WO2016195353A1 - A use of 1'-cyano-cytarabine for cancer treatment - Google Patents

A use of 1'-cyano-cytarabine for cancer treatment Download PDF

Info

Publication number
WO2016195353A1
WO2016195353A1 PCT/KR2016/005715 KR2016005715W WO2016195353A1 WO 2016195353 A1 WO2016195353 A1 WO 2016195353A1 KR 2016005715 W KR2016005715 W KR 2016005715W WO 2016195353 A1 WO2016195353 A1 WO 2016195353A1
Authority
WO
WIPO (PCT)
Prior art keywords
cancer
ara
cyano
compound
cda
Prior art date
Application number
PCT/KR2016/005715
Other languages
French (fr)
Inventor
Su-Sung OH
Hyangmi KIM
Seongrim BYEON
Original Assignee
Kainos Medicine, Inc.
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 Kainos Medicine, Inc. filed Critical Kainos Medicine, Inc.
Publication of WO2016195353A1 publication Critical patent/WO2016195353A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H19/00Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof
    • C07H19/02Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof sharing nitrogen
    • C07H19/04Heterocyclic radicals containing only nitrogen atoms as ring hetero atom
    • C07H19/06Pyrimidine radicals
    • 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
    • A61K31/7052Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides
    • A61K31/706Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom
    • A61K31/7064Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines
    • A61K31/7068Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines having oxo groups directly attached to the pyrimidine ring, e.g. cytidine, cytidylic acid

