WO2022189618A1 - Nitrogen-containing heterocycles as radiosensitizers - Google Patents

Nitrogen-containing heterocycles as radiosensitizers Download PDF

Info

Publication number
WO2022189618A1
WO2022189618A1 PCT/EP2022/056316 EP2022056316W WO2022189618A1 WO 2022189618 A1 WO2022189618 A1 WO 2022189618A1 EP 2022056316 W EP2022056316 W EP 2022056316W WO 2022189618 A1 WO2022189618 A1 WO 2022189618A1
Authority
WO
WIPO (PCT)
Prior art keywords
alkyl
formula
group
compound
carcinoma
Prior art date
Application number
PCT/EP2022/056316
Other languages
French (fr)
Inventor
Frédérique MEGNIN-CHANET
Florent POYER
Sandrine Piguel
Pierre Verrelle
Marie-Paule Teulade-Fichou
Elaine DEL NERY
Original Assignee
Institut Curie
Centre National De La Recherche Scientifique
Institut National De La Sante Et De La Recherche Medicale (Inserm)
Universite Paris-Saclay
Université Clermont Auvergne
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 Institut Curie, Centre National De La Recherche Scientifique, Institut National De La Sante Et De La Recherche Medicale (Inserm), Universite Paris-Saclay, Université Clermont Auvergne filed Critical Institut Curie
Publication of WO2022189618A1 publication Critical patent/WO2022189618A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic 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
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4418Non condensed pyridines; Hydrogenated derivatives thereof having a carbocyclic group directly attached to the heterocyclic ring, e.g. cyproheptadine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/506Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents

Definitions

  • the present invention concerns nitrogen-containing heterocycles as radiosensitizers, in particular for cancer treatment.
  • the present invention also relates to these nitrogen-containing heterocycles for use for the treatment of cancer, in combination with radiotherapy or in combination with radiotherapy and any anticancer drug.
  • Some tumours are treated in first line with radiotherapy or with chemoradiotherapy.
  • the challenge is to deliver a cure dose to the tumor while limiting the possibility of serious damages to normal tissues.
  • the balance between tumour control and the risk of normal tissue side-effects is given by the therapeutic index of the treatment. Improving the therapeutic index by modulating the tumour/normal tissue response is challenging.
  • small molecules could be used as radiosensitizing agents (radiosensitizers) in order to increase the radiosensitivity of the tumours.
  • the main characteristics of a radiosensitizer should be a low toxicity without ionizing radiations (healthy tissue) and an enhanced injury to the tumour in presence of ionizing radiations.
  • the expectation of getting a supra-additive antitumor effect greater than expected based on the simple addition of each cytotoxic used alone) with minimal secondary effects on healthy tissues is of importance in cancer therapy.
  • the aim of the present invention is thus to provide non-toxic nitrogen- containing heterocycles for use as radiosensitizers for the treatment of cancer in combination with radiotherapy or in combination with radiotherapy and any anticancer drug.
  • the present invention relates to a compound having the following formula (I): wherein:
  • Ri is H or is selected from the group consisting of: halogen, (Ci-C 6 )alkyl, aryl groups, and heteroaryl groups, and
  • R a and R b are independently H or a (Ci-C 6 )alkyl group; or a group of formula (III): wherein:
  • R2 is H or is selected from the group consisting of: halogen, (Ci-Ce)alkyl, aryl groups, and heteroaryl groups,
  • R is H or is selected from the group consisting of: halogen, (Ci-Ce)alkyl, aryl groups, and heteroaryl groups, said aryl and heteroaryl groups being optionally substituted with one or several substituents independently selected from: amino, hydroxyl, thiol (-SH), halogen, (Ci- C 6 )alkyl, (Ci-Ce)alkoxy, (Ci-Ce)alkylthio, (Ci-C 6 )alkylamino, halo(Ci-Ce)alkyl, carboxyl and carboxy(Ci-Ce)alkyl; .
  • R 3 is selected from the group consisting of: H, halogen, (Ci-C 6 )alkyl, aryl groups, and heteroaryl groups; said aryl and heteroaryl groups being optionally substituted with one or several substituents independently selected from: amino, hydroxyl, thiol (-SH), halogen, (Ci- C 6 )alkyl, (Ci-C 6 )alkoxy, (Ci-C 6 )alkylthio, (Ci-C 6 )alkylamino, halo(Ci-C 6 )alkyl, carboxyl and carboxy(Ci-C 6 )alkyl;
  • R 5 is chosen from the group consisting of: H, (Ci-C 6 )alkyl, and aryl, preferably phenyl, said aryl group being optionally substituted with one or several substituents independently selected from: amino, hydroxyl, thiol, halogen, (Ci- C 6 )alkyl, (Ci-C 6 )alkoxy, (Ci-C 6 )alkylthio, (Ci-C 6 )alkylamino, halo(Ci-C 6 )alkyl, carboxyl and carboxy(Ci-C 6 )alkyl;
  • R 7 groups are chosen from the group consisting of: H, halogen, and (Ci-C 6 )alkyl; or a pharmaceutically acceptable salt thereof, for use for the treatment of cancer as radiosensitizer.
  • the present invention thus concerns new radiosensitizers, preferably used in combination with radiation as explained later.
  • C t -C z means a carbon-based chain which can have from t to z carbon atoms, for example C 1 -C 3 means a carbon- based chain which can have from 1 to 3 carbon atoms.
  • halogen means: a fluorine, a chlorine, a bromine or an iodine.
  • alkyl means: a linear or branched, saturated, hydrocarbon-based aliphatic group comprising, unless otherwise mentioned, from 1 to 6 carbon atoms.
  • alkyl means: a linear or branched, saturated, hydrocarbon-based aliphatic group comprising, unless otherwise mentioned, from 1 to 6 carbon atoms.
  • alkoxy means: an -O-alkyl radical where the alkyl group is as previously defined.
  • alkyl group is as previously defined.
  • -0-(Ci-C 4 )alkyl groups and in particular the -O-methyl group, the -O-ethyl group, as -0-C 3 alkyl group, the -O-propyl group, the -O-isopropyl group, and as -0-C 4 alkyl group, the -O-butyl, -O-isobutyl or -O-tert-butyl group.
  • alkylamino means: an -NH-alkyl group, the alkyl group being as defined above.
  • alkylthio means: an -S-alkyl group, the alkyl group being as defined above.
  • haloalkyl means: an alkyl group as defined above, in which one or more of the hydrogen atoms is (are) replaced with a halogen atom.
  • fluoroalkyls in particular CF 3 or CHF 2 .
  • aryl means: a cyclic aromatic group comprising between 6 and 10 carbon atoms.
  • aryl groups mention may be made of phenyl or naphthyl groups.
  • heteroaryl means: a 5- to 10-membered aromatic monocyclic or bicyclic group containing from 1 to 4 heteroatoms selected from O, S or N.
  • heteroaryl means: a 5- to 10-membered aromatic monocyclic or bicyclic group containing from 1 to 4 heteroatoms selected from O, S or N.
  • heteroaryl comprising 5 to 6 atoms, including 1 to 4 nitrogen atoms
  • carboxyalkyl means: an FIOOC- alkyl- group, the alkyl group being as defined above.
  • carboxyalkyl groups mention may in particular be made of carboxymethyl or carboxyethyl.
  • carboxyl means: a COOFI group.
  • alkylcarbonyl means a -CO-alkyl group, the alkyl group being as defined above.
  • alkoxylcarbonyl means a -CO-O-alkyl group, the alkyl group being as defined above.
  • the radiosensitizers according to the present invention have the formula (I) as mentioned above.
  • a family of radiosensitizers according to the present invention consists of compounds having the formula (I) as defined above, wherein R is a group of formula (II). Such compounds have the below formula (1-1):
  • R a , R b , Ri, R 5 , R7, and i being as defined above in formula (I).
  • a preferred family of radiosensitizers according to the present invention consists of compounds having the formula (1-1), wherein R a and R b are H.
  • Another preferred family of radiosensitizers according to the present invention consists of compounds having the formula (1-1), wherein Ri is H.
  • a preferred family of radiosensitizers according to the present invention consists of compounds having the formula (1-1), wherein R a and R b are H and Ri is H.
  • a family of radiosensitizers according to the present invention consists of compounds having the formula (I) as defined above, wherein R is a group of formula (III). Such compounds have the below formula (I-2):
  • R 2 , R 3 , R4, R 5 , R7, and i being as defined above in formula (I).
  • a preferred family of radiosensitizers according to the present invention consists of compounds having the formula (1-2), wherein R4 is H.
  • radiosensitizers consists of compounds having the formula (1-2), wherein R 3 is H or a (Ci-Ce)alkyl group.
  • a preferred family of radiosensitizers according to the present invention consists of compounds having the formula (1-2), wherein R 2 is halogen, such as Br, or an aryl group, such as phenyl.
  • R 5 is an aryl group, substituted or not.
  • the compounds for the use according to the present invention have the following formula (IV): wherein:
  • R6 is chosen from the group consisting of: amino, hydroxyl, thiol, halogen, (Ci-C6)alkyl, (Ci-C6)alkoxy, (Ci-C6)alkylthio, (Ci-C6)alkylamino, halo(Ci-C6)alkyl, carboxyl and carboxy(Ci-C6)alkyl.
  • the compounds of formula (IV) correspond to compounds having the formula (I) as defined above, wherein R 5 is a phenyl group, optionally substituted by a R6 group.
  • a family of radiosensitizers according to the present invention consists of compounds having the formula (IV) as defined above, wherein R is a group of formula (II). Such compounds have the below formula (IV-1):
  • R a , R b , Ri, R7, and i being as defined above in formula (I), and R 6 being as defined above in formula (IV).
  • a preferred family of radiosensitizers according to the present invention consists of compounds having the formula (IV-1), wherein R a and R b are H.
  • Another preferred family of radiosensitizers according to the present invention consists of compounds having the formula (IV-1), wherein Ri is H.
  • a preferred family of radiosensitizers according to the present invention consists of compounds having the formula (IV-1), wherein R a and R b are H and Ri is H.
  • a family of radiosensitizers according to the present invention consists of compounds having the formula (IV) as defined above, wherein R is a group of formula (III). Such compounds have the below formula (IV-2): defined above in formula (IV).
  • a preferred family of radiosensitizers according to the present invention consists of compounds having the formula (IV-2), wherein FU is H.
  • Another preferred family of radiosensitizers according to the present invention consists of compounds having the formula (IV-2), wherein R 3 is H or a (Ci-C 6 )alkyl group.
  • a preferred family of radiosensitizers according to the present invention consists of compounds having the formula (IV-2), wherein R 2 is halogen, such as Br, or an aryl group, such as phenyl.
  • the compounds for the use according to the present invention have the following formula (IV), wherein R 6 is chosen from the group consisting of: hydroxyl, halogen, in particular F, (Ci-Ce)alkoxy, in particular OMe, and halo(Ci-Ce)alkyl, in particular CF 3 , preferably in para position.
  • the compounds for the use according to the present invention have the following formula (IV-1) or (IV-2), wherein R 6 is chosen from the group consisting of: hydroxyl, halogen, in particular F, (Ci-Ce)alkoxy, in particular OMe, and halo(Ci-Ce)alkyl, in particular CF 3 , preferably in para position.
  • Preferred groups R 6 are OH, F, OMe, and CF 3 , and are preferably in para position.
  • a preferred family of radiosensitizers according to the present invention consists of compounds having the formula (IV), wherein R 6 is halogen in para position, and preferably R 6 is F in para position.
  • a preferred family of radiosensitizers according to the present invention consists of compounds having the formula (IV-1) or (IV-2), wherein R 6 is halogen in para position, and preferably R 6 is F in para position.
  • the compound for the use according to the invention has the following formula (IV-3):
  • R, R , and i being as defined in formula (I), and R 6 being as defined in formula
  • a particular family of radiosensitizers according to the present invention consists of compounds having the below formula (IV-3-1 ):
  • R a , R b , Ri, R7, and i being as defined above in formula (I), and R 6 being as defined above in formula (IV).
  • a preferred family of radiosensitizers according to the present invention consists of compounds having the formula (IV-3-1), wherein R a and R b are H.
  • Another preferred family of radiosensitizers according to the present invention consists of compounds having the formula (IV-3-1), wherein Ri is H.
  • a preferred family of radiosensitizers according to the present invention consists of compounds having the formula (IV-3-1), wherein R a and R b are H and Ri is H.
  • a particular family of radiosensitizers according to the present invention consists of compounds having the below formula (IV-3-2):
  • R 2 , R3, R4, R7, and i being as defined above in formula (I), and R 6 being as defined above in formula (IV).
  • a preferred family of radiosensitizers according to the present invention consists of compounds having the formula (IV-3-2), wherein FU is H.
  • radiosensitizers consists of compounds having the formula (IV-3-2), wherein R 3 is H or a (Ci-C 6 )alkyl group.
  • a preferred family of radiosensitizers according to the present invention consists of compounds having the formula (IV-3-2), wherein R 2 is halogen, such as Br, or an aryl group, such as phenyl.
  • the compounds for the use according to the invention have the following formula (IV-4):
  • R, R , and R 6 being as defined above.
  • R 7 is F.
  • a family of radiosensitizers according to the present invention consists of compounds having the formula (IV-4) as defined above, wherein R is a group of formula (II). Such compounds have the below formula (IV-4-1 ):
  • R a , R b , Ri, and R being as defined above in formula (I), and R 6 being as defined above in formula (IV).
  • a preferred family of radiosensitizers according to the present invention consists of compounds having the formula (IV-4-1), wherein R a and R b are H.
  • Another preferred family of radiosensitizers according to the present invention consists of compounds having the formula (IV-4-1), wherein Ri is H.
  • a preferred family of radiosensitizers according to the present invention consists of compounds having the formula (IV-4-1), wherein R a and R b are H and Ri is H.
  • a family of radiosensitizers according to the present invention consists of compounds having the formula (IV-4) as defined above, wherein R is a group of formula (III). Such compounds have the below formula (IV-4-2):
  • R 2 , R3, R4, and R being as defined above in formula (I), and R6 being as defined above in formula (IV).
  • a preferred family of radiosensitizers according to the present invention consists of compounds having the formula (IV-4-2), wherein R4 is H.
  • Another preferred family of radiosensitizers according to the present invention consists of compounds having the formula (IV-4-2), wherein R 3 is H or a (Ci-Ce)alkyl group.
  • a preferred family of radiosensitizers according to the present invention consists of compounds having the formula (IV-4-2), wherein R 2 is halogen, such as Br, or an aryl group, such as phenyl.
  • the compounds for the use according to the invention have the following formula (IV-5):
  • R, R ? , and R6 being as defined above.
  • R7 is F.
  • a family of radiosensitizers according to the present invention consists of compounds having the formula (IV-5) as defined above, wherein R is a group of formula (II). Such compounds have the below formula (IV-5-1 ):
  • R a , R b , Ri, R7, and R6 being as defined above.
  • a preferred family of radiosensitizers according to the present invention consists of compounds having the formula (IV-5-1), wherein R a and R b are H.
  • Another preferred family of radiosensitizers according to the present invention consists of compounds having the formula (IV-5-1), wherein Ri is H.
  • a preferred family of radiosensitizers according to the present invention consists of compounds having the formula (IV-5-1), wherein R a and R b are H and Ri is H.
  • a family of radiosensitizers according to the present invention consists of compounds having the formula (IV-5) as defined above, wherein R is a group of formula (III). Such compounds have the below formula (IV-5-2): (IV-5-2)
  • R 2 , R3, R4, R7, and R6 being as defined above.
  • a preferred family of radiosensitizers according to the present invention consists of compounds having the formula (IV-5-2), wherein R4 is H.
  • Another preferred family of radiosensitizers according to the present invention consists of compounds having the formula (IV-5-2), wherein R 3 is H or a (Ci-Ce)alkyl group.
  • a preferred family of radiosensitizers according to the present invention consists of compounds having the formula (IV-5-2), wherein R 2 is halogen, such as Br, or an aryl group, such as phenyl.
  • the compound for the use according to the present invention has the following formula (V):
  • the compound for the use according to the present invention has the following formula (VI):
  • a preferred family of radiosensitizers according to the present invention consists of compounds having the formula (VI), wherein R a and R b are H.
  • Another preferred family of radiosensitizers according to the present invention consists of compounds having the formula (VI), wherein Ri is H.
  • the compound for the use according to the present invention has the following formula (VII):
  • R 2 , R 3 and R 4 being as defined above.
  • a preferred family of radiosensitizers according to the present invention consists of compounds having the formula (VII), wherein FU is H.
  • Another preferred family of radiosensitizers according to the present invention consists of compounds having the formula (VII), wherein R 3 is H or a (Ci-Ce)alkyl group.
  • a preferred family of radiosensitizers according to the present invention consists of compounds having the formula (VII), wherein R 2 is halogen, such as Br, or an aryl group, such as phenyl.
  • the compound for the use according to the invention is one of the following compounds:
  • the present invention also relates to a conjugate molecule comprising one or more compound(s) according to the invention, having the formula (I) as defined above, said compound(s) being covalently bound to at least one cell binding agent.
  • the present invention also relates to a conjugate molecule comprising one or more compound(s) according to the invention, having the formula (1-1), (1-1), (IV), (IV-1), (IV-2), (IV-3), (IV-3-1 ), (IV-3-2), (IV-4), (IV-4-1), (IV-4-2), (IV-5), (IV-5-1), (IV- 5-2), (v), (VI) or (VII) as defined above, said compound(s) being covalently bound to at least one cell binding agent.
  • the present invention also relates to a conjugate molecule comprising one or more compound(s) according to the invention, having the formula (1), (2), (3) or (4) as defined above, said compound(s) being covalently bound to at least one cell binding agent.
  • the present invention also relates to a conjugate molecule being a compound according to the invention to which is bound to at least one payload, optionally through a linker or covalently conjugated, thereby forming a single molecule.
  • Suitable payloads include, but are not limited to, peptides, polypeptides, proteins, antibodies, or antigen-binding fragments thereof, antigens, nucleic acid molecules, polymers, small molecules, mimetic agents, drugs, inorganic molecules, organic molecules, and radioisotopes.
  • Suitable payloads include, but are not limited to, cell binding agents, chemotherapeutic agents, targeted therapy agents, cytotoxic agents, ligands for cellular receptor(s), immunomodulatory agents, pro-apoptotic agents, anti- angiogenic agents, photodetectable labels, contrast agents, radiolabels, and the like.
  • a cell binding agent is a molecule with affinity for a biological target.
  • the cell binding agent may be, for example, a ligand, a protein, an antibody, more particularly a monoclonal antibody, a protein or antibody fragment, a peptide, an oligonucleotide or an oligosaccharide.
  • the function of the binding agent is to direct the biologically active compound towards the biological target.
  • the conjugate of the invention is an antibody drug conjugate (ADC).
  • the payload is an antibody, particularly monoclonal antibody, sdAb, VHH, intrabody, or single-chain antibodies (scFv).
  • the conjugate of the invention comprises a linker unit between the compound of the invention and the payload.
  • the linker is cleavable under intracellular conditions, such that cleavage of the linker releases the compound of the invention from the payload in the intracellular environment.
  • the linker unit is not cleavable and the compound of the invention is released, for example, by payload degradation.
  • the linker is cleavable by a cleaving agent that is present in the intracellular environment (e.g., within a lysosome or endosome or caveolea).
  • the linker can be, e.g., a peptidyl linker that is cleaved by an intracellular peptidase or protease enzyme, including, but not limited to, a lysosomal or endosomal protease.
  • the cleavable linker is pH-sensitive, i.e., sensitive to hydrolysis at certain pH values.
  • the pH-sensitive linker hydrolyzable under acidic conditions.
  • an acid-labile linker that is hydrolyzable in the lysosome e.g., a hydrazone, semicarbazone, thiosemicarbazone, cis-aconitic amide, orthoester, acetal, ketal, or the like
  • an acid-labile linker that is hydrolyzable in the lysosome e.g., a hydrazone, semicarbazone, thiosemicarbazone, cis-aconitic amide, orthoester, acetal, ketal, or the like
  • Such linkers are relatively stable under neutral pH conditions, such as those in the blood, but are unstable at below pH 5.5 or 5.0, the approximate pH of the lysosome.
  • the linker is cleavable under reducing conditions (e.g., a disulfide linker).
  • a disulfide linker e.g., a disulfide linker.
