WO2014170875A1 - Quinazolinedione componds with a sirtuin inhibiting activity - Google Patents

Quinazolinedione componds with a sirtuin inhibiting activity Download PDF

Info

Publication number
WO2014170875A1
WO2014170875A1 PCT/IB2014/060829 IB2014060829W WO2014170875A1 WO 2014170875 A1 WO2014170875 A1 WO 2014170875A1 IB 2014060829 W IB2014060829 W IB 2014060829W WO 2014170875 A1 WO2014170875 A1 WO 2014170875A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound according
cells
phenyl
sirt6
inflammatory
Prior art date
Application number
PCT/IB2014/060829
Other languages
French (fr)
Other versions
WO2014170875A8 (en
Inventor
Alberto DEL RIO
Claudio FRANCESCHI
Marco Daniele PARENTI
Inga Bauer
Santina Bruzzone
Alessia Grozio
Alessio NENCIONI
Original Assignee
Università Degli Studi Di Genova
Alma Mater Studiorum - Università di Bologna
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 Università Degli Studi Di Genova, Alma Mater Studiorum - Università di Bologna filed Critical Università Degli Studi Di Genova
Publication of WO2014170875A1 publication Critical patent/WO2014170875A1/en
Publication of WO2014170875A8 publication Critical patent/WO2014170875A8/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/517Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with carbocyclic ring systems, e.g. quinazoline, perimidine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents

Definitions

  • the present invention relates to the technical field of pharmaceutical industry and, in particular, concerns quinazolinedione compounds with sirtuin inhibiting activity, which are useful in the treatment of different diseases, such as for example metabolic, inflammatory and tumor diseases.
  • Sirtuins are NAD + -dependent enzymes, playing a role in ageing, metabolism, nutritional behavior, cancer and inflammation 1 . Due to their wide implication in the physiopathology of highly prevalent diseases, sirtuins represent an interesting therapeutic target 2 .
  • SIRTl-7 Seven sirtuins (SIRTl-7) are known, and activators of SIRT1 were described, with a potentially positive role in regulating metabolism and extending the duration of a healthy life ("healthspan"), although their mode of action is still controversial 2 . Inhibitors of SIRT1, SIRT2 and SIRT5 were reported too 3 .
  • SIRT6 is involved in genome stability and its deficiency was associated to the development of a progeroid syndrome in mice 4 .
  • SIRT6 promotes DNA repair by different mechanisms comprising deacetylation of histone H3 lysine 9 (H3K9) at telomeric chromatin with consequent association of WRN and telomere maintenance 5 , stabilization of DNA-dependent protein kinase (DNA-PK) at chromatin 6 , as well as poly(ADP- ribose) polymerase 1 (PARP1) mono-ADP-ribosylation and consequent promotion of its activity 7 .
  • H3K9 histone H3 lysine 9
  • PARP1 poly(ADP- ribose) polymerase 1
  • SIRT6 regulates glucose uptake by virtue of its capacity of co-repressing the transcription factor Hifla, a critical regulator of nutrient stress responses 8 .
  • SIRT6 deficient cells exhibit increased Hifla activity, increased glucose uptake and glycolysis, and diminished mitochondrial respiration.
  • SIRT6-deficient mice develop hypoglycaemia and exhibit a pronounced increase in glucose uptake in muscle and brown adipose tissue 4 - 8 .
  • SIRT6 plays a role in inflammation, as shown by the immune defects of SIRT6-deficient mice 4 and by the ability to promote the expression of TNFa, IFN- ⁇ 10 and IL8 11 in response to activating stimuli.
  • the immunogenic activity of SIRT6 reflects, at least in part, its propensity to increase intracellular ADP-ribose levels by virtue of its enzymatic activity 12 .
  • SIRT6-derived ADP-ribose appears to promote Ca 2+ responses, cytokine gene transcription (via the transcription factor NFAT) and cell motility.
  • SIRT6 inhibition appears as a viable strategy for the treatment of cancer (as a means to sensitize cancer cells to anticancer agents and radiotherapy and to reduce cancer-associated inflammation) 4 ' 6 - 12 , metabolic disorders (by virtue of its capacity to increase tissue glucose uptake) and inflammation.
  • no specific SIRT6 inhibitors have been described, whereas the availability of selective SIRT6 inhibitors could lead to the achievement of therapeutic agents with wide application possibilities 13 .
  • Patent application WO 2008/ 138943 relates to a sirtuin inhibitor other than suramin for use in the reduction of TNF- alpha and/ or reduction of local or systemic inflammation in a subject and/ or treatment of TNF-alpha mediated diseases.
  • the inhibitor in question is selected from sirtinol, m-sirtinol, p-sirtinol, splitomicin, dehydrosplitomicin, cambinol and dihydrocoumarin.
  • Patent application WO 2011 /038110 concerns the use of a SIRT6 inhibitor to reduce or inhibit hyperglycaemia or obesity in a subject.
  • SIRT6 inhibitors antibodies anti-SIRT6, interfering RNA molecules and antisense nucleic acids can be mentioned.
  • the problem underlying the present invention was that of providing compounds having inhibiting activity towards sirtuins, in particular, even if not exclusively, towards SIRT6, for the use in inducing glucose tissue uptake (and thus in obtaining a reduction of glycaemic levels) and /or in reducing the production of TNF-alpha and other proinflammatory, chemo tactic, or proangiogenic cytokines (such as IFN- ⁇ and IL8) and/or in interfering with DNA repair in tumor cells, so as to be used as therapeutic agents in the treatment of type I or II diabetes-related diseases and/or inflammatory diseases and/or neoplastic diseases.
  • R 1 H, d-Csalkyl
  • R2 H, Ci-Csalkyl
  • R 5 5- or 6-membered, saturated or aromatic heterocyclic ring containing one or more heteroatoms selected from N, S and O, optionally substituted with a Ci-Csalkyl;
  • R 4 H, Ci-ealkyl, Cs-ecycloalkyl, halogen, -NO 2 , -CN, -OH, -SH, -CF 3 , - CCI3, -COOH, -COOCi-salkyl, -SO 2 OC 1-5 alkyl, -OC 1-5 alkyl, or enantiomers, diastereomers and pharmaceutically acceptable salts thereof, for use as a medicament.
  • alkyl both straight chain and branched chain alkyls are meant.
  • R2 H, CH 3
  • compounds of formula (I) comprise the following compounds:
  • the abovementioned compounds of formula (I) can be used as inhibitors of one or more sirtuins and, in particular SIRT6, to increase glucose tissue uptake in a subject.
  • such compounds can be used in the treatment of: type I and type II diabetes mellitus and complications thereof, such as ketoacidotic coma, hyperglycaemic hyperosmolar condition, atherosclerosis, ischaemic heart disease (angina and myocardial infarction), stroke, peripheral vasculopathy, diabetic retinopathy, diabetic nephropathy, diabetic neuropathy and diabeteic foot.
  • type I and type II diabetes mellitus and complications thereof such as ketoacidotic coma, hyperglycaemic hyperosmolar condition, atherosclerosis, ischaemic heart disease (angina and myocardial infarction), stroke, peripheral vasculopathy, diabetic retinopathy, diabetic nephropathy, diabetic neuropathy and diabeteic foot.
  • compounds of formula (I) can be used as inhibitors of one or more sirtuins, and in particular SIRT6, to reduce the production of TNF-alpha and other proinflammatory, chemotactic, or proangiogenic cytokines (such as IFN- ⁇ and IL8) in a subject and/ or to reduce local or systemic inflammation in a subject and/ or to treat a pathology mediated by TNF-alpha or other proinflammatory, chemotactic, or proangiogenic cytokines (such as IFN- ⁇ and IL8) in a subject having local or systemic excessive or dysregulated cytokine production.
  • SIRT6 to reduce the production of TNF-alpha and other proinflammatory, chemotactic, or proangiogenic cytokines (such as IFN- ⁇ and IL8) in a subject and/ or to reduce local or systemic inflammation in a subject and/ or to treat a pathology mediated by TNF-alpha or other proinflammatory, chemotactic, or proangiogenic cyto
  • inflammatory diseases comprising rheumatoid arthritis, multiple sclerosis, inflammatory bowel disease, systemic lupus erythematosus, vasculitis, Goodpasture syndrome, scleroderma, atherosclerosis, graft versus host disease (GVHD), organ transplant rejection, myocardial infarction, stroke, reperfusion injury after revascularization in heart and other organs.
  • inflammatory diseases comprising rheumatoid arthritis, multiple sclerosis, inflammatory bowel disease, systemic lupus erythematosus, vasculitis, Goodpasture syndrome, scleroderma, atherosclerosis, graft versus host disease (GVHD), organ transplant rejection, myocardial infarction, stroke, reperfusion injury after revascularization in heart and other organs.
  • GVHD graft versus host disease
  • compounds of formula (I) are used as inhibitors of one or more sirtuins, and in particular SIRT6, to interfere with DNA repair in tumor cells and thus exert an anticancer effect and sensitize said cells to antineoplastic agents and radiotherapy.
  • neoplastic diseases comprising pancreatic cancer, breast cancer, colorectal cancer, prostatic cancer, ovarian cancer, melanoma, lung cancer, oesophageal cancer, hepatic carcinoma, lymphomas, leukemias, myeloma, sarcomas, neoplastic cachexia.
  • the present invention relates to a pharmaceutical composition containing a therapeutically effective amount of at least one compound of formula (I) or an enantiomer, diastereomer or pharmaceutically acceptable salt thereof, as defined above and a pharmaceutically acceptable vehicle.
  • Examples of pharmaceutically acceptable salts are those formed with organic acids such as oxalic, tartaric, maleic, succinic and citric, and with inorganic acids such as nitric, hydrochloric, sulphuric and phosphoric acid.
  • Compounds according to the invention having one or more asymmetric carbon atoms can exist as pure enantiomers, pure diastereomers, racemic mixtures of enantiomers, racemates and mixtures of racemates.
  • Compounds and compositions according to the invention can be administered with any available and efficient delivery system, comprising, but not limited to, oral, buccal, parenteral, inhalatory routes, topical application, injection, transdermal or rectal route (e.g. suppositories), in unit dose formulations containing conventional pharmaceutically acceptable and non-toxic supports, adjuvants and carriers.
  • Administration by parenteral route includes subcutaneous, intravenous, intramuscular, intracisternal injection or infusion techniques.
  • Solid dosage forms for administration by oral route comprise, for example, capsule, tablets, powders, granules and gels.
  • the active compound can be mixed with at least one inert diluent, such as for example saccharose, lactose or starch.
  • these dosage forms also comprise additional substances different from inert diluents, such as for example lubricating agents like magnesium stearate.
  • Injection preparations for example sterile, aqueous or oily solutions or suspensions for injection, can be formulated according to known art and optionally using suitable dispersing, wetting agents and/ or suspending agents.
  • Pharmaceutical preparations according to the present invention can be manufactured by using conventional pharmaceutical techniques, as described in various pharmacopoeias or handbooks in the field such as for example "Remington's Pharmaceutical Sciences Handbook", Mack Publishing, New York, 18 th Ed., 1990.
  • the average daily dose of the compounds according to the present invention depends on many factors, such as for example severity of the disease and state of the patient (age, weight, gender): the dose can generally range from 1 mg to 1500 mg/day of compound according to the invention, optionally partitioned in more administrations.
  • the present invention relates to a method for reducing the production of TNF-alpha and other proinflammatory, chemotactic, or proangiogenic cytokines in cells in vitro, comprising the step of exposing the cells to a compound as defined above, a method for increasing glucose uptake in cells in vitro, comprising the step of exposing the cells to said compound, as well as a method for increasing the antiproliferative and cytotoxic effect of antineoplastic drugs and ionizing radiations in vitro, comprising the step of exposing tumor cells to said compound.
  • compositions can be prepared with the compounds according to the present invention for the treatment of different pathologic conditions mediated by sirtuins and, in particular, SIRT6.
  • Some of these diseases such as type II diabetes mellitus, neoplastic diseases and inflammatory diseases, have a very strong epidemiological impact and hence pharmaceutical formulations containing compounds according to the present invention are susceptible of wide employment in different fields of medical therapy, with technical, economical and manufacturing advantages related to the fact of being based on low molecular weight and stable organic molecules.
  • the formulations can comprise further active substances, or can be used in combination with other formulations containing other active substances, such as for example antiinflammatory, blood glucose lowering agents, chemotherapies, or with radiotherapy.
  • subject reference is made, in the present application, to animals, preferably mammals and, in a particularly preferred way, human individuals.
  • a therapeutic treatment By the term “treating” or “treatment” both a therapeutic treatment and a preventive, prophylactic treatment are meant, as well as a treatment achieving a prolongation of the survival in comparison with what expected in the absence of treatment.
  • therapeutically effective amount refers to an amount able to determine a biological or therapeutic response in an animal or human tissue or system pursued by a researcher, physician or veterinary and, in particular, that can prevent or alleviate at least one local or systemic symptom of the treated disease.
  • the activity of the compounds according to the present invention was evaluated by a series of biological tests and in particular those summarized hereinafter.
  • IC50 values were determined using assays in commercial kits. All the compounds were dissolved at a 50mM concentration in DMSO. The compounds concentrations in the measurements for IC50 determination were in the range from 8 ⁇ to 5mM. IC50 values were determined from non-linear logarithmic regression curves by GraphPad Prism (GraphPad Software, the Jolla, CA - USA). Three independent IC50 measurements were performed for each compound. Western Blot analysis.
  • Pancreatic adenocarcinoma BxPC3 cells were incubated for 18 hours with various compounds ( ⁇ final concentration). Cells were lysed in lysis buffer (50mM Tris-HCl, pH 7.5, 150mM NaCl, 1% NP40 and cocktail of protease inhibitors) and 30 Ug of proteins were loaded onto 15% polyacrylamide gel and separated by SDS-PAGE. Proteins were transferred onto nitrocellulose membranes and incubated with the primary antibody specific against acetylated H3K9 (Sigma, Milan) and with the antibody against actin (Santa Cruz Biotechnology, Santa Cruz, USA). After incubation with suitable secondary antibodies and ECL detection (GE Healthcare, Milan), band intensity was measured with the ChemiDoc imaging system (BioRad) and the H3K9 acetylation level was normalized to actin.
  • lysis buffer 50mM Tris-HCl, pH 7.5, 150mM NaCl, 1% NP40 and cocktail of protease inhibitors
  • BxPC-3 cells overexpressing SIRT6 (Bauer I. et al., J. Biol. Chem. 2012 Oct 18) (3 x 10 5 cells/well) were seeded in 6-well plates and allowed to adhere for 24 hours before being incubated for 18 hours with the various compounds ( ⁇ final concentration). Subsequently, to induce cytokine expression, cells were stimulated for 48 hours with 25 ng/ml of phorbol myristate acetate (PMA; Sigma Aldrich). Subsequently, supernatants were collected and assayed for IL8 and TNF using DuoSet ® ELISA kits available on the market (R&D Systems, Minneapolis, USA). The concentration in supernatants was normalized to the cell density measured with sulforhodamine B. Glucose uptake experiments
  • Glucose uptake was stopped by immediately removing the labelling mixture and washing the cells 3 times with ice-cold KRH. The cells were then lysed with 0.1% sodium dodecyl sulfate (SDS) and an aliquot of each lysate was used for the scintillation count in a LS 6500 Beta-Counter (Beckman- Coulter, CA). Non-specific uptake in the presence of 20 ⁇ citocalasine B and 200 ⁇ phloretin was subtracted from each experimental value.
  • SDS sodium dodecyl sulfate
  • Reactions were performed in a total volume of 10 ⁇ , containing 20 pg of recombinant SIRT6 (produced following published protocols 13 ) in 50mM Tris-HCl, pH 8.0, 150mM NaCl, lOmM dithiothreitol, and 0.4 pCi [ 3 H]NAD (40 Ci mmol" 1 ; Perkin Elmer, Boston, MA), in the presence or absence of different compounds (200 ⁇ final concentration) or nicotinamide (lOOmM final concentration) for 45 minutes at 22°C.