Definitions

  • the present invention relates to a cytidine deaminase (CDA)-resistant compound selected from the group consisting of ⁇ -cyano-cytarabine (l'-CN- Ara-C), ⁇ -cyano-gemcitabine, l'-cyano-5-azacytidine and l'-cyano-5-aza-2'- deoxycytidine, a prodrug or an acceptable salt thereof; a pharmaceutical composition comprising the same; and a method for preventing or treating a cancer using the compound.
  • CDA cytidine deaminase
  • Nucleoside analogs as a class of therapeutic agents are prevalent in the clinical treatment of cancer and viral disease (X. Liu, et al. , Expert Opin. Investig. Drugs, 2012, 21 : 541-555).
  • anti-cancer nucleosides are grouped as antimetabolites, hypomethylating agents or others depending on their mechanism of action. They differ greatly in the means by which they cause cell death after they are incorporated into DNA.
  • the key biochemical pathway responsible for the cytotoxic action of nucleoside analogs is mediated mainly by the incorporation of the drug into DNA which subsequently results in interfere with DNA replication or termination of DNA synthesis (A. J. Townsend, et al , Molecular Pharmacology, 1987, 32: 330-339; P. Huang et al., J. Bio. Chem., 1990, 265 : 16617- 16625)
  • Anti-cancer nucleosides are typically analogs of natural nucleosides of cytidine, adenosine, guanosine and thymidine (B. Ewald, et al. , Oncogene, 2008, 27: 6522-6537).
  • CDA cytidine deaminase
  • CDA is an intracellular enzyme of the pyrimidine salvage pathways that catalyzes the deamination of cytidine and deoxycytidine to uridine and deoxyuridine, respectively (S. Costanzi et al, Chem. Med. Chem., 2012, 6: 1452-1458).
  • human CDA recognizes as substrates a number of cytidine-based anti-cancer drugs such as Ara-C, gemcitabine, decitabine and azacytidine for the degradation of cytosine to uracil.
  • Ara-C is a nucleoside cytotoxic anti-cancer agent useful for the chemotherapy of cancers, especially, hematological malignancies. It has been used as a standard treatment agent for acute myeloid leukemia since 1969. However, Ara-C rapidly undergoes deamination by CDA present in blood and tissues such as liver to become inactive metabolite, uracil- Ara-C. We excrete uracil-Ara-C through urine and Ara-C has no anti-cancer activity. Oral plasma exposure of Ara-C is very low, less than 20%, and clearance rate is high (O. Schiavon, et al, European Journal of Medicinal Chemistry, 2004, 39: 123-133).
  • Ara-C has no anti-cancer effect against solid tumors.
  • formation of uracil-Ara-C by CDA deamination is increased, active metabolite, triphosphate-Ara-C that plays a role in inhibiting DNA synthesis is diminished.
  • very low plasma exposure of Ara-C brings out poor distribution into solid tumor tissues (J. P. Godefridus, Deoxynucleoside Analogs in Cancer Therapy, 2006, 139; W. A. Bleyer, Clinical Cancer Research, 1999, 5 : 3349-3351)
  • cytidine-based nucleoside anti-cancer drugs do not meet the stability requirement against CDA deamination.
  • this modified structure was aimed at achieving improved oral pharmacokinetic properties including high oral absorption via oral administration, high in vivo efficacy with a low dose and expanding indication area to solid tumors, as well as hematological malignancies.
  • a cytidine deaminase (CDA)-resistant compound selected from the group consisting of ⁇ - cyano-cytarabine represented by formula (II), ⁇ -cyano-gemcitabine represented by formula (III), l'-cyano-5-azacytidine represented by formula (IV) and - cyano-5-aza-2'-deoxycytidine represented by formula (V), a prodrug or an acceptable salt thereof:
  • Fig. 1 View of the complex between human CDA (PDB ID: 1MQ0) and
  • Ara-C (a) as resulting from molecular docking -CN-Ara-C (b);
  • Fig. 2 Relative susceptibility of cytidine, Ara-C and -CN- Ara-C to CDA of Test Example 2;
  • Fig. 3 Pharmacokinetics of the Ara-C (mouse) and -CN-Ara-C (mouse, rat and dog) of Test Example 4 through IV and PO administration routes;
  • Fig. 4 The tumor volume of IP and PO Ara-C and ⁇ -CN- Ara-C administered mice in HL-60 human leukemia xenograft of Test Example 5; and Fig. 5: ⁇ -CN- Ara-C concentration in plasma and tumor tissues in HL-60 human leukemia xenograft of Test Example 6.
  • the present invention provides a cytidine deaminase (CDA)-resistant compound selected from the group consisting of -cyano-cytarabine represented by formula (II), ⁇ -cyano-gemcitabine represented by formula (III), l'-cyano-5- azacytidine represented by formula (IV) and l'-cyano-5-aza-2'-deoxy cytidine represented by formula (V), a prodrug or an acceptable salt thereof:
  • CDA cytidine deaminase
  • the compound of the present invention is highly resistant to deamination by CDA and highly stable against CDA.
  • the high level of CDA resistance of the compound of the present invention affords animals such as mouse, rat and dog excellent pharmacokinetic properties and in vivo efficacy by PO or IV administration.
  • the present invention provides a pharmaceutical composition for preventing or treating a cancer, comprising the compound of the present invention as an active ingredient, and a pharmaceutically acceptable carrier or an excipient.
  • the cancer may be selected from the group consisting of hematological malignancies, epithelial tumors, melanoma, leukemia, acute promyelocyte leukemia, lymphoma, osteogenic sarcoma, colon cancer, pancreatic cancer, breast cancer, prostate cancer, ovarian cancer, lung cancer and a combination thereof.
  • the compound may be formulated in a pharmaceutical composition, comprising one or more of the compound and a pharmaceutically acceptable carrier or an excipient.
  • the compound can be mixed with a pharmaceutically acceptable carrier or an excipient, diluted by an excipient or enclosed within such a carrier which can be in the form of a capsule, sachet, paper or other container.
  • a pharmaceutically acceptable carrier or an excipient which can be in the form of a capsule, sachet, paper or other container.
  • the excipient serves as a diluent, it can be a solid, semi-solid, or liquid material, which acts as a vehicle, carrier, or medium for the compound.
  • the pharmaceutical compositions can be in the form of tablets, pills, powers, lozenges, sachets, cachets, elixirs, suspensions, emulsions, solutions, syrups, soft and hard gelatin capsules, and other orally ingestible formulations.
  • excipients include lactose, dextrose, sucrose, sorbitol, mannitol, starches, gum acacia, calcium phosphate, alginates, tragacanth, gelatin, calcium silicate, microcrystalline cellulose, polyvinylpyrrolidone, cellulose, magnesium carbonate, water, ethanol, propylene glycol, syrup, and methyl cellulose.
  • the formulations can additionally include lubricating agents such as talc, magnesium stearate, and mineral oil; wetting agents; emulsifying and suspending agents; preserving agents such as methyl- and propyl-hydroxybenzoates, sweetening agents; and flavoring agents.
  • lubricating agents such as talc, magnesium stearate, and mineral oil
  • wetting agents such as talc, magnesium stearate, and mineral oil
  • emulsifying and suspending agents such as methyl- and propyl-hydroxybenzoates, sweetening agents
  • preserving agents such as methyl- and propyl-hydroxybenzoates, sweetening agents
  • sweetening agents such as methyl- and propyl-hydroxybenzoates, sweetening agents
  • flavoring agents such as talc, magnesium stearate, and mineral oil
  • the compositions of the present invention can also be formulated so as to provide quick, sustained or delayed release of the novel compound after administration to the patient by employing procedures known in the art.
  • the term "pharmaceutically acceptable carrier” refers to those components in the particular dosage form employed which are considered inert and are typically employed in the pharmaceutical fields to formulate a dosage form containing a particular active compound. This may include without limitation solids, liquids and gases, used to formulate the particular pharmaceutical product.
  • carriers include diluents, flavoring agents, solubilizers, suspending agents, binders or tablet disintegrating agents, encapsulating materials, penetration enhancers, solvents, emollients, thickeners, and dispersants, sustained release forms, such as matrices, transdermal delivery components, buffers, stabilizers, and the like.
  • composition of the present invention may be formulated in the absence or presence of carriers or excipients that provide a sustained released effect.
