WO2012123774A1 - Glycolytic inhibitor with cytotoxic agent for use in the treatment of a cancer - Google Patents

Glycolytic inhibitor with cytotoxic agent for use in the treatment of a cancer Download PDF

Info

Publication number
WO2012123774A1
WO2012123774A1 PCT/IB2011/000795 IB2011000795W WO2012123774A1 WO 2012123774 A1 WO2012123774 A1 WO 2012123774A1 IB 2011000795 W IB2011000795 W IB 2011000795W WO 2012123774 A1 WO2012123774 A1 WO 2012123774A1
Authority
WO
WIPO (PCT)
Prior art keywords
cancer
cytotoxic agent
glycolytic inhibitor
inhibitor
group
Prior art date
Application number
PCT/IB2011/000795
Other languages
French (fr)
Inventor
Jean-Ehrland RICCI
Original Assignee
INSERM (Institut National de la Santé et de la Recherche Médicale)
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) filed Critical INSERM (Institut National de la Santé et de la Recherche Médicale)
Priority to PCT/IB2011/000795 priority Critical patent/WO2012123774A1/en
Publication of WO2012123774A1 publication Critical patent/WO2012123774A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K33/00Medicinal preparations containing inorganic active ingredients
    • A61K33/24Heavy metals; Compounds thereof
    • A61K33/243Platinum; Compounds thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • A61K31/407Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil condensed with other heterocyclic ring systems, e.g. ketorolac, physostigmine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/4151,2-Diazoles
    • A61K31/4161,2-Diazoles condensed with carbocyclic ring systems, e.g. indazole
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41641,3-Diazoles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/473Quinolines; Isoquinolines ortho- or peri-condensed with carbocyclic ring systems, e.g. acridines, phenanthridines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/555Heterocyclic compounds containing heavy metals, e.g. hemin, hematin, melarsoprol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7004Monosaccharides having only carbon, hydrogen and oxygen atoms
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7012Compounds having a free or esterified carboxyl group attached, directly or through a carbon chain, to a carbon atom of the saccharide radical, e.g. glucuronic acid, neuraminic acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7028Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages
    • A61K31/7034Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages attached to a carbocyclic compound, e.g. phloridzin
    • A61K31/704Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages attached to a carbocyclic compound, e.g. phloridzin attached to a condensed carbocyclic ring system, e.g. sennosides, thiocolchicosides, escin, daunorubicin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7042Compounds having saccharide radicals and heterocyclic rings
    • A61K31/7048Compounds having saccharide radicals and heterocyclic rings having oxygen as a ring hetero atom, e.g. leucoglucosan, hesperidin, erythromycin, nystatin, digitoxin or digoxin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K33/00Medicinal preparations containing inorganic active ingredients
    • A61K33/24Heavy metals; Compounds thereof
    • A61K33/36Arsenic; Compounds thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/04Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
    • A61K38/14Peptides containing saccharide radicals; Derivatives thereof, e.g. bleomycin, phleomycin, muramylpeptides or vancomycin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca

Definitions

  • the present invention relates to a method for use in the treatment cancer.
  • Chemotherapeutic cytotoxic agents are the main clinical tool to control invasive malignancy.
  • the classes of cytotoxic agents used most widely in the oncology clinic are DNA-damaging agents; antimetabolites ; and antimitotics (Cancer immunotherapy : immune suppression and tumor growth, George C. et al.) (Chemotherapy at Dorland's Medical Dictionary)).
  • DNA- damaging agents may be alkylating agents, topoisomerase inhibitors or platinum compounds.
  • -Alkylating agents (Anatomical Therapeutic Chemical Classification (ATC) code L01A) are so named because of their ability to alkylate many nucleophilic functional groups under conditions present in cells. They impair cell function by forming covalent bonds with the amino, carboxyl, sulfhydryl, and phosphate groups in biologically important molecules.
  • alkylating agents examples include cyclophosphamide, chlorambucil, chlormethine, busulfan, treosulfan and thiotepa.
  • ATC code LOICB and L01XX are agents that blocks type I or type II topoisomerases interfering thus with both transcription and replication of DNA by upsetting proper DNA supercoiling.
  • type I topoisomerase inhibitors are camptothecins, irinotecan and topotecan.
  • type II inhibitors are amsacrine, etoposide, etoposide phosphate, and teniposide.
  • -Platinum compounds damage DNA by creating intrastrand and interstrand cross-links.
  • platinum compounds are cisplatin, carboplatin, oxaliplatin.
  • Anti-tumor antibiotics include mainly anthracyclines.
  • anthracyclines examples include doxorubicin, epirubicin, idarubicin, mitoxantrone, valrubicin.
  • Anti-metabolites (ATC code L01B) :
  • Anti-metabolites are similar in structure to naturally occurring compounds that are required for the viability and division of a cell.
  • the efficacy of the most important anti-metabolites against a range of tumor cells is based on the inhibition of purine or pyrimidine nucleoside synthesis pathway that are required for DNA synthesis.
  • Anti-metabolites can be divided into several classes, including folate antagonists such as methotrexate, purine antagonists such as fludarabine and pyrimidine antagonists such as 5-fluorouracil.
  • folate antagonists such as methotrexate
  • purine antagonists such as fludarabine
  • pyrimidine antagonists such as 5-fluorouracil.
  • ATC code L01CD
  • taxanes examples include paclitaxel, and docetaxel.
  • -Vinca alkaloids ATC code : L01CA
  • -Vinca alkaloids bind to specific sites on tubulin, inhibiting the assembly of tubulin into microtubules (M phase of the cell cycle). They are derived from the Madagascar periwinkle, Catharanthus roseus (formerly known as Vinca rosea).
  • vinca alkaloids examples include vincristine, vinblastine, vinorelbine,vindesine.
  • cancer therapies must target all tumor cells, including cancer stem cells or they must remove a fraction of the tumor and be accompanied by a "bystander effect" in which the immune system recognizes, attacks and eradicates the remaining tumor cells including cancer stem cells and chemotherapy resistant cancer cells.
  • the present invention relates to a method for the treatment of a cancer selected from the group consisting of colorectal cancer, cervical cancer and melanoma in a subject in need thereof comprising administering to said subject an effective amount of glycolytic inhibitor and an effective amount of cytotoxic agent.
  • the present invention relates to a glycolytic inhibitor for use in the treatment of a cancer selected from the group consisting of colorectal cancer, cervical cancer and melanoma in a subject to whom a cytotoxic agent is administered.
  • the present invention also provides a kit for use in the treatment of a cancer selected from the group consisting of colorectal cancer, cervical cancer and melanoma comprising : (a) glycolytic inhibitor ; and (b) a cytotoxic agent.
  • the present invention relates to a method for the treatment of a cancer selected from the group consisting of colorectal cancer, cervical cancer and melanoma in a subject in need thereof comprising administering to said subject an effective amount of glycolytic inhibitor and an effective amount of cytotoxic agent.
  • the present invention relates to a glycolytic inhibitor for use in the treatment of a cancer selected from the group consisting of colorectal cancer, cervical cancer and melanoma in a subject to whom a cytotoxic agent is administered.
  • the presence of a glycolytic inhibitor, in combination with a cytotoxic agent enables to extend the cytotoxic effect of the cytotoxic agent by inducing an anticancer immune response.
  • Glycolysis refers to the series of enzymatic reactions which convert a molecule of glucose into lactate with the generation of two molecules of ATP. Glycolytic inhibitors are being designed that target the enzymes involved in the glycolysis pathway.
  • Glycolytic inhibitors are well known in the state of art and some are developed in cancer therapy (see, for example, Pelicano H et al. (2006) ; Pathania D et al. (2009)) ; El Mjiyad N et al. (2011)).
  • the present invention relates to a cytotoxic agent for use in the treatment of a cancer selected from the group consisting of colorectal cancer, cervical cancer and melanoma in a subject to whom a glycolytic inhibitor is administered.
  • the present invention relates to the use of a glycolytic inhibitor for the preparation of a medicament for the treatment of a cancer selected from the group consisting of colorectal cancer, cervical cancer and melanoma in a subject to whom a cytotoxic agent is administered.
  • the present invention relates to the use of a cytotoxic agent for the preparation of a medicament for the treatment of a cancer selected from the group consisting of colorectal cancer, cervical cancer and melanoma in a subject to whom a glycolytic inhibitor is administered.
  • the subject is a subject with stage III or stage IV cancer.
  • the cancer according to the invention is a colorectal cancer.
  • colorectal cancer standard treatment with chemotherapy is quite effective in stage I disease and stage II disease. Indeed, 5 years survival rates approach 90% for colon cancer.
  • stage III disease denotes lymph node involvement
  • studies indicate that the number of lymph nodes involved affects prognosis (55% to 20%: 5 year survival) and stage IV colon cancer (metastatic) clearly has the worst prognosis ( ⁇ 5% 5 year survival) (National Cancer Institute).
  • the subject is a subject with stage III or stage IV colorectal cancer.
  • the cancer according to the invention is cervical cancer.
  • the 5-year relative survival rate for the earliest stage of invasive cervical cancer is superior to 90%. This survival rate decreases importantly to 25 to 35% of women with stage III cancer and 15% or fewer of those with stage IV cancer are alive after 5 years.
  • the subject is a subject with stage III or stage IV cervical cancer.
  • the cancer according to the invention is melanoma.
  • the subject is a subject with stage III or stage IV melanoma.
  • the cytotoxic agent according to the invention is a cytotoxic agent which does not induce an anticancer immune response.
  • a "cytotoxic agent which induces an anticancer immune response” refers to a cytotoxic agent which, upon administration to tumor cells induces the dying cells to express tumor antigen(s) which elicits a protective response when said dying cells are injected in an animal in the absence of any adjuvant (Casares et al., 2005; Martins et al., 2010). When living cells expressing the tumor antigen(s) is (are) injected into the animal, such a protective immune response precludes the growth of these living cells.
  • a "cytotoxic agent which does not induce an anticancer immune response” refers to the contrary to a cytotoxic agent which, when administered to cells, does not induce the dying cells to elicit a protective response in an animal in which they are injected (Casares et al., 2005; Martins et al., 2010).
  • cytotoxic agents which induce an anticancer immune response are anti- cancer agents which induce the expression of at least one tumor antigen selected from the group consisting of calreticuhn, Heat Shock Proteins (HSPs) or High-mobility group box 1 (HMGB1).
  • HSPs Heat Shock Proteins
  • HMGB1 High-mobility group box 1
  • oxaliplatin and anthracyclines such as doxorubicin are known to be cytotoxic agents which induce an anticancer immune response (Obeid M. et al. (2007); Casares N. et al. (2005) ; Martins I. et al., (2010)).
  • etoposide, cisplatin and mitomycin are known to be cytotoxic agents which do not induce an anticancer immune response (Obeid M. et al. (2007); Casares N. et al. (2005) ; Martins I. et al., (2010)).
  • the glycolytic inhibitor according to the present invention is a hexokinase (HK) inhibitor.
  • Hexokinase catalyzes the first step of glycolysis, phosphorylating hexose (e.g. glucose) to hexose-6-phosphate.
  • hexose e.g. glucose
  • Hexokinase inhibitors are well known in the state of art (see, for example,
  • the glycolytic inhibitor is a HK inhibitor selected from the group consisting of 2-deoxyglucose, 5-thioglucose, mannoheptulose, 3- bromopyruvate, lonidamine and methyl jasmonate.
