WO2016135139A1 - Dérivés de 2,3-dihydrocyclopenta[b]quinoléine en tant qu'inhibiteurs de mth1 pour la thérapie du cancer - Google Patents

Dérivés de 2,3-dihydrocyclopenta[b]quinoléine en tant qu'inhibiteurs de mth1 pour la thérapie du cancer Download PDF

Info

Publication number
WO2016135139A1
WO2016135139A1 PCT/EP2016/053767 EP2016053767W WO2016135139A1 WO 2016135139 A1 WO2016135139 A1 WO 2016135139A1 EP 2016053767 W EP2016053767 W EP 2016053767W WO 2016135139 A1 WO2016135139 A1 WO 2016135139A1
Authority
WO
WIPO (PCT)
Prior art keywords
alkylene
alkyl
compound
cancer
chloro
Prior art date
Application number
PCT/EP2016/053767
Other languages
English (en)
Inventor
Kilian Huber
Original Assignee
Cemm - Forschungszentrum Für Molekulare Medizin Gmbh
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 Cemm - Forschungszentrum Für Molekulare Medizin Gmbh filed Critical Cemm - Forschungszentrum Für Molekulare Medizin Gmbh
Publication of WO2016135139A1 publication Critical patent/WO2016135139A1/fr

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
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/473Quinolines; Isoquinolines ortho- or peri-condensed with carbocyclic ring systems, e.g. acridines, phenanthridines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents

Definitions

  • the present invention relates to 2,3-dihydrocyclopenta[b]quinoline derivatives of formula (I) for use as medicaments as well as pharmaceutical compositions comprising these compounds, particularly for use as inhibitors of MTH1 and for use in the treatment or prevention of cancer.
  • Cancer is a leading cause of death in both economically developed countries as well as in developing countries (Mathers C et al., The global burden of disease: 2004 update, World Health Organization, 2008; Jemal A et al., CA Cancer J Clin, 201 1 , 61 (2):69-90), and there is an ongoing and urgent need for novel therapeutic agents against cancer.
  • MTH1 The enzyme human MutT homolog 1 (MTH1 ) is a 2-hydroxy-dATP diphosphatase (EC 3.6.1 .56) which sanitizes oxidized dNTP pools to prevent incorporation of damaged nucleotides during DNA replication and has been described as a target for cancer therapy (Huber KV et al., Nature, 2014, 508(7495):222-7; Gad H et al., Nature, 2014, 508(7495):215- 21 ; Cancer Discovery, 2014, 4:631 , doi: 10.1 158/2159-8290.CD-RW2014-085). Since MTH1 is generally required for the survival of a variety of different cancers, regardless of their specific genetic changes, the inhibition of MTH1 provides an effective and versatile approach for the therapy of cancer, which does not rely on the targeting of specific genetic defects.
  • MTH1 inhibitors include the compound SCH 51344, the (S)-enantiomer of crizotinib and related derivatives (Huber KV et al., Nature, 2014, 508(7495):222-7; WO 2014/033136) as well as the compounds ai iu ⁇ ⁇ di iu ⁇ ⁇ i U i i -U lai l i t 16 UouVail eS
  • inhibitors of MTH1 which should be effective in the treatment of a wide range of different cancers having different genetic defects.
  • the 2,3-dihydro- cyclopenta[b]quinoline derivatives of formula (I) as described and defined in the following are potent inhibitors of MTH1 and can thus advantageously be used for the treatment or prevention of cancer.
  • the compounds of formula (I) according to the invention are particularly effective inhibitors of MTH1 due to the presence of their specific substituent R 1 in position 7 of the 2,3-dihydro-cyclopenta[b]quinoline scaffold.
  • compound (1 ) according to the invention has been found to exhibit a considerably stronger affinity to MTH1 than a corresponding reference compound (9) lacking the specific substituent R 1 in position 7, as reflected by a shift in the melting temperature (Tm) of MTH1 effected by compound (1)
  • DSF differential scanning fluorimetry
  • R 1 is halogen, methoxy, ethoxy, or -CF 3 .
  • Each R 2 is independently selected from C1.5 alkyl, C2.5 alkenyl, C 2 - 5 alkynyl, -(C 0 -3 alkylene)-OH, -(Co-3 alkylene)-0(Ci_ 5 alkyl), -(C c . 3 alkylene)-0(C -5 alkylene)-OH, -(C 0 _ 3 alkylene)-0(Ci_ 5 alkylene)-0(C 1-5 alkyl), -(C 0 - 3 alkylene)-SH, -(C 0 - 3 alkylene)-S(C 1 _ 5 alkyl), -(C 0-3 alkylene)-S(C 1 .
  • Each R 3 is independently selected from d. 5 alkyl, C 2 . 5 alkenyl, C 2 . 5 alkynyl, -(C 0 . 3 alkyiene)-OH, -(Co-3 alkylene)-0(C 1 - 5 alkyl), -(C 0 - 3 alkylene)-0(C 5 alkylene)-OH, -(C c 3 alkylene)-0(C; 5 alkylene)-0(C 1 -5 alkyl), -(C 0 . 3 a!kylene)-SH, -(C 0 . 3 alkylene)-S(C 1 5 alkyl), -(C 0 .
  • R 4 is hydrogen or d. 5 alkyl.
  • R 5 is a covending bond or C 1-3 alkyiene.
  • the group A is aryl or heteroaryl, wherein said aryl and said heteroaryl are each optionally substituted with one or more groups R 6 .
  • Each R 6 is independently selected from d. 5 alkyl, C 2 . 5 alkenyl, C 2 . 5 alkynyl, -(C 0 . 3 alkylene)-OH, -(Co-3 alkylene)-0(Ci. 5 alkyl), -(C 0 . 3 alkylene)-0(Ci. 5 alkylene)-OH, -(C 0 - 3 alky!ene)-0(C . 5 alkylene)-0(Ci. 5 alkyl), -(C 0 .
  • the present invention also provides a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of formula (I), as described and defined herein, or a pharmaceutically acceptable salt, solvate or prodrug thereof, in combination with a pharmaceutically acceptable excipient, for use in the treatment or prevention of cancer.
  • the present invention further relates to a compound of formula (I), as described and defined herein, or a pharmaceutically acceptable salt, solvate or prodrug thereof, wherein (i) R 1 is -F, -CI, -I, methoxy, ethoxy, or -CF 3 , or wherein (ii) R 5 is a covalent bond or C 2 . 3 alkylene, for use as a medicament.
  • the invention relates to a compound of formula (I), as described and defined herein, or a pharmaceutically acceptable salt, solvate or prodrug thereof, wherein R 1 is -F, -CI, -I, methoxy, ethoxy, or -CF 3 , for use as a medicament.
  • the invention also relates to a compound of formula (I), as described and defined herein, or a pharmaceutically acceptable salt, solvate or prodrug thereof, wherein R 5 is a covalent bond or C 2 .3 alkylene, for use as a medicament.
  • the invention refers to a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of formula (I) or a pharmaceutically acceptable salt, solvate or prodrug thereof, wherein R 1 is -F, -CI, -I, methoxy, ethoxy, or -CF 3 , in combination with a pharmaceutically acceptable excipient.
  • the invention likewise provides a pharmaceutical composition comprising a compound of formula (I) or a pharmaceutically acceptable salt, solvate or prodrug thereof, wherein R 5 is a covalent bond or C 2 . 3 alkylene, in combination with a pharmaceutically acceptable excipient.
  • the present invention relates to the use of a compound of formula (I) or a pharmaceutically acceptable salt, solvate or prodrug thereof, wherein R 1 is -F, -CI, -I, methoxy, ethoxy, or -CF 3 , in the preparation of a medicament, particularly for the treatment or prevention of a disease/disorder such as, e.g., cancer.
  • the invention also relates to the use of a compound of formula (I) or a pharmaceutically acceptable salt, solvate or prodrug thereof, wherein R 5 is a covalent bond or C 2 . 3 alkylene, in the preparation of a medicament, particularly for the treatment or prevention of a disease/disorder such as, e.g., cancer.
  • MTH1 is regarded as the major clearance enzyme for oxidized nucleotides such as 2-OH-dATP and 8-oxo-dGTP (Nakabeppu, Mutation Research/Genetic Toxicology and Environmental Mutagenesis, 2010, 703(1 ):51 -8).
  • Oxidized nucleotides which are generated by attack of reactive oxygen species (ROS) on DNA or the nucleotide pool, can cause DNA damage and mutations.
  • ROS reactive oxygen species
  • Sources of ROS include mitochondrial respiration, chemicals or radiation.
  • the frequently occurring 8-oxo-guanine (8-oxo-G) can lead to transversion mutations during replication.
  • 8-oxo-G In contrast to unoxidized guanine, 8-oxo-G is able to pair with either cytosine or adenine with almost equal efficiency, thereby generating mutations if 8-oxo-G is inserted opposite A in a nascent DNA strand or vice versa. If the misincorporated oxidized nucleotide is recognized by the DNA repair system, the lesion can be repaired by base excision repair (BER). BER involves induction of a temporary single strand break to remove the falsely inserted base and subsequent replacement. However, high amounts of 8-oxo-G can lead to accumulation of single strand breaks which eventually progress to double strand breaks (DSB), thus inducing cell cycle arrest (quiescence or senescence) and apoptosis.
  • DSB double strand breaks
  • MTH1 By converting the oxidized nucleoside triphosphates into the corresponding nucleoside monophosphates which can no longer be used as substrates by DNA polymerases, MTH1 prevents the integration of oxidized bases into DNA and thus prevents mutations and oxidative DNA damage induced by ROS. Transformation of cells by oncogenes such as mutant RAS which occurs in about 20% of all tumors can also lead to increased production of ROS (Rai, Oncogene, 201 1 , 30(12): 1489-96). As for normal cells, oxidative damage caused by ROS can force cancer cells into a state of quiescence or senescence (OIS), and eventually apoptosis.
  • OIS quiescence or senescence
  • RAS-transformed cells upregulate MTH1 which protects the cells from oxidative DNA damage.
  • MTH1 MTH1
  • HRAS human skin fibroblasts transfected with HRAS undergo senescence, but this phenotype can be rescued by concomitant overexpression of MTH1 (Rai, Oncogene, 201 1 , 30(12):1489-96). Reports consequently indicate that MTH1 suppression causes proliferative defects in cancer cells expressing mutant RAS (Rai, Mutation Research/Genetic Toxicology and Environmental Mutagenesis, 2010, 703(1 ):71 -81).
  • MTH1 -/- knockout mice show a very mild phenotype (Tsuzuki, Proceedings of the National Academy of Sciences, 2001 , 98(20): 1 1456- 61 )
  • targeting MTH1 with small molecules is considered to provide an effective and well-tolerated therapeutic approach for the treatment or prevention of cancer, including also cancer having an activating RAS mutation (i.e., RAS-dependent cancer).
  • the compounds of formula (I) have surprisingly been found to be potent inhibitors of MTH1 , as also demonstrated in the appended examples, in which the effect of a range of exemplary compounds of formula (i) on MTH1 was confirmed in an in vitro binding assay using differential scanning fluorimetry.
  • MTH1 has recently been identified as a critical factor for cancer cell survival in vitro and in vivo in a wide range of different cancers (Huber KV et al., Nature, 2014, 508(7495):222-7; Gad H et al., Nature, 2014, 508(7495):215-21 ; Cancer Discovery, 2014, 4:631 , doi: 10.1 158/2159-8290.
  • the present invention thus particularly relates to a compound of formula (I), as described and defined herein, or a pharmaceutically acceptable salt, solvate or prodrug thereof, for use in the treatment or prevention of cancer.
  • the invention also provides a pharmaceutical composition comprising a compound of formula (I) or a pharmaceutically acceptable salt, solvate or prodrug thereof, in combination with a pharmaceutically acceptable excipient, for use in the treatment or prevention of cancer.
  • the present invention furthermore relates to the use of a compound of formula (I) or a pharmaceutically acceptable salt, solvate or prodrug thereof in the preparation of a medicament for the treatment or prevention of cancer.
  • the invention likewise provides a method of treating or preventing cancer, the method comprising administering a compound of formula (I) or a pharmaceutically acceptable salt, solvate or prodrug thereof, or a pharmaceutical composition comprising any of the aforementioned entities and a pharmaceutically acceptable excipient, to a subject (e.g., a human) in need thereof.
  • a subject e.g., a human
  • the present invention relates to a compound of formula (I) or a pharmaceutically acceptable salt, solvate or prodrug thereof, or a pharmaceutical composition comprising any of the aforementioned entities and a pharmaceutically acceptable excipient, for use in inhibiting MTH1 or for use in treating or preventing cancer by inhibiting MTH1.
  • the invention further refers to the use of a compound of formula (I) or a pharmaceutically acceptable salt, solvate or prodrug thereof in the preparation of a medicament for inhibiting MTH1 or for treating or preventing cancer by inhibiting MTH1 .
  • the invention provides a method of inhibiting MTH1 in a subject, the method comprising administering a compound of formula (I) or a pharmaceutically acceptable salt, solvate or prodrug thereof, or a pharmaceutical composition comprising any of the aforementioned entities and a pharmaceutically acceptable excipient, to a subject (e.g., a human) in need thereof.
  • the invention also provides a method of treating or preventing cancer by inhibiting MTH1 , the method comprising administering a compound of formula (I) or a pharmaceutically acceptable salt, solvate or prodrug thereof, or a pharmaceutical composition comprising any of the aforementioned entities and a pharmaceutically acceptable excipient, to a subject (e.g. , a human) in need thereof.
  • a subject e.g. , a human
  • the cancer to be treated or prevented in accordance with the present invention is preferably selected from gastrointestinal cancer, colorectal cancer, liver cancer (e.g. , hepatocellular carcinoma), pancreatic cancer, stomach cancer, genitourinary cancer, bladder cancer, esophageal cancer, prostate cancer (e.g., hormone-refractory prostate cancer), lung cancer (e.g., small cell lung cancer or non-small cell lung cancer), breast cancer (e.g., triple-negative breast cancer, or breast cancer having a BRCA1 and/or BRCA2 gene mutation), hematological cancer, leukemia (e.g., acute lymphoblastic leukemia, acute myeloid leukemia, chronic lymphocytic leukemia, or chronic myeloid leukemia), lymphoma (e.g., Hodgkin lymphoma or non-Hodgkin lymphoma, such as, e.g., follicular lymphoma or diffuse large B-cell lymphoma), ovarian cancer,
  • the cancer to be treated or prevented may also be a cancer having an activating RAS mutation (e.g., an activating KRAS mutation, such as, e.g., G12D, G12V, G12C, or Q61 H) and/or an activating EGFR mutation, including any one of the above-mentioned specific types of cancer further having an activating RAS mutation and/or an activating EGFR mutation.
  • an activating RAS mutation e.g., an activating KRAS mutation, such as, e.g., G12D, G12V, G12C, or Q61 H
  • MTH1 specifically assists RAS-induced tumors in preventing tumor- suppressive effects such as senescence whilst enabling maintenance and progression of the tumor.
  • MTH1 inhibition is therefore considered to help impairing tumor growth by abrogating mitogenic signaling, epithelial-mesenchymal transition (EMT), a hallmark of progressing and aggressive tumors, anoikis inhibition and PI3K/Akt-mediated pro-survival signaling (Rai, Proteomics, 2003, 3(8): 1454-63).
  • EMT epithelial-mesenchymal transition
  • PI3K/Akt-mediated pro-survival signaling Rosteomics, 2003, 3(8): 1454-63
  • MTH1 may be a promising target for adenocarcinomas expressing EGFR as the micro-RNA MiR-145 which suppresses both EGFR and MTH1 is downregulated in these tumors (Cho, RNA Biology, 201 1 , 8(1 ):125-31 ) and as the reexpression of MiR-145 led to a downregulation of EGFR and MTH1 on both mRNA and protein level and impaired the growth of EGFR-positive cell lines.
  • the invention thus also relates to the compound of formula (I), as described and defined herein, or a pharmaceutically acceptable salt, solvate or prodrug thereof, or a pharmaceutical composition comprising any of the aforementioned entities and a pharmaceutically acceptable excipient, for use in the treatment or prevention of RAS-dependent and/or EGFR-dependent cancer, wherein the RAS-dependent and/or EGFR-dependent cancer may further be selected from any one of the specific types of cancer described in the preceding paragraph.
  • Triple negative breast cancer cell lines are also known to be sensitive to oxidative DNA damage which can sensitize these cancers to chemotherapeutics such as PARP inhibitors, i i vvi cu e uui iaiuci cu lu uAiuaii c L urt legions can tnereiore advantageously be used to treat this type of cancer.
  • chemotherapeutics such as PARP inhibitors, i i vvi cu e uui iaiuci cu lu uAiuaii c L urt legions can tnereiore advantageously be used to treat this type of cancer.
  • the present invention particularly relates to a compound of formula (I) or a pharmaceutically acceptable salt, solvate or prodrug thereof, or a pharmaceutical composition comprising any of the aforementioned entities and a pharmaceutically acceptable excipient, for use in the treatment or prevention of triple negative breast cancer (particularly breast cancer lacking expression of estrogen receptor-a, lacking expression of progesterone receptor, and lacking overexpression or amplification of the HER2/NEU oncogene).
  • the BRCA1 and BRCA2 genes which are mutated in certain types of breast cancer are known to be involved in the repair of oxidative DNA damage including 8-oxo-guanine-based lesions (Le Page, Cancer Res, 2000, 60(19):5548-52), which makes breast cancer cells deficient in BRCA1 and/or BRCA2 particularly sensitive towards oxidative DNA damage (Alii, Cancer Res, 2009, 69(8):3589-96).
  • the compounds of formula (I) are thus also highly advantageous for the treatment or prevention of breast cancer having a BRCA1 and/or BRCA2 gene mutation.
  • the breast cancer to be treated or prevented may further be EGFR-dependent.
  • MMR mismatch DNA repair pathway
  • Particularly tumor types with elevated oxidative stress and cancers with dysregulated hypoxia signaling including, e g., the VHL and/or HIF1-alpha pathways are considered to be sensitive towards MTH1 inhibition and are thus preferred to be treated or prevented using a compound of formula (I) according to the invention.