  • the pharmaceutical composition of the present invention may be prepared according to methods known in the art. It is contemplated that administration of such compositions may be via oral, injectable and/or parenteral routes depending upon the needs of the subject.
  • the pharmaceutical composition of the present invention can also be administered by nasal or oral inhalation, oral ingestion, intramuscular injection, intravenous injection, intraperitoneal injection, transdermally, or other forms of administration.
  • Aerosol formulations for use in this invention typically include propellants, such as a fluorinated alkane, surfactants and co-solvents and may be filled into aluminum or other conventional aerosol containers which are then closed by a suitable metering valve and pressurized with propellant, producing a metered dose inhaler. Aerosol preparations are typically suitable for nasal or oral inhalation, and may be in powder or solution form, in combination with a compressed gas, typically compressed air. Additionally, aerosols may be useful topically. Topical preparations useful herein include creams, ointments, solutions, suspensions and the like. These may be formulated to enable one to apply the appropriate dosage topically to the affected area once daily, up to 3-4 times daily as appropriate. Topical sprays may be included herein as well.
  • propellants such as a fluorinated alkane, surfactants and co-solvents
  • Aerosol preparations are typically suitable for nasal or oral inhalation, and may be in powder or solution form, in combination with a compressed gas, typically
  • transdermal delivery may be an option, which can provide a relatively steady state delivery of the medication.
  • Transdermal delivery typically involves the use of a compound in solution, with an alcoholic vehicle, optionally a penetration enhancer, such as a surfactant and other optional ingredients.
  • Transdermal delivery systems of matrix and reservoir type are examples of suitable transdermal systems.
  • Transdermal delivery differs from conventional topical treatment in that the dosage form delivers a systemic dose of medication to the patient.
  • the present invention includes a method for inhibiting the growth of a cell using a compound of the present invention.
  • the present invention provides a method for inhibiting the growth of a cancer cell in a subject, which comprises administering a therapeutically effective amount of the compound to the subject in need thereof.
  • the present invention provides a method for preventing or treating a cancer in a subject, which comprises administering a therapeutically effective amount of the compound to the subject in need thereof.
  • Combination therapy includes combining the method of preventing or treating cancer as described in the invention and one or more cancer therapeutic methods.
  • the cancer therapeutic methods include surgical therapy, radiation therapy, administering an anticancer agent (including, for example, antineoplastics, novantrone, bicalutamide, esterified estrogens, goserelin, histrelin, leuprolide, nilandron, triptorelin pamoate, docetaxel, taxotere, carboplatin, and cisplatin angiogenesis inhibitors), immunotherapy, antineoplastons, investigational drugs, vaccines, and less conventional therapies (sometimes referred to as novel or innovative therapies, which include, for example, chemoembolization, hormone therapy, local hyperthermia, photodynamic therapy, radiofrequency ablation, stem cell transplantation, and gene therapy).
  • an anticancer agent including, for example, antineoplastics, novantrone, bicalutamide, esterified estrogens, goserelin, histrelin
  • the compound may be administered in combination with other active agents.
  • the compound used in the methods of prevention or treatment is administered in an amount which effectively achieves the desired therapeutic result in a subject.
  • the compound may be administered as a pharmaceutical composition, or in the absence of a carrier or diluent.
  • the dosages of the compound will vary somewhat depending upon the components of the compound, the rate of in vivo hydrolysis, etc. Those skilled in the art can determine the optimal dose of the compound based on clinical experience and the treatment indication.
  • the amount of compound administered to a subject is about 0.1 to about 100 mg/kg of body weight, more preferably, about 5 to about 40 mg/kg.
  • Other preferred dosages include about 0.1 to about 10 mg/kg of body weight, about 1 to about 100 mg/kg of body weight, about 1 to about 60 mg/kg of body weight, about 1 to about 10 mg/kg of body weight, about 10 to about 100 mg/kg of body weight, about 10 to about 60 mg/kg of body weight, about 20 to about 60 mg/kg of body weight and about 30 to about 50 mg/kg of body weight.
  • the compound can also be converted into a pharmaceutically acceptable salt or pharmaceutically acceptable solvate or other physical forms (e.g., polymorphs) by methods known in the art field.
  • the subjects that may be treated using the compounds of the present invention may be mammals including humans.
  • the present invention also includes a use of the compound of the present invention for the manufacture of a medicament for preventing or treating a cancer.
  • the present invention also includes a kit comprising one or more of the compound of the present invention and instructions for its use.
  • Ara-C favorably interacts with the E67 through the H-bonds and with the hydrophobic V38 and C65 with preserved goldscore 51.9 compared to reference goldscore 52.6. However, this interaction is disrupted by the presence of CN in 1 '-position of Ara-C. As shown in Fig.l (b), ⁇ -CN group causes crash in binding pocket with reduced goldscore 45.8.
  • -CN- Ara-C was not a substrate of CD A, and completely resistant to deamination by CDA in vitro.
  • CellTiter-Glo ® Luminescent Cell Viability Assay (Promega #G7572) was used to measure the 50% inhibition concentration (IC 50 ) of the Ara-C and l '-CN- Ara-C against ML-2 (human acute myeloma leukemia), MOLT-4 (human acute lymphoblastic leukemia), MV-4- 1 1 (human biphenotypic B myelomonocytic leukemia), HL-60 (human acute promyelocyte leukemia), Hep G2 (human hepatocellular carcinoma), HCT 1 16 (human colorectal carcinoma) cell lines. Six hundred cells in 90 ⁇ media were plated per well in duplicates in a 96-well plate.
  • l '-CN- Ara-C exhibited the proliferative activities on hematological cancer, liver cancer and colon cancer cells.
  • LC-MS liquid chromatography-mass spectrometry
  • TEST EXAMPLE 5 Tumor growth inhibition efficacy of l'-CN-Ara- C in HL-60 human leukemia xenograft
  • Tumor growth inhibition (TGI) activities of IP-administered Ara-C, and IP/PO-administered l'-CN- Ara-C were compared in HL-60 human leukemia xenografts.
  • Tumor growth inhibitions were observed in in vivo subcutaneous mouse xenograft model (HL-60 cell line, Corning #3603).
  • NOD/SCID female mice of 6-8 week age and weighing approximately 18-22 g, were used for tumor inoculation.
  • the amounts of treated compounds are represented in milligram (mg) of compound per kilogram (kg) of animal body weight (mg/kg, mpk).
  • Administration route of the test compound was either intraperitoneal (IP) injection or peros (PO, oral) injection.
  • mice Three groups, i.e., vehicle group (Control group), Ara-C group (treatment at 40 mpk of cytarabine through IP injection; Comparative group) and ⁇ -CN- Ara-C group (treatment at 40 mpk through IP and 60 mpk through PO injection) of mice were used.
  • the overall administration schedule was 2 cycles of treatment for 5 days (5d ON), followed by 2 days of non-treatment (2d OFF). Tumor sizes were measured using a caliper, once every 4 days, and the tumor volume was expressed in mm using Equation 1 :
  • V 0.5 a x b 2
  • a and b are the long and short diameters of the tumor, respectively.
  • the tumor volume (mm ) and body weight (g) results of the three groups are shown in Fig. 4.
  • tumor growth inhibition (% TGI) and body weight change (% BW change) compared to that of Ara-C were shown in Table 4.
  • oral (PO) dosing of ⁇ -CN- Ara-C shows a good potency.
  • IP dosing of ⁇ -CN- Ara-C exhibited superior efficacy (73% TGI) to IP dosing of Ara-C at the same dose (40 mpk).
  • the concentration of Ara-C and -CN- Ara-C in plasma/tumor tissue were evaluated in Test Example 5 in HL-60 human leukemia xenografts.
  • ⁇ -CN- Ara-C retained high intracellular drug concentration in plasma/tumor tissue from leukemia HL-60 cell derived xenograft model.

