WO2018189403A1 - Méthodes et compositions pharmaceutiques destinées au traitement du cancer - Google Patents

Méthodes et compositions pharmaceutiques destinées au traitement du cancer Download PDF

Info

Publication number
WO2018189403A1
WO2018189403A1 PCT/EP2018/059613 EP2018059613W WO2018189403A1 WO 2018189403 A1 WO2018189403 A1 WO 2018189403A1 EP 2018059613 W EP2018059613 W EP 2018059613W WO 2018189403 A1 WO2018189403 A1 WO 2018189403A1
Authority
WO
WIPO (PCT)
Prior art keywords
vegf
cancer
cells
patient
radiotherapy
Prior art date
Application number
PCT/EP2018/059613
Other languages
English (en)
Inventor
Gilles PAGES
Jérôme DOYEN
Audrey CLAREN MERCATI
Marilena LUPU PLESU
Original Assignee
INSERM (Institut National de la Santé et de la Recherche Médicale)
Universite De Nice Sophia Antipolis
Centre National De La Recherche Scientifique
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 INSERM (Institut National de la Santé et de la Recherche Médicale), Universite De Nice Sophia Antipolis, Centre National De La Recherche Scientifique filed Critical INSERM (Institut National de la Santé et de la Recherche Médicale)
Publication of WO2018189403A1 publication Critical patent/WO2018189403A1/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6883Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
    • C12Q1/6886Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material for cancer
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/106Pharmacogenomics, i.e. genetic variability in individual responses to drugs and drug metabolism
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/118Prognosis of disease development
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/158Expression markers