  • disulfide linkers are known in the art, including, for example, those that can be formed using SATA (N-succinimidyl-S- acetylthioacetate), SPDP (N-succinimidyl-3-(2-pyridyldithio)propionate), SPDB (N- succinimidyl-3-(2-pyridyldithio)butyrate) and SMPT (N-succinimidyl-oxycarbonyl- alpha-methyl-alpha-(2-pyridyl-dithio)toluene), SPDB and SMPT.
  • SATA N-succinimidyl-S- acetylthioacetate
  • SPDP N-succinimidyl-3
  • the present invention also relates to the conjugate as mentioned above for use as drug.
  • the present invention also relates to a medicament comprising the conjugate as defined above.
  • the present invention also relates to a pharmaceutical composition, comprising the conjugate as defined above, and also at least one pharmaceutically acceptable excipient.
  • compositions contain an effective dose of at least one compound according to the invention, or a pharmaceutically acceptable salt, and also at least one pharmaceutically acceptable excipient.
  • Said excipients are selected, according to the pharmaceutical form and the mode of administration desired, from the usual excipients which are known to those skilled in the art.
  • compositions of the present invention for oral, sublingual, subcutaneous, intramuscular, intravenous, topical, local, intratracheal, intranasal, transdermal or rectal administration the active ingredient of formula (I) above, or the salt thereof, can be administered in unit administration form, as a mixture with conventional pharmaceutical excipients, to animals and to human beings for the treatment of the disorders and diseases below.
  • the suitable unit administration forms include oral forms such as tablets, soft or hard gel capsules, powders, granules and oral solutions or suspensions, sublingual, buccal, intratracheal, intraocular and intranasal administration forms, forms for administration by inhalation, topical, transdermal, subcutaneous, intramuscular or intravenous administration forms, rectal administration forms, and implants.
  • oral forms such as tablets, soft or hard gel capsules, powders, granules and oral solutions or suspensions
  • sublingual, buccal, intratracheal intraocular and intranasal administration forms, forms for administration by inhalation
  • topical, transdermal, subcutaneous, intramuscular or intravenous administration forms rectal administration forms, and implants.
  • the compounds according to the invention can be used in creams, gels, ointments or lotions.
  • the present invention also relates to a compound of formula (I) as defined above or the conjugate as defined above for use in treating cancer.
  • the present invention also relates to a compound of formula (1-1), (I-2), (IV), (IV-1), (IV-2), (IV-3), (IV-3-1 ), (IV-3-2), (IV-4), (IV-4-1), (IV-4-2), (IV-5), (IV-5-1), (IV- 5-2), (V), (VI) or (VII) as defined above for use in treating cancer.
  • the present invention also relates to a compound of formula (I) as defined above or the conjugate as defined above, for use in treating cancer, in combination with radiotherapy or in combination with radiotherapy and any anticancer drug.
  • the present invention thus concerns the field of cancer treatment and the use of new nitrogen-containing heterocycles derivatives as radiosensitizers in combined therapy with radiation and methods to improve the therapeutic index of tumours treated in first-line with radiotherapy or chemo-radiotherapy (soft tissue sarcoma, glioblastoma, head and neck carcinoma, lung carcinoma, cervix carcinoma, bladder carcinoma, rectum carcinoma and anal carcinoma) using these new radiosensitizers.
  • chemo-radiotherapy soft tissue sarcoma, glioblastoma, head and neck carcinoma, lung carcinoma, cervix carcinoma, bladder carcinoma, rectum carcinoma and anal carcinoma
  • the present invention also relates to a compound of formula (1-1), (I-2), (IV), (IV-1), (IV-2), (IV-3), (IV-3-1 ), (IV-3-2), (IV-4), (IV-4-1), (IV-4-2), (IV-5), (IV-5-1), (IV- 5-2), (V), (VI) or (VII) as defined above, for use in treating cancer, in combination with radiotherapy or in combination with radiotherapy and any anticancer drug.
  • the compound used according to the invention, the conjugate and/or the pharmaceutical composition according to the invention is administered in combination with additional cancer therapies.
  • this compound, the conjugate and/or the pharmaceutical composition may be administered in combination without or with targeted therapy, immunotherapy such as immune checkpoint therapy and immune checkpoint inhibitor, co-stimulatory antibodies, or chemotherapy.
  • Immune checkpoint therapy such as checkpoint inhibitors include, but are not limited to programmed death-1 (PD-1) inhibitors, programmed death ligand-1 (PD- L1) inhibitors, programmed death ligand-2 (PD-L2) inhibitors, lymphocyte-activation gene 3 (LAG3) inhibitors, T-cell immunoglobulin and mucin-domain containing protein 3 (TIM-3) inhibitors, T cell immunoreceptor with Ig and ITIM domains (TIGIT) inhibitors, B- and T-lymphocyte attenuator (BTLA) inhibitors, V-domain Ig suppressor of T-cell activation (VISTA) inhibitors, cytotoxic T-lymphocyte-associated protein 4 (CTLA4) inhibitors, Indoleamine 2,3-dioxygenase (IDO) inhibitors, killer immunoglobulin-like receptors (KIR) inhibitors, KIR2L3 inhibitors, KIR3DL2 inhibitors and carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM-1) inhibitors
  • checkpoint inhibitors include antibodies anti-PD1 , anti-PD- L1 , anti-CTLA-4, anti-TIM-3, anti-LAG3.
  • Co-stimulatory antibodies deliver positive signals through immune-regulatory receptors including but not limited to ICOS, CD137, CD27, OX-40 and GITR.
  • anti-PD1 antibodies include, but are not limited to, nivolumab, cemiplimab (REGN2810 or REGN-2810), tislelizumab (BGB-A317), tislelizumab, spartalizumab (PDR001 or PDR-001), ABBV-181 , JNJ-63723283, Bl 754091 , MAG012, TSR-042, AGEN2034, pidilizumab, nivolumab (ONO-4538, BMS-936558, MDX1106, GTPL7335 or Opdivo), pembrolizumab (MK-3475, MK03475, lambrolizumab, SCH-900475 or Keytruda) and antibodies described in international applications W02004004771 , W02004056875, W02006121168, WO2008156712, W02009014708, W02009114335, WO2013043569 and WO2014047350
  • anti-PD-L1 antibodies examples include, but are not limited to, LY3300054, atezolizumab, durvalumab and avelumab.
  • anti-CTLA-4 antibodies include, but are not limited to, ipilimumab (see, e.g., US patents US6,984,720 and US8, 017,114), tremelimumab (see, e.g., US patents US7, 109,003 and US8, 143,379), single chain anti-CTLA4 antibodies (see, e.g., international applications W01997020574 and W02007123737) and antibodies described in US patent US8,491 ,895.
  • ipilimumab see, e.g., US patents US6,984,720 and US8, 017,114
  • tremelimumab see, e.g., US patents US7, 109,003 and US8, 143,379
  • single chain anti-CTLA4 antibodies see, e.g., international applications W01997020574 and W02007123737
  • Example of KIR inhibitor is IPH4102 targeting KIR3DL2.
  • the compound used according to the invention, the conjugate and/or the pharmaceutical composition is administered to the patient in combination with targeted therapy.
  • Targeted therapy agents are drugs designed to interfere with specific molecules necessary for tumor growth and progression.
  • targeted therapy agents such as therapeutic monoclonal antibodies target specific antigens found on the cell surface, such as transmembrane receptors or extracellular growth factors.
  • Small molecules can penetrate the cell membrane to interact with targets inside a cell. Small molecules are usually designed to interfere with the enzymatic activity of the target protein such as for example proteasome inhibitor, tyrosine kinase or cyclin-dependent kinase inhibitor, histone deacetylase inhibitor.
  • Targeted therapy may also use cytokines.
  • Examples of such targeted therapy include with no limitations: Ado-trastuzumab emtansine (HER2), Afatinib (EGFR (HER1/ERBB1), HER2), Aldesleukin (Proleukin), alectinib (ALK), Alemtuzumab (CD52), axitinib (kit, PDGFRbeta, VEGFR1/2/3), Belimumab (BAFF), Belinostat (HDAC), Bevacizumab (VEGF ligand), Blinatumomab (CD19/CD3), bortezomib (proteasome), Brentuximab vedotin (CD30), bosutinib (ABL), brigatinib (ALK), cabozantinib (FLT3, KIT, MET, RET, VEGFR2), Canakinumab (IL-1 beta), carfilzomib (proteasome), ceritinib (ALK
  • the compound used according to the invention, the conjugate and/or the pharmaceutical composition is administered to the patient in combination with chemotherapy.
  • chemotherapy has its general meaning in the art and refers to the treatment that consists in administering to the patient a chemotherapeutic agent.
  • Chemotherapeutic agents include, but are not limited to alkylating agents such as thiotepa and cyclosphosphamide; alkyl sulfonates such as busulfan, improsulfan and piposulfan; aziridines such as benzodopa, carboquone, meturedopa, and uredopa; ethylenimines and methylamelamines including altretamine, triethylenemelamine, trietylenephosphoramide, triethiylenethiophosphoramide and trimethylolomelamine; acetogenins (especially bullatacin and bullatacinone); a camptothecin (including the synthetic analogue topotecan); bryostatin; callystatin; CC-1065 (including its adozelesin, carzelesin and bizelesin synthetic analogues); cryptophycins (particularly cryptophycin 1 and cryptophycin 8); dolastatin; du
  • the compound used according to the invention, the conjugate and/or the pharmaceutical composition is administered to the patient in combination with radiotherapy.
  • radiation therapies include, but are not limited to external beam radiotherapy (such as superficial X-rays therapy, orthovoltage X-rays therapy, megavoltage X-rays therapy, radiosurgery, stereotactic radiation therapy, fractionated stereotactic radiation therapy, hypofractionated radiotherapy, cobalt therapy, electron therapy, fast neutron therapy, neutron-capture therapy, proton therapy, intensity modulated radiation therapy (IMRT), 3-dimensional conformal radiation therapy (3D-CRT) and the like; brachytherapy; unsealed source radiotherapy; tomotherapy and the like; or minibeam radiation therapy.
  • external beam radiotherapy such as superficial X-rays therapy, orthovoltage X-rays therapy, megavoltage X-rays therapy, radiosurgery, stereotactic radiation therapy, fractionated stereotactic radiation therapy, hypofractionated radiotherapy, cobalt therapy, electron therapy, fast neutron therapy,
  • Gamma rays are another form of photons used in radiotherapy. Gamma rays are produced spontaneously as certain elements (such as radium, uranium, cesium and cobalt 60) release radiation as they decompose, or decay.
  • radiotherapy may be hadrontherapy (using beams from charged particles like protons or other ions such as carbon), proton radiotherapy or proton minibeam radiation therapy.
  • Proton radiotherapy is an ultra- precise form of radiotherapy that uses proton beams (Prezado Y, Jouvion G, Guardiola C, Gonzalez W, Juchaux M, Bergs J, Nauraye C, Labiod D, De Marzi L, Pouzoulet F, Patriarca A, Dendale R.
  • Radiotherapy may also be FLASH radiotherapy (FLASH-RT) or FLASH proton irradiation.
  • FLASH radiotherapy involves the ultra-fast delivery of radiation treatment at dose rates several orders of magnitude greater than those currently in routine clinical practice (ultra-high dose rate) (Favaudon V, Fouillade C, Vozenin MC. The radiotherapy FLASH to save healthy tissues. Med Sci (Paris) 2015 ; 31 : 121-123. DOI: 10.1051/medsci/20153102002); Patriarca A., Fouillade C. M., Martin F., Pouzoulet F., Nauraye C., et al.
  • Radiotherapy may also be hypofractionated radiotherapy (Shah JL, Li G, Shaffer JL, Azoulay Ml, Gibbs IC, Nagpal S, Soltys SG. Stereotactic Radiosurgery and Hypofractionated Radiotherapy for Glioblastoma. Neurosurgery.
  • the cancer is selected from the group consisting of: soft tissue carcinoma, glioblastoma, head and neck carcinoma, lung carcinoma, cervix carcinoma, bladder carcinoma, rectum carcinoma, and anal carcinoma.
  • cancers that may be treated by the compounds or conjugates of the invention include, but are not limited to: cardiac: sarcoma (angiosarcoma, fibrosarcoma, rhabdomyosarcoma, liposarcoma), myxoma, rhabdomyoma, fibroma, lipoma and teratoma; lung: bronchogenic carcinoma (squamous cell, undifferentiated small cell, undifferentiated large cell, adenocarcinoma), alveolar (bronchiolar) carcinoma, bronchial adenoma, sarcoma, lymphoma, chondromatous hamartoma, mesothelioma, gastrointestinal: esophagus (squamous cell carcinoma, adenocarcino
  • the cancer is selected from the group consisting of: benign, metastatic and malignant neoplasias, and also including acral lentiginous melanoma, actinic keratoses, adenocarcinoma, adenoid cycstic carcinoma, adenomas, adenosarcoma, adenosquamous carcinoma, astrocytic tumors, Bartholin gland carcinoma, basal cell carcinoma, bronchial gland carcinomas, capillary, carcinoids, carcinoma, carcinosarcoma, cavernous, cholangiocarcinoma, chondosarcoma, choriod plexus papilloma/carcinoma, clear cell carcinoma, cystadenoma, endodermal sinus tumor, endometrial hyperplasia, endometrial stromal sarcoma, endometrioid adenocarcinoma, ependymal, epitheloid,
  • the present invention also relates to a method for treating the pathological conditions indicated above, which comprises the administration, to a patient, of an effective dose of a compound according to the invention, or a pharmaceutically acceptable salt thereof.
  • Figure 1 Survival fraction of CCD-19Lu (triangles), ME-180 (circles) and LJ- 373 MG (squares) cell lines treated with increasing doses of X-rays (Philips/YXYLON generator) performed in the high throughput screening in 384- wells. Data represent mean ⁇ standard errors of at least three independent experiments.
  • Figure 2 Dose-response curves of compound 1 on CCD-19Lu (A), ME-180 (B) and U-373 MG (C) cell lines without (upper curve) or following 4 (medium curve) or 6 (lower curve) Gy irradiation. Each dot represents an independent experiment (three replicates).
  • Figure 3 Dose-response curves of compound 2 on CCD-19Lu (A), ME-180 (B) and U-373 MG (C) cell lines without (upper curve) or following 4 (medium curve) or 6 (lower curve) Gy irradiation. Each dot represents an independent experiment (three replicates).
  • Figure 4 Dose-response curves of compound 3 on CCD-19Lu (A) and U-373 MG (B) cell lines without (upper curve) or following 4 (medium curve) or 6 (lower curve) Gy irradiation. Each dot represents an independent experiment (three replicates).
  • Figure 5 Dose-response curves of compound 4 on CCD-19Lu (A), ME-180 (B) and U-373 MG (C) cell lines without (upper curve) or following 4 (medium curve) or 6 (lower curve) Gy irradiation. Each dot represents an independent experiment (three replicates).
  • Figure 6 Dose-response curves of Olaparib® on CCD-19Lu (A), ME-180 (B) and U-373 MG (C) cell lines without (upper curve) or following 4 (medium curve) or 6 (lower curve) Gy irradiation. Each dot represents an independent experiment (three replicates).
  • Figure 7 Isobolographic graph analysis of the association of compound 1 with radiation (4 and 6 Gy) on ME-180 (A) and U-373 MG (B) cell lines for a survival isoeffect of 50% (SI 5 o). The different replicates are all plotted under the envelope of additivity showing the supra-additive effect.
  • Figure 8 Isobolographic graph analysis of the association of compound 2 (A), compound 3 (B) and compound 4 (C) with 6 Gy-radiation on U-373 MG (B) cell line for a survival isoeffect of 50% (SI 5 o). The different replicates are all plotted under the envelope of additivity showing the supra-additive effect.
  • Example 1 High-throughput molecules and irradiation screening
  • the human glioblastoma cancer cell lines, U-373 MG (ECACC, 08061901 , radioresistant) and the cervical cancer cells ME-180 (ATCC, FITB-33, radiosensitive) were used in this study.
  • Fluman pulmonary normal fibroblasts CCD-19Lu ATCC, CCL-210) were used as normal tissue control. All cell lines were negative for mycoplasma contamination and were authentified by short tandem repeat (STR) profiling (Institut Curie, Genomic Platform). The appropriate cell plating density was adapted to a well of 384-well plate in the presence of their recommended cell media.
  • STR short tandem repeat
  • High-throughput chemo-irradiation screening Cells were seeded on barcoded 384-well plates and allowed to attach overnight. Cells were then exposed to 11 -point concentration range of the 160 compounds (from 0.17 nM to 10 mM) and incubated at 37°C for 24 h. Cells were further mock irradiated (0 Gy) or irradiated at 4 or 6 Gy and incubated for additional 3 days at 37°C prior cell fixation and DAPI nuclei labelling.
  • Cell survival was determined using an automated high-throughput wide-field microscopy system (INCell 2200 - GE Healthcare). Six individual images were acquired for each well of the experimental plates and each labelled nucleus was detected using an automated nuclei segmentation custom-developed method. All survival curves represent at least three independent experiments.
  • Isobolograms The combination of radiotherapy and chemotherapy is not easy to assess, as the dose-response curve of each treatment is nonlinear (according to the Hill equation for drug alone and to linear-quadratic model for radiation alone). In that case, to determine if the effect of the combined treatment is greater or less that would be expected on the basis of the response of each agent alone, Steel and Peckham developped the isobolographic concept based on the built of an envelope of additivity in an isoeffect plot (Steel G.G. and Peckham M.J., Int. J. Radiat. Oncol. Biol. Phys. (1979) 5:85-91 ).
  • This envelope of additivity is based on the calculation of isoeffect curves according to Mode I (independent mechanisms of action of the two agents) and Mode II (dependent mechanisms of action of the two agents).
  • This graph delimited three regions: the region of additivity inside the envelope, the region of supra-additivity (or synergy) under the envelope and the region of sub-additivity or antagonism above the envelope.
  • the cytotoxic interactions of the selected compounds and radiation at 50% survival isoeffect (SI 5 o) were evaluated on the ME-180 and U-373 MG cancer cell lines using this method.
  • the median and mean of the three SI 5 o replicates of combined treatments were calculated according to Hill equation after fitting the experimental curves obtained with 4 and/or 6 Gy and these points were plotted on the graph.
  • the effect of a given drug in combination with radiation was categorized as supra-additive, additive or antagonist.
  • Fibroblast toxicity In tumor treatment, when using chemo-radiotherapy combination, the balance between tumor control and the risk of normal tissue side- effects is given by the therapeutic index of the treatment. The challenge is to improve this therapeutic index with a good control of the tumor and a low toxicity against normal tissues. In this study, a minimal toxicity on healthy tissues was taken into account thanks to a parallel study on fibroblasts.
  • the second criterion based on IC 5 o filtered out the compounds that affected cell survival of the fibroblast cell line without irradiation (IC 5 o OGy CCD-19Lu > 9 x IC 5 o 4/6Gy cancer cells) to assess the drug toxicity and the third criterion filtered out the compounds that also affected cell survival of the fibroblast cell line with irradiation (IC 5 o 4/6 Gy CCD-19Lu > 9 x IC 5 o 4/6Gy cancer cells).
  • the selected compounds presented in this invention passed through these three criteria, providing new non-toxic synthetized nitrogen-containing heterocyles derivatives for use as radiosensitizers in combination with radiation.
  • Figure 2, 3, 4 and 5 showed the typical drug dose-response curves of increasing doses of compound 1 ( Figure 2), compound 2 (Figure 3), compound 3 ( Figure 4) and compound 4 ( Figure 5) of the present invention on CCD-19Lu- (A),
  • Our process was validated by the use in a blind position of Olaparib®, a radiosensitizer now in clinical trials, as positive control in our FITS ( Figure 6). As expected, this molecule was part of the selected supra-additive hits.
  • the first criterion of our FITS is the use of the isobolographic method. All the nitrogen-containing heterocycle derivatives compounds of the present invention showed supra-additive effects when combined with radiation. The cytotoxic interaction of the compound 1 , 2, 3 and 4 with radiation at 50% survival isoeffect (SI50) were evaluated on the cancer cell lines and is showed on Figure 7 and 8.