Abstract

The present invention provides to a cytidine deaminase (CDA)-resistant compound selected from the group consisting of 1'-cyano-cytarabine, 1'-cyano-gemcitabine, 1'-cyano-5-azacytidine and 1'-cyano-5-aza-2'-deoxycytidine, a prodrug or an acceptable salt thereof; a pharmaceutical composition comprising the same; and a method for preventing or treating a cancer using the compound. Further, the compound of the present invention can be used as therapeutic agents for the prevention or treatment of a cancer.

Description

DESCRIPTION
A USE OF l'-CYANO-CYTARABINE FOR CANCER TREATMENT
FIELD OF THE INVENTION
The present invention relates to a cytidine deaminase (CDA)-resistant compound selected from the group consisting of Γ-cyano-cytarabine (l'-CN- Ara-C), Γ-cyano-gemcitabine, l'-cyano-5-azacytidine and l'-cyano-5-aza-2'- deoxycytidine, a prodrug or an acceptable salt thereof; a pharmaceutical composition comprising the same; and a method for preventing or treating a cancer using the compound.
BACKGROUND OF THE INVENTION Nucleoside analogs as a class of therapeutic agents are prevalent in the clinical treatment of cancer and viral disease (X. Liu, et al. , Expert Opin. Investig. Drugs, 2012, 21 : 541-555). Among them, anti-cancer nucleosides are grouped as antimetabolites, hypomethylating agents or others depending on their mechanism of action. They differ greatly in the means by which they cause cell death after they are incorporated into DNA. Especially, the key biochemical pathway responsible for the cytotoxic action of nucleoside analogs is mediated mainly by the incorporation of the drug into DNA which subsequently results in interfere with DNA replication or termination of DNA synthesis (A. J. Townsend, et al , Molecular Pharmacology, 1987, 32: 330-339; P. Huang et al., J. Bio. Chem., 1990, 265 : 16617- 16625)
Anti-cancer nucleosides are typically analogs of natural nucleosides of cytidine, adenosine, guanosine and thymidine (B. Ewald, et al. , Oncogene, 2008, 27: 6522-6537). The cytidine nucleoside analog 5-azacytidine (azacitidine), 5- aza-2'-deoxycytidine (decitabine), 1 -beta-D-arabinofuranosyl-5-azacytosine (fazarabine), 1-beta-D-arabinofuranosyl cytosine (cytarabine; Ara-C) and 2'- deoxy-2',2'-difluorocytidine (gemcitabine) have been used clinically in cancer treatment.
However, these cytidine anti-cancer nucleosides have clinical limits due to susceptibility to cytidine deaminase (CDA). CDA is an intracellular enzyme of the pyrimidine salvage pathways that catalyzes the deamination of cytidine and deoxycytidine to uridine and deoxyuridine, respectively (S. Costanzi et al, Chem. Med. Chem., 2012, 6: 1452-1458). Importantly, human CDA recognizes as substrates a number of cytidine-based anti-cancer drugs such as Ara-C, gemcitabine, decitabine and azacytidine for the degradation of cytosine to uracil.
For example, Ara-C is a nucleoside cytotoxic anti-cancer agent useful for the chemotherapy of cancers, especially, hematological malignancies. It has been used as a standard treatment agent for acute myeloid leukemia since 1969. However, Ara-C rapidly undergoes deamination by CDA present in blood and tissues such as liver to become inactive metabolite, uracil- Ara-C. We excrete uracil-Ara-C through urine and Ara-C has no anti-cancer activity. Oral plasma exposure of Ara-C is very low, less than 20%, and clearance rate is high (O. Schiavon, et al, European Journal of Medicinal Chemistry, 2004, 39: 123-133). Therefore, it cannot be administered orally since it is rapidly deaminated once absorbed. In order to improve the efficacy of Ara-C, it is usually administerated in high doses by continuous-infusion. Accordingly, high dose to maximize a therapeutic effect results in association of unpleasant side effect for the patient such as colitis, intestinal mucostisis, myelosuppression, bone marrow suppression and thrombopenia (J. K. Christman, Oncogene 2002, 21 : 5483-5495). A small number of patients may have abnormal liver function, fever, rash and other side effects (J. M. Bennett, Leukemia Research, 2003, 27:761 ; and H. M. Kantarjian, Cancer, 2007, 109: 1007-1010). And also, Ara-C has no anti-cancer effect against solid tumors. As formation of uracil-Ara-C by CDA deamination is increased, active metabolite, triphosphate-Ara-C that plays a role in inhibiting DNA synthesis is diminished. Thus, very low plasma exposure of Ara-C brings out poor distribution into solid tumor tissues (J. P. Godefridus, Deoxynucleoside Analogs in Cancer Therapy, 2006, 139; W. A. Bleyer, Clinical Cancer Research, 1999, 5 : 3349-3351)
Therefore, cytidine-based nucleoside anti-cancer drugs do not meet the stability requirement against CDA deamination. In order to solve these problems and maximize anti-cancer efficacy, we have investigated the modified nucleoside structure not susceptible to CDA. Based on CDA resistance, this modified structure was aimed at achieving improved oral pharmacokinetic properties including high oral absorption via oral administration, high in vivo efficacy with a low dose and expanding indication area to solid tumors, as well as hematological malignancies. The synthesis of Γ-CN- Ara-C and its antiviral activity and in vitro anti-cancer activity in CCRF-HSB-2 and KB cell lines were reported in 1996 (Yoshimura et al , Nucleoside & Nucleotides, 1996, 15: 305- 324). Various compounds \ substituted at 2'-, 3'-, and 4'-position have been synthesized. Among them, the analog where a cyano group is attached at 2'- position with "up" configuration, l-(2-C-cyano-2-deoxy-l- -D- arabinosyl)cytosine (CNDAC) and its anticancer activities in vitro and in vivo were reported by Matsuda's group (Matsuda, A. et al , J. Med. Chem., 1993, 36, 4183-4189). The synthesis of thymidine analogs, 3'-C-cyano-3'-deoxythymidine (Matsuda, A et al , Tetrahedron 1988, 44: 625-636), and 4'-C-cyano-thymidine (Yang, et al , Tetrahedron letter 1992, 33 : 37-40) having cyano group instead of 3'-hydroxyl group and 4'-hyrogen, respectively, were reported (Prisbe, E. J. et al , Plenum Publishing Co. New York, 1993, 101- 1 13).
SUMMARY OF THE INVENTION