Landscapes

  • Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Diabetes (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Hematology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Endocrinology (AREA)
  • Emergency Medicine (AREA)
  • Obesity (AREA)
  • Pain & Pain Management (AREA)
  • Rheumatology (AREA)
  • Epidemiology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Compounds Of Unknown Constitution (AREA)

Abstract

Compounds of general formula (I) or enantiomers, diastereomers and pharmaceutically acceptable salts thereof, for use as a medicament, in particular as inhibitors of sirtuins, with particular reference to SIRT6; such compounds increase glucose tissue uptake and find employment in the treatment of type I and type II diabetes mellitus and complications thereof; reduce the production of TNF-alpha and other inflammatory, chemotactic, or proangiogenic cytokines, finding use in the treatment of inflammatory diseases; interfere with DNA repair in tumor cells and thus exert an anticancer effect and sensitize such cells to antineoplastic agents and radiotherapy, hence finding employment in the treatment of neoplastic diseases.

Description

Title: Quinazolinedione compounds with a sirtuin inhibiting activity
DESCRIPTION
The work that has led to this invention was funded by the Seventh Framework Programme of the European Union (FP7 2007-2013) under the grant agreement N° 256986.
Field of application
The present invention relates to the technical field of pharmaceutical industry and, in particular, concerns quinazolinedione compounds with sirtuin inhibiting activity, which are useful in the treatment of different diseases, such as for example metabolic, inflammatory and tumor diseases.
Known art
Sirtuins are NAD+-dependent enzymes, playing a role in ageing, metabolism, nutritional behavior, cancer and inflammation1. Due to their wide implication in the physiopathology of highly prevalent diseases, sirtuins represent an interesting therapeutic target2.
Seven sirtuins (SIRTl-7) are known, and activators of SIRT1 were described, with a potentially positive role in regulating metabolism and extending the duration of a healthy life ("healthspan"), although their mode of action is still controversial2. Inhibitors of SIRT1, SIRT2 and SIRT5 were reported too3.
SIRT6 is involved in genome stability and its deficiency was associated to the development of a progeroid syndrome in mice4. SIRT6 promotes DNA repair by different mechanisms comprising deacetylation of histone H3 lysine 9 (H3K9) at telomeric chromatin with consequent association of WRN and telomere maintenance5, stabilization of DNA- dependent protein kinase (DNA-PK) at chromatin6, as well as poly(ADP- ribose) polymerase 1 (PARP1) mono-ADP-ribosylation and consequent promotion of its activity7. In addition to its role in DNA repair, SIRT6 regulates glucose uptake by virtue of its capacity of co-repressing the transcription factor Hifla, a critical regulator of nutrient stress responses8. SIRT6 deficient cells exhibit increased Hifla activity, increased glucose uptake and glycolysis, and diminished mitochondrial respiration.
In line with these findings, SIRT6-deficient mice develop hypoglycaemia and exhibit a pronounced increase in glucose uptake in muscle and brown adipose tissue4-8.
Finally, SIRT6 plays a role in inflammation, as shown by the immune defects of SIRT6-deficient mice4 and by the ability to promote the expression of TNFa, IFN-γ10 and IL811 in response to activating stimuli. The immunogenic activity of SIRT6 reflects, at least in part, its propensity to increase intracellular ADP-ribose levels by virtue of its enzymatic activity12. In turn, SIRT6-derived ADP-ribose appears to promote Ca2+ responses, cytokine gene transcription (via the transcription factor NFAT) and cell motility. Thus, SIRT6 inhibition appears as a viable strategy for the treatment of cancer (as a means to sensitize cancer cells to anticancer agents and radiotherapy and to reduce cancer-associated inflammation)4 ' 6-12, metabolic disorders (by virtue of its capacity to increase tissue glucose uptake) and inflammation. However, to date, no specific SIRT6 inhibitors have been described, whereas the availability of selective SIRT6 inhibitors could lead to the achievement of therapeutic agents with wide application possibilities13.
Patent application WO 2008/ 138943 relates to a sirtuin inhibitor other than suramin for use in the reduction of TNF- alpha and/ or reduction of local or systemic inflammation in a subject and/ or treatment of TNF-alpha mediated diseases. The inhibitor in question is selected from sirtinol, m-sirtinol, p-sirtinol, splitomicin, dehydrosplitomicin, cambinol and dihydrocoumarin.
Patent application WO 2011 /038110 concerns the use of a SIRT6 inhibitor to reduce or inhibit hyperglycaemia or obesity in a subject. Among SIRT6 inhibitors, antibodies anti-SIRT6, interfering RNA molecules and antisense nucleic acids can be mentioned.
Summary of the invention The problem underlying the present invention was that of providing compounds having inhibiting activity towards sirtuins, in particular, even if not exclusively, towards SIRT6, for the use in inducing glucose tissue uptake (and thus in obtaining a reduction of glycaemic levels) and /or in reducing the production of TNF-alpha and other proinflammatory, chemo tactic, or proangiogenic cytokines (such as IFN-γ and IL8) and/or in interfering with DNA repair in tumor cells, so as to be used as therapeutic agents in the treatment of type I or II diabetes-related diseases and/or inflammatory diseases and/or neoplastic diseases.
Such a problem was solved according to the invention by the provision of compounds of formula (I)
Figure imgf000004_0001
in which:
R1 = H, d-Csalkyl; R2 = H, Ci-Csalkyl;
R3 = optionally substituted phenyl; optionally substituted phenoxy; -CH2- (CH2)n-phenyl optionally substituted on the benzene ring, where n = 0, 1 or 2; or -Y-R5, where Y = single bond, -O, -CH2, and
R5 = 5- or 6-membered, saturated or aromatic heterocyclic ring containing one or more heteroatoms selected from N, S and O, optionally substituted with a Ci-Csalkyl;
R4 = H, Ci-ealkyl, Cs-ecycloalkyl, halogen, -NO2, -CN, -OH, -SH, -CF3, - CCI3, -COOH, -COOCi-salkyl, -SO2OC1-5alkyl, -OC1-5alkyl, or enantiomers, diastereomers and pharmaceutically acceptable salts thereof, for use as a medicament.
With "alkyl" both straight chain and branched chain alkyls are meant.
Preferably, in the compounds of formula (I) the substituents have the following meanings: R1 = H, CH3
R2 = H, CH3
R3 = phenyl, phenoxy, benzyl or phenylethyl optionally substituted with a Ci-3alkyl on the aromatic ring or Y-R5, where Y = a single bond, -O-, -C¾-, and R5 = 5- or 6-membered heteroaromatic ring, containing 1 or 2 N atoms, optionally substituted with a Ci-Csalkyl;
R4 = H.
Particularly preferred are compounds of formula (I) in which: R1 = H, CH3 R2 = H, CH3
R3 = phenyl, benzyl, phenylethyl or Y-R5, where Y = a single bond, -0-, -CH2-, and R5 = pyridinyl or imidazolyl; R4 = H. Further preferably, compounds of formula (I) comprise the following compounds:
(1) 2,4-dioxo-N-[4-(pyridin-3-yloxy)phenyl]-l ,2,3,4-tetrahydroquinazoline- 6- sulfonamide;
(2) N-[4-(lH-imidazol-l-ylmethyl)phenyl]-2,4-dioxo-l ,2,3,4- tetrahydroquinazoline-6- sulfonamide; (3) 2,4-dioxo-N-{3-[2-(pyridin-4-yl)ethyl]phenyl}- 1,2,3,4- tetrahydroquinazoline-6-sulfonamide;
Figure imgf000006_0001
(5) 2,4-dioxo-N-(2-phenylphenyl)- l,2,3,4-tetrahydroquinazoline-6- sulfonamide;
(6) 1 ,3-dimethyl-2,4-dioxo-N-[4-(pyridin-4-ylmethyl)phenyl]- 1 ,2,3,4- tetrahydroquinazoline-6-sulfonamide.
The abovementioned compounds of formula (I) can be used as inhibitors of one or more sirtuins and, in particular SIRT6, to increase glucose tissue uptake in a subject.
According to an aspect of the present invention, such compounds can be used in the treatment of: type I and type II diabetes mellitus and complications thereof, such as ketoacidotic coma, hyperglycaemic hyperosmolar condition, atherosclerosis, ischaemic heart disease (angina and myocardial infarction), stroke, peripheral vasculopathy, diabetic retinopathy, diabetic nephropathy, diabetic neuropathy and diabeteic foot.