Definitions

  • the present invention relates to methods and pharmaceutical compositions for the treatment of cancer.
  • radiotherapy Approximately 50% of all cancer patients are subject to radiotherapy during the course of their illness with an estimation that radiotherapy contributes to approximately 40% towards curative treatment (1).
  • the goal of radiotherapy is to deliver loco-regionally a specific dose of radioactivity that will allow the destruction of cancer cells, while limiting the exposure of surrounding healthy tissues.
  • X photons
  • the main disadvantage of X radiotherapy is represented by the deposition of radiation also at the level of surrounding healthy tissues, leading to side effects.
  • P proton beams
  • the ionizing radiation by proton beams (P) is currently more expensive and more difficult to produce, it has the physical advantage of delivering no radiation outside of the intended targeted area, thanks to the so-called Bragg peak (2).
  • P radiotherapy is mainly proposed for the treatment of uveal melanoma, skull base and paraspinal tumors due to its high precision in tumor targeting with a very high irradiation dose next to radiosensitive structures (2). It is also proposed for the pediatric tumors based on the advantage to deliver a much lower integral dose, which significantly reduces the risk of radiation induced cancers in a long-life expectancy setting (2).
  • HNSCC head and neck squamous cell carcinoma
  • HNSCC head and neck cancers are among the ten most common types of cancer and the 7 th cause of mortality from cancer worldwide.
  • the treatment of HNSCC consists of either chemoradiotherapy and/or surgical excision (3).
  • conventional radiotherapy with X in HNSCC remains difficult, due to the proximity of numerous organs at risk (i.e. salivary glands, esophagus and larynx).
  • organs at risk i.e. salivary glands, esophagus and larynx.
  • Recent studies have shown an advantage of P, over X radiotherapy, in inducing lower toxicities (4) and lower dose delivery to organs of risk (5) in HNSCC patients.
  • VEGF-C Vascular Endothelial Growth Factor C
  • MMP matrix-metalloproteinases
  • P may have different biological properties, as compared to X radiation at a similar dose.
  • the purpose of the present study was thus to analyze the different biological behaviors of FiNSCC cells when exposed to P vs X radiation.
  • the present invention relates to methods and pharmaceutical compositions for the treatment of cancer.
  • HNSCC head and neck squamous cell carcinoma
  • RBE relative biological effectiveness
  • VEGF-C protein levels and regulations were documented in freshly irradiated and/or long-term surviving cells receiving low/high-dose, single (SI)/multiple (MI) irradiations with P/X.
  • the RBE was found to be 1.1 Key (lymph)angiogenesis and inflammation genes were down-regulated, except for vegf-c, after P and up-regulated after X irradiation in MI surviving cells, demonstrating a more favorable profile after P irradiation. Both irradiation types stimulated vegf-c promoter activity in a NF-KB-dependent transcriptional regulation manner, but at a lesser extent after P, as compared to X irradiation, which correlated with mRNA and protein levels. The cells surviving to MI by P or X generated tumors with higher volume, anarchic architecture and increased density of blood vessels.
  • the present invention demonstrates the biological advantage of P, as compared to X irradiation. In addition to its physical advantage in dose deposition, P irradiation may help to improve treatment approaches for HNSCC.
  • the inventors demonstrated that P and X radiation-induced VEGF-C over-expression at both gene and protein levels in HNSCC cells and in meduUoblastoma (MDB) cells and that VEGF-C is a major factor responsible for post-irradiation disease progression in FiNSCC patients, via promotion of lymphangiogenesis.
  • MDB meduUoblastoma
  • VEGF-C is an important therapeutic target for FiNSCC patients who relapse after radiotherapy with either P or X.
  • the inventors also investigated the presence of lymphatic markers in biopsies from primary and locally relapsed human FiNSCC, after conventional X radiotherapy.
  • the cohort of patients presented increased protein and/or mRNA levels of VEGF-C, PDPN, LYVE1 and PROX1, bringing evidence that X radiotherapy may promote lymphangiogenesis.
  • the present invention relates to a method for predicting the outcome of a cancer in patient afflicted with solid cancer after radiotherapy treatment, comprising the steps of: i) determining the expression level of VEGF-C in a biological sample obtained from said patient, ii) comparing the expression level determined at step i) with a predetermined reference value and iii) concluding that the patient has a good prognosis when the level determined at step i) is lower than the predetermined reference value or concluding that the patient has a poor prognosis when the level determined at step i) is higher than the predetermined reference value.
  • a patient denotes a mammal.
  • a patient according to the invention refers to any patient (preferably human) afflicted with solid cancer.
  • the term “patient” also refers to any patient afflicted with head and neck squamous cell carcinoma (FiNSCC) or with meduUoblastoma (MDB) for example.
  • the term “patient” also refers to any patient afflicted with solid cancer receiving radiotherapy.
  • patient also refers to any patient afflicted with head and neck squamous cell carcinoma (FiNSCC) or with meduUoblastoma (MDB) receiving radiotherapy.
  • solid cancer has its general meaning in the art and refers to solid cancer selected from the group consisting of, but not limited to, head and neck squamous cell carcinoma (FiNSCC), adrenal cortical cancer, anal cancer, bile duct cancer (e.g. periphilar cancer, distal bile duct cancer, intrahepatic bile duct cancer), bladder cancer, bone cancer (e.g.
  • osteoblastoma osteochrondroma, hemangioma, chondromyxoid fibroma, osteosarcoma, chondrosarcoma, fibrosarcoma, malignant fibrous histiocytoma, giant cell tumor of the bone, chordoma, multiple myeloma), brain and central nervous system cancer (e.g. meningioma, astocytoma, oligodendrogliomas, ependymoma, gliomas, meduUoblastoma (MDB), ganglioglioma, Schwannoma, germinoma, craniopharyngioma), breast cancer (e.g.
  • MDB meduUoblastoma
  • ductal carcinoma in situ infiltrating ductal carcinoma, infiltrating lobular carcinoma, lobular carcinoma in situ, gynecomastia
  • cervical cancer colorectal cancer
  • endometrial cancer e.g. endometrial adenocarcinoma, adenocanthoma, papillary serous adenocarcinoma, clear cell
  • esophagus cancer gallbladder cancer (mucinous adenocarcinoma, small cell carcinoma), gastrointestinal carcinoid tumors (e.g. choriocarcinoma, chorioadenoma destruens), Kaposi's sarcoma, kidney cancer (e.g.
  • liver cancer e.g. hemangioma, hepatic adenoma, focal nodular hyperplasia, hepatocellular carcinoma
  • lung cancer e.g. small cell lung cancer, non-small cell lung cancer
  • mesothelioma plasmacytoma
  • nasal cavity and paranasal sinus cancer e.g.
  • esthesioneuroblastoma midline granuloma
  • nasopharyngeal cancer neuroblastoma
  • oral cavity and oropharyngeal cancer ovarian cancer, pancreatic cancer, penile cancer, pituitary cancer, prostate cancer, retinoblastoma, rhabdomyosarcoma (e.g. embryonal rhabdomyosarcoma, alveolar rhabdomyosarcoma, pleomorphic rhabdomyosarcoma), salivary gland cancer, skin cancer (e.g. melanoma, nonmelanoma skin cancer), stomach cancer, testicular cancer (e.g.
  • thyroid cancer e.g. follicular carcinoma, anaplastic carcinoma, poorly differentiated carcinoma, medullary thyroid carcinoma
  • vaginal cancer e.g. vulvar cancer
  • uterine cancer e.g. uterine leiomyosarcoma
  • the cancer is head and neck squamous cell carcinoma (HNSCC).
  • HNSCC head and neck squamous cell carcinoma
  • the cancer is medulloblastoma (MDB).
  • biological sample refers to any biological sample derived from the patient such as solid cancer sample, biopsy sample, blood sample, plasma sample, or serum sample. Said biological sample is obtained for the purpose of the in vitro evaluation.
  • radiotherapy treatment has its general meaning in the art and refers to photon (X) radiotherapy and proton (P) radiotherapy.
  • radiation treatment also refers to photon irradiation and proton irradiation.
  • radiation treatment also refers to single irradiation (SI) and multiple irradiations (MI) such as described in the example.
  • radiation treatment also refers to radiation therapy using radio therapeutic agent administered to the patient afflicted with solid cancer.
  • Radiation therapy or radiotherapy is the medical use of irradiation (i.e. ionizing radiation) as part of cancer treatment to control malignant cells.
  • Ionizing radiation deposits energy that injures or destroys cells in the area being treated (the target tissue) by damaging their genetic material, making it impossible for these cells to continue to grow.
  • One type of radiation therapy commonly used involves photons, e.g. X-rays. Depending on the amount of energy they possess, the rays can be used to destroy cancer cells on the surface of or deeper in the body. The higher the energy of the x-ray beam, the deeper the x-rays can go into the target tissue. Linear accelerators and betatrons produce x-rays of increasingly greater energy.
  • the use of machines to focus radiation (such as x-rays) on a cancer site is called external beam radiotherapy.
  • Gamma rays are another form of photons used in radiotherapy.
  • Gamma rays are produced spontaneously as certain elements (such as radium, uranium, and cobalt 60) release radiation as they decompose, or decay.
  • Another technique for delivering radiation to cancer cells is to place radioactive implants directly in a tumor or body cavity. This is called internal radiotherapy.
  • Brachytherapy, interstitial irradiation, and intracavitary irradiation are types of internal radiotherapy. In this treatment, the radiation dose is concentrated in a small area.
  • a further technique is intra-operative irradiation, in which a large dose of external radiation is directed at the tumor and surrounding tissue during surgery.
  • particle beam radiation therapy differs from photon radiotherapy in that it involves the use of fast-moving subatomic particles to treat localized cancers. Some particles (protons, neutrons, pions, and heavy ions) deposit more energy along the path they take through tissue than do x-rays or gamma rays, thus causing more damage to the cells they hit. This type of radiation is often referred to as high linear energy transfer (high LET) radiation. Radio-sensitizers make the tumor cells more likely to be damaged, and radio -protectors protect normal tissues from the effects of radiation.
  • high LET high linear energy transfer
  • a person of ordinary skill in the radiotherapy art knows how to determine an appropriate dosing and application schedule, depending on the nature of the disease and the constitution of the patient. In particular, the person knows how to assess dose-limiting toxicity (DLT) and how to determine the maximum tolerated dose (MTD) accordingly. More particularly, the amount of radiation used in photon radiation therapy is measured in gray (Gy), and varies depending on the type and stage of cancer being treated. Many other factors are considered by radiation oncologists when selecting a dose, including whether the patient is receiving chemotherapy, patient co -morbidities, whether radiation therapy is being administered before or after surgery, and the degree of success of surgery. Moreover, the total dose is often fractionated. Fractionation regimes are individualized between different radiotherapy centers.
  • the radiotherapy can be applied at a dose in a range from about 1 to 80Gy, about 10 to 55Gy, preferably from about 15 to 50 Gy, such as 20 to 40Gy, concretely from about 20 to 35 Gy, and more concretely from about 25 to 30 Gy.
  • VEGF-C has its general meaning in the art and refers to Vascular Endothelial Growth Factor C, a member of the vascular endothelial growth factor family.
  • the term "Good Prognosis” refers to a patient afflicted with solid cancer receiving radiotherapy treatment that is likely to not present lymph node metastasis, and/or that is likely to not present cancer relapse, and/or that is likely to present a high overall survival (OS), event-free survival (EFS), and/or metastasis- free survival (MFS).
  • OS overall survival
  • EFS event-free survival
  • MFS metastasis- free survival
  • the term "Poor Prognosis” or “Bad Prognosis” refers to a patient afflicted with solid cancer receiving radiotherapy treatment that is likely to present lymph node metastasis, and/or that is likely to present cancer relapse, and/or that is likely to present short overall survival (OS), progression free survival (PFS) and/or metastasis.
  • OS overall survival
  • PFS progression free survival
  • the method of the invention also comprises determining the expression level of PDPN, LYVE1 and PROX1.
  • PDPN has its general meaning in the art and refers to podoplanin, a mucin- type transmembrane protein.
  • LYVE1 has its general meaning in the art and refers to lymphatic vessel endothelial hyaluronan receptor 1.
  • PROX1 has its general meaning in the art and refers to prospero homeobox 1 transcription factor.
  • the "reference value” refers to a threshold value or a cut-off value.
  • the setting of a single “reference value” thus allows discrimination between a poor and a good prognosis with respect to the lymph node metastasis, cancer relapse and overall survival (OS) for a patient.
  • a “threshold value” or “cut-off value” can be determined experimentally, empirically, or theoretically.
  • a threshold value can also be arbitrarily selected based upon the existing experimental and/or clinical conditions, as would be recognized by a person of ordinary skilled in the art. The threshold value has to be determined in order to obtain the optimal sensitivity and specificity according to the function of the test and the benefit/risk balance (clinical consequences of false positive and false negative).
  • the optimal sensitivity and specificity can be determined using a Receiver Operating Characteristic (ROC) curve based on experimental data.
  • ROC Receiver Operating Characteristic
  • the person skilled in the art may compare the expression level (obtained according to the method of the invention) with a defined threshold value.
  • the threshold value is derived from the expression level (or ratio, or score) determined in a biological sample derived from one or more patients having solid cancer.
  • retrospective measurement of the expression level (or ratio, or scores) in properly banked historical patient samples may be used in establishing these threshold values.
  • Predetermined reference values used for comparison may comprise "cut-off or "threshold” values that may be determined as described herein.
  • Each reference (“cut-off) value for the bio marker of interest may be predetermined by carrying out a method comprising the steps of
  • step e providing, for each sample provided at step a), information relating to the responsiveness of the patient or the actual clinical outcome for the corresponding cancer patient (i.e. the duration of the event-free survival (EFS), metastasis- free survival (MFS) or the overall survival (OS) or both);
  • EFS event-free survival
  • MFS metastasis- free survival
  • OS overall survival
  • the expression level of a biomarker has been assessed for 100 cancer samples of 100 patients.
  • the 100 samples are ranked according to their expression level.
  • Sample 1 has the best expression level and sample 100 has the worst expression level.
  • a first grouping provides two subsets: on one side sample Nr 1 and on the other side the 99 other samples.
  • the next grouping provides on one side samples 1 and 2 and on the other side the 98 remaining samples etc., until the last grouping: on one side samples 1 to 99 and on the other side sample Nr 100.
  • Kaplan Meier curves are prepared for each of the 99 groups of two subsets. Also for each of the 99 groups, the p value between both subsets was calculated.
  • the reference value is selected such as the discrimination based on the criterion of the minimum p value is the strongest.
  • the expression level corresponding to the boundary between both subsets for which the p value is minimum is considered as the reference value. It should be noted that the reference value is not necessarily the median value of expression levels.
  • the reference value (cut-off value) may be used in the present method to discriminate cancer samples and therefore the corresponding patients.
  • Kaplan-Meier curves of percentage of survival as a function of time are commonly to measure the fraction of patients living for a certain amount of time after treatment and are well known by the man skilled in the art.
  • the reference value may correspond to the expression level of the biomarker determined in a sample associated having a good prognosis. Accordingly, a higher expression level of the biomarker than the reference value is indicative of a patient having poor prognosis, and a lower or equal expression level of the biomarker than the reference value is indicative of a patient having a good prognosis.
  • the reference value may correspond to the expression level of the biomarker determined in a sample associated with a patient having a poor prognosis. Accordingly, a higher or equal expression level of the biomarker than the reference value is indicative of a patient having poor prognosis, and a lower expression level of the biomarker than the reference value is indicative of a patient having good prognosis.
  • a score which is a composite of the expression levels of the different biomarkers may also be determined and compared to a reference value wherein a difference between said score and said reference value is indicative of a patient having a good or poor prognosis
  • the score may be generated by a computer program. Analyzing the biomarker expression level may be assessed by any of a wide variety of well-known methods for detecting expression of a transcribed nucleic acid or translated protein.
  • the biomarker expression level is assessed by analyzing the expression of the protein translated from said gene. Said analysis can be assessed using an antibody (e.g., a radio-labeled, chromophore-labeled, fluorophore-labeled, or enzyme-labeled antibody), an antibody derivative (e.g., an antibody conjugate with a substrate or with the protein or ligand of a protein of a protein/ligand pair (e.g., biotin-streptavidin)), or an antibody fragment (e.g., a single-chain antibody, an isolated antibody hypervariable domain, etc.) which binds specifically to the protein translated from the gene encoding for the biomarker.
  • an antibody e.g., a radio-labeled, chromophore-labeled, fluorophore-labeled, or enzyme-labeled antibody
  • an antibody derivative e.g., an antibody conjugate with a substrate or with the protein or ligand of a protein
  • Methods for measuring the expression level of a biomarker in a sample may be assessed by any of a wide variety of well-known methods from one of skill in the art for detecting expression of a protein including, but not limited to, direct methods like mass spectrometry-based quantification methods, protein microarray methods, enzyme immunoassay (EIA), radioimmunoassay (RIA), Immunohistochemistry (IHC), Western blot analysis, ELISA, Luminex, ELISPOT and enzyme linked immunoabsorbant assay and indirect methods based on detecting expression of corresponding messenger ribonucleic acids (mRNAs).
  • the mRNA expression profile may be determined by any technology known by a man skilled in the art.
  • each mRNA expression level may be measured using any technology known by a man skilled in the art, including nucleic microarrays, quantitative Polymerase Chain Reaction (qPCR), next generation sequencing and hybridization with a labelled probe.
  • the binding partner may be an antibody that may be polyclonal or monoclonal, preferably monoclonal (e.g., a isotope-label, element-label, radio-labeled, chromophore-labeled, fluorophore-labeled, or enzyme-labeled antibody), an antibody derivative (e.g., an antibody conjugate with a substrate or with the protein or ligand of a protein of a protein/ligand pair (e.g., biotin- streptavidin)), or an antibody fragment (e.g., a single-chain antibody, an isolated antibody hypervariable domain, etc.) which binds specifically to the protein translated from the gene encoding for the biomarker of the invention.
  • monoclonal e.g., a isotope-label, element-label, radio-labeled, chromophore-labeled, fluorophore-labeled, or enzyme-labeled antibody
  • an antibody derivative
  • the binding partner may be an aptamer.
  • the binding partners of the invention such as antibodies or aptamers may be labelled with a detectable molecule or substance, such as an isotope, an element, a fluorescent molecule, a radioactive molecule or any others labels known in the art. Labels are known in the art that generally provide (either directly or indirectly) a signal.
  • the term "labelled", with regard to the antibody is intended to encompass direct labelling of the antibody or aptamer by coupling (i.e., physically linking) a detectable substance, such as an isotope, an element, a radioactive agent or a fluorophore (e.g. fluorescein isothiocyanate (FITC) or phycoerythrin (PE) or Indocyanine (Cy5)) to the antibody or aptamer, as well as indirect labelling of the probe or antibody by reactivity with a detectable substance.
  • a detectable substance such as an isotope, an element, a radioactive agent or a fluorophore (e.g. fluorescein isothiocyanate (FITC) or phycoerythrin (PE) or Indocyanine (Cy5)
  • FITC fluorescein isothiocyanate
  • PE phycoerythrin
  • Indocyanine Indocyanine
  • radioactive molecules include but are not limited to radioactive atom for scintigraphic studies such as 1123, 1124, Inl 11, Rel86, Rel88, specific isotopes include but are not limited to 13C, 15N, 1261, 79Br, 81 Br.
  • the abore mentioned assays generally involve the binding of the binding partner (ie. antibody or aptamer) to a solid support.
  • Solid supports which can be used in the practice of the invention include substrates such as nitrocellulose (e. g., in membrane or microtiter well form); polyvinylchloride (e. g., sheets or microtiter wells); polystyrene latex (e.g., beads or microtiter plates); polyvinylidene fluoride; diazotized paper; nylon membranes; activated beads, magnetically responsive beads, silicon wafers.
  • substrates such as nitrocellulose (e. g., in membrane or microtiter well form); polyvinylchloride (e. g., sheets or microtiter wells); polystyrene latex (e.g., beads or microtiter plates); polyvinylidene fluoride; diazotized paper; nylon membranes; activated beads, magnetically responsive beads, silicon wafer
  • an ELISA method can be used, wherein the wells of a microtiter plate are coated with a set of antibodies which recognize said biomarker. A sample containing or suspected of containing said biomarker is then added to the coated wells. After a period of incubation sufficient to allow the formation of antibody-antigen complexes, the plate(s) can be washed to remove unbound moieties and a detectably labelled secondary binding molecule added. The secondary binding molecule is allowed to react with any captured sample marker protein, the plate washed and the presence of the secondary binding molecule detected using methods well known in the art such as Singulex, Quanterix, MSD, Bioscale, Cytof.
  • an Enzyme-linked immunospot (ELISpot) method may be used.
  • the sample is transferred to a plate which has been coated with the desired anti- biomarker capture antibodies.
  • Revelation is carried out with biotinylated secondary Abs and standard colorimetric or fluorimetric detection methods such as streptavidin-alkaline phosphatase and NBT-BCIP and the spots counted.
  • the bead may be a cytometric bead for use in flow cytometry.
  • Such beads may for example correspond to BDTM Cytometric Beads commercialized by BD Biosciences (San Jose, California).
  • cytometric beads may be suitable for preparing a multiplexed bead assay.
  • a multiplexed bead assay such as, for example, the BD(TM) Cytometric Bead Array, is a series of spectrally discrete beads that can be used to capture and quantify soluble antigens.
  • beads are labelled with one or more spectrally distinct fluorescent dyes, and detection is carried out using a multiplicity of photodetectors, one for each distinct dye to be detected.
  • a number of methods of making and using sets of distinguishable beads have been described in the literature. These include beads distinguishable by size, wherein each size bead is coated with a different target-specific antibody (see e.g. Fulwyler and McHugh, 1990, Methods in Cell Biology 33:613-629), beads with two or more fluorescent dyes at varying concentrations, wherein the beads are identified by the levels of fluorescence dyes (see e.g. European Patent No.
  • beads distinguishably labelled with two different dyes, wherein the beads are identified by separately measuring the fluorescence intensity of each of the dyes (see e.g. U.S. patent Nos. 4,499,052 and 4,717,655).
  • Both one- dimensional and two-dimensional arrays for the simultaneous analysis of multiple antigens by flow cytometry are available commercially. Examples of one-dimensional arrays of singly dyed beads distinguishable by the level of fluorescence intensity include the BD(TM) Cytometric Bead Array (CBA) (BD Biosciences, San Jose, Calif.) and Cyto-Plex(TM) Flow Cytometry microspheres (Duke Scientific, Palo Alto, Calif).
  • An example of a two-dimensional array of beads distinguishable by a combination of fluorescence intensity (five levels) and size (two sizes) is the QuantumPlex(TM) microspheres (Bangs Laboratories, Fisher, Ind.).
  • An example of a two-dimensional array of doubly-dyed beads distinguishable by the levels of fluorescence of each of the two dyes is described in Fulton et al. (1997, Clinical Chemistry 43(9): 1749-1756).
  • the beads may be labelled with any fluorescent compound known in the art such as e.g. FITC (FL1), PE (FL2), fluorophores for use in the blue laser (e.g.
  • bead is a magnetic bead for use in magnetic separation. Magnetic beads are known to those of skill in the art. Typically, the magnetic bead is preferably made of a magnetic material selected from the group consisting of metals (e.g. ferrum, cobalt and nickel), an alloy thereof and an oxide thereof. In another particular embodiment, bead is bead that is dyed and magnetized.
  • metals e.g. ferrum, cobalt and nickel
  • bead is bead that is dyed and magnetized.
  • protein microarray methods may be used.
  • at least one antibody or aptamer directed against the biomarker is immobilized or grafted to an array(s), a solid or semi-solid surface(s).
  • a sample containing or suspected of containing the biomarker is then labelled with at least one isotope or one element or one fluorophore or one colorimetric tag that are not naturally contained in the tested sample.
  • the array is then washed and dried.
  • quantifying said biomarker may be achieved using any appropriate microarray scanner like fluorescence scanner, colorimetric scanner, SIMS (secondary ions mass spectrometry) scanner, maldi scanner, electromagnetic scanner or any technique allowing quantifying said labels.
  • the antibody or aptamer grafted on the array is labelled.
  • reverse phase arrays may be used.
  • at least one sample is immobilized or grafted to an array(s), a solid or semi-solid surface(s).
  • An antibody or aptamer against the suspected biomarker is then labelled with at least one isotope or one element or one fluorophore or one colorimetric tag that is not naturally contained in the tested sample. After a period of incubation of said antibody or aptamer with the array sufficient to allow the formation of antibody-antigen complexes, the array is then washed and dried.
  • detecting quantifying and counting by D-SIMS said biomarker containing said isotope or group of isotopes, and a reference natural element, and then calculating the isotopic ratio between the biomarker and the reference natural element may be achieve using any appropriate microarray scanner like fluorescence scanner, colorimetric scanner, SIMS (secondary ions mass spectrometry) scanner, maldi scanner, electromagnetic scanner or any technique allowing to quantify said labels.
  • any appropriate microarray scanner like fluorescence scanner, colorimetric scanner, SIMS (secondary ions mass spectrometry) scanner, maldi scanner, electromagnetic scanner or any technique allowing to quantify said labels.
  • said direct analysis can also be assessed by mass Spectrometry.