We surprisingly found that an addition of cyano (CN) group at the Γ- position of nucleoside in Ara-C represented by formula (I) maintained the intrinsic anti-cancer cell activity while selectively abolishing the degradation by CDA. This novel molecular mechanism of l'-CN- Ara-C represented by formula (II) having cyano group at l'-positon of nucleoside in Ara-C for CDA resistance was illustrated through molecular docking with human CDA enzyme (1MQ0) in Fig. 1 and CDA assay in Test Example 2 and Fig. 2. And also, this CDA resistance of -CN- Ara-C dramatically improved oral pharmacokinetic property and bioavailability compared to Ara-C in mouse, rat and dog in Fig. 3. Moreover, significantly enhanced efficacy in xenograft was achieved at the same dose compared to Ara-C (Fig. 4). Γ-CN- Ara-C was well distributed in HL-60 tumor tissues and exhibited high drug concentration in HL-60 tumor tissues implying that it will be effective against solid tumors (Fig. 5).
Figure imgf000005_0001
Accordingly, it is an object of the present invention to provide a cytidine deaminase (CDA)-resistant compound selected from the group consisting of Γ- cyano-cytarabine represented by formula (II), Γ-cyano-gemcitabine represented by formula (III), l'-cyano-5-azacytidine represented by formula (IV) and - cyano-5-aza-2'-deoxycytidine represented by formula (V), a prodrug or an acceptable salt thereof:
Figure imgf000006_0001
(H) (IV) X=OH
(V) X=H
It is another object of the present invention to provide a pharmaceutical composition for preventing or treating a cancer, comprising the compound of the present invention as an active ingredient, and a pharmaceutically acceptable carrier or an excipient.
It is a further object of the present invention to provide a method for preventing or treating a cancer comprising administering a therapeutically effective amount of the compound of the present invention to the subject in need thereof.
It is still further object of the present invention to provide a use of the compound of the present invention for the manufacture of a medicament for preventing or treating a cancer. BRIEF DESCRIPTION OF THE DRAWINGS
The above and other objects and features of the present invention will become apparent from the following description of the invention, when taken in conjunction with the accompanying drawings, which respectively show:
Fig. 1 : View of the complex between human CDA (PDB ID: 1MQ0) and
Ara-C (a) as resulting from molecular docking; -CN-Ara-C (b);
Fig. 2: Relative susceptibility of cytidine, Ara-C and -CN- Ara-C to CDA of Test Example 2; Fig. 3: Pharmacokinetics of the Ara-C (mouse) and -CN-Ara-C (mouse, rat and dog) of Test Example 4 through IV and PO administration routes;
Fig. 4: The tumor volume of IP and PO Ara-C and Γ-CN- Ara-C administered mice in HL-60 human leukemia xenograft of Test Example 5; and Fig. 5: Γ-CN- Ara-C concentration in plasma and tumor tissues in HL-60 human leukemia xenograft of Test Example 6.
DETAILED DESCRIPTION OF THE INVENTION
The present invention provides a cytidine deaminase (CDA)-resistant compound selected from the group consisting of -cyano-cytarabine represented by formula (II), Γ-cyano-gemcitabine represented by formula (III), l'-cyano-5- azacytidine represented by formula (IV) and l'-cyano-5-aza-2'-deoxy cytidine represented by formula (V), a prodrug or an acceptable salt thereof:
Figure imgf000007_0001
(ii) (lit) (IV) X-OH
(V) X=H
The compound of the present invention, a prodrug or an acceptable salt thereof is highly resistant to deamination by CDA and highly stable against CDA.
The high level of CDA resistance of the compound of the present invention affords animals such as mouse, rat and dog excellent pharmacokinetic properties and in vivo efficacy by PO or IV administration. Further, the present invention provides a pharmaceutical composition for preventing or treating a cancer, comprising the compound of the present invention as an active ingredient, and a pharmaceutically acceptable carrier or an excipient.
The cancer may be selected from the group consisting of hematological malignancies, epithelial tumors, melanoma, leukemia, acute promyelocyte leukemia, lymphoma, osteogenic sarcoma, colon cancer, pancreatic cancer, breast cancer, prostate cancer, ovarian cancer, lung cancer and a combination thereof.
In one embodiment of the present invention, the compound may be formulated in a pharmaceutical composition, comprising one or more of the compound and a pharmaceutically acceptable carrier or an excipient.
The compound can be mixed with a pharmaceutically acceptable carrier or an excipient, diluted by an excipient or enclosed within such a carrier which can be in the form of a capsule, sachet, paper or other container. When the excipient serves as a diluent, it can be a solid, semi-solid, or liquid material, which acts as a vehicle, carrier, or medium for the compound. Thus, the pharmaceutical compositions can be in the form of tablets, pills, powers, lozenges, sachets, cachets, elixirs, suspensions, emulsions, solutions, syrups, soft and hard gelatin capsules, and other orally ingestible formulations.
Some examples of suitable excipients include lactose, dextrose, sucrose, sorbitol, mannitol, starches, gum acacia, calcium phosphate, alginates, tragacanth, gelatin, calcium silicate, microcrystalline cellulose, polyvinylpyrrolidone, cellulose, magnesium carbonate, water, ethanol, propylene glycol, syrup, and methyl cellulose.
The formulations can additionally include lubricating agents such as talc, magnesium stearate, and mineral oil; wetting agents; emulsifying and suspending agents; preserving agents such as methyl- and propyl-hydroxybenzoates, sweetening agents; and flavoring agents. The compositions of the present invention can also be formulated so as to provide quick, sustained or delayed release of the novel compound after administration to the patient by employing procedures known in the art.
As used herein, the term "pharmaceutically acceptable carrier" refers to those components in the particular dosage form employed which are considered inert and are typically employed in the pharmaceutical fields to formulate a dosage form containing a particular active compound. This may include without limitation solids, liquids and gases, used to formulate the particular pharmaceutical product. Examples of carriers include diluents, flavoring agents, solubilizers, suspending agents, binders or tablet disintegrating agents, encapsulating materials, penetration enhancers, solvents, emollients, thickeners, and dispersants, sustained release forms, such as matrices, transdermal delivery components, buffers, stabilizers, and the like.
The pharmaceutical composition of the present invention may be formulated in the absence or presence of carriers or excipients that provide a sustained released effect.