  • glycolytic inhibitor is a glyceraldehyde-3 phosphate dehydrogenase (GAPDH) inhibitor.
  • GPDH glyceraldehyde-3 phosphate dehydrogenase
  • GAPDH converts glyceraldehydes-3 -phosphate to 1,3-bisphospho-glycerate with a simultaneous reduction of NAD+ to NADH (nicotin-amide adenine dinucleotide; oxidized and reduced respectively).
  • GAPDH inhibitors such as, for example, alpha-chlorhydrin, ornidazole, arsenic, iodoacetate, koningic acid (see, for example, Pelicano H et al. (2006) ; Pathania D et al. (2009) ; El Mjiyad N et al. (2011)).
  • glycolytic inhibitor is a GAPDH inhibitor selected from the group consisting of alpha-chlorhydrin, ornidazole, arsenic, iodoacetate, koningic acid.
  • the cytotoxic agent is a topoisomerase inhibitor. In a preferred embodiment, the cytotoxic agent is a topoisomerase II inhibitor.
  • the cytotoxic agent is a topoisomerase II inhibitor selected from the group consisting of amsacrine, etoposide, etoposide phosphate, and teniposide.
  • the cytotoxic agent is a topoisomerase I inhibitor.
  • the cytotoxic agent is an anti-tumor antibiotic.
  • the cytotoxic agent is an anti-tumor antibiotic selected in the group consisting from mitomycin, bleomycin and plicamycin.
  • the cytotoxic agent is a platinum compound.
  • the cytotoxic agent is a platinum compound selected from the group consisting of cisplatin and carboplatin.
  • the present invention also relates to a composition for use in the treatment of a cancer selected from the group consisting of colorectal cancer, cervical cancer and melanoma comprising a glycolytic inhibitor and a cytotoxic agent.
  • the cytotoxic agent of the composition according to the invention is a cytotxic agent which doesn't induce anticancer immune response.
  • the present invention also relates to a kit for use in the treatment of a cancer selected from the group consisting of colorectal cancer, cervical cancer and melanoma comprising :
  • the cytotoxic agent of the kit according to the invention is a cytotxic agent which doesn't induce anticancer immune response.
  • the glycolytic inhibitor and the cytotoxic agent are administered at the same time. In another embodiment, the glycolytic inhibitor and the cytotoxic agent are administered sequentially.
  • they can be administrated at different moments of the day.
  • FIG. 1 Toxicity of various concentrations of Etoposide (ETO) with or without 2- deoxy-glucose (2DG) on ⁇ -myc cells.
  • ETO Etoposide
  • 2DG 2- deoxy-glucose
  • FIG. 1 Toxicity of various concentrations of Etoposide (ETO) with or without 2- deoxy-glucose (2DG) on CT26.
  • ETO Etoposide
  • 2DG 2- deoxy-glucose
  • FIG. 3 Toxicity of various concentrations of Etoposide (ETO) with or without lonidamine (LND) on CT26.
  • ETO Etoposide
  • LND lonidamine
  • Figure 4 Toxicity of various concentrations of Mytomycin (mitom-c) with or without lonidamine (LND) on CT26.
  • Figure 5 Toxicity of various concentrations of Mytomycin (mitom-c) with or without 3-bromopyruvic acid (3-BrPA) on CT26.
  • Figure 6 Size of lymph nodes as a function of treatment with PBS, 2DG, ETO or ETO+2DG (IP 3times/week; 2DG : 500mg/kg; ETO 2.5mg/kg).
  • Figure 7 Survival probability as a function of treatment with PBS, 2DG, ETO or ETO+2DG.
  • Figure 8 Time of relapse as a function of treatment with ETO or ETO+2DG.
  • CT26, B 16 and HeLa cells were obtained from ATCC and cultured in RPMI 1640 or DMEM medium supplemented with 10% fetal calf serum, 1 mM pyruvate, 10 mM HEPES and 100 Units/ml each of penicillin and streptomycin. Cells were incubated at 37°C under 5% C02.
  • TRC Etoposide
  • Mytomicin c Sigma
  • Mitoxanthrone Sigma
  • 2 deoxy glucose TRC
  • 3-Bromopyruvic acid Sigma
  • Lonidamine Tocris
  • DAPI Molecular Probes, 0.5 ⁇ g/ml. Cell death was analyzed immediately by flow cytometry and definied as DAPI+ cells using a MACSQuant Analyzer (Miltenyi Biotec).
  • mice were maintained in specific pathogen-free conditions and all experiments followed the guidelines of the federation of European Animal Science Association. All animal experiments were approved by the Institutional Animal Care and Use Committee of the Centre Mediterraneen de Medecine Mole vide (INSERM U895).
  • C57BL/6 ⁇ -myc transgenic mice were purchased from Jackson Laboratories and were genotyped by PCR according to instructions from the supplier. Lymphoma bearing animals were killed by cervical dislocation as soon as any signs of illness can be detected. A single cell suspension was prepared from lymph nodes by teasing them on a 70 ⁇ nylon filter. Cells were either resuspended in complete medium (DMEM supplemented with 10% FCS, 10 mM HEPES, penicillin/streptomycin, 0.1 mM L-asparagine and 50 ⁇ 2-mercaptoethanol) for further ex vivo analysis or directly reimplanted in wild type mice.
  • complete medium DMEM supplemented with 10% FCS, 10 mM HEPES, penicillin/streptomycin, 0.1 mM L-asparagine and 50 ⁇ 2-mercaptoethanol
  • Lymphoma transfers were realized into syngenic, non-transgenic, 6-10 weeks old C57BL/6 females by tail vein injection (0.5xl0 6 viable cells per mouse, in 200 ⁇ sterile PBS). Recipient mice were monitored thrice a week for lymph nodes enlargement. As the inguinal lymph nodes reached 5 mm in the longest diameter, the animals were intraperitoneally injected, three times a week for the indicated period of time, with PBS, etoposide (2.5 mg/kg), 2DG (500 mg/kg) or both compounds and subsequently monitored for treatment response.
  • PBS etoposide
  • 2DG 500 mg/kg
  • the survival of the mice and, for some experiments, the weight of the lymph nodes were measured.
  • the "time of relapse" reflects the time between remission and re- palpability of a recurrent lymph-node enlargement.
  • mice BALB/c height-week-old female mice (Harlan) were injected subcutaneously on the left flank with lxlO 6 CT26 cells treated either with mitoxantrone, or with etoposide or with 2DG and etoposide until reaching 80% DAPI positive cells for each treatment.
  • mice were challenged by injecting 0.5x10 6 live cells on the contralateral flank. The animals were then checked twice a week for tumor development using calipers.
  • CT26 cell lines murin colon carcinomas
  • isolated primary non-Hodgkin lymphoma cells from ⁇ -myc mice were used.
  • Those transgenic animals are presenting a deregulated c-myc expression under the control of the immunoglobulin heavy chain (IgH) gene enhancer ( ⁇ ) causes abnormal growth and proliferation of pre-B and B cells and prevents differentiation, culminating in clonal pre-B or immature B-cell lymphoma.
  • IgH immunoglobulin heavy chain
  • gene enhancer
  • Genetics and histopathology of ⁇ -myc lymphomas resemble human non-Hodgkin's lymphomas (NHL) (Adams et al., 1985).
  • mice received 0.5x10 6 ⁇ -myc cells I.V. 20 days later all mice presented enlarged lymph nodes indicating the induction of lymphoma. Then mice were treated with PBS, 2DG, Eto or Eto+2DG. In a first experiment, mice were sacrificed 21 days following the beginning of the treatment and lymph-nodes enlargement was measured. Co-treated animals were presenting significantly smaller lymph nodes than Eto- or control- treated mice.
  • Cytotoxic agent combined with glycolytic inhibitor converts a non- immunogenic cancer cell death stimulus to an immunogenic one by controlling calreticulin exposure.
  • Figure 9B HeLa cells (Human cervical cancer cell line), 9C: B16 cells (murin melanoma cell lines) and 9D: CT26 (murin colon carcinoma cell lines).
  • Eto+2DG was inducing a significant increase of CRT exposure compared to Eto or 2DG alone.
  • an increase of CRT exposure to the plasma membrane could also be observed when Eto was combined with other hexokinase inhibitors: LND ( Figure 9E) or 3-BrPA ( Figure 9F).
  • CT26 cells were killed by Eto, Eto+2DG or mitoxantrone (as a positive control) until reaching 80% dead cells in each condition. Then lxl0 6 dead CT26 cells were injected into one flank of BALB/c immuno-competent syngenic mice. One week later, 0.5x10 6 of CT26 live cells were injected into the contra lateral flank of the mice. Therefore in absence of vaccination the mice will develop a tumor whereas if vaccinated, mice should be protected.
  • Figure 10 described that, as expected, 100% of non-vaccinated mice were dying within 25 days following the challenge with alive CT26 cells.
  • mice vaccinated with Eto-treated cells could survive up to 90 days. Very importantly, this percentage jump up to 66% in the group of mice vaccinated with Eto+2DG. Of note, the protection given by the co-treatment is very close to the one induced by mitoxantrone (75%).
  • Anthracyclines molecular advances and pharmacologic developments in antitumor activity and cardiotoxicity.
  • Calreticulin exposure dictates the immunogenicity of cancer cell death. Nat Med 13, 54-61.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Chemical & Material Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Molecular Biology (AREA)
  • Inorganic Chemistry (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Immunology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The present invention relates to a glycolytic inhibitor for use in the treatment of a cancer selected from the group consisting of colorectal cancer, cervical cancer and melanoma in a subject to whom a cytotoxic agent is administered.