Landscapes

  • Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Epidemiology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The present invention relates to a compound having the following formula (I), wherein: - R is a group of formula (II) or (III): wherein in particular R1 is H, and Ra and Rb are H; and in particular R2 is H, R4 is H, and R3 is selected from the group consisting of: H, halogen, (C1-C6)alkyl, aryl groups, and heteroaryl groups; - R5 is chosen from the group consisting of: H, (C1-C6)alkyl, and aryl; - i is 1 or 2; and the R7 groups, identical or different, are chosen from the group consisting of: H, halogen, and (C1-C6)alkyl; for use as radiosensitizer.

Description

NITROGEN-CONTAINING HETEROCYCLES AS RADIOSENSITIZERS
The present invention concerns nitrogen-containing heterocycles as radiosensitizers, in particular for cancer treatment. The present invention also relates to these nitrogen-containing heterocycles for use for the treatment of cancer, in combination with radiotherapy or in combination with radiotherapy and any anticancer drug.
Some tumours (most of them are carcinomas) are treated in first line with radiotherapy or with chemoradiotherapy. The challenge is to deliver a cure dose to the tumor while limiting the possibility of serious damages to normal tissues. The balance between tumour control and the risk of normal tissue side-effects is given by the therapeutic index of the treatment. Improving the therapeutic index by modulating the tumour/normal tissue response is challenging. To this aim, small molecules could be used as radiosensitizing agents (radiosensitizers) in order to increase the radiosensitivity of the tumours. The main characteristics of a radiosensitizer should be a low toxicity without ionizing radiations (healthy tissue) and an enhanced injury to the tumour in presence of ionizing radiations. The expectation of getting a supra-additive antitumor effect (greater than expected based on the simple addition of each cytotoxic used alone) with minimal secondary effects on healthy tissues is of importance in cancer therapy.
Until now, many anticancer agents are known to be toxic per se but are still used as radiosensitizers as they induce more cell death when combined with radiation than radiation alone. The development of new non-toxic (on healthy tissues) radiosensitizing agents is of importance in oncology.
The aim of the present invention is thus to provide non-toxic nitrogen- containing heterocycles for use as radiosensitizers for the treatment of cancer in combination with radiotherapy or in combination with radiotherapy and any anticancer drug. Thus, the present invention relates to a compound having the following formula (I):
Figure imgf000003_0001
wherein:
- R is: a group of formula (II):
Figure imgf000003_0002
wherein:
. Ri is H or is selected from the group consisting of: halogen, (Ci-C6)alkyl, aryl groups, and heteroaryl groups, and
. Ra and Rb are independently H or a (Ci-C6)alkyl group; or a group of formula (III):
Figure imgf000003_0003
wherein:
. R2 is H or is selected from the group consisting of: halogen, (Ci-Ce)alkyl, aryl groups, and heteroaryl groups,
. R is H or is selected from the group consisting of: halogen, (Ci-Ce)alkyl, aryl groups, and heteroaryl groups, said aryl and heteroaryl groups being optionally substituted with one or several substituents independently selected from: amino, hydroxyl, thiol (-SH), halogen, (Ci- C6)alkyl, (Ci-Ce)alkoxy, (Ci-Ce)alkylthio, (Ci-C6)alkylamino, halo(Ci-Ce)alkyl, carboxyl and carboxy(Ci-Ce)alkyl; . R3 is selected from the group consisting of: H, halogen, (Ci-C6)alkyl, aryl groups, and heteroaryl groups; said aryl and heteroaryl groups being optionally substituted with one or several substituents independently selected from: amino, hydroxyl, thiol (-SH), halogen, (Ci- C6)alkyl, (Ci-C6)alkoxy, (Ci-C6)alkylthio, (Ci-C6)alkylamino, halo(Ci-C6)alkyl, carboxyl and carboxy(Ci-C6)alkyl;
- R5 is chosen from the group consisting of: H, (Ci-C6)alkyl, and aryl, preferably phenyl, said aryl group being optionally substituted with one or several substituents independently selected from: amino, hydroxyl, thiol, halogen, (Ci- C6)alkyl, (Ci-C6)alkoxy, (Ci-C6)alkylthio, (Ci-C6)alkylamino, halo(Ci-C6)alkyl, carboxyl and carboxy(Ci-C6)alkyl;
- i is 1 or 2;
- the R7 groups, identical or different, are chosen from the group consisting of: H, halogen, and (Ci-C6)alkyl; or a pharmaceutically acceptable salt thereof, for use for the treatment of cancer as radiosensitizer.
The present invention thus concerns new radiosensitizers, preferably used in combination with radiation as explained later.
According to the invention, the expression "Ct-Cz" means a carbon-based chain which can have from t to z carbon atoms, for example C1-C3 means a carbon- based chain which can have from 1 to 3 carbon atoms.
According to the invention, the term "halogen" means: a fluorine, a chlorine, a bromine or an iodine.
Within the present application, the term "alkyl" means: a linear or branched, saturated, hydrocarbon-based aliphatic group comprising, unless otherwise mentioned, from 1 to 6 carbon atoms. By way of examples, mention may be made of methyl, ethyl, n-propyl, isopropyl, butyl, isobutyl, tert-butyl or pentyl groups.
According to the invention, the term "alkoxy" means: an -O-alkyl radical where the alkyl group is as previously defined. By way of examples, mention may be made of -0-(Ci-C4)alkyl groups, and in particular the -O-methyl group, the -O-ethyl group, as -0-C3alkyl group, the -O-propyl group, the -O-isopropyl group, and as -0-C4alkyl group, the -O-butyl, -O-isobutyl or -O-tert-butyl group.
According to the invention, the term "alkylamino" means: an -NH-alkyl group, the alkyl group being as defined above. According to the invention, the term "alkylthio" means: an -S-alkyl group, the alkyl group being as defined above.
According to the invention, the term "haloalkyl" means: an alkyl group as defined above, in which one or more of the hydrogen atoms is (are) replaced with a halogen atom. By way of example, mention may be made of fluoroalkyls, in particular CF3 or CHF2.
According to the invention, the term "aryl" means: a cyclic aromatic group comprising between 6 and 10 carbon atoms. By way of examples of aryl groups, mention may be made of phenyl or naphthyl groups.
According to the invention, the term "heteroaryl" means: a 5- to 10-membered aromatic monocyclic or bicyclic group containing from 1 to 4 heteroatoms selected from O, S or N. By way of examples, mention may be made of imidazolyl, thiazolyl, oxazolyl, furanyl, thiophenyl, pyrazolyl, oxadiazolyl, tetrazolyl, pyridinyl, pyrazinyl, pyrimidinyl, pyridazinyl, indolyl, benzofuranyl, benzothiophenyl, benzoxazolyl, benzimidazolyl, indazolyl, benzothiazolyl, isobenzothiazolyl, benzotriazolyl, quinolinyl and isoquinolinyl groups.
By way of a heteroaryl comprising 5 to 6 atoms, including 1 to 4 nitrogen atoms, mention may in particular be made of the following representative groups: pyrrolyl, pyrazolyl, 1 ,2,3-triazolyl, 1 ,2,4-triazolyl, tetrazolyl and 1 ,2,3-triazinyl.
Mention may also be made, by way of heteroaryl, of thiophenyl, oxazolyl, furazanyl, 1 ,2,4-thiadiazolyl, naphthyridinyl, quinoxalinyl, phthalazinyl, imidazo[1 ,2- a]pyridine, imidazo[2,1-b]thiazolyl, cinnolinyl, benzofurazanyl, azaindolyl, benzimidazolyl, benzothiophenyl, thienopyridyl, thienopyrimidinyl, pyrrolopyridyl, imidazopyridyl, benzoazaindole, 1 ,2,4-triazinyl, indolizinyl, isoxazolyl, isoquinolinyl, isothiazolyl, purinyl, quinazolinyl, quinolinyl, isoquinolyl, 1 ,3,4-thiadiazolyl, thiazolyl, isothiazolyl, carbazolyl, and also the corresponding groups resulting from their fusion or from fusion with the phenyl nucleus.
According to the present invention, the term "carboxyalkyl" means: an FIOOC- alkyl- group, the alkyl group being as defined above. As examples of carboxyalkyl groups, mention may in particular be made of carboxymethyl or carboxyethyl.
According to the present invention, the term "carboxyl" means: a COOFI group.
According to the invention, the term “alkylcarbonyl” means a -CO-alkyl group, the alkyl group being as defined above.
According to the invention, the term “alkoxylcarbonyl” means a -CO-O-alkyl group, the alkyl group being as defined above. The radiosensitizers according to the present invention have the formula (I) as mentioned above.
A family of radiosensitizers according to the present invention consists of compounds having the formula (I) as defined above, wherein R is a group of formula (II). Such compounds have the below formula (1-1):
Figure imgf000006_0001
Ra, Rb, Ri, R5, R7, and i being as defined above in formula (I).
A preferred family of radiosensitizers according to the present invention consists of compounds having the formula (1-1), wherein Ra and Rb are H.
Another preferred family of radiosensitizers according to the present invention consists of compounds having the formula (1-1), wherein Ri is H.
A preferred family of radiosensitizers according to the present invention consists of compounds having the formula (1-1), wherein Ra and Rb are H and Ri is H.
A family of radiosensitizers according to the present invention consists of compounds having the formula (I) as defined above, wherein R is a group of formula (III). Such compounds have the below formula (I-2):
Figure imgf000006_0002
R2, R3, R4, R5, R7, and i being as defined above in formula (I). A preferred family of radiosensitizers according to the present invention consists of compounds having the formula (1-2), wherein R4 is H.
Another preferred family of radiosensitizers according to the present invention consists of compounds having the formula (1-2), wherein R3 is H or a (Ci-Ce)alkyl group.
A preferred family of radiosensitizers according to the present invention consists of compounds having the formula (1-2), wherein R2 is halogen, such as Br, or an aryl group, such as phenyl.
Preferably, in formula (I) as defined above, or in formula (1-1) or (I-2), R5 is an aryl group, substituted or not.
Preferably, in formula (I) as defined above, or in formula (1-1) or (I-2), i=1 and R7 is F.
According to an embodiment, the compounds for the use according to the present invention have the following formula (IV):
Figure imgf000007_0001
wherein:
- R, R , and i are as defined above in formula (I), and
- R6 is chosen from the group consisting of: amino, hydroxyl, thiol, halogen, (Ci-C6)alkyl, (Ci-C6)alkoxy, (Ci-C6)alkylthio, (Ci-C6)alkylamino, halo(Ci-C6)alkyl, carboxyl and carboxy(Ci-C6)alkyl.
The compounds of formula (IV) correspond to compounds having the formula (I) as defined above, wherein R5 is a phenyl group, optionally substituted by a R6 group. A family of radiosensitizers according to the present invention consists of compounds having the formula (IV) as defined above, wherein R is a group of formula (II). Such compounds have the below formula (IV-1):
Figure imgf000008_0001
Ra, Rb, Ri, R7, and i being as defined above in formula (I), and R6 being as defined above in formula (IV).
A preferred family of radiosensitizers according to the present invention consists of compounds having the formula (IV-1), wherein Ra and Rb are H.
Another preferred family of radiosensitizers according to the present invention consists of compounds having the formula (IV-1), wherein Ri is H.
A preferred family of radiosensitizers according to the present invention consists of compounds having the formula (IV-1), wherein Ra and Rb are H and Ri is H.
A family of radiosensitizers according to the present invention consists of compounds having the formula (IV) as defined above, wherein R is a group of formula (III). Such compounds have the below formula (IV-2):
Figure imgf000008_0002
defined above in formula (IV). A preferred family of radiosensitizers according to the present invention consists of compounds having the formula (IV-2), wherein FU is H.
Another preferred family of radiosensitizers according to the present invention consists of compounds having the formula (IV-2), wherein R3 is H or a (Ci-C6)alkyl group.
A preferred family of radiosensitizers according to the present invention consists of compounds having the formula (IV-2), wherein R2 is halogen, such as Br, or an aryl group, such as phenyl.
According to an embodiment, the compounds for the use according to the present invention have the following formula (IV), wherein R6 is chosen from the group consisting of: hydroxyl, halogen, in particular F, (Ci-Ce)alkoxy, in particular OMe, and halo(Ci-Ce)alkyl, in particular CF3, preferably in para position.
According to an embodiment, the compounds for the use according to the present invention have the following formula (IV-1) or (IV-2), wherein R6 is chosen from the group consisting of: hydroxyl, halogen, in particular F, (Ci-Ce)alkoxy, in particular OMe, and halo(Ci-Ce)alkyl, in particular CF3, preferably in para position.
Preferred groups R6 are OH, F, OMe, and CF3, and are preferably in para position.
A preferred family of radiosensitizers according to the present invention consists of compounds having the formula (IV), wherein R6 is halogen in para position, and preferably R6 is F in para position.
A preferred family of radiosensitizers according to the present invention consists of compounds having the formula (IV-1) or (IV-2), wherein R6 is halogen in para position, and preferably R6 is F in para position.
According to an embodiment, the compound for the use according to the invention has the following formula (IV-3):
Figure imgf000009_0001
R, R , and i being as defined in formula (I), and R6 being as defined in formula
(IV). The compounds of formula (IV-3) correspond to compounds having the formula (IV) as defined above, wherein R6 is in para position.
A particular family of radiosensitizers according to the present invention consists of compounds having the below formula (IV-3-1 ):
Figure imgf000010_0001
Ra, Rb, Ri, R7, and i being as defined above in formula (I), and R6 being as defined above in formula (IV).
A preferred family of radiosensitizers according to the present invention consists of compounds having the formula (IV-3-1), wherein Ra and Rb are H.
Another preferred family of radiosensitizers according to the present invention consists of compounds having the formula (IV-3-1), wherein Ri is H.
A preferred family of radiosensitizers according to the present invention consists of compounds having the formula (IV-3-1), wherein Ra and Rb are H and Ri is H.
A particular family of radiosensitizers according to the present invention consists of compounds having the below formula (IV-3-2):
Figure imgf000010_0002
R2, R3, R4, R7, and i being as defined above in formula (I), and R6 being as defined above in formula (IV). A preferred family of radiosensitizers according to the present invention consists of compounds having the formula (IV-3-2), wherein FU is H.
Another preferred family of radiosensitizers according to the present invention consists of compounds having the formula (IV-3-2), wherein R3 is H or a (Ci-C6)alkyl group.
A preferred family of radiosensitizers according to the present invention consists of compounds having the formula (IV-3-2), wherein R2 is halogen, such as Br, or an aryl group, such as phenyl.
According to an embodiment, the compounds for the use according to the invention have the following formula (IV-4):
Figure imgf000011_0001
R, R , and R6 being as defined above. Preferably, in formula (IV-4), R7 is F.
A family of radiosensitizers according to the present invention consists of compounds having the formula (IV-4) as defined above, wherein R is a group of formula (II). Such compounds have the below formula (IV-4-1 ):
Figure imgf000011_0002
Ra, Rb, Ri, and R being as defined above in formula (I), and R6 being as defined above in formula (IV).
A preferred family of radiosensitizers according to the present invention consists of compounds having the formula (IV-4-1), wherein Ra and Rb are H. Another preferred family of radiosensitizers according to the present invention consists of compounds having the formula (IV-4-1), wherein Ri is H.
A preferred family of radiosensitizers according to the present invention consists of compounds having the formula (IV-4-1), wherein Ra and Rb are H and Ri is H.
A family of radiosensitizers according to the present invention consists of compounds having the formula (IV-4) as defined above, wherein R is a group of formula (III). Such compounds have the below formula (IV-4-2):
Figure imgf000012_0001
R2, R3, R4, and R being as defined above in formula (I), and R6 being as defined above in formula (IV).
A preferred family of radiosensitizers according to the present invention consists of compounds having the formula (IV-4-2), wherein R4 is H.
Another preferred family of radiosensitizers according to the present invention consists of compounds having the formula (IV-4-2), wherein R3 is H or a (Ci-Ce)alkyl group.
A preferred family of radiosensitizers according to the present invention consists of compounds having the formula (IV-4-2), wherein R2 is halogen, such as Br, or an aryl group, such as phenyl.
According to an embodiment, the compounds for the use according to the invention have the following formula (IV-5):
Figure imgf000012_0002
R, R?, and R6 being as defined above. Preferably, in formula (IV-5), R7 is F.
A family of radiosensitizers according to the present invention consists of compounds having the formula (IV-5) as defined above, wherein R is a group of formula (II). Such compounds have the below formula (IV-5-1 ):
Figure imgf000013_0001
Ra, Rb, Ri, R7, and R6 being as defined above.
A preferred family of radiosensitizers according to the present invention consists of compounds having the formula (IV-5-1), wherein Ra and Rb are H.
Another preferred family of radiosensitizers according to the present invention consists of compounds having the formula (IV-5-1), wherein Ri is H.
A preferred family of radiosensitizers according to the present invention consists of compounds having the formula (IV-5-1), wherein Ra and Rb are H and Ri is H.
A family of radiosensitizers according to the present invention consists of compounds having the formula (IV-5) as defined above, wherein R is a group of formula (III). Such compounds have the below formula (IV-5-2):
Figure imgf000013_0002
(IV-5-2)
R2, R3, R4, R7, and R6 being as defined above. A preferred family of radiosensitizers according to the present invention consists of compounds having the formula (IV-5-2), wherein R4 is H.
Another preferred family of radiosensitizers according to the present invention consists of compounds having the formula (IV-5-2), wherein R3 is H or a (Ci-Ce)alkyl group.
A preferred family of radiosensitizers according to the present invention consists of compounds having the formula (IV-5-2), wherein R2 is halogen, such as Br, or an aryl group, such as phenyl.
According to an embodiment, the compound for the use according to the present invention has the following formula (V):
Figure imgf000014_0001
R being as defined above in formula (I).
According to an embodiment, the compound for the use according to the present invention has the following formula (VI):