  • the cancer to be treated or prevented is a cancer (including, e.g., any one of the above-mentioned specific types of cancer) wherein MTH1 is involved in the development and/or progression of the cancer.
  • the status e.g., the genetic constitution, expression and/or activity
  • the MTH1 status of a cancer represents an appropriate indicator for the involvement of MTH1 in the development and/or progression of that cancer.
  • the present invention thus also relates to stratifying cancer patients with respect to their MTH1 status.
  • the present invention further relates to an in vitro method of determining the effectiveness of a compound of formula (I) in the treatment or prevention of cancer in a subject, the method comprising:
  • a sample e.g. , a cell or tissue sample, particularly a cancer cell sample or a cancer tissue sample
  • a positive MTH1 status is indicative of an effective treatment or prevention of cancer in the subject.
  • the MTH1 status may be, e.g., the level of MTH1 biological activity and/or the level of MTH1 expression.
  • the level of MTH1 biological activity may be monitored by purifying MTH1 from the sample obtained from the subject and measuring the production of pyrophosphate (PPi) generated by ⁇ 1 -mediated hydrolysis of 8-oxo-dGTP.
  • PPi pyrophosphate
  • purified MTH1 may be contacted with 8-oxo-dGTP and the production of PPi may be measured using, e.g., the PPiLight Inorganic Pyrophosphate Assay (Lonza Rockland Inc.).
  • the level of MTH1 biological activity (e.g., the quantity of produced PPi) in the sample obtained from the subject may be compared to the level of MTH1 biological activity (e.g., the quantity of produced PPi) in a sample obtained from of a healthy control subject.
  • the MTH1 status of the subject is positive if the level of MTH1 biological activity (e.g., the quantity of produced PPi) is higher in the sample from the subject as compared to the level of MTH1 biological activity in the sample of the healthy control subject.
  • the level of MTH1 expression may be determined, e.g., by PGR, RT-PCT or Western blot.
  • the MTH1 status of the subject is positive if the level of expression of MTH1 (e.g., the amount of MTH1 mRNA or of MTH1 protein) is higher in the sample from the subject as compared to the level of expression of MTH1 in a sample obtained from a healthy control subject.
  • Antibodies, probes and primers that can be used for determining/detecting a subject ' s MTH1 status are known in the art.
  • an antibody for determining a subject's MTH1 status is, e.g., Novus Biologicals MTH1 Antibody (NB100-109).
  • Primers for determining a subject's MTH1 status are described, e.g., in Kennedy (1998) FEBS Lett. 429(1 ): 17-20, which is herein incorporated by reference in its entirety.
  • the following exemplary primers can also be used for determining a subject's MTH1 status:
  • the MTH1 status can also be determined, e.g., as described in WO 2014/033136.
  • Antibodies, probes and primers that can be used for determining/detecting a subject ' s RAS status are known in the art and are also described, e.g., in WO 2014/033136.
  • an antibody for detecting a subject's RAS status is, e.g., Cell Signaling Ras Antibody #3965.
  • Primers for detecting a subject's RAS status are described, e.g.. in Keohavong (1996) Clin Cancer Res. 2(2):41 1 -8 or in Gerry (1999) Mol Biol. 292(2):251-62, which are herein incorporated by reference in their entirety.
  • the in vitro method described herein can be performed prior to the administration of a compound of formula (I) to a subject in order to evaluate whether the subject will particularly benefit from the treatment with this compound. Accordingly, the invention provides a method of treating or preventing cancer in a subject in need thereof, wherein the method comprises:
  • step (ii) administering a compound of formula (I) or a pharmaceutically acceptable salt, solvate or prodrug thereof to the subject if the result obtained in step (i) is indicative of an effective treatment or prevention of cancer.
  • the gene that encodes MTH1 is also known as NUDT1.
  • NUDT1 The gene that encodes MTH1 is also known as NUDT1.
  • four isoforms of NUDT1/MTH1 p18, p21 , p22 and p26
  • p18 is considered to be the dominant isoform
  • the isoform pi 8 has been used in the experiments described herein. Mutations have been reported for MTH1 , however, their physiological or clinical relevance has not been elucidated.
  • NUDT/MTH1 -status e.g., for detecting the level, such as the expression level, of NUDT/MTH1 or for determining whether the nucleotide sequence or amino acid sequence of NUDT/MTH1 contains a specific mutation
  • Activating and inactivating aberrations include genetic aberrations such as gene mutation, gene copy number increase, aberration of gene expression, and aberration of mRNA expression.
  • cytogenetic aberrations several detection methods are known in the art and are reviewed, e g., in Speicher (2005) Nat Rev Genet 6(10):782-92.
  • the present invention furthermore relates to the use of a compound of formula (I) or a pharmaceutically acceptable salt, solvate or prodrug thereof as an MTH1 inhibitor in research, particularly as a research tool compound.
  • the invention refers to the in vitro use of a compound of formula (I) or a pharmaceutically acceptable salt, solvate or prodrug thereof as an MTH1 inhibitor and, in particular, to the in vitro use of a compound of formula (I) or a pharmaceutically acceptable salt, solvate or prodrug thereof as a research tool compound acting as an MTH1 inhibitor.
  • the compounds of formula (I) will be described in more detail in the following:
  • R 1 is halogen (e.g., -F, -CI, -Br, or -I; preferably -F, -CI, or -Br; more preferably -C!), methoxy (i.e., -OCH 3 ), ethoxy (i.e., -OCH 2 CH 3 ), or -CF 3 .
  • R 1 is halogen or methoxy. More preferably, R 1 is halogen. Even more preferably, R 1 is halogen which is selected from -F, -CI and -Br. Most preferably, R 1 is -CI.
  • Each R 2 is independently selected from C 1-5 alkyl, C 2 . 5 alkenyl, C 2 - 5 alkynyl, -(C 0 . 3 alkylene)-OH, -(Co-3 alkylene)-0(C 1 . 5 alkyl), -(C 0 . 3 alkylene)-0(d. 5 alkylene)-OH, -(C 0 . 3 alkylene)-0(C 1-5 alkylene)-0(C 1 . 5 alkyl), -(C 0-3 alkylene)-SH, -(Co-3 alkylene)-S(C,. 5 alkyl), -(C 0 - 3 alkylene)-S(Ci.
  • each R 2 is independently selected from d- 5 alkyl, C 2 - 5 alkenyl, C 2 . 5 alkynyl, -OH, -0(Ci.5 alkyl), -0(d. 5 alkylene)-OH, -0(d. 5 alkylene)-0(d. 5 alkyl), -SH, -S(d. 5 alkyl), -S(Ci_ 5 alkylene)-SH, -S(C, 5 alkylene)-S(d. 5 alkyl), -NH 2 , -NH(d 5 alkyl), -N(d. 5 alkyl)(Ci. 5 alkyl), halogen, d.
  • each R 2 is independently selected from d. 5 alkyl, C 2 - 5 alkenyl, C 2 . 5 alkynyl, -OH, -0(d_5 alkyl), -0(d. 5 alkylene)-OH, -0(d-s alkylene)-0(C 1-5 alkyl), -SH, -S(d_ 5 alkyl), -S(d-s alkyiene)-SH, -S(d.
  • each R 2 is independently selected from d-4 alkyl (e.g., methyl or ethyl), -OH, -0(d_ 4 alkyl) (e.g., -OCH 3 or -OCH 2 CH 3 ), -NH 2 , -NH(d_ 4 alkyl) (e.g., -NHCH 3 ), -N(d_ 4 alkyl)(C 1 4 alkyl) (e.g., -N(CH 3 ) 2 ), halogen (e.g., -F, -CI, -Br, or -I), -CF 3 , and -CN.
  • d-4 alkyl e.g., methyl or ethyl
  • -OH e.g., -OCH 3 or -OCH 2 CH 3
  • -NH 2 e.g., -NH(d_ 4 alkyl) (e.g., -NHCH 3 ), -N(
  • Each R a is independently selected from d-s alkyl, C 2 . 5 alkenyl, C 2 . 5 alkynyl. -(C 0 .3 alkylene)-OH, -(C 0 -3 alkylene)-0(d.5 alkyl), -(C 0 . 3 alkylene)-0(d-5 alkylene)-OH, -(C 0 . 3 alkylene)-0(d. 5 alkylene)-0(d- 5 alkyl), -(C 0 . 3 alkylene)-SH, -(C 0 . 3 alkylene)-S(d_ 5 alkyl), -(C 0 . 3 alkylene)-S(d.
  • each R 3 is independently selected from d-5 alkyl, C 2 _ 5 alkenyl, C 2 . 5 alkynyl, -OH, -0(Ci. 5 alkyl), -0(d. 5 alkylene)-OH, -0(d-s alkylene)-0(d. 5 alkyl), -SH, -S(d. 5 alkyl), -S(d. 5 alkylene)-SH, -S(d 5 alkylene)-S(d-s alkyl), -NH 2 , -NH(d. 5 alkyl), -N(d-5 alkyl)(Ci. 5 alkyl), i idiuycn, 1-5
  • alkyl -0-CO-(d. 5 alkyl), -CO-NH 2 , -CO-NH(d. 5 alkyl), -CO-N(d. 5 alkyl)(d -5 alkyl), -NH-CO-(d. 5 alkyl), -N(d_ 5 alkyl)-CO-(d -5 alkyl), -S0 2 -NH 2 , -S0 2 -NH(C 1 5 alkyl), -S0 2 -N(C 1-5 alkyl)(d.
  • each R 3 is independently selected from C 1 -5 alkyl, C 2 _ 5 alkenyl, C 2 . 5 alkynyl, -OH, -0(d_5 alkyl), -0(d_ 5 alkylene)-OH, -0(d_ 5 alkylene)-0(d. 5 alkyl), -SH, -S(d- 5 alkyl), -S(d. 5 alkylene)-SH, -S(d. 5 alkylene)-S(d.
  • each R 3 is independently selected from d_ 4 alkyl (e.g., methyl or ethyl), -OH, -0(C 1-4 alkyl) (e.g., -OCH 3 or -0CH 2 CH 3 ), -NH 2 , -NH(d.4 alkyl) (e.g., -NHCH 3 ), -N(C 1 .
  • R 4 is hydrogen or d-s alkyl.
  • R 4 is hydrogen, methyl or ethyl. More preferably, R 4 is hydrogen.
  • R 5 is a covalent bond or d-s alkylene.
  • R 5 is a covalent bond, -CH 2 -, -CH 2 -CH 2 - or -CH 2 -CH 2 -CH 2 -. More preferably, R° is a covalent bond or -CH 2 -. Even more preferably, R 5 is a covalent bond.
  • the group A is aryl or heteroaryl, wherein said aryl and said heteroaryl are each optionally substituted with one or more (e.g., one, two, or three) groups R 6 .
  • A is aryl which is optionally substituted with one or more (e.g., one, two, or three) groups R 6 .
  • A is phenyl which is optionally substituted with one or more (e.g., one, two, or three) groups R 6 . It is furthermore preferred that the aforementioned groups A (particularly the aforementioned aryl, heteroaryl, or phenyl groups) are unsubstituted or are substituted with one or two groups R 6 .
  • Each R 6 is independently selected from C-,. 5 alkyl, -s alkenyl, d-s alkynyl, -(C 0 . 3 alkylene)-OH, -(C 0 - 3 alkylene)-0(d-s alkyl), -(C 0 . 3 aikylene)-G(d-s alkylene)-OH, -(C 0 . 3 a!kyiene)-0(Ci_ 5 alkylene)-0(d_ 5 alkyl), -(C 0 . 3 alkylene)-SH, -(C 0 . 3 alky!ene)-S(d-s alkyl), -(C 0 .
  • each R 6 is independently selected from d-5 alkyl, C 2 . 5 alkenyl, C 2 . 5 alkynyl, -OH, -0(d. 5 alkyl), -0(d.
  • each R 6 is independently selected from d_ 4 alkyl, -OH, -0(d_ 4 alkyl), -NH 2 , -NH(d- 4 alkyl), -N(C -4 alkyl)(d_ 4 alkyl), halogen, -CF 3 , and -CN, and two groups R 6 which are bound to adjacent carbon ring atoms may also be mutually linked to form a group -0-(CH 2 )-i.
  • each R 6 is independently selected from C 1-4 alkyl (e.g., methyl or ethyl), -OH, -0(d- 4 alkyl) (e.g., -OCH 3 ), halogen (e.g., -F or -CI), and -CF 3 , and two groups R 6 which are bound to adjacent carbon ring atoms may also be mutually linked to form a group -0-(CH 2 )i -3 -0- (e.g., -0-CH 2 -0- or -0-CH 2 -CH 2 -0-).
  • the group A is phenyl which is optionally substituted with one or two groups independently selected from d_ 4 alkyl (e.g., methyl or ethyl), -OH, -0(C 1-4 alkyl) (e.g., -OCH 3 ), -NH 2 , -NH(d_ 4 alkyl), -N(C 1-4 alkyl)(d.
  • halogen e.g., -F or -CI
  • -CF 3 e.g., -CF 3
  • -CN e.g., -CN
  • A phenyl which is substituted on adjacent ring atoms with two groups that are mutually linked to form a group (e.g., -0-CH 2 -0- or -0-CH 2 -CH 2 -0-).
  • n is 0, 1 , 2 or 3.
  • n is 0, 1 or 2. More preferably, n is 0 or 1 . Even more preferably, n is 0.
  • m is an integer of 0 to 6.
  • m is 0, 1 , 2 or 3.
  • n indicates the number of substituents R 2 that are bound to the phenyl moiety comprised in the cyclopenta[b]quinoline ring system of the compound of formula (I). If n is 0, then this phenyl moiety is not substituted with any group R 2 , i.e. is substituted with hydrogen instead of R 2 .
  • m indicates the number of substituents R a that are bound to the cyclopentyl moiety comprised in the cyclopenta[b]quinoline ring system of the compound of formula (I). If m is 0, then this cyclopentyl moiety is unsubstituted, i.e. is substituted with hydrogen instead of R 3 .
  • the compound of formula (I) is a compound of the following formula (la) or a pharmaceutically acceptable salt, solvate or prodrug thereof:
  • the group R 1 comprised in the compound of formula (la) is halogen (preferably -F, -CI, or -Br; more preferably -CI).
  • the remaining groups/variables comprised in the compound of formula (la), particularly R 2 , R 3 , R 4 , A, R 6 , n and m, have the same meanings, including the same preferred meanings, as described and defined herein for the corresponding groups comprised in compound of formula (!).
  • Preferred examples of the compounds of formula (I) or (la) are the following compounds (1) to (8) as well as pharmaceutically acceptable salts, solvates and prodrugs of any of these compounds:
  • the compounds of formula (I), including also the compounds of formula (la), can be prepared by methods known in the field of synthetic chemistry. For example, these compounds can be prepared in accordance with or in analogy to the synthetic routes described in the literature cited in this specification, including in particular Rosini M et al., Bioorg Med Chem, 2006, 14(23):7846-53.
  • An example of a synthetic procedure for the preparation of compounds of formula (I) is given in Scheme 1 below.
  • Substituted anthranilic acid derivatives may be converted into the corresponding 9-chloro-2,3-dihydro-1 H-cyclopenta[6]quinolones using phosphorus oxychloride and appropriately substituted cyclopentanone derivatives.
  • hydrocarbon group refers to a group consisting of carbon atoms and hydrogen atoms.
  • alkyl refers to a monovalent saturated acyclic (i.e., non-cyclic) hydrocarbon group which may be linear or branched. Accordingly, an “alkyl” group does not comprise any carbon-to-carbon double bond or any carbon-to-carbon triple bond.
  • a "Ci. 5 alkyl” denotes an alkyi group having 1 to 5 carbon atoms. Preferred exemplary alkyl groups are methyl, ethyl, propyl (e.g., n-propyl or isopropyl), or butyl (e.g., n-butyl, isobutyl, sec-butyl, or tert-butyl).
  • alkyl preferably refers to C 1-4 alkyl, more preferably to methyl or ethyl, and even more preferably to methyl.
  • alkenyl refers to a monovalent unsaturated acyclic hydrocarbon group which may be linear or branched and comprises one or more (e.g., one or two) carbon- to-carbon double bonds while it does not comprise any carbon-to-carbon triple bond.
  • C 2 -5 alkenyl denotes an alkenyl group having 2 to 5 carbon atoms.
  • Preferred exemplary alkenyl groups are ethenyl, propenyl (e.g., prop-1 -en-1-yl, prop-1 -en-2-yl, or prop-2-en-1 -yl), butenyl, butadienyl (e.g., buta-1 ,3-dien-1 -yl or buta-1 ,3-dien-2-yl), pentenyl, or pentadienyl (e.g., isoprenyl).
  • alkenyl preferably refers to C 2 . alkenyl.
  • alkynyl refers to a monovalent unsaturated acyclic hydrocarbon group which may be linear or branched and comprises one or more (e.g., one or two) carbon- to-carbon triple bonds and optionally one or more carbon-to-carbon double bonds.
  • C 2 -5 alkynyl denotes an alkynyl group having 2 to 5 carbon atoms.
  • Preferred exemplary alkynyl groups are ethynyl, propynyl, or butynyl.
  • alkynyl preferably refers to C 2 . 4 alkynyl.
  • alkylene refers to an alkanediyl group, i.e. a divalent saturated acyclic hydrocarbon group which may be linear or branched.
  • a "d. 5 alkylene” denotes an alkylene group having 1 to 5 carbon atoms, and the term “C 0 - 3 alkylene” indicates that a covalent bond (corresponding to the option "C 0 alkylene”) or a C 1 .3 alkylene is present.
  • Preferred exemplary alkylene groups are methylene (-CH 2 -), ethylene (e.g., -CH 2 -CH 2 - or -CH(-CH 3 )-), propylene (e.g., -CH 2 -CH 2 -CH 2 -, -CH(-CH 2 -CH 3 )-, -CH 2 -CH(-CH 3 )-, or -CH(-CH 3 )- CH 2 -), or butylene (e.g., -CH 2 -CH 2 -CH 2 -CH 2 -CH 2 -).
  • the term "alkylene” preferably refers to Ci_ 4 alkylene (including, in particular, linear C 1-4 alkylene), more preferably to methylene or ethylene, and even more preferably to methylene.
  • aryl refers to an aromatic hydrocarbon ring group, including monocyclic aromatic rings as well as bridged ring and/or fused ring systems containing at least one aromatic ring (e.g., ring systems composed of two or three fused rings, wherein at least one of these fused rings is aromatic; or bridged ring systems composed of two or three rings, wherein at least one of these bridged rings is aromatic).
  • Aryl may, e.g., refer to phenyl, naphthyl, dialinyl (i.e., 1 .2-dihydronaphthyl), tetralinyl (i.e., 1 ,2,3,4-tetrahydronaphthyl), anthracenyl, or phenanthrenyl.
  • an "aryl” preferably has 6 to 14 ring atoms, more preferably 6 to 10 ring atoms, and most preferably refers to phenyl.