  • Mass spectrometry-based quantification methods may be performed using either labelled or unlabelled approaches (DeSouza and Siu, 2012). Mass spectrometry-based quantification methods may be performed using chemical labeling, metabolic labelingor proteolytic labeling. Mass spectrometry-based quantification methods may be performed using mass spectrometry label free quantification, LTQ Orbitrap Velos, LTQ-MS/MS, a quantification based on extracted ion chromatogram EIC (progenesis LC-MS, Liquid chromatography-mass spectrometry) and then profile alignment to determine differential expression of the biomarker.
  • the biomarker expression level is assessed by analyzing the expression of mR A transcript or mRNA precursors, such as nascent R A, of biomarker gene. Said analysis can be assessed by preparing mRNA/cDNA from cells in a sample from a patient, and hybridizing the mRNA/cDNA with a reference polynucleotide. The prepared mRNA/cDNA can be used in hybridization or amplification assays that include, but are not limited to, Southern or Northern analyses, polymerase chain reaction analyses, such as quantitative PCR (TaqMan), and probes arrays such as GeneChip(TM) DNA Arrays (AFFYMETRIX).
  • mR A transcript or mRNA precursors such as nascent R A
  • the analysis of the expression level of mRNA transcribed from the gene encoding for biomarkers involves the process of nucleic acid amplification, e. g., by RT- PCR (the experimental embodiment set forth in U. S. Patent No. 4,683, 202), ligase chain reaction (Barany, 1991), self- sustained sequence replication (Guatelli et al, 1990), transcriptional amplification system (Kwoh et al., 1989), Q-Beta Replicase (Lizardi et al, 1988), rolling circle replication (U. S. Patent No. 5,854, 033) or any other nucleic acid amplification method, followed by the detection of the amplified molecules using techniques well known to those of skill in the art.
  • RT- PCR the experimental embodiment set forth in U. S. Patent No. 4,683, 202
  • ligase chain reaction Barany, 1991
  • self- sustained sequence replication (Guatelli et al, 1990)
  • transcriptional amplification system Kwoh
  • amplification primers are defined as being a pair of nucleic acid molecules that can anneal to 5' or 3' regions of a gene (plus and minus strands, respectively, or vice-versa) and contain a short region in between.
  • amplification primers are from about 10 to 30 nucleotides in length and flank a region from about 50 to 200 nucleotides in length. Under appropriate conditions and with appropriate reagents, such primers permit the amplification of a nucleic acid molecule comprising the nucleotide sequence flanked by the primers.
  • the present invention relates to a method for predicting lymph node metastasis in a patient afflicted with solid cancer after radiotherapy treatment, comprising the steps of: i) determining the expression level of VEGF-C in a biological sample obtained from said patient, ii) comparing the expression level determined at step i) with a predetermined reference value and iii) concluding that the patient will not have a lymph node metastasis when the level determined at step i) is lower than the predetermined reference value or concluding that the patient will have a lymph node metastasis when the level determined at step i) is higher than the predetermined reference value.
  • the present invention relates to a method for predicting cancer relapse in a patient afflicted with solid cancer after radiotherapy treatment, comprising the steps of: i) determining the expression level of VEGF-C in a biological sample obtained from said patient, ii) comparing the expression level determined at step i) with a predetermined reference value and iii) concluding that the patient will not have a cancer relapse when the level determined at step i) is lower than the predetermined reference value or concluding that the patient will have a cancer relapse when the level determined at step i) is higher than the predetermined reference value.
  • the present invention relates to a method for predicting the survival time of a patient afflicted with solid cancer after radiotherapy treatment, comprising the steps of: i) determining the expression level of VEGF-C in a biological sample obtained from the patient, ii) comparing the expression level determined at step i) with a predetermined reference value and iii) concluding that the patient will have a long survival time when the level determined at step i) is lower than the predetermined reference value or concluding that the patient will have a short survival time when the level determined at step i) is higher than the predetermined reference value.
  • the method of the present invention is particularly suitable for predicting the duration of the overall survival (OS), progression-free survival (PFS) and/or the disease-free survival (DFS) of the cancer patient.
  • OS survival time is generally based on and expressed as the percentage of people who survive a certain type of cancer for a specific amount of time.
  • OS rates do not specify whether cancer survivors are still undergoing treatment at five years or if they've become cancer-free (achieved remission).
  • DFS gives more specific information and is the number of people with a particular cancer who achieve remission.
  • progression-free survival (PFS) rates (the number of people who still have cancer, but their disease does not progress) includes people who may have had some success with treatment, but the cancer has not disappeared completely.
  • short survival time indicates that the patient will have a survival time that will be lower than the median (or mean) observed in the general population of patients suffering from said cancer.
  • long survival time indicates that the patient will have a survival time that will be higher than the median (or mean) observed in the general population of patients suffering from said cancer.
  • the method of the invention is performed for predicting the overall survival (OS), progression-free survival (PFS) and/or the disease-free survival (DFS) of a patient afflicted with solid cancer receiving radiotherapy treatment.
  • OS overall survival
  • PFS progression-free survival
  • DFS disease-free survival
  • a further aspect of the invention relates to a method of monitoring cancer progression in a patient afflicted with solid cancer after radiotherapy treatment by performing the method of the invention.
  • a further aspect of the invention relates to a method of monitoring radiotherapy treatment of a patient afflicted with solid cancer by performing the method of the invention.
  • the present invention relates to a VEGF-C inhibitor for use in the treatment of solid cancer in a patient in need thereof wherein the patient was being classified as having a poor prognosis by the method as above described.
  • the solid cancer referred to in the methods and uses described herein above may be for example a head and neck squamous cell carcinoma (FINSCC) or a medulloblastoma (MDB).
  • FINSCC head and neck squamous cell carcinoma
  • MDB medulloblastoma
  • treatment refers to both prophylactic or preventive treatment as well as curative or disease modifying treatment, including treatment of patients at risk of contracting the disease or suspected to have contracted the disease as well as patients who are ill or have been diagnosed as suffering from a disease or medical condition, and includes suppression of clinical relapse.
  • the treatment may be administered to a patient having a medical disorder or who ultimately may acquire the disorder, in order to prevent, cure, delay the onset of, reduce the severity of, or ameliorate one or more symptoms of a disorder or recurring disorder, or in order to prolong the survival of a patient beyond that expected in the absence of such treatment.
  • therapeutic regimen is meant the pattern of treatment of an illness, e.g., the pattern of dosing used during therapy.
  • a therapeutic regimen may include an induction regimen and a maintenance regimen.
  • the phrase “induction regimen” or “induction period” refers to a therapeutic regimen (or the portion of a therapeutic regimen) that is used for the initial treatment of a disease.
  • the general goal of an induction regimen is to provide a high level of drug to a patient during the initial period of a treatment regimen.
  • An induction regimen may employ (in part or in whole) a "loading regimen", which may include administering a greater dose of the drug than a physician would employ during a maintenance regimen, administering a drug more frequently than a physician would administer the drug during a maintenance regimen, or both.
  • maintenance regimen refers to a therapeutic regimen (or the portion of a therapeutic regimen) that is used for the maintenance of a patient during treatment of an illness, e.g., to keep the patient in remission for long periods of time (months or years).
  • a maintenance regimen may employ continuous therapy (e.g., administering a drug at a regular intervals, e.g., weekly, monthly, yearly, etc.) or intermittent therapy (e.g., interrupted treatment, intermittent treatment, treatment at relapse, or treatment upon achievement of a particular predetermined criteria [e.g., disease manifestation, etc.]).
  • VEGF-C inhibitor has its general meaning in the art and refers to a compound that selectively blocks or inactivates the VEGF-C.
  • VEGF-C inhibitor also refers to a compound that selectively blocks the binding of VEGF-C to its receptors (such as VEGFR-3 and VEGFR-2).
  • VEGF-C inhibitor also refers to a compound able to prevent the action of VEGF-C for example by inhibiting the VEGF-C controls of downstream effectors such as inhibiting the activation of the KRAS/RAF/MEK/ER and PI3K/AKT/mTOR pathways.
  • VEGF-C inhibitor also refers to a compound that inhibits VEGF-C expression.
  • a VEGF-C inhibitor is a small organic molecule, a polypeptide, an aptamer, an antibody, an oligonucleotide, a ribozyme or a CRISPR.
  • Tests and assays for determining whether a compound is a VEGF-C inhibitor are well known by the skilled person in the art such as described in WO2011/127519; WO2011/071577; WO0152875; WO2011127519.
  • VEGF-C inhibitors include but are not limited to the anti- VEGF-C antibodies.
  • Anti- VEGF-C antibodies are available and described in the art (such as in WO2011/127519; WO2011/071577; WO2011/127519).
  • anti- VEGF-C antibody which can be used in the context of the invention is identified as "VGX-100" in WO2011/127519 and WO2011/071577.
  • anti- VEGF-C antibodies are described in WO2011/071577, such as in particular those respectively identified as “VC4.5” and “VC1.12” which can, among others, be used in the context of the invention.
  • the VEGF-C inhibitor is a VEGFR-3 antagonist (WO2016/184793). In some embodiments, the VEGF-C inhibitor is a VEGFR-2 antagonist (WO2016/184793). In another embodiment, the VEGF-C inhibitor of the invention is an antibody (the term including "antibody portion") directed against VEGF-C, VEGFR-3 or VEGFR-2.
  • the antibody is a monoclonal antibody. In one embodiment of the antibodies or portions thereof described herein, the antibody is a polyclonal antibody. In one embodiment of the antibodies or portions thereof described herein, the antibody is a humanized antibody. In one embodiment of the antibodies or portions thereof described herein, the antibody is a chimeric antibody. In one embodiment of the antibodies or portions thereof described herein, the portion of the antibody comprises a light chain of the antibody. In one embodiment of the antibodies or portions thereof described herein, the portion of the antibody comprises a heavy chain of the antibody. In one embodiment of the antibodies or portions thereof described herein, the portion of the antibody comprises a Fab portion of the antibody.
  • the portion of the antibody comprises a F(ab')2 portion of the antibody. In one embodiment of the antibodies or portions thereof described herein, the portion of the antibody comprises a Fc portion of the antibody. In one embodiment of the antibodies or portions thereof described herein, the portion of the antibody comprises a Fv portion of the antibody. In one embodiment of the antibodies or portions thereof described herein, the portion of the antibody comprises a variable domain of the antibody. In one embodiment of the antibodies or portions thereof described herein, the portion of the antibody comprises one or more CDR domains of the antibody.
  • antibody includes both naturally occurring and non-naturally occurring antibodies. Specifically, “antibody” includes polyclonal and monoclonal antibodies, and monovalent and divalent fragments thereof. Furthermore, “antibody” includes chimeric antibodies, wholly synthetic antibodies, single chain antibodies, and fragments thereof. The antibody may be a human or nonhuman antibody. A nonhuman antibody may be humanized by recombinant methods to reduce its immunogenicity in man.
  • Antibodies are prepared according to conventional methodology. Monoclonal antibodies may be generated using the method of Kohler and Milstein (Nature, 256:495, 1975). To prepare monoclonal antibodies useful in the invention, a mouse or other appropriate host animal is immunized at suitable intervals (e.g., twice-weekly, weekly, twice-monthly or monthly) with antigenic forms of VEGF-C. The animal may be administered a final "boost" of antigen within one week of sacrifice. It is often desirable to use an immunologic adjuvant during immunization.
  • Suitable immunologic adjuvants include Freund's complete adjuvant, Freund's incomplete adjuvant, alum, Ribi adjuvant, Hunter's Titermax, saponin adjuvants such as QS21 or Quil A, or CpG-containing immunostimulatory oligonucleotides.
  • Other suitable adjuvants are well-known in the field.
  • the animals may be immunized by subcutaneous, intraperitoneal, intramuscular, intravenous, intranasal or other routes. A given animal may be immunized with multiple forms of the antigen by multiple routes.
  • the antigen may be provided as synthetic peptides corresponding to antigenic regions of interest in VEGF-C.
  • lymphocytes are isolated from the spleen, lymph node or other organ of the animal and fused with a suitable myeloma cell line using an agent such as polyethylene glycol to form a hydridoma.
  • cells are placed in media permissive for growth of hybridomas but not the fusion partners using standard methods, as described (Coding, Monoclonal Antibodies: Principles and Practice: Production and Application of Monoclonal Antibodies in Cell Biology, Biochemistry and Immunology, 3rd edition, Academic Press, New York, 1996).
  • cell supernatants are analyzed for the presence of antibodies of the desired specificity, i.e., that selectively bind the antigen.
  • Suitable analytical techniques include ELISA, flow cytometry, immunoprecipitation, and western blotting. Other screening techniques are well-known in the field. Preferred techniques are those that confirm binding of antibodies to conformationally intact, natively folded antigen, such as non-denaturing ELISA, flow cytometry, and immunoprecipitation.
  • an antibody from which the pFc' region has been enzymatically cleaved, or which has been produced without the pFc' region designated an F(ab')2 fragment
  • an antibody from which the Fc region has been enzymatically cleaved, or which has been produced without the Fc region designated as Fab fragment
  • Fab fragments consist of a covalently bound antibody light chain and a portion of the antibody heavy chain denoted Fd.
  • the Fd fragments are the major determinant of antibody specificity (a single Fd fragment may be associated with up to ten different light chains without altering antibody specificity) and Fd fragments retain epitope-binding ability in isolation.
  • CDRs complementarity determining regions
  • FRs framework regions
  • CDR1 through CDRS complementarity determining regions
  • compositions and methods that include humanized forms of antibodies.
  • humanized describes antibodies wherein some, most or all of the amino acids outside the CDR regions are replaced with corresponding amino acids derived from human immunoglobulin molecules.
  • Methods of humanization include, but are not limited to, those described in U.S. Pat. Nos. 4,816,567, 5,225,539, 5,585,089, 5,693,761, 5,693,762 and 5,859,205, which are hereby incorporated by reference.
  • the above U.S. Pat. Nos. 5,585,089 and 5,693,761, and WO 90/07861 also propose four possible criteria which may used in designing the humanized antibodies.
  • the first proposal was that for an acceptor to use a framework from a particular human immunoglobulin that is unusually homologous to the donor immunoglobulin to be humanized, or to use a consensus framework from many human antibodies.
  • the second proposal was that if an amino acid in the framework of the human immunoglobulin is unusual and the donor amino acid at that position is typical for human sequences, then the donor amino acid rather than the acceptor may be selected.
  • the third proposal was that in the positions immediately adjacent to the 3 CDRs in the humanized immunoglobulin chain, the donor amino acid rather than the acceptor amino acid may be selected.
  • the fourth proposal was to use the donor amino acid residue at the framework positions at which the amino acid is predicted to have a side chain atom within 3 A of the CDRs in a three dimensional model of the antibody and is predicted to be capable of interacting with the CDRs.
  • the above methods are merely illustrative of some of the methods that one skilled in the art could employ to make humanized antibodies.
  • One of ordinary skill in the art will be familiar with other methods for antibody humanization.
  • humanized forms of the antibodies some, most or all of the amino acids outside the CDR regions have been replaced with amino acids from human immunoglobulin molecules but where some, most or all amino acids within one or more CDR regions are unchanged. Small additions, deletions, insertions, substitutions or modifications of amino acids are permissible as long as they would not abrogate the ability of the antibody to bind a given antigen.
  • Suitable human immunoglobulin molecules would include IgGl, IgG2, IgG3, IgG4, IgA and IgM molecules.
  • a "humanized" antibody retains a similar antigenic specificity as the original antibody.
  • the affinity and/or specificity of binding of the antibody may be increased using methods of "directed evolution", as described by Wu et al, /. Mol. Biol. 294: 151, 1999, the contents of which are incorporated herein by reference.
  • Fully human monoclonal antibodies also can be prepared by immunizing mice transgenic for large portions of human immunoglobulin heavy and light chain loci. See, e.g., U.S. Pat. Nos. 5,591,669, 5,598,369, 5,545,806, 5,545,807, 6,150,584, and references cited therein, the contents of which are incorporated herein by reference. These animals have been genetically modified such that there is a functional deletion in the production of endogenous (e.g., murine) antibodies. The animals are further modified to contain all or a portion of the human germ-line immunoglobulin gene locus such that immunization of these animals will result in the production of fully human antibodies to the antigen of interest.
  • monoclonal antibodies can be prepared according to standard hybridoma technology. These monoclonal antibodies will have human immunoglobulin amino acid sequences and therefore will not provoke human anti-mouse antibody (KAMA) responses when administered to humans.
  • KAMA human anti-mouse antibody
  • the present invention also provides for F(ab') 2 Fab, Fv and Fd fragments; chimeric antibodies in which the Fc and/or FR and/or CDR1 and/or CDR2 and/or light chain CDR3 regions have been replaced by homologous human or non-human sequences; chimeric F(ab')2 fragment antibodies in which the FR and/or CDR1 and/or CDR2 and/or light chain CDR3 regions have been replaced by homologous human or non-human sequences; chimeric Fab fragment antibodies in which the FR and/or CDR1 and/or CDR2 and/or light chain CDR3 regions have been replaced by homologous human or non-human sequences; and chimeric Fd fragment antibodies in which the FR and/or CDR1 and/or CDR2 regions have been replaced by homologous human or non-human sequences.
  • the present invention also includes so-called single chain antibodies.
  • the various antibody molecules and fragments may derive from any of the commonly known immunoglobulin classes, including but not limited to IgA, secretory IgA, IgE, IgG and IgM.
  • IgG subclasses are also well known to those in the art and include but are not limited to human IgGl, IgG2, IgG3 and IgG4.
  • the VEGF-C inhibitor of the invention is a Human IgG4.
  • the invention provides a multi-specific antibody comprising a first antigen binding site from an antibody of the present invention directed against VEGF-C, VEGFR-3 or VEGFR-2 and at least one second antigen binding site.
  • the second antigen-binding site is used for recruiting a killing mechanism such as, for example, by binding an antigen on a human effector cell as a BiTE (Bispecific T-Cell engager) antibody which is a bispecific scFv2 directed against target antigen and CD3 on T cells described in US7235641, or by binding a cytotoxic agent or a second therapeutic agent.
  • a killing mechanism such as, for example, by binding an antigen on a human effector cell as a BiTE (Bispecific T-Cell engager) antibody which is a bispecific scFv2 directed against target antigen and CD3 on T cells described in US7235641, or by binding a cytotoxic agent or a second therapeutic agent.
  • effector cell refers to an immune cell which is involved in the effector phase of an immune response, as opposed to the cognitive and activation phases of an immune response.
  • Exemplary immune cells include a cell of a myeloid or lymphoid origin, for instance lymphocytes (such as B cells and T cells including cytolytic T cells (CTLs)), killer cells, natural killer cells, macrophages, monocytes, mast cells and granulocytes, such as neutrophils, eosinophils and basophils.
  • lymphocytes such as B cells and T cells including cytolytic T cells (CTLs)
  • CTLs cytolytic T cells
  • FcRs Fc receptors
  • an effector cell is capable of inducing ADCC, such as a natural killer cell.
  • monocytes, macrophages, which express FcRs are involved in specific killing of target cells and presenting antigens to other components of the immune system.
  • an effector cell may phagocytose a target antigen or target cell.
  • the expression of a particular FcR on an effector cell may be regulated by humoral factors such as cytokines.
  • An effector cell can phagocytose a target antigen or phagocytose or lyse a target cell.
  • Suitable cytotoxic agents and second therapeutic agents are exemplified below, and include toxins (such as radiolabeled peptides), chemotherapeutic agents and prodrugs
  • the second antigen-binding site binds a tissue- specific antigen, promoting localization of the bispecific antibody to a specific tissue.
  • the second antigen-binding site binds to an antigen located on the same type of cell as the [VEGF-C, VEGFR-3 or VEGFR-2] -expressing cell, typically a tumor-associated antigen (TAA), but has a binding specificity different from that of the first antigen-binding site.
  • TAA tumor-associated antigen
  • Such multi- or bispecific antibodies can enhance the specificity of the tumor cell binding and/or engage multiple effector pathways.
  • TAAs include carcinoembryonic antigen (CEA), prostate specific antigen (PSA), RAGE (renal antigen), a- fetoprotein, CAMEL (CTL-recognized antigen on melanoma), CT antigens (such as MAGE- B5, -B6, -C2, -C3, and D; Mage-12; CT10; NY-ESO-1, SSX-2, GAGE, BAGE, MAGE, and SAGE), mucin antigens (e.g., MUC1, mucin-CA125, etc.), ganglioside antigens, tyrosinase, gp75, c-Met, Marti, MelanA, MUM-1, MUM-2, MUM-3, HLA-B7, Ep-CAM or a cancer- associated integrin, such as ⁇ 5 ⁇ 3 integrin.
  • CEA carcinoembryonic antigen
  • PSA prostate specific antigen
  • RAGE renal antigen
  • the second antigen- binding site binds to a different epitope of [VEGF-C, VEGFR-3 or VEGFR-2].
  • the second antigen- binding site may alternatively bind an angiogenic factor or other cancer-associated growth factor, such as a vascular endothelial growth factor, a fibroblast growth factor, epidermal growth factor, angiogenin or a receptor of any of these, particularly receptors associated with cancer progression.
  • the second antigen-binding site is from a second antibody or ADCC of the invention, such as the antibody of the present invention.
  • Exemplary formats for the multispecific antibody molecules of the invention include, but are not limited to (i) two antibodies cross-linked by chemical heteroconjugation, one with a specificity to [VEGF-C, VEGFR-3 or VEGFR-2] and another with a specificity to a second antigen; (ii) a single antibody that comprises two different antigen-binding regions; (iii) a single-chain antibody that comprises two different antigen-binding regions, e.g., two scFvs linked in tandem by an extra peptide linker; (iv) a dual-variable-domain antibody (DVD-Ig), where each light chain and heavy chain contains two variable domains in tandem through a short peptide linkage (Wu et al., Generation and Characterization of a Dual Variable Domain Immunoglobulin (DVD-IgTM) Molecule, In : Antibody Engineering, Springer Berlin Heidelberg (2010)); (v) a chemically- linked bispecific (Fab')2 fragment; (vi)
  • IgG-like molecules with complementary CH3 domains to force heterodimerization is IgG-like molecules with complementary CH3 domains to force heterodimerization.
  • Such molecules can be prepared using known technologies, such as, e.g., those known as Triomab/Quadroma (Trion Pharma/Fresenius Biotech), Knob-into-Hole (Genentech), CrossMAb (Roche) and electrostatically-matched (Amgen), LUZ-Y (Genentech), Strand Exchange Engineered Domain body (SEEDbody)(EMD Serono), Biclonic (Merus) and DuoBody (Genmab A/S) technologies.
  • the bispecific antibody is obtained or obtainable via a controlled Fab-arm exchange, typically using DuoBody technology.
  • a controlled Fab-arm exchange typically using DuoBody technology.
  • In vitro methods for producing bispecific antibodies by controlled Fab-arm exchange have been described in WO2008119353 and WO 2011131746 (both by Genmab A/S).
  • a bispecific antibody is formed by "Fab-arm" or "half- molecule” exchange (swapping of a heavy chain and attached light chain) between two monospecific antibodies, both comprising IgG4-like CH3 regions, upon incubation under reducing conditions.
  • the resulting product is a bispecific antibody having two Fab arms which may comprise different sequences.
  • bispecific antibodies of the present invention are prepared by a method comprising the following steps, wherein at least one of the first and second antibodies is the antibody of the present invention : a) providing a first antibody comprising an Fc region of an immunoglobulin, said Fc region comprising a first CH3 region; b) providing a second antibody comprising an Fc region of an immunoglobulin, said Fc region comprising a second CH3 region; wherein the sequences of said first and second CH3 regions are different and are such that the heterodimeric interaction between said first and second CH3 regions is stronger than each of the homodimeric interactions of said first and second CH3 regions; c) incubating said first antibody together with said second antibody under reducing conditions; and d) obtaining said bispecific antibody, wherein the first antibody is the antibody of the present invention and the second antibody has a different binding specificity, or vice versa.
  • the reducing conditions may, for example, be provided by adding a reducing agent, e.g. selected from 2-mercaptoethylamine, dithiothreitol and tris(2-carboxyethyl)phosphine.
  • Step d) may further comprise restoring the conditions to become non-reducing or less reducing, for example by removal of a reducing agent, e.g. by desalting.
  • the sequences of the first and second CH3 regions are different, comprising only a few, fairly conservative, asymmetrical mutations, such that the heterodimeric interaction between said first and second CH3 regions is stronger than each of the homodimeric interactions of said first and second CH3 regions.
  • the antibody according to the invention is a single domain antibody.
  • the term “single domain antibody” (sdAb) or “VHH” refers to the single heavy chain variable domain of antibodies of the type that can be found in Camelid mammals which are naturally devoid of light chains. Such VHH are also called “nanobody®”. According to the invention, sdAb can particularly be llama sdAb.
  • VHH refers to the single heavy chain having 3 complementarity determining regions (CDRs): CDR1, CDR2 and CDR3.
  • CDRs complementarity determining region
  • CDR complementarity determining region
  • VHH according to the invention can readily be prepared by an ordinarily skilled artisan using routine experimentation.
  • VHH variants and modified form thereof may be produced under any known technique in the art such as in- vitro maturation.
  • VHHs or sdAbs are usually generated by PCR cloning of the V-domain repertoire from blood, lymph node, or spleen cDNA obtained from immunized animals into a phage display vector, such as pHEN2.