Description

GLYCOLYTIC INHIBITOR WITH CYTOTOXIC AGENT
FOR USE IN THE TREATMENT OF A CANCER
FIELD OF THE INVENTION
The present invention relates to a method for use in the treatment cancer.
BACKGROUND OF THE INVENTION
Chemotherapeutic cytotoxic agents are the main clinical tool to control invasive malignancy. The classes of cytotoxic agents used most widely in the oncology clinic are DNA-damaging agents; antimetabolites ; and antimitotics (Cancer immunotherapy : immune suppression and tumor growth, George C. et al.) (Chemotherapy at Dorland's Medical Dictionary)).
DNA-damaging agents :
DNA- damaging agents may be alkylating agents, topoisomerase inhibitors or platinum compounds.
-Alkylating agents (Anatomical Therapeutic Chemical Classification (ATC) code L01A) are so named because of their ability to alkylate many nucleophilic functional groups under conditions present in cells. They impair cell function by forming covalent bonds with the amino, carboxyl, sulfhydryl, and phosphate groups in biologically important molecules.
Examples of alkylating agents are cyclophosphamide, chlorambucil, chlormethine, busulfan, treosulfan and thiotepa.
-Topoisomerase inhibitors (ATC code LOICB and L01XX) are agents that blocks type I or type II topoisomerases interfering thus with both transcription and replication of DNA by upsetting proper DNA supercoiling.
Examples of type I topoisomerase inhibitors are camptothecins, irinotecan and topotecan. Examples of type II inhibitors are amsacrine, etoposide, etoposide phosphate, and teniposide.
-Platinum compounds damage DNA by creating intrastrand and interstrand cross-links.
Examples of platinum compounds are cisplatin, carboplatin, oxaliplatin.
Anti-tumor antibiotics :
Anti-tumor antibiotics include mainly anthracyclines.
-Anthracyclines (ATC code : L01DB )
Examples of anthracyclines are doxorubicin, epirubicin, idarubicin, mitoxantrone, valrubicin.
-Other anti-tumor antibiotics (ATC code : L01DC)
Examples of other anti-tumor antibiotics are mitomycin, bleomycin and plicamycin. Anti-metabolites (ATC code L01B) :
Anti-metabolites are similar in structure to naturally occurring compounds that are required for the viability and division of a cell. The efficacy of the most important anti-metabolites against a range of tumor cells is based on the inhibition of purine or pyrimidine nucleoside synthesis pathway that are required for DNA synthesis.
Anti-metabolites can be divided into several classes, including folate antagonists such as methotrexate, purine antagonists such as fludarabine and pyrimidine antagonists such as 5-fluorouracil.
Antimitotics :
-Taxanes (ATC code : L01CD) interfere with microtubules. They block cell growth by stopping mitosis.
Examples of taxanes are paclitaxel, and docetaxel. -Vinca alkaloids (ATC code : L01CA) bind to specific sites on tubulin, inhibiting the assembly of tubulin into microtubules (M phase of the cell cycle). They are derived from the Madagascar periwinkle, Catharanthus roseus (formerly known as Vinca rosea).
Examples of vinca alkaloids are vincristine, vinblastine, vinorelbine,vindesine.
Conventional anticancer chemotherapies are generally thought to reduce tumor progression by direct cytostatic and cytotoxic effects on tumor cells.
However, even after an initial therapeutic success, some patients fail to display a clinically relevant antitumor immune response and succumb to tumor cell variants that escape from chemotherapy. (Casares et al. (2005); Obeid et al. (2007))
To completely and permanently succeed against cancers, cancer therapies must target all tumor cells, including cancer stem cells or they must remove a fraction of the tumor and be accompanied by a "bystander effect" in which the immune system recognizes, attacks and eradicates the remaining tumor cells including cancer stem cells and chemotherapy resistant cancer cells.
There is a growing consensus that the use of an individual chemotherapeutic cytotoxic agent is unlikely to succeed as a therapeutic strategy in the long run. The best approach to circumvent such problem would be to identify a way not only to induce an efficient and specific death of the tumor cells but also to educate the immune system to react against any remaining tumor cells; thereby extending the cytotoxic effect of the chemotherapy and preventing metastasis apparition.
SUMMARY OF THE INVENTION The inventors have demonstrated that a treatment combining a glycolytic inhibitor and a cytotoxic agent will represent an ideal way to reach such goal, in particular in colorectal cancer, cervical cancer and melanoma The present invention relates to a method for the treatment of a cancer selected from the group consisting of colorectal cancer, cervical cancer and melanoma in a subject in need thereof comprising administering to said subject an effective amount of glycolytic inhibitor and an effective amount of cytotoxic agent.
The present invention relates to a glycolytic inhibitor for use in the treatment of a cancer selected from the group consisting of colorectal cancer, cervical cancer and melanoma in a subject to whom a cytotoxic agent is administered.
The present invention also provides a kit for use in the treatment of a cancer selected from the group consisting of colorectal cancer, cervical cancer and melanoma comprising : (a) glycolytic inhibitor ; and (b) a cytotoxic agent.
DETAILED DESCRIPTION OF THE INVENTION
The present invention relates to a method for the treatment of a cancer selected from the group consisting of colorectal cancer, cervical cancer and melanoma in a subject in need thereof comprising administering to said subject an effective amount of glycolytic inhibitor and an effective amount of cytotoxic agent.
The present invention relates to a glycolytic inhibitor for use in the treatment of a cancer selected from the group consisting of colorectal cancer, cervical cancer and melanoma in a subject to whom a cytotoxic agent is administered.
In the method of treatment according to the invention, the presence of a glycolytic inhibitor, in combination with a cytotoxic agent, enables to extend the cytotoxic effect of the cytotoxic agent by inducing an anticancer immune response.
Glycolysis refers to the series of enzymatic reactions which convert a molecule of glucose into lactate with the generation of two molecules of ATP. Glycolytic inhibitors are being designed that target the enzymes involved in the glycolysis pathway.
Glycolytic inhibitors are well known in the state of art and some are developed in cancer therapy (see, for example, Pelicano H et al. (2006) ; Pathania D et al. (2009)) ; El Mjiyad N et al. (2011)).
Indeed, tumor cells undergo accelerated aerobic glycolysis (Warburg effect). This peculiar metabolism of the cancer cells has led the field to develop new therapeutic strategies using glycolytic inhibitors (Scatena et al., 2008). The present invention relates to a cytotoxic agent for use in the treatment of a cancer selected from the group consisting of colorectal cancer, cervical cancer and melanoma in a subject to whom a glycolytic inhibitor is administered.
The present invention relates to the use of a glycolytic inhibitor for the preparation of a medicament for the treatment of a cancer selected from the group consisting of colorectal cancer, cervical cancer and melanoma in a subject to whom a cytotoxic agent is administered.
The present invention relates to the use of a cytotoxic agent for the preparation of a medicament for the treatment of a cancer selected from the group consisting of colorectal cancer, cervical cancer and melanoma in a subject to whom a glycolytic inhibitor is administered.
In one preferred embodiment, the subject is a subject with stage III or stage IV cancer.
In a particular embodiment, the cancer according to the invention is a colorectal cancer. In case of colorectal cancer, standard treatment with chemotherapy is quite effective in stage I disease and stage II disease. Indeed, 5 years survival rates approach 90% for colon cancer.
However, stage III disease denotes lymph node involvement, and studies indicate that the number of lymph nodes involved affects prognosis (55% to 20%: 5 year survival) and stage IV colon cancer (metastatic) clearly has the worst prognosis (<5% 5 year survival) (National Cancer Institute).
In one preferred embodiment, the subject is a subject with stage III or stage IV colorectal cancer.
In a particular embodiment, the cancer according to the invention is cervical cancer.
In case of cervical cancer, with treatment, the 5-year relative survival rate for the earliest stage of invasive cervical cancer is superior to 90%. This survival rate decreases importantly to 25 to 35% of women with stage III cancer and 15% or fewer of those with stage IV cancer are alive after 5 years.
In one preferred embodiment, the subject is a subject with stage III or stage IV cervical cancer.
In a particular embodiment, the cancer according to the invention is melanoma.
Melanoma confined to the skin is curable in 95% to 98% of cases but the prognosis become dismal when the lesion recurs, or spreads from the skin to the lymph nodes or distant organs. Indeed, until now, no treatment has increased survival significantly in melanoma patients with metastases. Average survival for Stage III and IV patients has been about eight months. In one preferred embodiment, the subject is a subject with stage III or stage IV melanoma.
In one embodiment, the cytotoxic agent according to the invention is a cytotoxic agent which does not induce an anticancer immune response.