  • heteroaryl refers to an aromatic ring group, including monocyclic aromatic rings as well as bridged ring and/or fused ring systems containing at least one aromatic ring (e.g., ring systems composed of two or three fused rings, wherein at least one of these fused rings is aromatic; or bridged ring systems composed of two or three rings, wherein at least one of these bridged rings is aromatic), wherein said aromatic ring group comprises one or more (such as, e.g., one, two, three, or four) ring heteroatoms independently selected from O, S and N, and the remaining ring atoms are carbon atoms, wherein one or more S ring atoms (if present) and/or one or more N ring atoms (if present) may optionally be oxidized, and further wherein one or more carbon ring atoms may optionally be oxidized (i.e., to form an oxo group).
  • aromatic ring group comprises one or more (such as, e.g., one, two,
  • Heteroaryl may, e.g., refer to thienyl (i.e., thiophenyl), benzo[b]thienyl, naphtho[2,3- bjihienyl, thianthrenyl, furyl (i.e., furanyl), benzofuranyi, isobenzofuranyl, chromenyi, xanthenyl, phenoxathiinyl, pyrrolyl (e.g., 2H-pyrrolyl), imidazolyl, pyrazolyl, pyridyl (i.e.
  • pyridinyl e.g., 2-pyridyl, 3-pyridyl, or 4-pyridyl
  • pyrazinyl e.g., 2-pyridyl, 3-pyridyl, or 4-pyridyl
  • pyrimidinyl pyridazinyl
  • indolizinyl isoindolyl, indolyl (e.g., 3H-indolyl), indazolyl
  • purinyl isoquinolyl, quinolyl, phthalazinyl, naphthyridinyl, quinoxalinyl, cinnolinyl, pteridinyl, carbazolyl, beta-carbolinyl, phenanthridinyl, acridinyl, perimidinyl, phenanthrolinyl (e.g. , [1 , 10]phenanthrolinyl, [1 ,7]phenanthro
  • furazanyl phenoxazinyl, pyrazolo[1 ,5-a]pyrimidinyl (e.g., pyrazolo[1 ,5-a]pyrimidin-3-yl), 1 ,2-benzoisoxazol-3-yl, benzothiazolyl, benzoxazolyl, benzisoxazolyl, benzimidazolyl, 1 H-tetrazolyl, 2H-tetrazolyl, coumarinyl, or chromonyl.
  • a “heteroaryl” preferably refers to a 5 to 14 membered (more preferably 5 to 10 membered) monocyclic ring or fused ring system comprising one or more (e.g., one, two, three or four) ring heteroatoms independently selected from O, S and N, wherein one or more S ring atoms (if present) and/or one or more N ring atoms (if present) are optionally oxidized, and wherein one or more carbon ring atoms are optionally oxidized; even more preferably, a “heteroaryl” refers to a 5 or 6 membered monocyclic ring comprising one or more (e.g., one, two or three) ring heteroatoms independently selected from O, S and N, wherein one or more S ring atoms (if present) and/or one or more N ring atoms (if present) are optionally oxidized, and wherein one or more carbon ring atoms are optionally oxidized;
  • halogen refers to fluoro (-F), chloro (-CI), bromo (-Br), or iodo (-1).
  • haloalkyl refers to an alkyl group substituted with one or more (preferably 1 to 6, more preferably 1 to 3) halogen atoms which are selected independently from fluoro, chloro, bromo and iodo, and are preferably all fluoro atoms. It will be understood that the maximum number of halogen atoms is limited by the number of available attachment sites and, thus, depends on the number of carbon atoms comprised in the alkyl moiety of the haloalkyl group.
  • Haloalkyl may, e.g., refer to -CF 3 , -CHF 2 , -CH 2 F, -CF 2 -CH 3 , -CH 2 -CF 3 , -CH 2 -CHF 2 , -CH 2 -CF 2 -CH 3 , -CH 2 -CF 2 -CF 3 , or -CH(CF 3 ) 2 .
  • substituents such as, e.g., one, two, three or four substituents. It will be understood that the maximum number of substituents is limited by the number of attachment sites available on the substituted moiety.
  • the "optionally substituted" groups referred to in this specification carry preferably not more than two substituents and may, in particular, carry only one substituent.
  • the optional substituents are absent, i.e. that the corresponding groups are unsubstituted.
  • the terms “optional”, “optionally” and “may” denote that the indicated feature may be present but can also be absent. Whenever the term “optional”, “optionally” or “may” is used, the present invention specifically relates to both possibilities, i.e., that the corresponding feature is present or, alternatively, that the corresponding feature is absent.
  • the expression “X is optionally substituted with Y” (or “X may be substituted with Y”) means that X is either substituted with Y or is unsubstituted.
  • a component of a composition is indicated to be “optional"
  • the invention specifically relates to both possibilities, i.e., that the corresponding component is present (contained in the composition) or that the corresponding component is absent from the composition.
  • substituent groups comprised in the compounds of formula (I) may be attached to the remainder of the respective compound via a number of different positions of the corresponding specific substituent group. Unless defined otherwise, the preferred attachment positions for the various specific substituent groups are as illustrated in the examples.
  • the term "about” preferably refers to ⁇ 10% of the indicated numerical value, more preferably to ⁇ 5% of the indicated numerical value, and in particular to the exact numerical value indicated.
  • the scope of the invention embraces all pharmaceutically acceptable salt forms of the compounds of formula (I) which may be formed, e.g., by protonation of an atom carrying an electron lone pair which is susceptible to protonation, such as an amino group, with an inorganic or organic acid, or as a salt of an acid group (such as a carboxylic acid group) with a physiologically acceptable cation.
  • Exemplary base addition salts comprise, for example: alkali metal salts such as sodium or potassium salts; alkaline earth metal salts such as calcium or magnesium salts; zinc salts; ammonium salts; aliphatic amine salts such as trimethylamine, triethylamine, dicyclohexylamine, ethanolamine, diethanolamine, triethanolamine, procaine salts, meglumine salts, ethylenediamine salts, or choline salts; aralkyl amine salts such as ⁇ , ⁇ -dibenzylethylenediamine salts, benzathine salts, benethamine salts; heterocyclic aromatic amine salts such as pyridine salts, picoline salts, quinoline salts or isoquinoline salts; quaternary ammonium salts such as tetramethylammonium salts, tetraethylammonium salts, benzyltrimethylammonium salts, benzyltriethyl
  • Exemplary acid addition salts comprise, for example: mineral acid salts such as hydrochloride, hydrobromide, hydroiodide, sulfate salts (such as, e.g., sulfate or hydrogensulfate salts), nitrate salts, phosphate salts (such as, e.g., phosphate, hydrogenphosphate, or dihydrogenphosphate salts), carbonate salts, hydrogencarbonate salts, perchlorate salts, borate salts, or thiocyanate salts; organic acid salts such as acetate, propionate, butyrate, pentanoate, hexanoate, heptanoate, octanoate, cyclopentanepropionate, decanoate, undecanoate, oleate, stearate, lactate, maleate, oxalate, fumarate, tartrate, malate, citrate, succinate, adipate, gluconate, glyco!ate,
  • Preferred pharmaceutically acceptable salts of the compounds of formula (I) include a hydrochloride salt, a hydrobromide salt, a mesylate salt, a sulfate salt, a tartrate salt, a fumarate salt, an acetate salt, a citrate salt, and a phosphate salt.
  • a particularly preferred pharmaceutically acceptable salt of the compound of formula (I) is a hydrochloride salt.
  • the compound of formula (I), including any one of the specific compounds of formula (I) described herein, is in the form of a hydrochloride salt, a hydrobromide salt, a mesylate salt, a sulfate salt, a tartrate salt, a fumarate salt, an acetate salt, a citrate salt, or a phosphate salt, and it is particularly preferred that the compound of formula (I) is in the form of a hydrochloride salt.
  • the scope of the invention embraces the compounds of formula (I) in any solvated form, including, e.g., solvates with water, for example hydrates, or with organic solvents such as, e.g., methanol, ethanol or acetonitrile, i.e., as a methanolate, ethanolate or acetonitrilate, respectively, or in the form of any polymorph. It is to be understood that such solvates of the compounds of the formula (I) also include solvates of pharmaceutically acceptable salts of the compounds of the formula (I).
  • the compounds of formula (I) may exist in the form of different isomers, in particular stereoisomers (including, e.g., geometric isomers (or cis/trans isomers), enantiomers and diastereomers) or tautomers. All such isomers of the compounds of formula (I) are contemplated as being part of the present invention, either in admixture or in pure or substantially pure form.
  • stereoisomers the invention embraces the isolated optical isomers of the compounds according to the invention as well as any mixtures thereof (including, in particular, racemic mixtures/racemates).
  • racemates can be resolved by physical methods, such as, e.g., fractional crystallization, separation or crystallization of diastereomeric derivatives, or separation by chiral column chromatography.
  • the individual optical isomers can also be obtained from the racemates via salt formation with an optically active acid followed by crystallization.
  • the present invention further encompasses any tautomers of the compounds provided herein.
  • the scope of the invention also embraces compounds of formula (I), in which one or more atoms are replaced by a specific isotope of the corresponding atom.
  • the invention encompasses compounds of formula (I), in which one or more hydrogen atoms (or, e.g., all hydrogen atoms) are replaced by deuterium atoms (i.e. , 2 H; also referred to as "D").
  • the invention also embraces compounds of formula (I) which are enriched in deuterium.
  • Naturally occurring hydrogen is an isotopic mixture comprising about 99.98 mol-% hydrogen-1 ( ⁇ ) and about 0.0156 mol-% deuterium ( 2 H or D).
  • the content of deuterium in one or more hydrogen positions in the compounds of formula (I) can be increased using deuteration techniques known in the art.
  • a compound of formula (I) or a reactant or precursor to be used in the synthesis of the compound of formula (I) can be subjected to an H/D exchange reaction using, e.g., heavy water (D 2 0).
  • deuteration techniques are described in: Atzrodt J et al., Bioorg Med Chem, 20(18), 5658-5667, 2012; William JS et al., Journal of Labelled Compounds and Radiopharmaceuticals, 53(1 1-12), 635-644, 2010; Modvig A et al., J Org Chem, 79, 5861 -5868, 2014.
  • the content of deuterium can be determined, e.g., using mass spectrometry or NMR spectroscopy.
  • it is preferred that the compound of formula (I) is not enriched in deuterium. Accordingly, the presence of naturally occurring hydrogen atoms or 1 H hydrogen atoms in the compounds of formula (I) is preferred.
  • the present invention also embraces compounds of formula (I), in which one or more atoms are replaced by a positron-emitting isotope of the corresponding atom, such as, e.g., 18 F, 11 C, 13 N, 15 0, 76 Br, 77 Br, 120 l and/or 124 l.
  • a positron-emitting isotope of the corresponding atom such as, e.g., 18 F, 11 C, 13 N, 15 0, 76 Br, 77 Br, 120 l and/or 124 l.
  • Such compounds can be used as tracers or imaging probes in positron emission tomography (PET).
  • the invention thus includes (i) compounds of formula (I), in which one or more fluorine atoms (or, e.g., all fluorine atoms) are replaced by 18 F atoms, (II) compounds of formula (I), in which one or more carbon atoms (or, e.g., all carbon atoms) are replaced by 11 C atoms, (iii) compounds of formula (I), in which one or more nitrogen atoms (or, e.g., all nitrogen atoms) are replaced by 13 N atoms, (iv) compounds of formula (I), in which one or more oxygen atoms (or, e.g., all oxygen atoms) are replaced by 5 0 atoms, (v) compounds of formula (I), in which one or more bromine atoms (or, e.g., all bromine atoms) are replaced by 76 Br atoms, (vi) compounds of formula (I), in which one or more bromine atoms (or, e.g.,
  • prodrugs of the compounds of formula (I) are derivatives which have chemically or metabolically cleavable groups and become, by solvolysis or under physiological conditions, the compounds of formula (I) which are pharmaceutically active in vivo.
  • Prodrugs of the compounds according to the the present invention may be formed in a conventional manner with a functional group of the compounds such as, e.g., with an amino, hydroxy or carboxy group.
  • the prodrug form often offers advantages in terms of solubility, tissue compatibility or delayed release in a mammalian organism (see, Bundgaard, H., Design of Prodrugs, pp. 7-9, 21 -24, Elsevier, Amsterdam 1985).
  • Prodrugs include acid derivatives, such as, e.g., esters prepared by reaction of the parent acidic compound with a suitable alcohol, or amides prepared by reaction of the parent acid compound with a suitable amine. If a compound of the present invention has a carboxyl group, an ester derivative prepared by reacting the carboxyl group with a suitable alcohol or an amide derivative prepared by reacting the carboxyl group with a suitable amine is exemplified as a prodrug.
  • An especially preferred ester derivative as a prodrug is methylester, ethylester, n-propylester, isopropylester, n- butylester, isobutylester, tert-butylester, morpholinoethylester, ⁇ , ⁇ -diethylglycolamidoester or a-acetoxyethylester.
  • a compound of the present invention has a hydroxy group
  • an acyloxy derivative prepared by reacting the hydroxyl group with a suitable acylhalide or a suitable acid anhydride is exemplified as a prodrug.
  • a compound of the present invention has an amino group
  • an amide derivative prepared by reacting the amino group with a suitable acid halide or a suitable mixed anhydride is exemplified as a prodrug.
  • the compounds provided herein may be administered as compounds per se or may be formulated as medicaments.
  • the medicaments/pharmaceutical compositions may optionally comprise one or more pharmaceutically acceptable excipients, such as carriers, diluents, fillers, disintegrants, lubricating agents, binders, colorants, pigments, stabilizers, preservatives, antioxidants, and/or solubility enhancers.
  • pharmaceutically acceptable excipients such as carriers, diluents, fillers, disintegrants, lubricating agents, binders, colorants, pigments, stabilizers, preservatives, antioxidants, and/or solubility enhancers.
  • the pharmaceutical compositions may comprise one or more solubility enhancers, such as, e.g., poly(ethylene glycol), including poly(ethylene glycol) having a molecular weight in the range of about 200 to about 5,000 Da, ethylene glycol, propylene glycol, non-ionic surfactants, tyloxapol, polysorbate 80, macrogol-15-hydroxystearate, phospholipids, lecithin, dimyristoyl phosphatidylcholine, dipalmitoyl phosphatidylcholine, distearoyl phosphatidylcholine, cyclodextrins, a-cyclodextrin, ⁇ -cyclodextrin, ⁇ -cyclodextrin, hydroxyethyl- ⁇ -cyclodextrin, hydroxypropyl-p-cyclodextrin, hydroxyethyl-y-cyclodextrin, hydroxypropyl- ⁇ - cyclodextrin,
  • compositions can be formulated by techniques known to the person skilled in the art, such as the techniques published in "Remington: The Science and Practice of Pharmacy", Pharmaceutical Press, 22 nd edition.
  • the pharmaceutical compositions can be formulated as dosage forms for oral, parenteral, such as intramuscular, intravenous, subcutaneous, intradermal, intraarterial, intracardial, rectal, nasal, topical, aerosol or vaginal administration.
  • Dosage forms for oral administration include coated and uncoated tablets, soft gelatin capsules, hard gelatin capsules, lozenges, troches, solutions, emulsions, suspensions, syrups, elixirs, powders and granules for reconstitution, dispersible powders and granules, medicated gums, chewing tablets and effervescent tablets.
  • Dosage forms for parenteral administration include solutions, emulsions, suspensions, dispersions and powders and granules for reconstitution. Emulsions are a preferred dosage form for parenteral administration.
  • Dosage forms for rectal and vaginal administration include suppositories and ovula.
  • Dosage forms for nasal administration can be administered via inhalation and insufflation, for example by a metered inhaler.
  • Dosage forms for topical administration include creams, gels, ointments, salves, patches and transdermal delivery systems.
  • the compounds of formula (I) or the above described pharmaceutical compositions comprising a compound of formula (I) may be administered to a subject by any convenient route of administration, whether systemically/peripherally or at the site of desired action, including but not limited to one or more of: oral (e.g., as a tablet, capsule, or as an ingestible solution), topical (e.g., transdermal, intranasal, ocular, buccal, and sublingual), parenteral (e.g., using injection techniques or infusion techniques, and including, for example, by injection, e.g., subcutaneous, intradermal, intramuscular, intravenous, intraarterial, intracardiac, intrathecal, intraspinal, intracapsular, subcapsular, intraorbital, intraperitoneal, intratracheal, subcuticular, intraarticular, subarachnoid, or intrasiernal by, e.g., implant of a depot, for example, subcutaneously or intramuscularly), pulmonary