  • Antigen- specific VHHs are commonly selected by panning phage libraries on immobilized antigen, e.g., antigen coated onto the plastic surface of a test tube, biotinylated antigens immobilized on streptavidin beads, or membrane proteins expressed on the surface of cells.
  • immobilized antigen e.g., antigen coated onto the plastic surface of a test tube, biotinylated antigens immobilized on streptavidin beads, or membrane proteins expressed on the surface of cells.
  • VHHs often show lower affinities for their antigen than VHHs derived from animals that have received several immunizations.
  • VHHs from immune libraries are attributed to the natural selection of variant VHHs during clonal expansion of B-cells in the lymphoid organs of immunized animals.
  • the affinity of VHHs from non-immune libraries can often be improved by mimicking this strategy in vitro, i.e., by site directed mutagenesis of the CDR regions and further rounds of panning on immobilized antigen under conditions of increased stringency (higher temperature, high or low salt concentration, high or low pH, and low antigen concentrations).
  • VHHs derived from camelid are readily expressed in and purified from the E. coli periplasm at much higher levels than the corresponding domains of conventional antibodies.
  • VHHs generally display high solubility and stability and can also be readily produced in yeast, plant, and mammalian cells.
  • the "Hamers patents” describe methods and techniques for generating VHH against any desired target (see for example US 5,800,988; US 5,874, 541 and US 6,015,695).
  • the "Hamers patents” more particularly describe production of VHHs in bacterial hosts such as E. coli (see for example US 6,765,087) and in lower eukaryotic hosts such as moulds (for example Aspergillus or Trichoderma) or in yeast (for example Saccharomyces, Kluyveromyces, Hansenula or Pichia) (see for example US 6,838,254).
  • the VEGF-C inhibitor of the invention is a VEGF-C expression inhibitor, VEGFR-3 and/or VEGFR-2 expression inhibitor.
  • a gene product can be the direct transcriptional product of a gene (e.g., mRNA, tRNA, rRNA, antisense RNA, ribozyme, structural RNA or any other type of RNA) or a protein produced by translation of a mRNA.
  • Gene products also include messenger RNAs, which are modified, by processes such as capping, polyadenylation, methylation, and editing, and proteins (e.g., VEGF-C, VEGFR-3 and VEGFR-2) modified by, for example, methylation, acetylation, phosphorylation, ubiquitination, SUMOylation, ADP-ribosylation, myristilation, and glycosylation.
  • proteins e.g., VEGF-C, VEGFR-3 and VEGFR-2
  • proteins e.g., VEGF-C, VEGFR-3 and VEGFR-2
  • an “inhibitor of expression” refers to a natural or synthetic compound that has a biological effect to inhibit the expression of a gene.
  • VEGF-C, VEGFR-3 and VEGFR-2 expression inhibitors for use in the present invention may be based on antisense oligonucleotide constructs.
  • Anti-sense oligonucleotides including anti-sense RNA molecules and anti-sense DNA molecules, would act to directly block the translation of VEGF-C, VEGFR-3 and VEGFR-2 mRNA by binding thereto and thus preventing protein translation or increasing mRNA degradation, thus decreasing the level of VEGF-C, VEGFR-3 and VEGFR-2 proteins, and thus activity, in a cell.
  • antisense oligonucleotides of at least about 15 bases and complementary to unique regions of the mRNA transcript sequence encoding VEGF-C, VEGFR-3 and VEGFR-2 can be synthesized, e.g., by conventional phosphodiester techniques and administered by e.g., intravenous injection or infusion.
  • Methods for using antisense techniques for specifically alleviating gene expression of genes whose sequence is known are well known in the art (e.g. see U.S. Pat. Nos. 6,566,135; 6,566,131; 6,365,354; 6,410,323; 6,107,091; 6,046,321; and 5,981,732).
  • Small inhibitory RNAs can also function as expression inhibitors for use in the present invention.
  • Gene expression can be reduced by contacting the subject or cell with a small double stranded RNA (dsRNA), or a vector or construct causing the production of a small double stranded RNA, such that VEGF-C, VEGFR-3 and VEGFR-2 expression is specifically inhibited (i.e. RNA interference or RNAi).
  • dsRNA small double stranded RNA
  • Methods for selecting an appropriate dsRNA or dsRNA-encoding vector are well known in the art for genes whose sequence is known (e.g. see Tuschl, T. et al. (1999); Elbashir, S. M. et al. (2001); Hannon, GJ.
  • Ribozymes can also function as expression inhibitors for use in the present invention.
  • Ribozymes are enzymatic RNA molecules capable of catalyzing the specific cleavage of RNA.
  • the mechanism of ribozyme action involves sequence specific hybridization of the ribozyme molecule to complementary target RNA, followed by endonucleo lytic cleavage.
  • Engineered hairpin or hammerhead motif ribozyme molecules that specifically and efficiently catalyze endonucleo lytic cleavage of mRNA sequences are thereby useful within the scope of the present invention.
  • ribozyme cleavage sites within any potential RNA target are initially identified by scanning the target molecule for ribozyme cleavage sites, which typically include the following sequences, GUA, GUU, and GUC. Once identified, short RNA sequences of between about 15 and 20 ribonucleotides corresponding to the region of the target gene containing the cleavage site can be evaluated for predicted structural features, such as secondary structure, that can render the oligonucleotide sequence unsuitable. The suitability of candidate targets can also be evaluated by testing their accessibility to hybridization with complementary oligonucleotides, using, e.g., ribonuclease protection assays.
  • antisense oligonucleotides and ribozymes useful as VEGF-C, VEGFR-3 and VEGFR-2 inhibitors can be prepared by known methods. These include techniques for chemical synthesis such as, e.g., by solid phase phosphoramadite chemical synthesis. Alternatively, anti-sense RNA molecules can be generated by in vitro or in vivo transcription of DNA sequences encoding the RNA molecule. Such DNA sequences can be incorporated into a wide variety of vectors that incorporate suitable RNA polymerase promoters such as the T7 or SP6 polymerase promoters. Various modifications to the oligonucleotides of the invention can be introduced as a means of increasing intracellular stability and half-life.
  • Antisense oligonucleotides siRNAs and ribozymes of the invention may be delivered in vivo alone or in association with a vector.
  • a "vector” is any vehicle capable of facilitating the transfer of the antisense oligonucleotide siRNA or ribozyme nucleic acid to the cells and preferably cells expressing VEGF-C, VEGFR-3 and VEGFR-2.
  • the vector transports the nucleic acid to cells with reduced degradation relative to the extent of degradation that would result in the absence of the vector.
  • the vectors useful in the invention include, but are not limited to, plasmids, phagemids, viruses, other vehicles derived from viral or bacterial sources that have been manipulated by the insertion or incorporation of the antisense oligonucleotide siRNA or ribozyme nucleic acid sequences.
  • Viral vectors are a preferred type of vector and include, but are not limited to nucleic acid sequences from the following viruses: retrovirus, such as moloney murine leukemia virus, harvey murine sarcoma virus, murine mammary tumor virus, and rouse sarcoma virus; adenovirus, adeno-associated virus; SV40-type viruses; polyoma viruses; Epstein-Barr viruses; papilloma viruses; herpes virus; vaccinia virus; polio virus; and RNA virus such as a retrovirus.
  • retrovirus such as moloney murine leukemia virus, harvey murine sarcoma virus, murine mammary tumor virus, and rouse sarcoma virus
  • retrovirus such as moloney murine leukemia virus, harvey murine sarcoma virus, murine mammary tumor virus, and rouse sarcoma virus
  • adenovirus adeno
  • Non-cytopathic viruses include retroviruses (e.g., lentivirus), the life cycle of which involves reverse transcription of genomic viral RNA into DNA with subsequent proviral integration into host cellular DNA. Retroviruses have been approved for human gene therapy trials. Most useful are those retroviruses that are replication-deficient (i.e., capable of directing synthesis of the desired proteins, but incapable of manufacturing an infectious particle). Such genetically altered retroviral expression vectors have general utility for the high-efficiency transduction of genes in vivo.
  • adeno-viruses and adeno-associated viruses are double-stranded DNA viruses that have already been approved for human use in gene therapy.
  • the adeno-associated virus can be engineered to be replication deficient and is capable of infecting a wide range of cell types and species. It further has advantages such as, heat and lipid solvent stability; high transduction frequencies in cells of diverse lineages, including hematopoietic cells; and lack of superinfection inhibition thus allowing multiple series of transductions.
  • the adeno-associated virus can integrate into human cellular DNA in a site-specific manner, thereby minimizing the possibility of insertional mutagenesis and variability of inserted gene expression characteristic of retroviral infection.
  • adeno-associated virus infections have been followed in tissue culture for greater than 100 passages in the absence of selective pressure, implying that the adeno-associated virus genomic integration is a relatively stable event.
  • the adeno- associated virus can also function in an extrachromosomal fashion.
  • Plasmid vectors have been extensively described in the art and are well known to those of skill in the art. See e.g., SANBROOK et al, "Molecular Cloning: A Laboratory Manual," Second Edition, Cold Spring Harbor Laboratory Press, 1989. In the last few years, plasmid vectors have been used as DNA vaccines for delivering antigen-encoding genes to cells in vivo. They are particularly advantageous for this because they do not have the same safety concerns as with many of the viral vectors. These plasmids, however, having a promoter compatible with the host cell, can express a peptide from a gene operatively encoded within the plasmid.
  • Plasmids may be delivered by a variety of parenteral, mucosal and topical routes.
  • the DNA plasmid can be injected by intramuscular, intradermal, subcutaneous, or other routes. It may also be administered by intranasal sprays or drops, rectal suppository and orally.
  • the plasmids may be given in an aqueous solution, dried onto gold particles or in association with another DNA delivery system including but not limited to liposomes, dendrimers, cochleate and microencapsulation.
  • VEGF-C, VEGFR-3 and VEGFR-2 expression inhibitors include but are not limited to siRNAs and shRNA such as described in Liu et al, 2015 and Ye et al, 2015.
  • inhibitors according to the invention as described above are administered to the patient in a therapeutically effective amount.
  • a “therapeutically effective amount” of the inhibitor of the present invention as above described is meant a sufficient amount of the inhibitor for treating solid cancer at a reasonable benefit/risk ratio applicable to any medical treatment. It will be understood, however, that the total daily usage of the inhibitors and compositions of the present invention will be decided by the attending physician within the scope of sound medical judgment.
  • the specific therapeutically effective dose level for any particular patient will depend upon a variety of factors including the disorder being treated and the severity of the disorder; activity of the specific inhibitor employed; the specific composition employed, the age, body weight, general health, sex and diet of the patient; the time of administration, route of administration, and rate of excretion of the specific inhibitor employed; the duration of the treatment; drugs used in combination or coincidential with the specific inhibitor employed; and like factors well known in the medical arts.
  • the daily dosage of the products may be varied over a wide range from 0.01 to 1,000 mg per adult per day.
  • the compositions contain 0.01, 0.05, 0.1, 0.5, 1.0, 2.5, 5.0, 10.0, 15.0, 25.0, 50.0, 100, 250 and 500 mg of the inhibitor of the present invention for the symptomatic adjustment of the dosage to the patient to be treated.
  • a medicament typically contains from about 0.01 mg to about 500 mg of the inhibitor of the present invention, preferably from 1 mg to about 100 mg of the inhibitor of the present invention.
  • An effective amount of the drug is ordinarily supplied at a dosage level from 0.0002 mg/kg to about 20 mg/kg of body weight per day, especially from about 0.001 mg/kg to 7 mg/kg of body weight per day.
  • the compound according to the invention may be used in a concentration between 0.01 ⁇ and 20 ⁇ , particularly, the compound of the invention may be used in a concentration of 0.01, 0.05, 0.1, 0.5, 1.0, 2.5, 5.0, 10.0, 15.0, 20.0 ⁇ .
  • the present invention relates to the compound according to the invention for use in the treatment of solid cancer in a patient in need thereof in combination with radiotherapy treatment wherein the patient was being classified as having a poor prognosis by the method as above described.
  • the VEGF-C inhibitor of the invention is administered sequentially or concomitantly with the radiotherapy treatment, herein defined as a photon (X) radiotherapy or as a proton (P) radiotherapy.
  • the radiotherapy treatment herein defined as a photon (X) radiotherapy or as a proton (P) radiotherapy.
  • the present invention relates to the compound according to the invention in combination with one or more anti-cancer compound for use in the treatment of solid cancer in a patient in need thereof wherein the patient was being classified as having a poor prognosis by the method as above described.
  • the present invention relates to the compound according to the invention in combination with one or more anti-angiogenic compound or anti- lymphangiogenic compound for use in the treatment of solid cancer in a patient in need thereof wherein the patient was being classified as having a poor prognosis by the method as above described.
  • anti-cancer compound has its general meaning in the art and refers to compounds used in anti-cancer therapy such as anti-angiogenic compound, tyrosine kinase inhibitors, tyrosine kinase receptor (TKR) inhibitors, Vascular Endothelial Growth Factor Receptors (VEGFRs) pathway inhibitors, interferon therapy, anti-HER2 compounds, anti- EGFR compounds, alkylating agents, anti-metabolites, immunotherapeutic agents, Interferons (IFNs), Interleukins, and chemotherapeutic agents such as described below.
  • anti-angiogenic compound has its general meaning in the art and refers to compounds used in anti-angiogenic therapy such as tyrosine kinase inhibitors, anti-angiogenic tyrosine kinase receptor (TKR) inhibitors, anti-angiogenics targeting the Vascular Endothelial Growth Factor Receptors (VEGFRs) pathway, interferon therapy and anti-HER2 compounds such as Trastuzumab (herceptin) and pertuzumab.
  • anti- angiogenic compound refers to Sunitinib (Sutent), an anti-angiogenic TKR inhibitor of VEGFRs, platelet-derived growth factor receptors (PDGF-Rs), and c-kit.
  • tyrosine kinase inhibitor refers to any of a variety of therapeutic agents or drugs that act as selective or non-selective inhibitors of receptor and/or non-receptor tyrosine kinases. Tyrosine kinase inhibitors and related compounds are well known in the art and described in U.S Patent Publication 2007/0254295, which is incorporated by reference herein in its entirety.
  • a compound related to a tyrosine kinase inhibitor will recapitulate the effect of the tyrosine kinase inhibitor, e.g., the related compound will act on a different member of the tyrosine kinase signaling pathway to produce the same effect as would a tyrosine kinase inhibitor of that tyrosine kinase.
  • tyrosine kinase inhibitors and related compounds suitable for use in methods of embodiments of the present invention include, but are not limited to, sunitinib (Sutent; SU11248), Axitinib, pazopanib (Votrient), cabozantinib, dasatinib (BMS-354825), PP2, BEZ235, saracatinib, gefitinib (Iressa), erlotinib (Tarceva; OSI-1774), lapatinib (GW572016; GW2016), canertinib (CI 1033), semaxinib (SU5416), vatalanib (PTK787/ZK222584), sorafenib (BAY 43-9006), imatinib (Gleevec; STI571), leflunomide (SU101), vandetanib (Zactima; ZD6474), MK-2206 (8-[4-aminocycl
  • the tyrosine kinase inhibitor is a small molecule kinase inhibitor that has been orally administered to the patient of at least one Phase I clinical trial, more preferably at least one Phase II clinical, even more preferably at least one Phase III clinical trial, and most preferably approved by the FDA for at least one hematological or oncological indication.
  • inhibitors include, but are not limited to, Gefitinib, Erlotinib, Lapatinib, Canertinib, BMS-599626 (AC-480), Neratinib, KR -633, CEP-11981, Imatinib, Nilotinib, Dasatinib, AZM-475271, CP-724714, TAK-165, Sunitinib, Vatalanib, CP-547632, Vandetanib, Bosutinib, Lestaurtinib, Tandutinib, Midostaurin, Enzastaurin, AEE- 788, Pazopanib, Axitinib, Motasenib, OSI-930, Cediranib, KR -951, Dovitinib, Seliciclib, SNS-032, PD-0332991, MKC-I (Ro-317453; R-440), Sorafenib, A
  • anti-angiogenic compound refers to compounds targeting the vascular endothelial growth factor (VEGF) pathway such anti-VEGF antibody bevacizumab (Avastin) and VEGF receptor tyrosine kinase inhibitor (TKI) compounds such as sunitinib (Sutent), vandetanib (Zactima), pazopanib (Votrient), sorafenib (Nexavar) and cediranib.
  • VEGF vascular endothelial growth factor
  • TKI VEGF receptor tyrosine kinase inhibitor
  • the compound of the invention is administered sequentially or concomitantly with one or more anti-cancer compound and/or anti-angiogenic compound.
  • the compound of the present invention is administered to the patient in the form of a pharmaceutical composition.
  • the compound of the present invention may be combined with pharmaceutically acceptable excipients, and optionally sustained-release matrices, such as biodegradable polymers, to form therapeutic compositions.
  • pharmaceutically acceptable excipients or “pharmaceutically acceptable” refer to molecular entities and compositions that do not produce an adverse, allergic or other untoward reaction when administered to a mammal, especially a human, as appropriate.
  • a pharmaceutically acceptable carrier or excipient refers to a non-toxic solid, semi-solid or liquid filler, diluent, encapsulating material or formulation auxiliary of any type.
  • the active principle in the pharmaceutical compositions of the present invention for oral, sublingual, subcutaneous, intramuscular, intravenous, transdermal, local or rectal administration, can be administered in a unit administration form, as a mixture with conventional pharmaceutical supports, to animals and human beings.
  • Suitable unit administration forms comprise oral-route forms such as tablets, gel capsules, powders, granules and oral suspensions or solutions, sublingual and buccal administration forms, aerosols, implants, subcutaneous, transdermal, topical, intraperitoneal, intramuscular, intravenous, subdermal, transdermal, intrathecal and intranasal administration forms and rectal administration forms.
  • the pharmaceutical compositions contain vehicles which are pharmaceutically acceptable for a formulation capable of being injected.
  • vehicles which are pharmaceutically acceptable for a formulation capable of being injected.
  • These may be in particular isotonic, sterile, saline solutions (monosodium or disodium phosphate, sodium, potassium, calcium or magnesium chloride and the like or mixtures of such salts), or dry, especially freeze-dried compositions which upon addition, depending on the case, of sterilized water or physiological saline, permit the constitution of injectable solutions.
  • the pharmaceutical forms suitable for injectable use include sterile aqueous solutions or dispersions; formulations including sesame oil, peanut oil or aqueous propylene glycol; and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions.
  • the form In all cases, the form must be sterile and must be fluid to the extent that easy syringability exists. It must be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms, such as bacteria and fungi.
  • Solutions comprising compounds of the invention as free base or pharmacologically acceptable salts can be prepared in water suitably mixed with a surfactant, such as hydroxypropylcellulose. Dispersions can also be prepared in glycerol, liquid polyethylene glycols, and mixtures thereof and in oils. Under ordinary conditions of storage and use, these preparations contain a preservative to prevent the growth of microorganisms.
  • the compound of the present invention can be formulated into a composition in a neutral or salt form.
  • Pharmaceutically acceptable salts include the acid addition salts (formed with the free amino groups of the protein) and which are formed with inorganic acids such as, for example, hydrochloric or phosphoric acids, or such organic acids as acetic, oxalic, tartaric, mandelic, and the like. Salts formed with the free carboxyl groups can also be derived from inorganic bases such as, for example, sodium, potassium, ammonium, calcium, or ferric hydroxides, and such organic bases as isopropylamine, trimethylamine, histidine, procaine and the like.
  • inorganic acids such as, for example, hydrochloric or phosphoric acids, or such organic acids as acetic, oxalic, tartaric, mandelic, and the like.
  • Salts formed with the free carboxyl groups can also be derived from inorganic bases such as, for example, sodium, potassium, ammonium, calcium, or ferric hydroxides, and such organic bases as isopropylamine, trimethylamine,
  • the carrier can also be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyethylene glycol, and the like), suitable mixtures thereof, and vegetables oils.
  • the proper fluidity can be maintained, for example, by the use of a coating, such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants.
  • the prevention of the action of microorganisms can be brought about by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, thimerosal, and the like.
  • isotonic agents for example, sugars or sodium chloride.
  • Prolonged absorption of the injectable compositions can be brought about by the use in the compositions of agents delaying absorption, for example, aluminium monostearate and gelatin.
  • Sterile injectable solutions are prepared by incorporating the active compounds in the required amount in the appropriate solvent with several of the other ingredients enumerated above, as required, followed by filtered sterilization.
  • dispersions are prepared by incorporating the various sterilized agent of the present inventions into a sterile vehicle which contains the basic dispersion medium and the required other ingredients from those enumerated above.
  • sterile powders for the preparation of sterile injectable solutions the typical methods of preparation are vacuum-drying and freeze-drying techniques which yield a powder of the compound of the present invention plus any additional desired ingredient from a previously sterile- filtered solution thereof.
  • the preparation of more, or highly concentrated solutions for direct injection is also contemplated, where the use of DMSO as solvent is envisioned to result in extremely rapid penetration, delivering high concentrations of the active agents to a small tumor area.
  • solutions Upon formulation, solutions will be administered in a manner compatible with the dosage formulation and in such amount as is therapeutically effective.
  • the formulations are easily administered in a variety of dosage forms, such as the type of injectable solutions described above, but drug release capsules and the like can also be employed.
  • aqueous solutions For parenteral administration in an aqueous solution, for example, the solution should be suitably buffered if necessary and the liquid diluent first rendered isotonic with sufficient saline or glucose.
  • aqueous solutions are especially suitable for intravenous, intramuscular, subcutaneous and intraperitoneal administration.
  • sterile aqueous media which can be employed will be known to those of skill in the art in light of the present disclosure. Some variation in dosage will necessarily occur depending on the condition of the patient being treated. The person responsible for administration will, in any event, determine the appropriate dose for the individual patient.
  • the compound of the present invention is administered sequentially or concomitantly with one or more therapeutic active agent such as chemotherapeutic.
  • the compound of the present invention is administered with a chemotherapeutic agent.
  • chemotherapeutic agent refers to chemical compounds that are effective in inhibiting tumor growth.
  • examples of chemotherapeutic agents include alkylating agents such as thiotepa and cyclosphosphamide; alkyl sulfonates such as busulfan, improsulfan and piposulfan; aziridines such as benzodopa, carboquone, meturedopa, and uredopa; ethylenimines and methylamelamines including altretamine, triethylenemelamine, trietylenephosphoramide, triethylenethiophosphaorarnide and trimethylolomelamine; acetogenins (especially bullatacin and bullatacinone); a carnptothecin (including the synthetic analogue topotecan); bryostatin; callystatin; CC-1065 (including its adozeles).
  • calicheamicin especially calicheamicin (11 and calicheamicin 21 1, see, e.g., Agnew Chem Intl. Ed. Engl. 33 : 183-186 (1994); dynemicin, including dynemicin A; an esperamicin; as well as neocarzinostatin chromophore and related chromoprotein enediyne antiobiotic chromomophores), aclacinomysins, actinomycin, authramycin, azaserine, bleomycins, cactinomycin, carabicin, canninomycin, carzinophilin, chromomycins, dactinomycin, daunorubicin, detorubicin, 6-diazo-5-oxo-L-norleucine, doxorubicin (including morpholino-doxorubicin, cyanomorpholino-doxorubicin, 2-pyrrolin
  • paclitaxel (TAXOL®, Bristol-Myers Squibb Oncology, Princeton, N.].) and doxetaxel (TAXOTERE®, Rhone-Poulenc Rorer, Antony, France); chlorambucil; gemcitabine; 6-thioguanine; mercaptopurine; methotrexate; platinum analogs such as cisplatin and carbop latin; vinblastine; platinum; etoposide (VP- 16); ifosfamide; mitomycin C; mitoxantrone; vincristine; vinorelbine; navelbine; novantrone; teniposide; daunomycin; aminopterin; xeloda; ibandronate; CPT-1 1 ; topoisomerase inhibitor RFS 2000; difluoromethylornithine (DMFO); retinoic acid; capecitabine; and pharmaceutically acceptable salts, acids or derivatives of any of the above.
  • anti-estrogens including for example tamoxifen, raloxifene, aromatase inhibiting 4(5)-imidazoles, 4-hydroxytamoxifen, trioxifene, keoxifene, LY117018, onapristone, and toremifene (Fareston); and anti-androgens such as flutamide, nilutamide, bicalutamide, leuprolide, and goserelin; and phannaceutically acceptable salts, acids or derivatives of any of the above.
  • the compound of the present invention is administered with a targeted cancer therapy.
  • Targeted cancer therapies are drugs or other substances that block the growth and spread of cancer by interfering with specific molecules ("molecular targets") that are involved in the growth, progression, and spread of cancer.
  • Targeted cancer therapies are sometimes called “molecularly targeted drugs", “molecularly targeted therapies”, “precision medicines”, or similar names.
  • the targeted therapy consists of administering the patient with a tyrosine kinase inhibitor as defined above.
  • compound of the present invention is administered with an immunotherapeutic agent.
  • immunotherapeutic agent refers to a compound, composition or treatment that indirectly or directly enhances, stimulates or increases the body's immune response against cancer cells and/or that decreases the side effects of other anticancer therapies. Immunotherapy is thus a therapy that directly or indirectly stimulates or enhances the immune system's responses to cancer cells and/or lessens the side effects that may have been caused by other anti-cancer agents. Immunotherapy is also referred to in the art as immunologic therapy, biological therapy biological response modifier therapy and biotherapy. Examples of common immunotherapeutic agents known in the art include, but are not limited to, cytokines, cancer vaccines, monoclonal antibodies and non- cytokine adjuvants.
  • the immunotherapeutic treatment may consist of administering the patient with an amount of immune cells (T cells, NK, cells, dendritic cells, B cells).
  • Immunotherapeutic agents can be non-specific, i.e. boost the immune system generally so that the human body becomes more effective in fighting the growth and/or spread of cancer cells, or they can be specific, i.e. targeted to the cancer cells themselves immunotherapy regimens may combine the use of non-specific and specific immunotherapeutic agents.