Figure imgf000014_0002
Ri, Ra and Rb being as defined above in formula (I).
A preferred family of radiosensitizers according to the present invention consists of compounds having the formula (VI), wherein Ra and Rb are H.
Another preferred family of radiosensitizers according to the present invention consists of compounds having the formula (VI), wherein Ri is H. According to an embodiment, the compound for the use according to the present invention has the following formula (VII):
Figure imgf000015_0001
R2, R3 and R4 being as defined above.
A preferred family of radiosensitizers according to the present invention consists of compounds having the formula (VII), wherein FU is H.
Another preferred family of radiosensitizers according to the present invention consists of compounds having the formula (VII), wherein R3 is H or a (Ci-Ce)alkyl group.
A preferred family of radiosensitizers according to the present invention consists of compounds having the formula (VII), wherein R2 is halogen, such as Br, or an aryl group, such as phenyl.
According to a preferred embodiment, the compound for the use according to the invention is one of the following compounds:
Figure imgf000015_0002
(3) (4) The present invention also relates to a conjugate molecule comprising one or more compound(s) according to the invention, having the formula (I) as defined above, said compound(s) being covalently bound to at least one cell binding agent.
The present invention also relates to a conjugate molecule comprising one or more compound(s) according to the invention, having the formula (1-1), (1-1), (IV), (IV-1), (IV-2), (IV-3), (IV-3-1 ), (IV-3-2), (IV-4), (IV-4-1), (IV-4-2), (IV-5), (IV-5-1), (IV- 5-2), (v), (VI) or (VII) as defined above, said compound(s) being covalently bound to at least one cell binding agent.
The present invention also relates to a conjugate molecule comprising one or more compound(s) according to the invention, having the formula (1), (2), (3) or (4) as defined above, said compound(s) being covalently bound to at least one cell binding agent.
The present invention also relates to a conjugate molecule being a compound according to the invention to which is bound to at least one payload, optionally through a linker or covalently conjugated, thereby forming a single molecule.
Examples of suitable payloads include, but are not limited to, peptides, polypeptides, proteins, antibodies, or antigen-binding fragments thereof, antigens, nucleic acid molecules, polymers, small molecules, mimetic agents, drugs, inorganic molecules, organic molecules, and radioisotopes.
Examples of suitable payloads include, but are not limited to, cell binding agents, chemotherapeutic agents, targeted therapy agents, cytotoxic agents, ligands for cellular receptor(s), immunomodulatory agents, pro-apoptotic agents, anti- angiogenic agents, photodetectable labels, contrast agents, radiolabels, and the like.
According to the invention, a cell binding agent is a molecule with affinity for a biological target. The cell binding agent may be, for example, a ligand, a protein, an antibody, more particularly a monoclonal antibody, a protein or antibody fragment, a peptide, an oligonucleotide or an oligosaccharide. The function of the binding agent is to direct the biologically active compound towards the biological target.
In some embodiments, the conjugate of the invention is an antibody drug conjugate (ADC). Accordingly, the payload is an antibody, particularly monoclonal antibody, sdAb, VHH, intrabody, or single-chain antibodies (scFv). In some embodiments, the conjugate of the invention comprises a linker unit between the compound of the invention and the payload. In some embodiments, the linker is cleavable under intracellular conditions, such that cleavage of the linker releases the compound of the invention from the payload in the intracellular environment. In yet other embodiments, the linker unit is not cleavable and the compound of the invention is released, for example, by payload degradation.
In some embodiments, the linker is cleavable by a cleaving agent that is present in the intracellular environment (e.g., within a lysosome or endosome or caveolea). The linker can be, e.g., a peptidyl linker that is cleaved by an intracellular peptidase or protease enzyme, including, but not limited to, a lysosomal or endosomal protease.
In some embodiments, the cleavable linker is pH-sensitive, i.e., sensitive to hydrolysis at certain pH values. Typically, the pH-sensitive linker hydrolyzable under acidic conditions. For example, an acid-labile linker that is hydrolyzable in the lysosome (e.g., a hydrazone, semicarbazone, thiosemicarbazone, cis-aconitic amide, orthoester, acetal, ketal, or the like) can be used. (See, e.g., U.S. Pat. Nos. 5,122,368; 5,824,805; 5,622,929; Dubowchik and Walker, 1999, Pharm. Therapeutics 83:67-123; Neville et al., 1989, Biol. Chem. 264:14653-14661). Such linkers are relatively stable under neutral pH conditions, such as those in the blood, but are unstable at below pH 5.5 or 5.0, the approximate pH of the lysosome.
In some embodiments, the linker is cleavable under reducing conditions (e.g., a disulfide linker). A variety of disulfide linkers are known in the art, including, for example, those that can be formed using SATA (N-succinimidyl-S- acetylthioacetate), SPDP (N-succinimidyl-3-(2-pyridyldithio)propionate), SPDB (N- succinimidyl-3-(2-pyridyldithio)butyrate) and SMPT (N-succinimidyl-oxycarbonyl- alpha-methyl-alpha-(2-pyridyl-dithio)toluene), SPDB and SMPT. (See, e.g., Thorpe et al., 1987, Cancer Res. 47:5924-5931 ; Wawrzynczak et al., In Immunoconjugates: Antibody Conjugates in Radioimagery and Therapy of Cancer (C. W. Vogel ed., Oxford U. Press, 1987. See also U.S. Pat. No. 4,880,935).
The present invention also relates to the conjugate as mentioned above for use as drug.
The present invention also relates to a medicament comprising the conjugate as defined above. The present invention also relates to a pharmaceutical composition, comprising the conjugate as defined above, and also at least one pharmaceutically acceptable excipient.
These pharmaceutical compositions contain an effective dose of at least one compound according to the invention, or a pharmaceutically acceptable salt, and also at least one pharmaceutically acceptable excipient.
Said excipients are selected, according to the pharmaceutical form and the mode of administration desired, from the usual excipients which are known to those skilled in the art.
In the pharmaceutical compositions of the present invention for oral, sublingual, subcutaneous, intramuscular, intravenous, topical, local, intratracheal, intranasal, transdermal or rectal administration, the active ingredient of formula (I) above, or the salt thereof, can be administered in unit administration form, as a mixture with conventional pharmaceutical excipients, to animals and to human beings for the treatment of the disorders and diseases below.
The suitable unit administration forms include oral forms such as tablets, soft or hard gel capsules, powders, granules and oral solutions or suspensions, sublingual, buccal, intratracheal, intraocular and intranasal administration forms, forms for administration by inhalation, topical, transdermal, subcutaneous, intramuscular or intravenous administration forms, rectal administration forms, and implants. For topical application, the compounds according to the invention can be used in creams, gels, ointments or lotions.
The present invention also relates to a compound of formula (I) as defined above or the conjugate as defined above for use in treating cancer.
The present invention also relates to a compound of formula (1-1), (I-2), (IV), (IV-1), (IV-2), (IV-3), (IV-3-1 ), (IV-3-2), (IV-4), (IV-4-1), (IV-4-2), (IV-5), (IV-5-1), (IV- 5-2), (V), (VI) or (VII) as defined above for use in treating cancer.
The present invention also relates to a compound of formula (I) as defined above or the conjugate as defined above, for use in treating cancer, in combination with radiotherapy or in combination with radiotherapy and any anticancer drug.
The present invention thus concerns the field of cancer treatment and the use of new nitrogen-containing heterocycles derivatives as radiosensitizers in combined therapy with radiation and methods to improve the therapeutic index of tumours treated in first-line with radiotherapy or chemo-radiotherapy (soft tissue sarcoma, glioblastoma, head and neck carcinoma, lung carcinoma, cervix carcinoma, bladder carcinoma, rectum carcinoma and anal carcinoma) using these new radiosensitizers.
The present invention also relates to a compound of formula (1-1), (I-2), (IV), (IV-1), (IV-2), (IV-3), (IV-3-1 ), (IV-3-2), (IV-4), (IV-4-1), (IV-4-2), (IV-5), (IV-5-1), (IV- 5-2), (V), (VI) or (VII) as defined above, for use in treating cancer, in combination with radiotherapy or in combination with radiotherapy and any anticancer drug.
In some embodiments, the compound used according to the invention, the conjugate and/or the pharmaceutical composition according to the invention is administered in combination with additional cancer therapies. In particular, this compound, the conjugate and/or the pharmaceutical composition may be administered in combination without or with targeted therapy, immunotherapy such as immune checkpoint therapy and immune checkpoint inhibitor, co-stimulatory antibodies, or chemotherapy.
Immune checkpoint therapy such as checkpoint inhibitors include, but are not limited to programmed death-1 (PD-1) inhibitors, programmed death ligand-1 (PD- L1) inhibitors, programmed death ligand-2 (PD-L2) inhibitors, lymphocyte-activation gene 3 (LAG3) inhibitors, T-cell immunoglobulin and mucin-domain containing protein 3 (TIM-3) inhibitors, T cell immunoreceptor with Ig and ITIM domains (TIGIT) inhibitors, B- and T-lymphocyte attenuator (BTLA) inhibitors, V-domain Ig suppressor of T-cell activation (VISTA) inhibitors, cytotoxic T-lymphocyte-associated protein 4 (CTLA4) inhibitors, Indoleamine 2,3-dioxygenase (IDO) inhibitors, killer immunoglobulin-like receptors (KIR) inhibitors, KIR2L3 inhibitors, KIR3DL2 inhibitors and carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM-1) inhibitors. In particular, checkpoint inhibitors include antibodies anti-PD1 , anti-PD- L1 , anti-CTLA-4, anti-TIM-3, anti-LAG3. Co-stimulatory antibodies deliver positive signals through immune-regulatory receptors including but not limited to ICOS, CD137, CD27, OX-40 and GITR.
Examples of anti-PD1 antibodies include, but are not limited to, nivolumab, cemiplimab (REGN2810 or REGN-2810), tislelizumab (BGB-A317), tislelizumab, spartalizumab (PDR001 or PDR-001), ABBV-181 , JNJ-63723283, Bl 754091 , MAG012, TSR-042, AGEN2034, pidilizumab, nivolumab (ONO-4538, BMS-936558, MDX1106, GTPL7335 or Opdivo), pembrolizumab (MK-3475, MK03475, lambrolizumab, SCH-900475 or Keytruda) and antibodies described in international applications W02004004771 , W02004056875, W02006121168, WO2008156712, W02009014708, W02009114335, WO2013043569 and WO2014047350.
Examples of anti-PD-L1 antibodies include, but are not limited to, LY3300054, atezolizumab, durvalumab and avelumab.
Examples of anti-CTLA-4 antibodies include, but are not limited to, ipilimumab (see, e.g., US patents US6,984,720 and US8, 017,114), tremelimumab (see, e.g., US patents US7, 109,003 and US8, 143,379), single chain anti-CTLA4 antibodies (see, e.g., international applications W01997020574 and W02007123737) and antibodies described in US patent US8,491 ,895.
Examples of anti- VISTA antibodies are described in US patent application US20130177557.
Examples of inhibitors of the LAG3 receptor are described in US patent US5,773,578.
Example of KIR inhibitor is IPH4102 targeting KIR3DL2.
In some embodiments, the compound used according to the invention, the conjugate and/or the pharmaceutical composition is administered to the patient in combination with targeted therapy. Targeted therapy agents are drugs designed to interfere with specific molecules necessary for tumor growth and progression. For example, targeted therapy agents such as therapeutic monoclonal antibodies target specific antigens found on the cell surface, such as transmembrane receptors or extracellular growth factors. Small molecules can penetrate the cell membrane to interact with targets inside a cell. Small molecules are usually designed to interfere with the enzymatic activity of the target protein such as for example proteasome inhibitor, tyrosine kinase or cyclin-dependent kinase inhibitor, histone deacetylase inhibitor. Targeted therapy may also use cytokines. Examples of such targeted therapy include with no limitations: Ado-trastuzumab emtansine (HER2), Afatinib (EGFR (HER1/ERBB1), HER2), Aldesleukin (Proleukin), alectinib (ALK), Alemtuzumab (CD52), axitinib (kit, PDGFRbeta, VEGFR1/2/3), Belimumab (BAFF), Belinostat (HDAC), Bevacizumab (VEGF ligand), Blinatumomab (CD19/CD3), bortezomib (proteasome), Brentuximab vedotin (CD30), bosutinib (ABL), brigatinib (ALK), cabozantinib (FLT3, KIT, MET, RET, VEGFR2), Canakinumab (IL-1 beta), carfilzomib (proteasome), ceritinib (ALK), Cetuximab (EGFR), cofimetinib (MEK), Crizotinib (ALK, MET, ROS1), Dabrafenib (BRAF), Daratumumab (CD38), Dasatinib (ABL), Denosumab (RANKL), Dinutuximab (B4GALNT1 (GD2)), Elotuzumab (SLAMF7), Enasidenib (IDH2), Erlotinib (EGFR), Everolimus (mTOR), Gefitinib (EGFR), Ibritumomab tiuxetan (CD20), Sonidegib (Smoothened), Sipuleucel-T, Siltuximab (IL-6), Sorafenib (VEGFR, PDGFR, KIT, RAF),(Tocilizumab (IL-6R), Temsirolimus (mTOR), Tofacitinib (JAK3), Trametinib (MEK), Tositumomab (CD20), Trastuzumab (HER2), Vandetanib (EGFR), Vemurafenib (BRAF), Venetoclax (BCL2), Vismodegib (PTCH, Smoothened), Vorinostat (HDAC), Ziv-aflibercept (PIGF, VEGFA/B).
In some embodiments, the compound used according to the invention, the conjugate and/or the pharmaceutical composition is administered to the patient in combination with chemotherapy. As used herein, the term “chemotherapy” has its general meaning in the art and refers to the treatment that consists in administering to the patient a chemotherapeutic agent. Chemotherapeutic agents include, but are not limited to alkylating agents such as thiotepa and cyclosphosphamide; alkyl sulfonates such as busulfan, improsulfan and piposulfan; aziridines such as benzodopa, carboquone, meturedopa, and uredopa; ethylenimines and methylamelamines including altretamine, triethylenemelamine, trietylenephosphoramide, triethiylenethiophosphoramide and trimethylolomelamine; acetogenins (especially bullatacin and bullatacinone); a camptothecin (including the synthetic analogue topotecan); bryostatin; callystatin; CC-1065 (including its adozelesin, carzelesin and bizelesin synthetic analogues); cryptophycins (particularly cryptophycin 1 and cryptophycin 8); dolastatin; duocarmycin (including the synthetic analogues, KW-2189 and CB1-TM1); eleutherobin; pancratistatin; a sarcodictyin; spongistatin; nitrogen mustards such as chlorambucil, chlornaphazine, cholophosphamide, estramustine, ifosfamide, mechlorethamine, mechlorethamine oxide hydrochloride, melphalan, novembichin, phenesterine, prednimustine, trofosfamide, uracil mustard; nitrosureas such as carmustine, chlorozotocin, fotemustine, lomustine, nimustine, and ranimnustine; antibiotics such as the enediyne antibiotics (e.g., calicheamicin, especially calicheamicin gammall and calicheamicin omegall; dynemicin, including dynemicin A; bisphosphonates, such as clodronate; an esperamicin; as well as neocarzinostatin chromophore and related chromoprotein enediyne antiobiotic chromophores, aclacinomysins, actinomycin, authrarnycin, azaserine, bleomycins, cactinomycin, carabicin, caminomycin, carzinophilin, chromomycinis, dactinomycin, daunorubicin, detorubicin, 6-diazo-5- oxo-L-norleucine, doxorubicin (including morpholino-doxorubicin, cyanomorpholino- doxorubicin, 2-pyrrolino-doxorubicin and deoxy doxorubicin), epirubicin, esorubicin, idarubicin, marcellomycin, mitomycins such as mitomycin C, mycophenolic acid, nogalamycin, olivomycins, peplomycin, potfiromycin, puromycin, quelamycin, rodorubicin, streptonigrin, streptozocin, tubercidin, ubenimex, zinostatin, zorubicin; anti-metabolites such as methotrexate and 5-fluorouracil (5-FU); folic acid analogues such as denopterin, methotrexate, pteropterin, trimetrexate; purine analogs such as fludarabine, 6-mercaptopurine, thiamiprine, thioguanine; pyrimidine analogs such as ancitabine, azacitidine, 6-azauridine, carmofur, cytarabine, dideoxyuridine, doxifluridine, enocitabine, floxuridine; androgens such as calusterone, dromostanolone propionate, epitiostanol, mepitiostane, testolactone; anti-adrenals such as aminoglutethimide, mitotane, trilostane; folic acid replenisher such as frolinic acid; aceglatone; aldophosphamide glycoside; aminolevulinic acid; eniluracil; amsacrine; bestrabucil; bisantrene; edatraxate; defofamine; demecolcine; diaziquone; elformithine; elliptinium acetate; an epothilone; etoglucid; gallium nitrate; hydroxyurea; lentinan; lonidainine; maytansinoids such as maytansine and ansamitocins; mitoguazone; mitoxantrone; mopidanmol; nitraerine; pentostatin; phenamet; pirarubicin; losoxantrone; podophyllinic acid; 2-ethylhydrazide; methylhydrazine derivatives including N-methylhydrazine (MIH) and procarbazine; PSK polysaccharide complex); razoxane; rhizoxin; sizofuran; spirogermanium; tenuazonic acid; triaziquone; 2,2',2"-trichlorotriethylamine; trichothecenes (especially T-2 toxin, verracurin A, roridin A and anguidine); urethan; vindesine; dacarbazine; mannomustine; mitobronitol; mitolactol; pipobroman; gacytosine; arabinoside ("Ara- C"); cyclophosphamide; thiotepa; taxoids, e.g., paclitaxel and doxetaxel; chlorambucil; gemcitabine; 6-thioguanine; mercaptopurine; methotrexate; platinum coordination complexes such as cisplatin, oxaliplatin and carboplatin; vinblastine; platinum; etoposide (VP- 16); ifosfamide; mitoxantrone; vincristine; vinorelbine; novantrone; teniposide; edatrexate; daunomycin; aminopterin; xeloda; ibandronate; irinotecan (e.g., CPT-1 1); topoisomerase inhibitor RFS 2000; difluoromethylomithine (DMFO); retinoids such as retinoic acid; capecitabine; anthracyclines, nitrosoureas, antimetabolites, epipodophylotoxins, enzymes such as L-asparaginase; anthracenediones; hormones and antagonists including adrenocorticosteroid antagonists such as prednisone and equivalents, dexamethasone and aminoglutethimide; progestin such as hydroxyprogesterone caproate, medroxyprogesterone acetate and megestrol acetate; estrogen such as diethylstilbestrol and ethinyl estradiol equivalents; antiestrogen such as tamoxifen; androgens including testosterone propionate and fluoxymesterone/equivalents; antiandrogens such as flutamide, gonadotropin-releasing hormone analogs and leuprolide; and non-steroidal antiandrogens such as flutamide; and pharmaceutically acceptable salts, acids or derivatives of any of the above.