According to "another aspect of the present invention, compounds of formula (I) can be used as inhibitors of one or more sirtuins, and in particular SIRT6, to reduce the production of TNF-alpha and other proinflammatory, chemotactic, or proangiogenic cytokines (such as IFN-γ and IL8) in a subject and/ or to reduce local or systemic inflammation in a subject and/ or to treat a pathology mediated by TNF-alpha or other proinflammatory, chemotactic, or proangiogenic cytokines (such as IFN-γ and IL8) in a subject having local or systemic excessive or dysregulated cytokine production.
Consequently, such compounds find application in the treatment of inflammatory diseases comprising rheumatoid arthritis, multiple sclerosis, inflammatory bowel disease, systemic lupus erythematosus, vasculitis, Goodpasture syndrome, scleroderma, atherosclerosis, graft versus host disease (GVHD), organ transplant rejection, myocardial infarction, stroke, reperfusion injury after revascularization in heart and other organs.
According to a further aspect of the present invention, compounds of formula (I) are used as inhibitors of one or more sirtuins, and in particular SIRT6, to interfere with DNA repair in tumor cells and thus exert an anticancer effect and sensitize said cells to antineoplastic agents and radiotherapy.
Consequently, such compounds find application in the treatment of neoplastic diseases comprising pancreatic cancer, breast cancer, colorectal cancer, prostatic cancer, ovarian cancer, melanoma, lung cancer, oesophageal cancer, hepatic carcinoma, lymphomas, leukemias, myeloma, sarcomas, neoplastic cachexia.
In a further aspect, the present invention relates to a pharmaceutical composition containing a therapeutically effective amount of at least one compound of formula (I) or an enantiomer, diastereomer or pharmaceutically acceptable salt thereof, as defined above and a pharmaceutically acceptable vehicle.
Examples of pharmaceutically acceptable salts are those formed with organic acids such as oxalic, tartaric, maleic, succinic and citric, and with inorganic acids such as nitric, hydrochloric, sulphuric and phosphoric acid.
Compounds according to the invention having one or more asymmetric carbon atoms can exist as pure enantiomers, pure diastereomers, racemic mixtures of enantiomers, racemates and mixtures of racemates.
Compounds and compositions according to the invention can be administered with any available and efficient delivery system, comprising, but not limited to, oral, buccal, parenteral, inhalatory routes, topical application, injection, transdermal or rectal route (e.g. suppositories), in unit dose formulations containing conventional pharmaceutically acceptable and non-toxic supports, adjuvants and carriers. Administration by parenteral route includes subcutaneous, intravenous, intramuscular, intracisternal injection or infusion techniques.
Solid dosage forms for administration by oral route comprise, for example, capsule, tablets, powders, granules and gels. In such solid dosage forms the active compound can be mixed with at least one inert diluent, such as for example saccharose, lactose or starch. Normally, these dosage forms also comprise additional substances different from inert diluents, such as for example lubricating agents like magnesium stearate.
Injection preparations, for example sterile, aqueous or oily solutions or suspensions for injection, can be formulated according to known art and optionally using suitable dispersing, wetting agents and/ or suspending agents. Pharmaceutical preparations according to the present invention can be manufactured by using conventional pharmaceutical techniques, as described in various pharmacopoeias or handbooks in the field such as for example "Remington's Pharmaceutical Sciences Handbook", Mack Publishing, New York, 18th Ed., 1990. The average daily dose of the compounds according to the present invention depends on many factors, such as for example severity of the disease and state of the patient (age, weight, gender): the dose can generally range from 1 mg to 1500 mg/day of compound according to the invention, optionally partitioned in more administrations. Finally, in a further aspect, the present invention relates to a method for reducing the production of TNF-alpha and other proinflammatory, chemotactic, or proangiogenic cytokines in cells in vitro, comprising the step of exposing the cells to a compound as defined above, a method for increasing glucose uptake in cells in vitro, comprising the step of exposing the cells to said compound, as well as a method for increasing the antiproliferative and cytotoxic effect of antineoplastic drugs and ionizing radiations in vitro, comprising the step of exposing tumor cells to said compound.
Pharmaceutical formulations can be prepared with the compounds according to the present invention for the treatment of different pathologic conditions mediated by sirtuins and, in particular, SIRT6. Some of these diseases, such as type II diabetes mellitus, neoplastic diseases and inflammatory diseases, have a very strong epidemiological impact and hence pharmaceutical formulations containing compounds according to the present invention are susceptible of wide employment in different fields of medical therapy, with technical, economical and manufacturing advantages related to the fact of being based on low molecular weight and stable organic molecules. Moreover, the formulations can comprise further active substances, or can be used in combination with other formulations containing other active substances, such as for example antiinflammatory, blood glucose lowering agents, chemotherapies, or with radiotherapy.
Detailed description
Compounds of formula (I) described above can be prepared starting from the corresponding 6-chlorosulfonyl-quinazoline-2,4-diones of formula
Figure imgf000009_0001
(II)
by reaction with a compound of formula (III)
Figure imgf000009_0002
in which R3 and R4 are as defined with reference to compounds of formula (II) .
The reaction is carried out under the conditions described in Kuryazov R. Sh. et al, Chemistry of Heterocyclic Compounds, Vol. 44, No. 3, 2008 "Quinazolines 1. Synthesis and chemical reactions of 6- chlorosulfonyl-quinazoline-2,4-diones".
Preferred compounds mentioned above are available on the market from the following sources: (1) : from the company Asinex, product identification: SYN 17739303;
(2) : from the company Asinex, product identification: SYN 17738896; (3): from the company Asinex, product identification: SYN
17742030;
(4) : from the company Asinex, product identification: SYN 17736818;
(5) : from the company Asinex, product identification: SYN 17735999;
(6) : from the company Princeton BioMolecular Research, Inc, product identification: OSSL_304544.
By the term "subject" reference is made, in the present application, to animals, preferably mammals and, in a particularly preferred way, human individuals.
By the term "treating" or "treatment" both a therapeutic treatment and a preventive, prophylactic treatment are meant, as well as a treatment achieving a prolongation of the survival in comparison with what expected in the absence of treatment. The expression "therapeutically effective amount" refers to an amount able to determine a biological or therapeutic response in an animal or human tissue or system pursued by a researcher, physician or veterinary and, in particular, that can prevent or alleviate at least one local or systemic symptom of the treated disease. The activity of the compounds according to the present invention was evaluated by a series of biological tests and in particular those summarized hereinafter.
Sirtuin inhibition test
In vitro activity of the compounds according to the present invention in terms of sirtuin inhibition was determined using commercial kits for SIRT6, SIRT1 and SIRT2 available from the company Cayman, Ann Arbor, USA, following the instructions of the manufacturer.
IC50 values were determined using assays in commercial kits. All the compounds were dissolved at a 50mM concentration in DMSO. The compounds concentrations in the measurements for IC50 determination were in the range from 8μΜ to 5mM. IC50 values were determined from non-linear logarithmic regression curves by GraphPad Prism (GraphPad Software, the Jolla, CA - USA). Three independent IC50 measurements were performed for each compound. Western Blot analysis.