  • Non-specific immunotherapeutic agents are substances that stimulate or indirectly improve the immune system.
  • Non-specific immunotherapeutic agents have been used alone as a main therapy for the treatment of cancer, as well as in addition to a main therapy, in which case the non-specific immunotherapeutic agent functions as an adjuvant to enhance the effectiveness of other therapies (e.g. cancer vaccines).
  • Non-specific immunotherapeutic agents can also function in this latter context to reduce the side effects of other therapies, for example, bone marrow suppression induced by certain chemotherapeutic agents.
  • Non-specific immunotherapeutic agents can act on key immune system cells and cause secondary responses, such as increased production of cytokines and immunoglobulins. Alternatively, the agents can themselves comprise cytokines.
  • Non-specific immunotherapeutic agents are generally classified as cytokines or non-cytokine adjuvants.
  • cytokines have found application in the treatment of cancer either as general nonspecific immunotherapies designed to boost the immune system, or as adjuvants provided with other therapies.
  • Suitable cytokines include, but are not limited to, interferons, interleukins and colony- stimulating factors.
  • Interferons (IFNs) contemplated by the present invention include the common types of IFNs, IFN-alpha (IFN-a), IFN-beta (IFN- ⁇ ) and IFN- gamma (IFN- ⁇ ).
  • IFNs can act directly on cancer cells, for example, by slowing their growth, promoting their development into cells with more normal behaviour and/or increasing their production of antigens thus making the cancer cells easier for the immune system to recognise and destroy.
  • IFNs can also act indirectly on cancer cells, for example, by slowing down angiogenesis, boosting the immune system and/or stimulating natural killer (NK) cells, T cells and macrophages.
  • Recombinant IFN-alpha is available commercially as Roferon (Roche Pharmaceuticals) and Intron A (Schering Corporation).
  • Interleukins contemplated by the present invention include IL-2, IL-4, IL-11 and IL-12. Examples of commercially available recombinant interleukins include Proleukin® (IL-2; Chiron Corporation) and Neumega® (IL- 12; Wyeth Pharmaceuticals). Zymogenetics, Inc.
  • Colony- stimulating factors contemplated by the present invention include granulocyte colony stimulating factor (G-CSF or filgrastim), granulocyte-macrophage colony stimulating factor (GM-CSF or sargramostim) and erythropoietin (epoetin alfa, darbepoietin). Treatment with one or more growth factors can help to stimulate the generation of new blood cells in patients undergoing traditional chemotherapy.
  • CSFs can be helpful in decreasing the side effects associated with chemotherapy and can allow for higher doses of chemo therapeutic agents to be used.
  • Various-recombinant colony stimulating factors are available commercially, for example, Neupogen® (G-CSF; Amgen), Neulasta (pelfilgrastim; Amgen), Leukine (GM-CSF; Berlex), Procrit (erythropoietin; Ortho Biotech), Epogen (erythropoietin; Amgen), Arnesp (erytropoietin).
  • immunotherapeutic agents can be active, i.e. stimulate the body's own immune response, or they can be passive, i.e.
  • Passive specific immunotherapy typically involves the use of one or more monoclonal antibodies that are specific for a particular antigen found on the surface of a cancer cell or that are specific for a particular cell growth factor.
  • Monoclonal antibodies may be used in the treatment of cancer in a number of ways, for example, to enhance a patient's immune response to a specific type of cancer, to interfere with the growth of cancer cells by targeting specific cell growth factors, such as those involved in angiogenesis, or by enhancing the delivery of other anticancer agents to cancer cells when linked or conjugated to agents such as chemotherapeutic agents, radioactive particles or toxins.
  • Monoclonal antibodies currently used as cancer immunotherapeutic agents that are suitable for inclusion in the combinations of the present invention include, but are not limited to, rituximab (Rituxan®), trastuzumab (Herceptin®), ibritumomab tiuxetan (Zevalin®), tositumomab (Bexxar®), cetuximab (C-225, Erbitux®), bevacizumab (Avastin®), gemtuzumab ozogamicin (Mylotarg®), alemtuzumab (Campath®), and BL22.
  • Other examples include anti-CTLA4 antibodies (e.g.
  • antibodies include B cell depleting antibodies.
  • Typical B cell depleting antibodies include but are not limited to anti-CD20 monoclonal antibodies [e.g.
  • the immunotherapeutic treatment may consist of allografting, in particular, allograft with hematopoietic stem cell HSC.
  • the immunotherapeutic treatment may also consist in an adoptive immunotherapy as described by Nicholas P. Restifo, Mark E.
  • NK cells circulating lymphocytes
  • the activated lymphocytes or NK cells are most preferably the patient's own cells that were earlier isolated from a blood or tumor sample and activated (or "expanded") in vitro.
  • said additional active compounds may be contained in the same composition or administrated separately.
  • the pharmaceutical composition of the invention relates to combined preparation for simultaneous, separate or sequential use in the treatment of solid cancer in a patient in need thereof wherein the patient was being classified as having a poor prognosis by the method as above described.
  • the present invention also relates to a method for treating solid cancer in a patient in need thereof wherein the patient was being classified as having a poor prognosis by the method as above described, comprising the step of administering to said patient the compound of the invention.
  • the solid cancer referred to in the methods and uses described herein above may be for example a head and neck squamous cell carcinoma (FiNSCC) or a medulloblastoma (MDB).
  • kits comprising the compound of the invention. Kits containing the compound of the invention find use in therapeutic methods.
  • the present invention relates to a method of screening a candidate compound for use as a drug for the treatment of solid cancer in a patient in need thereof wherein the patient was being classified as having a poor prognosis by the method as above described, wherein the method comprises the steps of:
  • VEGF-C and VEGF-C receptor VEGFR-3 and VEGFR-2
  • VEGFR-3 and VEGFR-2 providing a cell, tissue sample or organism expressing a VEGF-C and VEGF-C receptor
  • a candidate compound such as small organic molecule, an oligonucleotide, a polypeptide, an aptamer, antibody or an intra-antibody,
  • lymph node metastasis is measured such as described in the example.
  • measuring the lymphatic vessel density and lymph node metastasis involves determining a Ki on the VEGF-C receptor cloned and transfected in a stable manner into a CHO cell line, measuring the VEGF-C downstream signalling, measuring lymphatic vessel density, measuring inhibition of VEGF-C induced endothelial cell proliferation, measuring VEGFR-3 and VEGFR-2 phosphorylation, and measuring KRAS/RAF/MEK/ERK and PI3K/AKT/mTOR signaling.
  • the present invention relates to a method for treating solid cancer, for example a head and neck squamous cell carcinoma (FINSCC) or a medulloblastoma (MDB), in a patient in need thereof, comprising the step of administering to said patient a VEGF-C inhibitor in combination with radiotherapy treatment (herein defined as a photon (X) radiotherapy or a proton (P) radiotherapy), for example in combination with proton radiotherapy.
  • a VEGF-C inhibitor in combination with radiotherapy treatment
  • X photon
  • P proton
  • FIGURES are a diagrammatic representation of FIGURES.
  • the values correspond to fold increase, as compared to the viable cell number at 24h after cell seeding.
  • CT control (non-irradiated cells).
  • FIG. 1 VEGF-C protein expression levels and regulation in CAL33 cells following P or X irradiation.
  • A VEGF-C protein levels at 48h post-single irradiation (AR- SI): * and *, significantly (p ⁇ 0.05) increased levels after a low (2 Gy) or high (8 Gy) dose of P and X irradiation, respectively, as compared to CT; #, significantly decreased levels after a high dose of P, as compared to X irradiation; ⁇ , significantly increased levels after a high, as compared to a low X irradiation dose;
  • B VEGF-C protein levels after cell expansion following the third irradiation (CR-MI): * and *, significantly increased levels after low and high doses of P and X irradiation, respectively, as compared to CT; Concentration in ng/ml, normalized to 1 x 106 cells, and represented as percentage of CT.
  • FIG. 3 Evaluation of tumors generated following xenografting of either non- irradiated, P or X irradiated CAL33 cells in immunodeficient mice.
  • A Average tumor volume (mm3);
  • B Representative images of tumor xenografts;
  • C Heatmap of ten most up- and down-regulated mouse genes in tumors generated by non-irradiated cells vs P or X tumors, and in P vs X tumors;
  • D Heatmap of ten most up- and down-regulated human genes in tumors generated by non-irradiated cells vs P or X tumors, and in P vs X tumors;
  • E Venn diagrams showing common up-regulated and down-regulated human genes between P and X tumors. Framed genes are commonly expressed in P and X tumors. Selection is adjusted p value ⁇ 0.05 and lofFC > 1.
  • FIG. 4 Histology, immunofluorescence and quantitative gene expression of vascular and lymphatic markers in murine xenografts.
  • A Representative images of HES staining, indicating increased necrosis (black arrowhead, delimited by dashed black lines) in CT and increased blood vessels density (white arrowhead showing collagen surrounding the vessels) in the irradiated cells-derived tumors;
  • B Representative images of CD31 (endothelial cells, green) / aSMA (pericytes, red) / Hoechst (nuclei, blue) staining, showing anarchic blood vessels structures and lack of pericyte coverage of blood vessels in the irradiated cells-derived tumors;
  • C Representative images of LYVEl (lymphatic endothelial cells, red) / Hoechst (nuclei, blue) staining, showing different patterns of lymphatic vessels development in X (both periphery and interior of the tumor), P and CT (periphery of the tumor) groups
  • FIG. 1 Evaluation of vascular and lymphatic markers in biopsies from patients diagnosed with HNSCC. Representative images of immunohistochemistry for (A) PDPN and (B) CD31 expression: (1) oral and (2) hypopharyngeal localization; Left panels (l.a, 2.
  • FIG. 7 (A) Proliferation curves, (B) VEGF-C protein expression levels, activity of a (C) VEGF-C promoter and (D) artificial promotor having three NF- ⁇ binding sites, in CAL27 cells following high doses of either P or X irradiation (CR-MI setting).
  • FIG. 8 Heatmap of ten most up- and down-regulated mouse genes involved in angiogenesis, inflammation, metastasis, M1/M2 macrophage transition and proliferation in tumors generated by non-irradiated cells vs P or X tumors, and in P vs X tumors. Framed genes are commonly expressed in P and X tumors. Selection is Abs(logFC) > 1.
  • Figure 9 Heatmap of ten most up- and down-regulated human genes involved in angiogenesis, inflammation, metastasis, M1/M2 macrophage transition and proliferation in tumors generated by non-irradiated cells vs P or X tumors, and in P vs X tumors. Framed genes are commonly expressed in P and X tumors. Written in red are genes associated with disease progression; written in green are genes associated with favorable outcomes. Selection is adjusted p value ⁇ 0.05 and Abs(logFC) > 1.
  • FIG. 10 Density of (A) tumor vessels and (B) tumor blood vessels with CD31/aSMA colocalization. * and *, significantly increased vessel density in P and X tumors, respectively, as compared to CT; # and #, significantly decreased density of blood vessels with CD31/aSMA colocalization in P and X tumors, respectively, as compared to CT; CT, control (non-irradiated cells).
  • FIG. 11 cBioPortal data showing the correlation between VEGF-C over- expression and significantly lower (A) disease free and (B) overall survival rates in patients with HNSCC.
  • FIG 13 Relative levels of VEGFC mRNA in different medulloblastoma (MDB) lines from different genetic groups: Sonic hedgehog cells (SHH), HDMB03 cells (Group 3) and Group 4 (cf. example 2 and reference 51).
  • SHH Sonic hedgehog cells
  • HDMB03 cells Group 3
  • Group 4 cf. example 2 and reference 51.
  • the HDMB03 cells (Group 3 MDB) were incubated in the presence of various reference chemotherapies or irradiation with 8 Gray photons (CbPT: carbop latin, Eto, etoposide, CT: vehicle, MIX, CbPT/Eto combination, Radio: irradiation, 8 grays).
  • the chemotherapy concentrations correspond to the dose killing 50% of the cells (IC50).
  • VEGFC was measured in cell culture medium by ELISA. ** p ⁇ 0.01; *** p ⁇ 0.001.
  • Figure 15 qPCR analysis of the different genes involved in the lymphatic program in HDMB03 cells (Group3 MDB); * p>0.05; *** p ⁇ 0.001.
  • IHC Immunohistochemistry
  • CAL33 and CAL27 Two human HNSCC cell lines, CAL33 and CAL27, were provided through a Material Transfer Agreement with the Oncopharmacology Laboratory, Centre Antoine Lacassagne (CAL), where they had initially been isolated (50). The cells were cultured in Dulbecco's Modified Eagle's Medium supplemented with 7% fetal bovine serum (Thermo Fisher Scientific, Waltham, MA, USA).
  • the cells were irradiated either once or three times, one week apart (multiple irradiations, MI) with either 2 Gy (low dose) or 8 Gy (high dose), and processed 6h after irradiation.
  • MI multiple irradiations
  • cells were re-seeded after each irradiation and kept in culture until the next irradiation to reproduce the clinical situation where patients are usually given several irradiations.
  • the chronic response (CR) was evaluated to determine if the changes associated with the acute response (AR) persist late (three weeks) after irradiation.
  • Two cell groups were thus generated from each independent irradiation experiment. They consisted of cells subjected to: (1) SI and analysis 48h thereafter (AR-SI); (2) MI and culture expansion (three weeks) after the third irradiation (CR-MI).
  • the cell counting for the CR-MI group was done every day, for four days post- seeding, in triplicate, with an automatic cell counter (Advanced Detection Accurate Measurement system, LabTech, TAMPA, FL, USA), according to the manufacturer instructions.
  • cDNA samples were amplified by using the StepOnePlusTM RT-PCR System (Thermo Fisher Scientific) for 40 cycles with the TakyonTM Rox SYBR® Master Mix, dTTP Blue (Eurogentec, vide, Belgium) and specific oligonucleotides (Sigma Aldrich), to assess mRNA expression for VEGF-A, VEGF-C, VEGF-D, VEGFR-1, VEGFR-2, VEGFR-3, NRP1, NRP2, IL-6, IL-8, CCL2, TRF2, PLK1, PD-L1, LYVE1, PDPN and PROX1.
  • mRNA levels were normalized to a housekeeping mRNA coding for either the human or murine ribosomal protein, large, P0 (RPLPO).
  • the gene expression levels were given the individual scores of -1, 0 and 1 when they were significantly decreased, not significantly changed and significantly increased, respectively, as compared to control. For each irradiation setting, a global gene expression score was then calculated by cumulating the individual scores allocated to each gene expression level.
  • VEGF-C protein was quantified by using an enzyme-linked immunosorbent assay (human DuoSet ELISA kit, R&D Systems, MN, USA). Protein concentration was normalized to the viable cell number.
  • CAL33 cells belonging to the CR-MI group were transfected by using 50 ⁇ NaCl buffer, 1.25 ⁇ of polyethylenimine transfection reagent (Sigma Aldrich) and 0.5 ⁇ g of total test plasmid DNA-renilla luciferase.
  • the plasmids encoded either (i) a human vegf-c promoter fragment with either a non-mutated (wild type, WT) or a mutated (MUT) binding site for the nuclear factor kappa- light-chain-enhancer of activated B cells (NF-KB) (32), (ii) an artificial promoter containing three binding sites for human NF- ⁇ or (iii) a human VEGF-C 3 'UTR reporter (LightSwitchTM, S803537, Active Motif, Carlsbad, CA, USA), all cloned downstream of the luciferase reporter gene.
  • a CMV plasmid was used to control the variability of transfection efficiency in the reporter assays.
  • RNA degradation was documented (Bioanalyzer 2100, Agilent Technologies, Santa Clara, CA, USA).
  • the libraries were generated by using Truseq Stranded mRNA kit (Illumina, San Diego, CA, USA).
  • STAR options were set to the recommended Encode RNA-seq options "— outFilterType BySJout —outFilterMultimapNmax 20 —alignSJoverhangMin 8 alignSJDBoverhangMin 1 —outFilterMismatchNmax 999 —outFilterMismatchNoverLmax 0.04 -alignlntronMin 20 -alignlntronMax 1000000 -alignMatesGapMax 1000000".
  • Gene counts were obtained with featureCounts (subread-1.5.0-p3-Linux-x86_64) and "--primary -p -s 1 -C” options, by using the same GTF files used for STAR splice junctions training.
  • genes involved in angiogenesis, inflammation, metastasis and cell proliferation were selected by using the Ingenuity Pathway Analysis (Qiagen) database.
  • Qiagen Ingenuity Pathway Analysis
  • M1/M2 macrophages-related genes the GEO dataset GSE69607 has been reanalyzed by using geo2R online resource.
  • Genes up- and down-regulated (Abs (logFC)>2) in both Ml vs M0 and M2 vs M0 comparisons were selected as the "M1/M2 macrophages"-related gene list.
  • Murine tumor sections were handled as previously described 8. To assess tumor architecture, the sections were subjected to Hematoxylin Eosin Saffron (HES) staining. For immunofluorescence, the frozen sections were incubated overnight, at 4°C, with the following primary antibodies: polyclonal rabbit anti-mouse/human LYVE1 (1 :200; Abeam, Cambridge, United Kingdom), monoclonal mouse anti-mouse/human alpha smooth muscle actin (aSMA, 1 :400, Sigma Aldrich) and monoclonal rat anti-mouse CD31 (1 :50, clone MEC 13.3, BD Pharmigen, Heidelberg, Germany) primary antibodies, then incubated for 2h at room temperature, in the dark, with the secondary antibodies: anti-rabbit FP594, anti-mouse FP547 (1 : 1000, FluoroProbes, Interchim, Montlucon, France) and anti-rat AF488 (1 : 1000, AlexaFluor, Thermo
  • Table 2 Common up-regulated and down-regulated human genes in tumors generated with either X or P irradiated cells. In bullet are genes up-regulated in either P or X tumors, but down-regulated in tumors generated with non-irradiated cells. Selection is adjusted p value ⁇ 0.05 and lofFC > 1.
  • Table 4 Quantitative gene expression, as percentage of control (0 Gy), in either P or X irradiated CAL27 cells belonging to CR-MI group. Highlighted values - significantly different (p ⁇ 0.05) expression levels, as compared to control, for genes associated to antitumor (dark grey) and pro-tumor (black) effects; *, significantly different expression levels after 2 Gy, as compared to 8 Gy of either P or X irradiation; #, significantly different expression levels after either 2 Gy or 8 Gy of P, as compared to X irradiation.
  • the mRNA levels of the different tested genes overall increased in a dose-dependent manner and with the irradiation number after both P and X irradiation.
  • IL-8 was the gene whose mRNA was induced at the highest level after X (79-fold, as compared to control), but not after P irradiation, within the high dose CR-MI setting (p ⁇ 0.0001).
  • both P and X irradiation augmented PD-L1 mRNA expression in a dose-dependent manner within the AR-SI and CR-MI settings, and in an irradiation number-dependent manner within the AR-SI setting.
  • the generated gene expression scores showed that P irradiation is associated with a more favorable profile [reduced proliferation, (lymph)angiogenesis, inflammation)].
  • VEGF-C the major growth factor for lymphatic endothelial cells.
  • mRNA levels of VEGF-C were increased after both low and high dose(s) of P or X irradiation, they were lower after high dose(s) of P irradiation.
  • VEGF-C protein levels in CAL33 and CAL27 cells were assessed.
  • VEGF-C protein levels increased in a dose-dependent manner following both P and X irradiation. Furthermore, they were significantly lower after P irradiation.
  • Irradiation by either X or P stimulated the activity of the vegf-c promoter especially in CAL33 cells surviving to multiple X irradiations (6- and 18-fold increase, respectively, p ⁇ 0.001, Figure 2C).
  • This result is consistent with the induction of the VEGF-C mRNA within the CR-MI setting (Table 1) and suggests a chronic induction of vegf-c gene transcription, an increase in vegf-c mRNA half-life or a combination of both mechanisms.
  • Mutation of the NF- ⁇ binding site (MUT) had no effect on the basal vegf-c promoter activity in no n- irradiated cells.
  • VEGF-C was particularly discriminative between the two experimental irradiation conditions, we tested whether it had induced the development of lymphatic vessels.
  • LYVEl, PDPN and PROXl markers of lymphatic vessels were then tested (Fig. 4.D).
  • VEGF-C may represent an extrinsic mechanism responsible for the post-irradiation tumor dissemination/metastasis in FiNSCC.
  • VEGF-C expression was associated with lymph node metastasis, recurrence and a poorer five-year survival rate in patients with FiNSCC, being an independent prognostic factor (11, 29).
  • VEGF-C may be an important therapeutic target for FiNSCC patients who relapse after radiotherapy with either P or X.
  • VEGF-C might be a major factor responsible for post-irradiation disease progression in FiNSCC patients, via promotion of lymphangiogenesis, we further started investigating the mechanisms involved in its induction, which may serve to its therapeutic targeting. Regulation of VEGF-C expression has been poorly addressed (27, 31, 32). Irradiation-mediated induction of VEGF-C mRNA suggested stimulation of transcription, stabilization of mRNA or a combination of these mechanisms (31). Our data indicate that both P and X irradiation stimulated the activity of a short form of vegf-c promoter in CAL33 cells. The vegf-c promoter contained a binding site for NF- ⁇ .
  • VEGF-C regulation plays a key role in VEGF-C regulation, as suggested in another cancer type (32).
  • NF- ⁇ plays a key role in VEGF-C regulation, as suggested in another cancer type (32).
  • these cell lines came from two different patients, our results highlight the inter-patient variability in VEGF-C expression and regulation, stressing out the importance of implementing personalized diagnosis and treatment strategies.
  • the VEGF-A and VEGF-D genes were down-regulated by P and up-regulated by X irradiation.
  • VEGF-A expression significantly correlated with lymph node metastasis in patients with FiNSCC 11.
  • VEGF-A expression was also associated with higher clinical stages and worse overall survival, being a significant predictor of poor prognosis in patients with HNSCC (33).
  • VEGF-D expression correlated with lymphatic vessel density and lymph node metastasis in these patients (10).
  • VEGFR-2, VEGFR-3 and NRP1 highly expressed by HNSCC cells (34), were down-regulated in the surviving cells selected after three irradiations with P, but not with X.
  • High NRP1 and NRP2 levels correlated with poor prognosis in HNSCC patients, NRP2 being an independent prognostic markers for overall survival (35).
  • IL-6 expression predicted a poor response to radio-chemotherapy and a non-favorable prognosis in HNSCC patients (39). It was also linked to radiation resistance and development of chronic toxicities after irradiation (40). Depending on tumor location, the most common side effects after conventional radiotherapy of HNSCC include mucositis, xerostomia, dysphagia requiring short-term or permanent gastrostomy, soft tissue/bone necrosis, neck fibrosis, and thyroid dysfunction (41). Although the primary goal in radiotherapy is tumor control, a parallel essential goal is to spare normal tissues from radiation toxicity.
  • Irradiation leads to adaptive changes in the tumor microenvironment that may limit the generation of an anti-tumor immune response (24). Indeed, we showed a significant increase of PD-Ll expression after P, and confirmed the X radiation- induced PD-Ll expression in other cancers (24, 45). In patients with HNSCC, high PD-Ll expression in primary tumors correlated with metastasis and poor prognosis, being an independent prognostic factor (46). PD-Ll was also a significant predictor for poor treatment response and shorter survival in X radiotherapy-treated patients with HNSCC (45).
  • PDZK1IP1 PDZK1 interacting protein 1
  • FLRT2 fibronectin leucine rich transmembrane protein 2
  • mouse Car2 expression was down-regulated in P and X tumors, while up-regulated in tumors generated with non-irradiated cells.
  • low CAR2 protein expression has been associated with increased tumor size (26).
  • the X tumors showed up-regulation of human genes involved in metastasis, angiogenesis and epithelial mesenchymal transition, such as MMP2, MMP9, MMP13, MMP16, MMP28 and vimentinl5, while P tumors showed up-regulation of human C-C Motif Chemokine Ligand 5 chemokine gene involved in CD8 + T lymphocytes recruitment associated with better clinical outcomes (49).
  • lymphatic markers expression in patients with relapsed HNSCC after X radiotherapy we investigated lymphatic markers expression in patients with relapsed HNSCC after X radiotherapy. Biopsies at relapse are very rarely sampled in radiotherapy-treated patients. However, in this small cohort, all patients presented increased protein and/or mR A levels of PDPN, VEGF- C, LYVE1 and PROX1, bringing evidence that conventional radiotherapy may promote lymphangiogenesis. It has also been reported by others that high PDPN expression is associated with aggressive tumor behavior, poor prognosis and metastatic regulation through interaction with VEGF-C, suggesting that PDPN may be used as a potential prognostic biomarker for HNSCC (27). However, our in vitro studies did not reveal increased PDPN expression in HNSCC cells that resisted to MI ( Figure 12).
  • MDBs Medulloblastomas
  • MDBs Medulloblastomas
  • 70% of MDBs are diagnosed before the age of 10 and MDB are rare in adults.
  • MDB is a pathology composed of four molecular groups: wingless (WNT), sonic hedgehog (SHH), group 3 and group 4 (51). These groups are defined by clinical and molecular parameters of the tumor cells.
  • WNT and SHH groups exhibit aberrant activation of the WNT and SHH signaling pathways.
  • Group 3 tumors overexpress the OTX2 and c-MYC genes and those of group 4 N-MYC (52).
  • MDB The genetic landscape of MDB more precisely describes and illustrates tumor heterogeneity among the previously identified subgroups (53-55). These classifications represent important indicators for treatment decision at the first line but do not predict patients at risk of relapse after conventional therapies.
  • MDB treatments combine surgery, irradiation and/or multiple chemotherapies (carboplatin, etoposide). These heavy treatments induce physical, psychological or behavioral sequelae.
  • MDBs are highly vascularized by overexpression of VEGF and many other markers of angiogenesis (VEGFB, VEGFC, FGF, angiopoietin) (56). Anti-angiogenic treatments induce a poor response rate in MDB and are not devoid of side effects in developing children.
  • DAOY and HDMB03 cell lines were purchased from the ATCC. Stocks were made at the original date of obtaining the cells, and were usually passaged for no more than 4 months. These cell lines have been authenticated by DNA profiling using 8 different and highly polymorphic short tandem repeat loci (DSMZ).
  • Chemo/radiotherapeutic treatments result in the expression of VEGFC and several lymphatic markers in a Group 3 MDB model cell line.
  • the standard chemo therapeutic treatments for MDB are combinations of carboplatin/etoposide. We determined the IC50 for each of these compounds on different MDB lines. Our results show VEGFC induction by chemotherapies/irradiation in MDB cells ( Figure 14).
  • lymphatic vessels LV
  • LV lymphatic vessels
  • Pathologists advocate the absence of such vessels. However, their presence has been recently identified in the meninges (62, 63). The presence of LV in brain tumors has never been reported.
  • podoplanin labeling commonly used by pathologist to identify lymphatic embols
  • Figure 16 We believe these structures to be lymphatic vessels.
  • patients exhibiting such structures should beneficiate from an aggressive treatment of cancer, typically radiotherapy, preferably P radiotherapy, and/or chemotherapy using a VEGF-C inhibitor as the chemotherapeutic agent.
  • VEGF-C and VEGF-D expression is correlated with lymphatic vessel density and lymph node metastasis in oral squamous cell carcinoma: Implications for use as a prognostic marker. Int J Oncol 2009; 34: 673-680.
  • VEGF-A stimulates lymphangiogenesis and hemangiogenesis in inflammatory neovascularization via macrophage recruitment. J Clin Invest 2004; 113: 1040-1050.
  • VEGF-C Vascular endothelial growth factor C
  • Tumor necrosis factor-alpha promotes the lymphangiogenesis of gallbladder carcinoma through nuclear factor-kappaB- mediated upregulation of vascular endothelial growth factor-C. Cancer Sci 2014; 105: 1261- 1271.