According to an embodiment, the compound used according to the invention, the conjugate and/or the pharmaceutical composition is administered to the patient in combination with radiotherapy. Suitable examples of radiation therapies include, but are not limited to external beam radiotherapy (such as superficial X-rays therapy, orthovoltage X-rays therapy, megavoltage X-rays therapy, radiosurgery, stereotactic radiation therapy, fractionated stereotactic radiation therapy, hypofractionated radiotherapy, cobalt therapy, electron therapy, fast neutron therapy, neutron-capture therapy, proton therapy, intensity modulated radiation therapy (IMRT), 3-dimensional conformal radiation therapy (3D-CRT) and the like; brachytherapy; unsealed source radiotherapy; tomotherapy and the like; or minibeam radiation therapy. Gamma rays are another form of photons used in radiotherapy. Gamma rays are produced spontaneously as certain elements (such as radium, uranium, cesium and cobalt 60) release radiation as they decompose, or decay. In some embodiments, radiotherapy may be hadrontherapy (using beams from charged particles like protons or other ions such as carbon), proton radiotherapy or proton minibeam radiation therapy. Proton radiotherapy is an ultra- precise form of radiotherapy that uses proton beams (Prezado Y, Jouvion G, Guardiola C, Gonzalez W, Juchaux M, Bergs J, Nauraye C, Labiod D, De Marzi L, Pouzoulet F, Patriarca A, Dendale R. Tumor Control in RG2 Glioma-Bearing Rats: A Comparison Between Proton Minibeam Therapy and Standard Proton Therapy. Int J Radiat Oncol Biol Phys. 2019 Jun 1 ;104(2):266-271 . doi: 10.1016/j.ijrobp.2019.01.080; Prezado Y, Jouvion G, Patriarca A, Nauraye C, Guardiola C, Juchaux M, Lamirault C, Labiod D, Jourdain L, Sebrie C, Dendale R, Gonzalez W, Pouzoulet F. Proton minibeam radiation therapy widens the therapeutic index for high-grade gliomas. Sci Rep. 2018 Nov 7;8(1):16479. doi: 10.1038/s41598-018-34796-8). Radiotherapy may also be FLASH radiotherapy (FLASH-RT) or FLASH proton irradiation. FLASH radiotherapy involves the ultra-fast delivery of radiation treatment at dose rates several orders of magnitude greater than those currently in routine clinical practice (ultra-high dose rate) (Favaudon V, Fouillade C, Vozenin MC. The radiotherapy FLASH to save healthy tissues. Med Sci (Paris) 2015 ; 31 : 121-123. DOI: 10.1051/medsci/20153102002); Patriarca A., Fouillade C. M., Martin F., Pouzoulet F., Nauraye C., et al. Experimental set-up for FLASH proton irradiation of small animals using a clinical system. Int J Radiat Oncol Biol Phys, 102 (2018), pp. 619-626. doi: 10.1016/j.ijrobp.2018.06.403. Epub 2018 Jul 11). Radiotherapy may also be hypofractionated radiotherapy (Shah JL, Li G, Shaffer JL, Azoulay Ml, Gibbs IC, Nagpal S, Soltys SG. Stereotactic Radiosurgery and Hypofractionated Radiotherapy for Glioblastoma. Neurosurgery. 2018 Jan 1 ;82(1):24-34, doi: 10.1093/neuros/nyx115; Botti M, Kirova YM, Dendale R, Savignoni A, Fromantin I, Gautier C, Bollet MA, Campana F, Fourquet A. Hypofractionated breast radiotherapy in 13 fractions, perfect tolerance or delayed early reaction? Prospective study of Curie Institute. Cancer Radiother. 2009 Apr;13(2):92-6, doi: 10.1016/j.canrad.2008.11 .009).
According to an embodiment, the cancer is selected from the group consisting of: soft tissue carcinoma, glioblastoma, head and neck carcinoma, lung carcinoma, cervix carcinoma, bladder carcinoma, rectum carcinoma, and anal carcinoma.
The compounds used according to the invention or the conjugates as defined above are particularly deemed useful for the treatment of cancer including solid tumors such as skin, breast, brain, cervical carcinomas, testicular carcinomas, etc. More particularly, cancers that may be treated by the compounds or conjugates of the invention include, but are not limited to: cardiac: sarcoma (angiosarcoma, fibrosarcoma, rhabdomyosarcoma, liposarcoma), myxoma, rhabdomyoma, fibroma, lipoma and teratoma; lung: bronchogenic carcinoma (squamous cell, undifferentiated small cell, undifferentiated large cell, adenocarcinoma), alveolar (bronchiolar) carcinoma, bronchial adenoma, sarcoma, lymphoma, chondromatous hamartoma, mesothelioma, gastrointestinal: esophagus (squamous cell carcinoma, adenocarcinoma, leiomyosarcoma, lymphoma), stomach (carcinoma, lymphoma, leiomyosarcoma), pancreas (ductal adenocarcinoma, insulinoma, glucagonoma, gastrinoma, carcinoid tumors, vipoma), small bowel (adenocarcinoma, lymphoma, carcinoid tumors, Karposi's sarcoma, leiomyoma, hemangioma, lipoma, neurofibroma, fibroma), large bowel (adenocarcinoma, tubular adenoma, Villous adenoma, hamartoma, leiomyoma); Genitourinary tract: kidney (adenocarcinoma, Wilm's tumor nephroblastoma, lymphoma, leukemia), bladder and urethra (Squamous cell carcinoma, transitional cell carcinoma, adenocarcinoma), prostate (adenocarcinoma, Sarcoma), testis (seminoma, teratoma, embryonal carcinoma, teratocarcinoma, choriocarcinoma, Sarcoma, interstitial cell carcinoma, fibroma, fibroadenoma, adenomatoid tumors, lipoma); Liver: hepatoma (hepatocellular carcinoma), cholangiocarcinoma, hepatoblastom, angiosarcoma, hepatocellular adenoma, hemangioma; Bone: osteogenic sarcoma (osteosarcoma), fibrosarcoma, malignant fibrous histiocytoma, chondrosarcoma, Ewing's sarcoma, malignant lymphoma (reticulum cell Sarcoma), multiple myeloma, malignant giant cell tumor chordoma, osteochronfroma (osteocartilaginous exostoses), benign chondroma, chondroblastoma, chondromyxofibroma, osteoid osteoma and giant cell tumors, Nervous System: skull (osteoma, hemangioma, granuloma, Xanthoma, Osteitis deformans), meninges (meningioma, meningiosarcoma, gliomatosis), brain (astrocytoma, medulloblastoma, glioma, ependymoma, germinomapinealoma, glioblastoma multiform, oligodendroglioma, Schwannoma, retinoblastoma, congenital tumors), Spinal cord (neurofibroma, meningioma, glioma, Sarcoma); Gynecological: uterus (endometrial carcinoma), cervix (cervical carcinoma, pre tumor cervical dysplasia), ovaries (ovarian carcinoma, Serous cystadenocarcinoma, mucinous cystadenocarcinoma, unclassified carcinoma, granulosa-thecal cell tumors, Sertoli-Leydig cell tumors, dysgerminoma, malignant teratoma), Vulva (Squamous cell carcinoma, intraepithelial carcinoma, adenocarcinoma, fibrosarcoma, melanoma), vagina (clear cell carcinoma, Squamous cell carcinoma, botryoid Sarcoma embryonal rhabdomyosarcoma, fallopian tubes (carcinoma); Hematologic: blood (myeloid leukemia acute and chronic, acute lymphoblastic leukemia, chronic lymphocytic leukemia, myeloproliferative diseases, multiple myeloma, myelodysplastic Syndrome), Hodgkin’s disease, non-Hodgkin’s lymphoma malignant lymphoma; Skin: malignant melanoma, basal cell carcinoma, Squamous cell carcinoma, Karposi's Sarcoma, moles dysplastic nevi, lipoma, angioma, dermatofibroma, keloids, psoriasis, and Adrenal glands: neuroblastoma.
According to an embodiment, the cancer is selected from the group consisting of: benign, metastatic and malignant neoplasias, and also including acral lentiginous melanoma, actinic keratoses, adenocarcinoma, adenoid cycstic carcinoma, adenomas, adenosarcoma, adenosquamous carcinoma, astrocytic tumors, Bartholin gland carcinoma, basal cell carcinoma, bronchial gland carcinomas, capillary, carcinoids, carcinoma, carcinosarcoma, cavernous, cholangiocarcinoma, chondosarcoma, choriod plexus papilloma/carcinoma, clear cell carcinoma, cystadenoma, endodermal sinus tumor, endometrial hyperplasia, endometrial stromal sarcoma, endometrioid adenocarcinoma, ependymal, epitheloid, Ewing’s sarcoma, fibrolamellar, focal nodular hyperplasia, gastrinoma, germ cell tumors, glioblastoma, glucagonoma, hemangiblastomas, hemangioendothelioma, hemangiomas, hepatic adenoma, hepatic adenomatosis, hepatocellular carcinoma, insulinoma, intaepithelial neoplasia, interepithelial squamous cell neoplasia, invasive squamous cell carcinoma, large cell carcinoma, leiomyosarcoma, lentigo maligna melanomas, malignant melanoma, malignant mesothelial tumors, medulloblastoma, medulloepithelioma, melanoma, meningeal, mesothelial, metastatic carcinoma, mucoepidermoid carcinoma, neuroblastoma neuroepithelial adenocarcinoma nodular melanoma, oat cell carcinoma, oligodendroglial, osteosarcoma, pancreatic polypeptide papillary serous adenocarcinoma, pineal cell, pituitary tumors, plasmacytoma, pseudosarcoma, pulmonary blastoma, renal cell carcinoma, retinoblastoma, rhabdomyosarcoma, sarcoma, serous carcinoma, small cell carcinoma, soft tissue carcinomas, somatostatin-secreting tumor, squamous carcinoma, squamous cell carcinoma, submesothelial, superficial spreading melanoma, undifferentiated carcinoma, uveal melanoma, verrucous carcinoma, vipoma, well differentiated carcinoma and Wilm's tumor.
The present invention, also relates to a method for treating the pathological conditions indicated above, which comprises the administration, to a patient, of an effective dose of a compound according to the invention, or a pharmaceutically acceptable salt thereof.
EXAMPLES
FIGURES
Figure 1 : Survival fraction of CCD-19Lu (triangles), ME-180 (circles) and LJ- 373 MG (squares) cell lines treated with increasing doses of X-rays (Philips/YXYLON generator) performed in the high throughput screening in 384- wells. Data represent mean ± standard errors of at least three independent experiments.
Figure 2: Dose-response curves of compound 1 on CCD-19Lu (A), ME-180 (B) and U-373 MG (C) cell lines without (upper curve) or following 4 (medium curve) or 6 (lower curve) Gy irradiation. Each dot represents an independent experiment (three replicates).
Figure 3: Dose-response curves of compound 2 on CCD-19Lu (A), ME-180 (B) and U-373 MG (C) cell lines without (upper curve) or following 4 (medium curve) or 6 (lower curve) Gy irradiation. Each dot represents an independent experiment (three replicates).
Figure 4: Dose-response curves of compound 3 on CCD-19Lu (A) and U-373 MG (B) cell lines without (upper curve) or following 4 (medium curve) or 6 (lower curve) Gy irradiation. Each dot represents an independent experiment (three replicates).
Figure 5: Dose-response curves of compound 4 on CCD-19Lu (A), ME-180 (B) and U-373 MG (C) cell lines without (upper curve) or following 4 (medium curve) or 6 (lower curve) Gy irradiation. Each dot represents an independent experiment (three replicates).
Figure 6: Dose-response curves of Olaparib® on CCD-19Lu (A), ME-180 (B) and U-373 MG (C) cell lines without (upper curve) or following 4 (medium curve) or 6 (lower curve) Gy irradiation. Each dot represents an independent experiment (three replicates). Figure 7: Isobolographic graph analysis of the association of compound 1 with radiation (4 and 6 Gy) on ME-180 (A) and U-373 MG (B) cell lines for a survival isoeffect of 50% (SI5o). The different replicates are all plotted under the envelope of additivity showing the supra-additive effect.
Figure 8: Isobolographic graph analysis of the association of compound 2 (A), compound 3 (B) and compound 4 (C) with 6 Gy-radiation on U-373 MG (B) cell line for a survival isoeffect of 50% (SI5o). The different replicates are all plotted under the envelope of additivity showing the supra-additive effect.
EXAMPLES
Example 1 : High-throughput molecules and irradiation screening
Small molecule library. For this screen 160 different molecules were used. All compounds were synthetized in-house (Institut Curie, UMR9187 / U1196 unit), dissolved in 100% dimethyl sulfoxide (DMSO) or water.
Compounds (1), (2), and (3) are in particular disclosed in Baladi T et al., Bioorg Med Chem., 26 (2018), 5510-5530, and compound (4) is in particular disclosed in Traore T. et al., Eur. J. Med Chem., 70 (2013), 789-901.
Cell lines and cell culture. The human glioblastoma cancer cell lines, U-373 MG (ECACC, 08061901 , radioresistant) and the cervical cancer cells ME-180 (ATCC, FITB-33, radiosensitive) were used in this study. Fluman pulmonary normal fibroblasts CCD-19Lu (ATCC, CCL-210) were used as normal tissue control. All cell lines were negative for mycoplasma contamination and were authentified by short tandem repeat (STR) profiling (Institut Curie, Genomic Platform). The appropriate cell plating density was adapted to a well of 384-well plate in the presence of their recommended cell media.
Irradiation in 384-well plates. Cells were seeded in wells of 384-well plate and allowed to attach overnight in complete growth media. Cell plates were then exposed to an orthovoltage X-ray generator Philips/YXYLON (13.1 mA intensity, 320 kVp voltage, filtered by 0.4 mm Sn, 0.25 mm Cu, 1 mm Al). The source-sample distance was 60 cm and a 5-cm PMMA block was placed under the sample to provide backscatter. The average dose-rate, over all wells of the plate, was 1.15 Gy.mirr1. Plates were exposed to variable doses of X-ray radiation alone or to two doses (4 or 6 Gy) for the screen. Survival fractions were calculated and the data was adjusted to best fit to the classical linear-quadratic equation. From this model, the radiobiological alpha and beta parameters were calculated in order to establish isobolographic analysis. Survival curves represent at least three independent experiments.
High-throughput chemo-irradiation screening. Cells were seeded on barcoded 384-well plates and allowed to attach overnight. Cells were then exposed to 11 -point concentration range of the 160 compounds (from 0.17 nM to 10 mM) and incubated at 37°C for 24 h. Cells were further mock irradiated (0 Gy) or irradiated at 4 or 6 Gy and incubated for additional 3 days at 37°C prior cell fixation and DAPI nuclei labelling.
Cell survival was determined using an automated high-throughput wide-field microscopy system (INCell 2200 - GE Healthcare). Six individual images were acquired for each well of the experimental plates and each labelled nucleus was detected using an automated nuclei segmentation custom-developed method. All survival curves represent at least three independent experiments.
Data processing. To normalize the survival data, the signals from the compound-treated wells were processed and expressed as a ratio of the 0 Gy DMSO/H2O control wells from each plate. For each cell line, cell count by well was multiplied by a normalization factor (n). Cell count was then converted as survival fraction. A 4-parameter log-logistic model (Hill equation) was used to fit the data. From this model, the area under the curve, the minimum, the maximum doses and the IC5o (dose leading to 50% effect on cell survival) were also computed. The first criterion is based on the isobolographic analysis.
Isobolograms. The combination of radiotherapy and chemotherapy is not easy to assess, as the dose-response curve of each treatment is nonlinear (according to the Hill equation for drug alone and to linear-quadratic model for radiation alone). In that case, to determine if the effect of the combined treatment is greater or less that would be expected on the basis of the response of each agent alone, Steel and Peckham developped the isobolographic concept based on the built of an envelope of additivity in an isoeffect plot (Steel G.G. and Peckham M.J., Int. J. Radiat. Oncol. Biol. Phys. (1979) 5:85-91 ). This envelope of additivity is based on the calculation of isoeffect curves according to Mode I (independent mechanisms of action of the two agents) and Mode II (dependent mechanisms of action of the two agents). This graph delimited three regions: the region of additivity inside the envelope, the region of supra-additivity (or synergy) under the envelope and the region of sub-additivity or antagonism above the envelope. The cytotoxic interactions of the selected compounds and radiation at 50% survival isoeffect (SI5o) were evaluated on the ME-180 and U-373 MG cancer cell lines using this method. The median and mean of the three SI5o replicates of combined treatments were calculated according to Hill equation after fitting the experimental curves obtained with 4 and/or 6 Gy and these points were plotted on the graph. Depending on the position inside the isobologramm, the effect of a given drug in combination with radiation was categorized as supra-additive, additive or antagonist.
Fibroblast toxicity. In tumor treatment, when using chemo-radiotherapy combination, the balance between tumor control and the risk of normal tissue side- effects is given by the therapeutic index of the treatment. The challenge is to improve this therapeutic index with a good control of the tumor and a low toxicity against normal tissues. In this study, a minimal toxicity on healthy tissues was taken into account thanks to a parallel study on fibroblasts. The second criterion based on IC5o filtered out the compounds that affected cell survival of the fibroblast cell line without irradiation (IC5o OGy CCD-19Lu > 9 x IC5o 4/6Gy cancer cells) to assess the drug toxicity and the third criterion filtered out the compounds that also affected cell survival of the fibroblast cell line with irradiation (IC5o 4/6 Gy CCD-19Lu > 9 x IC5o 4/6Gy cancer cells). The selected compounds presented in this invention passed through these three criteria, providing new non-toxic synthetized nitrogen-containing heterocyles derivatives for use as radiosensitizers in combination with radiation.
Results.
The survival curves of CCD-19Lu, ME-180 and U-373 MG cells exposed to increased doses of X-rays were obtained 72h post-irradiation (Figure 1). Based on the survival fraction at 4 (SF ) and 6 (SF6) Gy, the results showed that the most radioresistant is the cell line U-373 MG (SF4=0.75 ± 0.058; SF6=0.63 ± 0.059), following by the other cancer cell line ME-180 (SF4=0.74 ± 0.12; SF6=0.53 ± 0.07). The normal lung fibroblasts displayed a typical radiation dose-response profile due to a higher radiosensitivity and activation of senescent death pathways (Schreiber V. et al. (2006) PARP-2: Structure-Function Relationship. In: Poly(ADP-Ribosyl)ation. Molecular Biology Intelligence Unit. Springer, Boston, MA).
Over 160 compounds tested in this high-throughput screening (HTS) in combination with radiation, generating 1 ,920 dose-response curves, four new non toxic nitrogen derivatives were found to radiosensitize cancer cells with no corresponding toxic effects on normal fibroblasts.
Figure 2, 3, 4 and 5 showed the typical drug dose-response curves of increasing doses of compound 1 (Figure 2), compound 2 (Figure 3), compound 3 (Figure 4) and compound 4 (Figure 5) of the present invention on CCD-19Lu- (A),
ME-180 (B) and U-373 MG (C) cell lines without (upper curve) or following 4 (medium curve) or 6 (lower curve) Gy irradiation. Our process was validated by the use in a blind position of Olaparib®, a radiosensitizer now in clinical trials, as positive control in our FITS (Figure 6). As expected, this molecule was part of the selected supra-additive hits.
The first criterion of our FITS is the use of the isobolographic method. All the nitrogen-containing heterocycle derivatives compounds of the present invention showed supra-additive effects when combined with radiation. The cytotoxic interaction of the compound 1 , 2, 3 and 4 with radiation at 50% survival isoeffect (SI50) were evaluated on the cancer cell lines and is showed on Figure 7 and 8.
The SI5o of each replicate and their mean were plotted on the graph under the envelope of additivity and were categorized as supra-additive.