Pancreatic adenocarcinoma BxPC3 cells were incubated for 18 hours with various compounds (ΙΟΟμΜ final concentration). Cells were lysed in lysis buffer (50mM Tris-HCl, pH 7.5, 150mM NaCl, 1% NP40 and cocktail of protease inhibitors) and 30 Ug of proteins were loaded onto 15% polyacrylamide gel and separated by SDS-PAGE. Proteins were transferred onto nitrocellulose membranes and incubated with the primary antibody specific against acetylated H3K9 (Sigma, Milan) and with the antibody against actin (Santa Cruz Biotechnology, Santa Cruz, USA). After incubation with suitable secondary antibodies and ECL detection (GE Healthcare, Milan), band intensity was measured with the ChemiDoc imaging system (BioRad) and the H3K9 acetylation level was normalized to actin.
Effects on TNF-alpha
BxPC-3 cells overexpressing SIRT6 (Bauer I. et al., J. Biol. Chem. 2012 Oct 18) (3 x 105 cells/well) were seeded in 6-well plates and allowed to adhere for 24 hours before being incubated for 18 hours with the various compounds (ΙΟΟμΜ final concentration). Subsequently, to induce cytokine expression, cells were stimulated for 48 hours with 25 ng/ml of phorbol myristate acetate (PMA; Sigma Aldrich). Subsequently, supernatants were collected and assayed for IL8 and TNF using DuoSet® ELISA kits available on the market (R&D Systems, Minneapolis, USA). The concentration in supernatants was normalized to the cell density measured with sulforhodamine B. Glucose uptake experiments
1 x 105 BxPC-3 cells engineered to express the pRETROSUPER vector (pRS) or a SIRT6-targeted shRNA (sh2 SIRT6) (Bauer I, et al. J Biol Chem. 2012 Oct 18) were plated in each well of a 12-well plate in 500 μΐ of standard culture medium. 72 hours later, cells were incubated (or not incubated) in the presence of the compounds at a concentration of 200μΜ for 18 h. Cells were then washed once with 1 ml of PBS buffer and glucose transport was measured by addition of D-glucose/0.5mM [14C]-2-deoxy-D- glucose (0.2 μCi/well) in KRH buffer for 5 mins at 37°C. Glucose uptake (absorption) was stopped by immediately removing the labelling mixture and washing the cells 3 times with ice-cold KRH. The cells were then lysed with 0.1% sodium dodecyl sulfate (SDS) and an aliquot of each lysate was used for the scintillation count in a LS 6500 Beta-Counter (Beckman- Coulter, CA). Non-specific uptake in the presence of 20μΜ citocalasine B and 200μΜ phloretin was subtracted from each experimental value.
AutoADP-ribosylation of SIRT6 assay
Reactions were performed in a total volume of 10 μΐ, containing 20 pg of recombinant SIRT6 (produced following published protocols13) in 50mM Tris-HCl, pH 8.0, 150mM NaCl, lOmM dithiothreitol, and 0.4 pCi [3H]NAD (40 Ci mmol"1; Perkin Elmer, Boston, MA), in the presence or absence of different compounds (200 μΜ final concentration) or nicotinamide (lOOmM final concentration) for 45 minutes at 22°C. At the end of incubation, samples were filtered on a nitrocellulose membrane (Bio-Rad), filters were washed twice with 3 ml of washing buffer (50mM Tris-HCl, pH 8.0, 150mM NaCl) and then dried, and radioactivity was measured in 3.0 ml of Ultima-Gold on a Packard β-counter.
The following table summarizes experimental data obtained subjecting some compounds according to the present invention to the tests described above.
Figure imgf000013_0001
* = percentage of SIRT6 inhibition > 10% and < 30%;
** = percentage of SIRT6 inhibition > 30% and < 70% *** = percentage of SIRT6 inhibition > 70%.
The data reported above demonstrate that the compounds according to the present invention display inhibitory activity against sirtuins SIRTl, SIRT2 and SIRT6, with good selectivity towards SIRT6, as well as inhibitory activity against the production of TNF-alpha by BxPC3 cells; ability to increase glucose uptake in BxPC3 cells; (limited) ability to reduce autoADP-ribosylation of SIRT6; and ability to reduce viability of BxPC3 cells in the presence of a low concentration (InM) of gemcitabine, which would be inactive as such in the absence of SIRT6 inhibitors (% of viability inhibition by InM gemcitabine alone = 4%). Literature references:
1. Finkel, T.; Deng, C. X.; Mostoslavsky, R., Recent progress in the biology and physiology of sirtuins. Nature 2009, 460 (7255), 587-91.
2. Feldman, J. L.; Dittenhafer-Reed, K. ed.; Denu, J. M., Sirtuin Catalysis and Regulation. J Biol Chem 2012. 3. Bruzzone, S.; Parenti, M. D.; Grozio, A.; Ballestrero, A.; Bauer,
I.; of the Rio, A.; Nencioni, A., Rejuvenating sirtuins: the rise of a new family of cancer drug targets. Current pharmaceutical design 2012.
4. Mostoslavsky, R.; Chua, K. F.; Lombard, D. B.; Pang, W. W.; Fischer, M. R.; Gellon, L.; Liu, P.; Mostoslavsky, G.; Franco, S.; Murphy, M. M.; Mills, K. D.; Patel, P.; Hsu, J. T.; Hong, A. L.; Ford, ed.; Cheng, H. L.; Kennedy, C; Nunez, N.; Bronson, R.; Frendewey, D.; Auerbach, W.; Valenzuela, D.; Karow, M.; Hottiger, M. O.; Hursting, S.; Barrett, J. C;
Guarente, L.; Mulligan, R.; Demple, B.; Yancopoulos, G. D.; Alt, F. W., Genomic instability and aging-like phenotype in the absence of mammalian SIRT6. Cell 2006, 124 (2), 315-29. 5. Michishita, ed.; McCord, R. A.; Berber, ed.; Kioi, M.; Padilla-
Nash, H.; Damian, M.; Cheung, P.; Kusumoto, R.; Kawahara, T. L.; Barrett, J. C; Chang, H. Y.; Bohr, V. A.; Ried, T.; Gozani, O.; Chua, K. F., SIRT6 is a histone H3 lysine 9 deacetylase that modulates telomeric chromatin. Nature 2008, 452 (7186), 492-6. 6. McCord, R. A.; Michishita, ed.; Hong, T.; Berber, ed.; Boxer, L.
D.; Kusumoto, R.; Guan, S.; Shi, X.; Gozani, O.; Burlingame, A. L.; Bohr, V. A.; Chua, K. F., SIRT6 stabilizes DNAdependent protein kinase at chromatin for DNA double- strand break repair. Aging (Albany, NY) 2009, 1 (1), 109-21. 7. Mao, Z.; Hine, C; Tian, X.; Van Meter, M.; Au, M.; Vaidya, A.;
Seluanov, A.; Gorbunova, V., SIRT6 promotes DNA repair under stress by activating PARP1. Science 201 1 , 332 (6036), 1443-6.
8. Zhong, L.; DUrso, A.; Toiber, D.; Sebastian, C; Henry, R. ed.; Vadysirisack, D. D.; Guimaraes, A.; Marinelli, B.; Wikstrom, J. D.; Nir, T.; Clish, C. B.; Vai thee s varan, B.; Iliopoulos, O.; Kurland, I.; Dor, Y.; Weissleder, R.; Shirihai, O. S.; Ellisen, L. W.; Espinosa, J. M.; Mostoslavsky, R., The histone deacetylase Sirt6 regulates glucose homeostasis via Hifl alpha. Cell 2010, 140 (2), 280-93.
9. Van Gool, F.; Galli, M.; Gueydan, C; Kruys, V.; Prevot, P. P.; Bedalov, A.; Mostoslavsky, R.; Alt, F. W.; De Smedt, T.; Leo, O.,
Intracellular NAD levels regulate tumor necrosis factor protein synthesis in a sirtuin-dependent manner. Nat Med 2009, 15 (2), 206- 10. 10. Bruzzone, S.; Fruscione, F.; Morando, S.; Ferrando, T.; Poggi, A.; Garuti, A.; DTJrso, A.; Selmo, M.; Benvenuto, F.; Cea, M.; Zoppoli, G.; Moran, ed.; Soncini, D.; Ballestrero, A.; Sordat, B.; Patrone, F.; Mostoslavsky, R.; birds, A.; Nencioni, A., Catastrophic NAD+ depletion in activated T lymphocytes through Nampt inhibition reduces demyelination and disability in EAE. PLoS One 2009, 4 (11), e7897.
11. Montecucco, F.; Bauer, I.; Braunersreuther, V.; Bruzzone, S.; Akhmedov, A.; Luscher, T. F.; Speer, T.; Poggi, A.; Mannino, ed.; Pelli, G.; Galan, K.; Bertolotto, M.; Lenglet, S.; Garuti, A.; Montessuit, C; Lerch, R.; Pellieux, C; Vuilleumier, N.; Dallegri, F.; Mage, J.;
Sebastian, C; Mostoslavsky, R.; Gayet-Ageron, A.; Patrone, F.; Mach, F.; Nencioni, A., Inhibition of Nicotinamide Phosphoribosyltransferase Reduces Neutrophil-Mediated Injury in Myocardial Infarction. An tioxid Redox Signal 2012. 12. Bauer, I.; Grozio, A.; Lasiglie, D.; Basile, G.; Sturla, L.;
Magnone, M.; Sociali, G.; Soncini, D.; Caffa, I.; Poggi, A.; Zoppoli, G.; Cea, M.; Feldmann, G.; Mostoslavsky, R.; Ballestrero, A.; Patrone, F.; Bruzzone, S.; Nencioni, A., The NAD+-dependent histone deacetylase SIRT6 promotes cytokine production and migration in pancreatic cancer cells by regulating Ca2+ responses. J Biol Chem 2012.
13. Pan, P. W.; Feldman, J. L.; Devries, M. K.; Dong, A.; Edwards, A. M.; Denu, J. M., Structure and biochemical functions of SIRT6. J Biol Chem 2011 , 286 (16), 14575-87.