Abstract

L'invention concerne un procédé de prédiction de l'issue d'un cancer chez un patient atteint d'un cancer solide après une radiothérapie. Les inventeurs ont étudié les effets biologiques comparatifs du rayonnement de protons (P) par rapport aux photons (X) dans les cellules de carcinome malpighien de la tête et du cou (FiNSCC). Les inventeurs ont démontré que le rayonnement P et X induisait une surexpression de VEGF-C au niveau des niveaux de gène et de protéine dans les cellules FiNSCC et dans les cellules MDB et que le VEGF-C est un facteur majeur responsable de la progression de la maladie après irradiation chez des patients atteints de FiNSCC, par l'intermédiaire de la promotion de la lymphangiogenèse. Les inventeurs ont démontré que le VEGF-C est une cible thérapeutique importante chez les patients atteints de FiNSCC qui rechutent après une radiothérapie. Les inventeurs ont examiné la présence de marqueurs lymphatiques dans des biopsies à partir de FiNSCC humain rechute et localement rechute, après une radiothérapie X. Les patients présentent un VEGF-C augmenté, mettant en évidence qu'une radiothérapie X favorise la lymphangiogenèse. Ainsi, l'invention concerne un procédé de prédiction du résultat d'un cancer chez un patient atteint d'un cancer solide après une radiothérapie, comprenant l'étape de détermination du niveau d'expression de VEGF-C. L'invention concerne également un inhibiteur de VEGF-C destiné à être utilisé dans le traitement du cancer solide chez un patient classé, par le procédé de l'invention, comme présentant un pronostic médiocre.
PCT/EP2018/059613 2017-04-14 2018-04-16 Méthodes et compositions pharmaceutiques destinées au traitement du cancer WO2018189403A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP17305448.7 2017-04-14
EP17305448 2017-04-14

Publications (1)

Publication Number Publication Date
WO2018189403A1 true WO2018189403A1 (fr) 2018-10-18

Family

ID=58644983

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2018/059613 WO2018189403A1 (fr) 2017-04-14 2018-04-16 Méthodes et compositions pharmaceutiques destinées au traitement du cancer

Country Status (1)

Country Link
WO (1) WO2018189403A1 (fr)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3885452A1 (fr) * 2020-03-23 2021-09-29 Koninklijke Philips N.V. Prédiction de la réponse à la radiothérapie pour un sujet atteint d'un cancer de la prostate sur la base des gènes de chimiokine
CN114681609A (zh) * 2022-05-05 2022-07-01 浙江大学 抗il-6抗体组合物在制备治疗肝细胞癌药物中的应用