The pharmaceutical composition of the present invention may be prepared according to methods known in the art. It is contemplated that administration of such compositions may be via oral, injectable and/or parenteral routes depending upon the needs of the subject. The pharmaceutical composition of the present invention can also be administered by nasal or oral inhalation, oral ingestion, intramuscular injection, intravenous injection, intraperitoneal injection, transdermally, or other forms of administration.
Aerosol formulations for use in this invention typically include propellants, such as a fluorinated alkane, surfactants and co-solvents and may be filled into aluminum or other conventional aerosol containers which are then closed by a suitable metering valve and pressurized with propellant, producing a metered dose inhaler. Aerosol preparations are typically suitable for nasal or oral inhalation, and may be in powder or solution form, in combination with a compressed gas, typically compressed air. Additionally, aerosols may be useful topically. Topical preparations useful herein include creams, ointments, solutions, suspensions and the like. These may be formulated to enable one to apply the appropriate dosage topically to the affected area once daily, up to 3-4 times daily as appropriate. Topical sprays may be included herein as well.
Depending upon the compound, transdermal delivery may be an option, which can provide a relatively steady state delivery of the medication. Transdermal delivery typically involves the use of a compound in solution, with an alcoholic vehicle, optionally a penetration enhancer, such as a surfactant and other optional ingredients. Transdermal delivery systems of matrix and reservoir type are examples of suitable transdermal systems. Transdermal delivery differs from conventional topical treatment in that the dosage form delivers a systemic dose of medication to the patient.
The present invention includes a method for inhibiting the growth of a cell using a compound of the present invention. In particular, the present invention provides a method for inhibiting the growth of a cancer cell in a subject, which comprises administering a therapeutically effective amount of the compound to the subject in need thereof.
Also, the present invention provides a method for preventing or treating a cancer in a subject, which comprises administering a therapeutically effective amount of the compound to the subject in need thereof.
The compound of the present invention, a prodrug or an acceptable salt thereof may be administered orally or intravenously. Combination therapy includes combining the method of preventing or treating cancer as described in the invention and one or more cancer therapeutic methods. The cancer therapeutic methods include surgical therapy, radiation therapy, administering an anticancer agent (including, for example, antineoplastics, novantrone, bicalutamide, esterified estrogens, goserelin, histrelin, leuprolide, nilandron, triptorelin pamoate, docetaxel, taxotere, carboplatin, and cisplatin angiogenesis inhibitors), immunotherapy, antineoplastons, investigational drugs, vaccines, and less conventional therapies (sometimes referred to as novel or innovative therapies, which include, for example, chemoembolization, hormone therapy, local hyperthermia, photodynamic therapy, radiofrequency ablation, stem cell transplantation, and gene therapy).
In embodiments of the method of preventing or treating cancer, the compound may be administered in combination with other active agents. Generally, the compound used in the methods of prevention or treatment is administered in an amount which effectively achieves the desired therapeutic result in a subject. The compound may be administered as a pharmaceutical composition, or in the absence of a carrier or diluent. Naturally, the dosages of the compound will vary somewhat depending upon the components of the compound, the rate of in vivo hydrolysis, etc. Those skilled in the art can determine the optimal dose of the compound based on clinical experience and the treatment indication.
Preferably, the amount of compound administered to a subject is about 0.1 to about 100 mg/kg of body weight, more preferably, about 5 to about 40 mg/kg. Other preferred dosages include about 0.1 to about 10 mg/kg of body weight, about 1 to about 100 mg/kg of body weight, about 1 to about 60 mg/kg of body weight, about 1 to about 10 mg/kg of body weight, about 10 to about 100 mg/kg of body weight, about 10 to about 60 mg/kg of body weight, about 20 to about 60 mg/kg of body weight and about 30 to about 50 mg/kg of body weight.
The compound can also be converted into a pharmaceutically acceptable salt or pharmaceutically acceptable solvate or other physical forms (e.g., polymorphs) by methods known in the art field.
The subjects that may be treated using the compounds of the present invention may be mammals including humans. The present invention also includes a use of the compound of the present invention for the manufacture of a medicament for preventing or treating a cancer.
The present invention also includes a kit comprising one or more of the compound of the present invention and instructions for its use.
The following Examples are intended to further illustrate the present invention without limiting its scope. TEST EXAMPLE 1: Molecular docking with CDA structure
The coordinates of the X-ray structures of the human CDA in complex with cytidine (PDB ID: 1MQ0) were retrieved from the protein data bank. The structure was then subjected to the Protein Preparation Wizard workflow implemented in the Schrodinger package. The molecules were docked in the Gold program with Goldscore. After the scoring, the pose of ligand was selected with the best Goldscore. In the co-crystal structure of human CDA (1MQ0) with l-bata-ribofuranosyl-l,3-diazepinone (reference), goldscore showed 52.6. For clarity, only the side chains of the residues are shown in Fig. 1. All the residues shown in Fig. 1 belong to human CDA monomer Al .
As shown in Fig.1 (a), Ara-C favorably interacts with the E67 through the H-bonds and with the hydrophobic V38 and C65 with preserved goldscore 51.9 compared to reference goldscore 52.6. However, this interaction is disrupted by the presence of CN in 1 '-position of Ara-C. As shown in Fig.l (b), Γ-CN group causes crash in binding pocket with reduced goldscore 45.8.
TEST EXAMPLE 2: Cytidine deaminase (CDA) assay