Claims

1. A compound having the following formula (I):
Figure imgf000032_0001
wherein:
R is: a group of formula (II):
Figure imgf000032_0002
wherein:
. Ri is H or is selected from the group consisting of: halogen, (Ci-C6)alkyl, aryl groups, and heteroaryl groups, and
. Ra and Rb are independently H or a (Ci-C6)alkyl group; or a group of formula (III):
Figure imgf000032_0003
wherein:
. R2 is H or is selected from the group consisting of: halogen, (Ci-Ce)alkyl, aryl groups, and heteroaryl groups;
. R is H or is selected from the group consisting of: halogen, (Ci-Ce)alkyl, aryl groups, and heteroaryl groups; said aryl and heteroaryl groups being optionally substituted with one or several substituents independently selected from: amino, hydroxyl, thiol, halogen, (Ci- C6)alkyl, (Ci-C6)alkoxy, (Ci-Ce)alkylthio, (Ci-C6)alkylamino, halo(Ci-C6)alkyl, carboxyl and carboxy(Ci-C6)alkyl;
. R3 is selected from the group consisting of: H, halogen, (Ci-C6)alkyl, aryl groups, and heteroaryl groups; said aryl and heteroaryl groups being optionally substituted with one or several substituents independently selected from: amino, hydroxyl, thiol, halogen, (Ci- C6)alkyl, (Ci-C6)alkoxy, (Ci-C6)alkylthio, (Ci-C6)alkylamino, halo(Ci-C6)alkyl, carboxyl and carboxy(Ci-C6)alkyl;
- R5 is chosen from the group consisting of: H, (Ci-C6)alkyl, and aryl, preferably phenyl, said aryl group being optionally substituted with one or several substituents independently selected from: amino, hydroxyl, thiol, halogen, (Ci- C6)alkyl, (Ci-C6)alkoxy, (Ci-C6)alkylthio, (Ci-C6)alkylamino, halo(Ci-C6)alkyl, carboxyl and carboxy(Ci-C6)alkyl;
- i is 1 or 2;
- the R groups, identical or different, are chosen from the group consisting of: H, halogen, and (Ci-C6)alkyl; or a pharmaceutically acceptable salt thereof, for use for the treatment of cancer as radiosensitizer.
2. The compound for the use of claim 1 , having the following formula (IV):
Figure imgf000033_0001
wherein:
- R, R , and i are as defined in claim 1 , and
- R6 is chosen from the group consisting of: amino, hydroxyl, thiol, halogen, (Ci-C6)alkyl, (Ci-C6)alkoxy, (Ci-C6)alkylthio, (Ci-C6)alkylamino, halo(Ci-C6)alkyl, carboxyl and carboxy(Ci-C6)alkyl.
3. The compound for the use of claim 2, wherein FTs is chosen from the group consisting of: hydroxyl, halogen, (Ci-C6)alkoxy, and halo(Ci-C6)alkyl.
4. The compound for the use of any one of claims 1 to 3, wherein said compound has the following formula (V):
Figure imgf000034_0001
R being as defined in claim 1 .
5. The compound for the use of any one of claims 1 to 4, wherein said compound has the following formula (VI):
Figure imgf000034_0002
Ri, Ra and Rb being as defined in claim 1 .
6. The compound for the use of any one of claims 1 to 4, wherein said compound has the following formula (VII):
Figure imgf000035_0001
R2, R3 and R4 being as defined in claim 1 .
7. The compound for the use of any one of claims 1 to 4, wherein said compound of formula (I) is one of the following compounds:
Figure imgf000035_0002
8. A conjugate molecule comprising one or more compound(s) of formula (I) as defined in any one of claims 1 to 7, covalently bound to at least one cell binding agent.
9. The conjugate of claim 8, for use as drug.
10. A medicament comprising the conjugate of claim or a pharmaceutically acceptable salt thereof.
11. The conjugate of claim 8, for use in treating cancer, optionally in combination with radiotherapy and/or with an anticancer drug.
12. The conjugate for the use of claim 11 , wherein the cancer is selected from the group consisting of: soft tissue carcinoma, glioblastoma, head and neck carcinoma, lung carcinoma, cervix carcinoma, bladder carcinoma, rectum carcinoma, and anal carcinoma.
13. A compound having the formula (I) as defined in any one of claims 1 to
7, for use in treating cancer, in combination with radiotherapy or in combination with radiotherapy and any anticancer drug.
14. The compound for the use of claim 13, wherein the cancer is selected from the group consisting of: soft tissue carcinoma, glioblastoma, head and neck carcinoma, lung carcinoma, cervix carcinoma, bladder carcinoma, rectum carcinoma, and anal carcinoma.
PCT/EP2022/056316 2021-03-12 2022-03-11 Nitrogen-containing heterocycles as radiosensitizers WO2022189618A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP21315035.2 2021-03-12
EP21315035 2021-03-12