Citations (61)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4499052A (en) 1982-08-30 1985-02-12 Becton, Dickinson And Company Apparatus for distinguishing multiple subpopulations of cells
US4717655A (en) 1982-08-30 1988-01-05 Becton, Dickinson And Company Method and apparatus for distinguishing multiple subpopulations of cells
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
WO1990007861A1 (fr) 1988-12-28 1990-07-26 Protein Design Labs, Inc. IMMUNOGLOBULINES CHIMERIQUES SPECIFIQUES CONTRE LA PROTEINE TAC p55 DU RECEPTEUR D'IL-2
US5225539A (en) 1986-03-27 1993-07-06 Medical Research Council Recombinant altered antibodies and methods of making altered antibodies
US5229275A (en) 1990-04-26 1993-07-20 Akzo N.V. In-vitro method for producing antigen-specific human monoclonal antibodies
US5545807A (en) 1988-10-12 1996-08-13 The Babraham Institute Production of antibodies from transgenic animals
US5545806A (en) 1990-08-29 1996-08-13 Genpharm International, Inc. Ransgenic non-human animals for producing heterologous antibodies
US5565332A (en) 1991-09-23 1996-10-15 Medical Research Council Production of chimeric antibodies - a combinatorial approach
US5567610A (en) 1986-09-04 1996-10-22 Bioinvent International Ab Method of producing human monoclonal antibodies and kit therefor
US5573905A (en) 1992-03-30 1996-11-12 The Scripps Research Institute Encoded combinatorial chemical libraries
US5585089A (en) 1988-12-28 1996-12-17 Protein Design Labs, Inc. Humanized immunoglobulins
US5591669A (en) 1988-12-05 1997-01-07 Genpharm International, Inc. Transgenic mice depleted in a mature lymphocytic cell-type
US5598369A (en) 1994-06-28 1997-01-28 Advanced Micro Devices, Inc. Flash EEPROM array with floating substrate erase operation
US5618829A (en) 1993-01-28 1997-04-08 Mitsubishi Chemical Corporation Tyrosine kinase inhibitors and benzoylacrylamide derivatives
US5639757A (en) 1995-05-23 1997-06-17 Pfizer Inc. 4-aminopyrrolo[2,3-d]pyrimidines as tyrosine kinase inhibitors
US5728868A (en) 1993-07-15 1998-03-17 Cancer Research Campaign Technology Limited Prodrugs of protein tyrosine kinase inhibitors
US5800988A (en) 1992-08-21 1998-09-01 Vrije Universiteit Brussel Immunoglobulins devoid of light chains
US5804396A (en) 1994-10-12 1998-09-08 Sugen, Inc. Assay for agents active in proliferative disorders
US5859205A (en) 1989-12-21 1999-01-12 Celltech Limited Humanised antibodies
WO1999032619A1 (fr) 1997-12-23 1999-07-01 The Carnegie Institution Of Washington Inhibition genetique par de l'arn double brin
US5981732A (en) 1998-12-04 1999-11-09 Isis Pharmaceuticals Inc. Antisense modulation of G-alpha-13 expression
US6046321A (en) 1999-04-09 2000-04-04 Isis Pharmaceuticals Inc. Antisense modulation of G-alpha-i1 expression
US6100254A (en) 1997-10-10 2000-08-08 Board Of Regents, The University Of Texas System Inhibitors of protein tyrosine kinases
US6107091A (en) 1998-12-03 2000-08-22 Isis Pharmaceuticals Inc. Antisense inhibition of G-alpha-16 expression
US6127374A (en) 1997-07-29 2000-10-03 Warner-Lambert Company Irreversible inhibitors of tyrosine kinases
US6150584A (en) 1990-01-12 2000-11-21 Abgenix, Inc. Human antibodies derived from immunized xenomice
US6245759B1 (en) 1999-03-11 2001-06-12 Merck & Co., Inc. Tyrosine kinase inhibitors
WO2001052875A1 (fr) 2000-01-18 2001-07-26 Ludwig Institute For Cancer Research Inhibiteur peptidomimetique de vegf-d/vegf-c/vegf
US6306874B1 (en) 1999-10-19 2001-10-23 Merck & Co., Inc. Tyrosine kinase inhibitors
US6313138B1 (en) 2000-02-25 2001-11-06 Merck & Co., Inc. Tyrosine kinase inhibitors
US6316444B1 (en) 1999-06-30 2001-11-13 Merck & Co., Inc. SRC kinase inhibitor compounds
US6329380B1 (en) 1999-06-30 2001-12-11 Merck & Co., Inc. SRC kinase inhibitor compounds
US6344459B1 (en) 1996-04-12 2002-02-05 Warner-Lambert Company Irreversible inhibitors of tyrosine kinases
US6365354B1 (en) 2000-07-31 2002-04-02 Isis Pharmaceuticals, Inc. Antisense modulation of lysophospholipase I expression
US6410323B1 (en) 1999-08-31 2002-06-25 Isis Pharmaceuticals, Inc. Antisense modulation of human Rho family gene expression
US6420382B2 (en) 2000-02-25 2002-07-16 Merck & Co., Inc. Tyrosine kinase inhibitors
US6479512B1 (en) 1999-10-19 2002-11-12 Merck & Co., Inc. Tyrosine kinase inhibitors
US6498165B1 (en) 1999-06-30 2002-12-24 Merck & Co., Inc. Src kinase inhibitor compounds
US6566135B1 (en) 2000-10-04 2003-05-20 Isis Pharmaceuticals, Inc. Antisense modulation of caspase 6 expression
US6566131B1 (en) 2000-10-04 2003-05-20 Isis Pharmaceuticals, Inc. Antisense modulation of Smad6 expression
US6573099B2 (en) 1998-03-20 2003-06-03 Benitec Australia, Ltd. Genetic constructs for delaying or repressing the expression of a target gene
US6586423B2 (en) 1999-09-10 2003-07-01 Merck & Co., Inc. Tyrosine kinase inhibitors
US6740665B1 (en) 1999-02-10 2004-05-25 Ramachandran Murali Tyrosine kinase inhibitors and methods of using the same
US6765087B1 (en) 1992-08-21 2004-07-20 Vrije Universiteit Brussel Immunoglobulins devoid of light chains
US6794393B1 (en) 1999-10-19 2004-09-21 Merck & Co., Inc. Tyrosine kinase inhibitors
US6838254B1 (en) 1993-04-29 2005-01-04 Conopco, Inc. Production of antibodies or (functionalized) fragments thereof derived from heavy chain immunoglobulins of camelidae
US6875767B2 (en) 2001-06-22 2005-04-05 Merck & Co., Inc. (5-cyano-2-thiazolyl)amino-4-pyridine tyrosine kinase inhibitors
US6927293B2 (en) 2001-08-30 2005-08-09 Merck & Co., Inc. Tyrosine kinase inhibitors
US6958340B2 (en) 2001-08-01 2005-10-25 Merck & Co., Inc. Tyrosine kinase inhibitors
US7109304B2 (en) 2003-07-31 2006-09-19 Immunomedics, Inc. Humanized anti-CD19 antibodies
WO2007015935A2 (fr) * 2005-07-29 2007-02-08 Bayer Healthcare Llc Procedes diagnostiques permettant de predire le succes therapeutique, d'eviter les rechutes et d'obtenir une bonne survivance generale en therapie du cancer
US7235641B2 (en) 2003-12-22 2007-06-26 Micromet Ag Bispecific antibodies
US20070254295A1 (en) 2006-03-17 2007-11-01 Prometheus Laboratories Inc. Methods of predicting and monitoring tyrosine kinase inhibitor therapy
WO2008119353A1 (fr) 2007-03-29 2008-10-09 Genmab A/S Anticorps bispécifiques et procédés de production de ceux-ci
WO2010053717A1 (fr) * 2008-10-29 2010-05-14 William Beaumont Hospital Procédés d'utilisation de biomarqueurs
WO2011071577A1 (fr) 2009-12-11 2011-06-16 Genentech, Inc. Anticorps anti-vegf-c et leurs procédés d'utilisation
WO2011127519A1 (fr) 2010-04-15 2011-10-20 Vegenics Pty Limited Traitement d'association avec des antagonistes de vegf-c
WO2011131746A2 (fr) 2010-04-20 2011-10-27 Genmab A/S Protéines contenant des anticorps fc hétérodimères et leurs procédés de production
CN104894229A (zh) * 2014-03-04 2015-09-09 中南大学 己糖激酶2作为鼻咽癌放疗预后预测的生物标志物
WO2016184793A1 (fr) 2015-05-15 2016-11-24 INSERM (Institut National de la Santé et de la Recherche Médicale) Méthodes de traitement de patients souffrant d'un adénocarcinome rénal métastatique résistant aux inhibiteurs du vegfr

Patent Citations (68)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4499052A (en) 1982-08-30 1985-02-12 Becton, Dickinson And Company Apparatus for distinguishing multiple subpopulations of cells
US4717655A (en) 1982-08-30 1988-01-05 Becton, Dickinson And Company Method and apparatus for distinguishing multiple subpopulations of cells
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
US5225539A (en) 1986-03-27 1993-07-06 Medical Research Council Recombinant altered antibodies and methods of making altered antibodies
US5567610A (en) 1986-09-04 1996-10-22 Bioinvent International Ab Method of producing human monoclonal antibodies and kit therefor
US5545807A (en) 1988-10-12 1996-08-13 The Babraham Institute Production of antibodies from transgenic animals
US5591669A (en) 1988-12-05 1997-01-07 Genpharm International, Inc. Transgenic mice depleted in a mature lymphocytic cell-type
US5693761A (en) 1988-12-28 1997-12-02 Protein Design Labs, Inc. Polynucleotides encoding improved humanized immunoglobulins
WO1990007861A1 (fr) 1988-12-28 1990-07-26 Protein Design Labs, Inc. IMMUNOGLOBULINES CHIMERIQUES SPECIFIQUES CONTRE LA PROTEINE TAC p55 DU RECEPTEUR D'IL-2
US5585089A (en) 1988-12-28 1996-12-17 Protein Design Labs, Inc. Humanized immunoglobulins
US5693762A (en) 1988-12-28 1997-12-02 Protein Design Labs, Inc. Humanized immunoglobulins
US5859205A (en) 1989-12-21 1999-01-12 Celltech Limited Humanised antibodies
US6150584A (en) 1990-01-12 2000-11-21 Abgenix, Inc. Human antibodies derived from immunized xenomice
US5229275A (en) 1990-04-26 1993-07-20 Akzo N.V. In-vitro method for producing antigen-specific human monoclonal antibodies
US5545806A (en) 1990-08-29 1996-08-13 Genpharm International, Inc. Ransgenic non-human animals for producing heterologous antibodies
US5565332A (en) 1991-09-23 1996-10-15 Medical Research Council Production of chimeric antibodies - a combinatorial approach
US5573905A (en) 1992-03-30 1996-11-12 The Scripps Research Institute Encoded combinatorial chemical libraries
US6015695A (en) 1992-08-21 2000-01-18 Vrije Universiteit Brussel Immunoglobulins devoid of light chains
US5800988A (en) 1992-08-21 1998-09-01 Vrije Universiteit Brussel Immunoglobulins devoid of light chains
US6765087B1 (en) 1992-08-21 2004-07-20 Vrije Universiteit Brussel Immunoglobulins devoid of light chains
US5618829A (en) 1993-01-28 1997-04-08 Mitsubishi Chemical Corporation Tyrosine kinase inhibitors and benzoylacrylamide derivatives
US6838254B1 (en) 1993-04-29 2005-01-04 Conopco, Inc. Production of antibodies or (functionalized) fragments thereof derived from heavy chain immunoglobulins of camelidae
US5728868A (en) 1993-07-15 1998-03-17 Cancer Research Campaign Technology Limited Prodrugs of protein tyrosine kinase inhibitors
US5598369A (en) 1994-06-28 1997-01-28 Advanced Micro Devices, Inc. Flash EEPROM array with floating substrate erase operation
US5804396A (en) 1994-10-12 1998-09-08 Sugen, Inc. Assay for agents active in proliferative disorders
US5639757A (en) 1995-05-23 1997-06-17 Pfizer Inc. 4-aminopyrrolo[2,3-d]pyrimidines as tyrosine kinase inhibitors
US6344459B1 (en) 1996-04-12 2002-02-05 Warner-Lambert Company Irreversible inhibitors of tyrosine kinases
US6127374A (en) 1997-07-29 2000-10-03 Warner-Lambert Company Irreversible inhibitors of tyrosine kinases
US6562818B1 (en) 1997-07-29 2003-05-13 Warner-Lambert Company Irreversible inhibitors of tyrosine kinases
US6100254A (en) 1997-10-10 2000-08-08 Board Of Regents, The University Of Texas System Inhibitors of protein tyrosine kinases
WO1999032619A1 (fr) 1997-12-23 1999-07-01 The Carnegie Institution Of Washington Inhibition genetique par de l'arn double brin
US6506559B1 (en) 1997-12-23 2003-01-14 Carnegie Institute Of Washington Genetic inhibition by double-stranded RNA
US6573099B2 (en) 1998-03-20 2003-06-03 Benitec Australia, Ltd. Genetic constructs for delaying or repressing the expression of a target gene
US6107091A (en) 1998-12-03 2000-08-22 Isis Pharmaceuticals Inc. Antisense inhibition of G-alpha-16 expression
US5981732A (en) 1998-12-04 1999-11-09 Isis Pharmaceuticals Inc. Antisense modulation of G-alpha-13 expression
US6740665B1 (en) 1999-02-10 2004-05-25 Ramachandran Murali Tyrosine kinase inhibitors and methods of using the same
US6544988B1 (en) 1999-03-11 2003-04-08 Merck & Co., Inc. Tyrosine kinase inhibitors
US6245759B1 (en) 1999-03-11 2001-06-12 Merck & Co., Inc. Tyrosine kinase inhibitors
US6046321A (en) 1999-04-09 2000-04-04 Isis Pharmaceuticals Inc. Antisense modulation of G-alpha-i1 expression
US6316444B1 (en) 1999-06-30 2001-11-13 Merck & Co., Inc. SRC kinase inhibitor compounds
US6498165B1 (en) 1999-06-30 2002-12-24 Merck & Co., Inc. Src kinase inhibitor compounds
US6329380B1 (en) 1999-06-30 2001-12-11 Merck & Co., Inc. SRC kinase inhibitor compounds
US6410323B1 (en) 1999-08-31 2002-06-25 Isis Pharmaceuticals, Inc. Antisense modulation of human Rho family gene expression
US6586424B2 (en) 1999-09-10 2003-07-01 Merck & Co., Inc. Tyrosine kinase inhibitors
US6586423B2 (en) 1999-09-10 2003-07-01 Merck & Co., Inc. Tyrosine kinase inhibitors
US6479512B1 (en) 1999-10-19 2002-11-12 Merck & Co., Inc. Tyrosine kinase inhibitors
US6794393B1 (en) 1999-10-19 2004-09-21 Merck & Co., Inc. Tyrosine kinase inhibitors
US6306874B1 (en) 1999-10-19 2001-10-23 Merck & Co., Inc. Tyrosine kinase inhibitors
WO2001052875A1 (fr) 2000-01-18 2001-07-26 Ludwig Institute For Cancer Research Inhibiteur peptidomimetique de vegf-d/vegf-c/vegf
US6313138B1 (en) 2000-02-25 2001-11-06 Merck & Co., Inc. Tyrosine kinase inhibitors
US6420382B2 (en) 2000-02-25 2002-07-16 Merck & Co., Inc. Tyrosine kinase inhibitors
US6365354B1 (en) 2000-07-31 2002-04-02 Isis Pharmaceuticals, Inc. Antisense modulation of lysophospholipase I expression
US6566135B1 (en) 2000-10-04 2003-05-20 Isis Pharmaceuticals, Inc. Antisense modulation of caspase 6 expression
US6566131B1 (en) 2000-10-04 2003-05-20 Isis Pharmaceuticals, Inc. Antisense modulation of Smad6 expression
US6875767B2 (en) 2001-06-22 2005-04-05 Merck & Co., Inc. (5-cyano-2-thiazolyl)amino-4-pyridine tyrosine kinase inhibitors
US6958340B2 (en) 2001-08-01 2005-10-25 Merck & Co., Inc. Tyrosine kinase inhibitors
US6927293B2 (en) 2001-08-30 2005-08-09 Merck & Co., Inc. Tyrosine kinase inhibitors
US7109304B2 (en) 2003-07-31 2006-09-19 Immunomedics, Inc. Humanized anti-CD19 antibodies
US7235641B2 (en) 2003-12-22 2007-06-26 Micromet Ag Bispecific antibodies
WO2007015935A2 (fr) * 2005-07-29 2007-02-08 Bayer Healthcare Llc Procedes diagnostiques permettant de predire le succes therapeutique, d'eviter les rechutes et d'obtenir une bonne survivance generale en therapie du cancer
US20070254295A1 (en) 2006-03-17 2007-11-01 Prometheus Laboratories Inc. Methods of predicting and monitoring tyrosine kinase inhibitor therapy
WO2008119353A1 (fr) 2007-03-29 2008-10-09 Genmab A/S Anticorps bispécifiques et procédés de production de ceux-ci
WO2010053717A1 (fr) * 2008-10-29 2010-05-14 William Beaumont Hospital Procédés d'utilisation de biomarqueurs
WO2011071577A1 (fr) 2009-12-11 2011-06-16 Genentech, Inc. Anticorps anti-vegf-c et leurs procédés d'utilisation
WO2011127519A1 (fr) 2010-04-15 2011-10-20 Vegenics Pty Limited Traitement d'association avec des antagonistes de vegf-c
WO2011131746A2 (fr) 2010-04-20 2011-10-27 Genmab A/S Protéines contenant des anticorps fc hétérodimères et leurs procédés de production
CN104894229A (zh) * 2014-03-04 2015-09-09 中南大学 己糖激酶2作为鼻咽癌放疗预后预测的生物标志物
WO2016184793A1 (fr) 2015-05-15 2016-11-24 INSERM (Institut National de la Santé et de la Recherche Médicale) Méthodes de traitement de patients souffrant d'un adénocarcinome rénal métastatique résistant aux inhibiteurs du vegfr

Non-Patent Citations (79)