  • examples of such administration include one or more of: i travenously, intraarterially, intraperitoneal ⁇ , intrathecally, intraventricularly, intraurethrally, intrasternally, intracardially, intracranial ⁇ , intramuscularly or subcutaneously administering the compounds or pharmaceutical compositions, and/or by using infusion techniques.
  • parenteral administration the compounds are best used in the form of a sterile aqueous solution which may contain other substances, for example, enough salts or glucose to make the solution isotonic with blood.
  • the aqueous solutions should be suitably buffered (preferably to a pH of from 3 to 9), if necessary.
  • the preparation of suitable parenteral formulations under sterile conditions is readily accomplished by standard pharmaceutical techniques well known to those skilled in the art.
  • Said compounds or pharmaceutical compositions can also be administered orally in the form of tablets, capsules, ovules, elixirs, solutions or suspensions, which may contain flavoring or coloring agents, for immediate-, delayed-, modified-, sustained-, pulsed- or controlled-release applications.
  • the tablets may contain excipients such as microcrystalline cellulose, lactose, sodium citrate, calcium carbonate, dibasic calcium phosphate and glycine, disintegrants such as starch (preferably corn, potato or tapioca starch), sodium starch glycolate, croscarmellose sodium and certain complex silicates, and granulation binders such as polyvinylpyrrolidone, hydroxypropylmethylcellulose (HPMC), hydroxypropylcellulose (HPC), sucrose, gelatin and acacia. Additionally, lubricating agents such as magnesium stearate, stearic acid, glyceryl behenate and talc may be included. Solid compositions of a similar type may also be employed as fillers in gelatin capsules.
  • excipients such as microcrystalline cellulose, lactose, sodium citrate, calcium carbonate, dibasic calcium phosphate and glycine
  • disintegrants such as starch (preferably corn, potato or tapioca starch), sodium starch glyco
  • Preferred excipients in this regard include lactose, starch, a cellulose, or high molecular weight polyethylene glycols.
  • the agent may be combined with various sweetening or flavoring agents, coloring matter or dyes, with emulsifying and/or suspending agents and with diluents such as water, ethanol, propylene glycol and glycerin, and combinations thereof.
  • said compounds or pharmaceutical compositions can be administered in the form of a suppository or pessary, or it may be applied topically in the form of a gel, hydrogel, lotion, solution, cream, ointment or dusting powder.
  • the compounds of the present invention may also be dermally or transdermal ⁇ administered, for example, by the use of a skin patch.
  • sustained-release compositions include semi-permeable polymer matrices in the form of shaped articles, e.g., films, or microcapsules.
  • Sustained-release matrices include, e.g., polylactides (see, e.g., US 3,773,919), copolymers of L-glutamic acid and gamma-ethyl-L-glutamate (Sidman, U. et al., Biopolymers 22:547-556 (1983)), poly(2-hydroxyethyl methacrylate) (R. Langer et al., J. Biomed. Mater. Res.
  • Sustained-release pharmaceutical compositions also include liposomally entrapped compounds.
  • Liposomes containing a compound of the present invention can be prepared by methods known in the art, such as, e.g., the methods described in any one of: DE3218121 ; Epstein et al., Proc. Natl. Acad. Sci. (USA) 82:3688-3692 (1985); Hwang et a!., Proc. Natl.
  • Said compounds or pharmaceutical compositions may also be administered by the pulmonary route, rectal routes, or the ocular route.
  • they can be formulated as micronized suspensions in isotonic, pH adjusted, sterile saline, or, preferably, as solutions in isotonic, pH adjusted, sterile saline, optionally in combination with a preservative such as a benza!konium chloride.
  • they may be formulated in an ointment such as petrolatum.
  • dry powder formulations of the compounds of formula (I) for pulmonary administration, particularly inhalation.
  • Such dry powders may be prepared by spray drying under conditions which result in a substantially amorphous glassy or a substantially crystalline bioactive powder.
  • dry powders of the compounds of the present invention can be made according to the emulsification/spray drying process disclosed in WO 99/16419 or WO 01/85136.
  • Spray drying of solution formulations of the compounds of the present invention can be carried out, e.g., as described generally in the "Spray Drying Handbook", 5th ed., K. Masters, John Wiley & Sons, Inc., NY (1991 ), and in WO 97/41833 or WO 03/05341 1.
  • said compounds or pharmaceutical compositions can be formulated as a suitable ointment containing the active compound suspended or dissolved in, for example, a mixture with one or more of the following: mineral oil, liquid petrolatum, white petrolatum, propylene glycol, emulsifying wax and water.
  • they can be formulated as a suitable lotion or cream, suspended or dissolved in, for example, a mixture of one or more of the following: mineral oil, sorbitan monostearate, a polyethylene glycol, liquid paraffin, poiysorbate 60, cetyl esters wax, 2-octyldodecanol, benzyl alcohol and water.
  • the present invention thus relates to the compounds or the pharmaceutical compositions provided herein, wherein the corresponding compound or pharmaceutical composition is to be administered by any one of: an oral route; topical route, including by transdermal, intranasal, ocular, buccal, or sublingual route; parenteral route using injection techniques or infusion techniques, including by subcutaneous, intradermal, intramuscular, intravenous, intraarterial, intracardiac, intrathecal, intraspinal, intracapsular, subcapsular, intraorbital, intraperitoneal, intratracheal, subcuticular, intraarticular, subarachnoid, intrasternal, intraventricular, intraurethral, or intracranial route; pulmonary route, including by inhalation or insufflation therapy; gastrointestinal route; intrauterine route; intraocular route; subcutaneous route; ophthalmic route, including by intravitreal, or intracameral route; rectal route; or vaginal route.
  • Particularly preferred routes of administration of the compounds or pharmaceutical compositions of the present invention
  • a physician will determine the actual dosage which will be most suitable for an individual subject.
  • the specific dose level and frequency of dosage for any particular individual 0 subject may be varied and will depend upon a variety of factors including the activity of the specific compound employed, the metabolic stability and length of action of that compound, the age, body weight, general health, sex, diet, mode and time of administration, rate of excretion, drug combination, the severity of the particular condition, and the individual subject undergoing therapy.
  • a proposed, yet non-limiting dose of the compounds according to the invention for oral administration to a human may be 0.05 to 2000 mg, preferably 0.1 mg to 1000 mg, of the active ingredient per unit dose.
  • the unit dose may be administered, e.g., 1 to 3 times per day.
  • the unit dose may also be administered 1 to 7 times 0 per week, e.g., with not more than one administration per day. It will be appreciated that it may be necessary to make routine variations to the dosage depending on the age and weight of the patient/subject as well as the severity of the condition to be treated. The precise dose and also the route of administration will ultimately be at the discretion of the attendant physician or veterinarian.
  • the compounds of formula (I) can be used in combination with other therapeutic agents, including in particular other anticancer agents.
  • a compound of the invention When a compound of the invention is used in combination with a second therapeutic agent active against the same disease, the dose of each compound may differ from that when the compound is used alone.
  • the combination of a0 compound of the present invention with a second therapeutic agent may comprise the administration of the second therapeutic agent simultaneously/concomitantly or sequentially/separately with the compound of the invention.
  • the second therapeutic agent to be administered in combination with a compound 5 of this invention is an anticancer drug.
  • the anticancer drug to be administered in combination with a compound of formula (I) according to the present invention may, e.g., be selected from: a tumor angiogenesis inhibitor (for example, a protease inhibitor, an epidermal growth factor receptor kinase inhibitor, or a vascular endothelial growth factor receptor kinase inhibitor); a cytotoxic drug (for example, an antimetabolite, such as purine and pyrimidine analogue antimetabolites); an antimitotic agent (for example, a microtubule stabilizing drug or an antimitotic alkaloid); a platinum coordination complex; an anti-tumor antibiotic; an alkylating agent (for example, a nitrogen mustard or a nitrosourea); an endocrine agent (for example, an adrenocorticosteroid, an androgen, an anti-androgen, an estrogen, an anti-ester
  • An alkylating agent which can be used as an anticancer drug in combination with a compound of the present invention may be, for example, a nitrogen mustard (such as cyclophosphamide, rnechlorethamine (ehlorrneth!ne), uramustine, melphalan, chlorambucil, ifosfamide, bendamustine, or trofosfamide), a nitrosourea (such as carmustine, streptozocin, fotemustine, lomustine, nimustine, prednimustine, ranimustine, or semustine), an alkyl sulfonate (such as busulfan, mannosulfan, or treosulfan), an aziridine (such as hexamethylmelamine (altretamine), triethylenemelamine, ThioTEPA ( ⁇ , ⁇ ' ⁇ '-triethylenethiophosphoramide), carboquone, or triaziquone), a hydrazine (such as pro
  • a platinum coordination complex which can be used as an anticancer drug in combination with a compound of the present invention may be, for example, cisplatin, carboplatin, nedaplatin, oxaliplatin, satraplatin, or triplatin tetranitrate.
  • a cytotoxic drug which can be used as an anticancer drug in combination with a compound of the present invention may be, for example, an antimetabolite, including folic acid analogue antimetabolites (such as aminopterin, methotrexate, pemetrexed, or raltitrexed), purine analogue antimetabolites (such as cladribine, clofarabine, fludarabine, 6-mercaptopurine (including its prodrug form azathioprine), pentostatin, or 6-thioguanine), and pyrimidine analogue antimetabolites (such as cytarabine, decitabine, 5-fluorouracil (including its prodrug forms capecitabine and tegafur), floxuridine, gemcitabine, enocitabine, or sapacitabine).
  • folic acid analogue antimetabolites such as aminopterin, methotrexate, pemetrexed, or raltitrexed
  • An antimitotic agent which can be used as an anticancer drug in combination with a compound of the present invention may be, for example, a taxane (such as docetaxel, larotaxel, ortataxel, paclitaxel/taxol, or tesetaxel), a Vinca alkaloid (such as vinblastine, vincristine, vinflunine, vindesine, or vinorelbine), an epothilone (such as epothilone A, epothilone B, epothilone C, epothilone D, epothilone E, or epothilone F) or an epothilone B analogue (such as ixabepilone/azaepothilone B).
  • a taxane such as docetaxel, larotaxel, ortataxel, paclitaxel/taxol, or tesetaxel
  • a Vinca alkaloid such as vinblastine
  • An anti-tumor antibiotic which can be used as an anticancer drug in combination with a compound of the present invention may be, for example, an anthracycline (such as aclarubicin, daunorubicin, doxorubicin, epirubicin, idarubicin, amrubicin, pirarubicin, valrubicin, or zorubicin), an anthracenedione (such as mitoxantrone, or pixantrone) or an anti-tumor antibiotic isolated from Streptomyces (such as actinomycin (including actinomycin D), bleomycin, mitomycin (including mitomycin C), or plicamycin).
  • an anthracycline such as aclarubicin, daunorubicin, doxorubicin, epirubicin, idarubicin, amrubicin, pirarubicin, valrubicin, or zorubicin
  • a tyrosine kinase inhibitor which can be used as an anticancer drug in combination with a compound of the present invention may be, for example, axitinib, bosutinib, cediranib, dasatinib, erlotinib, gefitinib, imatinib, lapatinib, lestaurtinib, nilotinib, semaxanib, sorafenib, sunitinib, or vandetanib.
  • a topoisomerase-inhibitor which can be used as an anticancer drug in combination with a compound of the present invention may be, for example, a topoisomerase I inhibitor (such as irinotecan, topotecan, camptothecin, belotecan, rubitecan, or lamellarin D) or a topoisomerase II inhibitor (such as amsacrine, etoposide, etoposide phosphate, teniposide, or doxorubicin).
  • a topoisomerase I inhibitor such as irinotecan, topotecan, camptothecin, belotecan, rubitecan, or lamellarin D
  • a topoisomerase II inhibitor such as amsacrine, etoposide, etoposide phosphate, teniposide, or doxorubicin.
  • a PARP inhibitor which can be used as an anticancer drug in combination with a compound of the present invention may be, for example, BMN-673, olaparib, rucaparib, veliparib, CEP 9722, MK 4827, BGB-290, or 3-aminobenzamide.
  • anticancer drugs may also be used in combination with a compound of the present invention.
  • the anticancer drugs may comprise biological or chemical molecules, like TNF- related apoptosis-inducing ligand (TRAIL), tamoxifen, amsacrine, bexarotene, estramustine, irofulven, trabectedin, cetuximab, panitumumab, tositumomab, alemtuzumab, bevacizumab, edrecolomab, gemtuzumab, alvocidib, seliciclib, aminolevulinic acid, methyl aminolevulinate, efaproxiral, porfimer sodium, talaporfin, temoporfin, verteporfin, alitretinoin, tretinoin, anagrelide, arsenic trioxide, atrasentan, bortezomib, carmofur,
  • biological drugs like antibodies, antibody fragments, antibody constructs (for example, single-chain constructs), and/or modified antibodies (like CDR-g rafted antibodies, humanized antibodies, "full humanized” antibodies, etc.) directed against cancer or tumor markers/factors/cytokines involved in proliferative diseases can be employed in co-therapy approaches with the compounds of the invention.
  • biological molecules are anti-HER2 antibodies (e.g. trastuzumab, Herceptin®), anti-CD20 antibodies (e.g.
  • the combinations referred to above may conveniently be presented for use in the form of a pharmaceutical formulation.
  • the individual components of such combinations may be administered either sequentially or simultaneously/concomitantly in separate or combined pharmaceutical formulations by any convenient route.
  • administration is sequential, either the compound of the present invention (i.e., the compound of formula (I) or a pharmaceutically acceptable salt, solvate or prodrug thereof) or the second therapeutic agent may be administered first.
  • administration is simultaneous, the combination may be administered either in the same pharmaceutical composition or in different pharmaceutical compositions.
  • the two compounds When combined in the same formulation, it will be appreciated that the two compounds must be stable and compatible with each other and the other components of the formulation. When formulated separately, they may be provided in any convenient formulation.
  • the compounds of formula (I) can also be administered in combination with physical therapy, such as radiotherapy.
  • Radiotherapy may commence before, after, or simultaneously with administration of the compounds of the invention.
  • radiotherapy may commence 1 -10 minutes, 1 -10 hours or 24-72 hours after administration of the compounds.
  • these time frames are not to be construed as limiting.
  • the subject is exposed to radiation, preferably gamma radiation, whereby the radiation may be provided in a single dose or in multiple doses that are administered over several hours, days and/or weeks.
  • Gamma radiation may be delivered according to standard radiotherapeutic protocols using standard dosages and regimens.
  • MTH1 inhibitors are considered to synergize with radiotherapy and/or chemotherapeutics/anticancer drugs, particularly with DNA damaging agents, agents that interfere with DNA repair mechanisms, and agents that induce the production or inhibit the clearance of reactive oxygen species (ROS).
  • ROS reactive oxygen species