A "cytotoxic agent which induces an anticancer immune response" refers to a cytotoxic agent which, upon administration to tumor cells induces the dying cells to express tumor antigen(s) which elicits a protective response when said dying cells are injected in an animal in the absence of any adjuvant (Casares et al., 2005; Martins et al., 2010). When living cells expressing the tumor antigen(s) is (are) injected into the animal, such a protective immune response precludes the growth of these living cells.
A "cytotoxic agent which does not induce an anticancer immune response" refers to the contrary to a cytotoxic agent which, when administered to cells, does not induce the dying cells to elicit a protective response in an animal in which they are injected (Casares et al., 2005; Martins et al., 2010).
One method for evaluating if an agent "induces an anticancer immune response" is well described by Casares N. et al. (2005).
Typically, cytotoxic agents which induce an anticancer immune response are anti- cancer agents which induce the expression of at least one tumor antigen selected from the group consisting of calreticuhn, Heat Shock Proteins (HSPs) or High-mobility group box 1 (HMGB1). The expression of said tumor antigen(s) can be detected by standard methods known in the art such as RT-PCR, immunoblot etc...
Typically, oxaliplatin and anthracyclines such as doxorubicin are known to be cytotoxic agents which induce an anticancer immune response (Obeid M. et al. (2007); Casares N. et al. (2005) ; Martins I. et al., (2010)). On the contrary, etoposide, cisplatin and mitomycin are known to be cytotoxic agents which do not induce an anticancer immune response (Obeid M. et al. (2007); Casares N. et al. (2005) ; Martins I. et al., (2010)).
In one embodiment, the glycolytic inhibitor according to the present invention is a hexokinase (HK) inhibitor.
Hexokinase catalyzes the first step of glycolysis, phosphorylating hexose (e.g. glucose) to hexose-6-phosphate.
Hexokinase inhibitors are well known in the state of art (see, for example,
Pelicano H et al. (2006) ; Pathania D et al. (2009)) ; El Mjiyad N et al. (2011)).
In a preferred embodiment, the glycolytic inhibitor is a HK inhibitor selected from the group consisting of 2-deoxyglucose, 5-thioglucose, mannoheptulose, 3- bromopyruvate, lonidamine and methyl jasmonate.
In another embodiment, glycolytic inhibitor is a glyceraldehyde-3 phosphate dehydrogenase (GAPDH) inhibitor.
GAPDH converts glyceraldehydes-3 -phosphate to 1,3-bisphospho-glycerate with a simultaneous reduction of NAD+ to NADH (nicotin-amide adenine dinucleotide; oxidized and reduced respectively).
In the state of art, there are many well known GAPDH inhibitors such as, for example, alpha-chlorhydrin, ornidazole, arsenic, iodoacetate, koningic acid (see, for example, Pelicano H et al. (2006) ; Pathania D et al. (2009) ; El Mjiyad N et al. (2011)). Several additional GAPDH inhibitors are currently under development. Preferably, glycolytic inhibitor is a GAPDH inhibitor selected from the group consisting of alpha-chlorhydrin, ornidazole, arsenic, iodoacetate, koningic acid.
In one embodiment, the cytotoxic agent is a topoisomerase inhibitor. In a preferred embodiment, the cytotoxic agent is a topoisomerase II inhibitor.
In a more preferred embodiment, the cytotoxic agent is a topoisomerase II inhibitor selected from the group consisting of amsacrine, etoposide, etoposide phosphate, and teniposide.
In another embodiment, the cytotoxic agent is a topoisomerase I inhibitor.
In another embodiment, the cytotoxic agent is an anti-tumor antibiotic.
In a preferred embodiment, the cytotoxic agent is an anti-tumor antibiotic selected in the group consisting from mitomycin, bleomycin and plicamycin.
In another embodiment, the cytotoxic agent is a platinum compound.
In a preferred embodiment, the cytotoxic agent is a platinum compound selected from the group consisting of cisplatin and carboplatin.
The present invention also relates to a composition for use in the treatment of a cancer selected from the group consisting of colorectal cancer, cervical cancer and melanoma comprising a glycolytic inhibitor and a cytotoxic agent.
In one embodiment, the cytotoxic agent of the composition according to the invention is a cytotxic agent which doesn't induce anticancer immune response.
The present invention also relates to a kit for use in the treatment of a cancer selected from the group consisting of colorectal cancer, cervical cancer and melanoma comprising :
a- a glycolytic inhibitor ; and
b- a cytotoxic agent.
In one embodiment, the cytotoxic agent of the kit according to the invention is a cytotxic agent which doesn't induce anticancer immune response. In one embodiment the glycolytic inhibitor and the cytotoxic agent are administered at the same time. In another embodiment, the glycolytic inhibitor and the cytotoxic agent are administered sequentially.
For example, they can be administrated at different moments of the day.
In the following, the invention will be illustrated by means of the following examples as well as the figures.
FIGURE LEGENDS
Figure 1 : Toxicity of various concentrations of Etoposide (ETO) with or without 2- deoxy-glucose (2DG) on Εμ-myc cells.
Figure 2 : Toxicity of various concentrations of Etoposide (ETO) with or without 2- deoxy-glucose (2DG) on CT26.
Figure 3 : Toxicity of various concentrations of Etoposide (ETO) with or without lonidamine (LND) on CT26.
Figure 4 : Toxicity of various concentrations of Mytomycin (mitom-c) with or without lonidamine (LND) on CT26.
Figure 5 : Toxicity of various concentrations of Mytomycin (mitom-c) with or without 3-bromopyruvic acid (3-BrPA) on CT26.
Figure 6 : Size of lymph nodes as a function of treatment with PBS, 2DG, ETO or ETO+2DG (IP 3times/week; 2DG : 500mg/kg; ETO 2.5mg/kg).
Figure 7 : Survival probability as a function of treatment with PBS, 2DG, ETO or ETO+2DG.
Figure 8 : Time of relapse as a function of treatment with ETO or ETO+2DG.
Figure 9 : Exposure of calreticulin as a function of treatment on different cell lines. EXAMPLE
Material and Methods:
Cell culture.
CT26, B 16 and HeLa cells were obtained from ATCC and cultured in RPMI 1640 or DMEM medium supplemented with 10% fetal calf serum, 1 mM pyruvate, 10 mM HEPES and 100 Units/ml each of penicillin and streptomycin. Cells were incubated at 37°C under 5% C02.
To induce cell death, cells were treated for 24 or 48 hours with the indicated amount of Etoposide (TRC), Mytomicin c (Sigma), Mitoxanthrone (Sigma), 2 deoxy glucose (TRC), 3-Bromopyruvic acid (Sigma) or Lonidamine (Tocris). Cell death measurements.
Cells were either trypsinized or centrifugated (Εμ-myc cells), wash in PBS and labelled with DAPI (Molecular Probes, 0.5 μg/ml). Cell death was analyzed immediately by flow cytometry and definied as DAPI+ cells using a MACSQuant Analyzer (Miltenyi Biotec).
Mice experiments
All mice were maintained in specific pathogen-free conditions and all experiments followed the guidelines of the federation of European Animal Science Association. All animal experiments were approved by the Institutional Animal Care and Use Committee of the Centre Mediterraneen de Medecine Moleculaire (INSERM U895).
- Transgenic mice and transplantation of lymphomas
C57BL/6 Εμ-myc transgenic mice were purchased from Jackson Laboratories and were genotyped by PCR according to instructions from the supplier. Lymphoma bearing animals were killed by cervical dislocation as soon as any signs of illness can be detected. A single cell suspension was prepared from lymph nodes by teasing them on a 70 μπι nylon filter. Cells were either resuspended in complete medium (DMEM supplemented with 10% FCS, 10 mM HEPES, penicillin/streptomycin, 0.1 mM L-asparagine and 50 μΜ 2-mercaptoethanol) for further ex vivo analysis or directly reimplanted in wild type mice.
Lymphoma transfers were realized into syngenic, non-transgenic, 6-10 weeks old C57BL/6 females by tail vein injection (0.5xl06 viable cells per mouse, in 200 μΐ sterile PBS). Recipient mice were monitored thrice a week for lymph nodes enlargement. As the inguinal lymph nodes reached 5 mm in the longest diameter, the animals were intraperitoneally injected, three times a week for the indicated period of time, with PBS, etoposide (2.5 mg/kg), 2DG (500 mg/kg) or both compounds and subsequently monitored for treatment response.
The survival of the mice and, for some experiments, the weight of the lymph nodes were measured. The "time of relapse" reflects the time between remission and re- palpability of a recurrent lymph-node enlargement.
- Antitumor vaccination
BALB/c height-week-old female mice (Harlan) were injected subcutaneously on the left flank with lxlO6 CT26 cells treated either with mitoxantrone, or with etoposide or with 2DG and etoposide until reaching 80% DAPI positive cells for each treatment. One week after, these mice were challenged by injecting 0.5x106 live cells on the contralateral flank. The animals were then checked twice a week for tumor development using calipers.
Detection of ecto-calreticulin
For the assessment of calreticulin exposure, cells were first washed in cold PBS, fixed for 5 minutes at 4°C with 0.25% paraformaldehyde in PBS, washed again and stained with rabbit anti-mouse calreticulin antibody (1:200, Abeam), for 30 min at 4°C in 2% FCS PBS. After another wash, cells were incubated with anti-rabbit Alexa Fluor 488 conjugated secondary antibody (1:200, Molecular Probes) and the pellet was resuspended in PBS buffer with DAPI (0.5 μg/ml). Samples were then analyzed by MACSQuant Analyzer (Miltenyi Biotec) and checked for calreticulin staining on DAPI negative cells. CRT exposure to the plasma membrane was expressed in Mean of fluorescence intensity.