Activities of human cytidine deaminase on cytidine, Ara-C and Γ-CN- Ara-C were compared. His6-tagged, E. co/ -purified recombinant human cytidine deaminase (CD A) was incubated with 0.01 mM of each of the cytidine, Ara-C and l '-CN- Ara-C, respectively, in a 1 mL reaction, containing 20 mM Tris-HCl (pH 8.0), 1 mM DTT, 2 mM EDTA, 100 mM NaCl and 40% glycerol at 25°C. Changes in observance at OD282 were recorded at every 10 seconds for 3 minutes using the continuous spectrophotometric method (A. Amici, et al., Br. J. Haematol , 1989, 73 : 392-395).
[Table 1]
Figure imgf000013_0001
As shown in Table 1 , and Fig. 2, -CN- Ara-C was not a substrate of CD A, and completely resistant to deamination by CDA in vitro.
TEST EXAMPLE 3: Anti-proliferative activity test
Anti-proliferative activities in IC50 (uM) of Ara-C and l '-CN- Ara-C against hematological cancer (ML-2, MOLT-4, MV-4- 1 1, and HL-60), colon cancer (HCT- 1 16) and liver cancer (Hep G2) cell lines were compared.
CellTiter-Glo® Luminescent Cell Viability Assay (Promega #G7572) was used to measure the 50% inhibition concentration (IC50) of the Ara-C and l '-CN- Ara-C against ML-2 (human acute myeloma leukemia), MOLT-4 (human acute lymphoblastic leukemia), MV-4- 1 1 (human biphenotypic B myelomonocytic leukemia), HL-60 (human acute promyelocyte leukemia), Hep G2 (human hepatocellular carcinoma), HCT 1 16 (human colorectal carcinoma) cell lines. Six hundred cells in 90 μΐ media were plated per well in duplicates in a 96-well plate. Cells were incubated in a humidified incubator at 37°C with 5% CO2 during the assay. DMSO was added to the test compounds with the final DMSO concentration being 0.1% [v/v] of the culture. 100 μΐ of CellTiter-GkT was added to the equal volume of cultured cells to read luminescence in En Vision Multi Label Reader. The results are shown in Table 2 below. [Table 2]
Figure imgf000014_0001
As shown in Table 2, l '-CN- Ara-C exhibited the proliferative activities on hematological cancer, liver cancer and colon cancer cells.
TEST EXAMPLE 4: Animal pharmacokinetics
Pharmacokinetics of Ara-C and l'-CN- Ara-C were evaluated in ICR mice following intravenous (IV) administration at 10 mg/kg and per oral (PO) administration at 10 mg/kg dose level, respectively.
For serum compound analysis, blood samples were collected by cardiac puncture over 24 hr time course. 20 μΐ of spiked plasma was added into 96 well plate, adding ten volumes of internal standard (IS) in acetonitrile (ACN) to precipitate proteins, mixed fully, centrifuged for 10 min at 4,000 rpm. 150 μΐ of supernatant was transferred to another pre-labeled 96 well plate, mixed with 150 μΐ of water, then injected 5 μΐ or 10 μΐ into the liquid chromatography-mass spectrometry (LC-MS) system under the following conditions:
Column: API-4000 + Waters UPLC (TCLM08);
Mobile phase: water : MeOH = 100:0, 30:70, 5 :95 and 100:0 (v/v %);
Flow rate: 0.45 mL/min. Lower limit of quantification (LLOQ) was at 1 ng/mL. Pharmacokinetic parameters of half-life (T1/2), clearance (CL), maximum plasma concentration (Cmax), time when the maximal serum concentration of compound is achieved (Tmax) and bioavailability (F%), exposure (AUC) and volume of distribution (Vss) of the compound were calculated using Phoenix WinNonlin 6.3 (non- compartmental model). The results are shown in Table 3, and Fig. 3.
Also, pharmacokinetics of Ara-C and -CN-Ara-C were evaluated in Sprague Dawley (SD) rats following IV administration at 3 mg/kg dose level, and PO administration at 3 mg/kg dose level, respectively. The analytical procedures were as described in the above and the results of rat pharmacokinetics are shown in Table 3, and Fig. 3. Further, pharmacokinetics of Ara-C and l'-CN- Ara-C were evaluated in beagle dogs following IV administration at 3 mg/kg and PO administration at 3 mg/kg dose level, respectively. The analytical procedures were as described in the above and the results are shown in Table 3, and Fig. 3. [Table 3]
Dose T C CI RT , vz
Compounds animal route F%
(mg/kg) (hr) (u hr) (uM) (mL/min/kg) (hr) (L/Kg)
IV 10 0.8 33.3 20.5 1.4
Ara-C mouse
PO 10 0.7 8.6 5 26
IV 10 4.4 25.0 24.9 1.1 9.5 l '-CN-Ara-C mouse
PO 10 4.4 17.2 5 2.3 69
0.5
IV 3 4.4 25.1 7.5 1.0
l '-CN- Ara-C rat (Vss)
PO 3 3.0 7.1 2.0 2.8 28
0.8
IV 3 3.9 47.4 3.9 3.1
l '-CN-Ara-C dog (Vss)
PO 3 3.7 40.8 7.4 4.5 87 As shown in Table 3, -CN- Ara-C showed significantly improved oral bioavailability, longer half-life and increased clearance in dog.
TEST EXAMPLE 5: Tumor growth inhibition efficacy of l'-CN-Ara- C in HL-60 human leukemia xenograft
Tumor growth inhibition (TGI) activities of IP-administered Ara-C, and IP/PO-administered l'-CN- Ara-C were compared in HL-60 human leukemia xenografts.
Tumor growth inhibitions were observed in in vivo subcutaneous mouse xenograft model (HL-60 cell line, Corning #3603). NOD/SCID female mice of 6-8 week age and weighing approximately 18-22 g, were used for tumor inoculation. Each mouse was inoculated subcutaneously at the right flank with HL-60 cancer cells (1 x 107 cells) in mixture of 100 μΐ of PBS and 100 μΐ of matrigel™ for tumor development. Eight mice were used for each group (n=8 / group). The amounts of treated compounds are represented in milligram (mg) of compound per kilogram (kg) of animal body weight (mg/kg, mpk). Administration route of the test compound was either intraperitoneal (IP) injection or peros (PO, oral) injection. Three groups, i.e., vehicle group (Control group), Ara-C group (treatment at 40 mpk of cytarabine through IP injection; Comparative group) and Γ-CN- Ara-C group (treatment at 40 mpk through IP and 60 mpk through PO injection) of mice were used. The overall administration schedule was 2 cycles of treatment for 5 days (5d ON), followed by 2 days of non-treatment (2d OFF). Tumor sizes were measured using a caliper, once every 4 days, and the tumor volume was expressed in mm using Equation 1 :
[Equation 1]
V = 0.5 a x b2
Wherein, a and b are the long and short diameters of the tumor, respectively. The tumor volume (mm ) and body weight (g) results of the three groups are shown in Fig. 4. Also, tumor growth inhibition (% TGI) and body weight change (% BW change) compared to that of Ara-C were shown in Table 4.
[Table 4]
Figure imgf000017_0001
As shown in Table 4 and Fig. 4, oral (PO) dosing of Γ-CN- Ara-C shows a good potency.
Further, IP dosing of Γ-CN- Ara-C exhibited superior efficacy (73% TGI) to IP dosing of Ara-C at the same dose (40 mpk).
TEST EXAMPLE 6: Drug concentration in plasma/tumor tissue
The concentration of Ara-C and -CN- Ara-C in plasma/tumor tissue were evaluated in Test Example 5 in HL-60 human leukemia xenografts.
As shown in Fig. 5, Γ-CN- Ara-C retained high intracellular drug concentration in plasma/tumor tissue from leukemia HL-60 cell derived xenograft model.