* Cited by examiner, † Cited by third party
Title
"Biochemistry and Immunology", 1996, ACADEMIC PRESS, article "Coding, Monoclonal Antibodies: Principles and Practice: Production and Application of Monoclonal Antibodies in Cell Biology"
A. ASPELUND ET AL.: "A dural lymphatic vascular system that drains brain interstitial fluid and macromolecules", J EXP MED, vol. 212, 2015, pages 991 - 999, XP055515828, DOI: doi:10.1084/jem.20142290
A. GAJJAR ET AL.: "Pediatric Brain Tumors: Innovative Genomic Information Is Transforming the Diagnostic and Clinical Landscape", J CLIN ONCOL, vol. 33, 2015, pages 2986 - 2998
A. LOUVEAU ET AL.: "Structural and functional features of central nervous system lymphatic vessels", NATURE, vol. 523, 2015, pages 337 - 341, XP055445074, DOI: doi:10.1038/nature14432
AGNEW CHEM INTL. ED. ENGL., vol. 33, 1994, pages 183 - 186
BEGG AC; STEWART FA; VENS C.: "Strategies to improve radiotherapy with targeted drugs", NAT REV CANCER, vol. 11, 2011, pages 239 - 253
BENHAMOU Y; PICCO V; RAYBAUD H; SUDAKA A; CHAMOREY E; BROLIH S ET AL.: "Telomeric repeat-binding factor 2: a marker for survival and anti-EGFR efficacy in oral carcinoma", ONCOTARGET, vol. 7, 2016, pages 44236 - 44251
BLANCHARD P; WONG AJ; GUNN GB; GARDEN AS; MOHAMED AS; ROSENTHAL DI ET AL.: "Toward a model-based patient selection strategy for proton therapy: External validation of photon-derived normal tissue complication probability models in a head and neck proton therapy cohort", RADIOTHER ONCOL, vol. 121, 2016, pages 381 - 386, XP029854929, DOI: doi:10.1016/j.radonc.2016.08.022
BOWLES DW; DIAMOND JR; LAM ET; WEEKES CD; ASTLING DP; ANDERSON RT ET AL.: "Phase I study of oral rigosertib (ON 01910.Na), a dual inhibitor of the PI3K and Plkl pathways, in adult patients with advanced solid malignancies", CLIN CANCER RES, vol. 20, 2014, pages 1656 - 1665, XP055483800, DOI: doi:10.1158/1078-0432.CCR-13-2506
CHEN MF; CHEN PT; CHEN WC; LU MS; LIN PY; LEE KD.: "The role of PD-Ll in the radiation response and prognosis for esophageal squamous cell carcinoma related to IL-6 and T-cell immunosuppression", ONCOTARGET, vol. 7, 2016, pages 7913 - 7924
CHEN YH; PAN SL; WANG JC; KUO SH; CHENG JC; TENG CM.: "Radiation-induced VEGF-C expression and endothelial cell proliferation in lung cancer", STRAHLENTHER ONKOL, vol. 190, 2014, pages 1154 - 1162
CHU W; SONG X; YANG X; MA L; ZHU J; HE M ET AL.: "Neuropilin-1 promotes epithelial-to-mesenchymal transition by stimulating nuclear factor-kappa B and is associated with poor prognosis in human oral squamous cell carcinoma", PLOS ONE, vol. 9, 2014, pages e101931
CLARK, W. R.: "The Experimental Foundations of Modern Immunology", 1986, WILEY & SONS, INC.
CURSIEFEN C; CHEN L; BORGES LP; JACKSON D; CAO J; RADZIEJEWSKI C ET AL.: "VEGF-A stimulates lymphangiogenesis and hemangiogenesis in inflammatory neovascularization via macrophage recruitment", J CLIN INVEST, vol. 113, 2004, pages 1040 - 1050
DALLAS J; IMANIRAD I; RAJANI R; DAGAN R; SUBBIAH S; GAA R ET AL.: "Response to sunitinib in combination with proton beam radiation in a patient with chondrosarcoma: a case report", J MED CASE REP, vol. 6, 2012, pages 41, XP021131883, DOI: doi:10.1186/1752-1947-6-41
DE BENEDETTI ET AL., J IMMUNOL, vol. 166, 2001, pages 4334 - 4340
DE MIGUEL-LUKEN MJ; CHAVES-CONDE M; DE MIGUEL-LUKEN V; MUNOZ-GALVAN S; LOPEZ-GUERRA JL; MATEOS JC ET AL.: "MAP17 (PDZKIP1) as a novel prognostic biomarker for laryngeal cancer", ONCOTARGET, vol. 6, 2015, pages 12625 - 12636
DING L; LI B; ZHAO Y; FU YF; HU EL; HU QG ET AL.: "Serum CCL2 and CCL3 as potential bio markers for the diagnosis of oral squamous cell carcinoma", TUMOUR BIOL, vol. 35, 2014, pages 10539 - 10546
DOVEDI SJ; ILLIDGE TM.: "The antitumor immune response generated by fractionated radiation therapy may be limited by tumor cell adaptive resistance and can be circumvented by PD-L1 blockade", ONCOIMMUNOLOGY, vol. 4, 2015, pages e1016709
DOYEN J; FALK AT; FLOQUET V; HERAULT J; HANNOUN-LEVI JM.: "Proton beams in cancer treatments: Clinical outcomes and dosimetric comparisons with photon therapy", CANCER TREAT REV, vol. 43, 2016, pages 104 - 112
DU Q; JIANG L; WANG X; WANG M; SHE F; CHEN Y.: "Tumor necrosis factor-alpha promotes the lymphangiogenesis of gallbladder carcinoma through nuclear factor-kappaB-mediated upregulation of vascular endothelial growth factor-C", CANCER SCI, vol. 105, 2014, pages 1261 - 1271
E. C. SCHWALBE ET AL.: "Novel molecular subgroups for clinical classification and outcome prediction in childhood medulloblastoma: a cohort study", LANCET ONCOL, vol. 18, 2017, pages 958 - 971, XP085110050, DOI: doi:10.1016/S1470-2045(17)30243-7
ENHOLM B; PAAVONEN K; RISTIMAKI A; KUMAR V; GUNJI Y; KLEFSTROM J ET AL.: "Comparison of VEGF, VEGF-B, VEGF-C and Ang-1 mRNA regulation by serum, growth factors, oncoproteins and hypoxia", ONCOGENE, vol. 14, 1997, pages 2475 - 2483, XP002913099, DOI: doi:10.1038/sj.onc.1201090
EPRATUZUMAB, LEONARD ET AL., CLINICAL CANCER RESEARCH, vol. 10, pages 53Z7 - 5334
F. M. G. CAVALLI ET AL.: "Intertumoral Heterogeneity within Medulloblastoma Subgroups", CANCER CELL, vol. 31, 2017, pages 737 - 754
FENG YB; LIN DC; SHI ZZ; WANG XC; SHEN XM; ZHANG Y ET AL.: "Overexpression of PLK1 is associated with poor survival by inhibiting apoptosis via enhancement of survivin level in esophageal squamous cell carcinoma", INT J CANCER, vol. 124, 2009, pages 578 - 588, XP055002858, DOI: doi:10.1002/ijc.23990
FRECH S; HORMANN K; RIEDEL F; GOTTE K.: "Lymphatic vessel density in correlation to lymph node metastasis in head and neck squamous cell carcinoma", ANTICANCER RES, vol. 29, 2009, pages 1675 - 1679
FUJITA Y; OKAMOTO M; GODA H; TANO T; NAKASHIRO K; SUGITA A.: "Prognostic significance of interleukin-8 and CD163-positive cell-infiltration in tumor tissues in patients with oral squamous cell carcinoma", PLOS ONE, vol. 9, 2014, pages el 10378
FULTON ET AL., CLINICAL CHEMISTRY, vol. 43, no. 9, 1997, pages 1749 - 1756
FULWYLER; MCHUGH, METHODS IN CELL BIOLOGY, vol. 33, 1990, pages 613 - 629
GIOANNI J; FISCHEL JL; LAMBERT JC; DEMARD F; MAZEAU C; ZANGHELLINI E ET AL.: "Two new human tumor cell lines derived from squamous cell carcinomas of the tongue: establishment, characterization and response to cytotoxic treatment", EUR J CANCER CLIN ONCOL, vol. 24, 1988, pages 1445 - 1455, XP026201081, DOI: doi:10.1016/0277-5379(88)90335-5
GIRDHANI S; LAMONT C; HAHNFELDT P; ABDOLLAHI A; HLATKY L.: "Proton irradiation suppresses angiogenic genes and impairs cell invasion and tumor growth", RADIAT RES, vol. 178, 2012, pages 33 - 45
GREPIN R; GUYOT M; JACQUIN M; DURIVAULT J; CHAMOREY E; SUDAKA A ET AL.: "Acceleration of clear cell renal cell carcinoma growth in mice following bevacizumab/Avastin treatment: the role of CXCL cytokines", ONCOGENE, vol. 31, 2012, pages 1683 - 1694, XP055078253, DOI: doi:10.1038/onc.2011.360
H. HUBER ET AL.: "Angiogenic profile of childhood primitive neuroectodermal brain tumours/medulloblastomas", EUR J CANCER, vol. 37, 2001, pages 2064 - 2072, XP004307926, DOI: doi:10.1016/S0959-8049(01)00225-8
HOLLIDAY EB; FRANK SJ.: "Proton radiation therapy for head and neck cancer: a review of the clinical experience to date", INT J RADIAT ONCOL BIOL PHYS, vol. 89, 2014, pages 292 - 302, XP028658008, DOI: doi:10.1016/j.ijrobp.2014.02.029
HU X; HUANG Z; LIAO Z; HE C; FANG X.: "Low CA II expression is associated with tumor aggressiveness and poor prognosis in gastric cancer patients", INT J CLIN EXP PATHOL, vol. 7, 2014, pages 6716 - 6724
HUANG D; DING Y; ZHOU M; RINI BI; PETILLO D; QIAN CN ET AL.: "Interleukin-8 mediates resistance to antiangiogenic agent sunitinib in renal cell carcinoma", CANCER RES, vol. 70, 2010, pages 1063 - 1071, XP002583999, DOI: doi:10.1158/0008-5472.CAN-09-3965
J. T. WIGLE; G. OLIVER: "Proxl function is required for the development of the murine lymphatic system", CELL, vol. 98, 1999, pages 769 - 778, XP002309908, DOI: doi:10.1016/S0092-8674(00)81511-1
JINNO T; KAWANO S; MARUSE Y; MATSUBARA R; GOTO Y; SAKAMOTO T ET AL.: "Increased expression of interleukin-6 predicts poor response to chemoradiotherapy and unfavorable prognosis in oral squamous cell carcinoma", ONCOL REP, vol. 33, 2015, pages 2161 - 2168
K. J. RANSOHOFF; K. Y. SARIN; J. Y. TANG: "Smoothened Inhibitors in Sonic Hedgehog Subgroup Medulloblastoma", J CLIN ONCOL, vol. 33, 2015, pages 2692 - 2694
KAJIOKA EH; ANDRES ML; MAO XW; MOYERS MF; NELSON GA; GRIDLEY DS.: "Hematological and TGF-beta variations after whole-body proton irradiation", IN VIVO, vol. 14, 2000, pages 703 - 708
KIM HY; RHA KS; SHIM GA; KIM JH; KIM JM; HUANG SM ET AL.: "Podoplanin is involved in the prognosis of head and neck squamous cell carcinoma through interaction with VEGF-C", ONCOL REP, vol. 34, 2015, pages 833 - 842
KIM SA; KWON SM; YOON JH; AHN SG: "The antitumor effect of PLK1 and HSF1 double knockdown on human oral carcinoma cells", INT J ONCOL, vol. 36, 2010, pages 867 - 872
KOHLER; MILSTEIN, NATURE, vol. 256, 1975, pages 495
KRIEGLER: "A Laboratory Manual", 1990, W.H. FREEMAN C.O.
LI M; ZHANG Y; FEURINO LW; WANG H; FISHER WE; BRUNICARDI FC ET AL.: "Interleukin-8 increases vascular endothelial growth factor and neuropilin expression and stimulates ERK activation in human pancreatic cancer", CANCER SCI, vol. 99, 2008, pages 733 - 737
LIU J; LI F; PING Y; WANG L; CHEN X; WANG D ET AL.: "Local production of the chemokines CCL5 and CXCL10 attracts CD8+ T lymphocytes into esophageal squamous cell carcinoma", ONCOTARGET, vol. 6, 2015, pages 24978 - 24989
LOBERG RD; DAY LL; HARWOOD J; YING C; ST JOHN LN; GILES R ET AL.: "CCL2 is a potent regulator of prostate cancer cell migration and proliferation", NEOPLASIA, vol. 8, 2006, pages 578 - 586, XP007910907, DOI: doi:10.1593/neo.06280
MAMELLE G; PAMPURIK J; LUBOINSKI B; LANCAR R; LUSINCHI A; BOSQ J.: "Lymph node prognostic factors in head and neck squamous cell carcinomas", AM J SURG, vol. 168, 1994, pages 494 - 498
MATSUI T; SHIGETA T; UMEDA M; KOMORI T.: "Vascular endothelial growth factor C (VEGF-C) expression predicts metastasis in tongue cancer", ORAL SURG ORAL MED ORAL PATHOL ORAL RADIOL, vol. 120, 2015, pages 436 - 442, XP029265801, DOI: doi:10.1016/j.oooo.2015.06.002
MURRY: "Methods in Molecular Biology", vol. 7, 1991, HUMANA PRESS, INC.
N. C. JOHNSON ET AL.: "Lymphatic endothelial cell identity is reversible and its maintenance requires Proxl activity", GENES DEV, vol. 22, 2008, pages 3282 - 3291
NICHOLAS P. RESTIFO; MARK E. DUDLEY; STEVEN A. ROSENBERG: "Adoptive immunotherapy for cancer: harnessing the T cell response", NATURE REVIEWS IMMUNOLOGY, 12 April 2012 (2012-04-12)
OGATA T; TESHIMA T; KAGAWA K; HISHIKAWA Y; TAKAHASHI Y; KAWAGUCHI A ET AL.: "Particle irradiation suppresses metastatic potential of cancer cells", CANCER RES, vol. 65, 2005, pages 113 - 120
OLIVEIRA-COSTA JP; DE CARVALHO AF; DA SILVEIRA DA GG; AMAYA P; WU Y; PARK KJ ET AL.: "Gene expression patterns through oral squamous cell carcinoma development: PD-L1 expression in primary tumor and circulating tumor cells", ONCOTARGET, vol. 6, 2015, pages 20902 - 20920
P. A. NORTHCOTT ET AL.: "Enhancer hijacking activates GFI1 family oncogenes in medulloblastoma", NATURE, vol. 511, 2014, pages 428 - 434
P. A. NORTHCOTT ET AL.: "The whole-genome landscape of medulloblastoma subtypes", NATURE, vol. 547, 2017, pages 311 - 317
PAN Y; WANG WD; YAGO T.: "Transcriptional regulation of podoplanin expression by Proxl in lymphatic endothelial cells", MICROVASC RES, vol. 94, 2014, pages 96 - 102
PARKIN DM; BRAY F; FERLAY J; PISANI P.: "Global cancer statistics", CA CANCER J CLIN 2005, vol. 55, 2002, pages 74 - 108
PRESCRIBING, RECORDING, AND REPORTING PROTON-BEAM THERAPY: INTERNATIONAL COMMISSION ON RADIATION UNITS AND MEASUREMENTS REPORT 78: "Prescribing, recording, and reporting proton-beam therapy: International Commission on Radiation Units and Measurements Report 78", J ICRU, vol. 7, 2007, pages 210
R. LEROY; N. BENAHMED; F. HULSTAERT; N. VAN DAMME; D. DE RUYSSCHER: "Proton Therapy in Children: A Systematic Review of Clinical Effectiveness in 15 Pediatric Cancers", INT JRADIAT ONCOL BIOL PHYS, vol. 95, 2016, pages 267 - 278
ROITT, I.: "Blackwell Scientific Publications", 1991, article "Essential Immunology"
S. JOSSON; Y. MATSUOKA; L. W. CHUNG; H. E. ZHAU; R. WANG: "Tumor-stroma co-evolution in prostate cancer progression and metastasis", SEMIN CELL DEV BIOL, vol. 21, 2010, pages 26 - 32
SANBROOK ET AL.: "Molecular Cloning: A Laboratory Manual", 1989, COLD SPRING HARBOR LABORATORY PRESS
SEIWERT TY; SALAMA JK; VOKES EE: "The chemoradiation paradigm in head and neck cancer", NAT CLIN PRACT ONCOL, vol. 4, 2007, pages 156 - 171
SHINRIKI S; JONO H; UEDA M; OTA K; OTA T; SUEYOSHI T ET AL.: "Interleukin-6 signalling regulates vascular endothelial growth factor-C synthesis and lymphangiogenesis in human oral squamous cell carcinoma", J PATHOL, vol. 225, 2011, pages 142 - 150
SIO TT; LIN HK; SHI Q; GUNN GB; CLEELAND CS; LEE JJ ET AL.: "Intensity Modulated Proton Therapy Versus Intensity Modulated Photon Radiation Therapy for Oropharyngeal Cancer: First Comparative Results of Patient-Reported Outcomes", INT J RADIAT ONCOL BIOL PHYS, vol. 95, 2016, pages 1107 - 1114, XP029618412, DOI: doi:10.1016/j.ijrobp.2016.02.044
SMITH BD; SMITH GL; CARTER D; SASAKI CT; HAFFTY BG: "Prognostic significance of vascular endothelial growth factor protein levels in oral and oropharyngeal squamous cell carcinoma", J CLIN ONCOL, vol. 18, 2000, pages 2046 - 2052
SUGIURA T; INOUE Y; MATSUKI R; ISHII K; TAKAHASHI M; ABE M ET AL.: "VEGF-C and VEGF-D expression is correlated with lymphatic vessel density and lymph node metastasis in oral squamous cell carcinoma: Implications for use as a prognostic marker", INT J ONCOL, vol. 34, 2009, pages 673 - 680
SUZUKI ET AL., EUROP J OF IMMUNOL, vol. 22, no. 8, 1992, pages 1989 - 1993
TIAN J; ZHAO W; TIAN S; SLATER JM; DENG Z; GRIDLEY DS.: "Expression of genes involved in mouse lung cell differentiation/regulation after acute exposure to photons and protons with or without low-dose preirradiation", RADIAT RES, vol. 176, 2011, pages 553 - 564
WILLERS H; AZZOLI CG; SANTIVASI WL; XIA F.: "Basic mechanisms of therapeutic resistance to radiation and chemotherapy in lung cancer", CANCER J, vol. 19, 2013, pages 200 - 207
WU CT; CHEN MF; CHEN WC; HSIEH CC.: "The role of IL-6 in the radiation response of prostate cancer", RADIAT ONCOL, vol. 8, 2013, pages 159, XP021157069, DOI: doi:10.1186/1748-717X-8-159
WU ET AL., MOL. BIOL., vol. 294, 1999, pages 151
WU ET AL.: "Antibody Engineering", 2010, SPRINGER BERLIN HEIDELBERG, article "Generation and Characterization of a Dual Variable Domain Immunoglobulin (DVD-IgTM) Molecule"
XU Y; WEI K; KULYK W; GONG SG.: "FLRT2 promotes cellular proliferation and inhibits cell adhesion during chondrogenesis", J CELL BIOCHEM, vol. 112, 2011, pages 3440 - 3448
YANASE M; KATO K; YOSHIZAWA K; NOGUCHI N; KITAHARA H; NAKAMURA H.: "Prognostic value of vascular endothelial growth factors A and C in oral squamous cell carcinoma", J ORAL PATHOL MED, vol. 43, 2014, pages 514 - 520
ZACHARY I.: "Neuropilins: role in signalling, angiogenesis and disease", CHEM IMMUNOL ALLERGY, vol. 99, 2014, pages 37 - 70
ZHANG B; GAO Z; SUN M; LI H; FAN H; CHEN D ET AL.: "Prognostic significance of VEGF-C, semaphorin 3F, and neuropilin-2 expression in oral squamous cell carcinomas and their relationship with lymphangiogenesis", J SURG ONCOL, vol. 111, 2015, pages 382 - 388

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3885452A1 (fr) * 2020-03-23 2021-09-29 Koninklijke Philips N.V. Prédiction de la réponse à la radiothérapie pour un sujet atteint d'un cancer de la prostate sur la base des gènes de chimiokine
WO2021190940A1 (fr) 2020-03-23 2021-09-30 Koninklijke Philips N.V. Prédiction de réponse à la radiothérapie pour un sujet atteint d'un cancer de la prostate sur la base de gènes de chimiokine
CN114681609A (zh) * 2022-05-05 2022-07-01 浙江大学 抗il-6抗体组合物在制备治疗肝细胞癌药物中的应用

Similar Documents

Publication Publication Date Title
US20180044422A1 (en) Treating solid tumor by targeting dectin-1 signaling
US20230088070A1 (en) Use of il-1beta binding antibodies
US20190263927A1 (en) Combination of a pd-1 antagonist and eribulin for treating urothelial cancer
WO2018185516A1 (fr) Méthodes et compositions pharmaceutiques destinées à traiter une toxicité cardiovasculaire induite par une thérapie anticancéreuse
WO2014153018A1 (fr) Utilisation de biomarqueurs du récepteur egfr pour le traitement du cancer de l'estomac par des agents anti-egfr
WO2018189403A1 (fr) Méthodes et compositions pharmaceutiques destinées au traitement du cancer
WO2019106126A1 (fr) Modulateurs mdm2 pour le diagnostic et le traitement du liposarcome
WO2019234221A1 (fr) Procédés de stratification et de traitement d'un patient souffrant de leucémie lymphoïde chronique
WO2019211370A1 (fr) Méthodes et compositions pharmaceutiques pour le traitement du cancer
US20170115293A1 (en) Methods for predicting the radiosensitivity of a cancer tumor and methods of treating cancer
US20210047696A1 (en) Methods and pharmaceutical compositions for treating cancer
US20210164984A1 (en) Methods for predicting outcome and treatment of patients suffering from prostate cancer or breast cancer
US20220290151A1 (en) Use of müllerian inhibiting substance inhibitors for treating cancer
WO2023165618A1 (fr) Méthodes de traitement du cancer
US20220025036A1 (en) Use of il-1beta binding antibodies
US20230266332A1 (en) Methods and compositions for preventing and treating a cancer
US20230076415A1 (en) Methods and compositions for treating melanoma
US20230250173A1 (en) Biomarkers for pd-1 axis binding antagonist therapy
WO2023078900A1 (fr) Méthodes et compositions pour traiter le cancer du sein triple négatif (tnbc)
US20210290633A1 (en) Combination for treating cancer
WO2020128637A1 (fr) UTILISATION D'ANTICORPS DE LIAISON À IL-1β DANS LE TRAITEMENT D'UN CANCER MSI-H
WO2020245054A1 (fr) Antagoniste de liaison à l'axe pd-1 pour traiter le cancer avec des mutations génétiques dans des gènes spécifiques
PA session A: gastrointestinal tumours

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: 18721291

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: 18721291

Country of ref document: EP

Kind code of ref document: A1