Claims

1. A compound of general formula (I)
Figure imgf000016_0001
in which:
R1 = H, Ci-Csalkyl; R2 = H, Ci-Csalkyl;
R3 = optionally substituted phenyl; optionally substituted phenoxy; -CH2-(CH2)n-phenyl optionally substituted on the benzene ring, where n = 0, 1 or 2; or -Y-R5, where Y = a single bond, -O, -CH2, and R5 = 5- or 6-membered, saturated or aromatic heterocyclic ring containing one or more heteroatoms selected from N, S and O, optionally substituted with a Ci-Csalkyl;
R4 = H, C1-6alkyl, C3-6cycloalkyl, halogen, -NO2, -CN, -OH, -SH, - CF3, -CCI3, -COOH, -COOC1-5alkyl, -SO2OC1-5alkyl, -OC1-5alkyl, or enantiomers, diastereomers and pharmaceutically acceptable salts thereof, for use as a medicament.
2. A compound according to claim 1, in which
R1 = H, CH3
R2 = H, CH3 R3 = phenyl, phenoxy, benzyl or phenylethyl optionally substituted with a Ci-3alkyl on the aromatic ring or Y-R5, where Y = a single bond, -O-, -CH2-, and
R5 = 5- or 6-membered heteroaromatic ring, containing 1 or 2 N atoms, optionally substituted with a Ci-Csalkyl;
R4 = H or enantiomers, diastereomers and pharmaceutically acceptable salts thereof, for use as a medicament.
3. A compound according to claim 2, in which R1 = H, CH3
R2 = H, CH3 R3 = phenyl, benzyl, phenylethyl or Y-R5, where Y = a single bond, -O-, -CH2-, and R5 = pyridinyl or imidazolyl; R4 = H or enantiomers, diastereomers and pharmaceutically acceptable salts thereof, for use as a medicament.
4. A compound according to claim 3, having formula:
2,4-dioxo-N-[4-(pyridin-3-yloxy)phenyl]- l,2,3,4- tetrahydroquinazoline-6-sulfonamide;
N-[4-(lH-imidazol-l-ylmethyl)phenyl]-2,4-dioxo- l,2,3,4- tetrahydroquinazoline-6-sulfonamide;
2,4-dioxo-N-{3-[2-(pyridin-4-yl)ethyl]phenyl}- 1,2,3,4- tetrahydroquinazoline-6-sulfonamide;
N-(2-benzylphenyl)-2,4-dioxo- 1 ,2,3,4-tetrahydroquinazoline-6- sulfonamide; 2,4-dioxo-N-(2-phenylphenyl)- l ,2,3,4-tetrahydroquinazoline-6- sulfonamide; or 1 , 3 -dimethyl-2 , 4-dioxo- N- [4 - (pyridin-4-ylmethyl) phenyl] - 1,2,3,4- tetrahydroquinazoline-6-sulfonamide, for use as a medicament.
5. A compound according to any one of claims 1 to 4, for use as an inhibitor of one or more sirtuins to increase glucose uptake in a subject.
6. A compound according to claim 5, wherein said compound inhibits sirtuin SIRT6.
7. A compound according to any one of claims 5 and 6, for use in the treatment of: type I and type II diabetes mellitus and complications thereof, such as ketoacidotic coma, hyperglycaemic hyperosmolar condition, atherosclerosis, ischaemic heart disease (angina and myocardial infarction), stroke, peripheral vasculopathy, diabetic retinopathy, diabetic nephropahty, diabetic neuropathy, diabetic foot.
8. A compound according to any one of claims 1 to 4 for use as an inhibitor of one or more sirtuins to reduce the production of TNF-alpha or other inflammatory, chemotactic, or proangiogenic cytokines in a subject and/ or to reduce local or systemic inflammation in a subject and/or to treat a pathology mediated by TNF-alpha or other inflammatory, chemotactic, or proangiogenic cytokines in a subject having local or systemic excessive or dysregulated production of TNF-alpha or other inflammatory, chemotactic, or proangiogenic cytokines.
9. A compound according to claim 8, wherein said compound inhibits sirtuin SIRT6.
10. A compound according to any one of claims 8 and 9, for use in the treatment of inflammatory diseases comprising rheumatoid arthritis, multiple sclerosis, inflammatory bowel disease, systemic lupus erythematosus, vasculitis, Goodpasture syndrome, scleroderma, atherosclerosis, graft versus host disease (GVHD), organ transplant rejection, myocardial infarction, stroke, reperfusion injury after revascularization in heart and other organs.
11. A compound according to any one of claims 1 to 4 for use as an inhibitor of one or more sirtuins to interfere with DNA repair in tumor cells and thus exert an anticancer effect and sensitize said cells to antineoplastic agents and radiotherapy.
12. A compound according to claim 1 1 , wherein said compound inhibits sirtuin SIRT6.
13. A compound according to any one of claims 11 and 12, for use in the treatment of neoplastic diseases comprising pancreatic cancer, breast cancer, colorectal cancer, prostatic cancer, ovarian cancer, melanoma, lung cancer, oesophageal cancer, hepatic carcinoma, lymphomas, leukemias, myeloma, sarcomas, neoplastic cachexia.
14. A pharmaceutical composition containing a therapeutically effective amount of at least one compound according to any one of claims 1 to 4 and a pharmaceutically acceptable vehicle.
15. A method for reducing the production of TNF-alpha or other inflammatory, chemotactic, or proangiogenic cytokines in cells in vitro, comprising the step of exposing said cells to a compound according to any one of claims 1 to 4.
16. A method for increasing glucose uptake in cells in vitro, comprising the step of exposing said cells to a compound according to any one of claims 1 to 4.
17. A method for increasing the antiproliferative and cytotoxic effect of antineoplastic drugs and ionizing radiations in vitro, comprising the step of exposing said tumor cells to a compound according to any one of claims 1 to 4.
PCT/IB2014/060829 2013-04-19 2014-04-18 Quinazolinedione componds with a sirtuin inhibiting activity WO2014170875A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
ITMI2013A000646 2013-04-19
IT000646A ITMI20130646A1 (en) 2013-04-19 2013-04-19 CHINAZOLINDIONIC COMPOUNDS WITH INHABITING ACTIVITIES ON SIRTUINES