Claims

WHAT IS CLAIMED IS:
1. A cytidine deaminase (CDA)-resistant compound selected from the group consisting of l'-cyano-cytarabine represented by formula (II), Γ- cyano-gemcitabine represented by formula (III), l'-cyano-5-azacytidine represented by formula (IV) and -cyano-5-aza-2'-deoxycytidine represented by formula (V), a prodrug or an acceptable salt thereof:
Figure imgf000018_0001
(II) H) (IV) X-OH
(V) X=H
2. A pharmaceutical composition for preventing or treating a cancer, comprising the compound according to claim 1 as an active ingredient, and a pharmaceutically acceptable carrier or an excipient.
3. The pharmaceutical composition of claim 2, wherein the cancer is selected from the group consisting of hematological malignancies, epithelial tumors, melanoma, leukemia, acute promyelocytic leukemia, lymphoma, osteogenic sarcoma, colon cancer, pancreatic cancer, breast cancer, prostate cancer, ovarian cancer, lung cancer and a combination thereof.
4. A method for preventing or treating a cancer, which comprises administering a therapeutically effective amount of the compound according to claim 1 to the subject in need thereof.
5. The method of claim 4, wherein the cancer is selected from the group consisting of hematological malignancies, epithelial tumors, melanoma, leukemia, acute promyelocytic leukemia, lymphoma, osteogenic sarcoma, colon cancer, pancreatic cancer, breast cancer, prostate cancer, ovarian cancer, lung cancer and a combination thereof.
6. The method of claim 4, wherein the compound according to claim 1 is administered orally or intravenously.
7. A use of the compound according to claim 1 for the manufacture of a medicament for preventing or treating a cancer.
PCT/KR2016/005715 2015-06-01 2016-05-30 A use of 1'-cyano-cytarabine for cancer treatment WO2016195353A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201562169030P 2015-06-01 2015-06-01
US62/169,030 2015-06-01

Publications (1)

Publication Number Publication Date
WO2016195353A1 true WO2016195353A1 (en) 2016-12-08

Family

ID=57441290

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/KR2016/005715 WO2016195353A1 (en) 2015-06-01 2016-05-30 A use of 1'-cyano-cytarabine for cancer treatment

Country Status (1)

Country Link
WO (1) WO2016195353A1 (en)

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1997023230A1 (en) * 1995-12-22 1997-07-03 East Carolina University Method of treating disorders characterized by overexpression of cytidine deaminase or deoxycytidine deaminase
US20130196941A1 (en) * 2010-07-13 2013-08-01 Clavis Pharma Asa Parenteral formulations of elacytarabine derivatives
WO2015072784A1 (en) * 2013-11-14 2015-05-21 Kainos Medicine, Inc. Mutual prodrug comprising short chain fatty acids and zebularine or 1'-cyano-cytarabine for cancer treatment

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1997023230A1 (en) * 1995-12-22 1997-07-03 East Carolina University Method of treating disorders characterized by overexpression of cytidine deaminase or deoxycytidine deaminase
US6136791A (en) * 1995-12-22 2000-10-24 East Carolina University Agent and method for treating disorders associated with cytidine deaminase or deoxycytidine deaminase overexpression
US20130196941A1 (en) * 2010-07-13 2013-08-01 Clavis Pharma Asa Parenteral formulations of elacytarabine derivatives
WO2015072784A1 (en) * 2013-11-14 2015-05-21 Kainos Medicine, Inc. Mutual prodrug comprising short chain fatty acids and zebularine or 1'-cyano-cytarabine for cancer treatment

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
YOSHIDA, TAKAHIRO ET AL.: "Influence of cytidine deaminase on antitumor activity of 2'-deoxycytidine analogs in vitro and in vivo", DRUG METABOLISM & DISPOSITION, vol. 38, no. 10, 2010, pages 1814 - 1819, XP055332831 *

Similar Documents

Publication Publication Date Title
US20110218170A1 (en) Use of 2'-deoxy-4'-thiocytidine and its analogues as dna hypomethylating anticancer agents
NZ583923A (en) Azacytidine analogues and uses thereof
WO2000076524A1 (en) Combination cancer therapy comprising adenosine and deaminase enzyme inhibitor
EP3068406B1 (en) Mutual prodrug comprising short chain fatty acids and zebularine or 1'-cyano-cytarabine for cancer treatment
Miura et al. Comparison of 1-(2-deoxy-2-fluoro-4-thio-β-D-arabinofuranosyl) cytosine with gemcitabine in its antitumor activity
WO2016195353A1 (en) A use of 1'-cyano-cytarabine for cancer treatment
EP2268287B1 (en) Dosage regimens of an antitumor agent comprising deoxycytidine derivative
JP6845332B2 (en) Pharmaceutical composition for cancer treatment and its use
US8586561B2 (en) Anti-tumor agent comprising cytidine derivative and carboplatin
US20190054103A1 (en) Method of treatment of tp53 wild-type tumors with 2',2'-difluoro-5-aza-2'-deoxycytidine or prodrugs thereof
WO2011109012A1 (en) Use of 2'-deoxy-4'-thiocytidine and its analogues as dna hypomethylating anticancer agents
US20230142647A1 (en) Method of treating cancer or a blood disorder
EP2711008A1 (en) N6,N6-dimethyladenosine for use in treating or preventing primary and metastatic breast cancer
US11179349B2 (en) Use of tumor gene methylation regulator and anti-tumor drugs
EP2854864B1 (en) Compositions comprising oligomers of gemcitabine for use in therapy
MXPA98005083A (en) Method of treatment of disorders characterizopopor the over-expression of citidina deaminasa or deoxicitidina deamin
WO2007095639A2 (en) Napht alimide compositions and uses thereof
EP2711009A1 (en) Compounds for use in treating or preventing primary and metastatic breast and prostate cancer
WO2011109383A1 (en) Use of 2'-deoxy -4'-thiocytidine and its analogues as dna hypomethylating anticancer agents

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: 16803708

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: 16803708

Country of ref document: EP

Kind code of ref document: A1