  • an anticancer drug to be administered in combination with a compound of formula (I) is a PARP inhibitor (e.g., BMN-673, olaparib, rucaparib, veliparib, CEP 9722, MK 4827, BGB-290, or 3-aminobenzamide) or an EGFR inhibitor/antagonist (e.g., gefitinib, erlotinib, lapatinib, afatinib, neratinib, ABT-414, dacomitinib, AV-412, PD 153035, vandetanib, PKI-166, pelitinib, canertinib, icotinib, poziotinib, BMS-690514, CUDC-101 , AP261 13, XL647, cetuximab, panitumumab, zalutumumab, nimotuzumab. or matu
  • PARP inhibitor
  • an anticancer drug to be administered in combination with a compound of formula (I) is an immunooncology therapeutic (e.g., a monoclonal antibody or a polyclonal antibody) targeting any one of CTLA-4, PD-1/PD-L1 , TIM3, LAG 3, 0X4, CSF1 R, IDO, or CD40.
  • an immunooncology therapeutic e.g., a monoclonal antibody or a polyclonal antibody targeting any one of CTLA-4, PD-1/PD-L1 , TIM3, LAG 3, 0X4, CSF1 R, IDO, or CD40.
  • Such immunooncology therapeutics include, e.g., an anti-CTLA-4 antibody (particularly an antagonistic or pathway-blocking anti-CTLA-4 antibody; e.g., ipilimumab or tremelimumab), an anti-PD-1 antibody (particularly an antagonistic or pathway-blocking anti- PD-1 antibody; e.g., nivolumab (BMS-936558), pembrolizumab (MK-3475), pidilizumab (CT-01 1 ), AMP-224, or APE02058), an anti-PD-LI antibody (particularly a pathway-blocking anti-PD-L1 antibody; e.g., BMS-936559, MEDI4736, MPDL3280A (RG7446), MDX-1 105, or MEDI6469), an anti-TIM3 antibody (particularly a pathway-blocking anti-TIM3 antibody), an anti-LAG3 antibody (particularly an antagonistic or pathway-blocking anti-LAG3 antibody; e.
  • the present invention thus relates to a compound of formula (I) or a pharmaceutically acceptable salt, solvate, or prodrug thereof, or a pharmaceutical composition comprising any of the aforementioned entities in combination with a pharmaceutically acceptable excipient, for use in the treatment or prevention of cancer, wherein the compound or the pharmaceutical 5 composition is to be administered in combination with an anticancer drug and/or in combination with radiotherapy.
  • the compounds of formula (I) can also be used in monotherapy, particularly in the monotherapeutic treatment or prevention of cancer (i.e., without administering any other 0 anticancer agents until the treatment with the compound(s) of formula (I) is terminated).
  • the invention also relates to a compound of formula (I) or a pharmaceutically acceptable salt, solvate, or prodrug thereof, or a pharmaceutical composition comprising any of the aforementioned entities in combination with a pharmaceutically acceptable excipient, for use in the monotherapeutic treatment or prevention of cancer.
  • the subject or patient may be an animal (e.g., a non-human animal), a vertebrate animal, a mammal, a rodent (e.g., a guinea pig, a hamster, a rat, a mouse), a murine (e.g., a mouse), a canine (e.g., a dog), a feline (e.g., a cat), a porcine (e.g., a pig), an equine (e.g., a horse), a primate, a simian (e.g., a monkey or 0 ape), a monkey (e.g., a marmoset, a baboon), an ape (e.g., a gorilla, chimpanzee, orang-utan, gibbon), or a human.
  • an animal e.g., a non-human animal
  • a vertebrate animal e.g
  • animals are to be treated which are economically, agronomically or scientifically important.
  • Scientifically important organisms include, but are not limited to, mice, rats, and rabbits. Lower organisms such as, e.g., fruit flies like Drosophila melagonaster and nematodes like Caenorhabditis 5 elegans may also be used in scientific approaches.
  • Non-limiting examples of agronomically important animals are sheep, cattle and pigs, while, for example, cats and dogs may be considered as economically important animals.
  • the subject/patient is a mammal; more preferably, the subject/patient is a human or a non-human mammal (such as, e.g.
  • a guinea pig a hamster, a rat, a mouse, a rabbit, a dog, a cat, a horse, a monkey, an ape, a 0 marmoset, a baboon, a gorilla, a chimpanzee, an orang-utan, a gibbon, a sheep, cattle, or a pig); most preferably, the subject/patient is a human.
  • treatment of a disorder or disease as used herein (e.g., “treatment” of cancer) is well known in the art.
  • Treatment of a disorder or disease implies that a disorder or disease is5 suspected or has been diagnosed in a patient/subject.
  • a patient/subject suspected of suffering from a disorder or disease typically shows specific clinical and/or pathological symptoms which a skilled person can easily attribute to a specific pathological condition (i.e., diagnose a disorder or disease).
  • the "treatment" of a disorder or disease may, for example, lead to a halt in the progression of the disorder or disease (e.g., no deterioration of symptoms) or a delay in the progression of the disorder or disease (in case the halt in progression is of a transient nature only).
  • the "treatment” of a disorder or disease may also lead to a partial response (e.g., amelioration of symptoms) or complete response (e.g., disappearance of symptoms) of the subject/patient suffering from the disorder or disease.
  • the "treatment” of a disorder or disease may also refer to an amelioration of the disorder or disease, which may, e.g., lead to a halt in the progression of the disorder or disease or a delay in the progression of the disorder or disease.
  • Such a partial or complete response may be followed by a relapse.
  • a subject/patient may experience a broad range of responses to a treatment (such as the exemplary responses as described herein above).
  • the treatment of a disorder or disease may, inter alia, comprise curative treatment (preferably leading to a complete response and eventually to healing of the disorder or disease) and palliative treatment (including symptomatic relief).
  • prevention of a disorder or disease as used herein is also well known in the art.
  • a patient/subject suspected of being prone to suffer from a disorder or disease may particularly benefit from a prevention of the disorder or disease.
  • the subject/patient may have a susceptibility or predisposition for a disorder or disease, including but not limited to hereditary predisposition.
  • Such a predisposition can be determined by standard methods or assays, using, e.g., genetic markers or phenotypic indicators.
  • a disorder or disease to be prevented in accordance with the present invention has not been diagnosed or cannot be diagnosed in the patient/subject (for example, the patient/subject does not show any clinical or pathological symptoms).
  • prevention comprises the use of a compound of the present invention before any clinical and/or pathological symptoms are diagnosed or determined or can be diagnosed or determined by the attending physician.
  • the present invention specifically relates to each and every combination of features and embodiments described herein, including any combination of general and/or preferred features/embodiments.
  • the invention specifically relates to each combination of meanings (including general and/or preferred meanings) for the various groups and variables comprised in formula (I).
  • a number of documents including patent applications and scientific literature are cited. The disclosure of these documents, while not considered relevant for the patentability of this invention, is herewith incorporated by reference in its entirety. More specifically, all referenced documents are incorporated by reference to the same extent as if each individual document was specifically and individually indicated to be incorporated by reference.
  • the present invention particularly relates to the following items:
  • R 1 is halogen, methoxy, ethoxy, or -CF 3 ; each R 2 is independently selected from C 1-5 alkyl, C 2 . 5 alkenyl, C 2 . 5 alkynyl, -(C 0 . 3 alkylene)-OH, -(C 0 . 3 alkylene)-0(C 1 _ 5 alkyl), -(C 0 . 3 alkylene)-0(C 1 _ 5 alkylene)-OH,
  • each R 3 is independently selected from Ci. 5 alkyl, C 2 . 5 alkenyl, C 2 .
  • R 4 is hydrogen or d. 5 alkyl
  • R 5 is a covalent bond or d. 3 alkylene
  • A is ary! or heteroaryl, wherein said aryl and said heteroaryl are each optlona!!y substituted with one or more groups R 6 ;
  • alkylene)-OH -(C 0 . 3 alkylene)-0(d. 5 alkyl), -(C 0 . 3 alkylene)-0(C, 5 alkylene)-OH, -(C 0-3 alkylene)-0(Ci. 5 alkylene)-0(d-5 alkyl), -(C 0 . 3 alkylene)-SH, -(C 0 . 3 alkylene)-S(d_ 5 alkyl), -(C 0 . 3 alkylene)-S(C 1 .
  • R 6 which are bound to adjacent carbon ring atoms may also be mutually linked to form a group -0-(CH 2 ) 1-3 -0-; n is 0, 1 , 2 or 3; and m is an integer of 0 to 6; or a pharmaceutically acceptable salt, solvate or prodrug thereof for use in the treatment or prevention of cancer.
  • R 1 is halogen.
  • R 1 is -CI.
  • each R 2 is independently selected from C 1-5 alkyl, C 2 . 5 alkenyi, C 2 . 5 alkynyl, -OH, -0(C 1 . 5 alkyl), -0(d-s alkylene)-OH, -0(d-5 alkylene)-0(d. 5 alkyl), -SH, -S(C 1 . 5 alkyl), -S(d combat 5 alkylene)-SH, -S(d. 5 alkylene)-S(d- 5 alkyl), -NH 2 , -NH(C 1-5 alkyl), -N(d.
  • each R 3 is independently selected from d-5 alkyl, C 2 _s alkenyi, C 2 _5 alkynyl, -OH, -Q(d_5 alkyl). -0(d_5 alkylene)-OH, -0(C 1-5 alkylene)-0(d_ 5 alkyl), -SH, -S(d_ 5 alkyl), -S(C V5 alkylene)-SH, -S(C 1-5 alkylene)-S(d.
  • each R 6 is independently selected from C 1 .5 alkyl, C 2 . 5 alkenyl, C 2 diligent 5 alkynyl, -OH, -0(Cv 5 alkyl), -0(d_ 5 alkylene)-OH, -0(C 1-5 alkylene)-0(C 1-5 alkyl), -SH, -S(C,.5 alkyl), -S(C 1-5 alkylene)-SH, -S(C 1-5 alkylene)-S(C 1 .
  • a pharmaceutical composition comprising a compound as defined in any one of items 1 to 12 and a pharmaceutically acceptable excipient for use in the treatment or prevention of cancer.
  • a method of treating or preventing cancer comprising administering a compound as defined in any one of items 1 to 12 or the pharmaceutical composition of item 13 to a subject in need thereof.
  • said cancer is selected from gastrointestinal cancer, colorectal cancer, liver cancer, pancreatic cancer, stomach cancer, genitourinary cancer, bladder cancer, esophageal cancer, prostate cancer, lung cancer, breast cancer, hematological cancer, leukemia, lymphoma, ovarian cancer, brain cancer, neuroblastoma, Ewing's sarcoma, kidney cancer, skin cancer, and head and/or neck cancer.
  • R 1 is -F, -CI, -I, methoxy, ethoxy, or -CF 3 ; each R 2 is independently selected from d -5 alkyl, C 2tician 5 alkenyl, C2-5 alkynyl, -(C 0-3 alkylene)-OH, -(C 0 . 3 alkylene)-0(C 1 _ 5 alkyl), -(C 0 . 3 alkylene)-0(Ci. 5 alkylene)-OH, -(Co-3 alkylene)-0(C . 5 alkylene)-0(d-5 alkyl), -(C 0 . 3 alkylene)-SH, -(C Q . 3 alkylene)-S(d.
  • each R 3 Is independently selected from C 1 -5 alkyl, G 2 . 5 alkenyl, C 2 -5 alkynyl, -(Co-3 alkylene)-OH, -(C 0 . 3 alkylene)-0(d. 5 alkyl), -(C 0 - 3 alkylene)-0(d.
  • R 5 is a covalent bond or d. 3 alkylene;
  • A is aryi or heteroaryl, wherein said aryl and said heteroaryl are each optionally substituted with one or more groups R 6 ;
  • each R 6 is independently selected from Ci_ 5 alkyl, C 2 . 5 alkenyl, C 2 . 5 alkynyl, -(C 0 . 3 alkylene)-OH, -(C c . 3 alkylene)-0(C 1 - 5 alkyl), -(C 0 -3 alkylene)-0(C 1-5 alkylene)-OH, -(Co-3 alkylene)-0(Ci. 5 alkylene)-0(C 1-5 alkyl), -(C 0 .
  • R 1 is halogen, methoxy, ethoxy, or -CF 3 ; each R 2 is independently selected from Ci. 5 alkyl, C 2 _ 5 alkenyl, C 2 . 5 alkynyl, -(C 0 . 3 alkylene)-OH, -(C Q _ 3 alkylene)-0(Ci. 5 alkyl). -(C 0 . 3 alkylene)-0(Ci. 5 aIkylene)-OH, -(Co-3 alkylene)-0(Ci_ 5 alkylene)-0(d-5 alkyl), -(C 0-3 alkylene)-SH, -(C 0 .
  • each R 3 is independently selected from d. 5 alkyl, C 2 . 5 alkenyl, C 2 . 5 alkynyl, -(C Q .
  • R 4 is hydrogen or C 1-5 alkyl
  • R 5 is a covending bond or C 2 . 3 alkylene;
  • A is aryl or heteroaryl, wherein said aryl and said heteroaryl are each optionally substituted with one or more groups R 6 ;
  • each R 6 is independently selected from C 1-5 alkyl, C 2 .. 5 alkenyl, C 2 . 5 alkynyl, -(C 0 . 3 alkylene)-OH, -(C 0-3 alkylene)-0(C 1-5 alkyl), -(C 0-3 alkylene)-0(C -5 alkylene)-OH, -(Co-3 alkylene)-0(C 1 . 5 alkylene)-0(C 1 .
  • each R 2 is independently selected from d -5 alkyl, C 2 remedy 5 alkenyl, C 2 . 5 alkynyl, -OH, -0(C -5 alkyl), -0(C 1-5 alkylene)-OH, -0(C 1-5 alkylene)-0(Ci_ 5 alkyl), -SH, -S(d_ 5 alkyl), -S(C 1-5 alkylene)-SH, -S(d_ 5 aIkylene)-S(d_ 5 alkyl), -NH 2 , -NH(Ci. 5 alkyl), ⁇ N(C 1-5 a!ky!(d_5 alkyl), halogen, d-5 haloalkyl, -CF 3 , and -CN.
  • each R 3 is independently selected from d -5 alkyl, C 2 . 5 alkenyl, C 2 . 5 alkynyl, -OH, -0(d. 5 alkyl), -0(d_ 5 alkylene)-OH, -0(d_ 5 alkylene)-0(d_ 5 alkyl), -SH, -S(d_ 5 alkyl), -S(d-s alkylene)-SH, -S(d. 5 alkylene)-S(d. 5 alkyl), -NH 2 , -NH(d- 5 alkyl), -N(d_ 5 alkyl)(d. 5 alkyl), halogen, d- 5 haloalkyl, -CF 3 , and -CN.
  • each R 6 is independently selected from d_ 5 alkyl, C 2 . 5 alkenyl, C 2 . 5 alkynyl, -OH, -0(Ci. 5 alkyl),
  • a pharmaceutical composition comprising a compound as defined in any one of items 24 to 33 and a pharmaceutically acceptable excipient.
  • codon-optimized human MTH1 cDNA subcloned into a pETM-1 1 vector (Gunther Stier, EMBL) featuring a His-tag and TEV site was obtained from GenScript (GenScript, NJ, USA) and expressed in the E. coli strain BL21 DE3.
  • bacteria were lysed using buffer (50 mM Tris-HCI pH 7.5, 500 mM NaCI, 5% glycerol, 5 mM 3-mercaptoethanol, 1 mM PMSF) containing lysozyme (Sigma-Aldrich) and DNase I (Roche). His-tagged protein was purified with NiNTA agarose (Qiagen), washed with buffer, and eluted with an imidazole gradient. Following removal of the His-tag by incubation with TEV protease, fractions were dialyzed and purified using size-exclusion chromatography (Sephadex, GE Healthcare). Protein concentration of the purified fractions was determined by UV (A 280 ).
  • the DSF assay was performed as follows. Compounds were dissolved in dimethyl sulfoxide (DMSO) and diluted in assay buffer (10 mM HEPES-NaOH, pH 7.5, 500 mM NaCI, final concentration 10 ⁇ ). Upon addition of MTH 1 recombinant protein (final concentration 2 ⁇ ), SYPRO Orange (Sigma Aldrich) was added and samples were analysed in a qPCR machine as described in literature (Niesen, F. H. , Berglund, H. & Vedadi, M. , Nat. Protocols 2, 2212- 2221 (2007)). Tm shift values were determined by fitting a curve to the data points using nonlinear regression analysis.
  • a strong binding affinity to MTH1 as determined in the DSF assay is known to correlate with an inhibition of the catalytic activity of MTH 1 (Huber KV et aL , Nature, 2014, 508(7495):222-7).
  • MTH 1 hydrolyzes oxidized nucleotides such as 2-OH-dATP and 8-oxo-dGTP, yielding the corresponding nucleoside monophosphate and pyrophosphate (PPi).
  • PPi nucleoside monophosphate and pyrophosphate
  • MTH1 The activity of MTH1 is monitored using dGTP (Fermentas) using the PPiLight Inorganic Pyrophosphate Assay (Lonza Rockland Inc.). IC 50 values are determined using non-linear regression analysis utilizing GraphPad Prism Software.
  • the MTH1 catalytic assay can be performed as follows. Half-maximal inhibitory concentrations (IC 50 ) are determined using a luminescence-based assay as described previously (Svensson (201 1 ) FEBS Letters 585, 2617-2621 ) with some minor modifications. Briefly, serial dilutions of compounds are dissolved in assay buffer (100 mM Tris-acetate pH 7.5, 40 mM NaCI and 10mM Mg(OAc) 2 containing 0.005% Tween-20 and 2 mM dithiothreitol (DTT). Upon addition of MTH1 recombinant protein (final concentration 2 nM), plates are incubated on a plate shaker for 15 min at room temperature.
  • assay buffer 100 mM Tris-acetate pH 7.5, 40 mM NaCI and 10mM Mg(OAc) 2 containing 0.005% Tween-20 and 2 mM dithiothreitol (DTT).
  • PPi pyrophosphate
  • IC 50 values are determined by fitting a dose-response curve to the data points using nonlinear regression analysis using the GraphPad Prism software.
  • MTH1 levels are increased in Ras-expressing cancers ranging from lung cancer (Speina Journal of the National Cancer Institute, 2005, 97:384-95; Kennedy, FEBS Letters, 998, 429: 17-20) to renal carcinoma (Okamoto, Int J Cancer, 1996, 65:437-41), indicating that there is a connection between oncogenic transformation and oxidative stress. This is also supported by the fact that the MTH1 inhibitor SCH 51344 was shown to prevent the growth of fibroblasts infected with a variety of different oncogenes such as v-abl.
  • MTH1 deficiency in knockout mice confers a mild phenotype, indicating that there is a therapeutic window for MTH1 inhibitors (Tsuzuki, Proceedings of the National Academy of Sciences, 2001 , 98: 1 1456-61 ).
  • the compounds of formula (I) as described and defined herein can thus advantageously be used in the treatment or prevention of cancer.