Publications (2)

Publication Number Publication Date
WO2014170875A1 true WO2014170875A1 (en) 2014-10-23
WO2014170875A8 WO2014170875A8 (en) 2015-01-08

Family

ID=48579265

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IB2014/060829 WO2014170875A1 (en) 2013-04-19 2014-04-18 Quinazolinedione componds with a sirtuin inhibiting activity

Country Status (2)

Country Link
IT (1) ITMI20130646A1 (en)
WO (1) WO2014170875A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11497740B2 (en) 2016-04-29 2022-11-15 The Board Of Regents Of The University Of Texas System Use of Jumonji C demethylase inhibitors for the treatment and prevention of chemotherapy resistance and radioresistance in cancer

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002064572A1 (en) * 2001-02-14 2002-08-22 Warner-Lambert Company Llc Quinazolines as mmp-13 inhibitors
WO2008138943A2 (en) 2007-05-14 2008-11-20 Universite Libre De Bruxelles Prophylactic and therapeutic use of sirtuin inhibitors in tnf-alpha mediated pathologies
WO2011038110A2 (en) 2009-09-23 2011-03-31 The General Hospital Corporation Methods of treating metabolic disease
WO2011109441A1 (en) * 2010-03-01 2011-09-09 Myrexis, Inc. Compounds and therapeutic uses thereof

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8076491B2 (en) * 2007-08-21 2011-12-13 Senomyx, Inc. Compounds that inhibit (block) bitter taste in composition and use thereof
FR2944282B1 (en) * 2009-04-09 2013-05-03 Sanofi Aventis QUINAZOLINEDIONE DERIVATIVES, THEIR PREPARATION AND THEIR VARIOUS THERAPEUTIC APPLICATIONS

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002064572A1 (en) * 2001-02-14 2002-08-22 Warner-Lambert Company Llc Quinazolines as mmp-13 inhibitors
WO2008138943A2 (en) 2007-05-14 2008-11-20 Universite Libre De Bruxelles Prophylactic and therapeutic use of sirtuin inhibitors in tnf-alpha mediated pathologies
WO2011038110A2 (en) 2009-09-23 2011-03-31 The General Hospital Corporation Methods of treating metabolic disease
WO2011109441A1 (en) * 2010-03-01 2011-09-09 Myrexis, Inc. Compounds and therapeutic uses thereof

Non-Patent Citations (19)