Publications (1)

Publication Number Publication Date
WO2022189618A1 true WO2022189618A1 (en) 2022-09-15

Family

ID=75438695

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2022/056316 WO2022189618A1 (en) 2021-03-12 2022-03-11 Nitrogen-containing heterocycles as radiosensitizers

Country Status (1)

Country Link
WO (1) WO2022189618A1 (en)

Citations (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4880935A (en) 1986-07-11 1989-11-14 Icrf (Patents) Limited Heterobifunctional linking agents derived from N-succinimido-dithio-alpha methyl-methylene-benzoates
US5122368A (en) 1988-02-11 1992-06-16 Bristol-Myers Squibb Company Anthracycline conjugates having a novel linker and methods for their production
US5622929A (en) 1992-01-23 1997-04-22 Bristol-Myers Squibb Company Thioether conjugates
WO1997020574A1 (en) 1995-12-04 1997-06-12 The Regents Of The University Of California Blockade of t lymphocyte down-regulation associated with ctla-4 signaling
US5773578A (en) 1990-01-08 1998-06-30 Institut National De La Sante Et De La Recherche Medicale Proteins produced by human lymphocytes, DNA sequence encoding these proteins and their pharmaceutical and biological use
US5824805A (en) 1995-12-22 1998-10-20 King; Dalton Branched hydrazone linkers
WO2004004771A1 (en) 2002-07-03 2004-01-15 Ono Pharmaceutical Co., Ltd. Immunopotentiating compositions
WO2004056875A1 (en) 2002-12-23 2004-07-08 Wyeth Antibodies against pd-1 and uses therefor
US20050245530A1 (en) * 2004-04-23 2005-11-03 Borzilleri Robert M Monocyclic heterocycles as kinase inhibitors
US6984720B1 (en) 1999-08-24 2006-01-10 Medarex, Inc. Human CTLA-4 antibodies
US7109003B2 (en) 1998-12-23 2006-09-19 Abgenix, Inc. Methods for expressing and recovering human monoclonal antibodies to CTLA-4
WO2006121168A1 (en) 2005-05-09 2006-11-16 Ono Pharmaceutical Co., Ltd. Human monoclonal antibodies to programmed death 1(pd-1) and methods for treating cancer using anti-pd-1 antibodies alone or in combination with other immunotherapeutics
WO2007123737A2 (en) 2006-03-30 2007-11-01 University Of California Methods and compositions for localized secretion of anti-ctla-4 antibodies
WO2008156712A1 (en) 2007-06-18 2008-12-24 N. V. Organon Antibodies to human programmed death receptor pd-1
WO2009014708A2 (en) 2007-07-23 2009-01-29 Cell Genesys, Inc. Pd-1 antibodies in combination with a cytokine-secreting cell and methods of use thereof
WO2009114335A2 (en) 2008-03-12 2009-09-17 Merck & Co., Inc. Pd-1 binding proteins
US8017114B2 (en) 1999-08-24 2011-09-13 Medarex, Inc. Human CTLA-4 antibodies and their uses
WO2013043569A1 (en) 2011-09-20 2013-03-28 Vical Incorporated Synergistic anti-tumor efficacy using alloantigen combination immunotherapy
US20130177557A1 (en) 2010-03-26 2013-07-11 Randolph J. Noelle Vista regulatory t cell mediator protein, vista binding agents and use thereof
WO2014047350A1 (en) 2012-09-20 2014-03-27 Morningside Technology Ventures Ltd. Oncolytic virus encoding pd-1 binding agents and uses of the same

Patent Citations (22)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4880935A (en) 1986-07-11 1989-11-14 Icrf (Patents) Limited Heterobifunctional linking agents derived from N-succinimido-dithio-alpha methyl-methylene-benzoates
US5122368A (en) 1988-02-11 1992-06-16 Bristol-Myers Squibb Company Anthracycline conjugates having a novel linker and methods for their production
US5773578A (en) 1990-01-08 1998-06-30 Institut National De La Sante Et De La Recherche Medicale Proteins produced by human lymphocytes, DNA sequence encoding these proteins and their pharmaceutical and biological use
US5622929A (en) 1992-01-23 1997-04-22 Bristol-Myers Squibb Company Thioether conjugates
WO1997020574A1 (en) 1995-12-04 1997-06-12 The Regents Of The University Of California Blockade of t lymphocyte down-regulation associated with ctla-4 signaling
US5824805A (en) 1995-12-22 1998-10-20 King; Dalton Branched hydrazone linkers
US7109003B2 (en) 1998-12-23 2006-09-19 Abgenix, Inc. Methods for expressing and recovering human monoclonal antibodies to CTLA-4
US8491895B2 (en) 1998-12-23 2013-07-23 Amgen Fremont Inc. Methods of treating cancer with human monoclonal antibodies to CTLA-4
US8143379B2 (en) 1998-12-23 2012-03-27 Amgen Fremont Inc. Human monoclonal antibodies to CTLA-4
US8017114B2 (en) 1999-08-24 2011-09-13 Medarex, Inc. Human CTLA-4 antibodies and their uses
US6984720B1 (en) 1999-08-24 2006-01-10 Medarex, Inc. Human CTLA-4 antibodies
WO2004004771A1 (en) 2002-07-03 2004-01-15 Ono Pharmaceutical Co., Ltd. Immunopotentiating compositions
WO2004056875A1 (en) 2002-12-23 2004-07-08 Wyeth Antibodies against pd-1 and uses therefor
US20050245530A1 (en) * 2004-04-23 2005-11-03 Borzilleri Robert M Monocyclic heterocycles as kinase inhibitors
WO2006121168A1 (en) 2005-05-09 2006-11-16 Ono Pharmaceutical Co., Ltd. Human monoclonal antibodies to programmed death 1(pd-1) and methods for treating cancer using anti-pd-1 antibodies alone or in combination with other immunotherapeutics
WO2007123737A2 (en) 2006-03-30 2007-11-01 University Of California Methods and compositions for localized secretion of anti-ctla-4 antibodies
WO2008156712A1 (en) 2007-06-18 2008-12-24 N. V. Organon Antibodies to human programmed death receptor pd-1
WO2009014708A2 (en) 2007-07-23 2009-01-29 Cell Genesys, Inc. Pd-1 antibodies in combination with a cytokine-secreting cell and methods of use thereof
WO2009114335A2 (en) 2008-03-12 2009-09-17 Merck & Co., Inc. Pd-1 binding proteins
US20130177557A1 (en) 2010-03-26 2013-07-11 Randolph J. Noelle Vista regulatory t cell mediator protein, vista binding agents and use thereof
WO2013043569A1 (en) 2011-09-20 2013-03-28 Vical Incorporated Synergistic anti-tumor efficacy using alloantigen combination immunotherapy
WO2014047350A1 (en) 2012-09-20 2014-03-27 Morningside Technology Ventures Ltd. Oncolytic virus encoding pd-1 binding agents and uses of the same

Non-Patent Citations (16)

* Cited by examiner, † Cited by third party
Title
BALADI T ET AL., BIOORG MED CHEM., vol. 26, 2018, pages 5510 - 5530
BALADI TOM ET AL: "Design, synthesis, biological evaluation and cellular imaging of imidazo[4,5-b]pyridine derivatives as potent and selective TAM inhibitors", BIOORGANIC & MEDICINAL CHEMISTRY, vol. 26, no. 20, 1 November 2018 (2018-11-01), AMSTERDAM, NL, pages 5510 - 5530, XP055827306, ISSN: 0968-0896, DOI: 10.1016/j.bmc.2018.09.031 *
BOTTI MKIROVA YMDENDALE RSAVIGNONI AFROMANTIN IGAUTIER CBOLLET MACAMPANA FFOURQUET A: "Hypofractionated breast radiotherapy in 13 fractions, perfect tolerance or delayed early reaction?", PROSPECTIVE STUDY OF CURIE INSTITUTE. CANCER RADIOTHER, vol. 13, no. 2, April 2009 (2009-04-01), pages 92 - 6
DUBOWCHIKWALKER, PHARM. THERAPEUTICS, vol. 83, 1999, pages 67 - 123
FAVAUDON VFOUILLADE CVOZENIN MC: "The radiotherapy FLASH to save healthy tissues", MED SCI (PARIS, vol. 31, 2015, pages 121 - 123
NEVILLE ET AL., BIOL. CHEM., vol. 264, 1989, pages 14653 - 14661
PATRIARCA A.FOUILLADE C. M.MARTIN F.POUZOULET F.NAURAYE C. ET AL.: "Experimental set-up for FLASH proton irradiation of small animals using a clinical system", INT J RADIAT ONCOL BIOL PHYS, vol. 102, 2018, pages 619 - 626, XP085474451, DOI: 10.1016/j.ijrobp.2018.06.403
PREZADO YJOUVION GGUARDIOLA CGONZALEZ WJUCHAUX MBERGS JNAURAYE CLABIOD DDE MARZI LPOUZOULET F: "Tumor Control in RG2 Glioma-Bearing Rats: A Comparison Between Proton Minibeam Therapy and Standard Proton Therapy", INT J RADIAT ONCOL BIOL PHYS, vol. 104, no. 2, 1 June 2019 (2019-06-01), pages 266 - 271
PREZADO YJOUVION GPATRIARCA ANAURAYE CGUARDIOLA CJUCHAUX MLAMIRAULT CLABIOD DJOURDAIN LSEBRIE C: "Proton minibeam radiation therapy widens the therapeutic index for high-grade gliomas", SCI REP, vol. 8, no. 1, 7 November 2018 (2018-11-07), pages 16479
SCHREIBER V ET AL.: "Poly(ADP-Ribosyl)ation. Molecular Biology Intelligence Unit", 2006, SPRINGER, article "PARP-2: Structure-Function Relationship"
SHAH JLLI GSHAFFER JLAZOULAY MIGIBBS ICNAGPAL SSOLTYS SG: "Stereotactic Radiosurgery and Hypofractionated Radiotherapy for Glioblastoma", NEUROSURGERY, vol. 82, no. 1, 1 January 2018 (2018-01-01), pages 24 - 34
STEEL G.G.PECKHAM M.J., INT. J. RADIAT. ONCOL. BIOL. PHYS., vol. 5, 1979, pages 85 - 91
THORPE ET AL., CANCER RES., vol. 47, 1987, pages 5924 - 5931
TRAORE T. ET AL., EUR. J. MED CHEM., vol. 70, 2013, pages 789 - 901
TRAORÉ TÉNIN ET AL: "New aminopyrimidine derivatives as inhibitors of the TAM family", EUROPEAN JOURNAL OF MEDICINAL CHEMISTRY, ELSEVIER, AMSTERDAM, NL, vol. 70, 22 October 2013 (2013-10-22), pages 789 - 801, XP028794184, ISSN: 0223-5234, DOI: 10.1016/J.EJMECH.2013.10.037 *
WAWRZYNCZAK ET AL.: "In Immunoconjugates: Antibody Conjugates in Radioimagery and Therapy of Cancer", 1987, OXFORD U. PRESS

Similar Documents

Publication Publication Date Title
Sanghera et al. Chemoradiotherapy for rectal cancer: an updated analysis of factors affecting pathological response
CN106572993B (en) Application of the EP4 antagonist in the drug of preparation treating cancer
US10836735B2 (en) Compositions and methods for treating cancers
KR20190018400A (en) Antibody-drug conjugate comprising antibody binding to epidermal growth factor receptor variant III
WO2023035611A1 (en) Pharmaceutical composition and use thereof
WO2019072220A1 (en) Use of pd-1 antibody combined with epigenetic regulator in preparation of drug for treating tumors
EP3493797A1 (en) Cobicistat for use in cancer treatments
EP4076465B1 (en) Once daily cancer treatment regimen with a prmt5 inhibitor
CN114302745A (en) Combination therapy comprising an anti-CD 19 antibody drug conjugate and a PI3K inhibitor or a second agent
WO2022189618A1 (en) Nitrogen-containing heterocycles as radiosensitizers
WO2021224186A1 (en) New pyridine derivatives as radiosensitizers
US20230303604A1 (en) Metallic trans-(n-heterocyclic carbene)-amine-platinum complexes and uses thereof for treating cancer
JPWO2021182571A5 (en)
JPWO2021182574A5 (en)
CA2717100A1 (en) Use of lucanthone in combination with an anti-metabolite to treat cancer
US20230372382A1 (en) Use of adenosine diphosphate ribose for adjuvant therapy with radiation and/or anti-cancer treatment
US20240091351A1 (en) FOCAL IONIZING RADIATION AND CD47/SIRPa DISRUPTION ANTICANCER COMBINATION THERAPY
WO2024061203A1 (en) Pharmaceutical composition and use thereof
WO2023011879A1 (en) Scanning dynamic device for minibeams production
WO2012145671A1 (en) Radioisotope-photodynamic therapy for cancer treatment

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: 22713418

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: 22713418

Country of ref document: EP

Kind code of ref document: A1