Results: - Glycolysis inhibition increases the sensitivity to cytotoxic agent-induced cell death.
In order to analyze the potentialization of cytotoxic agent-induced cell death, CT26 cell lines (murin colon carcinomas) and isolated primary non-Hodgkin lymphoma cells from Εμ-myc mice were used. Those transgenic animals are presenting a deregulated c-myc expression under the control of the immunoglobulin heavy chain (IgH) gene enhancer (Εμ) causes abnormal growth and proliferation of pre-B and B cells and prevents differentiation, culminating in clonal pre-B or immature B-cell lymphoma. Genetics and histopathology of Εμ-myc lymphomas resemble human non-Hodgkin's lymphomas (NHL) (Adams et al., 1985). Primary murin lymphoma or CT26 cells were treated with increasing amount of Etoposide in presence or absence of the glycolytic inhibitor 2-deoxyglucose (2DG) (Figure 1 and Figure 2). While glycolysis inhibition was leading to little toxicity on its own, in all cases, the cell death induction was synergized by the co-treatment, glycolytic inhibitor and cytotoxic agent, compared to each compound individually. Same results were obtained when CT26 cells were treated with Etoposide (ETO) and Lonidamine (LND) (Figure 3), Mitomycin C (Mitom-C) and Lonidamine (Figure 4) or Mitomycin C and 3- Bromopyruvic acid (3-BrPA) (Figure 5).
Subsequently the beneficial effect of the co-treatment was analyzed in vivo using the Εμ-myc model. For that matter WT syngenic immuno-competent C57BL6 mice received 0.5x106 Εμ-myc cells I.V. 20 days later all mice presented enlarged lymph nodes indicating the induction of lymphoma. Then mice were treated with PBS, 2DG, Eto or Eto+2DG. In a first experiment, mice were sacrificed 21 days following the beginning of the treatment and lymph-nodes enlargement was measured. Co-treated animals were presenting significantly smaller lymph nodes than Eto- or control- treated mice. In order to analyze the impact of the co-treatment on the animal lifespan, the experiment was repeated and mice were treated until reaching a point were they had to be sacrificed for ethical concerns (end-point experiments). Figure 7 indicates that co-treated animals can leave 30% longer (69 vs 47 days) compared to the Eto group. Relapse of the disease is always observed in the Εμ-myc model upon treatment. It appeared that co-treated animal presented a time of relapse increase by 30% compared to Eto treated mice (Figure 8).
Therefore combining glycolytic inhibitor with standard cytotoxic agent is able to significantly protect mice.
- Cytotoxic agent combined with glycolytic inhibitor converts a non- immunogenic cancer cell death stimulus to an immunogenic one by controlling calreticulin exposure.
Altogether those results indicated that the co-treatment could increase the cytotoxicity induced by Etoposide or Mitomycin c. The inventors next wondered if the co- treatment could impact on the induction of an efficient anti-cancer immune response. For that matter, the inventors investigated CRT exposure on the surface of primary Εμ-myc cells. As suggested earlier (Obeid et al., 2007), Eto is a weak inducer of CRT exposure (Figure 9A). However, Eto+2DG is very efficient to do so. Mitoxantrone is used as a positive control for CRT exposure (Obeid et al., 2007). The inventors therefore extended this observation to 3 others cancer cell lines, Figure 9B: HeLa cells (Human cervical cancer cell line), 9C: B16 cells (murin melanoma cell lines) and 9D: CT26 (murin colon carcinoma cell lines). In each case, Eto+2DG was inducing a significant increase of CRT exposure compared to Eto or 2DG alone. Interestingly, an increase of CRT exposure to the plasma membrane could also be observed when Eto was combined with other hexokinase inhibitors: LND (Figure 9E) or 3-BrPA (Figure 9F).
To analyze the impact of the co-treatment on the induction of an efficient anti-cancer immune response, anti-tumoral vaccination essays were performed. For that matter, CT26 cells were killed by Eto, Eto+2DG or mitoxantrone (as a positive control) until reaching 80% dead cells in each condition. Then lxl06dead CT26 cells were injected into one flank of BALB/c immuno-competent syngenic mice. One week later, 0.5x106 of CT26 live cells were injected into the contra lateral flank of the mice. Therefore in absence of vaccination the mice will develop a tumor whereas if vaccinated, mice should be protected. Figure 10 described that, as expected, 100% of non-vaccinated mice were dying within 25 days following the challenge with alive CT26 cells. However, only 33% of the mice vaccinated with Eto-treated cells could survive up to 90 days. Very importantly, this percentage jump up to 66% in the group of mice vaccinated with Eto+2DG. Of note, the protection given by the co-treatment is very close to the one induced by mitoxantrone (75%).
These results show than combining glycolytic inhibitor with a classical cytotoxic agent is able to increase CRT exposure to the plasma membrane and therefore increase the anti-cancer immune response.
References : Throughout this application, various references describe the state of the art to which this invention pertains. The disclosures of these references are hereby incorporated by reference into the present disclosure. Weiss RB.Semin Oncol. 1992 Dec;19(6):670-86. The anthracyclines: will we ever find a better doxorubicin?
Minotti G, Menna P, Salvatorelli E, Cairo G, Gianni L.Pharmacol Rev. 2004
Jun;56(2): 185-229. Anthracyclines: molecular advances and pharmacologic developments in antitumor activity and cardiotoxicity.
Obeid M, Tesniere A, Ghiringhelli F, Fimia GM, Apetoh L, Perfettini JL, Castedo M, Mignot G, Panaretakis T, Casares N, Metivier D, Larochette N, van Endert P, Ciccosanti F, Piacentini M, Zitvogel L, Kroemer G. Calreticuhn exposure dictates the immunogenicity of cancer cell death. Nat Med. 2007 Jan;13(l):54-61. Epub 2006 Dec 24.
Casares N, Pequignot MO, Tesniere A, Ghiringhelli F, Roux S, Chaput N, Schmitt E, Hamai A, Hervas-Stubbs S, Obeid M, Coutant F, Metivier D, Pichard E, Aucouturier P, Pierron G, Garrido C, Zitvogel L, Kroemer G. Caspase-dependent immunogenicity of doxorubicin-induced tumor cell death. J Exp Med. 2005 Dec 19;202(12): 1691-701. "Cervical Cancer". Cervical Cancer: Cancers of the Female Reproductive System: Merck Manual Home Edition. Merck Manual Home Edition.
http://www.merck.corn/mmhe/sec22/ch252/ch252d.html. Retrieved 2007-03-24. Pelicano H, Martin DS, Xu RH, Huang P. Glycolysis inhibition for anticancer treatment. Oncogene. 2006 Aug 7;25(34):4633-46. Review.
Pathania D, Millard M, Neamati N. Opportunities in discovery and delivery of anticancer drugs targeting mitochondria and cancer cell metabolism. Adv Drug Deliv Rev. 2009 Nov 30;61 (14): 1250-75. Epub 2009 Aug 27. Review.
El Mjiyad N, Caro-Maldonado A, Ramirez-Peinado S, Munoz-Pinedo C. Sugar-free approaches to cancer cell killing. Oncogene. 2011 Jan 20;30(3):253-64. Epub 2010 Oct 25.
S.P. Mathupala, Y.H. Ko, P.L. Pedersen, Hexokinase II: cancer's double-edged sword acting as both facilitator and gatekeeper of malignancy when bound to mitochondria, Oncogene 25 (2006) 4777-4781.
R.B. Robey, N. Hay, Mitochondrial hexokinases, novel mediators of the antiapoptotic effects of growth factors and Akt, Oncogene 25 (2006) 4683-4696 Adams, J.M., Harris, A.W., Pinkert, C.A., Corcoran, L.M., Alexander, W.S., Cory, S., Palmiter, R.D., and Brinster, R.L. (1985). The c-myc oncogene driven by immunoglobulin enhancers induces lymphoid malignancy in transgenic mice. Nature 318, 533-538.
Apetoh, L., Ghiringhelli, F., Tesniere, A., Obeid, M., Ortiz, C, Criollo, A., Mignot,
G., Maiuri, M.C., Ullrich, E., Saulnier, P., et al. (2007). Toll-like receptor 4- dependent contribution of the immune system to anticancer chemotherapy and radiotherapy. Nat Med 13, 1050-1059.
Obeid, M., Tesniere, A., Ghiringhelli, F., Fimia, G.M., Apetoh, L., Perfettini, J.L.,
Castedo, M., Mignot, G., Panaretakis, T., Casares, N., et al. (2007). Calreticulin exposure dictates the immunogenicity of cancer cell death. Nat Med 13, 54-61.
Pelicano, H., Martin, D.S., Xu, R.H., and Huang, P. (2006). Glycolysis inhibition for anticancer treatment. Oncogene 25, 4633-4646.
Scatena, R., Bottoni, P., Pontoglio, A., Mastrototaro, L., and Giardina, B. (2008).
Glycolytic enzyme inhibitors in cancer treatment. Expert Opin Investig Drugs 17,
1533-1545.
Singh, D., Banerji, A.K., Dwarakanath, B.S., Tripathi, R.P., Gupta, J.P., Mathew, T.L., Ravindranath, T., and Jain, V. (2005). Optimizing cancer radiotherapy with 2- deoxy-d-glucose dose escalation studies in patients with glioblastoma multiforme. Strahlenther Onkol 181, 507-514.
Vander Heiden, M.G., Cantley, L.C., and Thompson, C.B. (2009). Understanding the Warburg effect: the metabolic requirements of cell proliferation. Science 324, 1029- 1033.
Martins I, Kepp O, Schlemmer F, Adjemian S, Tailler M, Shen S, Michaud M, Menger L, Gdoura A, Tajeddine N, Tesniere A, Zitvogel L, Kroemer G. Restoration of the immunogenicity of cisplatin-induced cancer cell death by endoplasmic reticulum stress. Oncogene. 2010 Dec 13. Zitvogel et al. Decoding Cell Death Signals in Inflammation and Immunity. Cell (2010) vol. 140 (6) pp. 798-804.