* Cited by examiner, † Cited by third party
Title
"Remington's Pharmaceutical Sciences Handbook", 1990, MACK PUBLISHING
BAUER I ET AL., J BIOL CHEM., 18 October 2012 (2012-10-18)
BAUER I. ET AL., J. BIOL. CHEM., 18 October 2012 (2012-10-18)
BAUER, I.; GROZIO, A.; LASIGLIE, D.; BASILE, G.; STURLA, L.; MAGNONE, M.; SOCIALI, G.; SONCINI, D.; CAFFA, I.; POGGI, A.: "The NAD+-dependent histone deacetylase SIRT6 promotes cytokine production and migration in pancreatic cancer cells by regulating Ca2+ responses", J BIOL CHEM, 2012
BRUZZONE, S.; FRUSCIONE, F.; MORANDO, S.; FERRANDO, T; POGGI, A.; GARUTI, A.; D'URSO, A.; SELMO, M.; BENVENUTO, F.; CEA, M.: "Catastrophic NAD+ depletion in activated T lymphocytes through Nampt inhibition reduces demyelination and disability in EAE", PLOS ONE, vol. 4, no. 11, 2009, pages E7897
BRUZZONE, S.; PARENTI, M. D.; GROZIO, A.; BALLESTRERO, A.; BAUER, I.; RIO, A.; NENCIONI, A.: "Rejuvenating sirtuins: the rise of a new family of cancer drug targets", CURRENT PHARMACEUTICAL DESIGN, 2012
FELDMAN, J. L.; DITTENHAFER-REED, K. ED.; DENU, J. M.: "Sirtuin Catalysis and Regulation", J BIOL CHEM, 2012
FINKEL, T.; DENG, C. X.; MOSTOSLAVSKY, R.: "Recent progress in the biology and physiology of sirtuins", NATURE, vol. 460, no. 7255, 2009, pages 587 - 91
GUARENTE, L; MULLIGAN, R; DEMPLE, B.; YANCOPOULOS, G. D; ALT, F. W: "Genomic instability and aging-like phenotype in the absence of mammalian SIRT6", CELL, vol. 124, no. 2, 2006, pages 315 - 29
KURYAZOV R. SH. ET AL.: "Quinazolines 1. Synthesis and chemical reactions of 6-chlorosulfonyl-quinazoline-2,4-diones", CHEMISTRY OF HETEROCYCLIC COMPOUNDS, vol. 44, no. 3, 2008
MAO, Z.; HINE, C.; TIAN, X.; VAN METER, M.; AU, M.; VAIDYA, A.; SELUANOV, A.; GORBUNOVA, V.: "SIRT6 promotes DNA repair under stress by activating PARP1", SCIENCE, vol. 332, no. 6036, 2011, pages 1443 - 6
MARCO DANIELE PARENTI ET AL: "Discovery of Novel and Selective SIRT6 Inhibitors", JOURNAL OF MEDICINAL CHEMISTRY, vol. 57, no. 11, 12 June 2014 (2014-06-12), pages 4796 - 4804, XP055124815, ISSN: 0022-2623, DOI: 10.1021/jm500487d *
MCCORD, R. A; MICHISHITA, ED.; HONG, T.; BERBER, ED.; BOXER, L. D.; KUSUMOTO, R.; GUAN, S.; SHI, X.; GOZANI, O.; BURLINGAME, A. L.: "SIRT6 stabilizes DNAdependent protein kinase at chromatin for DNA double-strand break repair", AGING (ALBANY, NY, vol. 1, no. 1, 2009, pages 109 - 21
MICHISHITA, ED.; MCCORD, R. A.; BERBER, ED.; KIOI, M.; PADILLA-NASH, H.; DAMIAN, M.; CHEUNG, P.; KUSUMOTO, R.; KAWAHARA, T. L.; BA: "SIRT6 is a histone H3 lysine 9 deacetylase that modulates telomeric chromatin", NATURE, vol. 452, no. 7186, 2008, pages 492 - 6
PAN, P. W.; FELDMAN, J. L.; DEVRIES, M. K.; DONG, A.; EDWARDS, A. M.;; DENU, J. M.: "Structure and biochemical functions of SIRT6", J BIOL CHEM, vol. 286, no. 16, 2011, pages 14575 - 87
SANTINA BRUZZONE ET AL: "Rejuvenating Sirtuins: The Rise of a New Family of Cancer Drug Targets", CURRENT PHARMACEUTICAL DESIGN, 2013, 19, 1 February 2013 (2013-02-01), pages 614 - 623, XP055124678, Retrieved from the Internet <URL:http://docserver.ingentaconnect.com/deliver/connect/ben/13816128/v19n4/s4.pdf?expires=1403519381&id=78487964&titleid=3901&accname=Europaeisches Patentamt&checksum=F4EB9729DF46EB7CBD664F1337218163> [retrieved on 20140623] *
SEBASTIAN, C; MOSTOSLAVSKY, R; GAYET-AGERON, A; PATRONE, F; MACH, F; NENCIONI, A: "Inhibition of Nicotinamide Phosphoribosyltransferase Reduces Neutrophil-Mediated Injury in Myocardial Infarction", ANTIOXID REDOX SIGNAL, 2012
VAN GOOL, F.; GALLI, M.; GUEYDAN, C.; KRUYS, V.; PREVOT, P. P.; BEDALOV, A.; MOSTOSLAVSKY, R.; ALT, F. W.; DE SMEDT, T.; LEO, O.: "Intracellular NAD levels regulate tumor necrosis factor protein synthesis in a sirtuin-dependent manner", NAT MED, vol. 15, no. 2, 2009, pages 206 - 10
ZHONG, L.; D'URSO, A.; TOIBER, D.; SEBASTIAN, C.; HENRY, R. ED.; VADYSIRISACK, D. D.; GUIMARAES, A.; MARINELLI, B.; WIKSTROM, J. D: "The histone deacetylase Sirt6 regulates glucose homeostasis via Hiflalpha", CELL, vol. 140, no. 2, 2010, pages 280 - 93

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11497740B2 (en) 2016-04-29 2022-11-15 The Board Of Regents Of The University Of Texas System Use of Jumonji C demethylase inhibitors for the treatment and prevention of chemotherapy resistance and radioresistance in cancer

Also Published As

Publication number Publication date
ITMI20130646A1 (en) 2014-10-20
WO2014170875A8 (en) 2015-01-08

Similar Documents

Publication Publication Date Title
WO2014170873A1 (en) Compounds with a sirtuin inhibiting activity
RU2497809C2 (en) Naphthalene carboxamide derivatives as protein kinase and histone deacetylase inhibitors, methods for preparing and using them
US9636333B2 (en) Methyl/fluoro-pyridinyl-methoxy substituted pyridinone-pyridinyl compounds and fluoro-pyrimidinyl-methoxy substituted pyridinone-pyridinyl compounds
RU2632097C2 (en) TREATMENT OF MALIGNANT AND NON-MALIGNANT DISEASES WITH Ras ANTAGONISTS
CN107759564B (en) Triazole pyridine formyl glycine compound, method and medical application thereof
EA024703B1 (en) Combination of olanzapine and taar1 agonist
BRPI0808714A2 (en) USE OF COMPOUNDS, SUCH AS 4-AMINO-5-FLUORO-3- [6- (4-METHYL-PIPERAZINYL-1) -1H-BENZIMIDAZOLYL-2] QUINOLINONE- (1H) -2 AND TAUTOMERS, SALTS, AND MIXTURES OF THE SAME PREPARING MEDICINAL PRODUCTS FOR MELANOMA TREATMENT
US8841288B2 (en) Quinolin-4 (1H)-one derivatives as inhibitors of phosphatidylinositol 3-kinases
JP2019510744A (en) Substituted oxadiazole compounds, compositions containing the compounds and applications thereof
US11186596B2 (en) Anticancer indenes, indanes, azaindenes, azaindanes, pharmaceutical compositions and uses
ES2927959T3 (en) Heterocyclic imidazole compounds, pharmaceutical compositions thereof, method of preparation thereof and use thereof
US9359300B2 (en) Methyl/difluorophenyl-methoxy substituted pyridinone-pyridinyl compounds, methyl-pyridinyl-methoxy substituted pyridinone-pyridinyl compounds, and methyl-pyrimidinyl-methoxy substituted pyridinone-pyridinyl compounds
WO2014170875A1 (en) Quinazolinedione componds with a sirtuin inhibiting activity
CN101397295B (en) 2-dihydroindolemanone derivates as histone deacetylase inhibitor, preparation method and use thereof
KR20080044277A (en) Combination of organic compounds
KR20210015892A (en) Compound for the treatment of pain, composition comprising the same, and method of using the same
US9656984B2 (en) PI3K/AKT/mTOR inhibitors and pharmaceutical uses thereof
WO2018211277A1 (en) Ldha activity inhibitors
US20230348401A1 (en) Inhibitors of Glucose-6-phosphate Dehydrogenase and Uses Thereof
US20190151289A1 (en) Identification of Small Molecule Inhibitors of Jumonji AT-Rich Interactive Domain 1A (JARID1A) Histone Demethylase
US20210169850A1 (en) Indoline derivatives, compositions comprising them and uses thereof
JP4604147B2 (en) Coumarin derivative
KR20200030466A (en) 7-hydroxy-4h-thieno[3,2-b]pyridin-5-one derivatives and methods of use of thereof
CN111943911B (en) Acrylamidobenzazepine derivatives as GSK-3 beta inhibitors and uses thereof
US10800776B2 (en) Fluorine-containing triazolopyridine, and manufacturing method, pharmaceutical composition, and application thereof

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: 14731008

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: 14731008

Country of ref document: EP

Kind code of ref document: A1