC. NATL. ACAD. SCI. U. S. A, vol. 90, 1993, pages 8033 - 8037
DIMRI,G.P. ET AL., PROC. NATL. ACAD. SCI. U. S. A, vol. 92, 1995, pages 9363 - 9367
F. SAHM ET AL: "The Endogenous Tryptophan Metabolite and NAD+ Precursor Quinolinic Acid Confers Resistance of Gliomas to Oxidative Stress", CANCER RESEARCH, vol. 73, no. 11, 1 June 2013 (2013-06-01), US, pages 3225 - 3234, XP055261183, ISSN: 0008-5472, DOI: 10.1158/0008-5472.CAN-12-3831 *
GRAS,B. ET AL., PLOS. ONE, vol. 9, 2014, pages E92254
IRIZARRY,R.A. ET AL., BIOSTATISTICS, vol. 4, 2003, pages 249 - 264
ISHIDOH,K. ET AL., BIOCHIM. BIOPHYS. ACTA, vol. 1803, 2010, pages 527 - 533
JENSEN NIELS F ET AL: "Establishment and characterization of models of chemotherapy resistance in colorectal cancer: Towards a predictive signature of chemoresistance", MOLECULAR ONCOLOGY, vol. 9, no. 6, 24 February 2015 (2015-02-24), pages 1169 - 1185, XP029172021, ISSN: 1574-7891, DOI: 10.1016/J.MOLONC.2015.02.008 *
LIU,H. ET AL., J. MOL. BIOL., vol. 373, 2007, pages 755 - 763
MALIK,S.S. ET AL., PROTEINS, vol. 82, 2014, pages 405 - 14
MARISA,L. ET AL., PLOS. MED., vol. 10, 2013, pages EL001453
MERLOS-SUAREZ,A. ET AL., CELL STEM CELL, vol. 8, 2011, pages 511 - 524
MONTERO-MELENDEZ,T. ET AL., PLOS. ONE, vol. 8, 2013, pages E76235
MOREL,A.P. ET AL., PLOS. GENET., vol. 8, 2012, pages 1002723
PEKOW,J. ET AL., INFLAMM. BOWEL. DIS., vol. 19, 2013, pages 461 - 470
SAHM,F. ET AL., CANCER RES., 2013, pages 3225 - 34
SAYAN,A.E. ET AL., PROC. NATL. ACAD. SCI. U. S. A, vol. 106, 2009, pages 14884 - 14889
VALLERINI,G.P. ET AL., J. MED. CHEM., vol. 56, 2013, pages 9482 - 95
VECSEI, L. ET AL., NAT. REV. DRUD DISCOV., vol. 12, 2013, pages 64 - 82
WATANABE, T. ET AL., CLIN CAN. RES., vol. 13, 2013, pages 415 - 420
ZUFFEREY,R. ET AL., NAT. BIOTECHNOL., vol. 15, 1997, pages 871 - 875

Also Published As

Publication number Publication date
FR3037403A1 (en) 2016-12-16

Similar Documents

Publication Publication Date Title
Yuan et al. SETD2 restricts prostate cancer metastasis by integrating EZH2 and AMPK signaling pathways
Lin et al. Enhancer-driven lncRNA BDNF-AS induces endocrine resistance and malignant progression of breast cancer through the RNH1/TRIM21/mTOR cascade
Varga et al. AKT-dependent NOTCH3 activation drives tumor progression in a model of mesenchymal colorectal cancer
Watanabe et al. Loss of KDM6A characterizes a poor prognostic subtype of human pancreatic cancer and potentiates HDAC inhibitor lethality
Kim et al. Notch1 counteracts WNT/β-catenin signaling through chromatin modification in colorectal cancer
Alam et al. MicroRNA 375 regulates proliferation and migration of colon cancer cells by suppressing the CTGF‐EGFR signaling pathway
Hsu et al. TET1 suppresses cancer invasion by activating the tissue inhibitors of metalloproteinases
Liu et al. ErbB2 pathway activation upon Smad4 loss promotes lung tumor growth and metastasis
Zhou et al. MiR-339-5p regulates the growth, colony formation and metastasis of colorectal cancer cells by targeting PRL-1
Mello et al. Deregulated expression of SRC, LYN and CKB kinases by DNA methylation and its potential role in gastric cancer invasiveness and metastasis
Lorenzo-Martín et al. VAV2 signaling promotes regenerative proliferation in both cutaneous and head and neck squamous cell carcinoma
Prickett et al. Somatic mutations in MAP3K5 attenuate its proapoptotic function in melanoma through increased binding to thioredoxin
Olasz et al. MicroRNA-135b regulates leucine zipper tumor suppressor 1 in cutaneous squamous cell carcinoma
Liu et al. LncRNA MNX1-AS1 sustains inactivation of Hippo pathway through a positive feedback loop with USP16/IGF2BP3 axis in gallbladder cancer
Chou et al. Targeting of miR‐31/96/182 to the numb gene during head and neck oncogenesis
Li et al. RNA Splicing of the Abi1 Gene by MBNL1 contributes to macrophage‐like phenotype modulation of vascular smooth muscle cell during atherogenesis
Diazzi et al. Blockade of the pro‐fibrotic reaction mediated by the miR‐143/‐145 cluster enhances the responses to targeted therapy in melanoma
Calvet et al. YAP1 is essential for malignant mesothelioma tumor maintenance
Wang et al. Epigenetic silencing of ASPP1 confers 5‐FU resistance in clear cell renal cell carcinoma by preventing p53 activation
Hwang et al. Upregulation of S100A9 contributes to the acquired resistance to BRAF inhibitors
Chen et al. N6-methyladenosine modification in 18S rRNA promotes tumorigenesis and chemoresistance via HSF4b/HSP90B1/mutant p53 axis
Stoy et al. Transcriptional co‐factor Transducin beta‐like (TBL) 1 acts as a checkpoint in pancreatic cancer malignancy
Luo et al. CUL4B-DDB1-COP1-mediated UTX downregulation promotes colorectal cancer progression
Wang et al. A novel HIF1α-STIL-FOXM1 axis regulates tumor metastasis
WO2016198807A1 (en) Method for predicting the sensitivity of a gastrointestinal tumor to an inhibitor treatment

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: 16741075

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 16741075

Country of ref document: EP

Kind code of ref document: A1