Claims

1. A glycolytic inhibitor for use in the treatment of a cancer selected from the group consisting of colorectal cancer, cervical cancer and melanoma in a subject to whom a cytotoxic agent is administered.
2. The glycolytic inhibitor according to claim 1 wherein cytotoxic agent is a cytotoxic agent which doesn't induce anticancer immune response.
3. The glycolytic inhibitor according to claim 1 or claim 2 wherein the glycolytic inhibitor is a hexokinase inhibitor.
4. The glycolytic inhibitor according to any of claims 1 to 3 wherein the glycolytic inhibitor is a hexokinase inhibitor selected from the group consisting of 2- deoxyglucose, 5-thioglucose, mannoheptulose, 3-bromopyruvate, lonidamine and methyl jasmonate.
5. The glycolytic inhibitor according to claim 1 or claim 2 wherein the glycolytic inhibitor is a glyceraldehyde-3 phosphate dehydrogenase (GAPDH) inhibitor.
6. The glycolytic inhibitor according to claim 1 or claim 2 wherein the glycolytic inhibitor is a GAPDH inhibitor selected from the group consisting of alpha- chlorhydrin, ornidazole, arsenic, iodoacetate, koningic acid.
7. The glycolytic inhibitor according to any of claims 1 to 6 wherein the cytototxic agent is a topoisomerase II inhibitor selected from the group consisting of amsacrine, etoposide, etoposide phosphate, and teniposide.
8. The glycolytic inhibitor according to any of claims 1 to 6 wherein cytototxic agent is an anti-tumor antibiotic selected in the group consisting from mitomycin, bleomycin and plicamycin.
9. The glycolytic inhibitor according to any of claims 1 to 6 wherein the cytotoxic agent is a platinum compound selected from the group consisting of cisplatin and carboplatin.
10. The glycolytic inhibitor according to any of claims 1 to 9 wherein the subject is a subject with stage III or stage IV cancer.
11. A cytotoxic agent for use in the treatment of a cancer selected from the group consisting of colorectal cancer, cervical cancer and melanoma in a subject to whom a glycolytic inhibitor is administered.
12. A composition for use in the treatment of a cancer selected from the group consisting of colorectal cancer, cervical cancer and melanoma comprising a glycolytic inhibitor and a cytotoxic agent.
13. The composition according to claim 12 wherein the cytotoxic agent is a cytotoxic agent which doesn't induce anticancer immune response.
14. A kit for use in the treatment of a cancer selected from the group consisting of colorectal cancer, cervical cancer and melanoma comprising :
a- a glycolytic inhibitor ; and
b- a cytotoxic agent.
15. The kit according to claim 14 wherein the cytotoxic agent is a cytotoxic agent which doesn't induce anticancer immune response.
PCT/IB2011/000795 2011-03-11 2011-03-11 Glycolytic inhibitor with cytotoxic agent for use in the treatment of a cancer WO2012123774A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
PCT/IB2011/000795 WO2012123774A1 (en) 2011-03-11 2011-03-11 Glycolytic inhibitor with cytotoxic agent for use in the treatment of a cancer

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
PCT/IB2011/000795 WO2012123774A1 (en) 2011-03-11 2011-03-11 Glycolytic inhibitor with cytotoxic agent for use in the treatment of a cancer

Publications (1)

Publication Number Publication Date
WO2012123774A1 true WO2012123774A1 (en) 2012-09-20

Family

ID=44120176

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IB2011/000795 WO2012123774A1 (en) 2011-03-11 2011-03-11 Glycolytic inhibitor with cytotoxic agent for use in the treatment of a cancer

Country Status (1)

Country Link
WO (1) WO2012123774A1 (en)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014122326A1 (en) * 2013-02-11 2014-08-14 Laboratoires Expanscience Use of a composition comprising avocado perseose in the protection of epidermal stem cells
CN104546833A (en) * 2015-02-09 2015-04-29 江苏澳格姆生物科技有限公司 Application of ornidazole in preparation of medicines for inhibiting tumor cell metastasis and spread
WO2016100203A1 (en) * 2014-12-20 2016-06-23 Gregg John Malcolm Hall Antimicrobial drug synthesis and therapeutic compositions
WO2016196890A1 (en) * 2015-06-04 2016-12-08 Vtv Therapeutics Llc Inhibitors of hexokinase and methods of use thereof
WO2017216791A1 (en) * 2016-06-13 2017-12-21 Ariel Scientific Innovations Ltd. Anticancer drug conjugates
WO2018009539A1 (en) * 2016-07-05 2018-01-11 Vtv Therapeutics Llc Inhibitors of hexokinase and methods of use thereof

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004062604A2 (en) * 2003-01-10 2004-07-29 Threshold Pharmaceuticals, Inc. Treatment of cancer with 2-deoxyglucose
US20060074050A1 (en) * 2004-07-14 2006-04-06 Glycogenesys, Inc. Composition and method for treating hyperproliferative diseases
US20060276527A1 (en) * 2003-01-17 2006-12-07 Thresold Pharmaceuticals, Inc. Combination therapies for the treatment of cancer

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004062604A2 (en) * 2003-01-10 2004-07-29 Threshold Pharmaceuticals, Inc. Treatment of cancer with 2-deoxyglucose
US20060276527A1 (en) * 2003-01-17 2006-12-07 Thresold Pharmaceuticals, Inc. Combination therapies for the treatment of cancer
US20060074050A1 (en) * 2004-07-14 2006-04-06 Glycogenesys, Inc. Composition and method for treating hyperproliferative diseases

Non-Patent Citations (21)

* Cited by examiner, † Cited by third party
Title
"Cervical Cancer: Cancers of the Female Reproductive System: Merck Manual Home Edition", 24 March 2007, MERCK MANUAL HOME EDITION, article "Cervical Cancer"
ADAMS, J.M., HARRIS, A.W., PINKERT, C.A., CORCORAN, L.M., ALEXANDER, W.S., CORY, S., PALMITER, R.D., BRINSTER, R.L.: "The c-myc oncogene driven by immunoglobulin enhancers induces lymphoid malignancy in transgenic mice", NATURE, vol. 318, 1985, pages 533 - 538, XP001207098, DOI: doi:10.1038/318533a0
APETOH, L., GHIRINGHELLI, F., TESNIERE, A., OBEID, M., ORTIZ, C., CRIOLLO, A., MIGNOT, G., MAIURI, M.C., ULLRICH, E., SAULNIER, P.: "Toll-like receptor 4- dependent contribution of the immune system to anticancer chemotherapy and radiotherapy", NAT MED, vol. 13, 2007, pages 1050 - 1059
BELLELLI A ET AL: "EFFECTS OF VM-26 AND LONIDAMINE ON A B16 MELANOMA CELL LINE", ANTICANCER RESEARCH, INTERNATIONAL INSTITUTE OF ANTICANCER RESEARCH, GR, vol. 10, no. 3, 1 January 1990 (1990-01-01), pages 565 - 578, XP008138838, ISSN: 0250-7005 *
CASARES N, PEQUIGNOT MO, TESNIERE A, GHIRINGHELLI F, ROUX S, CHAPUT N, SCHMITT E, HAMAI A, HERVAS-STUBBS S, OBEID M: "Caspase-dependent immunogenicity of doxorubicin-induced tumor cell death", J EXP MED., vol. 202, no. 12, 19 December 2005 (2005-12-19), pages 1691 - 701, XP002440491, DOI: doi:10.1084/jem.20050915
EL MJIYAD N, CARO-MALDONADO A, RAMÍREZ-PEINADO S, MUNOZ-PINEDO C.: "Sugar-free approaches to cancer cell killing", ONCOGENE, vol. 30, no. 3, 25 October 2010 (2010-10-25), pages 253 - 64
GEORGE C., CANCER IMMUNOTHERAPY : IMMUNE SUPPRESSION AND TUMOR GROWTH
MARTINS I, KEPP O, SCHLEMMER F, ADJEMIAN S, TAILLER M, SHEN S, MICHAUD M, MENGER L, GDOURA A, TAJEDDINE N: "Restoration of the immunogenicity of cisplatin-induced cancer cell death by endoplasmic reticulum stress", ONCOGENE, 13 December 2010 (2010-12-13)
MINOTTI G, MENNA P, SALVATORELLI E, CAIRO G, GIANNI L., PHARMACOL REV., vol. 56, no. 2, June 2004 (2004-06-01), pages 185 - 229
OBEID M, TESNIERE A, GHIRINGHELLI F, FIMIA GM, APETOH L, PERFETTINI JL, CASTEDO M, MIGNOT G, PANARETAKIS T, CASARES N: "Calreticulin exposure dictates the immunogenicity of cancer cell death", NAT MED., vol. 13, no. L, 24 December 2006 (2006-12-24), pages 54 - 61, XP002440487, DOI: doi:10.1038/nm1523
OBEID, M., TESNIERE, A., GHIRINGHELLI, F., FIMIA, G.M., APETOH, L., PERFETTINI, J.L., CASTEDO, M., MIGNOT, G., PANARETAKIS, T., CA: "Calreticulin exposure dictates the immunogenicity of cancer cell death", NAT MED, vol. 13, 2007, pages 54 - 61, XP002440487, DOI: doi:10.1038/nm1523
PATHANIA D, MILLARD M, NEAMATI N.: "Opportunities in discovery and delivery of anticancer drugs targeting mitochondria and cancer cell metabolism", ADV DRUG DELIV, vol. 61, no. 14, 27 August 2009 (2009-08-27), pages 1250 - 75, XP026761106, DOI: doi:10.1016/j.addr.2009.05.010
PELICANO H, MARTIN DS, XU RH, HUANG P.: "Glycolysis inhibition for anticancer treatment", ONCOGENE, vol. 25, no. 34, 7 August 2006 (2006-08-07), pages 4633 - 46, XP055009657, DOI: doi:10.1038/sj.onc.1209597
PELICANO, H., MARTIN, D.S., XU, R.H., HUANG, P.: "Glycolysis inhibition for anticancer treatment", ONCOGENE, vol. 25, 2006, pages 4633 - 4646, XP055009657, DOI: doi:10.1038/sj.onc.1209597
R.B. ROBEY, N. HAY: "Mitochondrial hexokinases, novel mediators of the antiapoptotic effects of growth factors and Akt", ONCOGENE, vol. 25, 2006, pages 4683 - 4696, XP002487537, DOI: doi:10.1038/sj.onc.1209595
S.P. MATHUPALA, Y.H. KO, P.L. PEDERSEN: "Hexokinase II: cancer's double-edged sword acting as both facilitator and gatekeeper of malignancy when bound to mitochondria", ONCOGENE, vol. 25, no. 2006, pages 4777 - 4781
SCATENA, R., BOTTONI, P., PONTOGLIO, A., MASTROTOTARO, L., GIARDINA, B.: "Glycolytic enzyme inhibitors in cancer treatment", EXPERT OPIN INVESTIG DRUGS, vol. 17, 2008, pages 1533 - 1545, XP002661595, DOI: doi:10.1517/13543780802411053
SINGH, D., BANERJI, A.K., DWARAKANATH, B.S., TRIPATHI, R.P., GUPTA, J.P., MATHEW, T.L., RAVINDRANATH, T., JAIN, V.: "Optimizing cancer radiotherapy with 2-deoxy-d-glucose dose escalation studies in patients with glioblastoma multiforme", STRAHLENTHER ONKOL, vol. 181, 2005, pages 507 - 514, XP019384396, DOI: doi:10.1007/s00066-005-1320-z
VANDER HEIDEN, M.G., CANTLEY, L.C., THOMPSON, C.B.: "Understanding the Warburg effect: the metabolic requirements of cell proliferation", SCIENCE, vol. 324, 2009, pages 1029 - 1033
WEISS RB., SEMIN ONCOL., vol. 19, no. 6, December 1992 (1992-12-01), pages 670 - 86
ZITVOGEL ET AL.: "Decoding Cell Death Signals in Inflammation and Immunity", CELL, vol. 140, no. 6, 2010, pages 798 - 804