Abstract

La présente invention concerne des dérivés de 2,3-dihydrocyclopenta[b]quinoléine de formule (I) ainsi que des compositions pharmaceutiques comprenant ces composés pour leur utilisation dans le traitement ou la prévention du cancer, et en particulier pour leur utilisation en tant qu'inhibiteurs de MTH1.
PCT/EP2016/053767 2015-02-23 2016-02-23 Dérivés de 2,3-dihydrocyclopenta[b]quinoléine en tant qu'inhibiteurs de mth1 pour la thérapie du cancer WO2016135139A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP15156188 2015-02-23
EP15156188.3 2015-02-23

Publications (1)

Publication Number Publication Date
WO2016135139A1 true WO2016135139A1 (fr) 2016-09-01

Family

ID=52544386

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2016/053767 WO2016135139A1 (fr) 2015-02-23 2016-02-23 Dérivés de 2,3-dihydrocyclopenta[b]quinoléine en tant qu'inhibiteurs de mth1 pour la thérapie du cancer

Country Status (1)

Country Link
WO (1) WO2016135139A1 (fr)

Citations (34)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3773919A (en) 1969-10-23 1973-11-20 Du Pont Polylactide-drug mixtures
EP0036676A1 (fr) 1978-03-24 1981-09-30 The Regents Of The University Of California Procédé de préparation de liposomes de taille identique et les liposomes ainsi obtenus
EP0052322A2 (fr) 1980-11-10 1982-05-26 Gersonde, Klaus, Prof. Dr. Méthode de préparation de vésicules lipidiques par traitement aux ultra-sons, utilisation de ce procédé et l'appareillage ainsi utilisé
EP0088046A2 (fr) 1982-02-17 1983-09-07 Ciba-Geigy Ag Lipides en phase aqueuse
DE3218121A1 (de) 1982-05-14 1983-11-17 Leskovar, Peter, Dr.-Ing., 8000 München Arzneimittel zur tumorbehandlung
EP0102324A2 (fr) 1982-07-29 1984-03-07 Ciba-Geigy Ag Lipides et composés tensio-actifs en phase aqueuse
US4485045A (en) 1981-07-06 1984-11-27 Research Corporation Synthetic phosphatidyl cholines useful in forming liposomes
JPS607934A (ja) 1983-06-29 1985-01-16 Dai Ichi Seiyaku Co Ltd リポソ−ムの製造方法
EP0133988A2 (fr) 1983-08-02 1985-03-13 Hoechst Aktiengesellschaft Préparations pharmaceutiques contenant des peptides régulateurs à libération retardée et procédé pour leur préparation
EP0142641A2 (fr) 1983-09-26 1985-05-29 Udo Dr. Ehrenfeld Moyen et produit pour le diagnostic et la thérapie de tumeurs ainsi que pour le traitement de déficiences du système immunitaire cellulaire et humoral
EP0143949A1 (fr) 1983-11-01 1985-06-12 TERUMO KABUSHIKI KAISHA trading as TERUMO CORPORATION Composition pharmaceutique contenant de l'urokinase
US4544545A (en) 1983-06-20 1985-10-01 Trustees University Of Massachusetts Liposomes containing modified cholesterol for organ targeting
EP0268871A1 (fr) 1986-10-31 1988-06-01 Sumitomo Pharmaceuticals Company, Limited Dérivés de quinoléine
WO1997041833A1 (fr) 1996-05-08 1997-11-13 Inhale Therapeutic Systems Compositions dispersibles a base de macromolecules, procedes de preparation et techniques d'utilisation
WO1999016419A1 (fr) 1997-09-29 1999-04-08 Inhale Therapeutic Systems, Inc. Microparticules perforees et procedes d'utilisation
WO2001064645A2 (fr) 2000-03-01 2001-09-07 Orion Corporation Derives de quinoline utilises comme antagonistes alpha2
WO2001085136A2 (fr) 2000-05-10 2001-11-15 Alliance Pharmaceutical Corporation Poudres a base de phospholipides permettant de distribuer un medicament
WO2003053411A1 (fr) 2001-12-19 2003-07-03 Nektar Therapeutics Administration pulmonaire d'aminoglycosides
EP1071752B1 (fr) 1998-04-21 2003-07-09 Micromet AG Polypeptides specifiques a cd19 et cd3 et leurs utilisations
US20040209902A1 (en) 2003-04-11 2004-10-21 Chu-Chung Lin Aminoquinoline compounds
WO2005120497A2 (fr) 2004-06-04 2005-12-22 The Regents Of The University Of California Composes intervenant dans l'acceleration du transport ionique par le cftr mutant, et utilisations desdits composes
WO2006041900A2 (fr) 2004-10-07 2006-04-20 Cytovia, Inc. N-aryl-1h-pyrazolo[3,4-b]quinolin-4-amines substitues et analogues utilises comme activateurs de caspases et inducteurs de l'apoptose
US20070148185A1 (en) 2005-10-14 2007-06-28 Dharmender Rathore Novel therapeutic target for protozoal diseases
WO2008021250A2 (fr) 2006-08-10 2008-02-21 Fred Hutchinson Cancer Research Center Compositions et procédés de modulation de l'apoptose dans des cellules surexprimant des protéines membres de la famille bcl-2
WO2008089307A2 (fr) 2007-01-18 2008-07-24 Lexicon Pharmaceuticals, Inc. Méthodes et compositions utilisés dans le traitement de la douleur, de l'inflammation et du cancer
WO2009001060A2 (fr) 2007-06-27 2008-12-31 Summit Corporation Plc Utilisation de composés pour la préparation d'agents anti-tuberculose
WO2009023558A1 (fr) 2007-08-10 2009-02-19 Vm Discovery Inc. Compositions et procédés pour modulateurs d'apoptose
US20090088420A1 (en) 2007-04-12 2009-04-02 University Of Southern California Compounds with hiv-1 integrase inhibitory activity and use thereof as anti-hiv/aids therapeutics
US20090163545A1 (en) 2007-12-21 2009-06-25 University Of Rochester Method For Altering The Lifespan Of Eukaryotic Organisms
WO2010039538A2 (fr) 2008-09-23 2010-04-08 Georgetown University Inhibiteur de flavivirus et procédés d'utilisation de ceux-ci
WO2010151737A2 (fr) 2009-06-25 2010-12-29 Amgen Inc. Composés hétérocycliques et leurs utilisations
CN103224466A (zh) 2013-04-15 2013-07-31 北京大学 具有β-分泌酶抑制功能的化合物及其制备方法与应用
WO2014033136A1 (fr) 2012-08-27 2014-03-06 Cemm - Research Center For Molecular Medicine Of The Austrian Academy Of Sciences Composés aminohétéroaryles en tant qu'inhibiteurs de mth1
WO2014084778A1 (fr) 2012-11-27 2014-06-05 Thomas Helledays Stiftelse För Medicinsk Forskning Dérivés de pyrimidine-2,4-diamine utilisables en vue du traitement du cancer