Cited By (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014122326A1 (en) * 2013-02-11 2014-08-14 Laboratoires Expanscience Use of a composition comprising avocado perseose in the protection of epidermal stem cells
FR3001889A1 (en) * 2013-02-11 2014-08-15 Expanscience Lab USE OF A COMPOSITION COMPRISING AVOCADO PERSEOSE IN THE PROTECTION OF EPIDERMAL STEM CELLS.
US10092495B2 (en) 2013-02-11 2018-10-09 Laboratoire Expanscience Use of a composition comprising avocado perseose in the protection of epidermal stem cells
WO2016100203A1 (en) * 2014-12-20 2016-06-23 Gregg John Malcolm Hall Antimicrobial drug synthesis and therapeutic compositions
EP3244890A4 (en) * 2014-12-20 2018-11-21 Gregg, John Malcolm Hall Antimicrobial drug synthesis and therapeutic compositions
US11135200B2 (en) 2014-12-20 2021-10-05 John Malcolm Hall Gregg Antimicrobial drug synthesis and therapeutic compositions
CN104546833A (en) * 2015-02-09 2015-04-29 江苏澳格姆生物科技有限公司 Application of ornidazole in preparation of medicines for inhibiting tumor cell metastasis and spread
WO2016196890A1 (en) * 2015-06-04 2016-12-08 Vtv Therapeutics Llc Inhibitors of hexokinase and methods of use thereof
WO2017216791A1 (en) * 2016-06-13 2017-12-21 Ariel Scientific Innovations Ltd. Anticancer drug conjugates
US11007271B2 (en) 2016-06-13 2021-05-18 Ariel Scientific Innovations Ltd. Anticancer drug conjugates
WO2018009539A1 (en) * 2016-07-05 2018-01-11 Vtv Therapeutics Llc Inhibitors of hexokinase and methods of use thereof

Similar Documents

Publication Publication Date Title
Mueller et al. Cooperation of the HDAC inhibitor vorinostat and radiation in metastatic neuroblastoma: efficacy and underlying mechanisms
JP2023082102A (en) Combination cancer therapy
WO2012123774A1 (en) Glycolytic inhibitor with cytotoxic agent for use in the treatment of a cancer
JP5662309B2 (en) Compositions and methods for treating neoplastic diseases
US20220265592A1 (en) Use of bipolar trans carotenoids with chemotherapy and radiotherapy for treatment of cancer
Lee et al. Translational assessment of mitochondrial dysfunction of pancreatic cancer from in vitro gene microarray and animal efficacy studies, to early clinical studies, via the novel tumor-specific anti-mitochondrial agent, CPI-613
Zereshkian et al. The human polynucleotide kinase/phosphatase (hPNKP) inhibitor A12B4C3 radiosensitizes human myeloid leukemia cells to Auger electron-emitting anti-CD123 111In-NLS-7G3 radioimmunoconjugates
RU2396960C2 (en) Combined anticancer therapy and pharmaceutical compositions therefor
Hou et al. Targeted arginine metabolism therapy: A dilemma in glioma treatment
Hu et al. Synergistic effect of adoptive immunotherapy and docetaxel inhibits tumor growth in a mouse model
Obrador et al. Glioblastoma therapy: past, present and future
Chen et al. Cisplatin and albumin-based gold–cisplatin nanoparticles enhance ablative radiation therapy–induced antitumor immunity in local and distant tumor microenvironment
Mathew et al. 5-FU mediated depletion of myeloid suppressor cells enhances T-cell infiltration and anti-tumor response in immunotherapy–resistant lung tumor
WO2015191576A1 (en) Combination therapy comprising a liposomal prodrug of mitomycin c and radiotherapy
Li et al. Advanced nanomaterials for enhanced immunotherapy via metabolic regulation
WO2021155479A1 (en) Methods for enhancing t cells using venetoclax
Chouaib Autophagy in Immune Response: Impact on Cancer Immunotherapy
JP2014513698A (en) Use of arsenic for cancer treatment protection
WO2006095270A1 (en) Combination anticancer therapy or om-174 and pharmaceutical compositions therefor
Gao et al. Biosynthetic MnSe Nanobomb with Low Mn Content Activates the cGAS-STING Pathway and Induces Immunogenic Cell Death to Enhance Antitumour Immunity
Muradova Effects of Gadolinium-Based Nanoparticles AGuIX on Ionizing Radiation-Elicited Macrophage Functional Reprogramming
TWI548410B (en) Method of treating cancer using combination of a bifunctional alkylating agent and dna repair inhibitors
Wang et al. Combination therapy of irreversible electroporation and cytokine-induced killer cells for treating mice bearing panc02 pancreatic-cancer xenografts
Reda DEVELOPMENT OF NOVEL NANOPARTICLE BASED PLK1, EGFR, AND PD-L1 TARGETED THERAPIES FOR LUNG CANCER
Sun et al. Combretastatin A4 phosphate encapsulated in hyaluronic acid nanoparticles is highly cytotoxic to oral squamous cell carcinoma

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: 11721576

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: 11721576

Country of ref document: EP

Kind code of ref document: A1