Patent Citations (34)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3773919A (en) 1969-10-23 1973-11-20 Du Pont Polylactide-drug mixtures
EP0036676A1 (fr) 1978-03-24 1981-09-30 The Regents Of The University Of California Procédé de préparation de liposomes de taille identique et les liposomes ainsi obtenus
EP0052322A2 (fr) 1980-11-10 1982-05-26 Gersonde, Klaus, Prof. Dr. Méthode de préparation de vésicules lipidiques par traitement aux ultra-sons, utilisation de ce procédé et l'appareillage ainsi utilisé
US4485045A (en) 1981-07-06 1984-11-27 Research Corporation Synthetic phosphatidyl cholines useful in forming liposomes
EP0088046A2 (fr) 1982-02-17 1983-09-07 Ciba-Geigy Ag Lipides en phase aqueuse
DE3218121A1 (de) 1982-05-14 1983-11-17 Leskovar, Peter, Dr.-Ing., 8000 München Arzneimittel zur tumorbehandlung
EP0102324A2 (fr) 1982-07-29 1984-03-07 Ciba-Geigy Ag Lipides et composés tensio-actifs en phase aqueuse
US4544545A (en) 1983-06-20 1985-10-01 Trustees University Of Massachusetts Liposomes containing modified cholesterol for organ targeting
JPS607934A (ja) 1983-06-29 1985-01-16 Dai Ichi Seiyaku Co Ltd リポソ−ムの製造方法
EP0133988A2 (fr) 1983-08-02 1985-03-13 Hoechst Aktiengesellschaft Préparations pharmaceutiques contenant des peptides régulateurs à libération retardée et procédé pour leur préparation
EP0142641A2 (fr) 1983-09-26 1985-05-29 Udo Dr. Ehrenfeld Moyen et produit pour le diagnostic et la thérapie de tumeurs ainsi que pour le traitement de déficiences du système immunitaire cellulaire et humoral
EP0143949A1 (fr) 1983-11-01 1985-06-12 TERUMO KABUSHIKI KAISHA trading as TERUMO CORPORATION Composition pharmaceutique contenant de l'urokinase
EP0268871A1 (fr) 1986-10-31 1988-06-01 Sumitomo Pharmaceuticals Company, Limited Dérivés de quinoléine
WO1997041833A1 (fr) 1996-05-08 1997-11-13 Inhale Therapeutic Systems Compositions dispersibles a base de macromolecules, procedes de preparation et techniques d'utilisation
WO1999016419A1 (fr) 1997-09-29 1999-04-08 Inhale Therapeutic Systems, Inc. Microparticules perforees et procedes d'utilisation
EP1071752B1 (fr) 1998-04-21 2003-07-09 Micromet AG Polypeptides specifiques a cd19 et cd3 et leurs utilisations
WO2001064645A2 (fr) 2000-03-01 2001-09-07 Orion Corporation Derives de quinoline utilises comme antagonistes alpha2
WO2001085136A2 (fr) 2000-05-10 2001-11-15 Alliance Pharmaceutical Corporation Poudres a base de phospholipides permettant de distribuer un medicament
WO2003053411A1 (fr) 2001-12-19 2003-07-03 Nektar Therapeutics Administration pulmonaire d'aminoglycosides
US20040209902A1 (en) 2003-04-11 2004-10-21 Chu-Chung Lin Aminoquinoline compounds
WO2005120497A2 (fr) 2004-06-04 2005-12-22 The Regents Of The University Of California Composes intervenant dans l'acceleration du transport ionique par le cftr mutant, et utilisations desdits composes
WO2006041900A2 (fr) 2004-10-07 2006-04-20 Cytovia, Inc. N-aryl-1h-pyrazolo[3,4-b]quinolin-4-amines substitues et analogues utilises comme activateurs de caspases et inducteurs de l'apoptose
US20070148185A1 (en) 2005-10-14 2007-06-28 Dharmender Rathore Novel therapeutic target for protozoal diseases
WO2008021250A2 (fr) 2006-08-10 2008-02-21 Fred Hutchinson Cancer Research Center Compositions et procédés de modulation de l'apoptose dans des cellules surexprimant des protéines membres de la famille bcl-2
WO2008089307A2 (fr) 2007-01-18 2008-07-24 Lexicon Pharmaceuticals, Inc. Méthodes et compositions utilisés dans le traitement de la douleur, de l'inflammation et du cancer
US20090088420A1 (en) 2007-04-12 2009-04-02 University Of Southern California Compounds with hiv-1 integrase inhibitory activity and use thereof as anti-hiv/aids therapeutics
WO2009001060A2 (fr) 2007-06-27 2008-12-31 Summit Corporation Plc Utilisation de composés pour la préparation d'agents anti-tuberculose
WO2009023558A1 (fr) 2007-08-10 2009-02-19 Vm Discovery Inc. Compositions et procédés pour modulateurs d'apoptose
US20090163545A1 (en) 2007-12-21 2009-06-25 University Of Rochester Method For Altering The Lifespan Of Eukaryotic Organisms
WO2010039538A2 (fr) 2008-09-23 2010-04-08 Georgetown University Inhibiteur de flavivirus et procédés d'utilisation de ceux-ci
WO2010151737A2 (fr) 2009-06-25 2010-12-29 Amgen Inc. Composés hétérocycliques et leurs utilisations
WO2014033136A1 (fr) 2012-08-27 2014-03-06 Cemm - Research Center For Molecular Medicine Of The Austrian Academy Of Sciences Composés aminohétéroaryles en tant qu'inhibiteurs de mth1
WO2014084778A1 (fr) 2012-11-27 2014-06-05 Thomas Helledays Stiftelse För Medicinsk Forskning Dérivés de pyrimidine-2,4-diamine utilisables en vue du traitement du cancer
CN103224466A (zh) 2013-04-15 2013-07-31 北京大学 具有β-分泌酶抑制功能的化合物及其制备方法与应用

Non-Patent Citations (59)

* Cited by examiner, † Cited by third party
Title
"Remington: The Science and Practice of Pharmacy", PHARMACEUTICAL PRESS
ABOUZID K ET AL., BIOORG MED CHEM, vol. 16, no. 16, 2008, pages 7543 - 7551
ALII, CANCER RES, vol. 69, no. 8, 2009, pages 3589 - 3596
ATZRODT J ET AL., BIOORG MED CHEM, vol. 20, no. 18, 2012, pages 5658 - 5667
BLATTMAN JN ET AL., SCIENCE, vol. 305, no. 5681, 2004, pages 200 - 205
BORROK MJ ET AL., J AM CHEM SOC, vol. 129, no. 42, 2007, pages 12780 - 12785
BRAUTIGAM L ET AL., CANCER RES, 2016
BUNDGAARD, H.: "Design of Prodrugs", 1985, ELSEVIER, pages: 7 - 9,21-24
BURDI NZ, BIOL DEISTVIE PROD ORG SINT PRIR SOEDIN, 1978, pages 3 - 7
BURDI, N. Z.: "Antiinflammatory activity and structure-activity relation of 4-amino-2,3-trimethylene-quinoline derivatives", BIOL. DEISTVIE PROD. ORG. SINT. PRIR. SOEDIN. , 3-7. EDITOR(S): PIDEMSKII, E. L. PUBLISHER: PERMSKII UNIV., PERM, USSR. CODEN: 41LJA7, 1978 *
CALLAHAN MK ET AL., J LEUKOC BIOL, vol. 94, no. 1, 2013, pages 41 - 53
CANCER DISCOVERY, vol. 4, 2014, pages 631
CHO, RNA BIOLOGY, vol. 8, no. 1, 2011, pages 125 - 131
DATABASE CA [online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; 12 May 1984 (1984-05-12), BURDI, N. Z.: "Antiinflammatory activity and structure-activity relation of 4-amino-2,3-trimethylene-quinoline derivatives", XP002742780, retrieved from STN Database accession no. 1979:551096 *
EBERHARD, CLIN ONCOL, vol. 26, no. 6, 2008, pages 983 - 993
EGASHIRA A ET AL., DNA REPAIR, vol. 1, no. 11, 2002, pages 881 - 893
EPSTEIN ET AL., PROC. NATI. ACAD. SCI. (USA, vol. 82, 1985, pages 3688 - 3692
GAD H ET AL., NATURE, vol. 508, no. 7495, 2014, pages 215 - 221
GERRY, MOL BIOL., vol. 292, no. 2, 1999, pages 251 - 262
HUBER KV ET AL., NATURE, vol. 508, no. 7495, 2014, pages 222 - 227
HWANG ET AL., PROC. NATL. ACAD. SCI. (USA, vol. 77, 1980, pages 4030 - 4034
INTLEKOFER AM ET AL., J LEUKOC BIOL, vol. 94, no. 1, 2013, pages 25 - 39
JEMAL A ET AL., CA CANCER J CLIN, vol. 61, no. 2, 2011, pages 69 - 90
K. MASTERS: "Spray Drying Handbook", 1991, JOHN WILEY & SONS, INC.
KENNEDY, FEBS LETT., vol. 429, no. 1, 1998, pages 17 - 20
KENNEDY, FEBS LETTERS, vol. 429, 1998, pages 17 - 20
KEOHAVONG, CLIN CANCER RES., vol. 2, no. 2, 1996, pages 411 - 418
KILIAN V M HUBER ET AL: "Stereospecific targeting of MTH1 by (S)-crizotinib as an anticancer strategy", NATURE, NATURE PUBLISHING GROUP, UNITED KINGDOM, vol. 508, no. 7495, 10 April 2014 (2014-04-10), pages 222 - 227, XP002739608, ISSN: 0028-0836, [retrieved on 20140402], DOI: 10.1038/NATURE13194 *
KYI C ET AL., FEBS LETT, vol. 588, no. 2, 2014, pages 368 - 376
LE PAGE, CANCER RES, vol. 60, no. 19, 2000, pages 5548 - 5552
LEE AY ET AL., SCIENCE, vol. 344, no. 6180, 2014, pages 208 - 211
LIU P ET AL., EUR J MED CHEM, vol. 79, 2014, pages 413 - 421
MATHERS C ET AL., THE GLOBAL BURDEN OF DISEASE: 2004 UPDATE, WORLD HEALTH ORGANIZATION, 2008
MODVIG A ET AL., J ORG CHEM, vol. 79, 2014, pages 5861 - 5868
MORENO E ET AL., EUR J MED CHEM, vol. 47, no. 1, 2012, pages 283 - 298
NAKABEPPU, MUTATION RESEARCH/GENETIC TOXICOLOGY AND ENVIRONMENTAL MUTAGENESIS, vol. 703, no. 1, 2010, pages 51 - 58
NASR M ET AL., J MED CHEM, vol. 21, no. 3, 1978, pages 295 - 298
NGIOW SF ET AL., CANCER RES, vol. 71, no. 21, 2011, pages 6567 - 6571
NIESEN, F.H.; BERGLUND, H.; VEDADI, M., NAT. PROTOCOLS, vol. 2, 2007, pages 2212 - 2221
OKAMOTO, INT J CANCER, vol. 65, 1996, pages 437 - 441
PEDEMONTE N ET AL., MOL PHARMACOL, vol. 67, no. 5, 2005, pages 1797 - 1807
R. LANGER ET AL., J. BIOMED. MATER. RES., vol. 15, 1981, pages 167 - 277
R. LANGER, CHEM. TECH., vol. 12, 1982, pages 98 - 105
RAI P ET AL., ONCOGENE, vol. 30, no. 12, 2011, pages 1489 - 1496
RAI, MUTATION RESEARCH/GENETIC TOXICOLOGY AND ENVIRONMENTAL MUTAGENESIS, vol. 703, no. 1, 2010, pages 71 - 81
RAI, ONCOGENE, vol. 30, no. 12, 2011, pages 1489 - 1496
RAI, PROTEOMICS, vol. 3, no. 8, 2003, pages 1454 - 1463
ROSINI M ET AL., BIOORG MED CHEM, vol. 14, no. 23, 2006, pages 7846 - 7853
ROXANA A., MAEDICA, vol. 1, no. 1, 2006, pages 63 - 65
SIDMAN, U. ET AL., BIOPOLYMERS, vol. 22, 1983, pages 547 - 556
SPEICHER, NAT REV GENET, vol. 6, no. 10, 2005, pages 782 - 792
SPEINA JOURNAL OF THE NATIONAL CANCER INSTITUTE, vol. 97, 2005, pages 384 - 395
SVENSSON, FEBS LETTERS, vol. 585, 2011, pages 2617 - 2621
TAYLOR PC, CURR OPIN PHARMACOL., vol. 3, no. 3, 2003, pages 323 - 328
TAYLOR PC: "Antibody therapy for rheumatoid arthritis", CURR OPIN PHARMACOL., vol. 3, no. 3, 2003, pages 323 - 328
TOBE M ET AL., BIOORG MED CHEM LETT, vol. 11, no. 4, 2001, pages 545 - 548
TSUZUKI, PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES, vol. 98, 2001, pages 11456 - 11461
TSUZUKI, PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES, vol. 98, no. 20, 2001, pages 11456 - 11461
WILLIAM JS ET AL., JOURNAL OF LABELLED COMPOUNDS AND RADIOPHARMACEUTICALS, vol. 53, no. 11-12, 2010, pages 635 - 644

Similar Documents

Publication Publication Date Title
CN107108637B (zh) 三唑并嘧啶化合物及其用途
TWI699363B (zh) 四氫-吡啶并[3,4-b]吲哚雌激素受體調節劑及其用途
CN106459034B (zh) 作为成纤维细胞生长因子受体抑制剂的喹诺酮衍生物
US10702518B2 (en) TAF1 inhibitors for the therapy of cancer
JP2018524382A (ja) キラルジアリール大環状分子及びその使用
KR20130045341A (ko) 암 치료를 위한 저산소증 활성 프로드러그와 항혈관형성제의 투여
EP3386997B1 (fr) Composés de platine fonctionnalisés par monomaléimide pour la thérapie du cancer
CN112672744B (zh) 抑制二氢乳清酸脱氢酶的方法和组合物
EP2566851B1 (fr) Analogues de la combretastatin à être utilisés dans le traitement du cancer
TW201526896A (zh) 藉二氫吡并吡組合療法的癌症治療方法
EP3035939B1 (fr) Antagonistes v1a pour traiter des troubles du sommeil à décalage de phase
KR20210027382A (ko) 접히지 않은 단백질 반응의 활성화제
US20220048863A1 (en) Dna2 inhibitors for cancer treatment
AU2020360709B2 (en) Heterocyclic derivatives, pharmaceutical compositions and their use in the treatment or amelioration of cancer
AU2018352382A1 (en) Compounds and methods for treating cancer
WO2016135138A1 (fr) Dérivés d'oxoquinoléine en tant qu'inhibiteurs de mth1 pour la thérapie du cancer
WO2016135140A1 (fr) Dérivés de 4-aminoquinazoline en tant qu'inhibiteurs de mth1 pour la thérapie du cancer
WO2016135139A1 (fr) Dérivés de 2,3-dihydrocyclopenta[b]quinoléine en tant qu'inhibiteurs de mth1 pour la thérapie du cancer
AU2022243600A1 (en) Alk-5 inhibitors and uses thereof
WO2016135137A1 (fr) Dérivés de 4-(phénylamino)quinoléine substitués en tant qu'inhibiteurs de mth1 pour la thérapie du cancer
WO2021064141A1 (fr) Inhibiteurs de la kinase 1b régulée par phosphorylation de tyrosine à double spécificité
CN108348611A (zh) 使用pi3k抑制剂和mdm2抑制剂的联合疗法
WO2021046220A1 (fr) Composés et méthodes de traitement du cancer
EP3878841A1 (fr) Inhibiteur d'indazole kinase et son utilisation
WO2021046178A1 (fr) Composés et méthodes de traitement du cancer

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: 16709711

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: 16709711

Country of ref document: EP

Kind code of ref document: A1